US20040010134A1 - Albumin fusion proteins - Google Patents

Albumin fusion proteins Download PDF

Info

Publication number
US20040010134A1
US20040010134A1 US09/833,245 US83324501A US2004010134A1 US 20040010134 A1 US20040010134 A1 US 20040010134A1 US 83324501 A US83324501 A US 83324501A US 2004010134 A1 US2004010134 A1 US 2004010134A1
Authority
US
United States
Prior art keywords
cancer
immune
hematopoietic
fragment
variant
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US09/833,245
Inventor
Craig Rosen
William Haseltine
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Human Genome Sciences Inc
Original Assignee
Human Genome Sciences Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Family has litigation
First worldwide family litigation filed litigation Critical https://patents.darts-ip.com/?family=27394014&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=US20040010134(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Human Genome Sciences Inc filed Critical Human Genome Sciences Inc
Priority to US09/833,245 priority Critical patent/US20040010134A1/en
Assigned to HUMAN GENOME SCIENCES, INC. reassignment HUMAN GENOME SCIENCES, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: HASELTINE, WILLIAM A., ROSEN, CRAIG A.
Priority to US10/472,964 priority patent/US20070032414A1/en
Priority to US10/472,965 priority patent/US20070026454A1/en
Publication of US20040010134A1 publication Critical patent/US20040010134A1/en
Priority to US10/868,184 priority patent/US20070048818A1/en
Priority to US11/264,096 priority patent/US20060084794A1/en
Priority to US11/545,766 priority patent/US20070099833A1/en
Priority to US12/570,397 priority patent/US20100286048A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/43Enzymes; Proenzymes; Derivatives thereof
    • A61K38/46Hydrolases (3)
    • A61K38/48Hydrolases (3) acting on peptide bonds (3.4)
    • A61K38/482Serine endopeptidases (3.4.21)
    • A61K38/4846Factor VII (3.4.21.21); Factor IX (3.4.21.22); Factor Xa (3.4.21.6); Factor XI (3.4.21.27); Factor XII (3.4.21.38)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/19Cytokines; Lymphokines; Interferons
    • A61K38/21Interferons [IFN]
    • A61K38/212IFN-alpha
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/16Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • A61K38/17Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • A61K38/38Albumins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/18Drugs for disorders of the alimentary tract or the digestive system for pancreatic disorders, e.g. pancreatic enzymes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/02Drugs for disorders of the urinary system of urine or of the urinary tract, e.g. urine acidifiers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/08Drugs for disorders of the urinary system of the prostate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P13/00Drugs for disorders of the urinary system
    • A61P13/12Drugs for disorders of the urinary system of the kidneys
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/08Drugs for genital or sexual disorders; Contraceptives for gonadal disorders or for enhancing fertility, e.g. inducers of ovulation or of spermatogenesis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/10Drugs for genital or sexual disorders; Contraceptives for impotence
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • A61P15/18Feminine contraceptives
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/02Drugs for dermatological disorders for treating wounds, ulcers, burns, scars, keloids, or the like
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/12Keratolytics, e.g. wart or anti-corn preparations
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/14Drugs for dermatological disorders for baldness or alopecia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/08Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease
    • A61P19/10Drugs for skeletal disorders for bone diseases, e.g. rachitism, Paget's disease for osteoporosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/02Drugs for disorders of the nervous system for peripheral neuropathies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/14Drugs for disorders of the nervous system for treating abnormal movements, e.g. chorea, dyskinesia
    • A61P25/16Anti-Parkinson drugs
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/12Drugs for disorders of the metabolism for electrolyte homeostasis
    • A61P3/14Drugs for disorders of the metabolism for electrolyte homeostasis for calcium homeostasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/16Antivirals for RNA viruses for influenza or rhinoviruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/14Antivirals for RNA viruses
    • A61P31/18Antivirals for RNA viruses for HIV
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • A61P31/20Antivirals for DNA viruses
    • A61P31/22Antivirals for DNA viruses for herpes viruses
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/02Antiprotozoals, e.g. for leishmaniasis, trichomoniasis, toxoplasmosis
    • A61P33/06Antimalarials
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P33/00Antiparasitic agents
    • A61P33/10Anthelmintics
    • A61P33/12Schistosomicides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/08Antiallergic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P39/00General protective or antinoxious agents
    • A61P39/02Antidotes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P41/00Drugs used in surgical methods, e.g. surgery adjuvants for preventing adhesion or for vitreum substitution
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/10Drugs for disorders of the endocrine system of the posterior pituitary hormones, e.g. oxytocin, ADH
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/38Drugs for disorders of the endocrine system of the suprarenal hormones
    • A61P5/40Mineralocorticosteroids, e.g. aldosterone; Drugs increasing or potentiating the activity of mineralocorticosteroids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/04Antihaemorrhagics; Procoagulants; Haemostatic agents; Antifibrinolytic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/06Antiarrhythmics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/12Antihypertensives
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/52Cytokines; Lymphokines; Interferons
    • C07K14/555Interferons [IFN]
    • C07K14/56IFN-alpha
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/61Growth hormones [GH] (Somatotropin)
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/62Insulins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/575Hormones
    • C07K14/65Insulin-like growth factors (Somatomedins), e.g. IGF-1, IGF-2
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • C07K14/715Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons
    • C07K14/7151Receptors; Cell surface antigens; Cell surface determinants for cytokines; for lymphokines; for interferons for tumor necrosis factor [TNF], for lymphotoxin [LT]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/76Albumins
    • C07K14/765Serum albumin, e.g. HSA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/62DNA sequences coding for fusion proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/96Stabilising an enzyme by forming an adduct or a composition; Forming enzyme conjugates
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01KANIMAL HUSBANDRY; CARE OF BIRDS, FISHES, INSECTS; FISHING; REARING OR BREEDING ANIMALS, NOT OTHERWISE PROVIDED FOR; NEW BREEDS OF ANIMALS
    • A01K2217/00Genetically modified animals
    • A01K2217/05Animals comprising random inserted nucleic acids (transgenic)
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K48/00Medicinal preparations containing genetic material which is inserted into cells of the living body to treat genetic diseases; Gene therapy
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/21Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a His-tag
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/31Fusion polypeptide fusions, other than Fc, for prolonged plasma life, e.g. albumin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/50Fusion polypeptide containing protease site
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/70Fusion polypeptide containing domain for protein-protein interaction
    • C07K2319/74Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor
    • C07K2319/75Fusion polypeptide containing domain for protein-protein interaction containing a fusion for binding to a cell surface receptor containing a fusion for activation of a cell surface receptor, e.g. thrombopoeitin, NPY and other peptide hormones
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Definitions

  • the invention relates generally to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or fragments or variants of albumin.
  • the invention further relates to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or fragments or variants of albumin, that exhibit extended shelf-life and/or extended or therapeutic activity in solution.
  • These fusion proteins are herein collectively referred to as “albumin fusion proteins of the invention.”
  • the invention encompasses therapeutic albumin fusion proteins, compositions, pharmaceutical compositions, formulations and kits.
  • Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention using these nucleic acids, vectors, and/or host cells.
  • the invention is also directed to methods of in vitro stabilizing a Therapeutic protein via fusion or conjugation of the Therapeutic protein to albumin or fragments or variants of albumin.
  • HSA Human serum albumin
  • HA a protein of 585 amino acids in its mature form (as shown in FIG. 15 or in SEQ ID NO:18), is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands.
  • HSA Human serum albumin
  • rHA recombinant HA
  • albumin As a carrier molecule and its inert nature are desirable properties for use as a carrier and transporter of polypeptides in vivo.
  • the use of albumin as a component of an albumin fusion protein as a carrier for various proteins has been suggested in WO 93/15199, WO 93/15200, and EP 413 622.
  • the use of N-terminal fragments of HA for fusions to polypeptides has also been proposed (EP 399 666). Fusion of albumin to the Therapeutic protein may be achieved by genetic manipulation, such that the DNA coding for HA, or a fragment thereof, is joined to the DNA coding for the Therapeutic protein.
  • a suitable host is then transformed or transfected with the fused nucleotide sequences, so arranged on a suitable plasmid as to express a fusion polypeptide.
  • the expression may be effected in vitro from, for example, prokaryotic or eukaryotic cells, or in vivo e.g. from a transgenic organism.
  • Storage problems are particularly acute when pharmaceutical formulations must be stored and dispensed outside of the hospital environment.
  • Many protein and peptide drugs also require the addition of high concentrations of other protein such as albumin to reduce or prevent loss of protein due to binding to the container. This is a major concern with respect to proteins such as IFN. For this reason, many Therapeutic proteins are formulated in combination with large proportion of albumin carrier molecule (100-1000 fold excess), though this is an undesirable and expensive feature of the formulation.
  • the present invention is based, in part, on the discovery that Therapeutic proteins may be stabilized to extend the shelf-life, and/or to retain the Therapeutic protein's activity for extended periods of time in solution, in vitro and/or in vivo, by genetically or chemically fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to stabilize the protein and/or its activity.
  • albumin-fusion proteins or albumin conjugated proteins may reduce the need to formulate protein solutions with large excesses of carrier proteins (such as albumin, unfused) to prevent loss of Therapeutic proteins due to factors such as binding to the container.
  • the present invention encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin.
  • the present invention also encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life of the Therapeutic protein, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo.
  • Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention and using these nucleic acids, vectors, and/or host cells.
  • the invention also encompasses pharmaceutical formulations comprising an albumin fusion protein of the invention and a pharmaceutically acceptable diluent or carrier.
  • Such formulations may be in a kit or container.
  • kit or container may be packaged with instructions pertaining to the extended shelf life of the Therapeutic protein.
  • Such formulations may be used in methods of treating, preventing, ameliorating or diagnosing a disease or disease symptom in a patient, preferably a mammal, most preferably a human, comprising the step of administering the pharmaceutical formulation to the patient.
  • the present invention encompasses methods of preventing treating, or ameliorating a disease or disorder.
  • the present invention encompasses a method of treating a disease or disorder listed in the “Preferred Indication Y” column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein portion corresponding to a Therapeutic protein (or fragment or variant thereof) disclosed in the “Therapeutic Protein X” column of Table 1 (in the same row as the disease or disorder to be treated is listed in the “Preferred Indication Y” column of Table 1) in an amount effective to treat prevent or ameliorate the disease or disorder.
  • the invention includes a method of extending the shelf life of a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to extend the shelf-life of the Therapeutic protein.
  • a Therapeutic protein e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof
  • the Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin.
  • the Therapeutic protein used according to this method is fused to albumin, or a fragment or variant of albumin, via recombinant DNA technology or genetic engineering.
  • the invention includes a method of stabilizing a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) in solution, comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to stabilize the Therapeutic protein.
  • a Therapeutic protein e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof
  • the Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin.
  • the Therapeutic protein used according to this method is fused to albumin, or a fragment or variant of albumin, via recombinant DNA technology or genetic engineering.
  • the present invention further includes transgenic organisms modified to contain the nucleic acid molecules of the invention, preferably modified to express the albumin fusion proteins encoded by the nucleic acid molecules.
  • FIG. 1 depicts the extended shelf-life of an HA fusion protein in terms of the biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to 5 weeks at 37° C. Under these conditions, hGH has no observed activity by week 2.
  • FIG. 2 depicts the extended shelf-life of an HA fusion protein in terms of the stable biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to 3 weeks at 4, 37, or 50° C. Data is normalized to the biological activity of hGH at time zero.
  • FIGS. 3A and 3B compare the biological activity of HA-hGH with hGH in the Nb2 cell proliferation assay.
  • FIG. 3A shows proliferation after 24 hours of incubation with various concentrations of hGH or the albumin fusion protein
  • FIG. 3B shows proliferation after 48 hours of incubation with various concentrations of hGH or the albumin fusion protein.
  • FIG. 4 shows a map of a plasmid (pPPC0005) that can be used as the base vector into which polynucleotides encoding the Therapeutic proteins (including polypeptide and fragments and variants thereof) may be cloned to form HA-fusions.
  • Plasmid Map key PRB1p: PRB1 S. cerevisiae promoter; FL: Fusion leader sequence; rHA: cDNA encoding HA; ADH1t: ADH1 S. cerevisiae terminator; T3: T3 sequencing primer site; T7: T7 sequencing primer site; Amp R: ⁇ -lactamase gene; ori: origin of replication.
  • the plasmid in FIG. 4 was labeled pPPC0006, instead of pPPC0005.
  • the drawing of this plasmid did not show certain pertinent restriction sites in this vector.
  • the drawing is labeled pPPC0005 and more restriction sites of the same vector are shown.
  • FIG. 5 compares the recovery of vial-stored HA-IFN solutions of various concentrations with a stock solution after 48 or 72 hours of storage.
  • FIG. 6 compares the activity of an HA- ⁇ -IFN fusion protein after administration to monkeys via TV or SC.
  • FIG. 7 describes the bioavailability and stability of an HA- ⁇ -IFN fusion protein.
  • FIG. 8 is a map of an expression vector for the production of HA- -IFN.
  • FIG. 9 shows the location of loops in HA.
  • FIG. 10 is an example of the modification of an HA loop.
  • FIG. 11 is a representation of the HA loops.
  • FIG. 12 shows the HA loop IV.
  • FIG. 13 shows the tertiary structure of HA.
  • FIG. 14 shows an example of a scFv-HA fusion
  • FIG. 15 shows the amino acid sequence of the mature form of human albumin (SEQ ID NO:18) and a polynucleotide encoding it (SEQ ID NO:17).
  • the present invention is based, in part, on the discovery that a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) may be stabilized to extend the shelf-life and/or retain the Therapeutic protein's activity for extended periods of time in solution (or in a pharmaceutical composition) in vitro and/or in vivo, by genetically fusing or chemically conjugating the Therapeutic protein, polypeptide or peptide to all or a portion of albumin sufficient to stabilize the protein and its activity.
  • a Therapeutic protein e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof
  • albumin fusion protein refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof).
  • An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or chemical conjugation to one another.
  • the Therapeutic protein and albumin protein once part of the albumin fusion protein, may be referred to as a “portion”, “region” or “moiety” of the albumin fusion protein (e.g., a “Therapeutic protein portion” or an “albumin protein portion”).
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein.
  • the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin.
  • the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is the extracellular soluble domain of the Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is the active form of the Therapeutic protien.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin.
  • an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion or chemical conjugation.
  • Therapeutic protein refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities.
  • Therapeutic proteins encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, and biologics. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) It is specifically contemplated that the term “Therapeutic protein” encompasses antibodies and fragments and variants thereof.
  • an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody.
  • the term “Therapeutic protein” may refer to the endogenous or naturally occurring correlate of a Therapeutic protein.
  • a polypeptide displaying a “therapeutic activity” or a protein that is “therapeutically active” is meant a polypeptide that possesses one or more known biological and/or therapeutic activities associated with a therapeutic protein such as one or more of the Therapeutic proteins described herein or otherwise known in the art.
  • a “Therapeutic protein” is a protein that is useful to treat, prevent or ameliorate a disease, condition or disorder.
  • a “Therapeutic protein” may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically.
  • a particular cell type normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)
  • a compound drug, or cytotoxic agent
  • a “Therapeutic protein” is a protein that has a biological activity, and in particular, a biological activity that is useful for treating preventing or ameliorating a disease.
  • a non-inclusive list of biological activities that may be possessed by a Therapeutic protein includes, enhancing the immune response, promoting angiogenesis, inhibiting angiogenesis, regulating hematopoietic functions, stimulating nerve growth, enhancing an immune response, inhibiting an immune response, or any one or more of the biological activities described in the “Biological Activities” section below.
  • therapeutic activity may refer to an activity whose effect is consistent with a desirable therapeutic outcome in humans, or to desired effects in non-human mammals or in other species or organisms.
  • Therapeutic activity may be measured in vivo or in vitro.
  • a desirable effect may be assayed in cell culture.
  • hGH is the Therapeutic protein
  • the effects of hGH on cell proliferation as described in Example 1 may be used as the endpoint for which therapeutic activity is measured.
  • Such in vitro or cell culture assays are commonly available for many Therapeutic proteins as described in the art. Examples of assays include, but are not limited to those described herein in the Examples section or in the “Exemplary Activity Assay” column of Table 1.
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention are often modified by the attachment of one or more oligosaccharide groups.
  • the modification referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone.
  • glycosylation characterized by O-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser/Thr sequence, where X can be any amino acid except proline.
  • N-acetylneuramic acid also known as sialic acid
  • Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type.
  • Natural human interferon- ⁇ 2 is O-glycosylated at threonine 106, and N-glycosylation occurs at asparagine 72 in interferon- ⁇ 14 (Adolf et al., J. Biochem 276:511 (1991); Nyman T A et al., J. Biochem 329:295 (1998)).
  • the oligosaccharides at asparagine 80 in natural interferon- ⁇ 1 ⁇ may play an important factor in the solubility and stability of the protein, but may not be essential for its biological activity.
  • Interferon- ⁇ contains two N-linked oligosaccharide chains at positions 25 and 97, both important for the efficient formation of the bioactive recombinant protein, and having an influence on the pharmacokinetic properties of the protein (Sareneva et al., Eur. J. Biochem 242:191 (1996); Sareneva et al, Biochem J. 303:831 (1994); Sareneva et al., J. Interferon Res. 13:267 (1993)).
  • N-linked glycosylation occurs at asparagine residues located at positions 24, 38 and 83 while O-linked glycosylation occurs at a serine residue located at position 126 (Lai et al., J. Biol. Chem. 261:3116 (1986); Broudy et al., Arch. Biochem. Biophys. 265:329 (1988)).
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention may be modified so that glycosylation at one or more sites is altered as a result of manipulation(s) of their nucleic acid sequence, by the host cell in which they are expressed, or due to other conditions of their expression.
  • glycosylation isomers may be produced by abolishing or introducing glycosylation sites, e.g., by substitution or deletion of amino acid residues, such as substitution of glutamine for asparagine, or unglycosylated recombinant proteins may be produced by expressing the proteins in host cells that will not glycosylate them, e.g. in E. coli or glycosylation-deficient yeast.
  • Therapeutic proteins are well known in the art and available in public databases such as Chemical Abstracts Services Databases (e.g., the CAS Registry), GenBank, and GenSeq as shown in Table 1.
  • Additional Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, one or more of the Therapeutic proteins or peptides disclosed in the “Therapeutic Protein X” column of Table 1, or fragment or variable thereof.
  • Table 1 provides a non-exhaustive list of Therapeutic proteins that correspond to a Therapeutic protein portion of an albumin fusion protein of the invention.
  • the “Therapeutic Protein X” column discloses Therapeutic protein molecules followed by parentheses containing scientific and brand names that comprise, or alternatively consist of, that Therapeutic protein molecule or a fragment or variant thereof.
  • “Therapeutic protein X” as used herein may refer either to an individual Therapeutic protein molecule (as defined by the amino acid sequence obtainable from the CAS and Genbank accession numbers), or to the entire group of Therapeutic proteins associated with a given Therapeutic protein molecule disclosed in this column.
  • the “Exemplary Identifier” column provides Chemical Abstracts Services (CAS) Registry Numbers (published by the American Chemical Society) and/or Genbank Accession Numbers ((e.g., Locus ID, NP_XXXXX (Reference Sequence Protein), and XP_XXXXX (Model Protein) identifiers available through the national Center for Biotechnology Information (NCBI) webpage at www.ncbi.nlm.nih.gov) that correspond to entries in the CAS Registry or Genbank database which contain an amino acid sequence of the Therapeutic Protein Molecule or of a fragment or variant of the Therapeutic Protein Molecule.
  • CAS Chemical Abstracts Services
  • Genbank Accession Numbers (e.g., Locus ID, NP_XXXXX (Reference Sequence Protein), and XP_XXXXX (Model Protein) identifiers available through the national Center for Biotechnology Information (NCBI) webpage at www.ncbi.nlm.nih.gov) that correspond
  • GenSeq Accession numbers and/or journal publication citations are given to identify the exemplary amino acid sequence for some polypeptides.
  • the summary pages associated with each of these CAS and Genbank and GenSeq Accession Numbers as well as the cited journal publications are each incorporated by reference in their entireties, particularly with respect to the amino acid sequences described therein.
  • the “PCT/Patent Reference” column provides U.S. Patent numbers, or PCT International Publication Numbers corresponding to patents and/or published patent applications that describe the Therapeutic protein molecule.
  • Each of the patents and/or published patent applications cited in the “PCT/Patent Reference” column are herein incorporated by reference in their entireties.
  • the “Biological activity” column describes Biological activities associated with the Therapeutic protein molecule.
  • the “Exemplary Activity Assay” column provides references that describe assays which may be used to test the therapeutic and/or biological activity of a Therapeutic protein or an albumin fusion protein of the invention comprising a Therapeutic protein X portion.
  • Each of the references cited in the “Exemplary Activity Assay” column are herein incorporated by reference in their entireties, particularly with respect to the description of the respective activity assay described in the reference (see Methods section, for example) for assaying the corresponding biological activity set forth in the “Biological Activity” column of Table 1.
  • the “Preferred Indication Y” column describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, or ameliorated by Therapeutic protein X or an albumin fusion protein of the invention comprising a Therapeutic protein X portion.
  • Therapeutic protein in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., leukemias, cancers, and/or as described below under “Hyperproliferative Disorders”).
  • neoplastic diseases e.g., leukemias, cancers, and/or as described below under “Hyperproliferative Disorders”.
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a neoplasm located in a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.g., leukemia), endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), uterus, eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital.
  • a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a pre-neoplastic condition, selected from the group consisting of: hyperplasia (e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”), metaplasia (e.g., connective tissue metaplasia, a typical metaplasia, and/or as described in the section entitled “Hyperproliferative Disorders”), and/or dysplasia (e.g., cervical dysplasia, and bronchopulmonary dysplasia).
  • hyperplasia e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”
  • metaplasia e.
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a benign dysproliferative disorder selected from the group consisting of: benign tumors, fibrocystic conditions, tissue hypertrophy, and/or as described in the section entitled “Hyperproliferative Disorders”.
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), blood disorders (e.g., as described below under “Immune Activity” “Cardiovascular Disorders” and/or “Blood-Related Disorders”), and infections (e.g., as described below under “Infectious Disease”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • blood disorders e.g., as described below under “Immune Activity” “Cardiovascular Disorders” and/or “Blood-Related Disorders”
  • infections e.g., as described below under “Infectious Disease”.
  • a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: anemia, pancytopenia, leukopenia, thrombocytopenia, leukemias, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL), plasmacytomas, multiple myeloma, Burkitt's lymphoma, arthritis, asthma, A/DS, autoimmune disease, rheumatoid arthritis, granulomatous disease, immune deficiency, inflammatory bowel disease, sepsis, neutropenia, neutrophilia, psoriasis, immune reactions to transplanted organs and tissues, systemic lupus
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the reproductive system (e.g., as described below under “Reproductive System Disorders”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • reproductive system e.g., as described below under “Reproductive System Disorders”.
  • a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1 may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cryptorchism, prostatitis, inguinal hernia, varicocele, leydig cell tumors, verrucous carcinoma, prostatitis, malacoplakia, Peyronie's disease, penile carcinoma, squamous cell hyperplasia, dysmenorrhea, ovarian adenocarcinoma, Turner's syndrome, mucopurulent cervicitis, Sertoli-leydig tumors, ovarian cancer, uterine cancer, pelvic inflammatory disease, testicular cancer, prostate cancer, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes mellitus, cystic fibrosis
  • Therapeutic protein in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the immune system (e.g., as described below under “Immune Activity”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • Immune Activity disorders of the immune system
  • a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: bone cancers (e.g., osteochondromas, benign chondromas, chondroblastoma, chondromyxoid fibromas, osteoid osteomas, giant cell tumors, multiple myeloma, osteosarcomas), Paget's Disease, rheumatoid arthritis, systemic lupus erythematosus, osteomyelitis, Lyme Disease, gout, bursitis, tendonitis, osteoporosis, osteoarthritis, muscular dystrophy, mitochondrial myopathy, cachexia, and multiple sclerosis.
  • bone cancers e.g., osteochondromas, benign chondromas, chondroblast
  • Cardiovascular in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the cardiovascular system (e.g., as described below under “Cardiovascular Disorders”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • Cardiovascular Disorders e.g., as described below under “Cardiovascular Disorders”.
  • a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: myxomas, fibromas, rhabdomyomas, cardiovascular abnormalities (e.g., congenital heart defects, cerebral arterioyenous malformations, septal defects), heart disease (e.g., heart failure, congestive heart disease, arrhythmia, tachycardia, fibrillation, pericardial Disease, endocarditis), cardiac arrest, heart valve disease (e.g., stenosis, regurgitation, prolapse), vascular disease (e.g., hypertension, coronary artery disease, angina, aneurysm, arteriosclerosis, peripheral vascular disease), hyponatremia, hypernatremia, hypokal
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”).
  • a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: spina bifida, hydranencephaly, neurofibromatosis, fetal alcohol syndrome, diabetes mellitus, PKU, Down's syndrome, Patau syndrome, Edwards syndrome, Turner syndrome, Apert syndrome, Carpenter syndrome, Conradi syndrome, Crouzon syndrome, cutis laxa, Cornelia de Lange syndrome, Ellis-van Creveld syndrome, Holt-Oram syndrome, Kartagener syndrome, Meckel-Gruber syndrome, Noonan syndrome, Pallister-Hall syndrome, Rubinstein-Taybi syndrome, Scimitar syndrome, Smith-Lemli-Opitz syndrome, thromo
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and renal disorders (e.g., as described below under “Renal Disorders”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • renal disorders e.g., as described below under “Renal Disorders”.
  • a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: bladder cancer, prostate cancer, benign prostatic hyperplasia, bladder disorders (e.g., urinary incontinence, urinary retention, urinary obstruction, urinary tract Infections, interstitial cystitis, prostatitis, neurogenic bladder, hematuria), renal disorders (e.g., hydronephrosis, proteinuria, renal failure, pyelonephritis, urolithiasis, reflux nephropathy, and unilateral obstructive uropathy).
  • bladder cancer e.g., prostate cancer, benign prostatic hyperplasia
  • bladder disorders e.g., urinary incontinence, urinary retention, urinary
  • Neurological/Sensory in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the nervous system (e.g., as described below under “Neural Activity and Neurological Diseases”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • Neurological Diseases e.g., as described below under “Neural Activity and Neurological Diseases”.
  • a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: brain cancer (e.g., brain stem glioma, brain tumors, central nervous system (Primary) lymphoma, central nervous system lymphoma, cerebellar astrocytoma, and cerebral astrocytoma, neurodegenerative disorders (e.g., Alzheimer's Disease, Creutzfeldt-Jakob Disease, Parkinson's Disease, and Idiopathic Presenile Dementia), encephalomyelitis, cerebral malaria, meningitis, metabolic brain diseases (e.g., phenylketonuria and pyruvate carboxylase deficiency), cerebellar at a disease or disorder selected from the
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the respiratory system (e.g., as described below under “Respiratory Disorders”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • Respiratory Disorders e.g., as described below under “Respiratory Disorders”.
  • a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cancers of the respiratory system such as larynx cancer, pharynx cancer, trachea cancer, epiglottis cancer, lung cancer, squamous cell carcinomas, small cell (oat cell) carcinomas, large cell carcinomas, and adenocarcinomas.
  • cancers of the respiratory system such as larynx cancer, pharynx cancer, trachea cancer, epiglottis cancer, lung cancer, squamous cell carcinomas, small cell (oat cell) carcinomas, large cell carcinomas, and adenocarcinomas.
  • Allergic reactions cystic fibrosis, sarcoidosis, histiocytosis X, infiltrative lung diseases (e.g., pulmonary fibrosis and lymphoid interstitial pneumonia), obstructive airway diseases (e.g., asthma, emphysema, chronic or acute bronchitis), occupational lung diseases (e.g., silicosis and asbestosis), pneumonia, and pleurisy.
  • infiltrative lung diseases e.g., pulmonary fibrosis and lymphoid interstitial pneumonia
  • obstructive airway diseases e.g., asthma, emphysema, chronic or acute bronchitis
  • occupational lung diseases e.g., silicosis and asbestosis
  • pneumonia e.g., silicosis and asbestosis
  • “Endocrine” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the respiratory system (e.g., as described below under “Respiratory Disorders”), renal disorders (e.g., as described below under “Renal Disorders”), and disorders of the endocrine system (e.g., as described below under “Endocrine Disorders”.
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • diseases or disorders of the respiratory system e.g., as described below under “Respiratory Disorders”
  • renal disorders e.g., as described below under “Renal Disorders”
  • disorders of the endocrine system
  • a Therapeutic protein having a “Endocrine” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cancers of endocrine tissues and organs (e.g., cancers of the hypothalamus, pituitary gland, thyroid gland, parathyroid glands, pancreas, adrenal glands, ovaries, and testes), diabetes (e.g., diabetes insipidus, type I and type II diabetes mellitus), obesity, disorders related to pituitary glands (e.g., hyperpituitarism, hypopituitarism, and pituitary dwarfism), hypothyroidism, hyperthyroidism, goiter, reproductive disorders (e.g.
  • cancers of endocrine tissues and organs e.g., cancers
  • disorders related to adrenal glands e.g., Addison's Disease, corticosteroid deficiency, and Cushing's Syndrome
  • kidney cancer e.g., hypemephroma, transitional cell cancer, and Wilm's tumor
  • diabetic nephropathy e.g., interstitial nephritis
  • polycystic kidney disease e.g., glomenilonephritis (e.g., IgM mesangial proliferative glomerulonephritis and glomerulonephritis caused by autoimmune disorders; such as Goodpasture's syndrome), and nephrocalcinosis.
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the gastrointestinal system (e.g., as described below under “Gastrointestinal Disorders”.
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • Gastrointestinal Disorders e.g., as described below under “Gastrointestinal Disorders”.
  • a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: ulcerative colitis, appendicitis, Crohn's disease, hepatitis, hepatic encephalopathy, portal hypertension, cholelithiasis, cancer of the digestive system (e.g., biliary tract cancer, stomach cancer, colon cancer, gastric cancer, pancreatic cancer, cancer of the bile duct, tumors of the colon (e.g., polyps or cancers), and cirrhosis), pancreatitis, ulcerative disease, pyloric stenosis, gastroenteritis, gastritis, gastric atropy, benign tumors of the duodenum,
  • a disease or disorder selected
  • Therapeutic protein fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), cellular and genetic abnormalities (e.g., as described below under “Diseases at the Cellular Level”), angiogenesis (e.g., as described below under “Anti-Angiogenesis Activity”), and or to promote or inhibit regeneration (e.g., as described below under “Regeneration”), and wound healing (e.g., as described below under “Wound Healing and Epithelial Cell Proliferation”).
  • neoplastic diseases e.g., as described below under “Hyperproliferative Disorders”
  • cellular and genetic abnormalities e.g., as described below under “Diseases at the Cellular Level”
  • angiogenesis e.g., as described below under “Anti-Angiogenesis
  • a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: connective tissue metaplasia, mixed connective tissue disease, focal epithelial hyperplasia, epithelial metaplasia, mucoepithelial dysplasia, graft v.
  • the albumin fusion proteins of the invention are capable of a therapeutic activity and/or biologic activity corresponding to the therapeutic activity and/or biologic activity of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein listed in the corresponding row of Table 1.
  • the therapeutically active protein portions of the albumin fusion proteins of the invention are fragments or variants of the reference sequence cited in the “Exemplary Identifier” column of Table 1, and are capable of the therapeutic activity and/or biologic activity of the corresponding Therapeutic protein.
  • the present invention is further directed to fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention.
  • fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., a Therapeutic protein as disclosed in Table 1).
  • N-terminal deletions may be described by the general formula m ⁇ q, where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1), and m is defined as any integer ranging from 2 to q ⁇ 6.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • fragments of serum albumin polypeptides corresponding to an albumin protein portion of an albumin fusion protein of the invention include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., serum albumin).
  • N-terminal deletions may be described by the general formula m ⁇ 585 , where 585 is a whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO:18), and m is defined as any integer ranging from 2 to 579.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • fragments of albumin fusion proteins of the invention include the full length albumin fusion protein as well as polypeptides having one or more residues deleted from the amino terminus of the albumin fusion protein.
  • N-terminal deletions may be described by the general formula m ⁇ q, where q is a whole integer representing the total number of amino acid residues in the albumin fusion protein, and m is defined as any integer ranging from 2 to q ⁇ 6. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • a reference polypeptide e.g., a Therapeutic protein and/or serum albumin protein
  • other functional activities e.g., biological activities, ability to multimerize, ability to bind a ligand
  • Therapeutic activities may still be retained.
  • the ability of polypeptides with C-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptide generally will be retained when less than the majority of the residues of the complete or mature polypeptide are removed from the C-terminus.
  • Whether a particular polypeptide lacking the N-terminal and/or C-terminal residues of a reference polypeptide retains Therapeutic activity can readily be determined by routine methods described herein and/or otherwise known in the art.
  • the present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., a Therapeutic protein referred to in Table 1).
  • C-terminal deletions may be described by the general formula 1 ⁇ n, where n is any whole integer ranging from 6 to q ⁇ 1, and where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1).
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., serum albumin).
  • C-terminal deletions may be described by the general formula 1 ⁇ n, where n is any whole integer ranging from 6 to 584, where 584 is the whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO: 18) minus 1.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of an albumin fusion protein of the invention.
  • C-terminal deletions may be described by the general formula 1 ⁇ n, where n is any whole integer ranging from 6 to q ⁇ 1, and where q is a whole integer representing the total number of amino acid residues in an albumin fusion protein of the invention.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • any of the above described N- or C-terminal deletions can be combined to produce a N- and C-terminal deleted reference polypeptide.
  • the invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues m-n of a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or serum albumin (e.g., SEQ ID NO: 18), or an albumin fusion protein of the invention) where n and m are integers as described above.
  • a reference polypeptide e.g., a Therapeutic protein referred to in Table 1, or serum albumin (e.g., SEQ ID NO: 18), or an albumin fusion protein of the invention
  • n and m are integers as described above.
  • Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • the present application is also directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a reference polypeptide sequence (e.g., a Therapeutic protein, serum albumin protein or an albumin fusion protein of the invention) set forth herein, or fragments thereof.
  • a reference polypeptide sequence e.g., a Therapeutic protein, serum albumin protein or an albumin fusion protein of the invention
  • the application is directed to proteins comprising polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to reference polypeptides having the amino acid sequence of N- and C-terminal deletions as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • Preferred polypeptide fragments of the invention are fragments comprising, or alternatively, consisting of, an amino acid sequence that displays a Therapeutic activity and/or functional activity (e.g. biological activity) of the polypeptide sequence of the Therapeutic protein or serum albumin protein of which the amino acid sequence is a fragment.
  • Other preferred polypeptide fragments are biologically active fragments.
  • Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention.
  • the biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity.
  • Variant refers to a polynucleotide or nucleic acid differing from a reference nucleic acid or polypeptide, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the reference nucleic acid or polypeptide.
  • variant refers to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin portion of an albumin fusion protein of the invention, or albumin fusion protein differing in sequence from a Therapeutic protein (e.g. see “therapeutic” column of Table 1), albumin protein, and/or albumin fusion protein of the invention, respectively, but retaining at least one functional and/or therapeutic property thereof as described elsewhere herein or otherwise known in the art.
  • variants are overall very similar, and, in many regions, identical to the amino acid sequence of the Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion protein of the invention. Nucleic acids encoding these variants are also encompassed by the invention.
  • the present invention is also directed to proteins which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., an amino acid sequence disclosed in the “Exemplary Identifier” column of Table 1, or fragments or variants thereof), albumin proteins (e.g., SEQ ID NO:18 or fragments or variants thereof) corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion proteins of the invention.
  • an amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention e.g., an amino acid sequence disclosed in the “Exemplary Identifier” column of Table 1, or fragments or variants thereof
  • albumin proteins e.g., SEQ ID NO:18 or fragment
  • polypeptides encompassed by the invention are polypeptides encoded by polynucleotides which hybridize to the complement of a nucleic acid molecule encoding an amino acid sequence of the invention under stringent hybridization conditions (e.g., hybridization to filter bound DNA in 6 ⁇ Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.2 ⁇ SSC, 0.1% SDS at about 50-65 degrees Celsius), under highly stringent conditions (e.g., hybridization to filter bound DNA in 6 ⁇ sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.1 ⁇ SSC, 0.2% SDS at about 68 degrees Celsius), or under other stringent hybridization conditions which are known to those of skill in the art (see, for example, Ausubel, F.
  • stringent hybridization conditions e.g., hybridization to filter bound DNA in 6 ⁇ Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more
  • a polypeptide having an amino acid sequence at least, for example, 95% “identical” to a query amino acid sequence of the present invention it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence.
  • up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, or substituted with another amino acid.
  • These alterations of the reference sequence may occur at the amino- or carboxy-terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence.
  • any particular polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequence of an albumin fusion protein of the invention or a fragment thereof (such as the Therapeutic protein portion of the albumin fusion protein or the albumin portion of the albumin fusion protein), can be determined conventionally using known computer programs.
  • a preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990)).
  • the query and subject sequences are either both nucleotide sequences or both amino acid sequences.
  • the result of said global sequence alignment is expressed as percent identity.
  • the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment.
  • This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score.
  • This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence.
  • a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity.
  • the deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus.
  • the 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C-termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%.
  • a 90 residue subject sequence is compared with a 100 residue query sequence.
  • deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query.
  • percent identity calculated by FASTDB is not manually corrected.
  • residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention.
  • the variant will usually have at least 75% (preferably at least about 80%, 90%, 95% or 99%) sequence identity with a length of normal HA or Therapeutic protein which is the same length as the variant.
  • Homology or identity at the nucleotide or amino acid sequence level is determined by BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al., Proc. Natl. Acad. Sci. USA 87: 2264-2268 (1990) and Altschul, J. Mol. Evol. 36: 290-300 (1993), fully incorporated by reference) which are tailored for sequence similarity searching.
  • the approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified and finally to summarize only those matches which satisfy a preselected threshold of significance.
  • a threshold of significance For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al., (Nature Genetics 6: 119-129 (1994)) which is fully incorporated by reference.
  • the search parameters for histogram, descriptions, alignments, expect i.e., the statistical significance threshold for reporting matches against database sequences
  • cutoff matrix and filter
  • the default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al., Proc. Natl. Acad. Sci. USA 89: 10915-10919 (1992), fully incorporated by reference).
  • the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively.
  • the polynucleotide variants of the invention may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Moreover, polypeptide variants in which less than 50, less than 40, less than 30, less than 20, less than 10, or 5-50, 5-25, 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred.
  • Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host, such as, yeast or E. coli ).
  • a polynucleotide encoding an albumin portion of an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
  • a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
  • a polynucleotide encoding an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells.
  • a codon optimized polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein of the invention does not hybridize to the wild type polynucleotide encoding the Therapeutic protein under stringent hybridization conditions as described herein.
  • a codon optimized polynucleotide encoding an albumin portion of an albumin fusion protein of the invention do not hybridize to the wild type polynucleotide encoding the albumin protein under stringent hybridization conditions as described herein.
  • a codon optimized polynucleotide encoding an albumin fusion protein of the invention do not hybridize to the wild type polynucleotide encoding the Therapeutic protein portin or the albumin protein portion under stringent hybridization conditions as described herein.
  • polynucleotides encoding a Therapeutic protein portion of an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of that Therapeutic protein.
  • polynucleotides encoding an albumin protein portion of an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of albumin protein.
  • polynucleotides encoding an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of a Therapeutic protein portion or the albumin protein portion.
  • Naturally occurring variants are called “allelic variants,” and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis.
  • variants may be generated to improve or alter the characteristics of the polypeptides of the present invention.
  • one or more amino acids can be deleted from the N-terminus or C-terminus of the polypeptide of the present invention without substantial loss of biological function.
  • Ron et al. J. Biol. Chem. 268: 2984-2988 (1993)
  • variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues.
  • Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216 (1988).)
  • the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity).
  • the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein.
  • Such variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity.
  • the variants of the invention have conservative substitutions.
  • conservative substitutions is intended swaps within groups such as replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gln, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly.
  • the first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein.
  • the second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function. For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used. See Cunningham and Wells, Science 244:1081-1085 (1989). The resulting mutant molecules can then be tested for biological activity.
  • tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gln, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly.
  • variants of the present invention include (i) polypeptides containing substitutions of one or more of the non-conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) polypeptides containing substitutions of one or more of the amino acid residues having a substituent group, or (iii) polypeptides which have been fused with or chemically conjugated to another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), (iv) polypeptide containing additional amino acids, such as, for example, an IgG Fc fusion region peptide.
  • polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein.
  • polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993).
  • the polypeptides of the invention comprise, or alternatively, consist of, fragments or variants of the amino acid sequence of a Therapeutic protein described herein and/or human serum albumin, and/or albumin fusion protein of the invention, wherein the fragments or variants have 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, amino acid residue additions, substitutions, and/or deletions when compared to the reference amino acid sequence.
  • the amino acid substitutions are conservative. Nucleic acids encoding these polypeptides are also encompassed by the invention.
  • the polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids.
  • the polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini.
  • polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • a polypeptide having functional activity refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a Therapeutic protein.
  • Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide.
  • a polypeptide having biological activity refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a Therapeutic protein of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention).
  • an albumin fusion protein of the invention has at least one biological and/or therapeutic activity associated with the Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin.
  • the albumin fusion proteins of the invention can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Additionally, one of skill in the art may routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, for activity using assays referenced in its corresponding row of Table 1. Further, one of skill in the art may routinely assay fragments of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention, for activity using assays known in the art and/or as described in the Examples section below.
  • functional activity e.g., biological activity
  • routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention for activity using assays referenced in its corresponding row of Table 1.
  • one of skill in the art may routinely assay fragments of an albumin protein corresponding to an albumin protein portion of
  • an albumin fusion protein of the invention in one embodiment where one is assaying for the ability of an albumin fusion protein of the invention to bind or compete with a Therapeutic protein for binding to an anti-Therapeutic polypeptide antibody and/or anti-albumin antibody, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophores
  • antibody binding is detected by detecting a label on the primary antibody.
  • the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody.
  • the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention.
  • binding partner e.g., a receptor or a ligand
  • binding to that binding partner by an albumin fusion protein containing that Therapeutic protein as the Therapeutic protein portion of the fusion can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995).
  • the ability of physiological correlates of an albumin fusion protein of the present invention to bind to a substrate(s) of the Therapeutic polypeptide corresponding to the Therapeutic portion of the albumin fusion protein of the invention can be routinely assayed using techniques known in the art.
  • association with other components of the multimer can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., supra.
  • an albumin fusion protein of the invention comprising all or a portion of an antibody that binds a Therapeutic protein, has at least one biological and/or therapeutic activity (e.g., to specifically bind a polypeptide or epitope) associated with the antibody that binds a Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin.
  • the biological activity and/or therapeutic activity of an albumin fusion protein of the invention comprising all or a portion of an antibody that binds a Therapeutic protein is the inhibition (i.e. antagonism) or activation (i.e., agonism) of one or more of the biological activities and/or therapeutic activities associated with the polypeptide that is specifically bound by antibody that binds a Therapeutic protein.
  • Albumin fusion proteins of the invention may be characterized in a variety of ways.
  • albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
  • albumin fusion proteins of the invention e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein
  • Assays for the ability of the albumin fusion proteins of the invention to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Pat. No. 5,223,409), on spores (e.g., Patent Nos.
  • Albumin fusion proteins of the invention comprising at least a fragment or variant of a Therapeutic antibody may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.
  • the albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
  • antigens e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, aggluti
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the albumin fusion protein of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C., adding sepharose beads coupled to an anti-albumin antibody, for example, to the cell lysate, incubating for about an hour or more at 40 degrees C., washing the beads in lysis buffer and resus
  • albumin fusion protein of the invention to immunoprecipitate a particular antigen can be assessed by, e.g. western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10. 16.1.
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), applying the albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32 P or 125 I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the albumin fusion protein (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the albumin fusion proteins specifically bound to the antigen coating the well.
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase
  • the albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes albumin fusion protein) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the albumin fusion protein may be coated to the well.
  • the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase).
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase.
  • the binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays.
  • a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3 H or 125 I) with the albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen.
  • labeled antigen e.g., 3 H or 125 I
  • the affinity of the albumin fusion protein of the present invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis.
  • Competition with a second protein that binds the same protein, antigen or epitope as the albumin fusion protein can also be determined using radioimmunoassays.
  • the protein, antigen or epitope is incubated with an albumin fusion protein of the present invention conjugated to a labeled compound (e.g., 3 H or 125 I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • a labeled compound e.g., 3 H or 125 I
  • BIAcore kinetic analysis is used to determine the binding on and off rates of albumin fusion proteins of the invention to a protein, antigen or epitope.
  • BIAcore kinetic analysis comprises analyzing the binding and dissociation of albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes or albumin fusion proteins, respectively, on their surface.
  • Antibodies that bind a Therapeutic protein corresponding to the Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity for a given protein or antigen, preferably the antigen which they specifically bind.
  • Preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 2 M, 10 ⁇ 2 M, 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇ 4 M, 10 ⁇ 4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 5 M, 10 ⁇ 5 M, 5 ⁇ 10 ⁇ 6 M, 10 ⁇ 6 M, 5 ⁇ 10 ⁇ 7 M, 10 7 M, 5 ⁇ 10 ⁇ 8 M or 10 ⁇ 8 M.
  • Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M, 5 ⁇ 10 ⁇ 13 M, 10 ⁇ 13 M, 5 ⁇ 10 ⁇ 14 M, 10 ⁇ 14 M, 5 ⁇ 10 ⁇ 15 M, or 10 ⁇ 15 M.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
  • albumin fusion proteins of the present invention may routinely be applied to measure the ability of albumin fusion proteins of the present invention and fragments, variants and derivatives thereof to elicit biological activity and/or Therapeutic activity (either in vitro or in vivo) related to either the Therapeutic protein portion and/or albumin portion of the albumin fusion protein of the present invention.
  • Therapeutic activity either in vitro or in vivo
  • Other methods will be known to the skilled artisan and are within the scope of the invention.
  • an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion or chemical conjugation.
  • HSA human serum albumin
  • HA human albumin
  • albumin and HA are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof).
  • albumin refers collectively to albumin protein or amino acid sequence, or an albumin fragment or variant, having one or more functional activities (e.g., biological activities) of albumin.
  • albumin refers to human albumin or fragments thereof (see EP 201 239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin as shown in FIG. 15 and SEQ ID NO: 18, or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof.
  • the human serum albumin protein used in the albumin fusion proteins of the invention contains one or both of the following sets of point mutations with reference to SEQ ID NO:18: Leu-407 to Ala, Leu-408 to Val, Val-409 to Ala, and Arg-410 to Ala; or Arg-410 to A, Lys-413 to Gln, and Lys-414 to Gln (see, e.g., International Publication No. WO95/23857, hereby incorporated in its entirety by reference herein).
  • albumin fusion proteins of the invention that contain one or both of above-described sets of point mutations have improved stability/resistance to yeast Yap3p proteolytic cleavage, allowing increased production of recombinant albumin fusion proteins expressed in yeast host cells.
  • a portion of albumin sufficient to prolong the therapeutic activity or shelf-life of the Therapeutic protein refers to a portion of albumin sufficient in length or structure to stabilize or prolong the therapeutic activity of the protein so that the shelf life of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life in the non-fusion state.
  • the albumin portion of the albumin fusion proteins may comprise the full length of the HA sequence as described above or as shown in FIG. 15, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity.
  • Such fragments may be of 10 or more amino acids in length or may include about 15, 20, 25, 30, 50, or more contiguous amino acids from the HA sequence or may include part or all of specific domains of HA. For instance, one or more fragments of HA spanning the first two immunoglobulin-like domains may be used.
  • the albumin portion of the albumin fusion proteins of the invention may be a variant of normal HA.
  • the Therapeutic protein portion of the albumin fusion proteins of the invention may also be variants of the Therapeutic proteins as described herein.
  • variants includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins.
  • the albumin fusion proteins of the invention may include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419).
  • the albumin may be derived from any vertebrate, especially any mammal, for example human, cow, sheep, or pig.
  • Non-mammalian albumins include, but are not limited to, hen and salmon.
  • the albumin portion of the albumin fusion protein may be from a different animal than the Therapeutic protein portion.
  • an HA fragment or variant will be at least 100 amino acids long, preferably at least 150 amino acids long.
  • the HA variant may consist of or alternatively comprise at least one whole domain of HA, for example domains 1 (amino acids 1-194 of SEQ ID NO:18), 2 (amino acids 195-387 of SEQ ID NO:18), 3 (amino acids 388-585 of SEQ ID NO:18), 1+2 (1-387 of SEQ ID NO:18), 2+3 (195-585 of SEQ ID NO:18) or 1+3 (amino acids 1-194 of SEQ ID NO:18+amino acids 388-585 of SEQ ID NO:18).
  • Each domain is itself made up of two homologous subdomains namely 1-105, 120-194, 195-291, 316-387, 388-491 and 512-585, with flexible inter-subdomain linker regions comprising residues Lys106 to Glu119, Glu292 to Val315 and Glu492 to Ala511.
  • the albumin portion of an albumin fusion protein of the invention comprises at least one subdomain or domain of HA or conservative modifications thereof. If the fusion is based on subdomains, some or all of the adjacent linker is preferably used to link to the Therapeutic protein moiety.
  • Antibodies that Specifically Bind Therapeutic Proteins are also Therapeutic Proteins
  • the present invention also encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that specifically binds a Therapeutic protein disclosed in Table 1. It is specifically contemplated that the term “Therapeutic protein” encompasses antibodies that bind a Therapeutic protein (e.g., as Described in column I of Table 1) and fragments and variants thereof. Thus an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an an antibody that binds a Therapeutic protein.
  • the basic antibody structural unit is known to comprise a tetramer.
  • Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa).
  • the amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition.
  • the carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as kappa and lambda light chains.
  • Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, 1gG, IgA, and IgE, respectively. See generally, Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N.Y. (1998)) (incorporated by reference in its entirety for all purposes).
  • the variable regions of each light/heavy chain pair form the antibody binding site.
  • an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same.
  • the chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs.
  • the CDR regions in general, are the portions of the antibody which make contact with the antigen and determine its specificity.
  • the CDRs from the heavy and the light chains of each pair are aligned by the framework regions, enabling binding to a specific epitope.
  • both light and heavy chains variable regions comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4.
  • the variable regions are connected to the heavy or light chain constant region.
  • antibody refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen (e.g., a molecule containing one or more CDR regions of an antibody).
  • Antibodies that may correspond to a Therapeutic protein portion of an albumin fusion protein include, but are not limited to, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies (e.g., single chain Fvs), Fab fragments, F(ab′) fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies specific to antibodies of the invention), and epitope-binding fragments of any of the above (e.g., VH domains, VL domains, or one or more CDR regions).
  • single chain antibodies e.g., single chain Fvs
  • Fab fragments fragments
  • F(ab′) fragments fragments produced by a Fab expression library
  • anti-idiotypic antibodies including, e.g., anti-Id antibodies specific to antibodies of the invention
  • epitope-binding fragments of any of the above e.g., VH domains, V
  • the present invention encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that binds a Therapeutic Protein (e.g., as disclosed in Table 1) or fragment or variant thereof.
  • Antibodies that bind a Therapeutic protein may be from any animal origin, including birds and mammals.
  • the antibodies are human, murine (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken antibodies.
  • the antibodies are human antibodies.
  • “human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries and xenomice or other organisms that have been genetically engineered to produce human antibodies.
  • the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule.
  • the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG1.
  • the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG2. In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG4.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab′ and F(ab′) 2 , Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain.
  • Antigen-binding antibody fragments, including single-chain antibodies may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be monospecific, bispecific, trispecific or of greater multispecificity.
  • Multispecific antibodies may be specific for different epitopes of a Therapeutic protein or may be specific for both a Therapeutic protein as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Pat. Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992).
  • Antibodies that bind a Therapeutic protein may be bispecific or bifunctional which means that the antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab′ fragments. See, e.g., Songsivilai & Lachmann Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al. J. Immunol. 148:1547 1553 (1992).
  • bispecific antibodies may be formed as “diabodies” (Holliger et al.
  • the present invention also provides albumin fusion proteins that comprise, fragments or variants (including derivatives) of an antibody described herein or known elsewhere in the art. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding a molecule of the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions.
  • the variants encode less than 50 amino acid substitutions, less than 40 amino acid subsitutions, less than 30 amino acid substitutions, less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the reference VH domain, VHCDR1, VHCDR2, VHCDR3, VL domain, VLCDR1, VLCDR2, or VLCDR3.
  • the variants encode substitutions of VHCDR3.
  • the variants have conservative amino acid substitutions at one or more predicted non-essential amino acid residues.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be described or specified in terms of the epitope(s) or portion(s) of a Therapeutic protein which they recognize or specifically bind.
  • Antibodies which specifically bind a Therapeutic protein or a specific epitope of a Therapeutic protein may also be excluded. Therefore, the present invention encompasses antibodies that specifically bind Therapeutic proteins, and allows for the exclusion of the same.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, binds the same epitopes as the.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a Therapeutic protein are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention.
  • antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention cross-react with murine, rat and/or rabbit homologs of human proteins and the corresponding epitopes thereof.
  • Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention.
  • the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially identical cross reactivity characteristics compared to the.
  • antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding a Therapeutic protein under stringent hybridization conditions are also included in the present invention.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention.
  • Preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 2 M, 10 ⁇ 2 M, 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇ 4 M, 10 ⁇ 4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 5 M, 10 ⁇ 5 M, 5 ⁇ 10 ⁇ 6 M, 10 ⁇ 6 M, 5 ⁇ 10 ⁇ 7 M, 10 7 M, 5 ⁇ 10 ⁇ 8 M or 10 ⁇ 8 M.
  • Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M, 5 ⁇ 10 ⁇ 13 M, 10 ⁇ 13 M, 5 ⁇ 10 ⁇ 14 M, 10 ⁇ 14 M, 5 ⁇ 10 ⁇ 15 M, or 10 ⁇ 15 M.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
  • the invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of a Therapeutic protein as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein.
  • the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of an antibody to an epitope of a Therapeutic protein as well as the competitively inhibits binding of an antibody to an epitope of a Therapeutic protein.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of the to an epitope of a Therapeutic protein by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may act as agonists or antagonists of the Therapeutic protein.
  • the present invention includes antibodies which disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully.
  • the invention features both receptor-specific antibodies and ligand-specific antibodies.
  • the invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation. Receptor activation (i.e., signaling) may be determined by techniques described herein or otherwise known in the art.
  • receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by immunoprecipitation followed by western blot analysis (for example, as described supra).
  • antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially similar characteristics with regard to preventing ligand binding and/or preventing receptor activation compared to the.
  • the invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand.
  • receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand.
  • neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor.
  • antibodies which activate the receptor are also act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor.
  • the antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the Therapeutic protreins (e.g. as disclosed in Table 1).
  • the above antibody agonists can be made using methods known in the art. See, e.g., PCT publication WO 96/40281; U.S. Pat. No. 5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res. 58(16):3668-3678 (1998); Harrop et al., J. Immunol. 161(4):1786-1794 (1998); Zhu et al., Cancer Res.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, have similar or substantially identical agonist or antagonist properties as the.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be used, for example, to purify, detect, and target Therapeutic proteins, including both in in vitro and in vivo diagnostic and therapeutic methods.
  • the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the Therapeutic protein in biological samples. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); incorporated by reference herein in its entirety.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, may be used, for example, to purify, detect, and target Therapeutic proteins, including both in in vitro and in vivo diagnostic and therapeutic methods.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein include derivatives that are modified, i.e, by the covalent attachment of any type of molecule to the antibody.
  • the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc.
  • any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. Albumin fusion proteins of the invention may also be modified as described above.
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be generated by any suitable method known in the art.
  • Polyclonal antibodies to an antigen-of-interest can be produced by various procedures well known in the art.
  • a Therapeutic protein may be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen.
  • adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art.
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof.
  • monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N.Y., 1981) (said references incorporated by reference in their entireties).
  • the term “monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology.
  • the term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced.
  • mice can be immunized with a Therapeutic protein or fragment or variant thereof or a cell expressing such a Therapeutic protein or fragment or variant thereof.
  • an immune response e.g., antibodies specific for the antigen are detected in the mouse serum
  • the mouse spleen is harvested and splenocytes isolated.
  • the splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC.
  • Hybridomas are selected and cloned by limited dilution.
  • hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention.
  • Ascites fluid which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones.
  • the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention.
  • EBV Epstein Barr Virus
  • Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Current Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY, which is hereby incorporated in its entirety by reference.
  • the source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues.
  • Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B cell-containing samples, because T cells from individuals seropositive for anti-EBV antibodies can suppress B cell immortalization by EBV.
  • EBV lines are generally polyclonal. However, over prolonged periods of cell cultures, EBV lines may become monoclonal or polyclonal as a result of the selective outgrowth of particular B cell clones.
  • polyclonal EBV transformed lines may be subcloned (e.g., by limiting dilution culture) or fused with a suitable fusion partner and plated at limiting dilution to obtain monoclonal B cell lines.
  • suitable fusion partners for EBV transformed cell lines include mouse myeloma cell lines (e.g., SP2/0, X63-Ag8.653), heteromyeloma cell lines (human ⁇ mouse; e.g, SPAM-8, SBC-H 2 O, and CB-F7), and human cell lines (e.g., GM 1500, SKO-007, RPMI 8226, and KR-4).
  • the present invention also provides a method of generating polyclonal or monoclonal human antibodies against polypeptides of the invention or fragments thereof, comprising EBV-transformation of human B cells.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques.
  • Fab and F(ab′) 2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′) 2 fragments).
  • F(ab′)2 fragments contain the variable region, the light chain constant region and the CH1 domain of the heavy chain.
  • antibodies that bind to a Therapeutic protein can also be generated using various phage display methods known in the art.
  • phage display methods functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them.
  • phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine).
  • Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead.
  • Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein.
  • phage display methods that can be used to make antibodies that bind to a Therapeutic protein include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol.
  • the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below.
  • a chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region.
  • Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entirety.
  • Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and a framework regions from a human immunoglobulin molecule.
  • CDRs complementarity determining regions
  • framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding.
  • These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No.
  • Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Pat. No. 5,565,332).
  • Human antibodies are particularly desirable for therapeutic treatment of human patients.
  • Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Pat. Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety.
  • Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes.
  • the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells.
  • the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes.
  • the mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production.
  • the modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice.
  • the chimeric mice are then bred to produce homozygous offspring which express human antibodies.
  • the transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention.
  • Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology.
  • the human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation.
  • Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as “guided selection.”
  • a selected non-human monoclonal antibody e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al., Bio/technology 12:899-903 (1988)).
  • the invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody and fragments thereof.
  • the invention also encompasses polynucleotides that hybridize under stringent or alternatively, under lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a Therapeutic protein, and more preferably, an antibody that binds to a polypeptide having the amino acid sequence of a “therapeutic protein X as discosed in the “Exemplay Identifier” column of Table 1.
  • the polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art.
  • a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR.
  • a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3′ and 5′ ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then
  • nucleotide sequence and corresponding amino acid sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y.
  • the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability.
  • CDRs complementarity determining regions
  • one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra.
  • the framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. Mol. Biol.
  • the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention.
  • one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds.
  • Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art.
  • a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies.
  • Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide.
  • Techniques for the assembly of functional Fv fragments in E. coli may also be used (Skerra et al., Science 242:1038-1041 (1988)).
  • an antibody, or fragment, derivative or analog thereof e.g., a heavy or light chain of an antibody or a single chain antibody
  • an expression vector containing a polynucleotide that encodes the antibody Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein.
  • the invention provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter.
  • Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Pat. No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain.
  • the expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody.
  • the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody, operably linked to a heterologous promoter.
  • vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below.
  • host-expression vector systems may be utilized to express the antibody molecules of the invention.
  • Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ.
  • These include but are not limited to microorganisms such as bacteria (e.g., E. coli, B.
  • subtilis transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CAMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mamm
  • bacterial cells such as Escherichia coli
  • eukaryotic cells especially for the expression of whole recombinant antibody molecule
  • mammalian cells such as Chinese hamster ovary cells (CHO)
  • CHO Chinese hamster ovary cells
  • a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed.
  • vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable.
  • Such vectors include, but are not limited, to the E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res.
  • pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST).
  • GST glutathione S-transferase
  • fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione.
  • the pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes.
  • the virus grows in Spodoptera frugiperda cells.
  • the antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • a number of viral-based expression systems may be utilized.
  • the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence.
  • This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts. (e.g., see Logan & Shenk, Proc.
  • Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol. 153:51-544 (1987)).
  • a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein.
  • Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed.
  • eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used.
  • Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst.
  • cell lines which stably express the antibody molecule may be engineered.
  • host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker.
  • appropriate expression control elements e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.
  • engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media.
  • the selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines.
  • This method may advantageously be used to engineer cell lines which express the antibody molecule.
  • Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule.
  • a number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt-cells, respectively.
  • antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci.
  • the expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • vector amplification for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)).
  • a marker in the vector system expressing antibody is amplifiable
  • increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., Mol. Cell. Biol. 3:257 (1983)).
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NS0) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657 which are incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al, Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are incorporated in their entirities by reference herein.
  • the host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide.
  • the two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides.
  • a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)).
  • the coding sequences for the heavy and light chains may comprise cDNA or genomic DNA.
  • an antibody molecule of the invention may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins.
  • chromatography e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • centrifugation e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography
  • differential solubility e.g., differential solubility
  • the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification.
  • Antibodies that bind a Therapeutic protein or fragments or variants can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the “HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the “flag” tag.
  • the present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent.
  • the antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions.
  • the detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Pat. No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase;
  • suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin;
  • suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin;
  • an example of a luminescent material includes luminol;
  • examples of bioluminescent materials include luciferase, luciferin, and aequorin;
  • suitable radioactive material include 125I, 131I, 111In or 99Tc. Other examples of detectable substances have been described elsewwhere herein.
  • an antibody of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.
  • the conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al., Int.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an
  • VEGI See, International Publication No. WO 99/23105
  • a thrombotic agent or an anti-angiogenic agent e.g., angiostatin or endostatin
  • biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL- interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Pat. No. 4,676,980, which is incorporated herein by reference in its entirety.
  • An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, antibodies that bind a Therapeutic protein disclosed in the “Therapeutic Protein X” column of Table 1, or a fragment or variant thereof.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH domain.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VH CDRs.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of the VH CDR1.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR3.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL domain.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VL CDRs.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR1.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR3.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two, three, four, five, or six VH and/or VL CDRs.
  • the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, an scFv comprising the VH domain of the Therapeutic antibody, linked to the VL domain of the therapeutic antibody by a peptide linker such as (Gly 4 Ser) 3 (SEQ ID NO:36).
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be utilized for immunophenotyping of cell lines and biological samples.
  • Therapeutic proteins of the present invention may be useful as cell-specific markers, or more specifically as cellular markers that are differentially expressed at various stages of differentiation and/or maturation of particular cell types.
  • Monoclonal antibodies (or albumin fusion proteins comprsing at least a fragment or variant of an antibody that binds a Therapeutic protein) directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker.
  • Various techniques can be utilized using monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, “panning” with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S. Pat. No. 5,985,660; and Morrison et al., Cell, 96:737-49 (1999)).
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be characterized in a variety of ways.
  • Albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the antibody that binds a Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art.
  • Assays for the ability of the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Pat. No. 5,223,409), on spores (e.g., Patent Nos.
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art.
  • the albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art.
  • antigens e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few.
  • competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, aggluti
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding an antibody of the invention or albumin fusion protein of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C., adding protein A and/or protein G sepharose beads (or beads coated with an appropriate anti-iditoypic antibody or anti-albumin antibody in the case when an albumin fusion protein comprising at
  • the ability of the antibody or albumin fusion protein of the invention to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis.
  • One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody or albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads).
  • immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1.
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), applying the antibody or albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., 32 P or 125 I) diluted in blocking buffer, washing the membrane in wash buffer, and
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the antibody or albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the antibody or albumin fusion proteins specifically bound to the antigen coating the well.
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase
  • the antibody or albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody or albumin fusion protein, respectively) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, antibody or the albumin fusion protein may be coated to the well.
  • the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase).
  • an enzymatic substrate e.g., horseradish peroxidase or alkaline phosphatase.
  • the binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an antibody- or albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays.
  • a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., 3 H or 125 I) with the antibody or albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen.
  • the affinity of the antibody or albumin fusion protein of the present invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the antibody or albumin fusion protein, can also be determined using radioimmunoassays.
  • the protein, antigen or epitope is incubated with an antibody or albumin fusion protein of the present invention conjugated to a labeled compound (e.g. 3 H or 125 I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • a labeled compound e.g. 3 H or 125 I
  • BIAcore kinetic analysis is used to determine the binding on and off rates of antibody or albumin fusion proteins of the invention to a protein, antigen or epitope.
  • BIAcore kinetic analysis comprises analyzing the binding and dissociation of antibodies, albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes, antibodies or albumin fusion proteins, respectively, on their surface.
  • the present invention is further directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein), nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein), albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, and nucleic acids encoding such albumin fusion proteins.
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • the present invention is directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions., and/or as described elsewhere herein.
  • Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell; antibodies directed to an epitope of a Therapeutic protein and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein).
  • the antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein.
  • the treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions.
  • Antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein.
  • a summary of the ways in which the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be used therapeutically includes binding Therapeutic proteins locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below.
  • CDC complement
  • ADCC effector cells
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies.
  • lymphokines or hematopoietic growth factors such as, e.g., IL-2, IL-3 and IL-7
  • the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in a preferred embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis.
  • binding affinities include dissociation constants or Kd's less than 5 ⁇ 10 ⁇ 2 M, 10 ⁇ 2 M, 5 ⁇ 10 ⁇ 3 M, 10 ⁇ 3 M, 5 ⁇ 10 ⁇ 4 M, 10 ⁇ 4 M.
  • More preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 5 M, 10 ⁇ 5 M, 5 ⁇ 10 ⁇ 6 M, 10 ⁇ 6 M, 5 ⁇ 10 ⁇ 7 M, 10 7 M, 5 ⁇ 10 ⁇ 8 M or 10 ⁇ 8 M.
  • Even more preferred binding affinities include those with a dissociation constant or Kd less than 5 ⁇ 10 ⁇ 9 M, 10 ⁇ 9 M, 5 ⁇ 10 ⁇ 10 M, 10 ⁇ 10 M, 5 ⁇ 10 ⁇ 11 M, 10 ⁇ 11 M, 5 ⁇ 10 ⁇ 12 M, 10 ⁇ 12 M, 5 ⁇ 10 ⁇ 13 M, 10 ⁇ 13 M, 5 ⁇ 10 ⁇ 14 M, 10 ⁇ 14 M, 5 ⁇ 10 ⁇ 15 M, or 10 ⁇ 15 M.
  • nucleic acids comprising sequences encoding antibodies that bind therapeutic proteins or albumin fusion proteins comprising at least a fragment or varaint of an antibody that binds a Therapeutic protein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein, by way of gene therapy.
  • Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid.
  • the nucleic acids produce their encoded protein that mediates a therapeutic effect.
  • the compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans.
  • in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample.
  • the effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays.
  • in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed.
  • the invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention, preferably an antibody.
  • the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects).
  • the subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human.
  • Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above; additional appropriate formulations and routes of administration can be selected from among those described herein below.
  • Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc.
  • Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes.
  • the compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local.
  • Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent.
  • a protein, including an antibody, of the invention care must be taken to use materials to which the protein does not absorb.
  • the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.)
  • the compound or composition can be delivered in a controlled release system.
  • a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla.
  • a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)).
  • the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No.
  • a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination.
  • compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier.
  • pharmaceutically acceptable means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans.
  • carrier refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered.
  • Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like.
  • Water is a preferred carrier when the pharmaceutical composition is administered intravenously.
  • Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions.
  • Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like.
  • the composition if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents.
  • compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like.
  • the composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides.
  • Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin.
  • Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient.
  • the formulation should suit the mode of administration.
  • the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings.
  • compositions for intravenous administration are solutions in sterile isotonic aqueous buffer.
  • the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection.
  • the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent.
  • composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline.
  • an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.
  • the compounds of the invention can be formulated as neutral or salt forms.
  • Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc.
  • the amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques.
  • in vitro assays may optionally be employed to help identify optimal dosage ranges.
  • the precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems.
  • the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight.
  • the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight.
  • human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible.
  • the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation.
  • Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the aberrant expression and/or activity of Therapeutic protein.
  • the invention provides for the detection of aberrant expression of a Therapeutic protein, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein expression level compared to the standard expression level is indicative of aberrant expression.
  • the invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the Therapeutic protein or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein, and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein gene expression level compared to the standard expression level is indicative of a particular disorder.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer.
  • Antibodies of the invention or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen et al., J. Cell. Biol. 105:3087-3096 (1987)).
  • Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase
  • radioisotopes such as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc)
  • luminescent labels such as luminol
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the Therapeutic protein is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with aberrant expression of the therapeutic protein.
  • Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system.
  • the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99 mTc.
  • the labeled antibody, antibody fragment, or albumin fusion protein comprising at least a fragement or variant of an antibody that binds a Therapeutic protein will then preferentially accumulate at the location of cells which contain the specific Therapeutic protein. In vivo tumor imaging is described in S. W.
  • the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days.
  • monitoring of the disease or disorder is carried out by repeating the method for diagnosing the disease or disease, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc.
  • Presence of the labeled molecule can be detected in the patient using methods known in the art for in vivo scanning. These methods depend upon the type of label used. Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography.
  • CT computed tomography
  • PET position emission tomography
  • MRI magnetic resonance imaging
  • sonography sonography
  • the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S. Pat. No. 5,441,050).
  • the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument.
  • the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography.
  • the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI).
  • MRI magnetic resonance imaging
  • kits that can be used in the above methods.
  • a kit comprises an antibody, preferably a purified antibody, in one or more containers.
  • the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit.
  • the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest.
  • kits of the present invention contain a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate).
  • a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate.
  • the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides.
  • a kit may include a control antibody that does not react with the polypeptide of interest.
  • a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody.
  • a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry).
  • the kit may include a recombinantly produced or chemically synthesized polypeptide antigen.
  • the polypeptide antigen of the kit may also be attached to a solid support.
  • the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached.
  • a kit may also include a non-attached reporter-labeled anti-human antibody.
  • binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody.
  • the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention.
  • the diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody.
  • the antibody is attached to a solid support.
  • the antibody may be a monoclonal antibody.
  • the detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen.
  • test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention.
  • the reagent After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled-anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support.
  • the reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined.
  • the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, Mo.).
  • the solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plate or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s).
  • the invention provides an assay system or kit for carrying out this diagnostic method.
  • the kit generally includes a support with surface-bound recombinant antigens, and a reporter-labeled anti-human antibody for detecting surface-bound anti-antigen antibody.
  • albumin fusion protein refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof).
  • An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or chemical conjugation to one another.
  • the Therapeutic protein and albumin protein, once part of the albumin fusion protein may be referred to as a “portion”, “region” or “moiety” of the albumin fusion protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein.
  • the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin.
  • the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin.
  • the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin.
  • the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-terminal portion.
  • a Therapeutic protein as the C-terminal portion may also be used.
  • the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-terminus of albumin.
  • the Therapeutic proteins fused at the N- and C-termini are the same Therapeutic proteins.
  • the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins.
  • the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins which may be used to treat or prevent the same disease, disorder, or condition (e.g. as listed in the “Preferred Indication Y” column of Table 1).
  • the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins which may be used to treat or prevent diseases or disorders (e.g. as listed in the “Preferred Indication Y” column of Table 1) which are known in the art to commonly occur in patients simultaneously.
  • albumin fusion proteins of the invention may also be produced by inserting the Therapeutic protein or peptide of interest (e.g., a Therapeutic protein X as diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of HA.
  • a Therapeutic protein X as diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof
  • albumin fusion proteins of the invention may also be produced by inserting the Therapeutic protein or peptide of interest (e.g., a Therapeutic protein X as diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of HA.
  • a Therapeutic protein X as diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof
  • PDB identifiers IAO6, 1BJ5, 1BKE, 1BM0, 1E7E to 1E7I and 1UOR for the most part extend away from the body of the molecule.
  • These loops are useful for the insertion, or internal fusion, of therapeutically active peptides, particularly those requiring a secondary structure to be functional, or Therapeutic proteins, to essentially generate an albumin molecule with specific biological activity.
  • Loops in human albumin structure into which peptides or polypeptides may be inserted to generate albumin fusion proteins of the invention include: Val54-Asn61, Thr76-Asp89, Ala92-Glu100, Gln170-Ala176, His 247-Glu252, Glu 266-Glu277, Glu 280-His288, Ala362-Glu368, Lys439-Pro447,Val462-Lys475, Thr478-Pro486, and Lys560-Thr566.
  • peptides or polypeptides are inserted into the Val54-Asn61, Gln170-Ala176, and/or Lys560-Thr566 loops of mature human albumin (SEQ ID NO: 18).
  • Peptides to be inserted may be derived from either phage display or synthetic peptide libraries screened for specific biological activity or from the active portions of a molecule with the desired function. Additionally, random peptide libraries may be generated within particular loops or by insertions of randomized peptides into particular loops of the HA molecule and in which all possible combinations of amino acids are represented.
  • Such library(s) could be generated on HA or domain fragments of HA by one of the following methods:
  • the HA or HA domain fragment may also be made multifunctional by grafting the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment.
  • peptides inserted into a loop of human serum albumin are peptide fragments or peptide variants of the Therapeutic proteins disclosed in Table 1. More particulary, the invention encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids in length inserted into a loop of human serum albumin.
  • the invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the N-terminus of human serum albumin.
  • the invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the C-terminus of human serum albumin.
  • the albumin fusion proteins of the invention may have one HA-derived region and one Therapeutic protein-derived region. Multiple regions of each protein, however, may be used to make an albumin fusion protein of the invention. Similarly, more than one Therapeutic protein may be used to make an albumin fusion protein of the invention. For instance, a Therapeutic protein may be fused to both the N- and C-terminal ends of the HA. In such a configuration, the Therapeutic protein portions may be the same or different Therapeutic protein molecules.
  • the structure of bifunctional albumin fusion proteins may be represented as: X-HA-Y or Y-HA-X.
  • an anti-BLySTM scFv-HA-IFN ⁇ -2b fusion may be prepared to modulate the immune response to IFN ⁇ -2b by anti-BLySTM scFv.
  • An alternative is making a bi (or even multi) functional dose of HA-fusions e.g. HA-IFN ⁇ -2b fusion mixed with HA-anti-BLySTM scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc.
  • Bi- or multi-functional albumin fusion proteins may also be prepared to target the Therapeutic protein portion of a fusion to a target organ or cell type via protein or peptide at the opposite terminus of HA.
  • the peptides could be obtained by screening libraries constructed as fusions to the N-, C- or N- and C-termini of HA, or domain fragment of HA, of typically 6, 8, 12, 20 or 25 or X n (where X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented.
  • X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented.
  • a particular advantage of this approach is that the peptides may be selected in situ on the HA molecule and the properties of the peptide would therefore be as selected for rather than, potentially, modified as might be the case for a peptide derived by any other method then being attached to HA.
  • the albumin fusion proteins of the invention may include a linker peptide between the fused portions to provide greater physical separation between the moieties and thus maximize the accessibility of the Therapeutic protein portion, for instance, for binding to its cognate receptor.
  • the linker peptide may consist of amino acids such that it is flexible or more rigid.
  • the linker sequence may be cleavable by a protease or chemically to yield the growth hormone related moiety.
  • the protease is one which is produced naturally by the host, for example the S. cerevisiae protease kex2 or equivalent proteases.
  • the albumin fusion proteins of the invention may have the following formula R1-L-R2; R2-L-R1; or R1-L-R2-L-R1, wherein R1 is at least one Therapeutic protein, peptide or polypeptide sequence, and not necessarily the same Therapeutic protein, L is a linker and R2 is a serum albumin sequence.
  • Albumin fusion proteins of the invention comprising a Therapeutic protein have extended shelf life compared to the shelf life the same Therapeutic protein when not fused to albumin. Shelf-life typically refers to the time period over which the therapeutic activity of a Therapeutic protein in solution or in some other storage formulation, is stable without undue loss of therapeutic activity. Many of the Therapeutic proteins are highly labile in their unfused state. As described below, the typical shelf-life of these Therapeutic proteins is markedly prolonged upon incorporation into the albumin fusion protein of the invention.
  • Albumin fusion proteins of the invention with “prolonged” or “extended” shelf-life exhibit greater therapeutic activity relative to a standard that has been subjected to the same storage and handling conditions.
  • the standard may be the unfused full-length Therapeutic protein.
  • the Therapeutic protein portion of the albumin fusion protein is an analog, a variant, or is otherwise altered or does not include the complete sequence for that protein, the prolongation of therapeutic activity may alternatively be compared to the unfused equivalent of that analog, variant, altered peptide or incomplete sequence.
  • an albumin fusion protein of the invention may retain greater than about 100% of the therapeutic activity, or greater than about 105%, 110%, 120%, 130%, 150% or 200% of the therapeutic activity of a standard when subjected to the same storage and handling conditions as the standard when compared at a given time point.
  • Shelf-life may also be assessed in terms of therapeutic activity remaining after storage, normalized to therapeutic activity when storage began.
  • Albumin fusion proteins of the invention with prolonged or extended shelf-life as exhibited by prolonged or extended therapeutic activity may retain greater than about 50% of the therapeutic activity, about 60%, 70%, 80%, or 90% or more of the therapeutic activity of the equivalent unfused Therapeutic protein when subjected to the same conditions.
  • an albumin fusion protein of the invention comprising hGH fused to the full length HA sequence may retain about 80% or more of its original activity in solution for periods of up to 5 weeks or more under various temperature conditions.
  • the albumin fusion proteins of the invention may be produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line.
  • the polypeptide is secreted from the host cells.
  • Hiramatsu et al. Appl Environ Microbiol 56:2125 (1990); Appl Environ Microbiol 57:2052 (1991) found that the N-terminal portion of the pro sequence in the Mucor pusillus rennin pre-pro leader was important.
  • the pro sequences were believed to assist in the folding of the hGH by acting as an intramolecular chaperone.
  • the present invention shows that HA or fragments of HA can perform a similar function.
  • a particular embodiment of the invention comprises a DNA construct encoding a signal sequence effective for directing secretion in yeast, particularly a yeast-derived signal sequence (especially one which is homologous to the yeast host), and the fused molecule of the first aspect of the invention, there being no yeast-derived pro sequence between the signal and the mature polypeptide.
  • Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
  • the present invention also includes a cell, preferably a yeast cell transformed to express an albumin fusion protein of the invention.
  • a cell preferably a yeast cell transformed to express an albumin fusion protein of the invention.
  • the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away.
  • Many expression systems are known and may be used, including bacteria (for example E.
  • yeasts for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris
  • filamentous fungi for example Aspergillus
  • plant cells animal cells and insect cells.
  • D88 Preferred yeast strains to be used in the production of albumin fusion proteins
  • D88 [leu2-3, leu2-122, canI, praI, ubc4] is a derivative of parent strain AH22his + (also known as DB1; see, e.g., Sleep et al. Biotechnology 8:42-46 (1990)).
  • the strain contains a leu2 mutation which allows for auxotropic selection of 2 micron-based plasmids that contain the LEU2 gene.
  • D88 also exhibits a derepression of PRB1 in glucose excess.
  • the PRB1 promoter is normally controlled by two checkpoints that monitor glucose levels and growth stage.
  • the promoter is activated in wild type yeast upon glucose depletion and entry into stationary phase.
  • Strain D88 exhibits the repression by glucose but maintains the induction upon entry into stationary phase.
  • the PRA1 gene encodes a yeast vacuolar protease, YscA endoprotease A, that is localized in the ER.
  • the UBC4 gene is in the ubiquitination pathway and is involved in targeting short lived and abnormal proteins for ubiquitin dependant degradation. Isolation of this ubc4 mutation was found to increase the copy number of an expression plasmid in the cell and cause an increased level of expression of a desired protein expressed from the plasmid (see, e.g., International Publication No. WO99/00504, hereby incorporated in its entirety by reference herein).
  • DXY1 a derivative of D88, has the following genotype: [leu2-3, leu2-122, canI, pra1, ubc4, ura3::yap3].
  • this strain also has a knockout of the YAP3 protease.
  • This protease causes cleavage of mostly di-basic residues (RR, RK, KR, KK) but can also promote cleavage at single basic residues in proteins. Isolation of this yap3 mutation resulted in higher levels of full length HSA production (see, e.g., U.S. Pat. No. 5,965,386 and Kerry-Williams et al., Yeast 14:161-169 (1998), hereby incorporated in their entireties by reference herein).
  • BXP10 has the following genotype: leu2-3, leu2-122, can1, pra1, ubc4, ura3, yap3::URA3, lys2, hsp150::LYS2, pmt1::URA3.
  • this strain also has a knockout of the PMT1 gene and the HSP150 gene.
  • the PMT1 gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D-mannose protein O-mannosyltransferases (Pmts).
  • the transmembrane topology of Pmt1p suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in O-linked glycosylation.
  • This mutation serves to reduce/eliminate O-linked glycosylation of HSA fusions (see, e.g., International Publication No. WO00/44772, hereby incorporated in its entirety by reference herein).
  • Studies revealed that the Hsp150 protein is inefficiently separated from rHA by ion exchange chromatography.
  • the mutation in the HSP150 gene removes a potential contaminant that has proven difficult to remove by standard purification techniques. See, e.g., U.S. Pat. No. 5,783,423, hereby incorporated in its entirety by reference herein.
  • the desired protein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid.
  • the yeasts are transformed with a coding sequence for the desired protein in any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) Methods Enzymol. 194, 182.
  • Successfully transformed cells i.e., cells that contain a DNA construct of the present invention
  • cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide.
  • Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208.
  • the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA.
  • Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3.
  • Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps).
  • Preferred vectors for making albumin fusion proteins for expression in yeast include pPPC0005, pScCHSA, pScNHSA, and pC4:HSA which are described in detail in Example 2.
  • FIG. 4 shows a map of the pPPC0005 plasmid that can be used as the base vector into which polynucleotides encoding Therapeutic proteins may be cloned to form HA-fusions. It contains a PRB1 S. cerevisiae promoter (PRB1p), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADH1 S. cerevisiae terminator sequence.
  • sequence of the fusion leader sequence consists of the first 19 amino acids of the signal peptide of human serum albumin (SEQ ID NO:29) and the last five amino acids of the mating factor alpha 1 promoter (SLDKR, see EP-A-387 319 which is hereby incorporated by reference in its entirety.
  • plasmids pPPC0005, pScCHSA, pScNHSA, and pC4:HSA were deposited on Apr. 11, 2001 at the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209 and given accession numbers ATCC ______, ______, ______, and ______, respectively.
  • Another vector useful for expressing an albumin fusion protein in yeast the pSAC35 vector which is described in Sleep et al., BioTechnology 8:42 (1990) which is hereby incorporated by reference in its entirety.
  • a variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules.
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors.
  • the DNA segment generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA polymerase or E. coli DNA polymerase I, enzymes that remove protruding, -single-stranded termini with their 3′ 5′-exonucleolytic activities, and fill in recessed 3′-ends with their polymerizing activities.
  • the combination of these activities therefore generates blunt-ended DNA segments.
  • the blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
  • an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase.
  • the products of the reaction are DNA segments carrying polymeric linker sequences at their ends.
  • These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment.
  • a desirable way to modify the DNA in accordance with the invention is to use the polymerase chain reaction as disclosed by Saiki et al. (1988) Science 239, 487-491.
  • the DNA to be enzymatically amplified is flanked by two specific oligonucleotide primers which themselves become incorporated into the amplified DNA.
  • the specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin fusion proteins are Pichia (Hansenula), Saccharomyces, Kluyveromyces, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Debaromyces, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like.
  • Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are S. cerevisiae, S. italicus and S. rouxii.
  • Examples of Kluyveromyces spp. are K. fragilis, K. lactis and K. marxianus.
  • a suitable Torulaspora species is T. delbrueckii.
  • Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorpha ), P. anomala (formerly H. anomala ) and P. pastoris .
  • Methods for the transformation of S. cerevisiae are taught generally in EP 251 744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
  • Preferred exemplary species of Saccharomyces include S. cerevisiae, S. italicus, S. diastaticus, and Zygosaccharomyces rouxii.
  • Preferred exemplary species of Kluyveromyces include K. fragilis and K. lactis.
  • Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta ), H. anomala (now Pichia anomala ), and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris .
  • Preferred exemplary species of Aspergillus include A. niger and A. nidulans.
  • Preferred exemplary species of Yarrowia include Y. lipolytica.
  • yeast species are available from the ATCC.
  • the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmt1 mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos.
  • Suitable promoters for S. cerevisiae include those associated with the PGKI gene, GAL1 or GAL10 genes, CYCI, PHO5, TRPI, ADHI, ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5′ regulatory regions with parts of 5′ regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
  • Convenient regulatable promoters for use in Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbp1 gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, Calif.
  • Suitable promoters include AOXI and AOX2.
  • Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst EP 361 991, Fleer et al. (1991) and other-publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • the transcription termination signal is preferably the 3′ flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation.
  • Suitable 3′ flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the S. cerevisiae ADHI gene is preferred.
  • the desired albumin fusion protein may be initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen.
  • Leaders useful in S. cerevisiae include that from the mating factor polypeptide (MF -1) and the hybrid leaders of EP-A-387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 911036516), acid phosphatase (PH05), the pre-sequence of MFoz-1, 0 glucanase (BGL2) and killer toxin; S. diastaticus glucoamylase II; S. carlsbergensis -galactosidase (MEL1); K. lactis killer toxin; and Candida glucoarnylase.
  • SUC2 S. cerevisiae invertase
  • the present invention also relates to vectors containing a polynucleotide encoding an albumin fusion protein of the present invention, host cells, and the production of albumin fusion proteins by synthetic and recombinant techniques.
  • the vector may be, for example, a phage, plasmid, viral, or retroviral vector.
  • Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells.
  • the polynucleotides encoding albumin fusion proteins of the invention may be joined to a vector containing a selectable marker for propagation in a host.
  • a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells.
  • the polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan.
  • the expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation.
  • the coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • the expression vectors will preferably include at least one selectable marker.
  • markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in E. coli and other bacteria.
  • Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli , Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No.
  • insect cells such as Drosophila S2 and Spodoptera Sf9 cells
  • animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells
  • plant cells Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc.
  • preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia.
  • Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, Calif.).
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • polynucleotides encoding an albumin fusion protein of the invention may be fused to signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell.
  • E. coli one may wish to direct the expression of the protein to the periplasmic space.
  • Examples of signal sequences or proteins (or fragments thereof) to which the albumin fusion proteins of the invention may be fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase.
  • MBP maltose binding protein
  • ompA the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit
  • alkaline phosphatase Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-p series) available from New England Biolabs.
  • polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • Examples of signal peptides that may be fused to an albumin fusion protein of the invention in order to direct its secretion in mammalian cells include, but are not limited to, the MPIF-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134), the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:34), and a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID NO:35).
  • a suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1-19 of GenBank Accession Number AAA72759).
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively.
  • An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative.
  • Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene.
  • glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are herein incorporated by reference.
  • the present invention also relates to host cells containing the above-described vector constructs described herein, and additionally encompasses host cells containing nucleotide sequences of the invention that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art.
  • the host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell.
  • a host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired.
  • Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled.
  • different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed.
  • nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector.
  • the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Therapeutic protein may be replaced with an albumin fusion protein corresponding to the Therapeutic protein), and/or to include genetic material (e.g., heterologous polynucleotide sequences such as for example, an albumin fusion protein of the invention corresponding to the Therapeutic protein may be included).
  • the genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides.
  • heterologous polynucleotides e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof
  • heterologous control regions e.g., promoter and/or enhancer
  • endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication Number WO 96/29411; International Publication Number WO 94/12650; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
  • Albumin fusion proteins of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification.
  • HPLC high performance liquid chromatography
  • the albumin fusion proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAE, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAE, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns.
  • the albumin fusion proteins of the invention are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM columns and their equivalents and comparables.
  • the albumin fusion proteins of the invention are purified using Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharose, poros Phenyl, Butyl, Methyl, Octyl, Hexyl Toyopearl Phenyl, Butyl, Methyl, Octyl, Hexyl Resource/Source Phenyl, Butyl, Methyl, Octyl, Hexyl, Fractogel Phenyl, Butyl, Methyl, Octyl, Hexyl columns and their equivalents and comparables.
  • Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharose, poros Phenyl, Butyl, Methyl, Octyl, Hexyl Toyopearl Phenyl, Butyl, Me
  • albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but not limited to, sepharose S100, S200, S300, superdex resin columns and their equivalents and comparables.
  • albumin fusion proteins of the invention are purified using Affinity Chromatography including, but not limited to, Mimetic Dye affinity, peptide affinity and antibody affinity columns that are selective for either the HSA or the “fusion target” molecules.
  • albumin fusion proteins of the invention are purified using one or more Chromatography methods listed above. In other preferred embodiments, albumin fusion proteins of the invention are purified using one or more of the following Chromatography columns, Q sepharose FF column, SP Sepharose FF column, Q Sepharose High Performance Column, Blue Sepharose FF column, Blue Column, Phenyl Sepharose FF column, DEAE Sepharose FF, or Methyl Column.
  • albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44772 which is herein incorporated by reference in its entirety.
  • One of skill in the art could easily modify the process described therein for use in the purification of albumin fusion proteins of the invention.
  • Albumin fusion proteins of the present invention may be recovered from: products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, albumin fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes.
  • N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked.
  • the yeast Pichia pastoris is used to express albumin fusion proteins of the invention in a eukaryotic system.
  • Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source.
  • a main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using O 2 . This reaction is catalyzed by the enzyme alcohol oxidase.
  • Pichia pastoris In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O 2 .
  • alcohol oxidase produced from the AOX1 gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris . See Ellis, S. B., et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz, P. J, et al., Yeast 5:167-77 (1989); Tschopp, J. F., et al., Nucl. Acids Res. 15:3859-76 (1987).
  • a heterologous coding sequence such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the AOX1 regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
  • the plasmid vector pPIC9K is used to express DNA encoding an albumin fusion protein of the invention, as set forth herein, in a Pichea yeast system essentially as described in “Pichia Protocols: Methods in Molecular Biology,” D. R. Higgins and J. Cregg, eds. The Humana Press, Totowa, N.J., 1998.
  • This expression vector allows expression and secretion of a polypeptide of the invention by virtue of the strong AOX1 promoter linked to the Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
  • PHO alkaline phosphatase
  • yeast vectors could be used in place of pPIC9K, such as, pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, and PAO815, as one skilled in the art would readily appreciate, as long as the proposed expression construct provides appropriately located signals for transcription, translation, secretion (if desired), and the like, including an in-frame AUG as required.
  • high-level expression of a heterologous coding sequence such as, for example, a polynucleotide encoding an albumin fusion protein of the present invention, may be achieved by cloning the heterologous polynucleotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol.
  • albumin fusion proteins of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y., and Hunkapiller et al., Nature, 310:105-111 (1984)).
  • a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer.
  • nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence.
  • Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid
  • the invention encompasses albumin fusion proteins of the present invention which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH 4 ; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression.
  • the albumin fusion proteins may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein.
  • suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine ( 121 I, 123 I, 125 I, 131 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 111 In, 112 In, 113m In, 115m In), technetium ( 99 T
  • albumin fusion proteins of the present invention or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions, including but not limited to, 177 Lu, 90 Y, 166 Ho, and 153 Sm, to polypeptides.
  • the radiometal ion associated with the macrocyclic chelators is 111 In.
  • the radiometal ion associated with the macrocyclic chelator is 90 Y.
  • the macrocyclic chelator is 1,4,7,10-tetraazacyclododecane-N,N′,N′′,N′′′-tetraacetic acid (DOTA).
  • DOTA is attached to an antibody of the invention or fragment thereof via linker molecule.
  • linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art—see, for example, DeNardo et al., Clin Cancer Res. 4(10):2483-90 (1998); Peterson et al., Bioconjug. Chem. 10(4):553-7 (1999); and Zimmerman et al, Nucl. Med. Biol. 26(8):943-50 (1999); which are hereby incorporated by reference in their entirety.
  • the albumin fusion proteins of the invention may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide.
  • Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods.
  • Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination.
  • Albumin fusion proteins of the invention and antibodies that bind a Therapeutic protein or fragments or variants thereof can be fused to marker sequences, such as a peptide to facilitate purification.
  • the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311), among others, many of which are commercially available.
  • hexa-histidine provides for convenient purification of the fusion protein.
  • peptide tags useful for purification include, but are not limited to, the “HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the “flag” tag.
  • an albumin fusion protein of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi.
  • a cytotoxin or cytotoxic agent includes any agent that is detrimental to cells.
  • Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof.
  • Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g.
  • the conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents.
  • the drug moiety may be a protein or polypeptide possessing a desired biological activity.
  • Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, Int.
  • a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin
  • a protein such as tumor necrosis factor, alpha-interferon, ⁇ -interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an a
  • VEGI See, International Publication No. WO 99/23105
  • a thrombotic agent or an anti-angiogenic agent e.g., angiostatin or endostatin
  • biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-1”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • IL-1 interleukin-1
  • IL-2 interleukin-2
  • IL-6 interleukin-6
  • GM-CSF granulocyte macrophage colony stimulating factor
  • G-CSF granulocyte colony stimulating factor
  • Albumin fusion proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention.
  • solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene.
  • Albumin fusion proteins with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic.
  • the albumin fusion protein of the invention comprises only the VH domain of an antibody that binds a Therapeutic protein
  • the albumin fusion protein of the invention comprises only the VL domain of an antibody that binds a Therapeutic protein
  • Some Therapeutic antibodies are bispecific antibodies, meaning the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites.
  • the scFv fused to the N-terminus of albumin would correspond to one of the heavy/light (VH/VL) pairs of the original antibody that binds a Therapeutic protein and the scFv fused to the C-terminus of albumin would correspond to the other heavy/light (VH/VL) pair of the original antibody that binds a Therapeutic protein.
  • the chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like.
  • the albumin fusion proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties.
  • the polymer may be of any molecular weight, and may be branched or unbranched.
  • the preferred molecular weight is between about 1 kDa and about 100 kDa (the term “about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing.
  • Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog).
  • the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa.
  • the polyethylene glycol may have a branched structure.
  • Branched polyethylene glycols are described, for example, in U.S. Pat. No. 5,643,575; Morpurgo et al., Appl. Biochem. Biotechnol. 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al., Bioconjug. Chem. 10:638-646 (1999), the disclosures of each of which are incorporated herein by reference.
  • polyethylene glycol molecules should be attached to the protein with consideration of effects on functional or antigenic domains of the protein.
  • attachment methods available to those skilled in the art, such as, for example, the method disclosed in EP 0 401 384 (coupling PEG to G-CSF), herein incorporated by reference; see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride.
  • polyethylene glycol may be covalently bound through amino acid residues via reactive group, such as a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound.
  • the amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue.
  • Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
  • polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues.
  • polyethylene glycol can be linked to proteins via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues.
  • One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein.
  • polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein.
  • the method of obtaining the N-terminally pegylated preparation i.e., separating this moiety from other monopegylated moieties if necessary
  • Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved.
  • pegylation of the albumin fusion proteins of the invention may be accomplished by any number of means.
  • polyethylene glycol may be attached to the albumin fusion protein either directly or by an intervening linker.
  • Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. J. of Hematol. 68:1-18 (1998); U.S. Pat. No. 4,002,531; U.S. Pat. No. 5,349,052; WO 95/06058; and WO 98/32466, the disclosures of each of which are incorporated herein by reference.
  • One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (ClSO 2 CH 2 CF 3 ).
  • MPEG monmethoxy polyethylene glycol
  • ClSO 2 CH 2 CF 3 tresylchloride
  • polyethylene glycol is directly attached to amine groups of the protein.
  • the invention includes protein-polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
  • Polyethylene glycol can also be attached to proteins using a number of different intervening linkers.
  • U.S. Pat. No. 5,612,460 discloses urethane linkers for connecting polyethylene glycol to proteins.
  • Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuccinate, MPEG activated with 1,1′-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives.
  • the number of polyethylene glycol moieties attached to each albumin fusion protein of the invention may also vary.
  • the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules.
  • the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10-12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule. Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992).
  • polypeptides of the invention can be recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but are not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification.
  • HPLC high performance liquid chromatography
  • the presence and quantity of albumin fusion proteins of the invention may be determined using ELISA, a well known immunoassay known in the art.
  • ELISA protocol that would be useful for detecting/quantifying albumin fusion proteins of the invention, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the albumin fusion protein of the invention (at one or more different concentrations), adding a secondary anti-Therapeutic protein specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody.
  • the ELISA plate might be coated with the anti-Therapeutic protein specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody.
  • polynucleotides of the present invention are useful to produce the albumin fusion proteins of the invention.
  • polynucleotides of the invention encoding albumin fusion proteins
  • Polynucleotides of the present invention are also useful in gene therapy.
  • One goal of gene therapy is to insert a normal gene into an organism having a defective gene, in an effort to correct the genetic defect.
  • the polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner.
  • Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell. Additional non-limiting examples of gene therapy methods encompassed by the present invention are more thoroughly described elsewhere herein (see, e.g., the sections labeled “Gene Therapy”, and Examples 17 and 18).
  • polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques.
  • Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissue(s) (e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29:577-580 (1981)) or cell type(s) (e.g., immunocytochemistry assays).
  • tissue(s) e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29:577-580 (1981)
  • cell type(s) e.g., immunocytochemistry assays
  • Albumin fusion proteins can be used to assay levels of polypeptides in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell. Biol. 105:3087-3096 (1987)).
  • Other methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • Suitable assay labels include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine ( 131 I, 125 I, 123 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 115m In, 113m In, 112 In, 111 In), and technetium ( 99 Tc, 99m Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe), fluorine ( 18 F), 153 Sm, 177 Lu, 159 Gd, 149 Pm, 140 La, 175 Yb, 166 Ho, 90 Y, 47 Sc, 186 Re, 188 Re, 142 Pr, 105 Rh, 97 Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and bio
  • enzyme labels such
  • Albumin fusion proteins of the invention can also be detected in vivo by imaging.
  • Labels or markers for in vivo imaging of protein include those detectable by X-radiography, nuclear magnetic resonance (NMR) or electron spin relaxtion (ESR).
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject.
  • suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients given to a cell line expressing the albumin fusion protein of the invention.
  • An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, 131 I, 112 In, 99m Tc, ( 131 I, 125 I, 123 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 115m In, 113m In, 112 In, 111 In), and technetium ( 99 Tc, 99m Tc), thallium ( 201 Ti), gallium ( 68 Ga, 67 Ga), palladium ( 103 Pd), molybdenum ( 99 Mo), xenon ( 133 Xe), fluorine ( 18 F, 153 Sm, 177 Lu, 159 Gd, 149 Pm, 140 La, 175 Yb, 166 Ho, 90 Y, 47 Sc, 186 Re, 188 Re, 142 Pr, 105 Rh, 97 Ru), a radio-opaque substance, or a material detectable by nuclear magnetic resonance
  • the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images.
  • the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99m Tc.
  • the labeled albumin fusion protein will then preferentially accumulate at locations in the body (e.g., organs, cells, extracellular spaces or matrices) where one or more receptors, ligands or substrates (corresponding to that of the Therapeutic protein used to make the albumin fusion protein of the invention) are located.
  • the labeled albumin fusion protein will then preferentially accumulate at the locations in the body (e.g., organs, cells, extracellular spaces or matrices) where the polypeptides/epitopes corresponding to those bound by the Therapeutic antibody (used to make the albumin fusion protein of the invention) are located.
  • the locations in the body e.g., organs, cells, extracellular spaces or matrices
  • the polypeptides/epitopes corresponding to those bound by the Therapeutic antibody used to make the albumin fusion protein of the invention.
  • In vivo tumor imaging is described in S. W. Burchiel et al., “Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
  • the protocols described therein could easily be modified by one of skill in the
  • the invention provides a method for the specific delivery of albumin fusion proteins of the invention to cells by administering albumin fusion proteins of the invention (e.g., polypeptides encoded by polynucleotides encoding albumin fusion proteins of the invention and/or antibodies) that are associated with heterologous polypeptides or nucleic acids.
  • the invention provides a method for delivering a Therapeutic protein into the targeted cell.
  • the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell.
  • a single stranded nucleic acid e.g., antisense or ribozymes
  • double stranded nucleic acid e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering albumin fusion proteins of the invention in association with toxins or cytotoxic prodrugs.
  • toxin is meant one or more compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or on the surface of a cell that under defined conditions cause the cell's death.
  • Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin.
  • radioisotopes known in the art
  • compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseu
  • Toxin also includes a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213 Bi, or other radioisotopes such as, for example, 103 Pd, 133 Xe, 131 I, 68 Ge, 57 Co, 65 Zn, 85 Sr, 32 P, 35 S, 90 Y, 153 Sm, 153 Gd, 169 Yb, 51 Cr, 54 Mn, 75 Se, 113 Sn, 90 Yttrium, 117 Tin, 186 Rhenium, 166 Holmium, and 188 Rhenium; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • alpha-emitters such as, for example, 213 Bi
  • radioisotopes such as, for example, 103 Pd, 133 Xe, 131 I, 68 Ge,
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 90 Y.
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 111 In.
  • the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 131 I.
  • the albumin fusion proteins of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans. Such disorders include, but are not limited to, those described herein under the section heading “Biological Activities,” below.
  • the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder.
  • a diagnostic method of a disorder involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder.
  • the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting
  • albumin fusion proteins of the present invention can be used to treat or prevent diseases or conditions such as, for example, neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions.
  • patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of a polypeptide (e.g., an oncogene or tumor supressor), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of the immune response to proliferative cells or tissues).
  • a polypeptide e.g., insulin
  • a different polypeptide e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins
  • albumin fusion proteins comprising of at least a fragment or variant of a Therapeutic antibody can also be used to treat disease (as described supra, and elsewhere herein).
  • administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can bind, and/or neutralize the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, and/or reduce overproduction of the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds.
  • an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can activate the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, by binding to the polypeptide bound to a membrane (receptor).
  • the albumin fusion proteins of the invention of the present invention can be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art.
  • Albumin fusion proteins of the invention can also be used to raise antibodies, which in turn may be used to measure protein expression of the Therapeutic protein, albumin protein, and/or the albumin fusion protein of the invention from a recombinant cell, as a way of assessing transformation of the host cell, or in a biological sample.
  • the albumin fusion proteins of the present invention can be used to test the biological activities described herein.
  • the compounds of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans.
  • disorders include, but are not limited to, those described for each Therapeutic protein in the corresponding row of Table 1 and herein under the section headings “Immune Activity,” “Blood Related Disorders,” “Hyperproliferative Disorders,” “Renal Disorders,” “Cardiovascular Disorders,” “Respiratory Disorders,” “Anti-Angiogenesis Activity,” “Diseases at the Cellular Level,” “Wound Healing and Epithelial Cell Proliferation,” “Neural Activity and Neurological Diseases,” “Endocrine Disorders,” “Reproductive System Disorders,” “Infectious Disease,” “Regeneration,” and/or “Gastrointestinal Disorders,” infra.
  • substantially altered (increased or decreased) levels of gene expression can be detected in tissues, cells or bodily fluids (e.g., sera, plasma, urine, semen, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a “standard” gene expression level, that is, the expression level in tissues or bodily fluids from an individual not having the disorder.
  • a diagnostic method useful during diagnosis of a disorder which involves measuring the expression level of the gene encoding a polypeptide in tissues, cells or body fluid from an individual and comparing the measured gene expression level with a standard gene expression level, whereby an increase or decrease in the gene expression level(s) compared to the standard is indicative of a disorder.
  • diagnostic assays may be performed in vivo or in vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue.
  • the present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome
  • assaying the expression level of the gene encoding a polypeptide is intended qualitatively or quantitatively measuring or estimating the level of a particular polypeptide (e.g. a polypeptide corresponding to a Therapeutic protein disclosed in Table 1) or the level of the mRNA encoding the polypeptide of the invention in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA level in a second biological sample).
  • the polypeptide expression level or mRNA level in the first biological sample is measured or estimated and compared to a standard polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having the disorder.
  • a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison.
  • biological sample any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeptides of the invention (including portions thereof) or mRNA.
  • biological samples include body fluids (such as sera, plasma, urine, synovial fluid and spinal fluid) and tissue sources found to express the full length or fragments thereof of a polypeptide or mRNA. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy is the preferred source.
  • Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem. 162:156-159 (1987). Levels of mRNA encoding the polypeptides of the invention are then assayed using any appropriate method. These include Northern blot analysis, S1 nuclease mapping, the polymerase chain reaction (PCR), reverse transcription in combination with the polymerase chain reaction (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR).
  • PCR polymerase chain reaction
  • RT-PCR reverse transcription in combination with the polymerase chain reaction
  • RT-LCR reverse transcription in combination with the ligase chain reaction
  • the present invention also relates to diagnostic assays such as quantitative and diagnostic assays for detecting levels of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the invention, in a biological sample (e.g., cells and tissues), including determination of normal and abnormal levels of polypeptides.
  • a diagnostic assay in accordance with the invention for detecting abnormal expression of polypeptides that bind to, are bound by, or associate with albumin fusion proteins compared to normal control tissue samples may be used to detect the presence of tumors.
  • Assay techniques that can be used to determine levels of a polypeptide that bind to, are bound by, or associate with albumin fusion proteins of the present invention in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Assaying polypeptide levels in a biological sample can occur using any art-known method.
  • Assaying polypeptide levels in a biological sample can occur using a variety of techniques. For example, polypeptide expression in tissues can be studied with classical immunohistological methods (Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al., J. Cell. Biol. 105:3087-3096 (1987)). Other methods useful for detecting polypeptide gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA).
  • ELISA enzyme linked immunosorbent assay
  • RIA radioimmunoassay
  • Suitable antibody assay labels include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99m Tc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • enzyme labels such as, glucose oxidase, and radioisotopes, such as iodine ( 125 I, 121 I), carbon ( 14 C), sulfur ( 35 S), tritium ( 3 H), indium ( 112 In), and technetium ( 99m Tc)
  • fluorescent labels such as fluorescein and rhodamine, and biotin.
  • the tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the gene of interest (such as, for example, cancer).
  • the protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which is incorporated herein by reference in its entirety.
  • the isolated cells can be derived from cell culture or from a patient.
  • the analysis of cells taken from culture may be a necessary step in the assessment of cells that could be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the gene.
  • albumin fusion proteins may be used to quantitatively or qualitatively detect the presence of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the present invention. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled albumin fusion protein coupled with light microscopic, flow cytometric, or fluorimetric detection.
  • albumin fusion proteins comprising at least a fragment or variant of an antibody that specifically binds at least a Therapeutic protein disclosed herein (e.g., the Therapeutic proteins disclosed in Table 1) or otherwise known in the art may be used to quantitatively or qualitatively detect the presence of gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody coupled with light microscopic, flow cytometric, or fluorimetric detection.
  • the albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron microscopy or non-immunological assays, for in situ detection of polypeptides that bind to, are bound by, or associate with an albumin fusion protein of the present invention.
  • In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody or polypeptide of the present invention.
  • the albumin fusion proteins are preferably applied by overlaying the labeled albumin fusion proteins onto a biological sample.
  • Immunoassays and non-immunoassays that detect polypeptides that bind to, are bound by, or associate with albumin fusion proteins will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of binding gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art.
  • the biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins.
  • a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins.
  • the support may then be washed with suitable buffers followed by treatment with the detectably labeled albumin fusion protein of the invention.
  • the solid phase support may then be washed with the buffer a second time to remove unbound antibody or polypeptide.
  • the antibody is subsequently labeled.
  • the amount of bound label on solid support may then be detected by conventional means.
  • solid phase support or carrier any support capable of binding a polypeptide (e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.
  • a polypeptide e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.
  • Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite.
  • the nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention.
  • the support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to a polypeptide.
  • the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod.
  • the surface may be flat such as a sheet, test strip, etc.
  • Preferred supports include polystyrene beads.
  • the binding activity of a given lot of albumin fusion protein may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation.
  • polypeptide in addition to assaying polypeptide levels in a biological sample obtained from an individual, polypeptide can also be detected in vivo by imaging.
  • albumin fusion proteins of the invention are used to image diseased or neoplastic cells.
  • Labels or markers for in vivo imaging of albumin fusion proteins of the invention include those detectable by X-radiography, NMR, MRI, CAT-scans or ESR.
  • suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject.
  • suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients of a cell line (or bacterial or yeast strain) engineered.
  • albumin fusion proteins of the invention whose presence can be detected, can be administered.
  • albumin fusion proteins of the invention labeled with a radio-opaque or other appropriate compound can be administered and visualized in vivo, as discussed, above for labeled antibodies.
  • polypeptides can be utilized for in vitro diagnostic procedures.
  • a polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, 112 In, 99m Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for a disorder.
  • an appropriate detectable imaging moiety such as a radioisotope (for example, 131 I, 112 In, 99m Tc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for a disorder.
  • a radioisotope for example, 131 I, 112 In, 99m Tc
  • a radio-opaque substance for example, parenterally, subcutaneously or intraperitoneally
  • the quantity of radioactivity injected will normally range from
  • the labeled albumin fusion protein will then preferentially accumulate at the locations in the body which contain a polypeptide or other substance that binds to, is bound by or associates with an albumin fusion protein of the present invention.
  • In vivo tumor imaging is described in S. W. Burchiel et al., “Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
  • an albumin fusion protein of the present invention can be detectably labeled is by linking the same to a reporter enzyme and using the linked product in an enzyme immunoassay (EIA) (Voller, A., “The Enzyme Linked Immunosorbent Assay (ELISA)”, 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, Md.); Voller et al., J. Clin. Pathol. 31:507-520 (1978); Butler, J. E., Meth. Enzymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, Fla.,; Ishikawa, E.
  • EIA enzyme immunoassay
  • the reporter enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means.
  • Reporter enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the reporter enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
  • Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays.
  • radioimmunoassay RIA
  • the radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography.
  • the albumin fusion protein can also be detectably labeled using fluorescence emitting metals such as 152 Eu, or others of the lanthamide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • DTPA diethylenetriaminepentacetic acid
  • EDTA ethylenediaminetetraacetic acid
  • the albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged albumin fusion protein is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction.
  • chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester.
  • a bioluminescent compound may be used to label albumin fusion proteins of the present invention.
  • Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence.
  • Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin.
  • Transgenic organisms that express the albumin fusion proteins of the invention are also included in the invention.
  • Transgenic organisms are genetically modified organisms into which recombinant, exogenous or cloned genetic material has been transferred. Such genetic material is often referred to as a transgene.
  • the nucleic acid sequence of the transgene may include one or more transcriptional regulatory sequences and other nucleic acid sequences such as introns, that may be necessary for optimal expression and secretion of the encoded protein.
  • the transgene may be designed to direct the expression of the encoded protein in a manner that facilitates its recovery from the organism or from a product produced by the organism, e.g. from the milk, blood, urine, eggs, hair or seeds of the organism.
  • the transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal.
  • the transgene may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise normally found or at the normal locus for the transgene.
  • the term “germ cell line transgenic organism” refers to a transgenic organism in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability of the transgenic organism to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic information, then they too are transgenic organisms.
  • the alteration or genetic information may be foreign to the species of organism to which the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene.
  • a transgenic organism may be a transgenic animal or a transgenic plant.
  • Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Pat. No. 4,736,866; U.S. Pat. No. 5,602,307; Mullins et al. (1993) Hypertension 22(4):630-633; Brenin et al. (1997) Surg. Oncol. 6(2)99-110; Tuan (ed.), Recombinant Gene Expression Protocols, Methods in Molecular Biology No. 62, Humana Press (1997)).
  • the method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules.
  • Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Pat. No. 5,489,743 and U.S. Pat. No. 5,602,307.
  • mice A number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Pat. No. 4,736,866); express simian SV40 T-antigen (U.S. Pat. No. 5,728,915); lack the expression of interferon regulatory factor 1 (IRF-1) (U.S. Pat. No. 5,731,490); exhibit dopaminergic dysfunction (U.S. Pat. No. 5,723,719); express at least one human gene which participates in blood pressure control (U.S. Pat. No. 5,731,489); display greater similarity to the conditions existing in naturally occurring Alzheimer's disease (U.S. Pat. No.
  • mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species.
  • Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g., Kim et al. (1997) Mol. Reprod. Dev. 46(4):515-526; Houdebine (1995) Reprod. Nutr. Dev. 35(6):609-617; Petters (1994) Reprod. Fertil. Dev. 6(5):643-645; Schnieke et al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J. Animal Science 75(2):578-585).
  • transgene-encoded protein of the invention may be put under the control of a promoter that is preferentially activated in mammary epithelial cells.
  • Promoters that control the genes encoding milk proteins are preferred, for example the promoter for casein, beta lactoglobulin, whey acid protein, or lactalbumin (see, e.g., DiTullio (1992) BioTechnology 10:74-77; Clark et al. (1989) BioTechnology 7:487-492; Gorton et al. (1987) BioTechnology 5:1183-1187; and Soulier et al. (1992) FEBS Letts. 297:13).
  • the transgenic mammals of choice would produce large volumes of milk and have long lactating periods, for example goats, cows, camels or sheep.
  • An albumin fusion protein of the invention can also be expressed in a transgenic plant, e.g. a plant in which the DNA transgene is inserted into the nuclear or plastidic genome.
  • Plant transformation procedures used to introduce foreign nucleic acids into plant cells or protoplasts are known in the art (e.g., see Example 19). See, in general, Methods in Enzymology Vol. 153 (“Recombinant DNA Part D”) 1987, Wu and Grossman Eds., Academic Press and European Patent Application EP 693554. Methods for generation of genetically engineered plants are further described in U.S. Pat. No. 5,283,184, U.S. Pat. No. 5, 482,852, and European Patent Application EP 693 554, all of which are hereby incorporated by reference.
  • the albumin fusion proteins of the invention or formulations thereof may be administered by any conventional method including parenteral (e.g. subcutaneous or intramuscular) injection or intravenous infusion.
  • the treatment may consist of a single dose or a plurality of doses over a period of time.
  • albumin fusion protein of the invention While it is possible for an albumin fusion protein of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers.
  • the carrier(s) must be “acceptable” in the sense of being compatible with the albumin fusion protein and not deleterious to the recipients thereof.
  • the carriers will be water or saline which will be sterile and pyrogen free.
  • Albumin fusion proteins of the invention are particularly well suited to formulation in aqueous carriers such as sterile pyrogen free water, saline or other isotonic solutions because of their extended shelf-life in solution.
  • pharmaceutical compositions of the invention may be formulated well in advance in aqueous form, for instance, weeks or months or longer time periods before being dispensed.
  • formulations containing the albumin fusion protein may be prepared taking into account the extended shelf-life of the albumin fusion protein in aqueous formulations. As exhibited in Table 2, most Therapeutic proteins are unstable with short shelf-lives after formulation with an aqueous carrier. As discussed above, the shelf-life of many of these Therapeutic proteins are markedly increased or prolonged after fusion to HA. TABLE 2 Storage Conditions Tradename, of Non-Fusion Protein Manufacturer Route Formulation Protein Interferon, Roferon-A, sc sol n 4-8° C.
  • the albumin fusion proteins of the invention can be formulated as aerosols using standard procedures.
  • aerosol includes any gas-borne suspended phase of an albumin fusion protein of the instant invention which is capable of being inhaled into the bronchioles or nasal passages.
  • aerosol includes a gas-borne suspension of droplets of an albumin fusion protein of the instant invention, as may be produced in a metered dose inhaler or nebulizer, or in a mist sprayer.
  • Aerosol also includes a dry powder composition of a compound of the instant invention suspended in air or other carrier gas, which may be delivered by insufflation from an inhaler device, for example.
  • the formulations of the invention are also typically non-immunogenic, in part, because of the use of the components of the albumin fusion protein being derived from the proper species.
  • both the Therapeutic protein and albumin portions of the albumin fusion protein will typically be human.
  • that component may be humanized by substitution of key amino acids so that specific epitopes appear to the human immune system to be human in nature rather than foreign.
  • the formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the albumin fusion protein with the carrier that constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation appropriate for the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents.
  • the formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules, vials or syringes, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders.
  • Dosage formulations may contain the Therapeutic protein portion at a lower molar concentration or lower dosage compared to the non-fused standard formulation for the Therapeutic protein given the extended serum half-life exhibited by many of the albumin fusion proteins of the invention.
  • an albumin fusion protein of the invention comprises growth hormone as one or more of the Therapeutic protein regions
  • the dosage form can be calculated on the basis of the potency of the albumin fusion protein relative to the potency of hGH, while taking into account the prolonged serum half-life and shelf-life of the albumin fusion proteins compared to that of native hGH.
  • Growth hormone is typically administered at 0.3 to 30.0 IU/kg/week, for example 0.9 to 12.0 IU/kg/week, given in three or seven divided doses for a year or more.
  • an equivalent dose in terms of units would represent a greater weight of agent but the dosage frequency can be reduced, for example to twice a week, once a week or less.
  • Formulations or compositions of the invention may be packaged together with, or included in a kit with, instructions or a package insert referring to the extended shelf-life of the albumin fusion protein component.
  • instructions or package inserts may address recommended storage conditions, such as time, temperature and light, taking into account the extended or prolonged shelf-life of the albumin fusion proteins of the invention.
  • Such instructions or package inserts may also address the particular advantages of the albumin fusion proteins of the inventions, such as the ease of storage for formulations that may require use in the field, outside of controlled hospital, clinic or office conditions.
  • formulations of the invention may be in aqueous form and may be stored under less than ideal circumstances without significant loss of therapeutic activity.
  • albumin fusion proteins of the invention can also be included in nutraceuticals.
  • certain albumin fusion proteins of the invention may be administered in natural products, including milk or milk product obtained from a transgenic mammal which expresses albumin fusion protein.
  • Such compositions can also include plant or plant products obtained from a transgenic plant which expresses the albumin fusion protein.
  • the albumin fusion protein can also be provided in powder or tablet form, with or without other known additives, carriers, fillers and diluents. Nutraceuticals are described in Scott Hegenhart, Food Product Design, December 1993.
  • the invention also provides methods of treatment and/or prevention of diseases or disorders (such as, for example, any one or more of the diseases or disorders disclosed herein) by administration to a subject of an effective amount of an albumin fusion protein of the invention or a polynucleotide encoding an albumin fusion protein of the invention (“albumin fusion polynucleotide”) in a pharmaceutically acceptable carrier.
  • diseases or disorders such as, for example, any one or more of the diseases or disorders disclosed herein
  • the albumin fusion protein and/or polynucleotide will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with the albumin fusion protein and/or polynucleotide alone), the site of delivery, the method of administration, the scheduling of administration, and other factors known to practitioners.
  • the “effective amount” for purposes herein is thus determined by such considerations.
  • the total pharmaceutically effective amount of the albumin fusion protein administered parenterally per dose will be in the range of about 1 ug/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone.
  • the albumin fusion protein is typically administered at a dose rate of about 1 ug/kg/hour to about 50 ug/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect.
  • Albumin fusion proteins and/or polynucleotides can be are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems.
  • sustained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray.
  • “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type.
  • parenteral refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion.
  • sustained-release albumin fusion proteins and/or polynucleotides include suitable polymeric materials (such as, for example, semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules), suitable hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, and sparingly soluble derivatives (such as, for example, a sparingly soluble salt).
  • Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al., Biopolymers 22:547-556 (1983)), poly (2-hydroxyethyl methacrylate) (Langer et al., J. Biomed. Mater. Res. 15:167-277 (1981), and Langer, Chem. Tech. 12:98-105 (1982)), ethylene vinyl acetate (Langer et al., Id.) or poly-D-( ⁇ )-3-hydroxybutyric acid (EP 133,988).
  • polylactides U.S. Pat. No. 3,773,919, EP 58,481
  • copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al.,
  • Sustained-release albumin fusion proteins and/or polynucleotides also include liposomally entrapped albumin fusion proteins and/or polynucleotides of the invention (see generally, Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 317-327 and 353-365 (1989)).
  • Liposomes containing the albumin fusion protein and/or polynucleotide are prepared by methods known per se: DE 3,218,121; Epstein et al., Proc. Natl. Acad. Sci.
  • the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal Therapeutic.
  • the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)).
  • the albumin fusion protein and/or polynucleotide is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • a pharmaceutically acceptable carrier i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation.
  • the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the Therapeutic.
  • the formulations are prepared by contacting the albumin fusion protein and/or polynucleotide uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation.
  • the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes.
  • the carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability.
  • additives such as substances that enhance isotonicity and chemical stability.
  • Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbi
  • the albumin fusion protein is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of polypeptide salts.
  • Any pharmaceutical used for therapeutic administration can be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes). Albumin fusion proteins and/or polynucleotides generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • a sterile access port for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle.
  • Albumin fusion proteins and/or polynucleotides ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution.
  • a lyophilized formulation 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous albumin fusion protein and/or polynucleotide solution, and the resulting mixture is lyophilized.
  • the infusion solution is prepared by reconstituting the lyophilized albumin fusion protein and/or polynucleotide using bacteriostatic Water-for-Injection.
  • the Albumin fusion protein formulations comprises 0.01 M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2.
  • the Albumin fusion protein formulations consists 0.01 M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2.
  • the pH and buffer are chosen to match physiological conditions and the salt is added as a tonicifier.
  • Sodium octanoate has been chosen due to its reported ability to increase the thermal stability of the protein in solution.
  • polysorbate has been added as a generic surfactant, which lowers the surface tension of the solution and lowers non-specific adsorption of the albumin fusion protein to the container closure system.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention.
  • Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • the albumin fusion proteins and/or polynucleotides may be employed in conjunction with other therapeutic compounds.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with adjuvants.
  • Adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, alum, alum plus deoxycholate (ImmunoAg), MTP-PE (Biocine Corp.), QS21 (Genentech, Inc.), BCG (e.g., THERACYS®), MPL and nonviable preparations of Corynebacterium parvum .
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with alum.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with QS-21.
  • Further adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Monophosphoryl lipid immunomodulator, AdjuVax 100a, QS-21, QS-18, CRL1005, Aluminum salts, MF-59, and Virosomal adjuvant technology.
  • Vaccines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, vaccines directed toward protection against MMR (measles, mumps, rubella), polio, varicella, tetanus/diptheria, hepatitis A, hepatitis B, Haemophilus influenzae B, whooping cough, pneumonia, influenza, Lyme's Disease, rotavirus, cholera, yellow fever, Japanese encephalitis, poliomyelitis, rabies, typhoid fever, and pertussis.
  • MMR measles, mumps, rubella
  • polio varicella
  • tetanus/diptheria hepatitis A
  • hepatitis B Haemophilus influenzae B
  • whooping cough pneumonia, influenza, Lyme's Disease, rotavirus
  • cholera yellow fever
  • Japanese encephalitis poliomyelitis
  • Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially.
  • Administration “in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with other therapeutic agents.
  • Albumin fusion protein and/or polynucleotide agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include but not limited to, chemotherapeutic agents, antibiotics, steroidal and non-steroidal anti-inflammatories, conventional immunotherapeutic agents, and/or therapeutic treatments described below. Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially.
  • Administration “in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an anticoagulant.
  • Anticoagulants that may be administered with the compositions of the invention include, but are not limited to, heparin, low molecular weight heparin, warfarin sodium (e.g., COUMADIN®), dicumarol, 4-hydroxycoumarin, anisindione (e.g., MIRADONTM), acenocoumarol (e.g., nicoumalone, SINTHROMETM), indan-1,3-dione, phenprocoumon (e.g., MARCUMARTM), ethyl biscoumacetate (e.g., TROMEXANTM), and aspirin.
  • heparin low molecular weight heparin
  • warfarin sodium e.g., COUMADIN®
  • dicumarol e.g., 4-hydroxycoumarin
  • anisindione e.g., MIRADONTM
  • compositions of the invention are administered in combination with heparin and/or warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin and aspirin. In another specific embodiment, compositions of the invention are administered in combination with heparin. In another specific embodiment, compositions of the invention are administered in combination with heparin and aspirin.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with thrombolytic drugs.
  • thrombolytic drugs that may be administered with the compositions of the invention include, but are not limited to, plasminogen, lys-plasminogen, alpha2-antiplasmin, streptokinae (e.g., KABIKINASETM), antiresplace (e.g., EMINASETM), tissue plasminogen activator (t-PA, altevase, ACTIVASETM), urokinase (e.g., ABBOKINASETM), sauruplase, (Prourokinase, single chain urokinase), and aminocaproic acid (e.g., AMICARTM).
  • compositions of the invention are administered in combination with tissue plasminogen activator and aspirin.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiplatelet drugs.
  • Antiplatelet drugs that may be administered with the compositions of the invention include, but are not limited to, aspirin, dipyridamole (e.g., PERSANTINETM), and ticlopidine (e.g., TICLIDTM).
  • the use of anti-coagulants, thrombolytic and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention, diagnosis, and/or treatment of thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina.
  • the use of anticoagulants, thrombolytic drugs and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention of occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease.
  • occlusions in extracorporeal devices e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiretroviral agents, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and/or protease inhibitors (PIs).
  • NRTIs nucleoside/nucleotide reverse transcriptase inhibitors
  • NRTIs non-nucleoside reverse transcriptase inhibitors
  • PIs protease inhibitors
  • NRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, RETROVIRTM (zidovudine/AZT), VIDEXTM (didanosine/ddI), HVIDTM (zalcitabine/ddC), ZERITTM (stavudine/d4T), EPIVIRTM (lamivudine/3TC), and COMBIVIRTM (zidovudine/lamivudine).
  • RETROVIRTM zidovudine/AZT
  • VIDEXTM didanosine/ddI
  • HVIDTM zalcitabine/ddC
  • ZERITTM stavudine/d4T
  • EPIVIRTM lamvudine/3TC
  • COMBIVIRTM zidovudine/lamivudine
  • NNRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, VIRAMUNETM (nevirapine), RESCRIPTORTM (delavirdine), and SUSTIVATM (efavirenz).
  • Protease inhibitors that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, CRIXIVANTM (indinavir), NORVIRTM (ritonavir), INVRASETM (saquinavir), and VIRACEPTTM (nelfinavir).
  • antiretroviral agents nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, and/or protease inhibitors may be used in any combination with albumin fusion proteins and/or polynucleotides of the invention to treat AIDS and/or to prevent or treat HIV infection.
  • Additional NRTIs include LODENOSINETM (F-ddA; an acid-stable adenosine NRTI; Triangle/Abbott; COVIRACILTM (emtricitabine/FTC; structurally related to lamivudine (3TC) but with 3- to 10-fold greater activity in vitro; Triangle/Abbott); dOTC (BCH-10652, also structurally related to lamivudine but retains activity against a substantial proportion of lamivudine-resistant isolates; Biochem Pharma); Adefovir (refused approval for anti-HIV therapy by FDA; Gilead Sciences); PREVEON® (Adefovir Dipivoxil, the active prodrug of adefovir; its active form is PMEA-pp); TENOFOVIRTM (bis-POC PMPA, a PMPA prodrug; Gilead); DAPD/DXG (active metabolite of DAPD; Triangle/Abbot
  • Additional NNRTIs include COACTINONTM (Emivirine/MKC-442, potent NNRTI of the HEPT class; Triangle/Abbott); CAPRAVIRINETM (AG-1549/S-1153, a next generation NNRTI with activity against viruses containing the K103N mutation; Agouron); PNU-142721 (has 20- to 50-fold greater activity than its predecessor delavirdine and is active against K103N mutants; Pharmacia & Upjohn); DPC-961 and DPC-963 (second-generation derivatives of efavirenz, designed to be active against viruses with the K103N mutation; DuPont); GW-420867 ⁇ (has 25-fold greater activity than HBY097 and is active against K103N mutants; Glaxo Wellcome); CALANOLIDE A (naturally occurring agent from the latex tree; active against viruses containing either or both the Y181C and K103N mutations); and Propolis (WO 99/49830).
  • COACTINONTM Esmivirine/MKC
  • protease inhibitors include LOPINAVIRTM (ABT378/r; Abbott Laboratories); BMS-232632 (an azapeptide; Bristol-Myres Squibb); TIPRANAVIRTM (PNU-140690, a non-peptic dihydropyrone; Pharmacia & Upjohn); PD-178390 (a nonpeptidic dihydropyrone; Parke-Davis); BMS 232632 (an azapeptide; Bristol-Myers Squibb); L-756,423 (an indinavir analog; Merck); DMP-450 (a cyclic urea compound; Avid & DuPont); AG-1776 (a peptidomimetic with in vitro activity against protease inhibitor-resistant viruses; Agouron); VX-175/GW-433908 (phosphate prodrug of amprenavir; Vertex & Glaxo Welcome); CGP61755 (Ciba); and AGENERASETM (amprenavir; Glaxo
  • Additional antiretroviral agents include fusion inhibitors/gp41 binders.
  • Fusion inhibitors/gp41 binders include T-20 (a peptide from residues 643-678 of the HIV gp41 transmembrane protein ectodomain which binds to gp41 in its resting state and prevents transformation to the fusogenic state; Trimeris) and T-1249 (a second-generation fusion inhibitor; Trimeris).
  • Additional antiretroviral agents include fusion inhibitors/chemokine receptor antagonists.
  • Fusion inhibitors/chemokine receptor antagonists include CXCR4 antagonists such as AMD 3100 (a bicyclam), SDF-1 and its analogs, and ALX40-4C (a cationic peptide), T22 (an 18 amino acid peptide; Trimeris) and the T22 analogs T134 and T140; CCR5 antagonists such as RANTES (9-68), AOP-RANTES, NNY-RANTES, and TAK-779; and CCR5/CXCR4 antagonists such as NSC 651016 (a distamycin analog). Also included are CCR2B, CCR3, and CCR6 antagonists. Chemokine recpetor agonists such as RANTES, SDF-1, MIP-1 ⁇ , MIP-1 ⁇ , etc., may also inhibit fusion.
  • Additional antiretroviral agents include integrase inhibitors.
  • Integrase inhibitors include dicaffeoylquinic (DFQA) acids; L-chicoric acid (a dicaffeoyltartaric (DCTA) acid); quinalizarin (QLC) and related anthraquinones; ZINTEVIRTM (AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase inhibitor; Arondex); and naphthols such as those disclosed in WO 98/50347.
  • DFQA dicaffeoylquinic
  • DCTA dicaffeoyltartaric
  • QLC quinalizarin
  • ZINTEVIRTM AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase inhibitor
  • Arondex naphthols such as those disclosed in WO 98/50347.
  • Additional antiretroviral agents include hydroxyurea-like compunds such as BCX-34 (a purine nucleoside phosphorylase inhibitor; Biocryst); ribonucleotide reductase inhibitors such as DIDOXTM (Molecules for Health); inosine monophosphate dehydrogenase (IMPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCept (mycophenolate mofetil; Roche).
  • BCX-34 purine nucleoside phosphorylase inhibitor
  • Biocryst ribonucleotide reductase inhibitors
  • DIDOXTM Diotide reductase inhibitor
  • IMPDH inosine monophosphate dehydrogenase
  • VX-497 Verytex
  • mycopholic acids such as CellCept (mycophenolate mofetil; Roche).
  • Additional antiretroviral agents include inhibitors of viral integrase, inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds; inhibitors of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100; nucleocapsid zinc finger inhibitors such as dithiane compounds; targets of HIV Tat and Rev; and pharmacoenhancers such as ABT-378.
  • inhibitors of viral integrase inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds
  • inhibitors of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100
  • nucleocapsid zinc finger inhibitors such as dithiane compounds
  • cytokines and lymphokines such as MIP-1 ⁇ , MIP-1 ⁇ , SDF-1 ⁇ , IL-2, PROLEUKINTM (aldesleukin/L2-7001; Chiron), 1L-4, IL-10, IL-12, and IL-13; interferons such as IFN- ⁇ 2a; antagonists of TNFs, NF ⁇ B, GM-CSF, M-CSF, and IL-10; agents that modulate immune activation such as cyclosporin and prednisone; vaccines such as RemuneTM (HIV Immunogen), APL 400-003 (Apollon), recombinant gp120 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgp120CM235, MN rgp120, SF-2 rgp120, gp120/soluble CD4 complex, Delta JR-FL protein, branched synthetic peptide derived from discontinuous gp120 C
  • antibodies such as the anti-CXCR4 antibody 12G5, the anti-CCR5 antibodies 2D7, 5C7, PA8, PA9, PA10, PA11, PA12, and PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7B11, the anti-gp120 antibodies 17b, 48d, 447-52D, 257-D, 268-D and 50.1, anti-Tat antibodies, anti-TNF- ⁇ antibodies, and monoclonal antibody 33A; aryl hydrocarbon (AH) receptor agonists and antagonists such as TCDD, 3,3′,4,4′,5-pentachlorobiphenyl, 3,3′,4,4′-tetrachlorobiphenyl, and ⁇ -naphthoflavone (WO 98/30213); and antioxidants such as ⁇ -L-glutamyl-L-cysteine ethyl ester ( ⁇ -GCE; WO 99/56764
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiviral agent.
  • Antiviral agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, acyclovir, ribavirin, amantadine, and remantidine.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered in combination with anti-opportunistic infection agents.
  • Anti-opportunistic agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, TRIMETHOPRIM-SULFAMETHOXAZOLETM, DAPSONETM, PENTAMIDNETM, ATOVAQUONETM, ISONIAZIDTM, RIFAMPINTM, PYRAZINAMIDETM, ETHAMBUTOLTM, RIFABUTINTM, CLARITHROMYCINTM, AZITHROMYCINTM, GANCICLOVIRTM, FOSCARNETTM, CIDOFOVIRTM, FLUCONAZOLETM, ITRACONAZOLETM, KETOCONAZOLETM, ACYCLOVIRTM, FAMCICOLVIRTM, PYRIMETHAMINETM, LEUCOVORINTM, NEU
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with TRIMETHOPRIM-SULFAMETHOXAZOLETM, DAPSONETM, PENTAMIDINETM, and/or ATOVAQUONETM to prophylactically treat or prevent an opportunistic Pneumocystis carinii pneumonia infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ISONIAZIDTM, RIFAMPINTM, PYRAZINAMIDETM, and/or ETHAMBUTOLTM to prophylactically treat or prevent an opportunistic Mycobacterium avium complex infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with RIFABUTINTM, CLARITHROMYCINTM, and/or AZITHROMYCINTM to prophylactically treat or prevent an opportunistic Mycobacterium tuberculosis infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with GANCICLOVIRTM, FOSCARNETTM, and/or CIDOFOVIRTM to prophylactically treat or prevent an opportunistic cytomegalovirus infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with FLUCONAZOLETM, ITRACONAZOLETM, and/or KETOCONAZOLETM to prophylactically treat or prevent an opportunistic fungal infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ACYCLOVIRTM and/or FAMCICOLVIRTM to prophylactically treat or prevent an opportunistic herpes simplex virus type I and/or type II infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with PYRIMETHAMINETM and/or LEUCOVORINTM to prophylactically treat or prevent an opportunistic Toxoplasma gondii infection.
  • albumin fusion proteins and/or polynucleotides of the invention are used in any combination with LEUCOVORINTM and/or NEUPOGENTM to prophylactically treat or prevent an opportunistic bacterial infection.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antibiotic agent.
  • Antibiotic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, amoxicillin, beta-lactamases, aminoglycosides, beta-lactam (glycopeptide), beta-lactamases, Clindamycin, chloramphenicol, cephalosporins, ciprofloxacin, erythromycin, fluoroquinolones, macrolides, metronidazole, penicillins, quinolones, rapamycin, rifampin, streptomycin, sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamethoxazole, and vancomycin.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunostimulants.
  • Immunostimulants that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, levamisole (e.g., ERGAMISOLTM), isoprinosine (e.g. INOSIPLEXTM), interferons (e.g. interferon alpha), and interleukins (e.g., IL-2).
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunosuppressive agents.
  • Immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, steroids, cyclosporine, cyclosporine analogs, cyclophosphamide methylprednisone, prednisone, azathioprine, FK-506, 15-deoxyspergualin, and other immunosuppressive agents that act by suppressing the function of responding T cells.
  • immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, prednisolone, methotrexate, thalidomide, methoxsalen, rapamycin, leflunomide, mizoribine (BREDININTM), brequinar, deoxyspergualin, and azaspirane (SKF 105685), ORTHOCLONE OKT® 3 (muromonab-CD3), SANDIMMUNETM, NEORALTM, SANGDYATM (cyclosporine), PROGRAF® (FK506, tacrolimus), CELLCEPT® (mycophenolate motefil, of which the active metabolite is mycophenolic acid), IMURANTM (azathioprine), glucocorticosteroids, adrenocortical steroids such as DELTASONETM (prednisone) and HYDELTRASOLTM (predni
  • albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with one or more intravenous immune globulin preparations.
  • Intravenous immune globulin preparations that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but not limited to, GAMMARTM, IVEEGAMTM, SANDOGLOBULINTM, GAMMAGARD S/DTM, ATGAMTM (antithymocyte glubulin), and GAMIMUNETM.
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with intravenous immune globulin preparations in transplantation therapy (e.g., bone marrow transplant).
  • the albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with an anti-inflammatory agent.
  • Anti-inflammatory agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, corticosteroids (e.g.
  • compositions of the invention are administered alone or in combination with an anti-angiogenic agent.
  • Anti-angiogenic agents that may be administered with the compositions of the invention include, but are not limited to, Angiostatin (Entremed, Rockville, Md.), Troponin-1 (Boston Life Sciences, Boston, Mass.), anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel (Taxol), Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter “d group” transition metals.
  • Lighter “d group” transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition metal complexes.
  • vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes.
  • Suitable vanadate complexes include metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodium metavanadate, and sodium orthovanadate.
  • Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates.
  • tungsten and molybdenum complexes also include oxo complexes.
  • Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes.
  • Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid.
  • Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide.
  • Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes.
  • Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates.
  • Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid.
  • Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate.
  • Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars.
  • anti-angiogenic factors include, but are not limited to, platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res.
  • SP-PG Sulphated Polysaccharide Peptidoglycan Complex
  • steroids such as estrogen, and tamoxifen citrate
  • Staurosporine modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloffet al., J.
  • Thalidomide (Celgene, Warren, N.J.); Angiostatic steroid; AGM-1470 (H. Brem and J. Folkman J Pediatr. Surg. 28:445-51 (1993)); an integrin alpha v beta 3 antagonist (C. Storgard et al., J Clin. Invest.
  • Anti-angiogenic agents that may be administed in combination with the compounds of the invention may work through a variety of mechanisms including, but not limited to, inhibiting proteolysis of the extracellular matrix, blocking the function of endothelial cell-extracellular matrix adhesion molecules, by antagonizing the function of angiogenesis inducers such as growth factors, and inhibiting integrin receptors expressed on proliferating endothelial cells.
  • anti-angiogenic inhibitors that interfere with extracellular matrix proteolysis and which may be administered in combination with the compositons of the invention include, but are not lmited to, AG-3340 (Agouron, La Jolla, Calif.), BAY-12-9566 (Bayer, West Haven, Conn.), BMS-275291 (Bristol Myers Squibb, Princeton, N.J.), CGS-27032A (Novartis, East Hanover, N.J.), Marimastat (British Biotech, Oxford, UK), and Metastat (Aeterna, St-Foy, Quebec).
  • anti-angiogenic inhibitors that act by blocking the function of endothelial cell-extracellular matrix adhesion molecules and which may be administered in combination with the compositons of the invention include, but are not lmited to, EMD-121974 (Merck KcgaA Darmstadt, Germany) and Vitaxin (Ixsys, La Jolla, Calif./Medimmune, Gaithersburg, Md.).
  • anti-angiogenic agents that act by directly antagonizing or inhibiting angiogenesis inducers and which may be administered in combination with the compositons of the invention include, but are not lmited to, Angiozyme (Ribozyme, Boulder, Colo.), Anti-VEGF antibody (Genentech, S.
  • SU-101 S. San Francisco, Calif.
  • SU-5416 Sugen/Pharmacia Upjohn, Bridgewater, N.J.
  • SU-6668 Sugen.
  • Other anti-angiogenic agents act to indirectly inhibit angiogenesis.
  • indirect inhibitors of angiogenesis which may be administered in combination with the compositons of the invention include, but are not limited to, IM-862 (Cytran, Kirkland, Wash.), Interferon-alpha, IL-12 (Roche, Nutley, N.J.), and Pentosan polysulfate (Georgetown University, Washington, D.C.).
  • compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of an autoimmune disease, such as for example, an autoimmune disease described herein.
  • compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of arthritis.
  • use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of rheumatoid arthritis.
  • the polynucleotides encoding a polypeptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein.
  • angiogenic proteins include, but are not limited to, acidic and basic fibroblast growth factors, VEGF-1, VEGF-2, VEGF-3, epidermal growth factor alpha and beta, platelet-derived endothelial cell growth factor, platelet-derived growth factor, tumor necrosis factor alpha, hepatocyte growth factor, insulin-like growth factor, colony stimulating factor, macrophage colony stimulating factor, granulocyte/macrophage colony stimulating factor, and nitric oxide synthase.
  • compositions of the invention are administered in combination with a chemotherapeutic agent.
  • Chemotherapeutic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to alkylating agents such as nitrogen mustards (for example, Mechlorethamine, cyclophosphamide, Cyclophosphamide Ifosfamide, Melphalan (L-sarcolysin), and Chlorambucil), ethylenimines and methylmelamines (for example, Hexamethylmelamine and Thiotepa), alkyl sulfonates (for example, Busulfan), nitrosoureas (for example, Carmustine (BCNU), Lomustine (CCNU), Semustine (methyl-CCNU), and Streptozocin (streptozotocin)), triazenes (for example, dacarbazine (DTIC; dimethyltriazenoimidazo
  • compositions of the invention are administered in combination with one or more of the following drugs: infliximab (also known as RemicadeTM Centocor, Inc.), Trocade (Roche, RO-32-3555), Leflunomide (also known as AravaTM from Hoechst Marion Roussel), KineretTM (an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.)
  • infliximab also known as RemicadeTM Centocor, Inc.
  • Trocade Roche, RO-32-3555
  • Leflunomide also known as AravaTM from Hoechst Marion Roussel
  • KineretTM an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.
  • compositions of the invention are administered in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) or combination of one or more of the components of CHOP.
  • CHOP cyclophosphamide, doxorubicin, vincristine, and prednisone
  • the compositions of the invention are administered in combination with anti-CD20 antibodies, human monoclonal anti-CD20 antibodies.
  • the compositions of the invention are administered in combination with anti-CD20 antibodies and CHOP, or anti-CD20 antibodies and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • compositions of the invention are administered in combination with Rituximab.
  • compositions of the invention are administered with Rituximab and CHOP, or Rituximab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • compositions of the invention are administered in combination with tositumomab.
  • compositions of the invention are administered with tositumomab and CHOP, or tositumomab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • the anti-CD20 antibodies may optionally be associated with radioisotopes, toxins or cytotoxic prodrugs.
  • compositions of the invention are administered in combination ZevalinTM.
  • compositions of the invention are administered with ZevalinTM and CHOP, or ZevalinTM and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone.
  • ZevalinTM may be associated with one or more radisotopes. Particularly preferred isotopes are 90 Y and 111 In.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with cytokines.
  • Cytokines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, IL2, IL3, IL4, IL5, IL6, IL7, IL10, IL12, IL13, IL15, anti-CD40, CD40L, IFN-gamma and TNF-alpha.
  • albumin fusion proteins and/or polynucleotides of the invention may be administered with any interleukin, including, but not limited to, IL-1alpha, IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21.
  • interleukin including, but not limited to, IL-1alpha, IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with members of the TNF family.
  • TNF, TNF-related or TNF-like molecules that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, soluble forms of TNF-alpha, lymphotoxin-alpha (LT-alpha, also known as TNF-beta), LT-beta (found in complex heterotrimer LT-alpha2-beta), OPGL, FasL, CD27L, CD30L, CD40L, 4-1BBL, DcR3, OX40L, TNF-gamma (International Publication No.
  • WO 96/14328 AIM-I (International Publication No. WO 97/33899), endokine-alpha (International Publication No. WO 98/07880), OPG, and netitrokine-alpha (International Publication No. WO 98/18921, OX40, and nerve growth factor (NGF), and soluble forms of Fas, CD30, CD27, CD40 and 4-IBB, TR2 (International Publication No. WO 96/34095), DR3 (International Publication No. WO 97/33904), DR4 (International Publication No. WO 98/32856), TR5 (International Publication No. WO 98/30693), TRANK, TR9 (International Publication No. WO 98/56892),TR10 (International Publication No. WO 98/54202), 312C2 (International Publication No. WO 98/06842), and TR12, and soluble forms CD154, CD70, and CD153.
  • TR2 International Publication No.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with angiogenic proteins.
  • Angiogenic proteins that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Glioma Derived Growth Factor (GDGF), as disclosed in European Patent Number EP-399816; Platelet Derived Growth Factor-A (PDGF-A), as disclosed in European Patent Number EP-682110; Platelet Derived Growth Factor-B (PDGF-B), as disclosed in European Patent Number EP-282317; Placental Growth Factor (PIGF), as disclosed in International Publication Number WO 92/06194; Placental Growth Factor-2 (PIGF-2), as disclosed in Hauser et al., Growth Factors, 4:259-268 (1993); Vascular Endothelial Growth Factor (VEGF), as disclosed in International Publication Number WO 90/13649; Vascular Endothelial Growth Factor (
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with Fibroblast Growth Factors.
  • Fibroblast Growth Factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, FGF-1, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, and FGF-15.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with hematopoietic growth factors.
  • Hematopoietic growth factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, granulocyte macrophage colony stimulating factor (GM-CSF) (sargramostim, LEUKINETM, PROKINETM), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGENTM), macrophage colony stimulating factor (M-CSF, CSF-1) erythropoietin (epoetin alfa, EPOGENTM, PROCRITTM), stem cell factor (SCF, c-kit ligand, steel factor), megakaryocyte colony stimulating factor, PIXY321 (a GMCSF/IL-3 fusion protein), interleukins, especially any one or more of IL-1 through IL-12,
  • albumin fusion proteins and/or polynucleotides of the present invention are administered in combination with adrenergic blockers, such as, for example, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol.
  • adrenergic blockers such as, for example, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiarrhythmic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainamide, N-acetyl procainamide, propafenone, propranolol, quinidine, sotalol, tocainide, and verapamil).
  • an antiarrhythmic drug e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainamide,
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with diuretic agents, such as carbonic anhydrase-inhibiting agents (e.g., acetazolamide, dichlorphenamide, and methazolamide), osmotic diuretics (e.g., glycerin, isosorbide, mannitol, and urea), diuretics that inhibit Na + -K + -2Cl ⁇ symport (e.g., furosemide, bumetamide, azosemide, piretamide, tripamide, ethacrynic acid, muzolimine, and torsemide), thiazide and thiazide-like diuretics (e.g., bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazi
  • diuretic agents such
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for endocrine and/or hormone imbalance disorders.
  • Treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, 127 I, radioactive isotopes of iodine such as 131 I and 123 I; recombinant growth hormone, such as HUMATROPETM (recombinant somatropin); growth hormone analogs such as PROTROPINTM (somatrem); dopamine agonists such as PARLODELTM (bromocriptine); somatostatin analogs such as SANDOSTATINTM (octreotide); gonadotropin preparations such as PREGNYLTM, A.P.L.TM and PROFASITM (chorionic gonadotropin (CG)), PERGONALTM (menotropins), and METRODINTM (urofollitropin (uFSH)); synthetic human gonadotropin
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, estrogens or congugated estrogens such as ESTRACETM (estradiol), ESTINYLTM (ethinyl estradiol), PREMARINTM, ESTRATABTM, ORTHO-ESTTM, OGENTM and estropipate (estrone), ESTROVISTM (quinestrol), ESTRADERMTM (estradiol), DELESTROGENTM and VALERGENTM (estradiol valerate), DEPO-ESTRADIOL CYPIONATETM and ESTROJECT LATM (estradiol cypionate); antiestrogens such as NOLVADEXTM (tamoxifen), SEROPHENETM and CLOMIDTM (clomiphene); progestins such as DURALUTINTM (hydroxyprogesterone caproate), MPATM and DEPO-PROVERATM (medroxyprogesterone acetate), PROVERATM and
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, testosterone esters such as methenolone acetate and testosterone undecanoate; parenteral and oral androgens such as TESTOJECT-50TM (testosterone), TESTEXTM (testosterone propionate), DELATESTRYLTM (testosterone enanthate), DEPO-TESTOSTERONETM (testosterone cypionate), DANOCRINETM (danazol), HALOTESTINTM (fluoxymesterone), ORETON METHYLTM, TESTREDTM and VIRILONTM (methyltestosterone), and OXANDRINTM (oxandrolone); testosterone transdermal systems such as TESTODERMTM; androgen receptor antagonist and 5-alpha-reductase inhibitors such as ANDROCURTM (cyproterone acetate), EULEXNTM (flutamide), and PROSCARTM (finasteride); a
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for uterine motility disorders.
  • Treatments for uterine motility disorders include, but are not limited to, estrogen drugs such as conjugated estrogens (e.g., PREMARIN® and ESTRATAB®), estradiols (e.g., CLIMARA® and ALORA®), estropipate, and chlorotrianisene; progestin drugs (e.g., AMEN® (medroxyprogesterone), MICRONOR® (norethidrone acetate), PROMETRIUM® progesterone, and megestrol acetate); and estrogen/progesterone combination therapies such as, for example, conjugated estrogens/medroxyprogesterone (e.g., PREMPROTM and PREMPHASE®) and norethindrone acetate/ethinyl estsradiol (e.g., FEMHRTTM).
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with drugs effective in treating iron deficiency and hypochromic anemias, including but not limited to, ferrous sulfate (iron sulfate, FEOSOLTM), ferrous fumarate (e.g., FEOSTATTM), ferrous gluconate (e.g., FERGONTM), polysaccharide-iron complex (e.g., NIFEREXTM), iron dextran injection (e.g., INFEDTM), cupric sulfate, pyroxidine, riboflavin, Vitamin B 12 , cyancobalamin injection (e.g., REDISOLTM, RUBRAMIN PCTM), hydroxocobalamin, folic acid (e.g., FOLVITETM), leucovorin (folinic acid, 5-CHOH4PteGlu, citrovorum factor) or WELLCOVORIN (Calcium
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat psychiatric disorders.
  • Psychiatric drugs that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antipsychotic agents (e.g., chlorpromazine, chlorprothixene, clozapine, fluphenazine, haloperidol, loxapine, mesoridazine, molindone, olanzapine, perphenazine, pimozide, quetiapine, risperidone, thioridazine, thiothixene, trifluoperazine, and triflupromazine), antimanic agents (e.g., carbamazepine, divalproex sodium, lithium carbonate, and lithium citrate), antidepressants (e.g., amitriptyline, amoxa
  • antipsychotic agents
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat neurological disorders.
  • Neurological agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antiepileptic agents (e.g., carbamazepine, clonazepam, ethosuximide, phenobarbital, phenytoin, primidone, valproic acid, divalproex sodium, felbamate, gabapentin, lamotrigine, levetiracetam, oxcarbazepine, tiagabine, topiramate, zonisamide, diazepam, lorazepam, and clonazepam), antiparkinsonian agents (e.g., levodopa/carbidopa, selegiline, amantidine, bromocriptine, pergolide, ropinirole
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with vasodilating agents and/or calcium channel blocking agents.
  • Vasodilating agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Angiotensin Converting Enzyme (ACE) inhibitors (e.g., papaverine, isoxsuprine, benazepril, captopril, cilazapril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, spirapril, trandolapril, and nylidrin), and nitrates (e.g., isosorbide dinitrate, isosorbide mononitrate, and nitroglycerin).
  • ACE Angiotensin Con
  • calcium channel blocking agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to amlodipine, bepridil, diltiazem, felodipine, flunarizine, isradipine, nicardipine, nifedipine, nimodipine, and verapamil.
  • the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for gastrointestinal disorders.
  • Treatments for gastrointestinal disorders that may be administered with the albumin fusion protein and/or polynucleotide of the invention include, but are not limited to, H 2 histamine receptor antagonists (e.g., TAGAMETTM (cimetidine), ZANTACTM (ranitidine), PEPCIDTM (famotidine), and AXIDTM (nizatidine)); inhibitors of H + , K + ATPase (e.g., PREVACIDTM (lansoprazole) and PRILOSECTM (omeprazole)); Bismuth compounds (e.g., PEPTO-BISMOLTM (bismuth subsalicylate) and DE-NOLTM (bismuth subcitrate)); various antacids; sucralfate; prostaglandin analogs (e.g.
  • H 2 histamine receptor antagonists e.g.,
  • CYTOTECTM miprostol
  • muscarinic cholinergic antagonists e.g., laxatives (e.g., surfactant laxatives, stimulant laxatives, saline and osmotic laxatives); antidiarrheal agents (e.g., LOMOTILTM (diphenoxylate), MOTOFENTM (diphenoxin), and IMODIUMTM (loperamide hydrochloride)), synthetic analogs of somatostatin such as SANDOSTATINTM (octreotide), antiemetic agents (e.g., ZOFRANTM (ondansetron), KYTRILTM (granisetron hydrochloride), tropisetron, dolasetron, metoclopramide, chlorpromazine, perphenazine, prochlorperazine, promethazine, thiethylperazine, triflupromazine, domperidone, haloperidol, drop
  • albumin fusion proteins and/or polynucleotides of the invention are administered in combination with other therapeutic or prophylactic regimens, such as, for example, radiation therapy.
  • the invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising albumin fusion proteins of the invention.
  • a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising albumin fusion proteins of the invention.
  • Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration.
  • Constructs encoding albumin fusion proteins of the invention can be used as a part of a gene therapy protocol to deliver therapeutically effective doses of the albumin fusion protein.
  • a preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, encoding an albumin fusion protein of the invention. Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid.
  • Retrovirus vectors and adeno-associated virus vectors can be used as a recombinant gene delivery system for the transfer of exogenous nucleic acid molecules encoding albumin fusion proteins in vivo. These vectors provide efficient delivery of nucleic acids into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host.
  • the development of specialized cell lines (termed “packaging cells”) which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A. D. (1990) Blood 76:271).
  • a replication defective retrovirus can be packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al., (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
  • Another viral gene delivery system useful in the present invention uses adenovirus-derived vectors.
  • the genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See, for example, Berkner et al., BioTechniques 6:616 (1988); Rosenfeld et al., Science 252:431-434 (1991); and Rosenfeld et al., Cell 68:143-155 (1992).
  • adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus are known to those skilled in the art.
  • Recombinant adenoviruses can be advantageous in certain circumstances in that they are not capable of infecting nondividing cells and can be used to infect a wide variety of cell types, including epithelial cells (Rosenfeld et al., (1992) cited supra).
  • the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity.
  • introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA).
  • the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al., cited supra; Haj-Ahmand et al., J. Virol. 57:267 (1986)).
  • non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject nucleotide molecule by the targeted cell.
  • exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes.
  • a nucleic acid molecule encoding an albumin fusion protein of the invention can be entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et al. (1992) No Shinkei Geka 20:547-551; PCT publication WO91/06309; Japanese patent application 1047381; and European patent publication EP-A-43075).
  • Gene delivery systems for a gene encoding an albumin fusion protein of the invention can be introduced into a patient by any of a number of methods.
  • a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof.
  • initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized.
  • the gene delivery vehicle can be introduced by catheter (see U.S. Pat. No. 5,328,470) or by Stereotactic injection (e.g.
  • the pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded.
  • the albumin fusion protein can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the albumin fusion protein.
  • the gene therapy methods relate to the introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal to achieve expression of an albumin fusion protein of the invention.
  • This method requires a polynucleotide which codes for an albumin fusion protein of the present invention operatively linked to a promoter and any other genetic elements necessary for the expression of the fusion protein by the target tissue.
  • Such gene therapy and delivery techniques are known in the art, see, for example, WO90/11092, which is herein incorporated by reference.
  • cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising a promoter operably linked to a polynucleotide encoding an albumin fusion protein of the present invention ex vivo, with the engineered cells then being provided to a patient to be treated with the fusion protein of the present invention.
  • a polynucleotide DNA or RNA
  • Such methods are well-known in the art. For example, see Belldegrun, A., et al., J. Natl. Cancer Inst. 85: 207-216 (1993); Ferrantini, M. et al., Cancer Research 53: 1107-1112 (1993); Ferrantini, M. et al., J.
  • the cells which are engineered are arterial cells.
  • the arterial cells may be reintroduced into the patient through direct injection to the artery, the tissues surrounding the artery, or through catheter injection.
  • the polynucleotide constructs can be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, and the like).
  • the polynucleotide constructs may be delivered in a pharmaceutically acceptable liquid or aqueous carrier.
  • polynucleotides encoding the albumin fusion proteins of the present invention is delivered as a naked polynucleotide.
  • naked polynucleotide, DNA or RNA refers to sequences that are free from any delivery vehicle that acts to assist, promote or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like.
  • polynucleotides encoding the albumin fusion proteins of the present invention can also be delivered in liposome formulations and lipofectin formulations and the like can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in U.S. Pat. Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference.
  • the polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication.
  • Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL available from Pharmacia; and pEF1/V5, pcDNA3.1, and pRc/CMV2 available from Invitrogen.
  • Other suitable vectors will be readily apparent to the skilled artisan.
  • Suitable promoters include adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs; the b-actin promoter; and human growth hormone promoters.
  • the promoter also may be the native promoter for the gene corresponding to the Therapeutic protein portion of the albumin fusion proteins of the invention.
  • one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months.
  • the polynucleotide construct can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue.
  • Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone.
  • the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides.
  • an effective dosage amount of DNA or RNA will be in the range of from about 0.05 mg/kg body weight to about 50 mg/kg body weight.
  • the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg.
  • this dosage will vary according to the tissue site of injection.
  • the appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration.
  • the preferred route of administration is by the parenteral route of injection into the interstitial space of tissues.
  • parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose.
  • naked DNA constructs can be delivered to arteries during angioplasty by the catheter used in the procedure.
  • the naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, and so-called “gene guns”. These delivery methods are known in the art.
  • constructs may also be delivered with delivery vehicles such as viral sequences, viral particles, liposome formulations, lipofectin, precipitating agents, etc. Such methods of delivery are known in the art.
  • the polynucleotide constructs are complexed in a liposome preparation.
  • Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations.
  • cationic liposomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic nucleic acid.
  • Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al., Proc. Natl. Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference); mRNA (Malone et al., Proc. Natl.
  • Cationic liposomes are readily available.
  • N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N.Y. (See, also, Felgner et al., Proc. Natl. Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference).
  • Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehringer).
  • cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO 90/11092 (which is herein incorporated by reference) for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes. Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Felgner et al., Proc. Natl. Acad. Sci. USA 84:7413-7417, which is herein incorporated by reference. Similar methods can be used to prepare liposomes from other cationic lipid materials.
  • anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials.
  • Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others.
  • DOPC dioleoylphosphatidyl choline
  • DOPG dioleoylphosphatidyl glycerol
  • DOPE dioleoylphoshatidyl ethanolamine
  • DOPC dioleoylphosphatidyl choline
  • DOPG dioleoylphosphatidyl glycerol
  • DOPE dioleoylphosphatidyl ethanolamine
  • DOPG/DOPC vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water.
  • the sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15EC.
  • negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size.
  • Other methods are known and available to those of skill in the art.
  • the liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred.
  • MLVs multilamellar vesicles
  • SUVs small unilamellar vesicles
  • LUVs large unilamellar vesicles
  • the various liposome-nucleic acid complexes are prepared using methods well known in the art. See, e.g., Straubinger et al., Methods of Immunology (1983), 101:512-527, which is herein incorporated by reference.
  • MLVs containing nucleic acid can be prepared by depositing a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to be encapsulated.
  • SUVs are prepared by extended sonication of MLVs to produce a homogeneous population of unilamellar liposomes.
  • the material to be entrapped is added to a suspension of preformed MLVs and then sonicated.
  • liposomes containing cationic lipids the dried lipid film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCl, sonicated, and then the preformed liposomes are mixed directly with the DNA.
  • the liposome and DNA form a very stable complex due to binding of the positively charged liposomes to the cationic DNA.
  • SUVs find use with small nucleic acid fragments.
  • LUVs are prepared by a number of methods, well known in the art. Commonly used methods include Ca 2+ -EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta (1975) 394:483; Wilson et al., Cell 17:77 (1979)); ether injection (Deamer, D. and Bangham, A., Biochim. Biophys. Acta 443:629 (1976); Ostro et al., Biochem. Biophys. Res. Commun. 76:836 (1977); Fraley et al., Proc. Natl. Acad. Sci. USA 76:3348 (1979)); detergent dialysis (Enoch, H.
  • the ratio of DNA to liposomes will be from about 10:1 to about 1:10.
  • the ration will be from about 5:1 to about 1:5. More preferably, the ration will be about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1.
  • U.S. Pat. No. 5,676,954 reports on the injection of genetic material, complexed with cationic liposomes carriers, into mice.
  • WO 94/9469 (which are herein incorporated by reference) provide cationic lipids for use in transfecting DNA into cells and mammals.
  • WO 94/9469 provide methods for delivering DNA-cationic lipid complexes to mammals.
  • cells are engineered, ex vivo or in vivo, using a retroviral particle containing RNA which comprises a sequence encoding an albumin fusion protein of the present invention.
  • Retroviruses from which the retroviral plasmid vectors may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus.
  • the retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines.
  • packaging cells which may be transfected include, but are not limited to, the PE501, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE, RCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, Human Gene Therapy 1:5-14 (1990), which is incorporated herein by reference in its entirety.
  • the vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO 4 precipitation.
  • the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • the producer cell line generates infectious retroviral vector particles which include polynucleotide encoding an albumin fusion protein of the present invention. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express a fusion protin of the present invention.
  • cells are engineered, ex vivo or in vivo, with polynucleotide contained in an adenovirus vector.
  • Adenovirus can be manipulated such that it encodes and expresses fusion protein of the present invention, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis.
  • adenoviruses have been used as live enteric vaccines for many years with an excellent safety profile (Schwartz et al. Am. Rev. Respir. Dis. 109:233-238 (1974)).
  • adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1-antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld, M. A. et al. (1991) Science 252:431-434; Rosenfeld et al., (1992) Cell 68:143-155). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green, M. et al. (1979) Proc. Natl. Acad. Sci. USA 76:6606).
  • Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel. 3:499-503 (1993); Rosenfeld et al., Cell 68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993); Yang et al., Nature Genet. 7:362-369 (1994); Wilson et al., Nature 365:691-692 (1993); and U.S. Pat. No. 5,652,224, which are herein incorporated by reference.
  • the adenovirus vector Ad2 is useful and can be grown in human 293 cells.
  • These cells contain the E1 region of adenovirus and constitutively express E1a and E1b, which complement the defective adenoviruses by providing the products of the genes deleted from the vector.
  • Ad2 other varieties of adenovirus (e.g., Ad3, Ad5, and Ad7) are also useful in the present invention.
  • the adenoviruses used in the present invention are replication deficient.
  • Replication deficient adenoviruses require the aid of a helper virus and/or packaging cell line to form infectious particles.
  • the resulting virus is capable of infecting cells and can express a polynucleotide of interest which is operably linked to a promoter, but cannot replicate in most cells.
  • Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: E1a, E1b, E3, E4, E2a, or L1 through L5.
  • the cells are engineered, ex vivo or in vivo, using an adeno-associated virus (AAV).
  • AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, N., Curr. Topics in Microbiol. Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb. Methods for producing and using such AAVs are known in the art. See, for example, U.S. Pat. Nos. 5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377.
  • an appropriate AAV vector for use in the present invention will include all the sequences necessary for DNA replication, encapsidation, and host-cell integration.
  • the polynucleotide construct is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press (1989).
  • the recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc.
  • Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses.
  • the packaging cells Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the polynucleotide construct. These viral particles are then used to transduce eukaryotic cells, either ex vivo or in vivo.
  • the transduced cells will contain the polynucleotide construct integrated into its genome, and will express a fsuion protein of the invention.
  • Another method of gene therapy involves operably associating heterologous control regions and endogenous polynucleotide sequences (e.g. encoding a polypeptide of the present invention) via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication No. WO 96/29411, published Sep. 26, 1996; International Publication No. WO 94/12650, published Aug. 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), which are herein encorporated by reference.
  • This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired.
  • Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter with targeting sequences flanking the promoter. Suitable promoters are described herein.
  • the targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence.
  • the targeting sequence will be sufficiently near the 5′ end of the desired endogenous polynucleotide sequence so the promoter will be operably linked to the endogenous sequence upon homologous recombination.
  • the promoter and the targeting sequences can be amplified using PCR.
  • the amplified promoter contains distinct restriction enzyme sites on the 5′ and 3′ ends.
  • the 3′ end of the first targeting sequence contains the same restriction enzyme site as the 5′ end of the amplified promoter and the 5′ end of the second targeting sequence contains the same restriction site as the 3′ end of the amplified promoter.
  • the amplified promoter and targeting sequences are digested and ligated together.
  • the promoter-targeting sequence construct is delivered to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail above.
  • transfection-facilitating agents such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc.
  • the P promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, catheter infusion, particle accelerators, etc. The methods are described in more detail below.
  • the promoter-targeting sequence construct is taken up by cells. Homologous recombination between the construct and the endogenous sequence takes place, such that an endogenous sequence is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence.
  • the polynucleotide encoding an albumin fusion protein of the present invention may contain a secretory signal sequence that facilitates secretion of the protein.
  • the signal sequence is positioned in the coding region of the polynucleotide to be expressed towards or at the 5′ end of the coding region.
  • the signal sequence may be homologous or heterologous to the polynucleotide of interest and may be homologous or heterologous to the cells to be transfected. Additionally, the signal sequence may be chemically synthesized using methods known in the art.
  • any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect.
  • This includes direct needle injection, systemic injection, catheter infusion, biolistic injectors, particle accelerators (i.e., “gene guns”), gelfoam sponge depots, other commercially available depot materials, osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery.
  • a preferred method of local administration is by direct injection.
  • an albumin fusion protein of the present invention complexed with a delivery vehicle is administered by direct injection into or locally within the area of arteries.
  • Administration of a composition locally within the area of arteries refers to injecting the composition centimeters and preferably, millimeters within arteries.
  • Another method of local administration is to contact a polynucleotide construct of the present invention in or around a surgical wound.
  • a patient can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound.
  • compositions useful in systemic administration include fusion proteins of the present invention complexed to a targeted delivery vehicle of the present invention.
  • Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site.
  • suitable delivery vehicles for use with systemic administration comprise liposomes comprising albumin fusion proteins of the invention for targeting the vehicle to a particular site.
  • Preferred methods of systemic administration include intravenous injection, aerosol, oral and percutaneous (topical) delivery.
  • Intravenous injections can be performed using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad. Sci. USA 189:11277-11281, 1992, which is incorporated herein by reference).
  • Oral delivery can be performed by complexing a polynucleotide construct of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art.
  • Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin.
  • a lipophilic reagent e.g., DMSO
  • Determining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration.
  • the frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses will be determined by the attending physician or veterinarian.
  • Albumin fusion proteins of the present invention can be administered to any animal, preferably to mammals and birds.
  • Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly preferred.
  • Albumin fusion proteins and/or polynucleotides encoding albumin fusion proteins of the present invention can be used in assays to test for one or more biological activities. If an albumin fusion protein and/or polynucleotide exhibits an activity in a particular assay, it is likely that the Therapeutic protein corresponding to the fusion portein may be involved in the diseases associated with the biological activity. Thus, the fusion protein could be used to treat the associated disease.
  • albumin fusion proteins of the invention and polynucleotides encoding these protiens may be used in diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with aberrant activity of secreted polypeptides.
  • fusion proteins of the present invention may be used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders relating to diseases and disorders of the endocrine system, the nervous system (See, for example, “Neurological Disorders” section below), and the immune system (See, for example, “Immune Activity” section below), respiratory system (See, for example, “Respiratory Disorders” section below), cardiovascular system (See, for example, “Cardiovascular Disorders” section below), reproductive system (See, for example, “Reproductive System Disorders” section below) digestive system (See, for example, “Gastrointestinal Disorders” section below), diseases and/or disorders relating to cell proliferation (See, for example, “Hyperproliferative Disorders” section below), and/or diseases or disorders relating to the blood ((See, for example, “Blood-Related Disorders” section below).
  • the present invention encompasses a method of treating a disease or disorder listed in the “Preferred Indication Y” column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein portion corresponding to a Therapeutic protein disclosed in the “Therapeutic Protein X” column of Table 1 (in the same row as the disease or disorder to be treated is listed in the “Preferred Indication Y” column of Table 1) in an amount effective to treat, prevent or ameliorate the disease or disorder.
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to a Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., leukemia, cancer, and/or as described below under “Hyperproliferative Disorders”).
  • a neoplastic disease e.g., leukemia, cancer, and/or as described below under “Hyperproliferative Disorders”.
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a neoplasm located in a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.g., leukemia), endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), uterus, eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital.
  • a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.g., leukemia), endocrine glands
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a pre-neoplastic condition, selected from the group consisting of: hyperplasia (e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”), metaplasia (e.g., connective tissue metaplasia, a typical metaplasia, and/or as described in the section entitled “Hyperproliferative Disorders”), and dysplasia (e.g., cervical dysplasia, and bronchopulmonary dysplasia).
  • hyperplasia e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”
  • metaplasia e.g., connective tissue metaplasia, a typical meta
  • a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a benign dysproliferative disorder selected from the group consisting of: benign tumors, fibrocystic conditions, tissue hypertrophy, and/or as described in the section entitled “Hyperproliferative Disorders”.
  • a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a blood disorder (e.g., as described below under “Immune Activity”, “Cardiovascular Disorders” and/or “Blood-Related Disorders”), and/or an infection (e.g., as described below under “Infectious Disease”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a blood disorder e.g., as described below under “Immune Activity”, “Cardiovascular Disorders” and/or “Blo
  • a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: anemia, pancytopenia, leukopenia, thrombocytopenia, leukemias, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL), plasmacytomas, multiple myeloma, Burkitt's lymphoma, arthritis, asthma, AIDS, autoimmune disease, rheumatoid arthritis, granulomatous disease, immune deficiency, inflammatory bowel disease, sepsis, neutropenia, neutrophilia, psoriasis, an immune reaction to a transplanted organ and/or tissue, systemic lup
  • a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the reproductive system (e.g., as described below under “Reproductive System Disorders”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disorder of the reproductive system e.g., as described below under “Reproductive System Disorders”.
  • a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1 an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: cryptorchism, prostatitis, inguinal hernia, varicocele, a leydig cell tumor, verrucous carcinoma, prostatitis, malacoplakia, Peyronie's disease, penile carcinoma, squamous cell hyperplasia, dysmenorrhea, ovarian adenocarcinoma, Turner's syndrome, mucopurulent cervicitis, a Sertoli-leydig tumor, ovarian cancer, uterine cancer, pelvic inflammatory disease, testicular cancer, prostate cancer, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes mellitus, cystic fibrosis
  • a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the immune system (e.g., as described below under “Immune Activity”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disorder of the immune system e.g., as described below under “Immune Activity”.
  • a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of bone cancer (e.g., osteochondroma, benign chondroma, chondroblastoma, chondromyxoid fibroma, osteoid osteoma, giant cell tumor, multiple myeloma, and osteosarcoma), Paget's Disease, rheumatoid arthritis, systemic lupus erythematosus, osteomyelitis, Lyme Disease, gout, bursitis, tendonitis, osteoporosis, osteoarthritis, muscular dystrophy, mitochondrial myopathy, cachexia, and multiple sclerosis.
  • bone cancer e.g., osteochondroma, benign chondroma, chondroblast
  • a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the cardiovascular system (e.g., as described below under “Cardiovascular Disorders”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disorder of the cardiovascular system e.g., as described below under “Cardiovascular Disorders”.
  • a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: myxoma, fibroma, rhabdomyoma, cardiovascular abnormality (e.g., a congenital heart defect, cerebral arterioyenous malformation, septal defect), heart disease (e.g., heart failure, congestive heart disease, arrhythmia, tachycardia, fibrillation, pericardial Disease, endocarditis), cardiac arrest, heart valve disease (e.g., stenosis, regurgitation, prolapse), vascular disease (e.g., hypertension, coronary artery disease, angina, aneurysm, arteriosclerosis, peripheral vascular disease), hyponatremia, hypernatremia,
  • a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”).
  • a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: spina bifida, hydranencephaly, neurofibromatosis, fetal alcohol syndrome, diabetes mellitus, PKU, Down's syndrome, Patau syndrome, Edwards syndrome, Turner syndrome, Apert syndrome, Carpenter syndrome, Conradi syndrome, Crouzon syndrome, cutis laxa, Cornelia de Lange syndrome, Ellis-van Creveld syndrome, Holt-Oram syndrome, Kartagener syndrome, Meckel-Gruber syndrome, Noonan syndrome, Pallister-Hall syndrome, Rubinstein-Taybi syndrome, Scimitar syndrome, Smith-Lemli-Opitz syndrome, thromocytopenia-absent radius (TAR) syndrome
  • a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a renal disorder (e.g., as described below under “Renal Disorders”).
  • a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a renal disorder e.g., as described below under “Renal Disorders”.
  • a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: bladder cancer, prostate cancer, benign prostatic hyperplasia, bladder disorders (e.g., urinary incontinence, urinary retention, urinary obstruction, urinary tract Infections, interstitial cystitis, prostatitis, neurogenic bladder, hematuria), a renal disorder (e.g., hydronephrosis, proteinuria, renal failure, pyelonephritis, urolithiasis, reflux nephropathy, and unilateral obstructive uropathy).
  • a disease and/or disorder selected from the group consisting of: bladder cancer, prostate cancer, benign prostatic hyperplasia, bladder disorders (e.g., urinary incontinence, urinary retention, urinary
  • a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the nervous system (e.g., as described below under “Neural Activity and Neurological Diseases”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disease or disorder of the nervous system e.g., as described below under “Neural Activity and Neurological Diseases”.
  • a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: brain cancer (e.g., brain stem glioma, brain tumor, central nervous system (Primary) lymphoma, central nervous system lymphoma, cerebellar astrocytoma, and cerebral astrocytoma, a neurodegenerative disorder (e.g., Alzheimer's Disease, Creutzfeldt-Jakob Disease, Parkinson's Disease, and Idiopathic Presenile Dementia), encephalomyelitis, cerebral malaria, meningitis, a metabolic brain disease (e.g., phenylketonuria and pyruvate carboxylase deficiency), cerebellar ataxia, ataxia tel
  • a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the respiratory system (e.g., as described below under “Respiratory Disorders”).
  • a disease and/or disorder relating to a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disease or disorder of the respiratory system e.g., as described below under “Respiratory Disorders”.
  • a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: a cancer of the respiratory system (such as larynx cancer, pharynx cancer, trachea cancer, epiglottis cancer, lung cancer, squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcinoma), an allergic reaction, cystic fibrosis, sarcoidosis, histiocytosis X, an infiltrative lung disease (e.g., pulmonary fibrosis and lymphoid interstitial pneumonia), an obstructive airway disease (e.g., asthma, emphysema, chronic or acute bronchitis), an occupational lung disease (e.g.,
  • a Therapeutic protein having an “Endocrine” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a disease or disorder of the respiratory system (e.g., as described below under “Respiratory Disorders”), a renal disorder (e.g., as described below under “Renal Disorders”), and/or a disorder of the endocrine system (e.g., as described below under “Endocrine Disorders”).
  • a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disease or disorder of the respiratory system e.g., as described below under “Respiratory Disorders”
  • a renal disorder e.g., as described below
  • a Therapeutic protein having a “Endocrine” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: a cancer of endocrine tissues and/or organs (e.g., cancer of the hypothalamus, pituitary gland, thyroid gland, parathyroid glands, pancreas, adrenal glands, ovaries, and testes), diabetes (e.g., diabetes insipidus, type I and type II diabetes mellitus), obesity, a disorder related to pituitary glands (e.g., hyperpituitarism, hypopituitarism, and pituitary dwarfism), hypothyroidism, hyperthyroidism, goiter, reproductive disorders (e.g.
  • a cancer of endocrine tissues and/or organs e.g., cancer of the hypothala
  • kidney cancer e.g., hypemephroma, transitional cell cancer, and Wilm's tumor
  • diabetic nephropathy e.g., interstitial nephritis
  • polycystic kidney disease e.g., glomerulonephritis (e.g., IgM mesangial proliferative glomerulonephritis and glomerulonephritis caused by an autoimmune disorder; such as Goodpasture's syndrome), and nephrocalcinosis.
  • a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to, for example, a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the gastrointestinal system (e.g., as described below under “Gastrointestinal Disorders”).
  • a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a disease or disorder of the gastrointestinal system e.g., as described below under “Gastrointestinal Disorders”.
  • a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: ulcerative colitis, appendicitis, Crohn's disease, hepatitis, hepatic encephalopathy, portal hypertension, cholelithiasis, cancer of the digestive system (e.g., biliary tract cancer, stomach cancer, colon cancer, gastric cancer, pancreatic cancer, cancer of the bile duct, a tumor of the colon (e.g., polyps or cancers), and cirrhosis), pancreatitis, ulcerative disease, pyloric stenosis, gastroenteritis, gastritis, gastric atropy, a benign tumor of the duodenum, distension, irritable bowel syndrome
  • a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a cellular and/or genetic abnormality (e.g., as described below under “Diseases at the Cellular Level”), angiogenesis (e.g., as described below under “Anti-Angiogenesis Activity”), and/or to promote or inhibit regeneration (e.g., as described below under “Regeneration”), and/or wound healing (e.g., as described below under “Wound Healing and Epithelial Cell Proliferation”).
  • a neoplastic disease e.g., as described below under “Hyperproliferative Disorders”
  • a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: connective tissue metaplasia, mixed connective tissue disease, focal epithelial hyperplasia, epithelial metaplasia, mucoepithelial dysplasia, graft v.
  • an albumin fusion protein of the present invention may be used to diagnose and/or prognose diseases and/or disorders associated with the tissue(s) in which the gene corresponding to the Therapeutic protein portion of the fusion portien of the invention is expressed.
  • fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention are useful in the diagnosis, detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone activation, neurotransmitter activity, cellular signaling, cellular proliferation, cellular differentiation, and cell migration.
  • fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful for the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with the following systems.
  • Albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing diseases, disorders, and/or conditions of the immune system, by, for example, activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells.
  • Immune cells develop through a process called hematopoiesis, producing myeloid (platelets, red blood cells, neutrophils, and macrophages) and lymphoid (B and T lymphocytes) cells from pluripotent stem cells.
  • etiology of these immune diseases, disorders, and/or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy or toxins), or infectious.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as a marker or detector of a particular immune system disease or disorder.
  • a fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be used to treat diseases and disorders of the immune system and/or to inhibit or enhance an immune response generated by cells associated with the tissue(s) in which the polypeptide of the invention is expressed.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing, and/or prognosing immunodeficiencies, including both congenital and acquired immunodeficiencies.
  • B cell immunodeficiencies in which immunoglobulin levels B cell function and/or B cell numbers are decreased include: X-linked agammaglobulinemia (Bruton's disease), X-linked infantile agammaglobulinemia, X-linked immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM, X-linked lymphoproliferative syndrome (XLP), agammaglobulinemia including congenital and acquired agammaglobulinemia, adult onset agammaglobulinemia, late-onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia, unspecified hypogammaglobulinemia, recessive agammaglobulinemia (Swiss type), Selective IgM deficiency, selective IgA deficiency, selective IgG subclass deficiencies, IgG subclass deficiency (with or without IgA deficiency), I
  • Ataxia-telangiectasia or conditions associated with ataxia-telangiectasia are treated, prevented, diagnosed, and/or prognosing using the, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • Examples of congenital immunodeficiencies in which T cell and/or B cell function and/or number is decreased include, but are not limited to: DiGeorge anomaly, severe combined immunodeficiencies (SCID) (including, but not limited to, X-linked SCID, autosomal recessive SCID, adenosine deaminase deficiency, purine nucleoside phosphorylase (PNP) deficiency, Class II MHC deficiency (Bare lymphocyte syndrome), Wiskott-Aldrich syndrome, and ataxia telangiectasia), thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22q11.2 deletion, chronic mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic CD4+ T-lymphocytopenia, immunodeficiency with predominant T cell defect (unspecified), and unspecified immunodeficiency of cell mediated immunity.
  • SCID severe combined immunodeficiencies
  • DiGeorge anomaly or conditions associated with DiGeorge anomaly are treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • immunodeficiencies that may be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, chronic granulomatous disease, Chédiak-Higashi syndrome, myeloperoxidase deficiency, leukocyte glucose-6-phosphate dehydrogenase deficiency, X-linked lymphoproliferative syndrome (XLP), leukocyte adhesion deficiency, complement component deficiencies (including C1, C2, C3, C4, C5, C6, C7, C8 and/or C9 deficiencies), reticular dysgenesis, thymic alymphoplasia-aplasia, immunodeficiency with thymoma, severe congenital leukopenia, dysplasia with immunodeficiency, neonatal neutropenia, short limbed dwarfism, and Nezelof syndrome-combined immunodefici
  • the immunodeficiencies and/or conditions associated with the immunodeficiencies recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among immunodeficient individuals.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among B cell and/or T cell immunodeficient individuals.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing autoimmune disorders.
  • Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue. Therefore, the administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can inhibit an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune disorders.
  • Autoimmune diseases or disorders that may be treated, prevented, diagnosed and/or prognosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, one or more of the following: systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's thyroiditis, autoimmune hemolytic anemia, hemolytic anemia, thrombocytopenia, autoimmune thrombocytopenia purpura, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura), autoimmunocytopenia, Goodpasture's syndrome, Pemphigus vulgaris, myasthenia gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes mellit
  • Additional disorders that are likely to have an autoimmune component that may be treated, prevented, and/or diagnosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, type II collagen-induced arthritis, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, neuritis, uveitis ophthalmia, polyendocrinopathies, Reiter's Disease, Stiff-Man Syndrome, autoimmune pulmonary inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disorders.
  • Additional disorders that are likely to have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often characterized, e.g., by nonhistone ANA), pernicious anemia (often characterized, e.g., by antiparietal cell, microsomes, and intrinsic factor antibodies), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermat
  • Additional disorders that may have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g., by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitochondria antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-MI (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.
  • the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using for example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • rheumatoid arthritis is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • systemic lupus erythematosus is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • idiopathic thrombocytopenia purpura is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • IgA nephropathy is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a immunosuppressive agent(s).
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, prognosing, and/or diagnosing diseases, disorders, and/or conditions of hematopoietic cells.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with a decrease in certain (or many) types hematopoietic cells, including but not limited to, leukopenia, neutropenia, anemia, and thrombocytopenia.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with an increase in certain (or many) types of hematopoietic cells, including but not limited to, histiocytosis.
  • Allergic reactions and conditions such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Moreover, these molecules can be used to treat, prevent, prognose, and/or diagnose anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat, prevent, diagnose and/or prognose IgE-mediated allergic reactions.
  • allergic reactions include, but are not limited to, asthma, rhinitis, and eczema.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate IgE concentrations in vitro or in vivo.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention have uses in the diagnosis, prognosis, prevention, and/or treatment of inflammatory conditions.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the activation, proliferation and/or differentiation of cells involved in an inflammatory response, these molecules can be used to prevent and/or treat chronic and acute inflammatory conditions.
  • Such inflammatory conditions include, but are not limited to, for example, inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome), ischemia-reperfusion injury, endotoxin lethality, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, over production of cytokines (e.g., TNF or IL-1.), respiratory disorders (e.g., asthma and allergy); gastrointestinal disorders (e.g., inflammatory bowel disease); cancers (e.g., gastric, ovarian, lung, bladder, liver, and breast); CNS disorders (e.g., multiple sclerosis; ischemic brain injury and/or stroke, traumatic brain injury, neurodegenerative disorders (e.g., Parkinson's disease and Alzheimer's disease); AIDS-related dementia; and prion disease); cardiovascular disorders (e.g., atherosclerosis, myocarditis
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention have uses in the treatment of tissue-specific inflammatory disorders, including, but not limited to, adrenalitis, alveolitis, angiocholecystitis, appendicitis, balanitis, blepharitis, bronchitis, bursitis, carditis, cellulitis, cervicitis, cholecystitis, chorditis, cochlitis, colitis, conjunctivitis, cystitis, dermatitis, diverticulitis, encephalitis, endocarditis, esophagitis, eustachitis, fibrositis, folliculitis, gastritis, gastroenteritis, gingivitis, glossitis, hepatosplenitis, keratitis, labyrin
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are useful to diagnose, prognose, prevent, and/or treat organ transplant rejections and graft-versus-host disease.
  • Organ rejection occurs by host immune cell destruction of the transplanted tissue through an immune response.
  • an immune response is also involved in GVHD, but, in this case, the foreign transplanted immune cells destroy the host tissues.
  • Polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing experimental allergic and hyperacute xenograft rejection.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are useful to diagnose, prognose, prevent, and/or treat immune complex diseases, including, but not limited to, serum sickness, post streptococcal glomerulonephritis, polyarteritis nodosa, and immune complex-induced vasculitis.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat, detect, and/or prevent infectious agents. For example, by increasing the immune response, particularly increasing the proliferation activation and/or differentiation of B and/or T cells, infectious diseases may be treated, detected, and/or prevented.
  • the immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also directly inhibit the infectious agent (refer to section of application listing infectious agents, etc), without necessarily eliciting an immune response.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a vaccine adjuvant that enhances immune responsiveness to an antigen.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance tumor-specific immune responses.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-viral immune responses.
  • Anti-viral immune responses that may be enhanced using the compositions of the invention as an adjuvant include virus and virus associated diseases or symptoms described herein or otherwise known in the art.
  • the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of: AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis B).
  • compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of: HIV/AIDS, respiratory syncytial virus, Dengue, rotavirus, Japanese B encephalitis, influenza A and B, parainfluenza, measles, cytomegalovirus, rabies, Junin, Chikungunya, Rift Valley Fever, herpes simplex, and yellow fever.
  • a virus, disease, or symptom selected from the group consisting of: HIV/AIDS, respiratory syncytial virus, Dengue, rotavirus, Japanese B encephalitis, influenza A and B, parainfluenza, measles, cytomegalovirus, rabies, Junin, Chikungunya, Rift Valley Fever, herpes simplex, and yellow fever.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-bacterial or anti-fungal immune responses.
  • Anti-bacterial or anti-fungal immune responses that may be enhanced using the compositions of the invention as an adjuvant include bacteria or fungus and bacteria or fungus associated diseases or symptoms described herein or otherwise known in the art.
  • the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: tetanus, Diphtheria, botulism, and meningitis type B.
  • compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: Vibrio cholerae, Mycobacterium leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis, Streptococcus pneumoniae , Group B streptococcus, Shigella spp., Enterotoxigenic Escherichia coli , Enterohemorrhagic E. coli , and Borrelia burgdorferi.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-parasitic immune responses.
  • Anti-parasitic immune responses that may be enhanced using the compositions of the invention as an adjuvant include parasite and parasite associated diseases or symptoms described herein or otherwise known in the art.
  • the compositions of the invention are used as an adjuvant to enhance an immune response to a parasite.
  • the compositions of the invention are used as an adjuvant to enhance an immune response to Plasmodium (malaria) or Leishmania.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat infectious diseases including silicosis, sarcoidosis, and idiopathic pulmonary fibrosis; for example, by preventing the recruitment and activation of mononuclear phagocytes.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are administered to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and human, most preferably human) to boost the immune system to produce increased quantities of one or more antibodies (e.g., IgG, IgA, IgM, and IgE), to induce higher affinity antibody production and immunoglobulin class switching (e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response.
  • an animal e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and human, most preferably human
  • an animal e.g.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell responsiveness to pathogens.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an activator of T cells.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to induce higher affinity antibodies.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to increase serum immunoglobulin concentrations.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to accelerate recovery of immunocompromised individuals.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among aged populations and/or neonates.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an immune system enhancer prior to, during, or after bone marrow transplant and/or other transplants (e.g., allogeneic or xenogeneic organ transplantation).
  • compositions of the invention may be administered prior to, concomitant with, and/or after transplantation.
  • compositions of the invention are administered after transplantation, prior to the beginning of recovery of T-cell populations.
  • compositions of the invention are first administered after transplantation after the beginning of recovery of T cell populations, but prior to full recovery of B cell populations.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having an acquired loss of B cell function.
  • Conditions resulting in an acquired loss of B cell function that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, HIV Infection, AIDS, bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL).
  • CLL B cell chronic lymphocytic leukemia
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having a temporary immune deficiency.
  • Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a regulator of antigen presentation by monocytes, dendritic cells, and/or B-cells.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention enhance antigen presentation or antagonizes antigen presentation in vitro or in vivo.
  • this enhancement or antagonism of antigen presentation may be useful as an anti-tumor treatment or to modulate the immune system.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to direct an individual's immune system towards development of a humoral response (i.e. TH2) as opposed to a TH1 cellular response.
  • a humoral response i.e. TH2
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means to induce tumor proliferation and thus make it more susceptible to anti-neoplastic agents.
  • multiple myeloma is a slowly dividing disease and is thus refractory to virtually all anti-neoplastic regimens. If these cells were forced to proliferate more rapidly their susceptibility profile would likely change.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell production in pathologies such as AIDS, chronic lymphocyte disorder and/or Common Variable Immunodificiency.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for generation and/or regeneration of lymphoid tissues following surgery, trauma or genetic defect.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in the pretreatment of bone marrow samples prior to transplant.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a gene-based therapy for genetically inherited disorders resulting in immuno-incompetence/immunodeficiency such as observed among SCID patients.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leishmania.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of regulating secreted cytokines that are elicited by polypeptides of the invention.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in one or more of the applications decribed herein, as they may apply to veterinary medicine.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of blocking various aspects of immune responses to foreign agents or self.
  • diseases or conditions in which blocking of certain aspects of immune responses may be desired include autoimmune disorders such as lupus, and arthritis, as well as immunoresponsiveness to skin allergies, inflammation, bowel disease, injury and diseases/disorders associated with pathogens.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for preventing the B cell proliferation and Ig secretion associated with autoimmune diseases such as idiopathic thrombocytopenic purpura, systemic lupus erythematosus and multiple sclerosis.
  • polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the present fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention invention are used as a inhibitor of B and/or T cell migration in endothelial cells. This activity disrupts tissue architecture or cognate responses and is useful, for example in disrupting immune responses, and blocking sepsis.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for chronic hypergammaglobulinemia evident in such diseases as monoclonal gammopathy of undetermined significance (MGUS), Waldenstrom's disease, related idiopathic monoclonal gammopathies, and plasmacytomas.
  • MGUS monoclonal gammopathy of undetermined significance
  • Waldenstrom's disease related idiopathic monoclonal gammopathies
  • plasmacytomas MGUS
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed for instance to inhibit polypeptide chemotaxis and activation of macrophages and their precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain autoimmune and chronic inflammatory and infective diseases. Examples of autoimmune diseases are described herein and include multiple sclerosis, and insulin-dependent diabetes.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat idiopathic hyper-eosinophilic syndrome by, for example, preventing eosinophil production and migration.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit complement mediated cell lysis.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit antibody dependent cellular cytotoxicity.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed for treating atherosclerosis, for example, by preventing monocyte infiltration in the artery wall.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed to treat adult respiratory distress syndrome (ARDS).
  • ARDS adult respiratory distress syndrome
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful for stimulating wound and tissue repair, stimulating angiogenesis, and/or stimulating the repair of vascular or lymphatic diseases or disorders. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to stimulate the regeneration of mucosal surfaces.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, treat, and/or prevent a disorder characterized by primary or acquired immunodeficiency, deficient serum immunoglobulin production, recurrent infections, and/or immune system dysfunction.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat or prevent infections of the joints, bones, skin, and/or parotid glands, blood-borne infections (e.g., sepsis, meningitis, septic arthritis, and/or osteomyelitis), autoimmune diseases (e.g., those disclosed herein), inflammatory disorders, and malignancies, and/or any disease or disorder or condition associated with these infections, diseases, disorders and/or malignancies) including, but not limited to, CVID, other primary immune deficiencies, HIV disease, CLL, recurrent bronchitis, sinusitis, otitis media, conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g., severe herpes zoster), and/or pneumocystis carnii.
  • blood-borne infections e.g., sepsis, meningitis,
  • fusion proteins of the invention include, but are not limited to, HIV infection, HTLV-BLV infection, lymphopenia, phagocyte bactericidal dysfunction anemia, thrombocytopenia, and hemoglobinuria.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, and/or diagnose an individual having common variable immunodeficiency disease (“CVID”; also known as “acquired agammaglobulinemia” and “acquired hypogammaglobulinemia”) or a subset of this disease.
  • CVID common variable immunodeficiency disease
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to diagnose, prognose, prevent, and/or treat cancers or neoplasms including immune cell or immune tissue-related cancers or neoplasms.
  • cancers or neoplasms that may be prevented, diagnosed, or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, acute myelogenous leukemia, chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL) Chronic lymphocyte leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, EBV-transformed diseases, and/or diseases and disorders described in the section entitled “Hyperproliferative Disorders” elsewhere herein.
  • ALL acute lymphocytic anemia
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for decreasing cellular proliferation of Large B-cell Lymphomas.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of decreasing the involvement of B cells and Ig associated with Chronic Myelogenous Leukemia.
  • compositions of the invention are used as an agent to boost immunoresponsiveness among B cell immunodeficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hemostatic (the stopping of bleeding) or thrombolytic (clot dissolving) activity.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to treat or prevent blood coagulation diseases, disorders, and/or conditions (e.g., afibrinogenemia, factor deficiencies, hemophilia), blood platelet diseases, disorders, and/or conditions (e.g., thrombocytopenia), or wounds resulting from trauma, surgery, or other causes.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting. These molecules could be important in the treatment or prevention of heart attacks (infarction), strokes, or scarring.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, diagnose, prognose, and/or treat thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for the prevention of occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease.
  • albumin fusion proteins of the invention include, but are not limited to, the prevention of occlusions in extrcorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines).
  • extrcorporeal devices e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, diagnose, prognose, and/or treat diseases and disorders of the blood and/or blood forming organs associated with the tissue(s) in which the polypeptide of the invention is expressed.
  • the fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hematopoietic activity (the formation of blood cells).
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to increase the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets.
  • the ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of anemias and leukopenias described below.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to decrease the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets.
  • the ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of leukocytoses, such as, for example eosinophilia.
  • the fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, treat, or diagnose blood dyscrasia.
  • Anemias are conditions in which the number of red blood cells or amount of hemoglobin (the protein that carries oxygen) in them is below normal. Anemia may be caused by excessive bleeding, decreased red blood cell production, or increased red blood cell destruction (hemolysis).
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias.
  • Anemias that may be treated prevented or diagnosed by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include iron deficiency anemia, hypochromic anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia, idiopathic acquired sideroblastic anemia, red cell aplasia, megaloblastic anemia (e.g., pernicious anemia, (vitamin B12 deficiency) and folic acid deficiency anemia), aplastic anemia, hemolytic anemias (e.g., autoimmune helolytic anemia, microangiopathic hemolytic anemia, and paroxysmal nocturnal hemoglobinuria).
  • iron deficiency anemia e.g., hypochromic anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia, idiopathic acquired sideroblastic anemia, red cell aplasia,
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with diseases including but not limited to, anemias associated with systemic lupus erythematosus, cancers, lymphomas, chronic renal disease, and enlarged spleens.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias arising from drug treatments such as anemias associated with methyldopa, dapsone, and/or sulfadrugs.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with abnormal red blood cell architecture including but not limited to, hereditary spherocytosis, hereditary elliptocytosis, glucose-6-phosphate dehydrogenase deficiency, and sickle cell anemia.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing hemoglobin abnormalities, (e.g., those associated with sickle cell anemia, hemoglobin C disease, hemoglobin S-C disease, and hemoglobin E disease). Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating thalassemias, including, but not limited to, major and minor forms of alpha-thalassemia and beta-thalassemia.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating bleeding disorders including, but not limited to, thrombocytopenia (e.g., idiopathic thrombocytopenic purpura, and thrombotic thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet disorders (e.g., storage pool disease such as Chediak-Higashi and Hermansky-Pudlak syndromes, thromboxane A2 dysfunction, thromboasthenia, and Bernard-Soulier syndrome), hemolytic-uremic syndrome, hemophelias such as hemophelia A or Factor VII deficiency and Christmas disease or Factor IX deficiency, Hereditary Hemorhhagic Telangiectsia, also known as Rendu-Osler-Weber syndrome, allergic purpura
  • thrombocytopenia
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention on the clotting time of blood may be monitored using any of the clotting tests known in the art including, but not limited to, whole blood partial thromboplastin time (PTT), the activated partial thromboplastin time (aPTT), the activated clotting time (ACT), the recalcified activated clotting time, or the Lee-White Clotting time.
  • PTT whole blood partial thromboplastin time
  • aPTT activated partial thromboplastin time
  • ACT activated clotting time
  • Lee-White Clotting time the recalcified activated clotting time
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating acquired platelet dysfunction such as platelet dysfunction accompanying kidney failure, leukemia, multiple myeloma, cirrhosis of the liver, and systemic lupus erythematosus as well as platelet dysfunction associated with drug treatments, including treatment with aspirin, ticlopidine, nonsteroidal anti-inflammatory drugs (used for arthritis, pain, and sprains), and penicillin in high doses.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders characterized by or associated with increased or decreased numbers of white blood cells.
  • Leukopenia occurs when the number of white blood cells decreases below normal. Leukopenias include, but are not limited to, neutropenia and lymphocytopenia. An increase in the number of white blood cells compared to normal is known as leukocytosis. The body generates increased numbers of white blood cells during infection. Thus, leukocytosis may simply be a normal physiological parameter that reflects infection.
  • leukocytosis may be an indicator of injury or other disease such as cancer.
  • Leokocytoses include but are not limited to, eosinophilia, and accumulations of macrophages.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukopenia.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukocytosis.
  • Leukopenia may be a generalized decreased in all types of white blood cells, or may be a specific depletion of particular types of white blood cells.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating decreases in neutrophil numbers, known as neutropenia.
  • Neutropenias that may be diagnosed, prognosed, prevented, and/or treated by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, infantile genetic agranulocytosis, familial neutropenia, cyclic neutropenia, neutropenias resulting from or associated with dietary deficiencies (e.g., vitamin B 12 deficiency or folic acid deficiency), neutropenias resulting from or associated with drug treatments (e.g., antibiotic regimens such as penicillin treatment, sulfonamide treatment, anticoagulant treatment, anticonvulsant drugs, anti-thyroid drugs, and cancer chemotherapy), and neutropenias resulting from increased neutrophil destruction that may occur in association with some bacterial or viral infections, allergic disorders, autoimmune diseases, conditions in which an individual has an enlarged spleen (e.g., Felty syndrome, malaria and sarcoidosis), and some drug treatment regimens.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating lymphocytopenias (decreased numbers of B and/or T lymphocytes), including, but not limited to, lymphocytopenias resulting from or associated with stress, drug treatments (e.g., drug treatment with corticosteroids, cancer chemotherapies, and/or radiation therapies), AIDS infection and/or other diseases such as, for example, cancer, rheumatoid arthritis, systemic lupus erythematosus, chronic infections, some viral infections and/or hereditary disorders (e.g., DiGeorge syndrome, Wiskott-Aldrich Syndome, severe combined immunodeficiency, ataxia telangiectsia).
  • lymphocytopenias decreased numbers of B and/or T lymphocytes
  • drug treatments e.g., drug treatment with cortic
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with macrophage numbers and/or macrophage function including, but not limited to, Gaucher's disease, Niemann-Pick disease, Letterer-Siwe disease and Hand-Schuller-Christian disease.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with eosinophil numbers and/or eosinophil function including, but not limited to, idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-Schuller-Christian disease.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukemias and lymphomas including, but not limited to, acute lymphocytic (lymphpblastic) leukemia (ALL), acute myeloid (myelocytic, myelogenous, myeloblastic, or myelomonocytic) leukemia, chronic lymphocytic leukemia (e.g., B cell leukemias, T cell leukemias, Sezary syndrome, and Hairy cell leukenia), chronic myelocytic (myeloid, myelogenous, or granulocytic) leukemia, Hodgkin's lymphoma, non-hodgkin's lymphoma, Burkitt's lymphoma, and mycosis fungoides.
  • ALL acute lymphocytic leukemia
  • acute myeloid my
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders of plasma cells including, but not limited to, plasma cell dyscrasias, monoclonal gammaopathies, monoclonal gammopathies of undetermined significance, multiple myeloma, macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and Raynaud's phenomenon.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing myeloproliferative disorders, including but not limited to, polycythemia vera, relative polycythemia, secondary polycythemia, myelofibrosis, acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including both primary and seconday thrombocythemia) and chronic myelocytic leukemia.
  • myeloproliferative disorders including but not limited to, polycythemia vera, relative polycythemia, secondary polycythemia, myelofibrosis, acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including both primary and seconday thrombocythemia) and chronic myelocytic leukemia.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a treatment prior to surgery, to increase blood cell production.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to enhance the migration, phagocytosis, superoxide production, antibody dependent cellular cytotoxicity of neutrophils, eosionophils and macrophages.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to stem cells pheresis.
  • the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to platelet pheresis.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase cytokine production.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in preventing, diagnosing, and/or treating primary hematopoietic disorders.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect hyperproliferative disorders, including neoplasms.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the proliferation of the disorder through direct or indirect interactions.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may proliferate other cells which can inhibit the hyperproliferative disorder.
  • hyperproliferative disorders can be treated.
  • This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response.
  • decreasing an immune response may also be a method of treating hyperproliferative disorders, such as a chemotherapeutic agent.
  • Examples of hyperproliferative disorders that can be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.
  • neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvis, skin, soft tissue,
  • hyperproliferative disorders can also be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • hyperproliferative disorders include, but are not limited to: Acute Childhood Lymphoblastic Leukemia, Acute Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult (Primary) Hepatocellular Cancer, Adult (Primary) Liver Cancer, Adult Acute Lymphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, prevent, and/or treat premalignant conditions and to prevent progression to a neoplastic or malignant state, including but not limited to those disorders described above.
  • Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such abnormal growth conditions, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W. B. Saunders Co., Philadelphia, pp. 68-79.)
  • Hyperplasia is a form of controlled cell proliferation, involving an increase in cell number in a tissue or organ, without significant alteration in structure or function.
  • Hyperplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, angiofollicular mediastinal lymph node hyperplasia, angiolymphoid hyperplasia with eosinophilia, a typical melanocytic hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal hyperplasia, endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival
  • Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell.
  • Metaplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, agnogenic myeloid metaplasia, apocrine metaplasia, a typical metaplasia, autoparenchymatous metaplasia, connective tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia.
  • Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells. Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomorphism. Dysplasia characteristically occurs where there exists chronic irritation or inflammation.
  • Dysplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia
  • Additional pre-neoplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar keratosis.
  • benign dysproliferative disorders e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia
  • leukoplakia keratoses
  • Bowen's disease Farmer's Skin
  • solar cheilitis solar cheilitis
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to diagnose and/or prognose disorders associated with the tissue(s) in which the polypeptide of the invention is expressed.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein may be used to treat cancers and neoplasms, including, but not limited to, those described herein.
  • albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein may be used to treat acute myelogenous leukemia.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may affect apoptosis, and therefore, would be useful in treating a number of diseases associated with increased cell survival or the inhibition of apoptosis.
  • diseases associated with increased cell survival or the inhibition of apoptosis include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroidit
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above.
  • Additional diseases or conditions associated with increased cell survival that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and
  • Diseases associated with increased apoptosis that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include AIDS; neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v.
  • neurodegenerative disorders such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral s
  • ischemic injury such as that caused by myocardial infarction, stroke and reperfusion injury
  • liver injury e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer
  • toxin-induced liver disease such as that caused by alcohol
  • septic shock cachexia and anorexia.
  • Hyperproliferative diseases and/or disorders that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, neoplasms located in the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract.
  • neoplasms located in the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system
  • hyperproliferative disorders can also be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • hyperproliferative disorders include, but are not limited to: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above.
  • Another preferred embodiment utilizes polynucleotides encoding albumin fusion proteins of the invention to inhibit aberrant cellular division, by gene therapy using the present invention, and/or protein fusions or fragments thereof.
  • the present invention provides a method for treating cell proliferative disorders by inserting into an abnormally proliferating cell a polynucleotide encoding an albumin fusion protein of the present invention, wherein said polynucleotide represses said expression.
  • polynucleotides of the present invention is a DNA construct comprising a recombinant expression vector effective in expressing a DNA sequence encoding said polynucleotides.
  • the DNA construct encoding the fusion protein of the present invention is inserted into cells to be treated utilizing a retrovirus, or more preferably an adenoviral vector (See G J. Nabel, et. al., PNAS 1999 96: 324-326, which is hereby incorporated by reference).
  • the viral vector is defective and will not transform non-proliferating cells, only proliferating cells.
  • the polynucleotides of the present invention inserted into proliferating cells either alone, or in combination with or fused to other polynucleotides can then be modulated via an external stimulus (i.e. magnetic, specific small molecule, chemical, or drug administration, etc.), which acts upon the promoter upstream of said polynucleotides to induce expression of the encoded protein product.
  • an external stimulus i.e. magnetic, specific small molecule, chemical, or drug administration, etc.
  • the beneficial therapeutic affect of the present invention may be expressly modulated (i.e. to increase, decrease, or inhibit expression of the present invention) based upon said external stimulus.
  • Polynucleotides of the present invention may be useful in repressing expression of oncogenic genes or antigens.
  • repressing expression of the oncogenic genes is intended the suppression of the transcription of the gene, the degradation of the gene transcript (pre-message RNA), the inhibition of splicing, the destruction of the messenger RNA, the prevention of the post-translational modifications of the protein, the destruction of the protein, or the inhibition of the normal function of the protein.
  • polynucleotides of the present invention may be administered by any method known to those of skill in the art including, but not limited to transfection, electroporation, microinjection of cells, or in vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any other method described throughout the specification.
  • the polynucleotide of the present invention may be delivered by known gene delivery systems such as, but not limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (1986); Wilson, et al., Proc. Natl. Acad. Sci. U.S.A.
  • vaccinia virus system Chokrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA delivery systems (Yates et al., Nature 313:812 (1985)) known to those skilled in the art.
  • vaccinia virus system Chokrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA delivery systems (Yates et al., Nature 313:812 (1985)) known to those skilled in the art.
  • retrovirus or adenoviral (as described in the art and elsewhere herein) delivery system known to those of skill in the art. Since host DNA replication is required for retroviral DNA to integrate and the retrovirus will be unable to self replicate due to the lack of the retrovirus genes needed for its life cycle. Utilizing such a retroviral delivery system for polynucleotides of the present invention will target said gene and constructs to abnormally proliferating cells and will spare the non-dividing normal cells.
  • the polynucleotides of the present invention may be delivered directly to cell proliferative disorder/disease sites in internal organs, body cavities and the like by use of imaging devices used to guide an injecting needle directly to the disease site.
  • the polynucleotides of the present invention may also be administered to disease sites at the time of surgical intervention.
  • cell proliferative disease any human or animal disease or disorder, affecting any one or any combination of organs, cavities, or body parts, which is characterized by single or multiple local abnormal proliferations of cells, groups of cells, or tissues, whether benign or malignant.
  • any amount of the polynucleotides of the present invention may be administered as long as it has a biologically inhibiting effect on the proliferation of the treated cells. Moreover, it is possible to administer more than one of the polynucleotide of the present invention simultaneously to the same site.
  • biologically inhibiting is meant partial or total growth inhibition as well as decreases in the rate of proliferation or growth of the cells. The biologically inhibitory dose may be determined by assessing the effects of the polynucleotides of the present invention on target malignant or abnormally proliferating cell growth in tissue culture, tumor growth in animals and cell cultures, or any other method known to one of ordinary skill in the art.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention of the present invention are useful in inhibiting the angiogenesis of proliferative cells or tissues, either alone, as a protein fusion, or in combination with other polypeptides directly or indirectly, as described elsewhere herein.
  • said anti-angiogenesis effect may be achieved indirectly, for example, through the inhibition of hematopoietic, tumor-specific cells, such as tumor-associated macrophages (See Joseph I B, et al. J Natl Cancer Inst, 90(21):1648-53 (1998), which is hereby incorporated by reference).
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in inhibiting proliferative cells or tissues through the induction of apoptosis.
  • These fusion protieins and/or polynucleotides may act either directly, or indirectly to induce apoptosis of proliferative cells and tissues, for example in the activation of a death-domain receptor, such as tumor necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF-receptor-related apoptosis-mediated protein (TRAMP) and TNF-related apoptosis-inducing ligand (TRAIL) receptor-1 and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem 254(3):439-59 (1998), which is hereby incorporated by reference).
  • TNF tumor necrosis factor
  • TRAMP TNF-receptor-related apopto
  • these fusion proteins and/or polynucleotides may induce apoptosis through other mechanisms, such as in the activation of other proteins which will activate apoptosis, or through stimulating the expression of these proteins, either alone or in combination with small molecule drugs or adjuviants, such as apoptonin, galectins, thioredoxins, anti-inflammatory proteins (See for example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses. 50(5):423-33 (1998), Chem Biol Interact. Apr 24;111-112:23-34 (1998), J Mol Med. 76(6):402-12 (1998), Int J Tissue React;20(1):3-15 (1998), which are all hereby incorporated by reference).
  • small molecule drugs or adjuviants such as apoptonin, galectins, thioredoxins, anti-inflammatory proteins
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are useful in inhibiting the metastasis of proliferative cells or tissues. Inhibition may occur as a direct result of administering these albumin fusion proteins and/or polynucleotides, or indirectly, such as activating the expression of proteins known to inhibit metastasis, for example alpha 4 integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125-41, which is hereby incorporated by reference). Such thereapeutic affects of the present invention may be achieved either alone, or in combination with small molecule drugs or adjuvants.
  • the invention provides a method of delivering compositions containing the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to targeted cells expressing the a polypeptide bound by, that binds to, or associates with an albumin fuison protein of the invention.
  • Albumin fusion proteins of the invention may be associated with with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalent interactions.
  • Albumin fusion proteins of the invention are useful in enhancing the immunogenicity and/or antigenicity of proliferating cells or tissues, either directly, such as would occur if the albumin fusion proteins of the invention ‘vaccinated’ the immune response to respond to proliferative antigens and immunogens, or indirectly, such as in activating the expression of proteins known to enhance the immune response (e.g. chemokines), to said antigens and immunogens.
  • proteins known to enhance the immune response e.g. chemokines
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat, prevent, diagnose, and/or prognose disorders of the renal system.
  • Renal disorders which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, kidney failure, nephritis, blood vessel disorders of kidney, metabolic and congenital kidney disorders, urinary disorders of the kidney, autoimmune disorders, sclerosis and necrosis, electrolyte imbalance, and kidney cancers.
  • Kidney diseases which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, acute kidney failure, chronic kidney failure, atheroembolic renal failure, end-stage renal disease, inflammatory diseases of the kidney (e.g., acute glomerulonephritis, postinfectious glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, membranous glomerulonephritis, familial nephrotic syndrome, membranoproliferative glomerulonephritis I and II, mesangial proliferative glomerulonephritis, chronic glomerulonephritis, acute tubulointerstitial nephritis, chronic tubulointerstitial nephritis, acute post-streptococcal glomerulonephritis (PSGN), pyelonephritis, lupus nephriti
  • compositions of the invention can be used to diagnose, prognose, prevent, and/or treat metabolic and congenital disorders of the kidney (e.g., uremia, renal amyloidosis, renal osteodystrophy, renal tubular acidosis, renal glycosuria, nephrogenic diabetes insipidus, cystinuria, Fanconi's syndrome, renal fibrocystic osteosis (renal rickets), Hartnup disease, Bartter's syndrome, Liddle's syndrome, polycystic kidney disease, medullary cystic disease, medullary sponge kidney, Alport's syndrome, nail-patella syndrome, congenital nephrotic syndrome, CRUSH syndrome, horseshoe kidney, diabetic nephropathy, nephrogenic diabetes insipidus, analgesic nephropathy, kidney stones, and membranous nephropathy), and autoimmune disorders of the kidney (e.g., systemic lup
  • compositions of the invention can also be used to diagnose, prognose, prevent, and/or treat sclerotic or necrotic disorders of the kidney (e.g., glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis (FSGS), necrotizing glomerulonephritis, and renal papillary necrosis), cancers of the kidney (e.g., nephroma, hypemephroma, nephroblastoma, renal cell cancer, transitional cell cancer, renal adenocarcinoma, squamous cell cancer, and Wilm's tumor), and electrolyte imbalances (e.g., nephrocalcinosis, pyuria, edema, hydronephritis, proteinuria, hyponatremia, hypernatremia, hypokalemia, hyperkalemia, hypocalcemia, hypercalcemia, hypophosphatemia, and hyperphosphatemia).
  • sclerotic or necrotic disorders of the kidney
  • compositions of the invention may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art.
  • Compositions of the invention may be administered as part of a Therapeutic, described in more detail below. Methods of delivering polynucleotides of the invention are described in more detail herein.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat, prevent, diagnose, and/or prognose cardiovascular disorders, including, but not limited to, peripheral artery disease, such as limb ischemia.
  • Cardiovascular disorders include, but are not limited to, cardiovascular abnormalities, such as arterio-arterial fistula, arterioyenous fistula, cerebral arterioyenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome.
  • cardiovascular abnormalities such as arterio-arterial fistula, arterioyenous fistula, cerebral arterioyenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome.
  • Congenital heart defects include, but are not limited to, aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects.
  • Cardiovascular disorders also include, but are not limited to, heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis.
  • heart disease such as arrhythmias, carcinoid heart disease
  • Arrhythmias include, but are not limited to, sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson-White syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation.
  • Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia.
  • Heart valve diseases include, but are not limited to, aortic valve insufficiency, aortic valve stenosis, hear murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis.
  • Myocardial diseases include, but are not limited to, alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis.
  • Myocardial ischemias include, but are not limited to, coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
  • coronary disease such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning.
  • Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease, Klippel-Trenaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema, aortic diseases, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disorders, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease, Raynaud's disease, CREST syndrome
  • Aneurysms include, but are not limited to, dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms.
  • Arterial occlusive diseases include, but are not limited to, arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans.
  • Cerebrovascular disorders include, but are not limited to, carotid artery diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arterioyenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subaraxhnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency.
  • Embolisms include, but are not limited to, air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromoboembolisms.
  • Thrombosis include, but are not limited to, coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis.
  • Ischemic disorders include, but are not limited to, cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia.
  • Vasculitis includes, but is not limited to, aortitis, arteritis, Behcet's Syndrome, Churg-Strauss Syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art. Methods of delivering polynucleotides are described in more detail herein.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat, prevent, diagnose, and/or prognose diseases and/or disorders of the respiratory system.
  • Diseases and disorders of the respiratory system include, but are not limited to, nasal vestibulitis, nonallergic rhinitis (e.g., acute rhinitis, chronic rhinitis, atrophic rhinitis, vasomotor rhinitis), nasal polyps, and sinusitis, juvenile angiofibromas, cancer of the nose and juvenile papillomas, vocal cord polyps, nodules (singer's nodules), contact ulcers, vocal cord paralysis, laryngoceles, pharyngitis (e.g., viral and bacterial), tonsillitis, tonsillar cellulitis, parapharyngeal abscess, laryngitis, laryngoceles, and throat cancers (e.g., cancer of the nasopharynx, tonsil cancer, larynx cancer), lung cancer (e.g., squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcino
  • Additional diseases and disorders of the respiratory system include, but are not limited to bronchiolitis, polio (poliomyelitis), croup, respiratory syncytial viral infection, mumps, erythema infectiosum (fifth disease), roseola infantum, progressive rubella panencephalitis, german measles, and subacute sclerosing panencephalitis), fungal pneumonia (e.g., Histoplasmosis, Coccidioidomycosis, Blastomycosis, fungal infections in people with severely suppressed immune systems (e.g., cryptococcosis, caused by Cryptococcus neoformans ; aspergillosis, caused by Aspergillus spp.; candidiasis, caused by Candida; and mucormycosis)), Pneumocystis carinii (pneumocystis pneumonia), a typical pneumonias (e.g., Mycoplasma and Chlamycosis
  • angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail. Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases.
  • a number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye disorders, and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-634 (1991); Folkman et al., N. Engl. J. Med., 333:1757-1763 (1995); Auerbach et al., J. Microvasc. Res. 29:401-411 (1985); Folkman, Advances in Cancer Research, eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol.
  • the present invention provides for treatment of diseases or disorders associated with neovascularization by administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention.
  • Malignant and metastatic conditions which can be treated with the polynucleotides and polypeptides, or agonists or antagonists of the invention include, but are not limited to, malignancies, solid tumors, and cancers described herein and otherwise known in the art (for a review of such disorders, see Fishman et al., Medicine, 2d Ed., J. B.
  • the present invention provides a method of treating an angiogenesis-related disease and/or disorder, comprising administering to an individual in need thereof a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention.
  • fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a variety of additional methods in order to therapeutically treat a cancer or tumor.

Abstract

The present invention encompasses albumin fusion proteins. Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention and using these nucleic acids, vectors, and/or host cells. Additionally the present invention encompasses pharmaceutical compositions comprising albumin fusion proteins and methods of treating, preventing, or ameliorating diseases, disordrs or conditions using albumin fusion proteins of the invention.

Description

  • This application claims the benefit of priority under 35 U.S.C. §119(e) based on the following U.S. provisional applications No. 60/229,358 filed on Apr. 12, 2000; No. 60/199,384 filed on Apr. 25, 2000; and No. 60/256,931 filed on Dec. 21, 2000. Each of the provisional applications is hereby incorporated by reference in its entirety.[0001]
  • BACKGROUND OF THE INVENTION
  • The invention relates generally to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or fragments or variants of albumin. The invention further relates to Therapeutic proteins (including, but not limited to, a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or fragments or variants of albumin, that exhibit extended shelf-life and/or extended or therapeutic activity in solution. These fusion proteins are herein collectively referred to as “albumin fusion proteins of the invention.” The invention encompasses therapeutic albumin fusion proteins, compositions, pharmaceutical compositions, formulations and kits. Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention using these nucleic acids, vectors, and/or host cells. [0002]
  • The invention is also directed to methods of in vitro stabilizing a Therapeutic protein via fusion or conjugation of the Therapeutic protein to albumin or fragments or variants of albumin. [0003]
  • Human serum albumin (HSA, or HA), a protein of 585 amino acids in its mature form (as shown in FIG. 15 or in SEQ ID NO:18), is responsible for a significant proportion of the osmotic pressure of serum and also functions as a carrier of endogenous and exogenous ligands. At present, HA for clinical use is produced by extraction from human blood. The production of recombinant HA (rHA) in microorganisms has been disclosed in EP 330 451 and [0004] EP 361 991.
  • The role of albumin as a carrier molecule and its inert nature are desirable properties for use as a carrier and transporter of polypeptides in vivo. The use of albumin as a component of an albumin fusion protein as a carrier for various proteins has been suggested in WO 93/15199, WO 93/15200, and EP 413 622. The use of N-terminal fragments of HA for fusions to polypeptides has also been proposed (EP 399 666). Fusion of albumin to the Therapeutic protein may be achieved by genetic manipulation, such that the DNA coding for HA, or a fragment thereof, is joined to the DNA coding for the Therapeutic protein. A suitable host is then transformed or transfected with the fused nucleotide sequences, so arranged on a suitable plasmid as to express a fusion polypeptide. The expression may be effected in vitro from, for example, prokaryotic or eukaryotic cells, or in vivo e.g. from a transgenic organism. [0005]
  • Therapeutic proteins in their native state or when recombinantly produced, such as interferons and growth hormones, are typically labile molecules exhibiting short shelf-lives, particularly when formulated in aqueous solutions. The instability in these molecules when formulated for administration dictates that many of the molecules must be lyophilized and refrigerated at all times during storage, thereby rendering the molecules difficult to transport and/or store. Storage problems are particularly acute when pharmaceutical formulations must be stored and dispensed outside of the hospital environment. Many protein and peptide drugs also require the addition of high concentrations of other protein such as albumin to reduce or prevent loss of protein due to binding to the container. This is a major concern with respect to proteins such as IFN. For this reason, many Therapeutic proteins are formulated in combination with large proportion of albumin carrier molecule (100-1000 fold excess), though this is an undesirable and expensive feature of the formulation. [0006]
  • Few practical solutions to the storage problems of labile protein molecules have been proposed. Accordingly, there is a need for stabilized, long lasting formulations of proteinaceous therapeutic molecules that are easily dispensed, preferably with a simple formulation requiring minimal post-storage manipulation. [0007]
  • SUMMARY OF THE INVENTION
  • The present invention is based, in part, on the discovery that Therapeutic proteins may be stabilized to extend the shelf-life, and/or to retain the Therapeutic protein's activity for extended periods of time in solution, in vitro and/or in vivo, by genetically or chemically fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to stabilize the protein and/or its activity. In addition it has been determined that the use of albumin-fusion proteins or albumin conjugated proteins may reduce the need to formulate protein solutions with large excesses of carrier proteins (such as albumin, unfused) to prevent loss of Therapeutic proteins due to factors such as binding to the container. [0008]
  • The present invention encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin. The present invention also encompasses albumin fusion proteins comprising a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) fused to albumin or a fragment (portion) or variant of albumin, that is sufficient to prolong the shelf life of the Therapeutic protein, and/or stabilize the Therapeutic protein and/or its activity in solution (or in a pharmaceutical composition) in vitro and/or in vivo. Nucleic acid molecules encoding the albumin fusion proteins of the invention are also encompassed by the invention, as are vectors containing these nucleic acids, host cells transformed with these nucleic acids vectors, and methods of making the albumin fusion proteins of the invention and using these nucleic acids, vectors, and/or host cells. [0009]
  • The invention also encompasses pharmaceutical formulations comprising an albumin fusion protein of the invention and a pharmaceutically acceptable diluent or carrier. Such formulations may be in a kit or container. Such kit or container may be packaged with instructions pertaining to the extended shelf life of the Therapeutic protein. Such formulations may be used in methods of treating, preventing, ameliorating or diagnosing a disease or disease symptom in a patient, preferably a mammal, most preferably a human, comprising the step of administering the pharmaceutical formulation to the patient. [0010]
  • In other embodiments, the present invention encompasses methods of preventing treating, or ameliorating a disease or disorder. In preferred embodiments, the present invention encompasses a method of treating a disease or disorder listed in the “Preferred Indication Y” column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein portion corresponding to a Therapeutic protein (or fragment or variant thereof) disclosed in the “Therapeutic Protein X” column of Table 1 (in the same row as the disease or disorder to be treated is listed in the “Preferred Indication Y” column of Table 1) in an amount effective to treat prevent or ameliorate the disease or disorder. [0011]
  • In another embodiment, the invention includes a method of extending the shelf life of a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to extend the shelf-life of the Therapeutic protein. In a preferred embodiment, the Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin. In a most preferred embodiment, the Therapeutic protein used according to this method is fused to albumin, or a fragment or variant of albumin, via recombinant DNA technology or genetic engineering. [0012]
  • In another embodiment, the invention includes a method of stabilizing a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) in solution, comprising the step of fusing or conjugating the Therapeutic protein to albumin or a fragment (portion) or variant of albumin, that is sufficient to stabilize the Therapeutic protein. In a preferred embodiment, the Therapeutic protein used according to this method is fused to the albumin, or the fragment or variant of albumin. In a most preferred embodiment, the Therapeutic protein used according to this method is fused to albumin, or a fragment or variant of albumin, via recombinant DNA technology or genetic engineering. [0013]
  • The present invention further includes transgenic organisms modified to contain the nucleic acid molecules of the invention, preferably modified to express the albumin fusion proteins encoded by the nucleic acid molecules.[0014]
  • BRIEF DESCRIPTION OF THE FIGURES
  • FIG. 1 depicts the extended shelf-life of an HA fusion protein in terms of the biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to 5 weeks at 37° C. Under these conditions, hGH has no observed activity by [0015] week 2.
  • FIG. 2 depicts the extended shelf-life of an HA fusion protein in terms of the stable biological activity (Nb2 cell proliferation) of HA-hGH remaining after incubation in cell culture media for up to 3 weeks at 4, 37, or 50° C. Data is normalized to the biological activity of hGH at time zero. [0016]
  • FIGS. 3A and 3B compare the biological activity of HA-hGH with hGH in the Nb2 cell proliferation assay. FIG. 3A shows proliferation after 24 hours of incubation with various concentrations of hGH or the albumin fusion protein, and FIG. 3B shows proliferation after 48 hours of incubation with various concentrations of hGH or the albumin fusion protein. [0017]
  • FIG. 4 shows a map of a plasmid (pPPC0005) that can be used as the base vector into which polynucleotides encoding the Therapeutic proteins (including polypeptide and fragments and variants thereof) may be cloned to form HA-fusions. Plasmid Map key: PRB1p: PRB1 [0018] S. cerevisiae promoter; FL: Fusion leader sequence; rHA: cDNA encoding HA; ADH1t: ADH1 S. cerevisiae terminator; T3: T3 sequencing primer site; T7: T7 sequencing primer site; Amp R: α-lactamase gene; ori: origin of replication. Please note that in the provisional applications to which this application claims priority, the plasmid in FIG. 4 was labeled pPPC0006, instead of pPPC0005. In addition the drawing of this plasmid did not show certain pertinent restriction sites in this vector. Thus in the present application, the drawing is labeled pPPC0005 and more restriction sites of the same vector are shown.
  • FIG. 5 compares the recovery of vial-stored HA-IFN solutions of various concentrations with a stock solution after 48 or 72 hours of storage. [0019]
  • FIG. 6 compares the activity of an HA-α-IFN fusion protein after administration to monkeys via TV or SC. [0020]
  • FIG. 7 describes the bioavailability and stability of an HA-α-IFN fusion protein. [0021]
  • FIG. 8 is a map of an expression vector for the production of HA- -IFN. [0022]
  • FIG. 9 shows the location of loops in HA. [0023]
  • FIG. 10 is an example of the modification of an HA loop. [0024]
  • FIG. 11 is a representation of the HA loops. [0025]
  • FIG. 12 shows the HA loop IV. [0026]
  • FIG. 13 shows the tertiary structure of HA. [0027]
  • FIG. 14 shows an example of a scFv-HA fusion [0028]
  • FIG. 15 shows the amino acid sequence of the mature form of human albumin (SEQ ID NO:18) and a polynucleotide encoding it (SEQ ID NO:17).[0029]
  • DETAILED DESCRIPTION
  • As described above, the present invention is based, in part, on the discovery that a Therapeutic protein (e.g., a polypeptide, antibody, or peptide, or fragments and variants thereof) may be stabilized to extend the shelf-life and/or retain the Therapeutic protein's activity for extended periods of time in solution (or in a pharmaceutical composition) in vitro and/or in vivo, by genetically fusing or chemically conjugating the Therapeutic protein, polypeptide or peptide to all or a portion of albumin sufficient to stabilize the protein and its activity. [0030]
  • The present invention relates generally to albumin fusion proteins and methods of treating, preventing, or ameliorating diseases or disorders. As used herein, “albumin fusion protein” refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof). An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or chemical conjugation to one another. The Therapeutic protein and albumin protein, once part of the albumin fusion protein, may be referred to as a “portion”, “region” or “moiety” of the albumin fusion protein (e.g., a “Therapeutic protein portion” or an “albumin protein portion”). [0031]
  • In one embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin. [0032]
  • In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin. In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein. In a further preferred embodiment, the Therapeutic protein portion of the albumin fusion protein is the extracellular soluble domain of the Therapeutic protein. In an alternative embodiment, the Therapeutic protein portion of the albumin fusion protein is the active form of the Therapeutic protien. [0033]
  • In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin. In preferred embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin. [0034]
  • Therapeutic Proteins [0035]
  • As stated above, an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion or chemical conjugation. [0036]
  • As used herein, “Therapeutic protein” refers to proteins, polypeptides, antibodies, peptides or fragments or variants thereof, having one or more therapeutic and/or biological activities. Therapeutic proteins encompassed by the invention include but are not limited to, proteins, polypeptides, peptides, antibodies, and biologics. (The terms peptides, proteins, and polypeptides are used interchangeably herein.) It is specifically contemplated that the term “Therapeutic protein” encompasses antibodies and fragments and variants thereof. Thus an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an antibody. Additionally, the term “Therapeutic protein” may refer to the endogenous or naturally occurring correlate of a Therapeutic protein. [0037]
  • By a polypeptide displaying a “therapeutic activity” or a protein that is “therapeutically active” is meant a polypeptide that possesses one or more known biological and/or therapeutic activities associated with a therapeutic protein such as one or more of the Therapeutic proteins described herein or otherwise known in the art. As a non-limiting example, a “Therapeutic protein” is a protein that is useful to treat, prevent or ameliorate a disease, condition or disorder. As a non-limiting example, a “Therapeutic protein” may be one that binds specifically to a particular cell type (normal (e.g., lymphocytes) or abnormal e.g., (cancer cells)) and therefore may be used to target a compound (drug, or cytotoxic agent) to that cell type specifically. [0038]
  • In another non-limiting example, a “Therapeutic protein” is a protein that has a biological activity, and in particular, a biological activity that is useful for treating preventing or ameliorating a disease. A non-inclusive list of biological activities that may be possessed by a Therapeutic protein includes, enhancing the immune response, promoting angiogenesis, inhibiting angiogenesis, regulating hematopoietic functions, stimulating nerve growth, enhancing an immune response, inhibiting an immune response, or any one or more of the biological activities described in the “Biological Activities” section below. [0039]
  • As used herein, “therapeutic activity” or “activity” may refer to an activity whose effect is consistent with a desirable therapeutic outcome in humans, or to desired effects in non-human mammals or in other species or organisms. Therapeutic activity may be measured in vivo or in vitro. For example, a desirable effect may be assayed in cell culture. As an example, when hGH is the Therapeutic protein, the effects of hGH on cell proliferation as described in Example 1 may be used as the endpoint for which therapeutic activity is measured. Such in vitro or cell culture assays are commonly available for many Therapeutic proteins as described in the art. Examples of assays include, but are not limited to those described herein in the Examples section or in the “Exemplary Activity Assay” column of Table 1. [0040]
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, such as cell surface and secretory proteins, are often modified by the attachment of one or more oligosaccharide groups. The modification, referred to as glycosylation, can dramatically affect the physical properties of proteins and can be important in protein stability, secretion, and localization. Glycosylation occurs at specific locations along the polypeptide backbone. There are usually two major types of glycosylation: glycosylation characterized by O-linked oligosaccharides, which are attached to serine or threonine residues; and glycosylation characterized by N-linked oligosaccharides, which are attached to asparagine residues in an Asn-X-Ser/Thr sequence, where X can be any amino acid except proline. N-acetylneuramic acid (also known as sialic acid) is usually the terminal residue of both N-linked and O-linked oligosaccharides. Variables such as protein structure and cell type influence the number and nature of the carbohydrate units within the chains at different glycosylation sites. Glycosylation isomers are also common at the same site within a given cell type. [0041]
  • For example, several types of human interferon are glycosylated. Natural human interferon-α2 is O-glycosylated at threonine 106, and N-glycosylation occurs at asparagine 72 in interferon-α14 (Adolf et al., J. Biochem 276:511 (1991); Nyman T A et al., J. Biochem 329:295 (1998)). The oligosaccharides at [0042] asparagine 80 in natural interferon-β1α may play an important factor in the solubility and stability of the protein, but may not be essential for its biological activity. This permits the production of an unglycosylated analog (interferon-β1b) engineered with sequence modifications to enhance stability (Hosoi et al., J. Interferon Res. 8:375 (1988; Karpusas et al., Cell Mol Life Sci 54:1203 (1998); Knight, J. Interferon Res. 2:421 (1982); Runkel et al., Pharm Res 15:641 (1998); Lin, Dev. Biol. Stand. 96:97 (1998))1. Interferon-γ contains two N-linked oligosaccharide chains at positions 25 and 97, both important for the efficient formation of the bioactive recombinant protein, and having an influence on the pharmacokinetic properties of the protein (Sareneva et al., Eur. J. Biochem 242:191 (1996); Sareneva et al, Biochem J. 303:831 (1994); Sareneva et al., J. Interferon Res. 13:267 (1993)). Mixed O-linked and N-linked glycosylation also occurs, for example in human erythropoietin, N-linked glycosylation occurs at asparagine residues located at positions 24, 38 and 83 while O-linked glycosylation occurs at a serine residue located at position 126 (Lai et al., J. Biol. Chem. 261:3116 (1986); Broudy et al., Arch. Biochem. Biophys. 265:329 (1988)).
  • Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, as well as analogs and variants thereof, may be modified so that glycosylation at one or more sites is altered as a result of manipulation(s) of their nucleic acid sequence, by the host cell in which they are expressed, or due to other conditions of their expression. For example, glycosylation isomers may be produced by abolishing or introducing glycosylation sites, e.g., by substitution or deletion of amino acid residues, such as substitution of glutamine for asparagine, or unglycosylated recombinant proteins may be produced by expressing the proteins in host cells that will not glycosylate them, e.g. in [0043] E. coli or glycosylation-deficient yeast. These approaches are described in more detail below and are known in the art.
  • Therapeutic proteins (particularly those disclosed in Table 1) and their nucleic acid sequences are well known in the art and available in public databases such as Chemical Abstracts Services Databases (e.g., the CAS Registry), GenBank, and GenSeq as shown in Table 1. [0044]
  • Additional Therapeutic proteins corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, one or more of the Therapeutic proteins or peptides disclosed in the “Therapeutic Protein X” column of Table 1, or fragment or variable thereof. [0045]
  • Table 1 provides a non-exhaustive list of Therapeutic proteins that correspond to a Therapeutic protein portion of an albumin fusion protein of the invention. The “Therapeutic Protein X” column discloses Therapeutic protein molecules followed by parentheses containing scientific and brand names that comprise, or alternatively consist of, that Therapeutic protein molecule or a fragment or variant thereof. “Therapeutic protein X” as used herein may refer either to an individual Therapeutic protein molecule (as defined by the amino acid sequence obtainable from the CAS and Genbank accession numbers), or to the entire group of Therapeutic proteins associated with a given Therapeutic protein molecule disclosed in this column. The “Exemplary Identifier” column provides Chemical Abstracts Services (CAS) Registry Numbers (published by the American Chemical Society) and/or Genbank Accession Numbers ((e.g., Locus ID, NP_XXXXX (Reference Sequence Protein), and XP_XXXXX (Model Protein) identifiers available through the national Center for Biotechnology Information (NCBI) webpage at www.ncbi.nlm.nih.gov) that correspond to entries in the CAS Registry or Genbank database which contain an amino acid sequence of the Therapeutic Protein Molecule or of a fragment or variant of the Therapeutic Protein Molecule. In addition GenSeq Accession numbers and/or journal publication citations are given to identify the exemplary amino acid sequence for some polypeptides. The summary pages associated with each of these CAS and Genbank and GenSeq Accession Numbers as well as the cited journal publications (e.g., PubMed ID number (PMID)) are each incorporated by reference in their entireties, particularly with respect to the amino acid sequences described therein. The “PCT/Patent Reference” column provides U.S. Patent numbers, or PCT International Publication Numbers corresponding to patents and/or published patent applications that describe the Therapeutic protein molecule. Each of the patents and/or published patent applications cited in the “PCT/Patent Reference” column are herein incorporated by reference in their entireties. In particular, the amino acid sequences of the specified polypeptide set forth in the sequence listing of each cited “PCT/Patent Reference”, the variants of these amino acid sequences (mutations, fragments, etc.) set forth, for example, in the detailed description of each cited “PCT/Patent Reference”, the therapeutic indications set forth, for example, in the detailed description of each cited “PCT/Patent Reference”, and the activity asssaysfor the specified polypeptide set forth in the detailed description, and more particularly, the examples of each cited “PCT/Patent Reference” are incorporated herein by reference. The “Biological activity” column describes Biological activities associated with the Therapeutic protein molecule. The “Exemplary Activity Assay” column provides references that describe assays which may be used to test the therapeutic and/or biological activity of a Therapeutic protein or an albumin fusion protein of the invention comprising a Therapeutic protein X portion. Each of the references cited in the “Exemplary Activity Assay” column are herein incorporated by reference in their entireties, particularly with respect to the description of the respective activity assay described in the reference (see Methods section, for example) for assaying the corresponding biological activity set forth in the “Biological Activity” column of Table 1. The “Preferred Indication Y” column describes disease, disorders, and/or conditions that may be treated, prevented, diagnosed, or ameliorated by Therapeutic protein X or an albumin fusion protein of the invention comprising a Therapeutic protein X portion. [0046]
  • The recitation of “Cancer” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., leukemias, cancers, and/or as described below under “Hyperproliferative Disorders”). [0047]
  • In specific embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a neoplasm located in a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.g., leukemia), endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), uterus, eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital. [0048]
  • In specific embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a pre-neoplastic condition, selected from the group consisting of: hyperplasia (e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”), metaplasia (e.g., connective tissue metaplasia, a typical metaplasia, and/or as described in the section entitled “Hyperproliferative Disorders”), and/or dysplasia (e.g., cervical dysplasia, and bronchopulmonary dysplasia). [0049]
  • In another specific embodiment, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a benign dysproliferative disorder selected from the group consisting of: benign tumors, fibrocystic conditions, tissue hypertrophy, and/or as described in the section entitled “Hyperproliferative Disorders”. [0050]
  • The recitation of “Immune/Hematopoietic” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), blood disorders (e.g., as described below under “Immune Activity” “Cardiovascular Disorders” and/or “Blood-Related Disorders”), and infections (e.g., as described below under “Infectious Disease”). [0051]
  • In specific embodiments, a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: anemia, pancytopenia, leukopenia, thrombocytopenia, leukemias, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL), plasmacytomas, multiple myeloma, Burkitt's lymphoma, arthritis, asthma, A/DS, autoimmune disease, rheumatoid arthritis, granulomatous disease, immune deficiency, inflammatory bowel disease, sepsis, neutropenia, neutrophilia, psoriasis, immune reactions to transplanted organs and tissues, systemic lupus erythematosis, hemophilia, hypercoagulation, diabetes mellitus, endocarditis, meningitis, Lyme Disease, and allergies. [0052]
  • The recitation of “Reproductive” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the reproductive system (e.g., as described below under “Reproductive System Disorders”). [0053]
  • In specific embodiments, a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cryptorchism, prostatitis, inguinal hernia, varicocele, leydig cell tumors, verrucous carcinoma, prostatitis, malacoplakia, Peyronie's disease, penile carcinoma, squamous cell hyperplasia, dysmenorrhea, ovarian adenocarcinoma, Turner's syndrome, mucopurulent cervicitis, Sertoli-leydig tumors, ovarian cancer, uterine cancer, pelvic inflammatory disease, testicular cancer, prostate cancer, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes mellitus, cystic fibrosis, Kartagener's syndrome, testicular atrophy, testicular feminization, anorchia, ectopic testis, epididymitis, orchitis, gonorrhea, syphilis, testicular torsion, vasitis nodosa, germ cell tumors, stromal tumors, dysmenorrhea, retroverted uterus, endometriosis, fibroids, adenomyosis, anovulatory bleeding, amenorrhea, Cushing's syndrome, hydatidiform moles, Asherman's syndrome, premature menopause, precocious puberty, uterine polyps, dysfunctional uterine bleeding, cervicitis, chronic cervicitis, mucopurulent cervicitis, cervical dysplasia, cervical polyps, Nabothian cysts, cervical erosion, cervical incompetence, cervical neoplasms, pseudohermaphroditism, and premenstrual syndrome. [0054]
  • The recitation of “Musculoskeletal” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the immune system (e.g., as described below under “Immune Activity”). [0055]
  • In specific embodiments, a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: bone cancers (e.g., osteochondromas, benign chondromas, chondroblastoma, chondromyxoid fibromas, osteoid osteomas, giant cell tumors, multiple myeloma, osteosarcomas), Paget's Disease, rheumatoid arthritis, systemic lupus erythematosus, osteomyelitis, Lyme Disease, gout, bursitis, tendonitis, osteoporosis, osteoarthritis, muscular dystrophy, mitochondrial myopathy, cachexia, and multiple sclerosis. [0056]
  • The recitation of “Cardiovascular” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), and disorders of the cardiovascular system (e.g., as described below under “Cardiovascular Disorders”). [0057]
  • In specific embodiments, a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, a fusion protein containing this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: myxomas, fibromas, rhabdomyomas, cardiovascular abnormalities (e.g., congenital heart defects, cerebral arterioyenous malformations, septal defects), heart disease (e.g., heart failure, congestive heart disease, arrhythmia, tachycardia, fibrillation, pericardial Disease, endocarditis), cardiac arrest, heart valve disease (e.g., stenosis, regurgitation, prolapse), vascular disease (e.g., hypertension, coronary artery disease, angina, aneurysm, arteriosclerosis, peripheral vascular disease), hyponatremia, hypernatremia, hypokalemia, and hyperkalemia. [0058]
  • The recitation of “Mixed Fetal” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”). [0059]
  • In specific embodiments, a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: spina bifida, hydranencephaly, neurofibromatosis, fetal alcohol syndrome, diabetes mellitus, PKU, Down's syndrome, Patau syndrome, Edwards syndrome, Turner syndrome, Apert syndrome, Carpenter syndrome, Conradi syndrome, Crouzon syndrome, cutis laxa, Cornelia de Lange syndrome, Ellis-van Creveld syndrome, Holt-Oram syndrome, Kartagener syndrome, Meckel-Gruber syndrome, Noonan syndrome, Pallister-Hall syndrome, Rubinstein-Taybi syndrome, Scimitar syndrome, Smith-Lemli-Opitz syndrome, thromocytopenia-absent radius (TAR) syndrome, Treacher Collins syndrome, Williams syndrome, Hirschsprung's disease, Meckel's diverticulum, polycystic kidney disease, Turner's syndrome, and gonadal dysgenesis, Klippel-Feil syndrome, Ostogenesis imperfecta, muscular dystrophy, Tay-Sachs disease, Wilm's tumor, neuroblastoma, and retinoblastoma. [0060]
  • The recitation of “Excretory” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and renal disorders (e.g., as described below under “Renal Disorders”). [0061]
  • In specific embodiments, a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: bladder cancer, prostate cancer, benign prostatic hyperplasia, bladder disorders (e.g., urinary incontinence, urinary retention, urinary obstruction, urinary tract Infections, interstitial cystitis, prostatitis, neurogenic bladder, hematuria), renal disorders (e.g., hydronephrosis, proteinuria, renal failure, pyelonephritis, urolithiasis, reflux nephropathy, and unilateral obstructive uropathy). [0062]
  • The recitation of “Neural/Sensory” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the nervous system (e.g., as described below under “Neural Activity and Neurological Diseases”). [0063]
  • In specific embodiments, a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: brain cancer (e.g., brain stem glioma, brain tumors, central nervous system (Primary) lymphoma, central nervous system lymphoma, cerebellar astrocytoma, and cerebral astrocytoma, neurodegenerative disorders (e.g., Alzheimer's Disease, Creutzfeldt-Jakob Disease, Parkinson's Disease, and Idiopathic Presenile Dementia), encephalomyelitis, cerebral malaria, meningitis, metabolic brain diseases (e.g., phenylketonuria and pyruvate carboxylase deficiency), cerebellar ataxia, ataxia telangiectasia, and AIDS Dementia Complex, schizophrenia, attention deficit disorder, hyperactive attention deficit disorder, autism, and obsessive compulsive disorders. [0064]
  • The recitation of “Respiratory” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the respiratory system (e.g., as described below under “Respiratory Disorders”). [0065]
  • In specific embodiments, a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cancers of the respiratory system such as larynx cancer, pharynx cancer, trachea cancer, epiglottis cancer, lung cancer, squamous cell carcinomas, small cell (oat cell) carcinomas, large cell carcinomas, and adenocarcinomas. Allergic reactions, cystic fibrosis, sarcoidosis, histiocytosis X, infiltrative lung diseases (e.g., pulmonary fibrosis and lymphoid interstitial pneumonia), obstructive airway diseases (e.g., asthma, emphysema, chronic or acute bronchitis), occupational lung diseases (e.g., silicosis and asbestosis), pneumonia, and pleurisy. [0066]
  • The recitation of “Endocrine” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the respiratory system (e.g., as described below under “Respiratory Disorders”), renal disorders (e.g., as described below under “Renal Disorders”), and disorders of the endocrine system (e.g., as described below under “Endocrine Disorders”. [0067]
  • In specific embodiments, a Therapeutic protein having a “Endocrine” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: cancers of endocrine tissues and organs (e.g., cancers of the hypothalamus, pituitary gland, thyroid gland, parathyroid glands, pancreas, adrenal glands, ovaries, and testes), diabetes (e.g., diabetes insipidus, type I and type II diabetes mellitus), obesity, disorders related to pituitary glands (e.g., hyperpituitarism, hypopituitarism, and pituitary dwarfism), hypothyroidism, hyperthyroidism, goiter, reproductive disorders (e.g. male and female infertility), disorders related to adrenal glands (e.g., Addison's Disease, corticosteroid deficiency, and Cushing's Syndrome), kidney cancer (e.g., hypemephroma, transitional cell cancer, and Wilm's tumor), diabetic nephropathy, interstitial nephritis, polycystic kidney disease, glomenilonephritis (e.g., IgM mesangial proliferative glomerulonephritis and glomerulonephritis caused by autoimmune disorders; such as Goodpasture's syndrome), and nephrocalcinosis. [0068]
  • The recitation of “Digestive” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”) and diseases or disorders of the gastrointestinal system (e.g., as described below under “Gastrointestinal Disorders”. [0069]
  • In specific embodiments, a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: ulcerative colitis, appendicitis, Crohn's disease, hepatitis, hepatic encephalopathy, portal hypertension, cholelithiasis, cancer of the digestive system (e.g., biliary tract cancer, stomach cancer, colon cancer, gastric cancer, pancreatic cancer, cancer of the bile duct, tumors of the colon (e.g., polyps or cancers), and cirrhosis), pancreatitis, ulcerative disease, pyloric stenosis, gastroenteritis, gastritis, gastric atropy, benign tumors of the duodenum, distension, irritable bowel syndrome, malabsorption, congenital disorders of the small intestine, bacterial and parasitic infection, megacolon, Hirschsprung's disease, aganglionic megacolon, acquired megacolon, colitis, anorectal disorders (e.g., anal fistulas, hemorrhoids), congenital disorders of the liver (e.g., Wilson's disease, hemochromatosis, cystic fibrosis, biliary atresia, and alpha1-antitrypsin deficiency), portal hypertension, cholelithiasis, and jaundice. [0070]
  • The recitation of “Connective/Epithelial” in the “Preferred Indication Y” column indicates that corresponding Therapeutic protein, fusion protein containing the Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate diseases and/or disorders relating to neoplastic diseases (e.g., as described below under “Hyperproliferative Disorders”), cellular and genetic abnormalities (e.g., as described below under “Diseases at the Cellular Level”), angiogenesis (e.g., as described below under “Anti-Angiogenesis Activity”), and or to promote or inhibit regeneration (e.g., as described below under “Regeneration”), and wound healing (e.g., as described below under “Wound Healing and Epithelial Cell Proliferation”). [0071]
  • In specific embodiments, a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used for example, to diagnose, treat, prevent, and/or ameliorate a disease or disorder selected from the group consisting of: connective tissue metaplasia, mixed connective tissue disease, focal epithelial hyperplasia, epithelial metaplasia, mucoepithelial dysplasia, graft v. host disease, polymyositis, cystic hyperplasia, cerebral dysplasia, tissue hypertrophy, Alzheimer's disease, lymphoproliferative disorder, Waldenstron's macroglobulinemia, Crohn's disease, pernicious anemia, idiopathic Addison's disease, glomerulonephritis, bullous pemphigoid, Sjogren's syndrome, diabetes mellitus, cystic fibrosis, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, osteoporosis, osteocarthritis, periodontal disease, wound healing, relapsing polychondritis, vasculitis, polyarteritis nodosa, Wegener's granulomatosis, cellulitis, rheumatoid arthritis, psoriatic arthritis, discoid lupus erythematosus, systemic lupus erythematosus, scleroderma, CREST syndrome, Sjogren's syndrome, polymyositis, dermatomyositis, mixed connective tissue disease, relapsing polychondritis, vasculitis, Henoch-Schonlein syndrome, erythema nodosum, polyarteritis nodosa, temporal (giant cell) arteritis, Takayasu's arteritis, Wegener's granulomatosis, Reiter's syndrome, Behcet's syndrome, ankylosing spondylitis, cellulitis, keloids, Ehler Danlos syndrome, Marfan syndrome, pseudoxantoma elasticum, osteogenese imperfecta, chondrodysplasias, epidermolysis bullosa, Alport syndrome, and cutis laxa. [0072]
    Therapeutic Exemplary
    Protein X Identifier PCT/Patent Reference Preferred Indication Y
    HETFO52 B03768 US6066724-A Neural/Sensory,
    Reproductive
    HETEZ10 B03769 US6066724-A Cancer
    HLICR58 B08775 WO200052160-A1 Cancer
    HMCIS41 B08776 WO200052160-A1 Cancer
    HCESA34 B08891 WO200017222-A1 Cancer
    HCRMZ90 B08892 WO200017222-A1 Cancer
    HDPXQ54 B08893 WO200017222-A1 Immune/Hematopoietic
    HETCL11 B08894 WO200017222-A1 Cancer
    HFXDN34 B08895 WO200017222-A1 Neural/Sensory
    HKAAV24 B08896 WO200017222-A1 Cancer
    HMTBE31 B08897 WO200017222-A1 Cancer
    HRADL70 B08898 WO200017222-A1 Excretory,
    Immune/Hematopoietic
    HTXGG31 B08899 WO200017222-A1 Cancer
    HWHHL34 B08900 WO200017222-A1 Cancer
    HYAAY40 B08901 WO200017222-A1 Immune/Hematopoietic
    HPASA81 B08902 WO200017222-A1 Digestive,
    Endocrine, Reproductive
    HCNDA61 B08903 WO200017222-A1 Digestive, Reproductive
    HTHCZ41 B08904 WO200017222-A1 Cancer
    HKADJ17 B08905 WO200017222-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HMSII78 B08906 WO200017222-A1 Cancer
    HCFBL76 B08907 WO200017222-A1 Cancer
    HFVHR84 B08908 WO200017222-A1 Connective/Epithelial,
    Digestive
    HIBCB67 B08909 WO200017222-A1 Cancer
    HCELI29 B08910 WO200017222-A1 Cancer
    HAHDZ77 B08911 WO200017222-A1 Cardiovascular,
    Mixed Fetal
    HDHMA45 B08912 WO200017222-A1 Cardiovascular,
    Neural/Sensory
    HELAW45 B08913 WO200017222-A1 Cardiovascular
    HFIAB31 B08914 WO200017222-A1 Cancer
    HLWBK05 B08915 WO200017222-A1 Cancer
    HLDBX13 B08916 WO200017222-A1 Digestive
    HMAGA15 B08917 WO200017222-A1 Cancer
    HMWFT53 B08918 WO200017222-A1 Immune/Hematopoietic
    HNFJD91 B08919 WO200017222-A1 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    HTGCM55 B08920 WO200017222-A1 Cardiovascular,
    Digestive,
    Immune/Hematopoietic
    HTTEX77 B08921 WO200017222-A1 Cancer
    HFXDN34 B08922 WO200017222-A1 Neural/Sensory
    HDPMI18 B08923 WO200017222-A1 Cancer
    HETGL41 B08924 WO200017222-A1 Cancer
    HPASA81 B08925 WO200017222-A1 Digestive,
    Endocrine,
    Reproductive
    HCNDA61 B08926 WO200017222-A1 Digestive,
    Reproductive
    HTTEX77 B08927 WO200017222-A1 Cancer
    HFXDN34 B08934 WO200017222-A1 Neural/Sensory
    HETGL41 B08935 WO200017222-A1 Cancer
    HPASA81 B08936 WO200017222-A1 Digestive,
    Endocrine,
    Reproductive
    HCNDA61 B08940 WO200017222-A1 Digestive,
    Reproductive
    HTTEX77 B08982 WO200017222-A1 Cancer
    HAOAB14 B12301 WO200029422-A1 Digestive, Musculoskeletal
    HHFBY53 B12302 WO200029422-A1 Cancer
    HE2FE69 B12303 WO200029422-A1 Cancer
    HNHFI33 B12305 WO200029422-A1 Immune/Hematopoietic
    HAMFE15 B12306 WO200029422-A1 Cancer
    HAMFE82 B12307 WO200029422-A1 Cancer
    HCWEM59 B12308 WO200029422-A1 Immune/Hematopoietic
    HDPGE10 B12309 WO200029422-A1 Immune/Hematopoietic
    HDPGP94 B12310 WO200029422-A1 Digestive,
    Immune/Hematopoietic
    HFPBY77 B12311 WO200029422-A1 Cancer
    HFXHK32 B12312 WO200029422-A1 Neural/Sensory
    HMTAK05 B12313 WO200029422-A1 Cancer
    HMWDC93 B12314 WO200029422-A1 Immune/Hematopoietic
    HSPBY40 B12315 WO200029422-A1 Cancer
    HODDO08 B12316 WO200029422-A1 Cancer
    HCFNK47 B12317 WO200029422-A1 Cancer
    HE2FL70 B12318 WO200029422-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    H2MBY03 B12319 WO200029422-A1 Cancer
    HACBS38 B12320 WO200029422-A1 Cancer
    HAGFG51 B12321 WO200029422-A1 Neural/Sensory
    HBQAB44 B12322 WO200029422-A1 Neural/Sensory,
    Reproductive,
    Respiratory
    HHEMA59 B12323 WO200029422-A1 Cancer
    HJBAV55 B12324 WO200029422-A1 Cancer
    HLHEY02 B12325 WO200029422-A1 Endocrine,
    Respiratory
    HSAAO94 B12326 WO200029422-A1 Cancer
    HTXKP61 B12327 WO200029422-A1 Cancer
    HWABC21 B12328 WO200029422-A1 Cancer
    HWBDI30 B12329 WO200029422-A1 Cancer
    HYBAR26 B12330 WO200029422-A1 Musculoskeletal
    HAJAF57 B12331 WO200029422-A1 Cancer
    HAMFE15 B12332 WO200029422-A1 Cancer
    HAMFE82 B12333 WO200029422-A1 Cancer
    HAMFE15 B12338 WO200029422-A1 Cancer
    HAMFE82 B12339 WO200029422-A1 Cancer
    HLDOK36 B15551 WO200056752-A2 Cancer
    HDPBW68 B15552 WO200056752-A2 Cancer
    HHEFO24 B15553 WO200056752-A2 Cardiovascular,
    Immune/Hematopoietic,
    Neural/Sensory
    HEGAL46 B15554 WO200056752-A2 Cancer
    HFOYC02 B15555 WO200056752-A2 Cancer
    HDABV82 B15556 WO200056752-A2 Cancer
    HCEMU42 B15557 WO200056752-A2 Cancer
    HSIFO61 B15558 WO200056752-A2 Cancer
    HDPBW68 B15559 WO200056752-A2 Cancer
    HDPBW68 B15562 WO200056752-A2 Cancer
    HSIFO61 B15566 WO200056752-A2 Cancer
    HOEAL47 B18715 WO200054651-A2 Cancer
    HE9SF68 B18755 WO200055204-A1 Cancer
    HLICQ90 B24437 WO200035937-A1 Cancer
    HNTSM04 B24438 WO200035937-A1 Cancer
    HMCAL59 B24439 WO200035937-A1 Cancer
    HMACO04 B24440 WO200035937-A1 Cancer
    HMAHY59 B24441 WO200035937-A1 Cancer
    HFXLL52 B24442 WO200035937-A1 Neural/Sensory
    HKABY55 B24443 WO200035937-A1 Cancer
    HCQCF36 B24444 WO200035937-A1 Digestive,
    Immune/Hematopoietic
    HTADO22 B24445 WO200035937-A1 Immune/Hematopoietic
    HHFHD92 B24446 WO200035937-A1 Cancer
    HNGFW58 B24447 WO200035937-A1 Cancer
    HOEFV61 B24448 WO200035937-A1 Cancer
    HPIAQ68 B24449 WO200035937-A1 Immune/Hematopoietic,
    Reproductive
    HNFFY60 B24450 WO200035937-A1 Cancer
    HCE3H20 B24451 WO200035937-A1 Cancer
    HE8EW79 B24452 WO200035937-A1 Cancer
    HTTDF41 B24453 WO200035937-A1 Cancer
    HSSGJ45 B24454 WO200035937-A1 Cancer
    HLWBY76 B24455 WO200035937-A1 Cancer
    HDPBN34 B24456 WO200035937-A1 Immune/Hematopoietic
    HMSHY73 B24457 WO200035937-A1 Cancer
    HPRBF19 B24458 WO200035937-A1 Cancer
    HNFJE06 B24459 WO200035937-A1 Immune/Hematopoietic,
    Musculoskeletal
    HCHCF61 B24460 WO200035937-A1 Reproductive
    HBJLH40 B24461 WO200035937-A1 Cancer
    HDPMV72 B24462 WO200035937-A1 Cancer
    HEMFA84 B24463 WO200035937-A1 Cancer
    HTOHW95 B24464 WO200035937-A1 Cancer
    HUNAH63 B24465 WO200035937-A1 Reproductive
    HISBT59 B24466 WO200035937-A1 Cancer
    HNTAS52 B24467 WO200035937-A1 Cancer
    HRACM44 B24468 WO200035937-A1 Excretory,
    Immune/Hematopoietic
    HFPES77 B24469 WO200035937-A1 Cancer
    HUSXU29 B24470 WO200035937-A1 Cancer
    HOHBB49 B24471 WO200035937-A1 Musculoskeletal
    HRABX31 B24472 WO200035937-A1 Excretory,
    Immune/Hematopoietic,
    Musculoskeletal
    HROBD68 B24473 WO200035937-A1 Cancer
    HMHBE18 B24474 WO200035937-A1 Cancer
    HNHDY21 B24475 WO200035937-A1 Immune/Hematopoietic
    HOEBZ89 B24476 WO200035937-A1 Cancer
    HYAAJ71 B24477 WO200035937-A1 Immune/Hematopoietic
    HTEKS16 B24478 WO200035937-A1 Connective/Epithelial,
    Mixed Fetal,
    Reproductive
    HCUFX40 B24479 WO200035937-A1 Immune/Hematopoietic
    HCWDL75 B24480 WO200035937-A1 Cardiovascular,
    Immune/Hematopoietic
    HNHKJ57 B24481 WO200035937-A1 Immune/Hematopoietic
    HCMSS06 B24482 WO200035937-A1 Cancer
    HIBCE35 B24483 WO200035937-A1 Cancer
    HE8EW79 B24484 WO200035937-A1 Cancer
    HTTDF41 B24485 WO200035937-A1 Cancer
    HSSGJ45 B24486 WO200035937-A1 Cancer
    HCMSS06 B24487 WO200035937-A1 Cancer
    HCMSS06 B24597 WO200035937-A1 Cancer
    HAOAB64 B25576 WO200029435-A1 Musculoskeletal,
    Reproductive
    HOHCH55 B25577 WO200029435-A1 Cancer
    HTLEW81 B25578 WO200029435-A1 Cancer
    HARAO44 B25579 WO200029435-A1 Neural/Sensory
    HDPCL05 B25580 WO200029435-A1 Immune/Hematopoietic
    HDPUW68 B25581 WO200029435-A1 Cancer
    HOHBY69 B25582 WO200029435-A1 Cancer
    HCDDP40 B25583 WO200029435-A1 Immune/Hematopoietic,
    Musculoskeletal
    HUSAQ05 B25585 WO200029435-A1 Cancer
    HOUDJ81 B25586 WO200029435-A1 Cancer
    HPWCM76 B25587 WO200029435-A1 Reproductive
    HOHCH55 B25588 WO200029435-A1 Cancer
    HDPCL05 B25589 WO200029435-A1 Immune/Hematopoietic
    HOHBY69 B25590 WO200029435-A1 Cancer
    HUSAQ05 B25592 WO200029435-A1 Cancer
    HOUDJ81 B25593 WO200029435-A1 Cancer
    HUSAQ05 B25618 WO200029435-A1 Cancer
    HE8NG02 B25665 WO200043495-A2 Mixed Fetal,
    Reproductive
    HWBDM37 B25666 WO200043495-A2 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HODFN71 B25668 WO200043495-A2 Mixed Fetal,
    Reproductive
    HODGE68 B25669 WO200043495-A2 Reproductive
    HCECN54 B25670 WO200043495-A2 Excretory,
    Neural/Sensory
    HE8UB86 B25671 WO200043495-A2 Cancer
    HNHKY10 B25672 WO200043495-A2 Immune/Hematopoietic
    HNHLB93 B25673 WO200043495-A2 Immune/Hematopoietic
    HNHON23 B25674 WO200043495-A2 Immune/Hematopoietic,
    Musculoskeletal
    HTEPG70 B25675 WO200043495-A2 Reproductive
    HNHOJ75 B25676 WO200043495-A2 Immune/Hematopoietic
    HDTIT10 B25677 WO200043495-A2 Cancer
    HKAOS84 B25678 WO200043495-A2 Connective/Epithelial
    HAPUC89 B25679 WO200043495-A2 Cancer
    HWAAD63 B25680 WO200043495-A2 Endocrine,
    Excretory,
    Immune/Hematopoietic
    HUCPD31 B25681 WO200043495-A2 Cancer
    HDQHD03 B25682 WO200043495-A2 Immune/Hematopoietic,
    Neural/Sensory
    HKAKK09 B25683 WO200043495-A2 Connective/Epithelial,
    Digestive,
    Mixed Fetal
    HOCNF19 B25684 WO200043495-A2 Digestive
    HTLIT32 B25685 WO200043495-A2 Reproductive
    HODEJ32 B25686 WO200043495-A2 Reproductive
    HNHMV54 B25687 WO200043495-A2 Immune/Hematopoietic
    HODEE95 B25688 WO200043495-A2 Reproductive
    HLHAM10 B25689 WO200043495-A2 Cancer
    HNHOG73 B25690 WO200043495-A2 Immune/Hematopoietic
    HBGNM47 B25691 WO200043495-A2 Cancer
    HAUBA08 B25692 WO200043495-A2 Cancer
    HYBBE75 B25693 WO200043495-A2 Musculoskeletal
    HTLGY87 B25694 WO200043495-A2 Cancer
    HNHPD10 B25695 WO200043495-A2 Immune/Hematopoietic
    HODEI83 B25696 WO200043495-A2 Reproductive
    HMUAI20 B25697 WO200043495-A2 Cancer
    HE9OW20 B25698 WO200043495-A2 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HDTIT10 B25699 WO200043495-A2 Cancer
    HWAAD63 B25700 WO200043495-A2 Endocrine,
    Excretory,
    Immune/Hematopoietic
    HWAAD63 B25701 WO200043495-A2 Endocrine,
    Excretory,
    Immune/Hematopoietic
    HEMCV19 B25703 WO200043495-A2 Cancer
    HEMCV19 B25704 WO200043495-A2 Cancer
    HEMCV19 B25705 WO200043495-A2 Cancer
    HAUBA08 B25706 WO200043495-A2 Cancer
    HEMCV19 B25707 WO200043495-A2 Cancer
    HE9OW20 B25715 WO200043495-A2 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HT4SB02 B27560 WO200055175-A1 Immune/Hematopoietic
    HCHAC68 B27562 WO200055175-A1 Cancer
    HCHCA79 B27563 WO200055175-A1 Digestive,
    Neural/Sensory,
    Reproductive
    HCHMY57 B27564 WO200055175-A1 Cancer
    HCHOY52 B27566 WO200055175-A1 Cancer
    HCHQB93 B27567 WO200055175-A1 Cancer
    HCMSA37 B27568 WO200055175-A1 Cardiovascular
    HCMSX51 B27570 WO200055175-A1 Cancer
    HCNAI74 B27571 WO200055175-A1 Digestive
    HCPAE41 B27578 WO200055175-A1 Cancer
    HCQAQ47 B27580 WO200055175-A1 Cancer
    HCQBH72 B27581 WO200055175-A1 Digestive,
    Excretory,
    Immune/Hematopoietic
    HCQDD32 B27585 WO200055175-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HCQDT67 B27586 WO200055175-A1 Cancer
    HCRAY10 B27587 WO200055175-A1 Cancer
    HCRBI79 B27589 WO200055175-A1 Cancer
    HNFAD50 B27591 WO200055175-A1 Cancer
    HCRNF78 B27592 WO200055175-A1 Cancer
    HCUAF85 B27594 WO200055175-A1 Immune/Hematopoietic
    HCUBM41 B27598 WO200055175-A1 Immune/Hematopoietic
    HCUBN69 B27599 WO200055175-A1 Immune/Hematopoietic
    HCUDD64 B27602 WO200055175-A1 Cancer
    HCUEC55 B27604 WO200055175-A1 Immune/Hematopoietic
    HCUFC77 B27607 WO200055175-A1 Cancer
    HBJBR40 B27686 WO200055201-A1 Immune/Hematopoietic
    HBJCH46 B27687 WO200055201-A1 Immune/Hematopoietic,
    Musculoskeletal
    HBJFU30 B27698 WO200055201-A1 Cancer
    HBJAY14 B27704 WO200055201-A1 Immune/Hematopoietic
    HBJND04 B27708 WO200055201-A1 Cancer
    HBKEA94 B27711 WO200055201-A1 Cancer
    HBJDS79 B27712 WO200055201-A1 Cancer
    HBKEI41 B27713 WO200055201-A1 Endocrine,
    Mixed Fetal,
    Reproductive
    HBJHO83 B27720 WO200055201-A1 Immune/Hematopoietic,
    Reproductive
    HBMCT40 B27721 WO200055201-A1 Cancer
    HBMTX26 B27724 WO200055201-A1 Immune/Hematopoietic
    HBMTY48 B27725 WO200055201-A1 Immune/Hematopoietic,
    Reproductive
    HBMUD59 B27726 WO200055201-A1 Cancer
    HBMUI10 B27727 WO200055201-A1 Cancer
    HCEEU18 B27794 WO200055199-A1 Cancer
    HCDCB03 B27795 WO200055199-A1 Cancer
    HCE1G78 B27797 WO200055199-A1 Cancer
    HCDEB19 B27799 WO200055199-A1 Cancer
    HCEDR26 B27801 WO200055199-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HCDBW67 B27804 WO200055199-A1 Cancer
    HCDDX81 B27808 WO200055199-A1 Musculoskeletal
    HBZAI75 B27809 WO200055199-A1 Digestive,
    Reproductive
    HCDEN46 B27810 WO200055199-A1 Cancer
    HCE1D45 B27811 WO200055199-A1 Cancer
    HCE1Y27 B27813 WO200055199-A1 Digestive,
    Neural/Sensory,
    Reproductive
    HCE2I23 B27816 WO200055199-A1 Neural/Sensory
    HCE2P90 B27817 WO200055199-A1 Neural/Sensory
    HCE3A54 B27818 WO200055199-A1 Neural/Sensory
    HCE3D89 B27819 WO200055199-A1 Endocrine,
    Neural/Sensory
    HCE3N23 B27820 WO200055199-A1 Cancer
    HCE4T64 B27821 WO200055199-A1 Cancer
    HCE5J64 B27823 WO200055199-A1 Digestive,
    Neural/Sensory
    HCECO77 B27824 WO200055199-A1 Cancer
    HCEDH42 B27825 WO200055199-A1 Neural/Sensory
    HCEDJ05 B27826 WO200055199-A1 Neural/Sensory
    HCEEE79 B27829 WO200055199-A1 Neural/Sensory
    HCEFH31 B27837 WO200055199-A1 Cancer
    HCDDL48 B27838 WO200055199-A1 Musculoskeletal
    HFVIC33 B27908 WO200055171-A1 Cancer
    HEMAH05 B27909 WO200055171-A1 Cancer
    HHSBI65 B27911 WO200055171-A1 Cancer
    HLEAA24 B27917 WO200055171-A1 Immune/Hematopoietic
    HPTTQ91 B27919 WO200055171-A1 Cancer
    HPMGY89 B27923 WO200055171-A1 Cancer
    H2LAO03 B27933 WO200055171-A1 Cancer
    H2MBA76 B27937 WO200055171-A1 Cancer
    H2MBF60 B27938 WO200055171-A1 Cancer
    H6BSF56 B27939 WO200055171-A1 Cancer
    H6BSM88 B27940 WO200055171-A1 Cancer
    H6EEU40 B27941 WO200055171-A1 Cancer
    HACAB68 B27943 WO200055171-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HACBA04 B27945 WO200055171-A1 Cancer
    HACBJ11 B27946 WO200055171-A1 Cancer
    HACBS86 B27947 WO200055171-A1 Cancer
    HACBT91 B27948 WO200055171-A1 Cancer
    HADAB60 B27951 WO200055171-A1 Cancer
    HADDE71 B27956 WO200055171-A1 Cancer
    HAGFU31 B28014 WO200055177-A2 Neural/Sensory
    HAPBR13 B28017 WO200055177-A2 Cancer
    HAQBG57 B28020 WO200055177-A2 Cancer
    HARAE26 B28022 WO200055177-A2 Neural/Sensory
    HAHEM51 B28032 WO200055177-A2 Cardiovascular
    HAICL90 B28035 WO200055177-A2 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HAMFC67 B28038 WO200055177-A2 Cancer
    HAPBU09 B28041 WO200055177-A2 Cancer
    HAPNL62 B28043 WO200055177-A2 Cancer
    HAPNY10 B28045 WO200055177-A2 Cancer
    HAPQU71 B28048 WO200055177-A2 Cancer
    HARAT69 B28054 WO200055177-A2 Cancer
    HAGEU26 B28059 WO200055177-A2 Neural/Sensory
    HANGB24 B28060 WO200055177-A2 Cancer
    HNGER85 B28286 WO200058355-A1 Immune/Hematopoietic
    HNGET33 B28287 WO200058355-A1 Immune/Hematopoietic
    HNGFA25 B28292 WO200058355-A1 Immune/Hematopoietic
    HNGFG04 B28297 WO200058355-A1 Immune/Hematopoietic
    HNGFG74 B28298 WO200058355-A1 Immune/Hematopoietic
    HNGFI21 B28301 WO200058355-A1 Cancer
    HNGFM31 B28302 WO200058355-A1 Immune/Hematopoietic
    HCUCK44 B28303 WO200058355-A1 Cancer
    HNGFQ18 B28304 WO200058355-A1 Immune/Hematopoietic
    HNGFR54 B28305 WO200058355-A1 Immune/Hematopoietic
    HNGGF13 B28309 WO200058355-A1 Immune/Hematopoietic
    HNGGL11 B28311 WO200058355-A1 Immune/Hematopoietic
    HNGGP65 B28312 WO200058355-A1 Immune/Hematopoietic
    HNGHM47 B28316 WO200058355-A1 Immune/Hematopoietic
    HNGIH40 B28318 WO200058355-A1 Immune/Hematopoietic
    HNGIM83 B28321 WO200058355-A1 Immune/Hematopoietic
    HNGIS27 B28322 WO200058355-A1 Immune/Hematopoietic
    HADFB84 B28707 WO200055198-A1 Cancer
    HADFD01 B28708 WO200055198-A1 Cancer
    HADFK11 B28709 WO200055198-A1 Connective/Epithelial
    HADFT44 B28710 WO200055198-A1 Connective/Epithelial,
    Mixed Fetal,
    Neural/Sensory
    HADGD93 B28714 WO200055198-A1 Cardiovascular,
    Connective/Epithelial
    HAFBB15 B28716 WO200055198-A1 Cancer
    HAGAB62 B28718 WO200055198-A1 Cancer
    HAGAF75 B28720 WO200055198-A1 Digestive,
    Neural/Sensory
    HAGAZ36 B28722 WO200055198-A1 Neural/Sensory
    H2CBH91 B28725 WO200055198-A1 Cancer
    HAGBV29 B28730 WO200055198-A1 Immune/Hematopoietic,
    Neural/Sensory
    HAGCH67 B28732 WO200055198-A1 Neural/Sensory
    HAGCI69 B28733 WO200055198-A1 Neural/Sensory,
    Reproductive
    HAGCT33 B28734 WO200055198-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HAGDG84 B28737 WO200055198-A1 Immune/Hematopoietic,
    Neural/Sensory
    HAGDO70 B28743 WO200055198-A1 Cancer
    HAGDW68 B28746 WO200055198-A1 Endocrine,
    Neural/Sensory
    HAGEG10 B28748 WO200055198-A1 Cancer
    HAGEK37 B28750 WO200055198-A1 Cancer
    HTADO61 B29803 WO200061779-A1 Cancer
    HELDH39 B29805 WO200061779-A1 Cancer
    HSLCV16 B29806 WO200061779-A1 Cancer
    HTOJL95 B29807 WO200061779-A1 Cancer
    HOEBK60 B29810 WO200061779-A1 Cancer
    HSAWN53 B29813 WO200061779-A1 Immune/Hematopoietic
    HORBS82 B29814 WO200061779-A1 Cancer
    HORBV76 B29815 WO200061779-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HTEHA56 B29822 WO200061779-A1 Cancer
    HMWER46 B29828 WO200061779-A1 Cancer
    HPRAD30 B29829 WO200061779-A1 Cancer
    HTLDD89 B29832 WO200061779-A1 Reproductive
    HMCJC19 B29835 WO200061779-A1 Immune/Hematopoietic
    HROBW46 B29841 WO200061779-A1 Digestive,
    Immune/Hematopoietic
    HSOBB94 B29842 WO200061779-A1 Cancer
    HSOBP75 B29844 WO200061779-A1 Cancer
    HNHKS19 B29848 WO200061779-A1 Immune/Hematopoietic,
    Reproductive
    HNHKV56 B29849 WO200061779-A1 Immune/Hematopoietic
    HWLIL31 B29850 WO200061779-A1 Cancer
    HMVBC31 B32002 WO200058350-A1 Cancer
    HMVBC84 B32003 WO200058350-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HMWAO82 B32006 WO200058350-A1 Immune/Hematopoietic
    HMWBK35 B32008 WO200058350-A1 Cancer
    HHENT16 B32010 WO200058350-A1 Cancer
    HMWEF46 B32020 WO200058350-A1 Immune/Hematopoietic
    HMWEX02 B32022 WO200058350-A1 Cancer
    HKGCK41 B32027 WO200058350-A1 Cancer
    HMWHR36 B32029 WO200058350-A1 Immune/Hematopoietic
    HMWIQ26 B32031 WO200058350-A1 Cancer
    HMWIU49 B32032 WO200058350-A1 Cancer
    HMWJJ64 B32035 WO200058350-A1 Cancer
    HNEAK38 B32040 WO200058350-A1 Immune/Hematopoietic
    HNECD52 B32043 WO200058350-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNECL75 B32045 WO200058350-A1 Cancer
    HNECW49 B32046 WO200058350-A1 Immune/Hematopoietic
    HNEDA05 B32048 WO200058350-A1 Immune/Hematopoietic
    HETKD92 B32371 WO200047602-A1 Cancer
    HNTSN12 B32372 WO200047602-A1 Cancer
    HMQBV64 B32373 WO200047602-A1 Cancer
    HTELM16 B32374 WO200047602-A1 Reproductive
    HSDFJ26 B32375 WO200047602-A1 Cancer
    HNGND37 B32376 WO200047602-A1 Cancer
    HWBDV80 B32377 WO200047602-A1 Cancer
    HDPOR60 B32378 WO200047602-A1 Cancer
    HWBFY57 B32379 WO200047602-A1 Digestive,
    Immune/Hematopoietic
    HNHOL24 B32380 WO200047602-A1 Immune/Hematopoietic
    HWHIB26 B32381 WO200047602-A1 Cancer
    HHAAF20 B32382 WO200047602-A1 Cancer
    HNHNE04 B32383 WO200047602-A1 Immune/Hematopoietic
    HSAAO65 B32384 WO200047602-A1 Cancer
    HTENO07 B32385 WO200047602-A1 Reproductive
    HTLHI35 B32386 WO200047602-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HTLHY14 B32387 WO200047602-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTXKY63 B32388 WO200047602-A1 Immune/Hematopoietic
    HOUDC53 B32389 WO200047602-A1 Cancer
    HWLGV78 B32390 WO200047602-A1 Cancer
    HCGLB30 B32391 WO200047602-A1 Cancer
    HTELP17 B32392 WO200047602-A1 Cancer
    HFXBS43 B32393 WO200047602-A1 Neural/Sensory
    HNGOM56 B32394 WO200047602-A1 Immune/Hematopoietic
    HTXON32 B32395 WO200047602-A1 Immune/Hematopoietic
    HDQHO40 B32396 WO200047602-A1 Cancer
    HKBAB11 B32397 WO200047602-A1 Immune/Hematopoietic
    HRAAN56 B32398 WO200047602-A1 Excretory,
    Reproductive
    HFIDS78 B32399 WO200047602-A1 Connective/Epithelial,
    Digestive,
    Musculoskeletal
    HSDIB20 B32400 WO200047602-A1 Cancer
    HHEPU04 B32401 WO200047602-A1 Cancer
    HNGKN89 B32402 WO200047602-A1 Immune/Hematopoietic
    HE9TH18 B32403 WO200047602-A1 Cancer
    HHFHM89 B32404 WO200047602-A1 Cancer
    HASAV70 B32405 WO200047602-A1 Cancer
    HSDFJ26 B32406 WO200047602-A1 Cancer
    HODAA16 B32407 WO200047602-A1 Cancer
    HODAA16 B32408 WO200047602-A1 Cancer
    HGBIB74 B32409 WO200047602-A1 Cancer
    HCRMU04 B32410 WO200047602-A1 Cancer
    HSAAO65 B32411 WO200047602-A1 Cancer
    HSAAO65 B32412 WO200047602-A1 Cancer
    HSMBB92 B32413 WO200047602-A1 Cancer
    HHEPU04 B32414 WO200047602-A1 Cancer
    HLDNA86 B32415 WO200047602-A1 Cancer
    HE9TH18 B32416 WO200047602-A1 Cancer
    HHFHM33 B32447 WO200047602-A1 Cancer
    HUUAV63 B32481 WO200047602-A1 Cancer
    HE2CJ53 B33721 WO200056753-A1 Cancer
    HE2HF76 B33724 WO200056753-A1 Cancer
    HDTDE66 B33729 WO200056753-A1 Cancer
    HDTKJ29 B33735 WO200056753-A1 Cancer
    HDTLM18 B33736 WO200056753-A1 Immune/Hematopoietic
    HE2AI94 B33740 WO200056753-A1 Cancer
    HE2BD72 B33741 WO200056753-A1 Cancer
    HE2CH58 B33744 WO200056753-A1 Digestive,
    Mixed Fetal
    HE2NW57 B33753 WO200056753-A1 Mixed Fetal
    HE2PJ56 B33755 WO200056753-A1 Cancer
    HE2PO93 B33756 WO200056753-A1 Cancer
    HE6AU52 B33757 WO200056753-A1 Mixed Fetal
    HAEAV42 B33758 WO200056753-A1 Cancer
    HE2AT61 B33759 WO200056753-A1 Cancer
    HE2CK47 B33761 WO200056753-A1 Cancer
    HE2DJ84 B33763 WO200056753-A1 Cancer
    HE2CJ53 B33770 WO200056753-A1 Cancer
    HSHAS72 B33832 WO200056753-A1 Cancer
    HEMDR05 B33845 WO200056881-A1 Cardiovascular,
    Digestive,
    Immune/Hematopoietic
    HADXA10 B33846 WO200056881-A1 Cancer
    HEOMF59 B33847 WO200056881-A1 Immune/Hematopoietic
    HEONP08 B33854 WO200056881-A1 Immune/Hematopoietic
    HEPAD15 B33855 WO200056881-A1 Endocrine,
    Reproductive
    HEIAC52 B33860 WO200056881-A1 Cancer
    HEQAP92 B33862 WO200056881-A1 Cancer
    HEQBM94 B33865 WO200056881-A1 Cancer
    HETAA62 B33870 WO200056881-A1 Cancer
    HETEY67 B33873 WO200056881-A1 Connective/Epithelial,
    Reproductive
    HETDP76 B33874 WO200056881-A1 Cancer
    HEQBF89 B33875 WO200056881-A1 Reproductive
    HETIN36 B33877 WO200056881-A1 Cancer
    HFAUA23 B33881 WO200056881-A1 Cancer
    HFCAG75 B33882 WO200056881-A1 Cancer
    HFCAQ17 B33883 WO200056881-A1 Cancer
    HFCDN13 B33887 WO200056881-A1 Cancer
    HFCDT67 B33888 WO200056881-A1 Cancer
    HFCEI04 B33891 WO200056881-A1 Neural/Sensory
    HTSGY89 B33946 WO200056881-A1 Cancer
    HFCAQ17 B33947 WO200056881-A1 Cancer
    HGLAH86 B33965 WO200056765-A1 Immune/Hematopoietic
    HHEBP28 B33971 WO200056765-A1 Cancer
    HHEMC55 B33973 WO200056765-A1 Immune/Hematopoietic
    HHEMM20 B33974 WO200056765-A1 Immune/Hematopoietic
    HHEMP35 B33976 WO200056765-A1 Cancer
    HHEMZ08 B33977 WO200056765-A1 Cancer
    HHENR74 B33980 WO200056765-A1 Immune/Hematopoietic
    HHEOK77 B33983 WO200056765-A1 Cancer
    HHEQI04 B33986 WO200056765-A1 Connective/Epithelial,
    Excretory,
    Immune/Hematopoietic
    HHFBA31 B33987 WO200056765-A1 Cancer
    HHFFF87 B33992 WO200056765-A1 Cancer
    HHFFL34 B33993 WO200056765-A1 Cancer
    HHFFS40 B33994 WO200056765-A1 Cancer
    HHGBF91 B34005 WO200056765-A1 Cancer
    HE9NB82 B34092 WO200056755-A1 Cancer
    HEAAC21 B34095 WO200056755-A1 Cancer
    HEAAM54 B34100 WO200056755-A1 Reproductive
    HEAAU28 B34102 WO200056755-A1 Reproductive
    HEBAT05 B34104 WO200056755-A1 Cancer
    HEBCN80 B34107 WO200056755-A1 Neural/Sensory
    HEBCY54 B34108 WO200056755-A1 Cancer
    HEBDW31 B34111 WO200056755-A1 Cancer
    HEBFL36 B34112 WO200056755-A1 Neural/Sensory
    HEBGE07 B34114 WO200056755-A1 Neural/Sensory
    HEBGE23 B34115 WO200056755-A1 Cancer
    HEGAI20 B34119 WO200056755-A1 Reproductive
    HEBCI18 B34121 WO200056755-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HEBDF90 B34125 WO200056755-A1 Cancer
    HELDK79 B34127 WO200056755-A1 Cardiovascular
    HELEL76 B34130 WO200056755-A1 Cancer
    HELFO30 B34131 WO200056755-A1 Cancer
    HEMCJ80 B34138 WO200056755-A1 Cancer
    HDPAR73 B34201 WO200056755-A1 Cancer
    HDPAR73 B34202 WO200056755-A1 Cancer
    HATBI94 B34222 WO200055352-A2 Cancer
    HATCB45 B34224 WO200055352-A2 Endocrine,
    Immune/Hematopoietic
    HATDW05 B34229 WO200055352-A2 Endocrine
    HATEH20 B34231 WO200055352-A2 Cancer
    HAWBA65 B34233 WO200055352-A2 Cancer
    HBBBA42 B34236 WO200055352-A2 Cancer
    HBBBE83 B34238 WO200055352-A2 Cancer
    HBBMA11 B34239 WO200055352-A2 Neural/Sensory
    HBCGE46 B34244 WO200055352-A2 Musculoskeletal
    HBGML95 B34249 WO200055352-A2 Reproductive
    HBHAA05 B34251 WO200055352-A2 Neural/Sensory
    HBHAA53 B34252 WO200055352-A2 Neural/Sensory
    HBIAA59 B34253 WO200055352-A2 Cancer
    HBICW51 B34262 WO200055352-A2 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HFCET43 B34299 WO200056883-A1 Cancer
    HFEAG55 B34302 WO200056883-A1 Cancer
    HFEAY59 B34304 WO200056883-A1 Connective/Epithelial
    HFFAV61 B34308 WO200056883-A1 Neural/Sensory
    HFGAN63 B34312 WO200056883-A1 Cancer
    HFICH70 B34316 WO200056883-A1 Musculoskeletal
    HFIHQ57 B34317 WO200056883-A1 Musculoskeletal,
    Reproductive
    HFIHZ75 B34318 WO200056883-A1 Cancer
    HFIJA29 B34321 WO200056883-A1 Cancer
    HFIJD81 B34322 WO200056883-A1 Cancer
    HFIUK66 B34324 WO200056883-A1 Cancer
    HFIXC39 B34326 WO200056883-A1 Cancer
    HFIXC69 B34327 WO200056883-A1 Cancer
    HLWAU42 B34329 WO200056883-A1 Cancer
    HFIZF51 B34331 WO200056883-A1 Musculoskeletal
    HFKDX53 B34333 WO200056883-A1 Cancer
    HFKEG63 B34335 WO200056883-A1 Excretory
    HFKES05 B34336 WO200056883-A1 Cancer
    HFKES35 B34337 WO200056883-A1 Cancer
    HFKEU12 B34338 WO200056883-A1 Excretory
    HFOYR54 B34344 WO200056883-A1 Cancer
    HFPBJ64 B34347 WO200056883-A1 Musculoskeletal,
    Neural/Sensory
    HFXBV67 B34441 WO200056767-A1 Digestive,
    Neural/Sensory
    HFXBY20 B34442 WO200056767-A1 Neural/Sensory
    HFXGT51 B34462 WO200056767-A1 Neural/Sensory
    HFXGW04 B34463 WO200056767-A1 Cancer
    HFXHL83 B34466 WO200056767-A1 Neural/Sensory
    HFXJB21 B34468 WO200056767-A1 Neural/Sensory
    HFXJN93 B34469 WO200056767-A1 Neural/Sensory
    HFXJT53 B34470 WO200056767-A1 Cancer
    HFXLK91 B34472 WO200056767-A1 Cancer
    HFXHM49 B34473 WO200056767-A1 Neural/Sensory
    HGBDV35 B34474 WO200056767-A1 Cancer
    HEPCU48 B34476 WO200056767-A1 Cancer
    HGBGN34 B34478 WO200056767-A1 Connective/Epithelial,
    Digestive,
    Reproductive
    HGBGX31 B34479 WO200056767-A1 Cancer
    HGBHP91 B34482 WO200056767-A1 Digestive
    HCEFN51 B34580 WO200056751-A1 Cancer
    HCEFZ82 B34581 WO200056751-A1 Cancer
    HCEJL08 B34585 WO200056751-A1 Cancer
    HCENN67 B34588 WO200056751-A1 Digestive,
    Endocrine,
    Neural/Sensory
    HCEPC90 B34592 WO200056751-A1 Neural/Sensory
    HCETL19 B34597 WO200056751-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCFAT42 B34602 WO200056751-A1 Immune/Hematopoietic
    HCFAT66 B34603 WO200056751-A1 Immune/Hematopoietic
    HCFBM77 B34605 WO200056751-A1 Immune/Hematopoietic
    HCFLJ52 B34611 WO200056751-A1 Cancer
    HCFLP48 B34613 WO200056751-A1 Immune/Hematopoietic
    HCFLQ12 B34614 WO200056751-A1 Cancer
    HCFMA39 B34617 WO200056751-A1 Immune/Hematopoietic
    HCFML07 B34619 WO200056751-A1 Cancer
    HCFMX88 B34620 WO200056751-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCFNN16 B34623 WO200056751-A1 Cancer
    HCFNN75 B34624 WO200056751-A1 Cancer
    HLYBI48 B34774 WO200058356-A1 Immune/Hematopoietic
    HFKES35 B34777 WO200058356-A1 Cancer
    HLYBU15 B34781 WO200058356-A1 Immune/Hematopoietic
    HLYDX01 B34789 WO200058356-A1 Cancer
    HLYEA60 B34790 WO200058356-A1 Cancer
    HLYEU59 B34793 WO200058356-A1 Immune/Hematopoietic
    HLYGE16 B34794 WO200058356-A1 Cancer
    HLYGV19 B34795 WO200058356-A1 Cancer
    HLYGY91 B34796 WO200058356-A1 Cancer
    HMADJ14 B34797 WO200058356-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HMAGF01 B34798 WO200058356-A1 Cancer
    HMDAB44 B34806 WO200058356-A1 Neural/Sensory
    HMDAE88 B34808 WO200058356-A1 Neural/Sensory
    HMDAG62 B34809 WO200058356-A1 Cancer
    HMDAK20 B34810 WO200058356-A1 Neural/Sensory
    HMECM77 B34815 WO200058356-A1 Cardiovascular
    HMECQ59 B34816 WO200058356-A1 Cancer
    HMEEZ07 B34821 WO200058356-A1 Cardiovascular,
    Reproductive
    HDPCV29 B34856 WO200056766-A1 Immune/Hematopoietic
    HDPCW16 B34857 WO200056766-A1 Cancer
    HDPFG13 B34859 WO200056766-A1 Cancer
    HDPFU43 B34860 WO200056766-A1 Cancer
    HDPFZ05 B34861 WO200056766-A1 Immune/Hematopoietic,
    Neural/Sensory.
    HDPGA84 B34862 WO200056766-A1 Cancer
    HDPIH25 B34864 WO200056766-A1 Cancer
    HDPKC55 B34866 WO200056766-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HDPNC21 B34868 WO200056766-A1 Cancer
    HDPOL37 B34869 WO200056766-A1 Immune/Hematopoietic,
    Reproductive
    HDPSZ07 B34871 WO200056766-A1 Immune/Hematopoietic
    HDPXN20 B34872 WO200056766-A1 Immune/Hematopoietic
    HDQGN08 B34874 WO200056766-A1 Immune/Hematopoietic
    HDPGE24 B34877 WO200056766-A1 Cancer
    HDPJV53 B34878 WO200056766-A1 Immune/Hematopoietic
    HDPOC24 B34881 WO200056766-A1 Cancer
    HDPPC19 B34883 WO200056766-A1 Immune/Hematopoietic
    HBCAO31 B34886 WO200056766-A1 Cancer
    HDQGD06 B34889 WO200056766-A1 Cancer
    HDRAA17 B34890 WO200056766-A1 Cancer
    HDRAC68 B34891 WO200056766-A1 Cancer
    HDSAH37 B34893 WO200056766-A1 Connective/Epithelial
    HDSAP15 B34896 WO200056766-A1 Cancer
    HDTAS57 B34897 WO200056766-A1 Cancer
    HDPFU43 B34916 WO200056766-A1 Cancer
    HDPKC55 B34932 WO200056766-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HPRCB54 B36696 WO200071150-A1 Cancer
    HMQAT69 B37348 WO200058335-A1 Cancer
    HMQBL90 B37349 WO200058335-A1 Digestive,
    Immune/Hematopoietic
    HMQCX41 B37354 WO200058335-A1 Immune/Hematopoietic
    HMQDU07 B37356 WO200058335-A1 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HMSDI67 B37365 WO200058335-A1 Digestive,
    Immune/Hematopoietic
    HMSHC86 B37372 WO200058335-A1 Immune/Hematopoietic
    HMSII36 B37376 WO200058335-A1 Immune/Hematopoietic
    HMSIT42 B37377 WO200058335-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HMSKQ91 B37381 WO200058335-A1 Immune/Hematopoietic
    HMTAT36 B37384 WO200058335-A1 Cancer
    HMUBK53 B37390 WO200058335-A1 Cancer
    HMUBO15 B37392 WO200058335-A1 Cancer
    HMUBZ15 B37393 WO200058335-A1 Cancer
    HMSLF15 B37984 WO200055371-A1 Cancer
    HKAET41 B37985 WO200055371-A1 Connective/Epithelial,
    Digestive,
    Reproductive
    HE9RJ42 B37987 WO200055371-A1 Mixed Fetal
    HDPAS92 B37988 WO200055371-A1 Cancer
    HATDF29 B37989 WO200055371-A1 Cancer
    HWLHH15 B37990 WO200055371-A1 Digestive
    HBXFL29 B37991 WO200055371-A1 Cancer
    HKGBF67 B37992 WO200055371-A1 Cancer
    HWHGP71 B37993 WO200055371-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HLWCU38 B37994 WO200055371-A1 Cancer
    HMTAX46 B37995 WO200055371-A1 Cancer
    HIBEU15 B37996 WO200055371-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPQV66 B37997 WO200055371-A1 Cancer
    HFXGW52 B37998 WO200055371-A1 Neural/Sensory
    HHEQR55 B37999 WO200055371-A1 Immune/Hematopoietic
    HNHNW84 B38000 WO200055371-A1 Immune/Hematopoietic
    HAJBX74 B38001 WO200055371-A1 Cancer
    HCUGE72 B38002 WO200055371-A1 Cancer
    HTEQI22 B38003 WO200055371-A1 Cancer
    HDPYE41 B38004 WO200055371-A1 Immune/Hematopoietic
    HDTII23 B38005 WO200055371-A1 Immune/Hematopoietic
    HAMFL84 B38007 WO200055371-A1 Cancer
    HTELW37 B38008 WO200055371-A1 Reproductive
    HNGOU56 B38009 WO200055371-A1 Immune/Hematopoietic
    HOUHD63 B38010 WO200055371-A1 Cancer
    HPJCX13 B38011 WO200055371-A1 Cancer
    HNHCT15 B38012 WO200055371-A1 Cancer
    HKGBF67 B38013 WO200055371-A1 Cancer
    HWHGP71 B38014 WO200055371-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HTEQI22 B38016 WO200055371-A1 Cancer
    HJBCI01 B38017 WO200055371-A1 Cancer
    HTSFV18 B38018 WO200055371-A1 Cancer
    HPJBF63 B38019 WO200055371-A1 Cancer
    HWHGP71 B38044 WO200055371-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HNEDU46 B38119 WO200058468-A2 Cancer
    HNFDY31 B38124 WO200058468-A2 Cancer
    HNFEA17 B38125 WO200058468-A2 Cancer
    HNFET12 B38127 WO200058468-A2 Immune/Hematopoietic
    HNFGR08 B38129 WO200058468-A2 Immune/Hematopoietic
    HNFGW37 B38130 WO200058468-A2 Immune/Hematopoietic
    HNFGW53 B38131 WO200058468-A2 Cancer
    HNFHA34 B38132 WO200058468-A2 Cancer
    HNFJE27 B38137 WO200058468-A2 Immune/Hematopoietic
    HNGAM58 B38143 WO200058468-A2 Immune/Hematopoietic
    HNGAT83 B38144 WO200058468-A2 Immune/Hematopoietic
    HNGBE63 B38148 WO200058468-A2 Immune/Hematopoietic
    HNGBH53 B38149 WO200058468-A2 Immune/Hematopoietic
    HNGBJ74 B38150 WO200058468-A2 Immune/Hematopoietic
    HNGBQ61 B38152 WO200058468-A2 Immune/Hematopoietic
    HNGBW25 B38154 WO200058468-A2 Immune/Hematopoietic
    HNGCF64 B38156 WO200058468-A2 Immune/Hematopoietic
    HNGDH22 B38158 WO200058468-A2 Immune/Hematopoietic
    HNGDQ38 B38161 WO200058468-A2 Immune/Hematopoietic
    HNGDR39 B38162 WO200058468-A2 Immune/Hematopoietic
    HNGEA34 B38165 WO200058468-A2 Digestive,
    Immune/Hematopoietic
    HTOJB02 B38205 WO200058469-A1 Immune/Hematopoietic
    HSDEA26 B38207 WO200058469-A1 Neural/Sensory
    HATCF80 B38209 WO200058469-A1 Cancer
    HTOAK03 B38215 WO200058469-A1 Cancer
    HSLAB11 B38216 WO200058469-A1 Cancer
    HSJAU93 B38218 WO200058469-A1 Cancer
    HSHBT15 B38221 WO200058469-A1 Cancer
    HSLCS31 B38234 WO200058469-A1 Cancer
    HSLCS34 B38235 WO200058469-A1 Cancer
    HOBNF51 B38237 WO200058469-A1 Cancer
    HSFAM19 B38242 WO200058469-A1 Cancer
    HNHEY29 B38245 WO200058469-A1 Immune/Hematopoietic
    HTHDB20 B38248 WO200058469-A1 Immune/Hematopoietic
    HPMGM06 B38250 WO200058469-A1 Digestive,
    Neural/Sensory,
    Reproductive
    HDPMA04 B38321 WO200061623-A1 Immune/Hematopoietic
    HEMFQ46 B38322 WO200061623-A1 Cancer
    HKAJK47 B38324 WO200061623-A1 Cancer
    HCGMF16 B38325 WO200061623-A1 Cancer
    HMSGU01 B38326 WO200061623-A1 Cancer
    HNTCE26 B38327 WO200061623-A1 Cancer
    HPTTI70 B38328 WO200061623-A1 Cancer
    HNSAD53 B38329 WO200061623-A1 Digestive
    HTEBV72 B38330 WO200061623-A1 Reproductive
    HCE3Z61 B38331 WO200061623-A1 Cancer
    HSSGD52 B38332 WO200061623-A1 Cancer
    HAPSA79 B38333 WO200061623-A1 Cancer
    HASAU84 B38334 WO200061623-A1 Cancer
    HLWEA51 B38335 WO200061623-A1 Cancer
    HNFIZ34 B38336 WO200061623-A1 Cancer
    HTELS08 B38337 WO200061623-A1 Reproductive
    HTLEJ24 B38338 WO200061623-A1 Cancer
    HCEHF62 B38339 WO200061623-A1 Cancer
    HUFBY15 B38340 WO200061623-A1 Digestive,
    Musculoskeletal,
    Reproductive
    HELHD85 B38341 WO200061623-A1 Cancer
    HOFNY91 B38342 WO200061623-A1 Cancer
    HEGAK44 B38343 WO200061623-A1 Cancer
    HETBA14 B38344 WO200061623-A1 Cancer
    HBAFV19 B38345 WO200061623-A1 Cancer
    HTXDO17 B38346 WO200061623-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Respiratory
    HE8DS15 B38347 WO200061623-A1 Cancer
    HLDOW79 B38348 WO200061623-A1 Cardiovascular,
    Digestive
    HOFND85 B38349 WO200061623-A1 Cancer
    HBIBU30 B38350 WO200061623-A1 Cancer
    HODFG71 B38351 WO200061623-A1 Reproductive
    HNHGE28 B38352 WO200061623-A1 Cancer
    HYASD09 B38355 WO200061623-A1 Cancer
    HDPCL63 B38356 WO200061623-A1 Cancer
    HBDAD07 B38357 WO200061623-A1 Immune/Hematopoietic
    HTOHG09 B38361 WO200061623-A1 Cancer
    HWBFX31 B38362 WO200061623-A1 Cancer
    HLHDP16 B38363 WO200061623-A1 Cancer
    HSDBC88 B38364 WO200061623-A1 Cancer
    HOVBX78 B38365 WO200061623-A1 Cancer
    HWADJ89 B38367 WO200061623-A1 Immune/Hematopoietic
    HYABE50 B38368 WO200061623-A1 Cancer
    HSJAQ17 B38369 WO200061623-A1 Cancer
    HCUGM86 B38370 WO200061623-A1 Immune/Hematopoietic
    HLDQC46 B38371 WO200061623-A1 Cancer
    HOFOA59 B38372 WO200061623-A1 Reproductive
    HFABG18 B38373 WO200061623-A1 Cancer
    HNHLY33 B38374 WO200061623-A1 Immune/Hematopoietic
    HFCFJ18 B38375 WO200061623-A1 Cancer
    HANGG89 B38376 WO200061623-A1 Cancer
    HNHOD46 B38377 WO200061623-A1 Immune/Hematopoietic
    HLYBI58 B38379 WO200061623-A1 Cancer
    HAJBG14 B38381 WO200061623-A1 Cancer
    HE9NN84 B38382 WO200061623-A1 Cancer
    HAPSA79 B38383 WO200061623-A1 Cancer
    HAPSA79 B38384 WO200061623-A1 Cancer
    HTLEJ24 B38385 WO200061623-A1 Cancer
    HEGAK44 B38386 WO200061623-A1 Cancer
    HTXDO17 B38387 WO200061623-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Respiratory
    HBIBB20 B38388 WO200061623-A1 Cancer
    HSIDL71 B38389 WO200061623-A1 Cancer
    HOVBX78 B38390 WO200061623-A1 Cancer
    HYABE50 B38391 WO200061623-A1 Cancer
    HFCFJ18 B38392 WO200061623-A1 Cancer
    HPRAL78 B38394 WO200061623-A1 Cancer
    HCE5F84 B38395 WO200061623-A1 Cancer
    HAMHE82 B38396 WO200061623-A1 Cancer
    HACBZ59 B38472 WO200061623-A1 Cancer
    HACBZ59 B38475 WO200061623-A1 Cancer
    HWLQU40 B38514 WO200061623-A1 Cancer
    HFKFI35 B38527 WO200056882-A1 Excretory
    HFPCZ55 B38529 WO200056882-A1 Cancer
    HFPDR39 B38533 WO200056882-A1 Cancer
    HFPDX08 B38536 WO200056882-A1 Cancer
    HFRAB10 B38539 WO200056882-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HFSBE94 B38541 WO200056882-A1 Immune/Hematopoietic
    HFTAR30 B38545 WO200056882-A1 Cancer
    HFTBB50 B38546 WO200056882-A1 Cancer
    HFSAY91 B38551 WO200056882-A1 Cancer
    HFSBC10 B38552 WO200056882-A1 Immune/Hematopoietic,
    Mixed Fetal
    HFTDK11 B38555 WO200056882-A1 Cancer
    HFVHW43 B38560 WO200056882-A1 Digestive
    HFXBI64 B38567 WO200056882-A1 Neural/Sensory
    HFXBL05 B38568 WO200056882-A1 Mixed Fetal,
    Neural/Sensory
    HHGBV02 B38971 WO200056880-A1 Immune/Hematopoietic,
    Reproductive
    HHGBW55 B38972 WO200056880-A1 Immune/Hematopoietic,
    Reproductive
    HHGDI12 B38976 WO200056880-A1 Neural/Sensory
    HHPBG90 B38983 WO200056880-A1 Cancer
    HHPFP26 B38987 WO200056880-A1 Cancer
    HHPGU74 B38990 WO200056880-A1 Neural/Sensory
    HHPEB61 B38991 WO200056880-A1 Cancer
    HHSBJ92 B39002 WO200056880-A1 Cancer
    HHPSE03 B39004 WO200056880-A1 Neural/Sensory
    HHSCQ67 B39005 WO200056880-A1 Cancer
    HHSDB43 B39007 WO200056880-A1 Cancer
    HHTLH79 B39015 WO200056880-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HIABC70 B39016 WO200056880-A1 Cancer
    HIBCR82 B39017 WO200056880-A1 Mixed Fetal,
    Neural/Sensory
    HIBEC45 B39019 WO200056880-A1 Cancer
    HILCA24 B39020 WO200056880-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HLHBS54 B39093 WO200058513-A1 Cancer
    HLMCT95 B39098 WO200058513-A1 Cancer
    HLDRU08 B39100 WO200058513-A1 Cancer
    HLDXF43 B39101 WO200058513-A1 Cancer
    HLEAA10 B39102 WO200058513-A1 Immune/Hematopoietic
    HLHCB33 B39104 WO200058513-A1 Digestive,
    Reproductive,
    Respiratory
    HLHCF14 B39105 WO200058513-A1 Connective/Epithelial,
    Respiratory
    HLHCN51 B39107 WO200058513-A1 Digestive,
    Immune/Hematopoietic,
    Respiratory
    HLHDM38 B39109 WO200058513-A1 Cancer
    HLHEX62 B39111 WO200058513-A1 Excretory,
    Immune/Hematopoietic,
    Respiratory
    HLHSG15 B39114 WO200058513-A1 Cancer
    HLIBD74 B39117 WO200058513-A1 Digestive
    HLICO10 B39120 WO200058513-A1 Cancer
    HLJBI22 B39121 WO200058513-A1 Cancer
    HLLAX95 B39123 WO200058513-A1 Immune/Hematopoietic
    HLMBZ14 B39127 WO200058513-A1 Immune/Hematopoietic
    HLMDH01 B39129 WO200058513-A1 Immune/Hematopoietic
    HLMDU23 B39130 WO200058513-A1 Immune/Hematopoietic
    HLMFU53 B39133 WO200058513-A1 Cancer
    HLMHG68 B39135 WO200058513-A1 Cancer
    HLMIM84 B39137 WO200058513-A1 Cancer
    HLMIQ83 B39139 WO200058513-A1 Immune/Hematopoietic
    HLDRT09 B39140 WO200058513-A1 Cancer
    HE9EA10 B39181 WO200056754-A1 Cancer
    HE6CS65 B39182 WO200056754-A1 Cancer
    HE8BE20 B39190 WO200056754-A1 Cancer
    HE8BT58 B39193 WO200056754-A1 Cancer
    HE8CA13 B39195 WO200056754-A1 Cancer
    HE8FC10 B39201 WO200056754-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HE8FG24 B39202 WO200056754-A1 Cancer
    HE8FL24 B39203 WO200056754-A1 Mixed Fetal
    HE8MA27 B39204 WO200056754-A1 Cancer
    HE8MG56 B39205 WO200056754-A1 Mixed Fetal
    HE8QU21 B39208 WO200056754-A1 Immune/Hematopoietic,
    Mixed Fetal
    HE8UX34 B39210 WO200056754-A1 Mixed Fetal
    HE9CY05 B39216 WO200056754-A1 Mixed Fetal
    HE9DG54 B39217 WO200056754-A1 Cancer
    HE9DZ47 B39218 WO200056754-A1 Endocrine,
    Immune/Hematopoietic,
    Mixed Fetal
    HE8EX86 B39226 WO200056754-A1 Cancer
    HMEIH57 B39310 WO200057903-A2 Cardiovascular,
    Immune/Hematopoietic
    HFEBA88 B39312 WO200057903-A2 Cancer
    HMEKW44 B39317 WO200057903-A2 Cardiovascular,
    Immune/Hematopoietic,
    Neural/Sensory
    HMELM75 B39318 WO200057903-A2 Cancer
    HMIAC52 B39322 WO200057903-A2 Cancer
    HMIAL39 B39325 WO200057903-A2 Cancer
    HMIBD93 B39329 WO200057903-A2 Cancer
    HMIBE95 B39330 WO200057903-A2 Neural/Sensory
    HMMAL32 B39344 WO200057903-A2 Immune/Hematopoietic
    HMMBK55 B39348 WO200057903-A2 Immune/Hematopoietic
    HMMBR63 B39350 WO200057903-A2 Cancer
    HMMBS55 B39351 WO200057903-A2 Immune/Hematopoietic,
    Reproductive
    HMMBT47 B39352 WO200057903-A2 Immune/Hematopoietic
    HMMCD35 B39353 WO200057903-A2 Immune/Hematopoietic
    HFKCZ13 B39362 WO200057903-A2 Cancer
    HFKCZ13 B39363 WO200057903-A2 Cancer
    HOAAL10 B39402 WO200058340-A2 Musculoskeletal
    HTXCV44 B39406 WO200058340-A2 Immune/Hematopoietic,
    Neural/Sensory
    HTXDJ75 B39407 WO200058340-A2 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HSIDZ25 B39410 WO200058340-A2 Cancer
    HTXEN33 B39413 WO200058340-A2 Immune/Hematopoietic,
    Reproductive
    HJPDK61 B39419 WO200058340-A2 Cancer
    HNHBM16 B39420 WO200058340-A2 Immune/Hematopoietic,
    Neural/Sensory
    HNHDE58 B39422 WO200058340-A2 Cancer
    HTTDT67 B39425 WO200058340-A2 Cancer
    HTLEP55 B39427 WO200058340-A2 Cancer
    HCUBA28 B39430 WO200058340-A2 Cancer
    HSNAT08 B39433 WO200058340-A2 Cancer
    HTOEV01 B39437 WO200058340-A2 Immune/Hematopoietic,
    Reproductive
    HSQBF66 B39439 WO200058340-A2 Cancer
    HSJBY32 B39445 WO200058340-A2 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HPMCV30 B39447 WO200058340-A2 Cancer
    HPMDF45 B39448 WO200058340-A2 Excretory,
    Immune/Hematopoietic,
    Reproductive
    HPIBI40 B40153 WO200058496-A1 Cancer
    HRDBA20 B40154 WO200058496-A1 Musculoskeletal
    HTJMA64 B40156 WO200058496-A1 Cancer
    HTHDF86 B40157 WO200058496-A1 Immune/Hematopoietic
    HRDBA20 B40158 WO200058496-A1 Musculoskeletal
    HSAAN03 B40161 WO200058496-A1 Cancer
    HRABZ80 B40162 WO200058496-A1 Excretory,
    Immune/Hematopoietic,
    Musculoskeletal
    HPDDQ28 B40163 WO200058496-A1 Endocrine,
    Musculoskeletal
    HOSBX14 B40166 WO200058496-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HCRAI29 B40168 WO200058496-A1 Neural/Sensory
    HSAXI10 B40171 WO200058496-A1 Digestive,
    Immune/Hematopoietic
    HTSFV18 B40172 WO200058496-A1 Cancer
    HNHGK22 B40174 WO200058496-A1 Immune/Hematopoietic
    HPMFP48 B40176 WO200058496-A1 Cancer
    HUSGL67 B40177 WO200058496-A1 Cancer
    HOFMO16 B40180 WO200058496-A1 Reproductive
    HPMAI31 B40182 WO200058496-A1 Cancer
    HTTAP37 B40184 WO200058496-A1 Immune/Hematopoietic,
    Reproductive
    HSABA15 B40187 WO200058496-A1 Cancer
    HPEAA65 B40188 WO200058496-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HSSAN03 B40189 WO200058496-A1 Cancer
    HTTDG27 B40194 WO200058496-A1 Reproductive
    HTPDV75 B40195 WO200058496-A1 Digestive,
    Reproductive
    HMJAC12 B40197 WO200058496-A1 Neural/Sensory
    HTLEJ24 B40198 WO200058496-A1 Cancer
    HBAMC47 B43415 WO200055350-A1 Excretory
    HBGDH11 B43571 WO200055350-A1 Cancer
    HCHAK72 B43572 WO200055350-A1 Cancer
    HSRAB84 B43594 WO200055350-A1 Cancer
    HMWBH91 B43612 WO200055350-A1 Cancer
    HUFFG07 B43619 WO200055350-A1 Cancer
    HMEIY69 B43625 WO200055350-A1 Cancer
    HIBCN93 B43651 WO200055350-A1 Cancer
    HDPQE64 B43656 WO200055350-A1 Cancer
    HSYCY88 B43723 WO200055350-A1 Cancer
    HCMSD61 B43779 WO200055350-A1 Cancer
    H2CBK94 B43801 WO200055350-A1 Digestive,
    Neural/Sensory,
    Respiratory
    HE9NK60 B43812 WO200055350-A1 Cancer
    HTXDT74 B43844 WO200055350-A1 Cancer
    HTTKN45 B43848 WO200055350-A1 Cancer
    HSSGC06 B43861 WO200055350-A1 Cancer
    HE8AM92 B43893 WO200055350-A1 Cancer
    HWLHZ28 B43933 WO200055350-A1 Cancer
    HCEDM42 B43935 WO200055350-A1 Cancer
    HMWFM73 B43952 WO200055350-A1 Cancer
    HCHBQ27 B44028 WO200055350-A1 Reproductive
    HCHBQ27 B44029 WO200055350-A1 Reproductive
    HCEGS49 B44338 WO200058358-A1 Connective/Epithelial,
    Neural/Sensory,
    Reproductive
    HTODH57 B44349 WO200058358-A1 Immune/Hematopoietic
    HRTAR24 B44351 WO200058358-A1 Digestive,
    Immune/Hematopoietic
    HSLAW59 B44355 WO200058358-A1 Immune/Hematopoietic,
    Musculoskeletal
    HODAG07 B44358 WO200058358-A1 Reproductive
    HODBA45 B44360 WO200058358-A1 Reproductive
    HODBD79 B44361 WO200058358-A1 Immune/Hematopoietic,
    Reproductive
    HODDG72 B44363 WO200058358-A1 Cancer
    HSVAE42 B44367 WO200058358-A1 Connective/Epithelial,
    Neural/Sensory
    HNHAG83 B44369 WO200058358-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Musculoskeletal
    HSIDA39 B44374 WO200058358-A1 Digestive
    HYBAW56 B44375 WO200058358-A1 Musculoskeletal
    HSATI91 B44376 WO200058358-A1 Immune/Hematopoietic
    HPWAO89 B44379 WO200058358-A1 Immune/Hematopoietic,
    Reproductive
    HOUCD12 B44380 WO200058358-A1 Connective/Epithelial
    HWTAM38 B44381 WO200058358-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HKGCE62 B44597 WO200058339-A2 Immune/Hematopoietic
    HKGAK45 B44601 WO200058339-A2 Musculoskeletal,
    Reproductive
    HKGBC33 B44602 WO200058339-A2 Immune/Hematopoietic
    HKGBH54 B44606 WO200058339-A2 Cancer
    HKGCK41 B44608 WO200058339-A2 Cancer
    HKGCX05 B44610 WO200058339-A2 Cancer
    HKGDA95 B44611 WO200058339-A2 Cancer
    HKIME53 B44612 WO200058339-A2 Cancer
    HKIXR91 B44616 WO200058339-A2 Cancer
    HKMLT89 B44625 WO200058339-A2 Excretory,
    Immune/Hematopoietic,
    Reproductive
    HKMMU76 B44629 WO200058339-A2 Cancer
    HL3AE69 B44635 WO200058339-A2 Cancer
    HLDBV54 B44638 WO200058339-A2 Cancer
    HLDNF18 B44639 WO200058339-A2 Cancer
    HLDOL74 B44642 WO200058339-A2 Cancer
    HKIMG23 B44643 WO200058339-A2 Cancer
    HLDBV18 B44645 WO200058339-A2 Cancer
    HMQBU44 B44647 WO200058339-A2 Cancer
    HNHDM21 B44703 WO200058494-A1 Immune/Hematopoietic
    HNHDM43 B44704 WO200058494-A1 Immune/Hematopoietic
    HNHDX28 B44708 WO200058494-A1 Immune/Hematopoietic
    HNHEG30 B44713 WO200058494-A1 Immune/Hematopoietic
    HNHEL22 B44715 WO200058494-A1 Immune/Hematopoietic
    HNHFF60 B44720 WO200058494-A1 Immune/Hematopoietic
    HTHDH18 B44723 WO200058494-A1 Immune/Hematopoietic
    HTHDP65 B44724 WO200058494-A1 Cancer
    HTHDT25 B44725 WO200058494-A1 Immune/Hematopoietic
    HTGAQ29 B44727 WO200058494-A1 Immune/Hematopoietic
    HTGAS70 B44728 WO200058494-A1 Cancer
    HNHGD95 B44731 WO200058494-A1 Cardiovascular,
    Immune/Hematopoietic
    HNHGR82 B44733 WO200058494-A1 Immune/Hematopoietic
    HSAYL24 B44763 WO200058336-A1 Immune/Hematopoietic
    HTGDS92 B44764 WO200058336-A1 Cancer
    HTWDJ17 B44767 WO200058336-A1 Cancer
    HNGJL07 B44776 WO200058336-A1 Immune/Hematopoietic,
    Neural/Sensory
    HSSGS62 B44785 WO200058336-A1 Musculoskeletal,
    Reproductive
    HOVCC73 B44786 WO200058336-A1 Immune/Hematopoietic,
    Reproductive
    HSAYO82 B44788 WO200058336-A1 Endocrine,
    Immune/Hematopoietic
    HMACT74 B44791 WO200058336-A1 Immune/Hematopoietic
    HMIAD75 B44792 WO200058336-A1 Neural/Sensory
    HUFAO92 B44795 WO200058336-A1 Digestive,
    Reproductive
    HTWFA21 B44799 WO200058336-A1 Immune/Hematopoietic
    HSDKE82 B44802 WO200058336-A1 Neural/Sensory
    HMAJS26 B44803 WO200058336-A1 Cancer
    HKGAP57 B44811 WO200058336-A1 Immune/Hematopoietic
    HCWUW24 B44831 WO200055176-A2 Immune/Hematopoietic
    HDPAE80 B44832 WO200055176-A2 Cancer
    HCUFE33 B44833 WO200055176-A2 Immune/Hematopoietic
    HCUFJ09 B44834 WO200055176-A2 Cancer
    HCUGR26 B44840 WO200055176-A2 Immune/Hematopoietic
    HCUHM71 B44844 WO200055176-A2 Immune/Hematopoietic,
    Musculoskeletal
    HCWAK88 B44847 WO200055176-A2 Immune/Hematopoietic
    HCWFK03 B44855 WO200055176-A2 Cancer
    HCWHT52 B44858 WO200055176-A2 Immune/Hematopoietic
    HCWKO32 B44859 WO200055176-A2 Immune/Hematopoietic
    HCWUF93 B44861 WO200055176-A2 Cancer
    HDACJ52 B44865 WO200055176-A2 Cancer
    HDFAB86 B44866 WO200055176-A2 Mixed Fetal,
    Neural/Sensory
    HDHAA55 B44870 WO200055176-A2 Immune/Hematopoietic,
    Neural/Sensory
    HDHEB12 B44871 WO200055176-A2 Immune/Hematopoietic,
    Neural/Sensory
    HDHIA16 B44874 WO200055176-A2 Cancer
    HDHIA26 B44875 WO200055176-A2 Neural/Sensory
    HARNB17 B44909 WO200055176-A2 Cancer
    HBKEE60 B44917 WO200055200-A1 Digestive
    HBMWJ92 B44920 WO200055200-A1 Cancer
    HBMXW83 B44924 WO200055200-A1 Cancer
    HCE3R01 B44926 WO200055200-A1 Cancer
    HBNBJ76 B44930 WO200055200-A1 Cancer
    HBQAC72 B44935 WO200055200-A1 Neural/Sensory
    HBSAJ63 B44937 WO200055200-A1 Cancer
    HATDE03 B44938 WO200055200-A1 Cancer
    HBSDD24 B44939 WO200055200-A1 Cancer
    HBWBD25 B44940 WO200055200-A1 Immune/Hematopoietic,
    Neural/Sensory
    HBXBM24 B44943 WO200055200-A1 Neural/Sensory
    HBXBM78 B44944 WO200055200-A1 Cancer
    HBNAX40 B44947 WO200055200-A1 Cancer
    HBXCQ03 B44949 WO200055200-A1 Cancer
    HBXFI33 B44955 WO200055200-A1 Immune/Hematopoietic,
    Neural/Sensory
    HBXGE12 B44958 WO200055200-A1 Cancer
    HCDAA24 B44960 WO200055200-A1 Cancer
    HCDAH02 B44962 WO200055200-A1 Immune/Hematopoietic,
    Musculoskeletal
    HCDAR40 B44964 WO200055200-A1 Cardiovascular,
    Immune/Hematopoietic,
    Musculoskeletal
    HCDBE76 B44965 WO200055200-A1 Cancer
    HCDBO32 B44966 WO200055200-A1 Cancer
    HTDAF68 B45025 WO200058357-A1 Immune/Hematopoietic
    HTWFA88 B45027 WO200058357-A1 Digestive,
    Immune/Hematopoietic
    HSDJV40 B45028 WO200058357-A1 Immune/Hematopoietic,
    Neural/Sensory
    HSDKA64 B45029 WO200058357-A1 Immune/Hematopoietic,
    Neural/Sensory
    HRAAC36 B45030 WO200058357-A1 Excretory,
    Immune/Hematopoietic
    HPRCF50 B45031 WO200058357-A1 Cancer
    HUSXP50 B45034 WO200058357-A1 Cardiovascular,
    Reproductive
    HSSFP88 B45035 WO200058357-A1 Cancer
    HOVBS68 B45037 WO200058357-A1 Cancer
    HOVCO53 B45039 WO200058357-A1 Reproductive
    HOVBI16 B45040 WO200058357-A1 Cancer
    HOSFR35 B45041 WO200058357-A1 Cancer
    HNTRB25 B45042 WO200058357-A1 Cancer
    HUFBP77 B45043 WO200058357-A1 Cancer
    HUFAP33 B45044 WO200058357-A1 Cancer
    HSDHD05 B45045 WO200058357-A1 Neural/Sensory
    HTWFM85 B45046 WO200058357-A1 Cancer
    HSDJC96 B45048 WO200058357-A1 Cancer
    HTXKH40 B45050 WO200058357-A1 Cancer
    HPQCI62 B45051 WO200058357-A1 Cancer
    HOFMJ65 B45053 WO200058357-A1 Cancer
    HMUAN45 B45057 WO200058357-A1 Cancer
    HAJAB88 B45059 WO200058357-A1 Cancer
    HPTTT62 B45067 WO200058357-A1 Cancer
    HSRDW57 B45071 WO200058357-A1 Cancer
    HSRDW57 B45118 WO200058357-A1 Cancer
    HJBCG74 B45121 WO200058467-A1 Cancer
    HCECO77 B45124 WO200058467-A1 Cancer
    HJABC58 B45129 WO200058467-A1 Cancer
    HJMBK59 B45140 WO200058467-A1 Cancer
    HJMBP01 B45141 WO200058467-A1 Cancer
    HJMBW62 B45143 WO200058467-A1 Reproductive
    HJPAQ19 B45146 WO200058467-A1 Cancer
    HJPBN96 B45148 WO200058467-A1 Cancer
    HJPBK28 B45149 WO200058467-A1 Cancer
    HKABN63 B45152 WO200058467-A1 Cancer
    HKAFF50 B45159 WO200058467-A1 Cancer
    HKMSB01 B45166 WO200058467-A1 Cancer
    HISEJ52 B45171 WO200058467-A1 Cancer
    HJBCG74 B45174 WO200058467-A1 Cancer
    HJBCG74 B45175 WO200058467-A1 Cancer
    HOFNY15 B45227 WO200063230-A2 Reproductive
    HNTAF42 B45228 WO200063230-A2 Cancer
    HPJAW78 B45233 WO200063230-A2 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HPJBS16 B45234 WO200063230-A2 Connective/Epithelial,
    Reproductive
    HPJCV35 B45236 WO200063230-A2 Reproductive
    HSNAH56 B45239 WO200063230-A2 Cancer
    HE2FE89 B45246 WO200063230-A2 Cardiovascular,
    Digestive,
    Mixed Fetal
    HPVAF86 B45249 WO200063230-A2 Reproductive
    HOGCD78 B45257 WO200063230-A2 Reproductive
    HRABU56 B45264 WO200063230-A2 Cardiovascular,
    Excretory,
    Musculoskeletal
    HCUBY47 B45267 WO200063230-A2 Digestive,
    Immune/Hematopoietic
    HUDBE20 B45270 WO200063230-A2 Reproductive
    HUDBK47 B45271 WO200063230-A2 Immune/Hematopoietic,
    Reproductive
    HSOAH16 B45318 WO200061628-A1 Digestive
    HWTBX66 B45320 WO200061628-A1 Cancer
    HTXDO17 B45321 WO200061628-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Respiratory
    HSSDQ20 B45325 WO200061628-A1 Musculoskeletal,
    Neural/Sensory
    HTOHM82 B45329 WO200061628-A1 Cancer
    HTOIH51 B45333 WO200061628-A1 Immune/Hematopoietic
    HHLBA86 B45334 WO200061628-A1 Digestive
    HTAEH58 B45335 WO200061628-A1 Immune/Hematopoietic
    HLTCO22 B45338 WO200061628-A1 Cancer
    HTOJS23 B45343 WO200061628-A1 Immune/Hematopoietic
    HNHBE21 B45356 WO200061628-A1 Immune/Hematopoietic
    HSSFE38 B45387 WO200061627-A1 Cancer
    HTOGB79 B45388 WO200061627-A1 Cancer
    HKABU43 B45389 WO200061627-A1 Cancer
    HSYBV44 B45398 WO200061627-A1 Immune/Hematopoietic
    HOHBZ10 B45399 WO200061627-A1 Cancer
    HWAAQ28 B45400 WO200061627-A1 Cancer
    HWBBQ70 B45402 WO200061627-A1 Immune/Hematopoietic,
    Neural/Sensory
    HWBCN36 B45403 WO200061627-A1 Immune/Hematopoietic
    HWBCP16 B45404 WO200061627-A1 Immune/Hematopoietic
    HWHGW09 B45406 WO200061627-A1 Cancer
    HWHHA21 B45407 WO200061627-A1 Connective/Epithelial
    HYABE50 B45408 WO200061627-A1 Cancer
    HBXFA04 B45411 WO200061627-A1 Neural/Sensory
    HPRCA64 B45412 WO200061627-A1 Cancer
    HTXAA20 B45414 WO200061627-A1 Cancer
    HOFAA78 B45423 WO200061627-A1 Reproductive
    HTOGR38 B45427 WO200061627-A1 Immune/Hematopoietic
    HUKBT67 B45431 WO200061627-A1 Cancer
    HCEMU42 B45432 WO200061627-A1 Cancer
    HWHPB78 B45433 WO200061627-A1 Cancer
    HSYBV44 B45452 WO200061627-A1 Immune/Hematopoietic
    HNHEN70 B45699 WO200071584-A1 Cancer
    HLYBN81 B45700 WO200071584-A1 Cancer
    H7TME50 B45701 WO200071584-A1 Cancer
    HDPWP65 B45702 WO200071584-A1 Cancer
    HDTIE58 B45703 WO200071584-A1 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    H7TPC96 B45704 WO200071584-A1 Cancer
    H7MAD52 B45705 WO200071584-A1 Reproductive
    HYASC03 B49502 WO200070076-A1 Endocrine,
    Immune/Hematopoietic
    HCE1K90 B49503 WO200070076-A1 Cancer
    HNTMH2C B49504 WO200070076-A1 Cancer
    HE8EJ16 B49505 WO200070076-A1 Mixed Fetal,
    Neural/Sensory,
    Reproductive
    HFEBD57 B49506 WO200070076-A1 Cancer
    HOFAD65 B49507 WO200070076-A1 Cancer
    HAPRB43 B49508 WO200070076-A1 Cancer
    HCE1K90 B49509 WO200070076-A1 Cancer
    HE8EJ16 B49510 WO200070076-A1 Mixed Fetal,
    Neural/Sensory,
    Reproductive
    HFEBD57 B49511 WO200070076-A1 Cancer
    HFEBD57 B49512 WO200070076-A1 Cancer
    HOFAD65 B49513 WO200070076-A1 Cancer
    HFPEY75 B49533 WO200061774-A2 Cancer
    HOHEC84 B49534 WO200061774-A2 Immune/Hematopoietic,
    Musculoskeletal
    HFKCD20 B49535 WO200061774-A2 Cancer
    HKMLR17 B49536 WO200061774-A2 Cancer
    HTHCW70 B49537 WO200061774-A2 Cancer
    HOHEC84 B49538 WO200061774-A2 Immune/Hematopoietic,
    Musculoskeletal
    HOUCQ17 B50011 WO200071577-A1 Cancer
    HMADD44 B50378 WO200061614-A2 Cancer
    HDQER52 B50379 WO200061614-A2 Cancer
    HTELM46 B50380 WO200061614-A2 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPUS73 B50381 WO200061614-A2 Cancer
    HFCDT50 B50382 WO200061614-A2 Cancer
    HEMGR64 B50383 WO200061614-A2 Cancer
    HHFDM26 B50384 WO200061614-A2 Cancer
    HTTIA36 B50385 WO200061614-A2 Cancer
    HDQHP22 B50387 WO200061614-A2 Cancer
    HRDCD90 B50388 WO200061614-A2 Cancer
    HEOMG91 B50389 WO200061614-A2 Cancer
    HSLGK66 B50390 WO200061614-A2 Cancer
    HSIFX64 B50391 WO200061614-A2 Cancer
    HETCD80 B50392 WO200061614-A2 Reproductive
    HHSGB09 B50393 WO200061614-A2 Cancer
    HLWBT44 B50394 WO200061614-A2 Cancer
    HTLJG95 B50395 WO200061614-A2 Cancer
    HDPDH32 B50935 WO200073323-A2 Immune/Hematopoietic
    HHFMQ22 B50936 WO200073323-A2 Cancer
    HETCM67 B50937 WO200073323-A2 Cancer
    HWBDU78 B50938 WO200073323-A2 Cancer
    HTXEM16 B50939 WO200073323-A2 Cancer
    HBJEM23 B50940 WO200073323-A2 Cardiovascular,
    Musculoskeletal,
    Reproductive
    H7TMD22 B50941 WO200073323-A2 Neural/Sensory
    HDPDH32 B50942 WO200073323-A2 Immune/Hematopoietic
    HDPDH32 B50943 WO200073323-A2 Immune/Hematopoietic
    HSQAX94 B51382 WO200058495-A1 Cancer
    HTOFA11 B51383 WO200058495-A1 Cancer
    HFFAH01 B51384 WO200058495-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HNHBI65 B51385 WO200058495-A1 Immune/Hematopoietic
    HNHCP14 B51386 WO200058495-A1 Immune/Hematopoietic
    HEAAW54 B51387 WO200058495-A1 Reproductive
    HSRDM56 B51393 WO200058495-A1 Cancer
    HSAXL82 B51397 WO200058495-A1 Immune/Hematopoietic
    HCE3L04 B51398 WO200058495-A1 Neural/Sensory
    HEBGM06 B51402 WO200058495-A1 Cancer
    HTWBO30 B51403 WO200058495-A1 Cancer
    HSSJF26 B51404 WO200058495-A1 Musculoskeletal
    HUKAD46 B51412 WO200058495-A1 Endocrine,
    Immune/Hematopoietic,
    Reproductive
    HPDDT14 B51413 WO200058495-A1 Cancer
    HTEDF78 B51415 WO200058495-A1 Reproductive
    HSUSB73 B51416 WO200058495-A1 Immune/Hematopoietic,
    Reproductive
    HSRAA81 B51417 WO200058495-A1 Cancer
    HCABW10 B51420 WO200058495-A1 Cancer
    HTWAM19 B51422 WO200058495-A1 Immune/Hematopoietic
    HSDZO08 B51620 WO200061620-A1 Cancer
    HSLHX15 B51624 WO200061620-A1 Musculoskeletal
    HSNBM34 B51625 WO200061620-A1 Digestive
    HSWBE76 B51629 WO200061620-A1 Cancer
    HSXAS59 B51630 WO200061620-A1 Neural/Sensory
    HSXAY60 B51631 WO200061620-A1 Cancer
    HTEDX07 B51635 WO200061620-A1 Cancer
    HTEJY20 B51640 WO200061620-A1 Cancer
    HLDQU79 B51645 WO200061620-A1 Cancer
    HTLBT80 B51646 WO200061620-A1 Cancer
    HTEAF65 B51648 WO200061620-A1 Excretory,
    Reproductive
    HTNBJ15 B51649 WO200061620-A1 Cancer
    HOUEP77 B51650 WO200061620-A1 Cancer
    HTOJL95 B51651 WO200061620-A1 Cancer
    HTTDN24 B51653 WO200061620-A1 Cancer
    HTXAD75 B51655 WO200061620-A1 Cancer
    HTXDJ21 B51657 WO200061620-A1 Immune/Hematopoietic
    HKB1E57 B51658 WO200061620-A1 Cancer
    HAQBZ15 B51659 WO200061620-A1 Cancer
    HMWAB92 B51661 WO200061620-A1 Cancer
    HSNBL85 B51663 WO200061620-A1 Cancer
    HWBDJ08 B51664 WO200061620-A1 Cancer
    HYABC84 B51667 WO200061620-A1 Cancer
    HTEDX07 B51682 WO200061620-A1 Cancer
    HOUHQ36 B51724 WO200061625-A1 Connective/Epithelial
    HOUIG92 B51726 WO200061625-A1 Cancer
    HSAZP90 B51728 WO200061625-A1 Immune/Hematopoietic
    HSPAY90 B51730 WO200061625-A1 Cancer
    HWHPU44 B51731 WO200061625-A1 Connective/Epithelial
    HWACZ33 B51734 WO200061625-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HRADA42 B51736 WO200061625-A1 Cancer
    HRADN25 B51737 WO200061625-A1 Cancer
    HRADN25 B51738 WO200061625-A1 Cancer
    HRADT25 B51739 WO200061625-A1 Digestive,
    Excretory
    HRADT25 B51740 WO200061625-A1 Digestive,
    Excretory
    HOHEC84 B51741 WO200061625-A1 Immune/Hematopoietic,
    Musculoskeletal
    HRADU15 B51742 WO200061625-A1 Excretory
    HWDAG96 B51743 WO200061625-A1 Cancer
    HWDAJ01 B51745 WO200061625-A1 Connective/Epithelial
    HMBSF85 B51749 WO200061625-A1 Cancer
    HRGSE38 B51752 WO200061625-A1 Cancer
    HTLBF46 B51755 WO200061625-A1 Cancer
    HSRHB59 B51761 WO200061625-A1 Cancer
    HRDDQ39 B51762 WO200061625-A1 Cancer
    HJPCH08 B51766 WO200061625-A1 Cancer
    HOEBJ70 B51767 WO200061625-A1 Cancer
    HDQER52 B51795 WO200061625-A1 Cancer
    HTLBF63 B51827 WO200061626-A1 Cancer
    HTOAT56 B51828 WO200061626-A1 Cancer
    HSSMY35 B51829 WO200061626-A1 Cancer
    HBHAA81 B51840 WO200061626-A1 Cancer
    HOQBG21 B51841 WO200061626-A1 Cancer
    HTPCG10 B51842 WO200061626-A1 Cancer
    HSHAX04 B51848 WO200061626-A1 Cancer
    HPEAD23 B51850 WO200061626-A1 Cancer
    HODDN21 B51857 WO200061626-A1 Reproductive
    HOABH36 B51859 WO200061626-A1 Cancer
    HOACG07 B51860 WO200061626-A1 Cancer
    HPMDA80 B51864 WO200061626-A1 Cancer
    HSKCQ51 B51865 WO200061626-A1 Cancer
    HRDDS22 B51870 WO200061626-A1 Cancer
    HTXDT72 B51871 WO200061626-A1 Cancer
    HTXDG92 B51873 WO200061626-A1 Cancer
    HTXES13 B51874 WO200061626-A1 Cancer
    HSSEF77 B51875 WO200061626-A1 Cancer
    HTEGH03 B51883 WO200061626-A1 Cancer
    HLMJB64 B51933 WO200058334-A1 Cancer
    HLMNA19 B51936 WO200058334-A1 Cardiovascular,
    Immune/Hematopoietic
    HLQAM30 B51937 WO200058334-A1 Cancer
    HLQCX36 B51942 WO200058334-A1 Digestive
    HLQCY09 B51943 WO200058334-A1 Digestive
    HLQDM47 B51947 WO200058334-A1 Digestive
    HLQDU77 B51948 WO200058334-A1 Cancer
    HLTDA14 B51952 WO200058334-A1 Immune/Hematopoietic
    HLTDK30 B51955 WO200058334-A1 Cancer
    HLTDX04 B51958 WO200058334-A1 Cancer
    HLWAU42 B51964 WO200058334-A1 Cancer
    HLWAW73 B51965 WO200058334-A1 Cancer
    HLWAX50 B51966 WO200058334-A1 Cancer
    HLWBJ93 B51968 WO200058334-A1 Cancer
    HLWCN37 B51970 WO200058334-A1 Cancer
    HLYAL28 B51975 WO200058334-A1 Immune/Hematopoietic
    HFXDR47 B52012 WO200061596-A1 Cancer
    HNHHB10 B52017 WO200061596-A1 Immune/Hematopoietic,
    Reproductive
    HPTRI42 B52019 WO200061596-A1 Cancer
    HTWCE14 B52020 WO200061596-A1 Cancer
    HPTVH59 B52025 WO200061596-A1 Endocrine,
    Neural/Sensory
    HUSGU40 B52027 WO200061596-A1 Cancer
    HUSYG26 B52028 WO200061596-A1 Cancer
    HOVCJ71 B52029 WO200061596-A1 Reproductive
    HSKYR49 B52034 WO200061596-A1 Cancer
    HTWEG06 B52040 WO200061596-A1 Immune/Hematopoietic
    HSDJF04 B52041 WO200061596-A1 Cancer
    HPQAN50 B52044 WO200061596-A1 Reproductive
    HT5FX76 B52051 WO200061596-A1 Cancer
    HT5FX79 B52052 WO200061596-A1 Cancer
    HNTRQ40 B52053 WO200061596-A1 Cancer
    HOUFS04 B52057 WO200061596-A1 Cancer
    HOGAR71 B52059 WO200061596-A1 Cancer
    HOFNB74 B52060 WO200061596-A1 Reproductive
    HOGAR71 B52101 WO200061596-A1 Cancer
    H7TDB54 B52104 WO200061624-A1 Cancer
    HOSEM81 B52105 WO200061624-A1 Cancer
    HTXKF95 B52108 WO200061624-A1 Cancer
    HTGGM44 B52113 WO200061624-A1 Immune/Hematopoietic,
    Musculoskeletal
    HROBJ10 B52114 WO200061624-A1 Cancer
    HTXLC05 B52118 WO200061624-A1 Digestive,
    Immune/Hematopoietic,
    Respiratory
    HTXLC45 B52119 WO200061624-A1 Immune/Hematopoietic
    HNHLD80 B52120 WO200061624-A1 Immune/Hematopoietic
    HNGKT41 B52124 WO200061624-A1 Immune/Hematopoietic
    HNHMP15 B52125 WO200061624-A1 Immune/Hematopoietic
    HNHMY76 B52127 WO200061624-A1 Immune/Hematopoietic,
    Reproductive
    HNHND14 B52129 WO200061624-A1 Immune/Hematopoietic
    HNHOF09 B52131 WO200061624-A1 Immune/Hematopoietic
    HODEM38 B52132 WO200061624-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HNGMW45 B52137 WO200061624-A1 Immune/Hematopoietic
    HNGNK44 B52139 WO200061624-A1 Immune/Hematopoietic
    HTLGL33 B52145 WO200061624-A1 Reproductive
    HTLGY50 B52146 WO200061624-A1 Cancer
    HNGKY94 B52147 WO200061624-A1 Immune/Hematopoietic
    HTXNV66 B52150 WO200061624-A1 Cancer
    HRODG74 B53274 WO200055351-A1 Cancer
    HTTDO45 B53323 WO200055351-A1 Cancer
    HSIFY77 B53335 WO200055351-A1 Cancer
    HWMIW26 B53358 WO200055351-A1 Cancer
    HEAHA84 B53397 WO200055351-A1 Cancer
    HBKDN33 B53414 WO200055351-A1 Cancer
    HKAIL83 B53430 WO200055351-A1 Cancer
    HBMSK08 B53503 WO200055351-A1 Cancer
    HTELE03 B53617 WO200055351-A1 Cancer
    HSWAR63 B53774 WO200055351-A1 Reproductive
    HFXAM85 B54142 WO200055320-A1 Cancer
    HISCO10 B54185 WO200055320-A1 Digestive
    HISBT02 B54226 WO200055320-A1 Digestive
    HNHLV34 B54251 WO200055320-A1 Cancer
    HUSXO71 B54257 WO200055320-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HLWBY67 B54277 WO200055320-A1 Cancer
    HUVDP63 B54282 WO200055320-A1 Cancer
    HSTAH26 B54290 WO200055320-A1 Connective/Epithelial
    HWLXE16 B54305 WO200055320-A1 Digestive
    HDQEG93 B54316 WO200055320-A1 Cancer
    HSLJG12 B54341 WO200055320-A1 Cancer
    HAOSL81 B54358 WO200055320-A1 Cancer
    HOFNH33 B54374 WO200055320-A1 Reproductive
    HAJBV26 B56077 WO200070042-A1 Cancer
    HAPOC74 B56078 WO200070042-A1 Excretory,
    Immune/Hematopoietic,
    Reproductive
    HATEI47 B56079 WO200070042-A1 Endocrine
    HNHGD15 B56080 WO200070042-A1 Immune/Hematopoietic
    HRKAB52 B56081 WO200070042-A1 Cancer
    HKGAT94 B56082 WO200070042-A1 Digestive,
    Reproductive
    HODAH46 B56083 WO200070042-A1 Cancer
    HASCE69 B56084 WO200070042-A1 Cancer
    HBNBE21 B56085 WO200070042-A1 Cancer
    HFLSH80 B56086 WO200070042-A1 Cancer
    HRACM44 B56087 WO200070042-A1 Excretory,
    Immune/Hematopoietic
    HBXFR04 B56090 WO200070042-A1 Neural/Sensory
    HNHFM14 B56094 WO200070042-A1 Cancer
    HEBCM27 B56095 WO200070042-A1 Cancer
    HNHBM80 B56096 WO200070042-A1 Immune/Hematopoietic,
    Reproductive
    HTEAR66 B56098 WO200070042-A1 Reproductive
    HTLDW38 B56099 WO200070042-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTOFD28 B56100 WO200070042-A1 Immune/Hematopoietic
    HFPBW41 B56101 WO200070042-A1 Neural/Sensory
    HTSAC80 B56102 WO200070042-A1 Cancer
    HANGD45 B56103 WO200070042-A1 Musculoskeletal
    HNGJL11 B56106 WO200070042-A1 Immune/Hematopoietic,
    Musculoskeletal
    HYBBE75 B56108 WO200070042-A1 Musculoskeletal
    HCWDS72 B56109 WO200070042-A1 Cancer
    HKPAD17 B56110 WO200070042-A1 Excretory
    HTLGY87 B56114 WO200070042-A1 Cancer
    HULAG01 B56115 WO200070042-A1 Cardiovascular
    HYAAJ71 B56116 WO200070042-A1 Immune/Hematopoietic
    HE9HY07 B56118 WO200070042-A1 Mixed Fetal,
    Reproductive
    HLSAF81 B56121 WO200070042-A1 Cancer
    HNGAU09 B56123 WO200070042-A1 Immune/Hematopoietic
    HTEID16 B56124 WO200070042-A1 Reproductive
    HTWKE60 B56125 WO200070042-A1 Immune/Hematopoietic
    HHLAB61 B56127 WO200070042-A1 Digestive
    HRLMB56 B56128 WO200070042-A1 Cancer
    HCUDW10 B56131 WO200070042-A1 Cancer
    HNGIQ46 B56133 WO200070042-A1 Immune/Hematopoietic
    H7TMD22 B56135 WO200070042-A1 Neural/Sensory
    HHFCJ31 B56136 WO200070042-A1 Cardiovascular,
    Connective/Epithelial,
    Reproductive
    HLDBW08 B56137 WO200070042-A1 Digestive
    H6EEW15 B56139 WO200070042-A1 Cancer
    HNHBM26 B56142 WO200070042-A1 Immune/Hematopoietic,
    Reproductive
    HFIVA74 B56144 WO200070042-A1 Musculoskeletal,
    Reproductive
    HPWAG46 B56145 WO200070042-A1 Cancer
    HBJFM34 B56146 WO200070042-A1 Immune/Hematopoietic
    HRDDS01 B56147 WO200070042-A1 Musculoskeletal
    HGBDH53 B56152 WO200070042-A1 Cancer
    HMKCV28 B56154 WO200070042-A1 Neural/Sensory
    HPLAT69 B56155 WO200070042-A1 Cancer
    HNGBJ27 B56157 WO200070042-A1 Immune/Hematopoietic
    HFXHO83 B56158 WO200070042-A1 Cancer
    HKGAM07 B56159 WO200070042-A1 Digestive,
    Endocrine
    HTXFE73 B56160 WO200070042-A1 Cancer
    HEBBN36 B56163 WO200070042-A1 Cancer
    HJPCY06 B56164 WO200070042-A1 Cancer
    HTEGS19 B56165 WO200070042-A1 Cancer
    HMJAK63 B56166 WO200070042-A1 Neural/Sensory
    HNHGE75 B56170 WO200070042-A1 Immune/Hematopoietic
    HMELA16 B56171 WO200070042-A1 Cardiovascular,
    Immune/Hematopoietic
    HNGAJ15 B56172 WO200070042-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNHHD40 B56173 WO200070042-A1 Cancer
    HFPAA06 B56177 WO200070042-A1 Cancer
    HNGIH43 B56180 WO200070042-A1 Immune/Hematopoietic,
    Reproductive
    HLSAD65 B56182 WO200070042-A1 Cancer
    HMDAK33 B56183 WO200070042-A1 Neural/Sensory
    HNALC70 B56184 WO200070042-A1 Cancer
    HOUCW42 B56185 WO200070042-A1 Connective/Epithelial
    HLMMX46 B56186 WO200070042-A1 Immune/Hematopoietic
    HHSDI68 B56188 WO200070042-A1 Neural/Sensory
    HLMIV11 B56190 WO200070042-A1 Immune/Hematopoietic
    HBMCI50 B56193 WO200070042-A1 Immune/Hematopoietic
    HCRAI47 B56195 WO200070042-A1 Cancer
    HNHEU34 B56198 WO200070042-A1 Immune/Hematopoietic
    HPFCR15 B56199 WO200070042-A1 Digestive,
    Mixed Fetal,
    Reproductive
    HNGJM27 B56201 WO200070042-A1 Immune/Hematopoietic
    HNHEL19 B56202 WO200070042-A1 Immune/Hematopoietic,
    Reproductive
    HGOCD38 B56204 WO200070042-A1 Cancer
    HHGCG53 B56205 WO200070042-A1 Cancer
    HHSBJ93 B56206 WO200070042-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HLHDS79 B56208 WO200070042-A1 Cancer
    HNHEA64 B56209 WO200070042-A1 Immune/Hematopoietic
    HOVAZ13 B56211 WO200070042-A1 Cancer
    HHGBK24 B56214 WO200070042-A1 Cancer
    HILCG67 B56215 WO200070042-A1 Cancer
    HOSFC36 B56218 WO200070042-A1 Cancer
    HKGAT94 B56220 WO200070042-A1 Digestive,
    Reproductive
    HBNBE21 B56221 WO200070042-A1 Cancer
    HFLSH80 B56222 WO200070042-A1 Cancer
    HGBDH53 B56227 WO200070042-A1 Cancer
    HMELA16 B56230 WO200070042-A1 Cardiovascular,
    Immune/Hematopoietic
    HNGAJ15 B56231 WO200070042-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCRAI47 B56232 WO200070042-A1 Cancer
    HNHEL19 B56233 WO200070042-A1 Immune/Hematopoietic,
    Reproductive
    HOSFC36 B56236 WO200070042-A1 Cancer
    HHSBJ93 B56351 WO200070042-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HJAAT54 B56364 WO200055174-A1 Cancer
    HPCQS73 B56431 WO200055174-A1 Cancer
    HPRTL26 B56435 WO200055174-A1 Reproductive
    HPFEA08 B56518 WO200055174-A1 Reproductive
    HTEPF49 B56536 WO200055174-A1 Cancer
    HOFAC67 B56601 WO200055174-A1 Reproductive
    HNTBH70 B56606 WO200055174-A1 Cancer
    HMQAI69 B56630 WO200055174-A1 Cancer
    HDPBI36 B56671 WO200055174-A1 Cancer
    HL2AH06 B56687 WO200055174-A1 Immune/Hematopoietic
    HHEPI21 B56725 WO200055174-A1 Cancer
    HAIBC14 B56739 WO200055174-A1 Cancer
    HDQGM08 B56749 WO200055174-A1 Cancer
    HWHPD31 B56788 WO200055174-A1 Cancer
    HPJCY94 B56791 WO200055174-A1 Musculoskeletal,
    Reproductive
    HDSAK19 B56816 WO200055174-A1 Cancer
    HTTFG83 B56820 WO200055174-A1 Reproductive
    HDPVR90 B56824 WO200055174-A1 Cancer
    HSLJW05 B56860 WO200055174-A1 Cancer
    HHEPE84 B56876 WO200055174-A1 Cancer
    HE2LW65 B56909 WO200055174-A1 Cancer
    HPRTI79 B56925 WO200055174-A1 Cancer
    HEQAN39 B56926 WO200055174-A1 Cancer
    HEMFC70 B56931 WO200055174-A1 Cancer
    HELGM94 B56937 WO200055174-A1 Cancer
    HDHMB78 B56950 WO200055174-A1 Cancer
    HDPQE64 B56987 WO200055174-A1 Cancer
    HDPBQ32 B56996 WO200055174-A1 Cancer
    HBXGB85 B57056 WO200055174-A1 Neural/Sensory
    HBUAC02 B57061 WO200055174-A1 Cancer
    HOCOT88 B57077 WO200055174-A1 Cancer
    HAJAT72 B57101 WO200055174-A1 Reproductive
    HPJAV43 B57106 WO200055174-A1 Immune/Hematopoietic,
    Reproductive,
    Respiratory
    HRODJ79 B57121 WO200055174-A1 Cancer
    HE8FD92 B57128 WO200055174-A1 Cancer
    HETAM53 B57137 WO200055174-A1 Cancer
    HCHBQ27 B57159 WO200055174-A1 Reproductive
    HAPSG03 B58194 WO200055180-A2 Cancer
    HHFFR04 B58240 WO200055180-A2 Cancer
    HCLSC85 B58320 WO200055180-A2 Respiratory
    HPAMC60 B58368 WO200055180-A2 Cancer
    HCE5E24 B58386 WO200055180-A2 Cancer
    HMADJ17 B58396 WO200055180-A2 Cancer
    HADCL25 B58403 WO200055180-A2 Cancer
    HWDAO40 B58434 WO200055180-A2 Cancer
    HRGBG45 B58718 WO200055173-A1 Cancer
    HOFMM27 B58772 WO200055173-A1 Reproductive
    HOFMH95 B58829 WO200055173-A1 Reproductive
    HEMFK40 B58912 WO200055173-A1 Cancer
    HODBC01 B58913 WO200055173-A1 Reproductive
    HOGAV29 B58914 WO200055173-A1 Immune/Hematopoietic,
    Reproductive
    HOFNL25 B58917 WO200055173-A1 Reproductive
    HOFMH12 B58929 WO200055173-A1 Reproductive
    HOFOC33 B58932 WO200055173-A1 Reproductive
    HSIFL06 B58996 WO200055173-A1 Cancer
    HNHBE38 B59469 WO200077173-A1 Cancer
    HOPBP13 B59470 WO200077173-A1 Cancer
    HOUDE92 B59472 WO200077173-A1 Cancer
    HPQAJ27 B59473 WO200077173-A1 Cancer
    HTLEV48 B59476 WO200077173-A1 Reproductive
    HSPAI20 B59478 WO200077173-A1 Digestive,
    Neural/Sensory
    HSPAA89 B59479 WO200077173-A1 Digestive
    HTWEH94 B59482 WO200077173-A1 Immune/Hematopoietic
    HTEGS48 B59490 WO200077173-A1 Reproductive
    HTEIV65 B59491 WO200077173-A1 Reproductive
    HOSEI81 B59494 WO200077173-A1 Digestive,
    Musculoskeletal
    HOEFL74 B59495 WO200077173-A1 Cardiovascular,
    Digestive,
    Musculoskeletal
    HOGAA41 B59497 WO200077173-A1 Cancer
    HOFMT59 B59499 WO200077173-A1 Reproductive
    HSYBD33 B59502 WO200077173-A1 Immune/Hematopoietic
    HTOHO21 B59503 WO200077173-A1 Immune/Hematopoietic
    HTAED89 B59504 WO200077173-A1 Immune/Hematopoietic
    HUSCA09 B60703 WO200076531-A1 Cancer
    HBXCD59 B60705 WO200076531-A1 Immune/Hematopoietic,
    Neural/Sensory
    HDPSZ07 B60706 WO200076531-A1 Immune/Hematopoietic
    HCLCU75 B60712 WO200076531-A1 Respiratory
    HDABR74 B60714 WO200076531-A1 Cancer
    HSSEA64 B60721 WO200076531-A1 Cancer
    HSSJF96 B60722 WO200076531-A1 Musculoskeletal
    HT4FV41 B60724 WO200076531-A1 Cancer
    HBSAJ63 B60725 WO200076531-A1 Cancer
    HTODA92 B60726 WO200076531-A1 Cancer
    HTTCB60 B60727 WO200076531-A1 Cancer
    HTTEO25 B60728 WO200076531-A1 Cancer
    HTXDD61 B60733 WO200076531-A1 Cancer
    HTXJM94 B60734 WO200076531-A1 Cancer
    HWHRC51 B60736 WO200076531-A1 Cancer
    HAGFJ67 B60737 WO200076531-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HBSAK32 B60739 WO200076531-A1 Cancer
    HCHCG33 B60741 WO200076531-A1 Cancer
    HE8FD92 B60747 WO200076531-A1 Cancer
    HSAWB58 B63049 WO200061748-A1 Immune/Hematopoietic
    HCE1T53 B63050 WO200061748-A1 Neural/Sensory
    HODCY44 B63057 WO200061748-A1 Reproductive
    HTEJF31 B63072 WO200061748-A1 Reproductive
    HTPCW21 B63077 WO200061748-A1 Digestive,
    Neural/Sensory
    HPFBA54 B63082 WO200061748-A1 Reproductive
    HSABG81 B63083 WO200061748-A1 Cancer
    HTECB02 B63084 WO200061748-A1 Cancer
    HTEDJ28 B63086 WO200061748-A1 Cancer
    HSLEC18 B63092 WO200061748-A1 Cancer
    HTSGO13 B63095 WO200061748-A1 Cancer
    HTLEM16 B63096 WO200061748-A1 Cancer
    HTXPD86 B63136 WO200061629-A1 Cancer
    HWLGP26 B63137 WO200061629-A1 Cancer
    HAJAY92 B63141 WO200061629-A1 Cancer
    HSYBZ94 B63145 WO200061629-A1 Cancer
    HPRBH85 B63146 WO200061629-A1 Cancer
    HTECE87 B63147 WO200061629-A1 Cancer
    HNHGD95 B63148 WO200061629-A1 Cardiovascular,
    Immune/Hematopoietic
    HNKAA76 B63150 WO200061629-A1 Cancer
    HOABP31 B63152 WO200061629-A1 Cancer
    HOHBC57 B63154 WO200061629-A1 Cancer
    HOSCZ41 B63156 WO200061629-A1 Cancer
    HMVEC89 B63157 WO200061629-A1 Cancer
    HPJBJ51 B63162 WO200061629-A1 Cancer
    HGBBR29 B63164 WO200061629-A1 Cancer
    HPMDD27 B63165 WO200061629-A1 Cancer
    HPRCM72 B63169 WO200061629-A1 Cancer
    HPTRM02 B63171 WO200061629-A1 Cancer
    HPTRW28 B63172 WO200061629-A1 Cancer
    HRAAZ12 B63175 WO200061629-A1 Cancer
    HRDEX93 B63177 WO200061629-A1 Cancer
    HRDFE30 B63178 WO200061629-A1 Cancer
    HBJHT01 B63180 WO200061629-A1 Immune/Hematopoietic,
    Reproductive
    HE8FC45 B64422 WO200077255-A1 Cancer
    HETCI16 B64423 WO200077255-A1 Cancer
    HFCBL53 B64427 WO200077255-A1 Cancer
    HE6GR29 B64429 WO200077255-A1 Cancer
    HBJEA15 B64432 WO200077255-A1 Cancer
    HAPNJ33 B64434 WO200077255-A1 Cancer
    HTXKB57 B64435 WO200077255-A1 Cancer
    HDLAL94 B64438 WO200077255-A1 Cancer
    HFVHX08 B64440 WO200077255-A1 Cancer
    HMVCS92 B64441 WO200077255-A1 Cancer
    HNEDQ02 B64443 WO200077255-A1 Cancer
    HPWTF53 B64450 WO200077255-A1 Cancer
    HRTAP63 B64451 WO200077255-A1 Cancer
    HASAU26 B64453 WO200077255-A1 Cancer
    HPWTF23 B64456 WO200077255-A1 Cancer
    HSLHG78 B64457 WO200077255-A1 Cancer
    HTNBJ15 B64461 WO200077255-A1 Cancer
    HTXJW06 B64463 WO200077255-A1 Cancer
    HUKFV37 B64466 WO200077255-A1 Cancer
    HWBBU75 B64468 WO200077255-A1 Cancer
    HWBEF34 B64469 WO200077255-A1 Immune/Hematopoietic,
    Neural/Sensory
    HDLAL94 B64492 WO200077255-A1 Cancer
    HUKFV37 B64539 WO200077255-A1 Cancer
    HUKFV37 B64540 WO200077255-A1 Cancer
    HMSGU30 B64549 WO200077197-A1 Cancer
    HMSHU20 B64550 WO200077197-A1 Immune/Hematopoietic,
    Reproductive
    HMTAB77 B64551 WO200077197-A1 Cancer
    HMUBX48 B64552 WO200077197-A1 Musculoskeletal,
    Reproductive
    HMWCG28 B64553 WO200077197-A1 Cancer
    HMWFO89 B64555 WO200077197-A1 Cancer
    HMWGM41 B64556 WO200077197-A1 Cancer
    HMWGV85 B64557 WO200077197-A1 Cancer
    HNDAC35 B64558 WO200077197-A1 Cancer
    HNGDN07 B64559 WO200077197-A1 Immune/Hematopoietic,
    Reproductive
    HOFMF63 B64564 WO200077197-A1 Cancer
    HOSEO83 B64567 WO200077197-A1 Cancer
    HPIAL55 B64568 WO200077197-A1 Cancer
    HRAAF59 B64569 WO200077197-A1 Excretory
    HSDIK31 B64571 WO200077197-A1 Cancer
    HSDJG47 B64573 WO200077197-A1 Cancer
    HSOAT44 B64574 WO200077197-A1 Cancer
    HTAEF02 B64578 WO200077197-A1 Immune/Hematopoietic
    HTLCX82 B64579 WO200077197-A1 Cancer
    HADDP51 B64581 WO200077197-A1 Cancer
    HAOAG15 B64584 WO200077197-A1 Cancer
    HATCS79 B64585 WO200077197-A1 Endocrine,
    Immune/Hematopoietic
    HMWGM41 B64604 WO200077197-A1 Cancer
    HOFMF63 B64612 WO200077197-A1 Cancer
    HEMBP72 B64627 WO200077197-A1 Cancer
    HEMBP72 B64628 WO200077197-A1 Cancer
    HAOAG15 B64657 WO200077197-A1 Cancer
    HAOAG15 B64658 WO200077197-A1 Cancer
    HKGDO12 B64667 WO200077237-A1 Cancer
    HLQAD72 B64668 WO200077237-A1 Cancer
    HFXHM93 B64672 WO200077237-A1 Neural/Sensory
    HEBCW57 B64675 WO200077237-A1 Mixed Fetal,
    Neural/Sensory
    HAECD28 B64676 WO200077237-A1 Cancer
    HADXA10 B64677 WO200077237-A1 Cancer
    HELEL76 B64678 WO200077237-A1 Cancer
    HETBJ88 B64680 WO200077237-A1 Cancer
    HETCM67 B64681 WO200077237-A1 Cancer
    HFCBL53 B64682 WO200077237-A1 Cancer
    HFEBK75 B64683 WO200077237-A1 Connective/Epithelial
    HFIIZ61 B64684 WO200077237-A1 Cancer
    HGBDL51 B64688 WO200077237-A1 Cancer
    HGLDA95 B64689 WO200077237-A1 Cancer
    HGLDB06 B64690 WO200077237-A1 Cancer
    HHBEI14 B64691 WO200077237-A1 Cancer
    HLDBG17 B64697 WO200077237-A1 Cancer
    HMCIH27 B64701 WO200077237-A1 Cancer
    HMEKW71 B64702 WO200077237-A1 Cancer
    HNGEA90 B64705 WO200077237-A1 Immune/Hematopoietic
    HNHEN70 B64708 WO200077237-A1 Cancer
    HOAAH51 B64711 WO200077237-A1 Cancer
    HORBL77 B64712 WO200077237-A1 Cancer
    HODAH46 B64713 WO200077237-A1 Cancer
    HMWEM23 B64718 WO200077237-A1 Cancer
    HNEBX72 B64775 WO200077256-A1 Immune/Hematopoietic,
    Neural/Sensory
    H6EEA48 B64776 WO200077256-A1 Cancer
    HACBS22 B64778 WO200077256-A1 Cancer
    HADDP23 B64779 WO200077256-A1 Cancer
    HAGFD75 B64782 WO200077256-A1 Cancer
    HAGFS57 B64783 WO200077256-A1 Cancer
    HAHSD51 B64785 WO200077256-A1 Cancer
    HANKC93 B64787 WO200077256-A1 Musculoskeletal
    HAPSH37 B64789 WO200077256-A1 Cancer
    HATAL05 B64790 WO200077256-A1 Cancer
    HCBAB34 B64799 WO200077256-A1 Cancer
    HCE5H86 B64802 WO200077256-A1 Cancer
    HCEBF54 B64803 WO200077256-A1 Cancer
    HCEDN07 B64804 WO200077256-A1 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HCEGH74 B64805 WO200077256-A1 Cancer
    HCELB04 B64806 WO200077256-A1 Cancer
    HDHEA33 B64813 WO200077256-A1 Cancer
    HDPIF65 B64814 WO200077256-A1 Immune/Hematopoietic
    HDPMC52 B64816 WO200077256-A1 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HDSAO14 B64819 WO200077256-A1 Cancer
    HDTAR39 B64820 WO200077256-A1 Cancer
    HBXFW01 B64844 WO200077256-A1 Neural/Sensory
    HLYEJ14 B64883 WO200076530-A1 Cancer
    HPJCX13 B64888 WO200076530-A1 Cancer
    HFKEV77 B64889 WO200076530-A1 Cancer
    HCEOV48 B64890 WO200076530-A1 Cancer
    HCRMR35 B64891 WO200076530-A1 Cancer
    HDPNJ26 B64893 WO200076530-A1 Cancer
    HDQGD06 B64894 WO200076530-A1 Cancer
    HDQHM36 B64895 WO200076530-A1 Immune/Hematopoietic
    HFCBL53 B64901 WO200076530-A1 Cancer
    HIBCO70 B64905 WO200076530-A1 Cancer
    HLDCR26 B64908 WO200076530-A1 Cancer
    HLDDM27 B64909 WO200076530-A1 Cancer
    HMABK52 B64910 WO200076530-A1 Immune/Hematopoietic
    HNGBC53 B64915 WO200076530-A1 Immune/Hematopoietic
    HNGJB41 B64916 WO200076530-A1 Immune/Hematopoietic
    HNHLS76 B64917 WO200076530-A1 Immune/Hematopoietic
    HSJAQ10 B64925 WO200076530-A1 Cancer
    HSLDW54 B64927 WO200076530-A1 Cancer
    HSLFR59 B64928 WO200076530-A1 Cancer
    HSNAN38 B64930 WO200076530-A1 Cancer
    HLYEJ14 B64931 WO200076530-A1 Cancer
    HLYEJ14 B64932 WO200076530-A1 Cancer
    HPMCV08 B64993 WO200075375-A1 Cancer
    HFKEM67 B64994 WO200075375-A1 Excretory,
    Neural/Sensory,
    Reproductive
    HDTBW53 B64996 WO200075375-A1 Cancer
    HFICL62 B64997 WO200075375-A1 Cancer
    HKAIA52 B64998 WO200075375-A1 Cancer
    HEGAK44 B64999 WO200075375-A1 Cancer
    HFXHC85 B65000 WO200075375-A1 Cancer
    HSXCV85 B65001 WO200075375-A1 Neural/Sensory,
    Reproductive
    HPMCU14 B65002 WO200075375-A1 Cancer
    HYACJ27 B65003 WO200075375-A1 Immune/Hematopoietic
    HAQBZ15 B65004 WO200075375-A1 Cancer
    HBIBX03 B65005 WO200075375-A1 Cancer
    HBMVI06 B65006 WO200075375-A1 Cancer
    HDPBA28 B65007 WO200075375-A1 Cancer
    HDPUH26 B65008 WO200075375-A1 Cancer
    HJTAD07 B65009 WO200075375-A1 Cancer
    HDPPJ60 B65010 WO200075375-A1 Cancer
    HLTAU74 B65011 WO200075375-A1 Cancer
    HCFNN16 B65012 WO200075375-A1 Cancer
    HCWUI05 B65013 WO200075375-A1 Immune/Hematopoietic
    HCEBC76 B65014 WO200075375-A1 Neural/Sensory
    HTEGT82 B65015 WO200075375-A1 Digestive,
    Reproductive
    HLHTP35 B65016 WO200075375-A1 Cancer
    HSYAZ63 B65017 WO200075375-A1 Cancer
    HLTCR13 B65018 WO200075375-A1 Cancer
    HPMCV08 B65019 WO200075375-A1 Cancer
    HEGAK44 B65021 WO200075375-A1 Cancer
    HEGAK44 B65022 WO200075375-A1 Cancer
    HSXCV85 B65024 WO200075375-A1 Neural/Sensory,
    Reproductive
    HAQBZ15 B65025 WO200075375-A1 Cancer
    HDPMM34 R76127 WO9517092-A Cancer
    HAPBR31 R76128 WO9517092-A Cancer
    HPTXE69 R76818 WO9520398-A Cancer
    HPTGA39 R76820 WO9520398-A Cancer
    HFTDJ13 R81567 WO9606169-A1 Cancer
    HPAAA47 R88481 WO9601270-A1 Cancer
    HDQAC88 R93087 WO9605856-A1 Cancer
    HCHBM70 W06550 WO9639419-A1 Cancer
    HAPBR31 W07203 WO9634891-A1 Cancer
    HDPMM34 W07204 WO9634891-A1 Cancer
    HDTAX72 W07606 WO9639522-A1 Cancer
    HPBCW46 W09405 WO9639158-A1 Mixed Fetal,
    Neural/Sensory
    HE9ME29 W09408 WO9639486-A1 Cancer
    HDTAX72 W10574 WO9624668-A1 Cancer
    HGCOP28 W12692 WO9639424-A1 Cancer
    HDQAC88 W22670 WO9731098-A1 Cancer
    HDPJJ70 W22732 WO9724929-A1 Cancer
    HSKHZ53 W23663 WO9729189-A1 Cancer
    HATCY89 W27087 WO9725349-A1 Cancer
    HNTCF82 W27224 WO9735870-A1 Cardiovascular,
    Connective/Epithelial,
    Reproductive
    HFEBJ25 W29291 WO9735010-A1 Cancer
    HKMMP34 W29292 WO9735010-A1 Cancer
    HBMBJ94 W31527 WO9737022-A1 Digestive,
    Immune/Hematopoietic
    HDPVA94 W31902 WO9737021-A1 Cancer
    HFIZF58 W32110 WO9738012-A1 Cancer
    HSLDE91 W32112 WO9734998-A1 Cancer
    HDPVA94 W32323 WO9736915-A1 Cancer
    HETFO52 W35803 WO9734997-A1 Neural/Sensory,
    Reproductive
    HETEZ10 W35804 WO9734997-A1 Cancer
    HBMTO47 W35904 WO9738003-A1 Cancer
    HDQDX59 W37002 WO9733902-A1 Cancer
    H2LAJ93 W37844 WO9807749-A1 Cancer
    HMWIP18 W37845 WO9807749-A1 Cancer
    HMQDN51 W37935 WO9808870-A1 Cancer
    HOECW07 W37946 WO9821236-A1 Cancer
    HTHBJ48 W41938 WO9748807-A1 Digestive,
    Immune/Hematopoietic
    HCHBM70 W46882 US5733748-A Cancer
    HAHSD23 W48334 WO9807881-A1 Cancer
    HTEHH47 W48335 WO9807754-A1 Cancer
    HPASD50 W48391 WO9807735-A1 Cancer
    HDQMC88 W52842 WO9807862-A2 Connective/Epithelial,
    Immune/Hematopoietic
    HKACN58 W53897 WO9808969-A1 Cancer
    HOEAL47 W57635 WO9812344-A1 Cancer
    HAPBR31 W57697 WO9814582-A1 Cancer
    HDPMM34 W57698 WO9814582-A1 Cancer
    HE9ME29 W58704 US5780263-A Cancer
    HDTAX72 W58901 WO9814477-A1 Cancer
    HISCH47 W61621 WO9831799-A2 Cancer
    HDPIR89 W61622 WO9831799-A2 Digestive,
    Immune/Hematopoietic
    HAIDQ59 W61623 WO9831799-A2 Cancer
    HHFEK40 W61624 WO9831799-A2 Cancer
    HGBGV89 W61625 WO9831799-A2 Digestive
    HUVBB80 W61626 WO9831799-A2 Cancer
    HFGAG96 W61629 WO9831799-A2 Cancer
    HDTEA84 W63622 WO9830694-A2 Cancer
    HPRCB54 W64668 WO9830693-A2 Cancer
    HAGFY16 W67808 WO9842738-A1 Cancer
    HASAV70 W67811 WO9842738-A1 Cancer
    HBNAF22 W67812 WO9842738-A1 Cancer
    HCDDR90 W67814 WO9842738-A1 Cancer
    HCEEF50 W67815 WO9842738-A1 Cardiovascular,
    Neural/Sensory
    HCEMU42 W67816 WO9842738-A1 Cancer
    HCENE16 W67817 WO9842738-A1 Cancer
    HMSJJ74 W67818 WO9842738-A1 Cancer
    HCUBF15 W67819 WO9842738-A1 Immune/Hematopoietic
    HE2DE47 W67820 WO9842738-A1 Cancer
    HKMLH01 W67821 WO9842738-A1 Cancer
    HE9DG49 W67822 WO9842738-A1 Cancer
    HELBA06 W67823 WO9842738-A1 Cancer
    HSLFM29 W67824 WO9842738-A1 Cancer
    HELBW38 W67825 WO9842738-A1 Cancer
    HFEAF41 W67828 WO9842738-A1 Connective/Epithelial,
    Digestive
    HFTBE43 W67831 WO9842738-A1 Cancer
    HLHSV96 W67835 WO9842738-A1 Respiratory
    HLTBX31 W67837 WO9842738-A1 Cancer
    HLTCJ63 W67838 WO9842738-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HMQAJ64 W67840 WO9842738-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HOABG65 W67841 WO9842738-A1 Musculoskeletal
    HODCL36 W67842 WO9842738-A1 Cancer
    HODCL50 W67843 WO9842738-A1 Reproductive
    HODCZ16 W67845 WO9842738-A1 Cancer
    HTOEU03 W67846 WO9842738-A1 Cancer
    HPBCJ74 W67847 WO9842738-A1 Cancer
    HSIDQ18 W67850 WO9842738-A1 Cancer
    HSJBQ79 W67852 WO9842738-A1 Cancer
    HTEJN13 W67854 WO9842738-A1 Neural/Sensory,
    Reproductive
    HTHBL86 W67855 WO9842738-A1 Immune/Hematopoietic
    HTSFO71 W67856 WO9842738-A1 Cancer
    HAPNO80 W67857 WO9842738-A1 Cancer
    HBIBZ09 W67858 WO9842738-A1 Cancer
    HCFLD84 W67859 WO9842738-A1 Cancer
    HE8EZ48 W67861 WO9842738-A1 Cancer
    HEBGF73 W67862 WO9842738-A1 Cancer
    HFEBF41 W67863 WO9842738-A1 Cancer
    HFRBU14 W67864 WO9842738-A1 Neural/Sensory
    HHGCO88 W67867 WO9842738-A1 Cancer
    HHGDB72 W67869 WO9842738-A1 Cancer
    HHGDI71 W67870 WO9842738-A1 Excretory
    HHSDI45 W67871 WO9842738-A1 Cancer
    HHSEB66 W67872 WO9842738-A1 Cancer
    HAUAI83 W67873 WO9842738-A1 Reproductive
    HKDBL30 W67874 WO9842738-A1 Cancer
    HLDBQ19 W67875 WO9842738-A1 Cancer
    HMSGT42 W67876 WO9842738-A1 Cancer
    HMWIC78 W67877 WO9842738-A1 Cancer
    HMWIR31 W67878 WO9842738-A1 Cancer
    HNTAC73 W67880 WO9842738-A1 Cancer
    HOSEI45 W67881 WO9842738-A1 Cancer
    HOSFD58 W67882 WO9842738-A1 Cancer
    HSAUM95 W67883 WO9842738-A1 Cancer
    HSAUR67 W67884 WO9842738-A1 Immune/Hematopoietic
    HSKDI81 W67885 WO9842738-A1 Cancer
    HOUFJ08 W67886 WO9842738-A1 Cancer
    HTLEX50 W67887 WO9842738-A1 Cancer
    HSKHL65 W67888 WO9842738-A1 Cancer
    HHFGA11 W67889 WO9842738-A1 Cancer
    HAQCF47 W67890 WO9842738-A1 Cancer
    HBXFG80 W67891 WO9842738-A1 Cancer
    HFLQB16 W67895 WO9842738-A1 Cancer
    HBMCP89 W67896 WO9842738-A1 Cancer
    HE6DG34 W67897 WO9842738-A1 Cancer
    HE9DG49 W67898 WO9842738-A1 Cancer
    HELBA06 W67899 WO9842738-A1 Cancer
    HMQAJ64 W67900 WO9842738-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HODCL36 W67901 WO9842738-A1 Cancer
    HODCL36 W67902 WO9842738-A1 Cancer
    HCMSD80 W67903 WO9842738-A1 Cancer
    HPBCJ74 W67904 WO9842738-A1 Cancer
    HHGDU04 W67905 WO9842738-A1 Cancer
    HTEJN13 W67907 WO9842738-A1 Neural/Sensory,
    Reproductive
    HAUCC47 W67909 WO9842738-A1 Cancer
    HOSFD58 W67913 WO9842738-A1 Cancer
    HSKHL65 W67916 WO9842738-A1 Cancer
    HHFGA11 W67917 WO9842738-A1 Cancer
    HOEBX83 W67918 WO9842738-A1 Cancer
    HHFGA11 W67919 WO9842738-A1 Cancer
    HTSFO71 W67967 WO9842738-A1 Cancer
    HKFBC53 W68002 WO9842738-A1 Cancer
    HSPBS71 W69221 WO9828420-A1 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HDPBT77 W69232 WO9831806-A2 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HNFFL83 W69233 WO9831806-A2 Digestive,
    Immune/Hematopoietic
    HETHE81 W71593 WO9833912-A1 Cancer
    HSVBZ80 W73397 WO9854206-A1 Cancer
    HTAAU21 W73398 WO9854206-A1 Cancer
    HUSIR91 W73400 WO9854206-A1 Cancer
    HADMC21 W73401 WO9854206-A1 Cancer
    HAGFM45 W73402 WO9854206-A1 Cancer
    HAIBE65 W73403 WO9854206-A1 Cancer
    HAQBH57 W73404 WO9854206-A1 Cancer
    HATCX80 W73405 WO9854206-A1 Cancer
    HLDOT61 W73408 WO9854206-A1 Cancer
    HEMCM42 W73409 WO9854206-A1 Cancer
    HFCDW34 W73411 WO9854206-A1 Cancer
    HTTEU91 W73412 WO9854206-A1 Cancer
    HHGBF89 W73413 WO9854206-A1 Mixed Fetal
    HKMLN27 W73415 WO9854206-A1 Cancer
    HLYAZ61 W73419 WO9854206-A1 Immune/Hematopoietic
    HMQDT36 W73420 WO9854206-A1 Cancer
    HETFI51 W73428 WO9854206-A1 Cancer
    HUSIR91 W73429 WO9854206-A1 Cancer
    HHGBF89 W73430 WO9854206-A1 Mixed Fetal
    HPWBA10 W73432 WO9854206-A1 Immune/Hematopoietic,
    Reproductive
    HPMBQ91 W74413 EP892053-A2 Reproductive
    HBGBW52 W74732 WO9839448-A2 Cancer
    HCUFQ22 W74734 WO9839448-A2 Immune/Hematopoietic
    HLDOU93 W74738 WO9839448-A2 Digestive,
    Musculoskeletal,
    Reproductive
    HNGJJ68 W74741 WO9839448-A2 Cancer
    HCFAW04 W74742 WO9839448-A2 Immune/Hematopoietic
    HLMAV65 W74743 WO9839448-A2 Cancer
    HPMFD84 W74744 WO9839448-A2 Cancer
    HE6DB26 W74745 WO9839448-A2 Cancer
    HODBD33 W74747 WO9839448-A2 Reproductive
    HBJAE44 W74750 WO9839448-A2 Immune/Hematopoietic
    HCFME41 W74751 WO9839448-A2 Cancer
    HOGCO71 W74752 WO9839448-A2 Cancer
    HOSEX08 W74753 WO9839448-A2 Cancer
    HSKNJ72 W74754 WO9839448-A2 Digestive,
    Musculoskeletal
    HEBEB69 W74755 WO9839448-A2 Neural/Sensory,
    Reproductive
    HE6EH18 W74756 WO9839448-A2 Mixed Fetal,
    Neural/Sensory
    HSSDM73 W74758 WO9839448-A2 Musculoskeletal,
    Neural/Sensory,
    Reproductive
    HMKCU94 W74761 WO9839448-A2 Cancer
    HRDEW41 W74762 WO9839448-A2 Cancer
    HBGDA21 W74764 WO9839448-A2 Cancer
    HFGAK75 W74765 WO9839448-A2 Cancer
    HFSAU96 W74766 WO9839448-A2 Cancer
    HOVCL83 W74767 WO9839448-A2 Cancer
    HBICM48 W74769 WO9839448-A2 Cancer
    HLTCL35 W74770 WO9839448-A2 Cancer
    HRSAN45 W74771 WO9839448-A2 Cancer
    HSNBB14 W74772 WO9839448-A2 Cancer
    HMABL38 W74773 WO9839448-A2 Cancer
    HSKDK47 W74774 WO9839448-A2 Cancer
    HOSFH03 W74775 WO9839448-A2 Cancer
    HOGAV75 W74776 WO9839448-A2 Cancer
    HBXDO23 W74777 WO9839448-A2 Cancer
    HAGBI17 W74778 WO9839448-A2 Cancer
    HPRCA31 W74780 WO9839448-A2 Cancer
    HPRCE95 W74781 WO9839448-A2 Cancer
    HHTLC66 W74782 WO9839448-A2 Cancer
    HMADJ02 W74783 WO9839448-A2 Cancer
    HPRCU93 W74784 WO9839448-A2 Cancer
    HSAXS65 W74785 WO9839448-A2 Cancer
    HHFHN61 W74787 WO9839448-A2 Cancer
    HCWEF90 W74788 WO9839448-A2 Cancer
    HFRAU10 W74790 WO9839448-A2 Neural/Sensory
    HATDT67 W74791 WO9839448-A2 Cancer
    HOUBG93 W74792 WO9839448-A2 Cancer
    HMWEX24 W74793 WO9839448-A2 Cancer
    HTOCD52 W74795 WO9839448-A2 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HTGCP16 W74796 WO9839448-A2 Cancer
    HKIXR69 W74797 WO9839448-A2 Cancer
    HE6CN34 W74800 WO9839448-A2 Cancer
    HSQEL25 W74802 WO9839448-A2 Cancer
    HEBEG68 W74803 WO9839448-A2 Cancer
    HBIAB39 W74804 WO9839448-A2 Cancer
    HOEAS24 W74805 WO9839448-A2 Cancer
    HETDD75 W74806 WO9839448-A2 Cancer
    HSKNE46 W74807 WO9839448-A2 Cancer
    HPMFL27 W74808 WO9839448-A2 Cancer
    HPRAX55 W74810 WO9839448-A2 Cancer
    HE2PL77 W74812 WO9839448-A2 Cancer
    HLHAU92 W74813 WO9839448-A2 Cancer
    HTPEG42 W74814 WO9839448-A2 Cancer
    HAUAV32 W74816 WO9839448-A2 Cancer
    HNEBI60 W74817 WO9839448-A2 Cancer
    HTSEL31 W74819 WO9839448-A2 Cancer
    HAUBL57 W74820 WO9839448-A2 Cancer
    HE6CT48 W74822 WO9839448-A2 Digestive,
    Mixed Fetal
    HMDAA61 W74823 WO9839448-A2 Cancer
    HAQBK61 W74824 WO9839448-A2 Cancer
    HAQBF73 W74825 WO9839448-A2 Cancer
    HAQBT94 W74826 WO9839448-A2 Cancer
    HLQAB52 W74828 WO9839448-A2 Cancer
    HE2BG03 W74830 WO9839448-A2 Cancer
    HCUBC79 W74832 WO9839448-A2 Cancer
    HSVAF07 W74833 WO9839448-A2 Cancer
    HT3AM65 W74834 WO9839448-A2 Cancer
    HE6DK18 W74835 WO9839448-A2 Cancer
    HEBEK93 W74836 WO9839448-A2 Cancer
    HJPCM10 W74837 WO9839448-A2 Cancer
    HSXBL78 W74838 WO9839448-A2 Cancer
    HOEAW81 W74839 WO9839448-A2 Cancer
    HEAAR60 W74841 WO9839448-A2 Cancer
    HOVBA03 W74843 WO9839448-A2 Cancer
    HGBGK76 W74844 WO9839448-A2 Digestive,
    Neural/Sensory
    HBMUW78 W74845 WO9839448-A2 Cancer
    HATCM76 W74848 WO9839448-A2 Cancer
    H6EBJ64 W74849 WO9839448-A2 Cancer
    HDDAD77 W74850 WO9839448-A2 Cancer
    HSPAG15 W74853 WO9839448-A2 Cancer
    HUSHH48 W74855 WO9839448-A2 Cancer
    HHSCV65 W74857 WO9839448-A2 Cancer
    HHSDQ41 W74858 WO9839448-A2 Cancer
    HEBFU93 W74860 WO9839448-A2 Excretory,
    Neural/Sensory,
    Reproductive
    HSGSC60 W74861 WO9839448-A2 Cancer
    HPMGD24 W74862 WO9839448-A2 Cancer
    HPTVC60 W74863 WO9839448-A2 Cancer
    HSKNE18 W74864 WO9839448-A2 Cancer
    HMWIF35 W74865 WO9839448-A2 Cancer
    HMWGI25 W74866 WO9839448-A2 Cancer
    HSKGF03 W74867 WO9839448-A2 Cancer
    HMSKE75 W74868 WO9839448-A2 Cancer
    HCMSH30 W74869 WO9839448-A2 Cancer
    HTWCB92 W74870 WO9839448-A2 Cancer
    HBMDM46 W74871 WO9839448-A2 Cancer
    HFXHL79 W74873 WO9839448-A2 Cancer
    HBJFJ73 W74874 WO9839448-A2 Cancer
    HSJAP03 W74875 WO9839448-A2 Cancer
    H6EAD09 W74876 WO9839448-A2 Cancer
    HTLEF62 W74879 WO9839448-A2 Cancer
    HTLAD94 W74880 WO9839448-A2 Cancer
    HTSFQ12 W74881 WO9839448-A2 Cancer
    HCE2K05 W74882 WO9839448-A2 Cancer
    HLTED27 W74884 WO9839448-A2 Cancer
    HMKBA64 W74885 WO9839448-A2 Cancer
    HNFCO49 W74886 WO9839448-A2 Cancer
    HCELB21 W74887 WO9839448-A2 Cancer
    HSAAS44 W74889 WO9839448-A2 Cancer
    HAFAL73 W74890 WO9839448-A2 Cancer
    HSAWF26 W74891 WO9839448-A2 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HMQDN51 W74892 WO9839448-A2 Cancer
    H2LAO11 W74894 WO9839448-A2 Cancer
    HPTTU11 W74896 WO9839448-A2 Cancer
    HTEDJ34 W74898 WO9839448-A2 Cancer
    HFTAR26 W74900 WO9839448-A2 Cancer
    H2MBF44 W74901 WO9839448-A2 Cancer
    HE8BI92 W74902 WO9839448-A2 Cancer
    HFTBR48 W74903 WO9839448-A2 Cancer
    HE9CM64 W74904 WO9839448-A2 Cancer
    HATAV51 W74905 WO9839448-A2 Cancer
    HCEEK08 W74907 WO9839448-A2 Cancer
    HAFAU18 W74908 WO9839448-A2 Cancer
    HETBY74 W74909 WO9839448-A2 Cancer
    HTOAF35 W74910 WO9839448-A2 Cancer
    HCRBX32 W74911 WO9839448-A2 Cancer
    HEBGB80 W74912 WO9839448-A2 Cancer
    HFAMH74 W74913 WO9839448-A2 Cancer
    HLMAV65 W74920 WO9839448-A2 Cancer
    HMAGF23 W74922 WO9839448-A2 Cancer
    HE6EH18 W74929 WO9839448-A2 Mixed Fetal,
    Neural/Sensory
    HMKCU94 W74930 WO9839448-A2 Cancer
    HBGDA21 W74931 WO9839448-A2 Cancer
    HFKFN58 W74932 WO9839448-A2 Cancer
    HSNBB14 W74935 WO9839448-A2 Cancer
    HOSFH03 W74937 WO9839448-A2 Cancer
    HAGBI17 W74939 WO9839448-A2 Cancer
    HPRCA31 W74940 WO9839448-A2 Cancer
    HPRCU93 W74943 WO9839448-A2 Cancer
    HPDDK44 W74944 WO9839448-A2 Cancer
    HCWEF90 W74946 WO9839448-A2 Cancer
    HFRAU10 W74947 WO9839448-A2 Neural/Sensory
    HBIAB39 W74953 WO9839448-A2 Cancer
    HBIAB39 W74954 WO9839448-A2 Cancer
    HOEAS24 W74955 WO9839448-A2 Cancer
    HOEAS24 W74956 WO9839448-A2 Cancer
    HPRAX55 W74958 WO9839448-A2 Cancer
    HTPEG42 W74960 WO9839448-A2 Cancer
    HAUAV32 W74961 WO9839448-A2 Cancer
    HNEBI60 W74962 WO9839448-A2 Cancer
    HAUBL57 W74963 WO9839448-A2 Cancer
    HAUBL57 W74964 WO9839448-A2 Cancer
    HE6CT48 W74965 WO9839448-A2 Digestive,
    Mixed Fetal
    HMDAA61 W74966 WO9839448-A2 Cancer
    HAQBK61 W74967 WO9839448-A2 Cancer
    HCUHB01 W74968 WO9839448-A2 Cancer
    HETHE07 W74970 WO9839448-A2 Cancer
    HETHE07 W74971 WO9839448-A2 Cancer
    HLQAB52 W74972 WO9839448-A2 Cancer
    HEONN58 W74973 WO9839448-A2 Cancer
    HIBEK16 W74974 WO9839448-A2 Cancer
    HE2BG03 W74975 WO9839448-A2 Cancer
    HCUBC79 W74976 WO9839448-A2 Cancer
    HSVAF07 W74978 WO9839448-A2 Cancer
    HSVAF07 W74979 WO9839448-A2 Cancer
    HT3AM65 W74980 WO9839448-A2 Cancer
    HT3AM65 W74981 WO9839448-A2 Cancer
    HJPCM10 W74983 WO9839448-A2 Cancer
    HJPCM10 W74984 WO9839448-A2 Cancer
    HOVBA03 W74987 WO9839448-A2 Cancer
    H6EBJ64 W74990 WO9839448-A2 Cancer
    HUSHH48 W74991 WO9839448-A2 Cancer
    HEBFU93 W74992 WO9839448-A2 Excretory,
    Neural/Sensory,
    Reproductive
    HPTVC60 W74993 WO9839448-A2 Cancer
    HMWIF35 W74995 WO9839448-A2 Cancer
    HSKGF03 W74996 WO9839448-A2 Cancer
    HBJFJ73 W75000 WO9839448-A2 Cancer
    HCFBC03 W75001 WO9839448-A2 Cancer
    HSJAP03 W75002 WO9839448-A2 Cancer
    HE6FL83 W75005 WO9839448-A2 Cancer
    HPTTU11 W75013 WO9839448-A2 Cancer
    H2MBF44 W75015 WO9839448-A2 Cancer
    HE9CM64 W75018 WO9839448-A2 Cancer
    HAFAU18 W75021 WO9839448-A2 Cancer
    HSHCC16 W75050 WO9839448-A2 Cancer
    HGCMD20 W75057 WO9839446-A2 Cancer
    HLDBG33 W75058 WO9839446-A2 Cancer
    HLHEJ14 W75059 WO9839446-A2 Cancer
    HKCSR70 W75060 WO9839446-A2 Cancer
    HBMCY91 W75062 WO9839446-A2 Immune/Hematopoietic
    HSSGE07 W75063 WO9839446-A2 Cancer
    HBMBX59 W75064 WO9839446-A2 Immune/Hematopoietic,
    Reproductive
    HNGIT22 W75065 WO9839446-A2 Immune/Hematopoietic
    HERAD57 W75066 WO9839446-A2 Connective/Epithelial
    HCENJ40 W75067 WO9839446-A2 Cancer
    HCSRA90 W75068 WO9839446-A2 Cardiovascular,
    Musculoskeletal
    HBJFC03 W75069 WO9839446-A2 Immune/Hematopoietic
    HTEBY26 W75071 WO9839446-A2 Cancer
    HMABH07 W75072 WO9839446-A2 Cancer
    HSKNY94 W75073 WO9839446-A2 Cancer
    HMCDA67 W75074 WO9839446-A2 Immune/Hematopoietic
    HOSFF45 W75075 WO9839446-A2 Cancer
    HMJAA51 W75076 WO9839446-A2 Cancer
    HTEBF05 W75077 WO9839446-A2 Reproductive
    HTEAL31 W75078 WO9839446-A2 Cancer
    HSKXE91 W75080 WO9839446-A2 Cancer
    HPWTB39 W75081 WO9839446-A2 Mixed Fetal,
    Reproductive
    HTLEV12 W75082 WO9839446-A2 Reproductive
    HSPAF93 W75083 WO9839446-A2 Digestive
    HHFGL62 W75084 WO9839446-A2 Cardiovascular
    HCE1U14 W75085 WO9839446-A2 Cancer
    HTHBA79 W75087 WO9839446-A2 Cancer
    HAGBB70 W75088 WO9839446-A2 Cancer
    HETDG84 W75089 WO9839446-A2 Cancer
    HTEGA81 W75090 WO9839446-A2 Cancer
    HTXAK60 W75091 WO9839446-A2 Cancer
    HMHBN40 W75092 WO9839446-A2 Cancer
    HFVGS85 W75093 WO9839446-A2 Cancer
    HERAH81 W75094 WO9839446-A2 Cancer
    HMSEU04 W75095 WO9839446-A2 Cancer
    HNEDJ57 W75096 WO9839446-A2 Cancer
    HNTME13 W75097 WO9839446-A2 Cancer
    HSXBI25 W75098 WO9839446-A2 Cancer
    HSXCK41 W75099 WO9839446-A2 Cancer
    HE8CJ26 W75100 WO9839446-A2 Cancer
    HTTDS54 W75101 WO9839446-A2 Cancer
    HHFCW44 W75102 WO9839446-A2 Cancer
    HMCBP63 W75103 WO9839446-A2 Cancer
    HEMGE83 W75104 WO9839446-A2 Cancer
    HHSDC22 W75105 WO9839446-A2 Digestive,
    Neural/Sensory
    HHSDZ57 W75106 WO9839446-A2 Cancer
    HCRBS80 W75107 WO9839446-A2 Cancer
    HMMAB12 W75108 WO9839446-A2 Immune/Hematopoietic,
    Neural/Sensory
    HSKDW02 W75109 WO9839446-A2 Cancer
    HWHHL34 W75110 WO9839446-A2 Cancer
    HODAZ50 W75111 WO9839446-A2 Reproductive
    HCEWC82 W75112 WO9839446-A2 Cancer
    HE6ES13 W75113 WO9839446-A2 Cancer
    HSSEP68 W75114 WO9839446-A2 Cancer
    HRDEV41 W75115 WO9839446-A2 Cancer
    HILCJ01 W75116 WO9839446-A2 Cancer
    HSATP28 W75117 WO9839446-A2 Cancer
    HBJEM49 W75119 WO9839446-A2 Cancer
    HSLDJ95 W75120 WO9839446-A2 Cancer, Immune
    HSREG44 W75121 WO9839446-A2 Cancer
    HTXCT40 W75122 WO9839446-A2 Cancer
    HRGDF73 W75123 WO9839446-A2 Cancer
    HKMND45 W75124 WO9839446-A2 Cancer
    HPEBD70 W75125 WO9839446-A2 Cancer
    HLMDX11 W75126 WO9839446-A2 Cancer
    HKCSR70 W75128 WO9839446-A2 Cancer
    HETBI87 W75129 WO9839446-A2 Reproductive
    HSSGE07 W75130 WO9839446-A2 Cancer
    HCENJ40 W75132 WO9839446-A2 Cancer
    HSNBL85 W75135 WO9839446-A2 Cancer
    HMAAD57 W75137 WO9839446-A2 Cancer
    HMAAD57 W75138 WO9839446-A2 Cancer
    HSKNY94 W75139 WO9839446-A2 Cancer
    HOSFF45 W75140 WO9839446-A2 Cancer
    HMJAA51 W75141 WO9839446-A2 Cancer
    HTEAL31 W75142 WO9839446-A2 Cancer
    HSPAF93 W75145 WO9839446-A2 Digestive
    HHFGL62 W75146 WO9839446-A2 Cardiovascular
    HTHBA79 W75148 WO9839446-A2 Cancer
    HTEGA81 W75151 WO9839446-A2 Cancer
    HTEGA81 W75152 WO9839446-A2 Cancer
    HMHBN40 W75154 WO9839446-A2 Cancer
    HLHDL62 W75155 WO9839446-A2 Cancer
    HSXBI25 W75156 WO9839446-A2 Cancer
    HSXCK41 W75157 WO9839446-A2 Cancer
    HTTDS54 W75159 WO9839446-A2 Cancer
    HHFCW44 W75160 WO9839446-A2 Cancer
    HHSDZ57 W75161 WO9839446-A2 Cancer
    HAICS58 W75162 WO9839446-A2 Cancer
    HAICS58 W75163 WO9839446-A2 Cancer
    HSKDW02 W75165 WO9839446-A2 Cancer
    HETGL41 W75166 WO9839446-A2 Cancer
    HODAZ50 W75167 WO9839446-A2 Reproductive
    HE6ES13 W75168 WO9839446-A2 Cancer
    HSSEP68 W75169 WO9839446-A2 Cancer
    HRDEV41 W75171 WO9839446-A2 Cancer
    HHFGL41 W75172 WO9839446-A2 Cancer
    HBJEM49 W75173 WO9839446-A2 Cancer
    HFTAK35 W75174 WO9839446-A2 Cancer
    HTXCT40 W75175 WO9839446-A2 Cancer
    HRDBF52 W75176 WO9839446-A2 Cancer
    HKMND45 W75177 WO9839446-A2 Cancer
    HDTBJ30 W75178 WO9839446-A2 Cancer
    HLMDX11 W75179 WO9839446-A2 Cancer
    HCEAB46 W75196 WO9840483-A2 Cancer
    HCEDH81 W75197 WO9840483-A2 Cancer
    HELDY41 W75200 WO9840483-A2 Cancer
    HETDM20 W75201 WO9840483-A2 Cancer
    HE2DX30 W75202 WO9840483-A2 Cancer
    HJBCD89 W75204 WO9840483-A2 Cancer
    HJTAA17 W75205 WO9840483-A2 Cancer
    HLTBS22 W75206 WO9840483-A2 Cancer
    HNFCV70 W75208 WO9840483-A2 Cancer
    HNFGF45 W75210 WO9840483-A2 Cancer
    HOVAB12 W75211 WO9840483-A2 Cancer
    HPMBQ91 W75212 WO9840483-A2 Reproductive
    HRSMC69 W75214 WO9840483-A2 Cancer
    HSQFP46 W75216 WO9840483-A2 Cancer
    HTEAE62 W75218 WO9840483-A2 Cardiovascular,
    Reproductive
    HTEBY11 W75219 WO9840483-A2 Reproductive
    HTEEB42 W75220 WO9840483-A2 Cancer
    HTPBY11 W75221 WO9840483-A2 Cancer
    HCEDH81 W75224 WO9840483-A2 Cancer
    HJBCD89 W75226 WO9840483-A2 Cancer
    HNFCV70 W75227 WO9840483-A2 Cancer
    HPMBQ91 W75228 WO9840483-A2 Reproductive
    HBMSH54 W75231 WO9840483-A2 Cancer
    HSDEG01 W75232 WO9840483-A2 Cancer
    HSQFP46 W75233 WO9840483-A2 Cancer
    HTEBY11 W75234 WO9840483-A2 Reproductive
    HYACC84 W75245 WO9840483-A2 Cancer
    HETAG43 W76253 WO9831818-A2 Digestive,
    Reproductive
    HOSBI96 W78128 WO9856804-A1 Cancer
    HPDDC77 W78131 WO9856804-A1 Cancer
    HPEBD85 W78132 WO9856804-A1 Digestive,
    Reproductive
    HPMGQ80 W78135 WO9856804-A1 Cancer
    HSDES04 W78140 WO9856804-A1 Cancer
    HSHBQ68 W78141 WO9856804-A1 Cancer
    HSKBO20 W78142 WO9856804-A1 Cancer
    HSKZE52 W78145 WO9856804-A1 Cancer
    HWTAZ75 W78146 WO9856804-A1 Cancer
    HSVAG05 W78148 WO9856804-A1 Cancer
    HSVBF78 W78149 WO9856804-A1 Cancer
    HSXBO51 W78150 WO9856804-A1 Cancer
    HT4AI54 W78152 WO9856804-A1 Cancer
    HTEHU93 W78153 WO9856804-A1 Reproductive
    HMSDG61 W78154 WO9856804-A1 Cancer
    HTLDQ11 W78157 WO9856804-A1 Reproductive
    HTOBX52 W78158 WO9856804-A1 Cancer
    HTTCN24 W78159 WO9856804-A1 Cancer
    HTXCS21 W78160 WO9856804-A1 Cancer
    HBMBB80 W78164 WO9856804-A1 Digestive,
    Immune/Hematopoietic
    HSXBP68 W78166 WO9856804-A1 Cancer
    HFFAT33 W78167 WO9856804-A1 Cancer
    HFGAG96 W78168 WO9856804-A1 Cancer
    HETFJ05 W78169 WO9856804-A1 Cancer
    HE8BX01 W78170 WO9856804-A1 Cancer
    HMSJU68 W78171 WO9856804-A1 Cancer
    HOSCZ41 W78172 WO9856804-A1 Cancer
    HSQEA85 W78174 WO9856804-A1 Cancer
    HSTAG52 W78175 WO9856804-A1 Cancer
    HBXGP76 W78177 WO9856804-A1 Immune/Hematopoietic,
    Neural/Sensory
    HE6GL64 W78178 WO9856804-A1 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HESAL35 W78179 WO9856804-A1 Connective/Epithelial,
    Mixed Fetal
    HNHAL34 W78183 WO9856804-A1 Cancer
    HOSFF78 W78184 WO9856804-A1 Cancer
    HPMCC16 W78188 WO9856804-A1 Cancer
    HOUCQ17 W78189 WO9856804-A1 Cancer
    HTOFC34 W78192 WO9856804-A1 Cancer
    H2CBJ08 W78193 WO9856804-A1 Cancer
    HAGFT48 W78194 WO9856804-A1 Cancer
    HCE5M29 W78195 WO9856804-A1 Cancer
    HCFNN01 W78197 WO9856804-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HE7TF86 W78198 WO9856804-A1 Cancer
    HHGAU81 W78200 WO9856804-A1 Cancer
    HPTRF90 W78206 WO9856804-A1 Cancer
    HSRDH01 W78207 WO9856804-A1 Cancer
    HSAWD74 W78208 WO9856804-A1 Cancer
    HTEJO12 W78209 WO9856804-A1 Digestive,
    Reproductive
    HTLAB43 W78210 WO9856804-A1 Cancer
    HTWCT03 W78211 WO9856804-A1 Immune/Hematopoietic
    HSDES04 W78213 WO9856804-A1 Cancer
    HT3BE24 W78214 WO9856804-A1 Cancer
    HTTCN24 W78216 WO9856804-A1 Cancer
    HCRAZ77 W78221 WO9856804-A1 Cancer
    HFGAG96 W78222 WO9856804-A1 Cancer
    HADTN61 W78223 WO9856804-A1 Cancer
    HLYBF81 W78224 WO9856804-A1 Cancer
    HSTBE27 W78225 WO9856804-A1 Cancer
    HMSDG61 W78263 WO9856804-A1 Cancer
    HTOBX52 W78274 WO9856804-A1 Cancer
    HFGAG96 W78295 WO9856804-A1 Cancer
    HCE5M29 W78316 WO9856804-A1 Cancer
    HLCAA05 W78321 WO9856804-A1 Cancer
    HTLEF68 W78326 WO9856804-A1 Cancer
    HSJAR34 W79739 WO9846746-A1 Cancer
    HOUCQ17 W80285 EP874050-A2 Cancer
    HCWHZ93 W83931 WO9845712-A2 Immune/Hematopoietic,
    Neural/Sensory
    HE2FV03 W83933 WO9845712-A2 Cancer
    HCDAG36 W83934 WO9845712-A2 Cancer
    HMQBU44 W83935 WO9845712-A2 Cancer
    HLHCM89 W83938 WO9845712-A2 Cancer
    HLHEF26 W83939 WO9845712-A2 Cancer
    HLHEO50 W83940 WO9845712-A2 Cancer
    HDSAE10 W83941 WO9845712-A2 Cancer
    HSKNK73 W83942 WO9845712-A2 Cancer
    HSSMS41 W83943 WO9845712-A2 Cancer
    HNGBV36 W83944 WO9845712-A2 Cancer
    HNGDE27 W83945 WO9845712-A2 Immune/Hematopoietic
    HPFDU90 W83947 WO9845712-A2 Cancer
    HRLMD77 W83948 WO9845712-A2 Cancer
    HRLMF92 W83949 WO9845712-A2 Cancer
    HLHDZ58 W88535 WO9854963-A2 Respiratory
    HLMMJ13 W88536 WO9854963-A2 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HNFED65 W88539 WO9854963-A2 Excretory,
    Immune/Hematopoietic
    HNHDX07 W88540 WO9854963-A2 Immune/Hematopoietic
    HNHGC82 W88541 WO9854963-A2 Immune/Hematopoietic
    HNHGO09 W88542 WO9854963-A2 Immune/Hematopoietic
    HOUBE18 W88543 WO9854963-A2 Cancer
    HOUDL69 W88544 WO9854963-A2 Cancer
    HPMFI71 W88545 WO9854963-A2 Cancer
    HPTBB03 W88548 WO9854963-A2 Cancer
    HPTWA66 W88549 WO9854963-A2 Cancer
    HPTWC08 W88550 WO9854963-A2 Cancer
    HRGCZ46 W88551 WO9854963-A2 Cancer
    HSAVU34 W88552 WO9854963-A2 Cancer
    HSDFW61 W88553 WO9854963-A2 Cancer
    HSQEO84 W88556 WO9854963-A2 Cancer
    HSXAM05 W88557 WO9854963-A2 Cancer
    HSXAS67 W88558 WO9854963-A2 Neural/Sensory
    HTDAF28 W88559 WO9854963-A2 Cancer
    HTOAM21 W88562 WO9854963-A2 Immune/Hematopoietic
    HETCH46 W88563 WO9854963-A2 Cancer
    HJPCD40 W88564 WO9854963-A2 Cancer
    HTWBY48 W88565 WO9854963-A2 Immune/Hematopoietic
    HWTBF59 W88568 WO9854963-A2 Cancer
    HAGFB60 W88570 WO9854963-A2 Neural/Sensory
    HATEF60 W88571 WO9854963-A2 Cancer
    HCDAR68 W88573 WO9854963-A2 Cancer
    HMDAN54 W88575 WO9854963-A2 Immune/Hematopoietic,
    Neural/Sensory
    HCEEC15 W88577 WO9854963-A2 Cancer
    HCESF40 W88578 WO9854963-A2 Immune/Hematopoietic,
    Neural/Sensory
    HCFMV39 W88579 WO9854963-A2 Cancer
    HCNAP62 W88581 WO9854963-A2 Cancer
    HCUDC07 W88583 WO9854963-A2 Immune/Hematopoietic
    HCWBB42 W88584 WO9854963-A2 Immune/Hematopoietic
    HE9ND48 W88592 WO9854963-A2 Mixed Fetal
    HEBBW11 W88593 WO9854963-A2 Cancer
    HEMAE80 W88595 WO9854963-A2 Cardiovascular,
    Musculoskeletal,
    Reproductive
    HFEBA88 W88596 WO9854963-A2 Cancer
    HGBAJ93 W88599 WO9854963-A2 Cancer
    HGBBQ69 W88600 WO9854963-A2 Cancer
    HHFHJ59 W88602 WO9854963-A2 Cancer
    HHPFD63 W88606 WO9854963-A2 Endocrine,
    Immune/Hematopoietic,
    Neural/Sensory
    HHSEG23 W88607 WO9854963-A2 Neural/Sensory
    HKIXL73 W88609 WO9854963-A2 Cancer
    HKMNC43 W88610 WO9854963-A2 Excretory
    HMEJE31 W88611 WO9854963-A2 Cardiovascular
    HNFAE54 W88613 WO9854963-A2 Cancer
    HNFJH45 W88614 WO9854963-A2 Immune/Hematopoietic
    HNGBT31 W88615 WO9854963-A2 Immune/Hematopoietic
    HNGIN60 W88616 WO9854963-A2 Immune/Hematopoietic,
    Neural/Sensory
    HNHDW42 W88618 WO9854963-A2 Immune/Hematopoietic
    HNHFL57 W88619 WO9854963-A2 Immune/Hematopoietic
    HOGAR52 W88620 WO9854963-A2 Cancer
    HOSBZ55 W88621 WO9854963-A2 Cancer
    HOSDI92 W88622 WO9854963-A2 Cancer
    HPBCU51 W88623 WO9854963-A2 Cancer
    HNTSU23 W88628 WO9854963-A2 Cancer
    HRDFB85 W88629 WO9854963-A2 Cancer
    HSKGN81 W88631 WO9854963-A2 Cancer
    HSPAH56 W88632 WO9854963-A2 Cancer
    HDTAL71 W88633 WO9854963-A2 Cancer
    HSXCS62 W88634 WO9854963-A2 Cancer
    HTEKM35 W88636 WO9854963-A2 Neural/Sensory,
    Reproductive
    HTGEP89 W88637 WO9854963-A2 Immune/Hematopoietic,
    Neural/Sensory
    HTPCN79 W88640 WO9854963-A2 Digestive,
    Neural/Sensory
    HTSGM54 W88641 WO9854963-A2 Cancer
    HTWAF58 W88643 WO9854963-A2 Immune/Hematopoietic
    HTWBY29 W88644 WO9854963-A2 Cancer
    HUKFC71 W88645 WO9854963-A2 Cancer
    HCE2V74 W88646 WO9854963-A2 Cancer
    HFXBW82 W88652 WO9854963-A2 Neural/Sensory
    HIBED17 W88654 WO9854963-A2 Cancer
    HPMCJ92 W88657 WO9854963-A2 Musculoskeletal,
    Reproductive
    HPWAZ95 W88658 WO9854963-A2 Reproductive
    HSUBW09 W88660 WO9854963-A2 Digestive,
    Immune/Hematopoietic
    HALSQ59 W88666 WO9854963-A2 Cancer
    HAIBP89 W88667 WO9854963-A2 Cancer
    HBXGK12 W88669 WO9854963-A2 Cancer
    HFKFJ07 W88670 WO9854963-A2 Cancer
    HCWHZ24 W88672 WO9854963-A2 Immune/Hematopoietic
    HE2GT20 W88673 WO9854963-A2 Cancer
    HFTCT67 W88676 WO9854963-A2 Cancer
    HUSIT49 W88680 WO9854963-A2 Cancer
    HNHED86 W88684 WO9854963-A2 Immune/Hematopoietic
    HNHFQ63 W88686 WO9854963-A2 Immune/Hematopoietic
    HAGDQ47 W88692 WO9854963-A2 Cancer
    HAICP19 W88693 WO9854963-A2 Cancer
    HCEQA68 W88699 WO9854963-A2 Neural/Sensory
    HCFNF11 W88701 WO9854963-A2 Cancer
    HCRBL20 W88702 WO9854963-A2 Cancer
    HDSAP81 W88704 WO9854963-A2 Cancer
    HE2CT29 W88705 WO9854963-A2 Mixed Fetal
    HE8MG65 W88706 WO9854963-A2 Cancer
    HE9FB42 W88707 WO9854963-A2 Cancer
    HEMAM41 W88708 WO9854963-A2 Cancer
    HEMCV19 W88709 WO9854963-A2 Cancer
    HETAR54 W88711 WO9854963-A2 Cancer
    HETBX14 W88712 WO9854963-A2 Cancer
    HFKFI40 W88714 WO9854963-A2 Cancer
    HFXHN68 W88715 WO9854963-A2 Cancer
    HGBFO79 W88716 WO9854963-A2 Cancer
    HGLAM56 W88717 WO9854963-A2 Cancer
    HHLBA89 W88718 WO9854963-A2 Digestive
    HIASB53 W88723 WO9854963-A2 Cancer
    HJABZ65 W88724 WO9854963-A2 Cancer
    HJPBB39 W88725 WO9854963-A2 Cancer
    HLHSK94 W88726 WO9854963-A2 Cancer
    HLMIW92 W88728 WO9854963-A2 Cancer
    HLTDB65 W88730 WO9854963-A2 Cancer
    HNFAH08 W88733 WO9854963-A2 Cancer
    HNGBE45 W88735 WO9854963-A2 Immune/Hematopoietic,
    Reproductive
    HNHCM59 W88737 WO9854963-A2 Cancer
    HCDEO95 W88740 WO9854963-A2 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HLMMJ13 W88741 WO9854963-A2 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HPTWA66 W88742 WO9854963-A2 Cancer
    HSAVU34 W88743 WO9854963-A2 Cancer
    HSQEO84 W88744 WO9854963-A2 Cancer
    HETCH46 W88745 WO9854963-A2 Cancer
    HWTBF59 W88746 WO9854963-A2 Cancer
    HCESF40 W88747 WO9854963-A2 Immune/Hematopoietic,
    Neural/Sensory
    HOFNZ45 W88748 WO9854963-A2 Reproductive
    HPWAN23 W88749 WO9854963-A2 Cancer
    HCRBL20 W88754 WO9854963-A2 Cancer
    HE8MG65 W88755 WO9854963-A2 Cancer
    HEMAM41 W88756 WO9854963-A2 Cancer
    HSAVU34 W88760 WO9854963-A2 Cancer
    HFHDN80 W88824 WO9854963-A2 Cardiovascular,
    Digestive,
    Immune/Hematopoietic
    HHFHR32 W88830 WO9854963-A2 Cancer
    HSKCP69 W89024 WO9854963-A2 Cancer
    HTLCU04 W89076 WO9854963-A2 Cancer
    HSKHZ53 W92460 US5871969-A Cancer
    HFIZH13 W94466 WO9900415-A1 Cancer
    HE9SF68 W97350 WO9903982-A1 Cancer
    HTECE94 Y00258 WO9906423-A1 Cancer
    HTWAH05 Y00259 WO9906423-A1 Cancer
    HAQAN31 Y00260 WO9906423-A1 Cancer
    HAUAQ39 Y00261 WO9906423-A1 Cancer
    HBNAU27 Y00262 WO9906423-A1 Cancer
    HSIDD28 Y00263 WO9906423-A1 Cancer
    HCABR41 Y00264 WO9906423-A1 Cancer
    HCUAQ30 Y00265 WO9906423-A1 Immune/Hematopoietic
    HE2AF21 Y00266 WO9906423-A1 Mixed Fetal
    HE2DC87 Y00267 WO9906423-A1 Mixed Fetal
    HE2PO86 Y00269 WO9906423-A1 Cancer
    HFCBD73 Y00272 WO9906423-A1 Cancer
    HSVAJ05 Y00273 WO9906423-A1 Cancer
    HLHSA86 Y00274 WO9906423-A1 Cancer
    H2CAA57 Y00278 WO9906423-A1 Cancer
    HADFV30 Y00279 WO9906423-A1 Cancer
    HAIBO71 Y00280 WO9906423-A1 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HAPAT76 Y00281 WO9906423-A1 Cancer
    HLHEB47 Y00282 WO9906423-A1 Cancer
    HLHEF54 Y00283 WO9906423-A1 Cancer
    HLMMJ78 Y00286 WO9906423-A1 Immune/Hematopoietic
    HLQBQ85 Y00287 WO9906423-A1 Cancer
    HLQBR11 Y00288 WO9906423-A1 Cancer
    HLWBZ56 Y00289 WO9906423-A1 Cancer
    HMCAR20 Y00292 WO9906423-A1 Cancer
    HMCAV55 Y00293 WO9906423-A1 Immune/Hematopoietic
    HMEFS61 Y00294 WO9906423-A1 Cardiovascular
    HMEJY78 Y00295 WO9906423-A1 Cancer
    HMWHH16 Y00298 WO9906423-A1 Immune/Hematopoietic
    HNFFC27 Y00300 WO9906423-A1 Immune/Hematopoietic
    HNFFC39 Y00301 WO9906423-A1 Immune/Hematopoietic,
    Reproductive
    HNGAM20 Y00302 WO9906423-A1 Immune/Hematopoietic
    HNGDS53 Y00304 WO9906423-A1 Immune/Hematopoietic
    HNGEW13 Y00307 WO9906423-A1 Immune/Hematopoietic
    HNGEY51 Y00308 WO9906423-A1 Immune/Hematopoietic
    HNGEZ47 Y00309 WO9906423-A1 Immune/Hematopoietic
    HNGFQ33 Y00310 WO9906423-A1 Immune/Hematopoietic
    HNGFU38 Y00311 WO9906423-A1 Immune/Hematopoietic
    HSKXE22 Y00313 WO9906423-A1 Cancer
    HNHBE49 Y00314 WO9906423-A1 Immune/Hematopoietic
    HNHEC59 Y00315 WO9906423-A1 Immune/Hematopoietic
    HNHEI54 Y00317 WO9906423-A1 Immune/Hematopoietic,
    Reproductive
    HNHER77 Y00318 WO9906423-A1 Immune/Hematopoietic
    HNHES40 Y00319 WO9906423-A1 Immune/Hematopoietic
    HNHEV43 Y00320 WO9906423-A1 Immune/Hematopoietic
    HNHFL46 Y00321 WO9906423-A1 Immune/Hematopoietic
    HNHFP80 Y00322 WO9906423-A1 Immune/Hematopoietic
    HNHFS63 Y00323 WO9906423-A1 Immune/Hematopoietic
    HNHGC56 Y00324 WO9906423-A1 Immune/Hematopoietic
    HRDEL61 Y00328 WO9906423-A1 Musculoskeletal
    HSAUC38 Y00329 WO9906423-A1 Immune/Hematopoietic
    HSAUF49 Y00330 WO9906423-A1 Immune/Hematopoietic
    HSAUK57 Y00331 WO9906423-A1 Immune/Hematopoietic
    HSAUL82 Y00332 WO9906423-A1 Immune/Hematopoietic
    HSAXI90 Y00333 WO9906423-A1 Immune/Hematopoietic
    HSDGW43 Y00335 WO9906423-A1 Neural/Sensory
    HSDJM31 Y00336 WO9906423-A1 Digestive,
    Neural/Sensory
    HSDJR23 Y00337 WO9906423-A1 Digestive,
    Neural/Sensory
    HSDMA90 Y00338 WO9906423-A1 Digestive,
    Endocrine,
    Neural/Sensory
    HSVAJ05 Y00340 WO9906423-A1 Cancer
    HAPAT76 Y00341 WO9906423-A1 Cancer
    HNGAM20 Y00344 WO9906423-A1 Immune/Hematopoietic
    HTXBK30 Y01135 WO9901020-A2 Cancer
    H2MBB56 Y01136 WO9901020-A2 Cancer
    HIBCW32 Y01138 WO9901020-A2 Cancer
    HLHCI58 Y01139 WO9901020-A2 Cancer
    HLMFG37 Y01140 WO9901020-A2 Cancer
    HBCAO31 Y01141 WO9901020-A2 Cancer
    HRDDR94 Y01142 WO9901020-A2 Cancer
    HSIDY06 Y01143 WO9901020-A2 Cancer
    HSKGO49 Y01144 WO9901020-A2 Cancer
    HBXGM67 Y01146 WO9901020-A2 Neural/Sensory
    HUFAC36 Y01147 WO9901020-A2 Cancer
    HAGBZ81 Y01148 WO9901020-A2 Excretory,
    Neural/Sensory
    HBJCK69 Y01150 WO9901020-A2 Immune/Hematopoietic
    HCACJ81 Y01152 WO9901020-A2 Cancer
    HBMWP47 Y01154 WO9901020-A2 Cancer
    HIBCW32 Y01155 WO9901020-A2 Cancer
    HCACJ81 Y01158 WO9901020-A2 Cancer
    HCE3F11 Y01206 WO9901020-A2 Digestive,
    Neural/Sensory
    HSXBV35 Y01383 WO9903990-A1 Neural/Sensory
    HTGAW51 Y01385 WO9903990-A1 Immune/Hematopoietic
    HTEGM07 Y01387 WO9903990-A1 Cancer
    HTWFK09 Y01389 WO9903990-A1 Immune/Hematopoietic
    HTXDJ88 Y01390 WO9903990-A1 Immune/Hematopoietic
    HUSGC54 Y01391 WO9903990-A1 Cardiovascular,
    Immune/Hematopoietic,
    Neural/Sensory
    HWTAD49 Y01392 WO9903990-A1 Cancer
    HWTBK81 Y01393 WO9903990-A1 Cancer
    HACBH16 Y01394 WO9903990-A1 Connective/Epithelial
    HCUDE16 Y01395 WO9903990-A1 Cancer
    HLWBZ73 Y01397 WO9903990-A1 Cancer
    HNGFR75 Y01398 WO9903990-A1 Immune/Hematopoietic
    HNHFO29 Y01400 WO9903990-A1 Immune/Hematopoietic
    HONAH29 Y01401 WO9903990-A1 Cancer
    HGCAB62 Y01402 WO9903990-A1 Cancer
    HAQBI01 Y01403 WO9903990-A1 Cancer
    HDPBA48 Y01405 WO9903990-A1 Immune/Hematopoietic
    HE6CT22 Y01406 WO9903990-A1 Mixed Fetal,
    Reproductive
    HE6CT56 Y01407 WO9903990-A1 Mixed Fetal,
    Neural/Sensory
    HE6CY88 Y01408 WO9903990-A1 Mixed Fetal
    HE9FT63 Y01409 WO9903990-A1 Cancer
    HE9ND43 Y01410 WO9903990-A1 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HERAN63 Y01411 WO9903990-A1 Connective/Epithelial,
    Reproductive
    HHBAG14 Y01413 WO9903990-A1 Cancer
    HMADU73 Y01417 WO9903990-A1 Cancer
    HMEAI74 Y01418 WO9903990-A1 Cancer
    HPMBZ15 Y01421 WO9903990-A1 Cancer
    HROAE16 Y01422 WO9903990-A1 Cancer
    HSAYM40 Y01423 WO9903990-A1 Immune/Hematopoietic
    HTBAB28 Y01426 WO9903990-A1 Immune/Hematopoietic
    HAQBT52 Y01428 WO9903990-A1 Cancer
    HBIBL04 Y01429 WO9903990-A1 Cancer
    HBJCI95 Y01430 WO9903990-A1 Cancer
    HBNBQ61 Y01431 WO9903990-A1 Reproductive
    HE2ID06 Y01432 WO9903990-A1 Cancer
    HEBCM63 Y01433 WO9903990-A1 Cancer
    HFFAK76 Y01434 WO9903990-A1 Neural/Sensory
    HFRBF28 Y01435 WO9903990-A1 Neural/Sensory
    HGBHM89 Y01436 WO9903990-A1 Cancer
    HLMBP18 Y01437 WO9903990-A1 Immune/Hematopoietic
    HAGFG63 Y01439 WO9903990-A1 Cancer
    HODAZ55 Y01440 WO9903990-A1 Reproductive
    HODDF08 Y01441 WO9903990-A1 Reproductive
    HOSDK95 Y01442 WO9903990-A1 Musculoskeletal
    HOUAR65 Y01443 WO9903990-A1 Connective/Epithelial
    HSVAC77 Y01444 WO9903990-A1 Cancer
    HRSMC69 Y01445 WO9903990-A1 Cancer
    HNECF34 Y01446 WO9903990-A1 Immune/Hematopoietic
    HAQAI46 Y01447 WO9903990-A1 Cancer
    HAQBI01 Y01448 WO9903990-A1 Cancer
    HJAAT30 Y01453 WO9903990-A1 Cancer
    HPMJI58 Y01458 WO9903990-A1 Cancer
    HNECF34 Y01477 WO9903990-A1 Immune/Hematopoietic
    HCEIA77 Y02650 WO9902546-A1 Cancer
    HCFCE10 Y02651 WO9902546-A1 Immune/Hematopoietic
    HCHAA63 Y02653 WO9902546-A1 Cancer
    HCNSP40 Y02654 WO9902546-A1 Cancer
    HDAAC10 Y02655 WO9902546-A1 Cardiovascular,
    Digestive,
    Reproductive
    HE8CV18 Y02656 WO9902546-A1 Cancer
    HFGAL10 Y02659 WO9902546-A1 Mixed Fetal,
    Neural/Sensory,
    Reproductive
    HFKEB72 Y02660 WO9902546-A1 Excretory,
    Reproductive
    HFTCU19 Y02661 WO9902546-A1 Cancer
    HFXHN31 Y02662 WO9902546-A1 Neural/Sensory
    HCEND31 Y02663 WO9902546-A1 Cancer
    HJABB94 Y02664 WO9902546-A1 Cancer
    HLTAI94 Y02666 WO9902546-A1 Immune/Hematopoietic,
    Reproductive
    HMELR03 Y02668 WO9902546-A1 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HMKAH10 Y02669 WO9902546-A1 Neural/Sensory,
    Reproductive
    HMKCW19 Y02670 WO9902546-A1 Cancer
    HMSJW18 Y02671 WO9902546-A1 Cancer
    HMWGY01 Y02672 WO9902546-A1 Immune/Hematopoietic
    HNFID82 Y02673 WO9902546-A1 Immune/Hematopoietic
    HNFIG36 Y02674 WO9902546-A1 Immune/Hematopoietic
    HNGEV29 Y02675 WO9902546-A1 Immune/Hematopoietic
    HNGJJ65 Y02677 WO9902546-A1 Immune/Hematopoietic
    HSLBF69 Y02687 WO9902546-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HSVBH58 Y02689 WO9902546-A1 Cancer
    HTADX17 Y02692 WO9902546-A1 Immune/Hematopoietic,
    Reproductive
    HTDAD22 Y02693 WO9902546-A1 Cancer
    HTEDS39 Y02694 WO9902546-A1 Cancer
    HTEHH53 Y02695 WO9902546-A1 Reproductive
    HTLDP69 Y02696 WO9902546-A1 Cancer
    HTPCS60 Y02698 WO9902546-A1 Cancer
    HUKBH05 Y02699 WO9902546-A1 Cancer
    HADFK68 Y02703 WO9902546-A1 Connective/Epithelial
    HADGG19 Y02704 WO9902546-A1 Connective/Epithelial,
    Musculoskeletal
    HAEAV45 Y02705 WO9902546-A1 Cardiovascular,
    Reproductive
    HARAA15 Y02706 WO9902546-A1 Neural/Sensory
    HBAFQ54 Y02708 WO9902546-A1 Cancer
    HBIAS26 Y02710 WO9902546-A1 Cancer
    HBJFU48 Y02711 WO9902546-A1 Immune/Hematopoietic
    HBJFV28 Y02712 WO9902546-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HBMWB01 Y02713 WO9902546-A1 Immune/Hematopoietic
    HBMXN79 Y02714 WO9902546-A1 Cancer
    HBMXP84 Y02715 WO9902546-A1 Cancer
    HCFMM26 Y02716 WO9902546-A1 Immune/Hematopoietic
    HCNAV36 Y02717 WO9902546-A1 Cancer
    HCNSB01 Y02718 WO9902546-A1 Cancer
    HCRBR74 Y02719 WO9902546-A1 Cancer
    HCUBN59 Y02720 WO9902546-A1 Immune/Hematopoietic
    HCUDB38 Y02721 WO9902546-A1 Immune/Hematopoietic
    HCUFZ62 Y02722 WO9902546-A1 Immune/Hematopoietic
    HDPCO25 Y02724 WO9902546-A1 Immune/Hematopoietic
    HDPHI51 Y02725 WO9902546-A1 Immune/Hematopoietic
    HE9FE83 Y02727 WO9902546-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Musculoskeletal
    HFPDE69 Y02731 WO9902546-A1 Neural/Sensory
    HGBGV89 Y02732 WO9902546-A1 Digestive
    HGLDE38 Y02733 WO9902546-A1 Cancer
    HHGDU58 Y02734 WO9902546-A1 Musculoskeletal
    HHTLF25 Y02735 WO9902546-A1 Cancer
    HKAFB88 Y02737 WO9902546-A1 Cancer
    HLHFP03 Y02738 WO9902546-A1 Respiratory
    HLYAF80 Y02741 WO9902546-A1 Immune/Hematopoietic
    HMKDD07 Y02743 WO9902546-A1 Immune/Hematopoietic,
    Neural/Sensory
    HMKDS08 Y02744 WO9902546-A1 Excretory,
    Neural/Sensory
    HMSHM14 Y02745 WO9902546-A1 Immune/Hematopoietic
    HMWDC28 Y02746 WO9902546-A1 Cancer
    HNFIU96 Y02749 WO9902546-A1 Immune/Hematopoietic
    HNGAX58 Y02751 WO9902546-A1 Immune/Hematopoietic
    HNHDL85 Y02754 WO9902546-A1 Immune/Hematopoietic
    HNHFU59 Y02755 WO9902546-A1 Immune/Hematopoietic
    HNHFW22 Y02756 WO9902546-A1 Immune/Hematopoietic
    HODCJ90 Y02758 WO9902546-A1 Cancer
    HPEBT80 Y02760 WO9902546-A1 Reproductive
    HSDAG05 Y02761 WO9902546-A1 Cancer
    HSDGR57 Y02762 WO9902546-A1 Cancer
    HSDJJ82 Y02763 WO9902546-A1 Neural/Sensory
    HSDZM95 Y02764 WO9902546-A1 Cancer
    HSKYU29 Y02766 WO9902546-A1 Cancer
    HSNAA55 Y02767 WO9902546-A1 Cancer
    HSQFP66 Y02768 WO9902546-A1 Excretory,
    Neural/Sensory
    HJPBB94 Y02769 WO9902546-A1 Cancer
    HSSJN64 Y02770 WO9902546-A1 Musculoskeletal
    HSVAQ28 Y02771 WO9902546-A1 Cancer
    HFTCU19 Y02775 WO9902546-A1 Cancer
    HGLAM53 Y02777 WO9902546-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HMKCW19 Y02778 WO9902546-A1 Cancer
    HMWGY01 Y02779 WO9902546-A1 Immune/Hematopoietic
    HSOAH66 Y02782 WO9902546-A1 Digestive
    HUKEX85 Y02785 WO9902546-A1 Musculoskeletal,
    Reproductive
    HSIDI15 Y02975 WO9902546-A1 Digestive,
    Immune/Hematopoietic
    HUKEJ46 Y03850 WO9909198-A1 Digestive,
    Reproductive
    HPASD50 Y04120 WO9909161-A1 Cancer
    HPASD50 Y04121 WO9909161-A1 Cancer
    HSDIT06 Y04295 WO9910363-A1 Neural/Sensory,
    Reproductive
    HSKEI54 Y04297 WO9910363-A1 Cancer
    HTNAG39 Y04300 WO9910363-A1 Cancer
    HTODL90 Y04301 WO9910363-A1 Immune/Hematopoietic
    HTWDC20 Y04302 WO9910363-A1 Immune/Hematopoietic
    HUFAT34 Y04303 WO9910363-A1 Cancer
    HAICJ23 Y04305 WO9910363-A1 Cancer
    HAPOF67 Y04306 WO9910363-A1 Digestive,
    Excretory,
    Musculoskeletal
    HE8DG53 Y04308 WO9910363-A1 Cancer
    HFSAY85 Y04309 WO9910363-A1 Cancer
    HHEDD41 Y04310 WO9910363-A1 Cancer
    HKCSO46 Y04311 WO9910363-A1 Cancer
    HKGAV60 Y04312 WO9910363-A1 Cancer
    HKGDJ66 Y04314 WO9910363-A1 Cancer
    HMCDK27 Y04315 WO9910363-A1 Cancer
    HMCDX48 Y04316 WO9910363-A1 Cancer
    HMIAS24 Y04317 WO9910363-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNFEG11 Y04318 WO9910363-A1 Immune/Hematopoietic
    HNGEP09 Y04319 WO9910363-A1 Immune/Hematopoietic
    HTXKK52 Y04320 WO9910363-A1 Immune/Hematopoietic
    HNGJP90 Y04321 WO9910363-A1 Immune/Hematopoietic
    HFVIF40 Y06461 WO9931116-A1 Cancer
    HFCCQ50 Y06462 WO9931116-A1 Cancer
    HDPIE88 Y06511 WO9936565-A1 Cancer
    HCWHN10 Y07746 WO9909155-A1 Immune/Hematopoietic
    HDTAE40 Y07748 WO9909155-A1 Digestive,
    Immune/Hematopoietic
    HE8DY08 Y07751 WO9909155-A1 Cancer
    HE9ND27 Y07753 WO9909155-A1 Cancer
    HCE3G69 Y07754 WO9909155-A1 Cancer
    HEAAX57 Y07755 WO9909155-A1 Reproductive
    HEMGD15 Y07759 WO9909155-A1 Cancer
    HEQBR95 Y07760 WO9909155-A1 Cancer
    HFKGE44 Y07764 WO9909155-A1 Cancer
    HFPCY39 Y07765 WO9909155-A1 Cancer
    HFXDX75 Y07768 WO9909155-A1 Neural/Sensory
    HFXJC53 Y07770 WO9909155-A1 Neural/Sensory,
    Reproductive,
    Respiratory
    HFXJW48 Y07771 WO9909155-A1 Cancer
    HGBGO11 Y07772 WO9909155-A1 Cancer
    HGBHM10 Y07773 WO9909155-A1 Cancer
    HSWAY58 Y07776 WO9909155-A1 Cancer
    HTEIM65 Y07779 WO9909155-A1 Immune/Hematopoietic,
    Reproductive
    HTHBX95 Y07780 WO9909155-A1 Cancer
    HTLDQ56 Y07781 WO9909155-A1 Reproductive
    HTOFU06 Y07782 WO9909155-A1 Immune/Hematopoietic,
    Musculoskeletal
    HTWEE31 Y07785 WO9909155-A1 Immune/Hematopoietic
    HUSAO56 Y07789 WO9909155-A1 Cancer
    HUSIJ08 Y07790 WO9909155-A1 Cancer
    HAGBD57 Y07791 WO9909155-A1 Excretory,
    Neural/Sensory
    HBAFA04 Y07793 WO9909155-A1 Cancer
    HBJES16 Y07794 WO9909155-A1 Cancer
    HCEFZ05 Y07796 WO9909155-A1 Mixed Fetal,
    Neural/Sensory,
    HCFMX95 Y07797 WO9909155-A1 Immune/Hematopoietic
    HLYHA71 Y07798 WO9909155-A1 Cancer
    HEBAL06 Y07800 WO9909155-A1 Neural/Sensory
    HEIAB33 Y07801 WO9909155-A1 Cancer
    HEPBC02 Y07802 WO9909155-A1 Cancer
    HFTBY96 Y07803 WO9909155-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HKMMM61 Y07804 WO9909155-A1 Cancer
    HLQBQ38 Y07806 WO9909155-A1 Cancer
    HMKCP66 Y07807 WO9909155-A1 Neural/Sensory
    HWTAL40 Y07808 WO9909155-A1 Cancer
    HNHDR03 Y07809 WO9909155-A1 Immune/Hematopoietic
    HNHFH41 Y07810 WO9909155-A1 Immune/Hematopoietic
    HNHFI81 Y07811 WO9909155-A1 Immune/Hematopoietic
    HOSFQ28 Y07812 WO9909155-A1 Cancer
    HPRAL78 Y07813 WO9909155-A1 Cancer
    HEAAA85 Y07814 WO9909155-A1 Cancer
    HDTAR09 Y07816 WO9909155-A1 Cancer
    HLYHA71 Y07843 WO9909155-A1 Cancer
    HCWCH14 Y07852 WO9918208-A1 Immune/Hematopoietic
    HE9MI43 Y07855 WO9918208-A1 Cancer
    HE2PI29 Y07859 WO9918208-A1 Cancer
    HLHDP83 Y07862 WO9918208-A1 Cancer
    HSIAS17 Y07863 WO9918208-A1 Cancer
    HOSDG32 Y07866 WO9918208-A1 Cancer
    HMUBU59 Y07867 WO9918208-A1 Cancer
    HWTCE21 Y07868 WO9918208-A1 Cancer
    HFIUM15 Y07869 WO9918208-A1 Cancer
    HTLAF13 Y07872 WO9918208-A1 Reproductive
    HTLFI93 Y07873 WO9918208-A1 Immune/Hematopoietic,
    Reproductive,
    Respiratory
    HBXGI20 Y07874 WO9918208-A1 Cancer
    HTPBH21 Y07875 WO9918208-A1 Connective/Epithelial,
    Digestive,
    Reproductive
    HSQAB87 Y07876 WO9918208-A1 Cancer
    HTEDJ94 Y07877 WO9918208-A1 Cancer
    HKMLM11 Y07878 WO9918208-A1 Cancer
    HNEAC05 Y07879 WO9918208-A1 Immune/Hematopoietic
    HETEW02 Y07880 WO9918208-A1 Cancer
    HLMCA59 Y07882 WO9918208-A1 Immune/Hematopoietic
    HOAAC90 Y07883 WO9918208-A1 Musculoskeletal
    HMEJQ68 Y07884 WO9918208-A1 Cancer
    HRTAE58 Y07888 WO9918208-A1 Digestive,
    Reproductive
    HSKNB54 Y07889 WO9918208-A1 Cancer
    HSKNT34 Y07890 WO9918208-A1 Cancer
    HTEDY42 Y07891 WO9918208-A1 Reproductive
    HTLAA40 Y07892 WO9918208-A1 Reproductive
    HTNBO91 Y07893 WO9918208-A1 Cancer
    H6BSD90 Y07894 WO9918208-A1 Cancer
    HBJBQ35 Y07895 WO9918208-A1 Immune/Hematopoietic
    HCE1Q89 Y07896 WO9918208-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCNSB61 Y07897 WO9918208-A1 Digestive,
    Immune/Hematopoietic
    HCDBO20 Y07898 WO9918208-A1 Musculoskeletal,
    Respiratory
    HBNAW17 Y07899 WO9918208-A1 Reproductive
    HEAAH81 Y07902 WO9918208-A1 Cancer
    HEBAE88 Y07903 WO9918208-A1 Immune/Hematopoietic,
    Neural/Sensory
    HFXGV31 Y07904 WO9918208-A1 Neural/Sensory
    HEAAJ57 Y07905 WO9918208-A1 Immune/Hematopoietic,
    Reproductive
    HCFMV71 Y07906 WO9918208-A1 Immune/Hematopoietic
    HGBDL30 Y07910 WO9918208-A1 Digestive
    HFKEN81 Y07911 WO9918208-A1 Excretory,
    Neural/Sensory
    HFPCX36 Y07912 WO9918208-A1 Neural/Sensory
    HFRAN90 Y07913 WO9918208-A1 Neural/Sensory
    HHGBO91 Y07915 WO9918208-A1 Digestive,
    Reproductive
    HERAN54 Y07917 WO9918208-A1 Connective/Epithelial
    HFXDE67 Y07918 WO9918208-A1 Neural/Sensory
    HFFAD59 Y07921 WO9918208-A1 Neural/Sensory
    HMDAE65 Y07923 WO9918208-A1 Neural/Sensory
    HMEGF92 Y07925 WO9918208-A1 Cardiovascular
    HNGIK36 Y07926 WO9918208-A1 Immune/Hematopoietic
    HMEJJ27 Y07927 WO9918208-A1 Cardiovascular
    HNHCY64 Y07928 WO9918208-A1 Immune/Hematopoietic
    HNHCY94 Y07929 WO9918208-A1 Immune/Hematopoietic
    HNEBN76 Y07930 WO9918208-A1 Immune/Hematopoietic,
    Reproductive,
    Respiratory
    HMEFT54 Y07931 WO9918208-A1 Cardiovascular,
    Musculoskeletal,
    Reproductive
    HLQBE09 Y07932 WO9918208-A1 Digestive
    HMWBC11 Y07933 WO9918208-A1 Immune/Hematopoietic
    HNGJR78 Y07934 WO9918208-A1 Immune/Hematopoietic
    HNGDP26 Y07935 WO9918208-A1 Immune/Hematopoietic
    HNGJH63 Y07936 WO9918208-A1 Immune/Hematopoietic
    HMDAL04 Y07937 WO9918208-A1 Neural/Sensory
    HMWHX28 Y07938 WO9918208-A1 Immune/Hematopoietic
    HNHGB09 Y07942 WO9918208-A1 Immune/Hematopoietic
    HNHHA15 Y07943 WO9918208-A1 Immune/Hematopoietic
    HHGDC01 Y07944 WO9918208-A1 Cancer
    HMWGU74 Y07945 WO9918208-A1 Immune/Hematopoietic
    HNGCF72 Y07946 WO9918208-A1 Immune/Hematopoietic
    HOACB38 Y07947 WO9918208-A1 Musculoskeletal
    HLMFD11 Y07950 WO9918208-A1 Immune/Hematopoietic
    HLYBA22 Y07952 WO9918208-A1 Immune/Hematopoietic
    HCWCH14 Y07953 WO9918208-A1 Immune/Hematopoietic
    HBMWF85 Y10797 WO9907891-A1 Immune/Hematopoietic
    HCDEJ37 Y10798 WO9907891-A1 Immune/Hematopoietic,
    Musculoskeletal
    HCE3L18 Y10799 WO9907891-A1 Neural/Sensory
    HCYBI42 Y10800 WO9907891-A1 Cancer
    HE6FB81 Y10801 WO9907891-A1 Mixed Fetal
    HFAMB72 Y10802 WO9907891-A1 Cancer
    HFCDW42 Y10803 WO9907891-A1 Cancer
    HFPAE26 Y10804 WO9907891-A1 Neural/Sensory
    HFXJM91 Y10805 WO9907891-A1 Cancer
    HJABX32 Y10807 WO9907891-A1 Cancer
    HJMBW30 Y10808 WO9907891-A1 Cancer
    HSVAT02 Y10810 WO9907891-A1 Cancer
    HSVBM90 Y10811 WO9907891-A1 Cancer
    HSYBL17 Y10812 WO9907891-A1 Cancer
    HTEBI28 Y10813 WO9907891-A1 Reproductive
    HTPDS14 Y10814 WO9907891-A1 Cancer
    HTSGG36 Y10815 WO9907891-A1 Cancer
    HODCJ27 Y10816 WO9907891-A1 Cancer
    HTXDB52 Y10819 WO9907891-A1 Immune/Hematopoietic,
    Musculoskeletal
    HTXDP60 Y10820 WO9907891-A1 Cancer
    HTXEB42 Y10821 WO9907891-A1 Cancer
    HBAFZ29 Y10824 WO9907891-A1 Cancer
    HBAHA77 Y10826 WO9907891-A1 Cancer
    HBJEW84 Y10827 WO9907891-A1 Immune/Hematopoietic
    HBJFE12 Y10828 WO9907891-A1 Immune/Hematopoietic
    HCFBM53 Y10830 WO9907891-A1 Cancer
    HCFBQ81 Y10831 WO9907891-A1 Immune/Hematopoietic
    HCFCI07 Y10832 WO9907891-A1 Immune/Hematopoietic
    HCFDD76 Y10833 WO9907891-A1 Cancer
    HCFMJ81 Y10834 WO9907891-A1 Cancer
    HCFOG45 Y10835 WO9907891-A1 Cancer
    HCUBN71 Y10836 WO9907891-A1 Immune/Hematopoietic,
    Reproductive
    HHEMA75 Y10837 WO9907891-A1 Cancer
    HHPTJ65 Y10839 WO9907891-A1 Cardiovascular,
    Musculoskeletal,
    Neural/Sensory
    HHSDR11 Y10840 WO9907891-A1 Neural/Sensory
    HLJDQ62 Y10842 WO9907891-A1 Cancer
    HKGBS49 Y10843 WO9907891-A1 Reproductive
    HKISA27 Y10844 WO9907891-A1 Cancer
    HKIXE06 Y10845 WO9907891-A1 Cancer
    HKMMV77 Y10846 WO9907891-A1 Excretory,
    Reproductive
    HLYAB80 Y10850 WO9907891-A1 Cancer
    HLYAG19 Y10851 WO9907891-A1 Digestive,
    Immune/Hematopoietic
    HLYBY48 Y10852 WO9907891-A1 Immune/Hematopoietic
    HMUAW28 Y10853 WO9907891-A1 Immune/Hematopoietic,
    Musculoskeletal
    HMWHC36 Y10854 WO9907891-A1 Cancer
    HNFIS82 Y10856 WO9907891-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HNGBO16 Y10859 WO9907891-A1 Immune/Hematopoietic
    HNGBQ90 Y10860 WO9907891-A1 Cancer
    HNGBV72 Y10861 WO9907891-A1 Immune/Hematopoietic
    HNGEG08 Y10863 WO9907891-A1 Immune/Hematopoietic
    HNGFI02 Y10864 WO9907891-A1 Immune/Hematopoietic
    HNGGF85 Y10865 WO9907891-A1 Immune/Hematopoietic
    HNGHM75 Y10866 WO9907891-A1 Immune/Hematopoietic
    HNGIN84 Y10867 WO9907891-A1 Digestive,
    Endocrine,
    Immune/Hematopoietic
    HNGJH08 Y10869 WO9907891-A1 Immune/Hematopoietic
    HNHAH01 Y10870 WO9907891-A1 Immune/Hematopoietic
    HNHET53 Y10871 WO9907891-A1 Immune/Hematopoietic
    HOABP21 Y10872 WO9907891-A1 Cancer
    HODAA12 Y10873 WO9907891-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HFKDH44 Y10874 WO9907891-A1 Cancer
    HOVAP06 Y10875 WO9907891-A1 Reproductive
    HPEAE34 Y10876 WO9907891-A1 Reproductive
    HPTRO86 Y10877 WO9907891-A1 Cancer
    HSAXJ60 Y10878 WO9907891-A1 Immune/Hematopoietic
    HSAXM32 Y10879 WO9907891-A1 Cancer
    HSKND71 Y10882 WO9907891-A1 Mixed Fetal,
    Musculoskeletal,
    Neural/Sensory
    HSOAC84 Y10883 WO9907891-A1 Digestive
    HFKCF34 Y10884 WO9907891-A1 Cancer
    HSAAO30 Y12916 WO9911293-A1 Cancer
    HSQBL21 Y12917 WO9911293-A1 Cancer
    HTEFU41 Y12919 WO9911293-A1 Immune/Hematopoietic,
    Reproductive
    HDPSP54 Y12920 WO9911293-A1 Cancer
    HELFQ07 Y12921 WO9911293-A1 Cancer
    HBSAJ16 Y12923 WO9911293-A1 Connective/Epithelial,
    Musculoskeletal,
    Reproductive
    HCEOC41 Y12924 WO9911293-A1 Cancer
    HCUEO60 Y12926 WO9911293-A1 Immune/Hematopoietic
    HDHEB60 Y12927 WO9911293-A1 Cancer
    HE6AJ31 Y12928 WO9911293-A1 Mixed Fetal
    HFCED59 Y12929 WO9911293-A1 Immune/Hematopoietic,
    Neural/Sensory
    HFXKJ03 Y12931 WO9911293-A1 Cardiovascular,
    Immune/Hematopoietic,
    Neural/Sensory
    HHFDG44 Y12932 WO9911293-A1 Cardiovascular,
    Endocrine,
    Immune/Hematopoietic
    HJACG02 Y12933 WO9911293-A1 Digestive,
    Immune/Hematopoietic
    HKGAJ54 Y12934 WO9911293-A1 Cancer
    HKMAB92 Y12935 WO9911293-A1 Cancer
    HLMFC54 Y12937 WO9911293-A1 Immune/Hematopoietic
    HLWBZ21 Y12939 WO9911293-A1 Immune/Hematopoietic,
    Reproductive
    HMJAX71 Y12940 WO9911293-A1 Neural/Sensory
    HNECU95 Y12941 WO9911293-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HNFCK41 Y12942 WO9911293-A1 Cancer
    HNFHD08 Y12943 WO9911293-A1 Cancer
    HNGEW65 Y12944 WO9911293-A1 Endocrine,
    Immune/Hematopoietic
    HNHEN68 Y12946 WO9911293-A1 Immune/Hematopoietic
    HNHFG05 Y12947 WO9911293-A1 Immune/Hematopoietic
    HODBF19 Y12948 WO9911293-A1 Cancer
    HOEBK34 Y12949 WO9911293-A1 Digestive,
    Musculoskeletal
    HPBCC51 Y12950 WO9911293-A1 Cancer
    HRGDC48 Y12951 WO9911293-A1 Immune/Hematopoietic,
    Musculoskeletal
    HSDJB13 Y12952 WO9911293-A1 Cancer
    HTEHR24 Y12953 WO9911293-A1 Cancer
    HARAO51 Y12957 WO9911293-A1 Cancer
    HATAA15 Y12958 WO9911293-A1 Cancer
    HATCK44 Y12959 WO9911293-A1 Cancer
    HBIAE26 Y12960 WO9911293-A1 Neural/Sensory,
    Reproductive
    HBMXG32 Y12961 WO9911293-A1 Immune/Hematopoietic
    HCDAT43 Y12963 WO9911293-A1 Cancer
    HSLJB89 Y12964 WO9911293-A1 Cancer
    HBAFC77 Y12966 WO9911293-A1 Cancer
    HSAAO30 Y12969 WO9911293-A1 Cancer
    HFCET92 Y14078 WO9921575-A1 Cancer
    HSIDU19 Y14411 WO9919339-A1 Digestive
    HPRSB76 Y14412 WO9919339-A1 Reproductive
    HTEIL66 Y14413 WO9919339-A1 Reproductive
    HSABG21 Y14415 WO9919339-A1 Cancer
    HSAXB32 Y14416 WO9919339-A1 Immune/Hematopoietic
    HPEAD48 Y14417 WO9919339-A1 Reproductive
    HPVAB94 Y14418 WO9919339-A1 Reproductive
    HSAXB81 Y14419 WO9919339-A1 Immune/Hematopoietic
    HSLCU73 Y14421 WO9919339-A1 Musculoskeletal
    HTEIP36 Y14423 WO9919339-A1 Reproductive
    HYBAY77 Y14424 WO9919339-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HROAE78 Y14425 WO9919339-A1 Digestive
    HSAVP17 Y14426 WO9919339-A1 Immune/Hematopoietic
    HSIEA14 Y14427 WO9919339-A1 Digestive
    HPEAD79 Y14430 WO9919339-A1 Reproductive
    HRDED19 Y14431 WO9919339-A1 Musculoskeletal
    HSAYS89 Y14432 WO9919339-A1 Immune/Hematopoietic
    HTODK73 Y14433 WO9919339-A1 Cancer
    HSVAM10 Y14434 WO9919339-A1 Cancer
    HSPAA60 Y14439 WO9919339-A1 Digestive
    HFAEF57 Y14440 WO9919339-A1 Neural/Sensory
    HEGAH43 Y14441 WO9919339-A1 Digestive,
    Reproductive
    HNGBX63 Y14443 WO9919339-A1 Immune/Hematopoietic
    HE2AG50 Y14444 WO9919339-A1 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HCUIN80 Y14445 WO9919339-A1 Immune/Hematopoietic
    HADCL29 Y14446 WO9919339-A1 Connective/Epithelial
    HAPPS89 Y14447 WO9919339-A1 Cancer
    HFGAH44 Y14448 WO9919339-A1 Cancer
    HFIHZ96 Y14449 WO9919339-A1 Musculoskeletal
    HFIUR10 Y14450 WO9919339-A1 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HLDNA86 Y14451 WO9919339-A1 Cancer
    HCUIO20 Y14453 WO9919339-A1 Immune/Hematopoietic
    HLTEF12 Y14454 WO9919339-A1 Cancer
    HCFBJ91 Y14455 WO9919339-A1 Immune/Hematopoietic
    HHFHP90 Y14456 WO9919339-A1 Cardiovascular
    HLYCQ48 Y14457 WO9919339-A1 Immune/Hematopoietic
    HHLAB07 Y14458 WO9919339-A1 Digestive,
    Immune/Hematopoietic
    HFOXE30 Y14459 WO9919339-A1 Musculoskeletal
    HBJEL68 Y14460 WO9919339-A1 Immune/Hematopoietic,
    Neural/Sensory
    HFIUR35 Y14462 WO9919339-A1 Musculoskeletal
    HFIZF58 Y16587 US5916769-A Cancer
    HNGDJ72 Y19443 WO9922243-A1 Immune/Hematopoietic
    HNGEO29 Y19444 WO9922243-A1 Immune/Hematopoietic
    HNHDL95 Y19445 WO9922243-A1 Immune/Hematopoietic
    HAGDS35 Y19446 WO9922243-A1 Cancer
    HNGEQ48 Y19447 WO9922243-A1 Immune/Hematopoietic
    HNGDG40 Y19448 WO9922243-A1 Immune/Hematopoietic
    HNGEN81 Y19449 WO9922243-A1 Immune/Hematopoietic
    H2MAC30 Y19450 WO9922243-A1 Cancer
    HNHFB16 Y19451 WO9922243-A1 Immune/Hematopoietic
    HPFCL43 Y19452 WO9922243-A1 Cancer
    HSATR82 Y19453 WO9922243-A1 Immune/Hematopoietic
    HNHIC21 Y19455 WO9922243-A1 Immune/Hematopoietic
    HOVCA92 Y19456 WO9922243-A1 Immune/Hematopoietic,
    Reproductive
    HSDIL30 Y19458 WO9922243-A1 Neural/Sensory
    HATDB65 Y19459 WO9922243-A1 Endocrine,
    Reproductive,
    Respiratory
    HTTEA24 Y19461 WO9922243-A1 Digestive,
    Reproductive
    HAGDS20 Y19462 WO9922243-A1 Neural/Sensory,
    Reproductive
    HSDJM30 Y19463 WO9922243-A1 Digestive,
    Neural/Sensory
    HNHEE88 Y19464 WO9922243-A1 Immune/Hematopoietic
    HSLFD55 Y19465 WO9922243-A1 Musculoskeletal
    HSAXJ29 Y19466 WO9922243-A1 Immune/Hematopoietic
    HSFAM39 Y19467 WO9922243-A1 Reproductive
    HADDZ85 Y19469 WO9922243-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPCM26 Y19470 WO9922243-A1 Cancer
    HSZAA13 Y19471 WO9922243-A1 Cancer
    HDTBP04 Y19472 WO9922243-A1 Digestive,
    Immune/Hematopoietic
    HHGCQ54 Y19473 WO9922243-A1 Cancer
    HSNAB12 Y19474 WO9922243-A1 Cardiovascular
    HBJID05 Y19475 WO9922243-A1 Immune/Hematopoietic
    HSNBM49 Y19476 WO9922243-A1 Cancer
    HJMBF77 Y19477 WO9922243-A1 Cancer
    HJMBM38 Y19478 WO9922243-A1 Cancer
    HHGCL33 Y19479 WO9922243-A1 Cancer
    HCEWE20 Y19480 WO9922243-A1 Endocrine,
    Immune/Hematopoietic,
    Neural/Sensory
    HCUHL13 Y19481 WO9922243-A1 Immune/Hematopoietic
    HBJHO68 Y19482 WO9922243-A1 Immune/Hematopoietic
    HCWDV84 Y19483 WO9922243-A1 Immune/Hematopoietic
    HBXFC78 Y19484 WO9922243-A1 Cancer
    HE2F145 Y19485 WO9922243-A1 Cancer
    HEOMG13 Y19486 WO9922243-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HFAMH77 Y19487 WO9922243-A1 Cancer
    HSVCF20 Y19488 WO9922243-A1 Cancer
    HISAG02 Y19489 WO9922243-A1 Cancer
    HCDAF84 Y19490 WO9922243-A1 Musculoskeletal
    HHAAC17 Y19491 WO9922243-A1 Digestive,
    Musculoskeletal,
    Neural/Sensory
    HEQAG39 Y19493 WO9922243-A1 Cancer
    HKACH44 Y19494 WO9922243-A1 Cancer
    HBNBG49 Y19495 WO9922243-A1 Cancer
    HE2EN04 Y19496 WO9922243-A1 Cancer
    HSVAA10 Y19497 WO9922243-A1 Cardiovascular
    HFPBA88 Y19498 WO9922243-A1 Cancer
    HHEBW54 Y19500 WO9922243-A1 Cancer
    HFEBH21 Y19501 WO9922243-A1 Connective/Epithelial,
    Reproductive
    HFTDZ36 Y19502 WO9922243-A1 Cancer
    HGLAW96 Y19503 WO9922243-A1 Immune/Hematopoietic,
    Neural/Sensory
    HKAFK41 Y19504 WO9922243-A1 Cancer
    HOSEG51 Y19505 WO9922243-A1 Endocrine,
    Immune/Hematopoietic,
    Musculoskeletal
    HTEJT39 Y19506 WO9922243-A1 Neural/Sensory,
    Reproductive
    HPTRH45 Y19507 WO9922243-A1 Cancer
    HDHMA72 Y19508 WO9922243-A1 Cancer
    HNTBL27 Y19509 WO9922243-A1 Cancer
    HCFMX35 Y19510 WO9922243-A1 Immune/Hematopoietic
    HMUAO21 Y19512 WO9922243-A1 Cancer
    HCHAR28 Y19513 WO9922243-A1 Cancer
    HLYDU25 Y19514 WO9922243-A1 Immune/Hematopoietic
    HOEJH89 Y19515 WO9922243-A1 Cancer
    HPFDG48 Y19516 WO9922243-A1 Immune/Hematopoietic,
    Reproductive
    HWTBM18 Y19517 WO9922243-A1 Immune/Hematopoietic,
    Musculoskeletal
    HCFOM18 Y19518 WO9922243-A1 Immune/Hematopoietic
    HMWFO02 Y19519 WO9922243-A1 Immune/Hematopoietic
    HNGAV42 Y19520 WO9922243-A1 Immune/Hematopoietic
    HSDSE75 Y19522 WO9922243-A1 Musculoskeletal,
    Neural/Sensory,
    Respiratory
    HLMFD85 Y19523 WO9922243-A1 Immune/Hematopoietic
    HLQCJ74 Y19524 WO9922243-A1 Digestive,
    Immune/Hematopoietic
    HTEFU65 Y19526 WO9922243-A1 Excretory,
    Immune/Hematopoietic,
    Reproductive
    HLYBF22 Y19527 WO9922243-A1 Immune/Hematopoietic,
    Mixed Fetal
    HMDAP35 Y19528 WO9922243-A1 Neural/Sensory
    HWBCN75 Y19530 WO9922243-A1 Cancer
    HROAH06 Y19531 WO9922243-A1 Digestive,
    Immune/Hematopoietic
    HSAXA83 Y19532 WO9922243-A1 Immune/Hematopoietic
    HSDJE10 Y19533 WO9922243-A1 Cancer
    HBAMA40 Y19534 WO9922243-A1 Excretory
    HBAMB34 Y19535 WO9922243-A1 Excretory,
    Reproductive
    HCWKC15 Y19536 WO9922243-A1 Immune/Hematopoietic
    HDTDM65 Y19537 WO9922243-A1 Cancer
    HMMBF71 Y19538 WO9922243-A1 Immune/Hematopoietic
    HPBDH41 Y19539 WO9922243-A1 Immune/Hematopoietic,
    Musculoskeletal
    HPBEN24 Y19540 WO9922243-A1 Cancer
    HCUIM65 Y19541 WO9922243-A1 Cancer
    HKNAA95 Y19542 WO9922243-A1 Digestive,
    Excretory,
    Immune/Hematopoietic
    HKIYH57 Y19543 WO9922243-A1 Cancer
    HBJMG49 Y19546 WO9922243-A1 Immune/Hematopoietic
    H6EDC19 Y19547 WO9922243-A1 Cancer
    HSKHZ81 Y19548 WO9922243-A1 Cancer
    HBJFX78 Y19549 WO9922243-A1 Cancer
    HEMFS60 Y19550 WO9922243-A1 Cancer
    HKACB56 Y19551 WO9922243-A1 Connective/Epithelial
    HTXJX80 Y19552 WO9922243-A1 Digestive,
    Immune/Hematopoietic
    HAFBD61 Y19553 WO9922243-A1 Cancer
    HBJJU28 Y19554 WO9922243-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNHEI47 Y19555 WO9922243-A1 Immune/Hematopoietic
    HPMFY74 Y19556 WO9922243-A1 Reproductive
    HLYAP91 Y19559 WO9922243-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HSKNB56 Y19560 WO9922243-A1 Cancer
    HHGCW91 Y19561 WO9922243-A1 Digestive,
    Immune/Hematopoietic
    HKIYE96 Y19562 WO9922243-A1 Excretory
    HLYAN59 Y19563 WO9922243-A1 Immune/Hematopoietic
    HNEEE24 Y19564 WO9922243-A1 Immune/Hematopoietic
    HAPRK85 Y19565 WO9922243-A1 Cancer
    HLTEJ06 Y19566 WO9922243-A1 Immune/Hematopoietic
    HMEKT48 Y19567 WO9922243-A1 Cancer
    HNGHR74 Y19568 WO9922243-A1 Immune/Hematopoietic
    HNHED17 Y19569 WO9922243-A1 Immune/Hematopoietic
    HNHEP59 Y19570 WO9922243-A1 Immune/Hematopoietic
    HNHFJ25 Y19571 WO9922243-A1 Immune/Hematopoietic
    HCPAA69 Y19572 WO9922243-A1 Neural/Sensory
    HEAAR07 Y19573 WO9922243-A1 Reproductive
    HHGDW43 Y19574 WO9922243-A1 Cancer
    HHSDX28 Y19575 WO9922243-A1 Immune/Hematopoietic,
    Neural/Sensory
    HE8ER60 Y19576 WO9922243-A1 Cancer
    HMEJQ66 Y19577 WO9922243-A1 Cardiovascular
    HRDAD66 Y19578 WO9922243-A1 Cancer
    HCMST14 Y19579 WO9922243-A1 Cancer
    HCEBA03 Y19580 WO9922243-A1 Neural/Sensory
    HJAAM10 Y19582 WO9922243-A1 Cancer
    HOHCC74 Y19584 WO9922243-A1 Cancer
    HPMPY57 Y19585 WO9922243-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HFXDN63 Y19586 WO9922243-A1 Neural/Sensory
    HADCL76 Y19587 WO9922243-A1 Cancer
    HMMAS76 Y19588 WO9922243-A1 Endocrine,
    Immune/Hematopoietic
    HMKCG09 Y19589 WO9922243-A1 Digestive,
    Endocrine,
    Neural/Sensory
    HFPBA88 Y19590 WO9922243-A1 Cancer
    HMIAH29 Y19596 WO9922243-A1 Cancer
    HEMFS60 Y19757 WO9922243-A1 Cancer
    HDPVA94 Y25708 WO9938882-A1 Cancer
    HDPNE25 Y25709 WO9938882-A1 Cancer
    HASCG84 Y25711 WO9938881-A1 Cancer
    HDPCY37 Y25712 WO9938881-A1 Cancer
    HHEBB10 Y25713 WO9938881-A1 Cancer
    HNGJA38 Y25714 WO9938881-A1 Immune/Hematopoietic
    HHENL07 Y25715 WO9938881-A1 Immune/Hematopoietic
    HKADQ91 Y25716 WO9938881-A1 Cancer
    HPMCV18 Y25717 WO9938881-A1 Musculoskeletal,
    Reproductive
    HKGAK22 Y25718 WO9938881-A1 Endocrine,
    Excretory,
    Neural/Sensory
    HTEHU31 Y25719 WO9938881-A1 Cancer
    HFXAM76 Y25720 WO9938881-A1 Cancer
    HFXDZ79 Y25721 WO9938881-A1 Neural/Sensory
    HOHBC68 Y25722 WO9938881-A1 Cancer
    HSVAM81 Y25723 WO9938881-A1 Cancer
    HTXDG40 Y25724 WO9938881-A1 Immune/Hematopoietic
    HE2FC81 Y25725 WO9938881-A1 Mixed Fetal
    HJACE05 Y25726 WO9938881-A1 Cancer
    HADCW30 Y25727 WO9938881-A1 Connective/Epithelial
    HBMDK25 Y25728 WO9938881-A1 Immune/Hematopoietic
    HFXKK25 Y25729 WO9938881-A1 Cancer
    HHEMO80 Y25730 WO9938881-A1 Immune/Hematopoietic
    HNGEJ53 Y25731 WO9938881-A1 Immune/Hematopoietic
    HTBAA70 Y25732 WO9938881-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HSAYB43 Y25734 WO9938881-A1 Immune/Hematopoietic
    HSLDS32 Y25735 WO9938881-A1 Cancer
    HMIAV27 Y25736 WO9938881-A1 Cancer
    HSQEH50 Y25737 WO9938881-A1 Cancer
    HKMMU22 Y25738 WO9938881-A1 Excretory
    HKMMD13 Y25739 WO9938881-A1 Excretory
    HLDNK64 Y25740 WO9938881-A1 Cancer
    HRDES01 Y25741 WO9938881-A1 Musculoskeletal
    HDTDZ50 Y25742 WO9938881-A1 Cancer
    HETAB45 Y25743 WO9938881-A1 Cancer
    HFPBD47 Y25744 WO9938881-A1 Cancer
    HJMBI18 Y25745 WO9938881-A1 Cancer
    HFXHK73 Y25746 WO9938881-A1 Neural/Sensory
    HJMBT65 Y25747 WO9938881-A1 Cancer
    HWHGZ26 Y25748 WO9938881-A1 Cancer
    HADFY83 Y25749 WO9938881-A1 Cancer
    HBMTV78 Y25750 WO9938881-A1 Digestive,
    Immune/Hematopoietic
    HTXJM03 Y25751 WO9938881-A1 Cancer
    HUSAT94 Y25752 WO9938881-A1 Cancer
    HCUEN88 Y25753 WO9938881-A1 Immune/Hematopoietic
    HCE3F70 Y25754 WO9938881-A1 Cancer
    HCE5F43 Y25755 WO9938881-A1 Cancer
    HL2AC08 Y25756 WO9938881-A1 Cancer
    HCNSM70 Y25757 WO9938881-A1 Cancer
    HDPTQ73 Y25758 WO9938881-A1 Cancer
    HTODG13 Y25759 WO9938881-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HE8DR25 Y25760 WO9938881-A1 Excretory,
    Mixed Fetal,
    Neural/Sensory
    HSAAO65 Y25761 WO9938881-A1 Cancer
    HKGDE09 Y25762 WO9938881-A1 Cancer
    HMVBS69 Y25763 WO9938881-A1 Cardiovascular,
    Immune/Hematopoietic
    HSIDU42 Y25764 WO9938881-A1 Cancer
    HSKCT36 Y25765 WO9938881-A1 Cancer
    HSXBU59 Y25766 WO9938881-A1 Immune/Hematopoietic,
    Neural/Sensory
    HSSGG82 Y25767 WO9938881-A1 Cancer
    HE8CH92 Y25768 WO9938881-A1 Cancer
    HYBAR01 Y25769 WO9938881-A1 Musculoskeletal
    HTLEF73 Y25770 WO9938881-A1 Cancer
    HEOMW84 Y25771 WO9938881-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HKGAR66 Y25772 WO9938881-A1 Cancer
    HHPDX20 Y25773 WO9938881-A1 Neural/Sensory
    HSICV24 Y25774 WO9938881-A1 Cancer
    HCWBE20 Y25775 WO9938881-A1 Immune/Hematopoietic
    HSXBM30 Y25776 WO9938881-A1 Cancer
    HDPCY37 Y25778 WO9938881-A1 Cancer
    HOSFQ65 Y25791 WO9938881-A1 Cancer
    HSXBH24 Y25807 WO9938881-A1 Cancer
    HUSIG64 Y27567 WO9924836-A1 Cancer
    HATCI78 Y27568 WO9924836-A1 Endocrine
    HSIDR70 Y27569 WO9924836-A1 Digestive
    HFADD53 Y27570 WO9924836-A1 Excretory,
    Neural/Sensory
    HPMGT51 Y27571 WO9924836-A1 Immune/Hematopoietic,
    Reproductive
    HFVAB79 Y27572 WO9924836-A1 Cardiovascular,
    Digestive,
    Reproductive
    HDTBP51 Y27573 WO9924836-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HLHFR19 Y27574 WO9924836-A1 Neural/Sensory,
    Respiratory
    HMEET96 Y27575 WO9924836-A1 Cancer
    HTXCV12 Y27576 WO9924836-A1 Cancer
    HCEFB70 Y27577 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HDTAV25 Y27578 WO9924836-A1 Cancer
    HSATA21 Y27579 WO9924836-A1 Immune/Hematopoietic
    HKIXI03 Y27580 WO9924836-A1 Excretory
    HDTDC56 Y27581 WO9924836-A1 Cancer
    HLTBF35 Y27582 WO9924836-A1 Cancer
    HEPAB80 Y27583 WO9924836-A1 Reproductive
    HFOXB13 Y27584 WO9924836-A1 Musculoskeletal
    HTOAK16 Y27585 WO9924836-A1 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    HBXDC63 Y27586 WO9924836-A1 Neural/Sensory
    HASAU43 Y27587 WO9924836-A1 Immune/Hematopoietic
    HAGEA31 Y27588 WO9924836-A1 Cancer
    HTXHB33 Y27590 WO9924836-A1 Immune/Hematopoietic
    HMWFT65 Y27591 WO9924836-A1 Immune/Hematopoietic
    HNGAZ68 Y27592 WO9924836-A1 Cardiovascular,
    Immune/Hematopoietic
    HTWFH07 Y27593 WO9924836-A1 Immune/Hematopoietic
    HMQDF12 Y27594 WO9924836-A1 Cancer
    HFABH95 Y27595 WO9924836-A1 Digestive,
    Neural/Sensory,
    Reproductive
    HNGDD48 Y27596 WO9924836-A1 Immune/Hematopoietic
    HPMBY46 Y27597 WO9924836-A1 Cancer
    HRKPA09 Y27598 WO9924836-A1 Cancer
    HAGAQ26 Y27599 WO9924836-A1 Cancer
    HCWFL55 Y27600 WO9924836-A1 Immune/Hematopoietic
    HKAAE44 Y27601 WO9924836-A1 Cancer
    HNGEU90 Y27602 WO9924836-A1 Immune/Hematopoietic
    HCFCC07 Y27603 WO9924836-A1 Digestive,
    Immune/Hematopoietic
    HLWBI63 Y27604 WO9924836-A1 Cancer
    HDUAC77 Y27605 WO9924836-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HFOYV27 Y27606 WO9924836-A1 Cancer
    HGBHI35 Y27607 WO9924836-A1 Cancer
    HRDEU27 Y27608 WO9924836-A1 Musculoskeletal
    HNGJE50 Y27609 WO9924836-A1 Immune/Hematopoietic
    HNHDU48 Y27610 WO9924836-A1 Immune/Hematopoietic
    HFXJU68 Y27611 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HMMAH60 Y27612 WO9924836-A1 Immune/Hematopoietic
    HNGFR31 Y27613 WO9924836-A1 Immune/Hematopoietic
    HFPDB26 Y27614 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HFRAW86 Y27615 WO9924836-A1 Neural/Sensory
    HTEDX90 Y27616 WO9924836-A1 Reproductive
    HTXGG45 Y27617 WO9924836-A1 Immune/Hematopoietic
    HTXJI95 Y27618 WO9924836-A1 Immune/Hematopoietic,
    Reproductive
    HLYBD32 Y27619 WO9924836-A1 Immune/Hematopoietic
    HROAJ03 Y27621 WO9924836-A1 Cancer
    HTXAJ12 Y27622 WO9924836-A1 Immune/Hematopoietic
    HKAEL80 Y27623 WO9924836-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HNHFL04 Y27624 WO9924836-A1 Immune/Hematopoietic
    HPCAM01 Y27625 WO9924836-A1 Cancer
    HJACA79 Y27626 WO9924836-A1 Immune/Hematopoietic
    HMSFI26 Y27628 WO9924836-A1 Immune/Hematopoietic
    HMSJR08 Y27629 WO9924836-A1 Immune/Hematopoietic
    HMWIO93 Y27630 WO9924836-A1 Cancer
    HNGAK47 Y27631 WO9924836-A1 Immune/Hematopoietic
    HNGAL31 Y27632 WO9924836-A1 Immune/Hematopoietic
    HNGIZ06 Y27633 WO9924836-A1 Immune/Hematopoietic
    HNHBI75 Y27634 WO9924836-A1 Immune/Hematopoietic
    HOFNT24 Y27635 WO9924836-A1 Reproductive
    HSAXI95 Y27636 WO9924836-A1 Immune/Hematopoietic
    HCMTB45 Y27637 WO9924836-A1 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HE9CP41 Y27638 WO9924836-A1 Immune/Hematopoietic,
    Mixed Fetal
    HHENV10 Y27639 WO9924836-A1 Immune/Hematopoietic
    HSKDD72 Y27640 WO9924836-A1 Digestive,
    Musculoskeletal
    HAGDO20 Y27641 WO9924836-A1 Cancer
    HCFBH15 Y27642 WO9924836-A1 Immune/Hematopoietic
    HSYBX48 Y27643 WO9924836-A1 Cancer
    HATDQ62 Y27644 WO9924836-A1 Cancer
    HMEJE13 Y27645 WO9924836-A1 Cancer
    HNAAF65 Y27646 WO9924836-A1 Cancer
    HNFHY30 Y27647 WO9924836-A1 Immune/Hematopoietic
    HNFIR81 Y27648 WO9924836-A1 Cancer
    HNTBI57 Y27649 WO9924836-A1 Cancer
    HSAYR13 Y27650 WO9924836-A1 Immune/Hematopoietic
    HTOHV49 Y27651 WO9924836-A1 Immune/Hematopoietic
    HSFAG37 Y27652 WO9924836-A1 Cancer
    HTXBU52 Y27653 WO9924836-A1 Cancer
    HLHFP18 Y27654 WO9924836-A1 Cancer
    HFXBW09 Y27655 WO9924836-A1 Neural/Sensory
    HNGIO59 Y27656 WO9924836-A1 Immune/Hematopoietic
    HNGJF92 Y27657 WO9924836-A1 Immune/Hematopoietic
    HMEED18 Y27658 WO9924836-A1 Cancer
    HMIAM45 Y27659 WO9924836-A1 Neural/Sensory
    HSAVK10 Y27660 WO9924836-A1 Immune/Hematopoietic
    HSDHC81 Y27661 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HSLCT04 Y27662 WO9924836-A1 Mixed Fetal,
    Musculoskeletal
    HMDAB56 Y27663 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HUDBZ89 Y27664 WO9924836-A1 Cancer
    HLYCT47 Y27665 WO9924836-A1 Digestive,
    Immune/Hematopoietic
    HOSDJ25 Y27666 WO9924836-A1 Cancer
    HADAO89 Y27667 WO9924836-A1 Connective/Epithelial
    HMSGB14 Y27668 WO9924836-A1 Cancer
    HPMGD01 Y27669 WO9924836-A1 Cancer
    HNHFU32 Y27670 WO9924836-A1 Immune/Hematopoietic
    HMIAL40 Y27671 WO9924836-A1 Musculoskeletal,
    Neural/Sensory
    HAMFY69 Y27672 WO9924836-A1 Cancer
    HBMCT17 Y27673 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HEBFI91 Y27674 WO9924836-A1 Neural/Sensory
    HHEAH86 Y27675 WO9924836-A1 Cancer
    HTPCS72 Y27677 WO9924836-A1 Cancer
    HFFAL36 Y27678 WO9924836-A1 Neural/Sensory
    HFXBT12 Y27679 WO9924836-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNGJF70 Y27680 WO9924836-A1 Immune/Hematopoietic
    HATEE46 Y27681 WO9924836-A1 Cancer
    HJMBN89 Y27682 WO9924836-A1 Cancer
    HNHEK61 Y27683 WO9924836-A1 Immune/Hematopoietic
    HEQAO65 Y27684 WO9924836-A1 Cancer
    HFCDV54 Y27685 WO9924836-A1 Cancer
    HHEAD14 Y27686 WO9924836-A1 Cancer
    HGBHE57 Y27687 WO9924836-A1 Cancer
    HGLAF75 Y27688 WO9924836-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HHEMQ28 Y27689 WO9924836-A1 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HERAR44 Y27691 WO9924836-A1 Connective/Epithelial,
    Reproductive
    HYBAV65 Y28640 WO9940183-A1 Immune/Hematopoietic,
    Musculoskeletal
    HETBA38 Y28643 WO9940183-A1 Digestive,
    Mixed Fetal,
    Reproductive
    HCE1Q30 Y30701 WO9943693-A1 Immune/Hematopoietic,
    Neural/Sensory
    HAGBP70 Y30702 WO9943693-A1 Cancer
    HBCAY27 Y30703 WO9943693-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCACU58 Y30704 WO9943693-A1 Immune/Hematopoietic
    HCWLD74 Y30705 WO9943693-A1 Immune/Hematopoietic
    HDPFP29 Y30706 WO9943693-A1 Cancer
    HDPPH47 Y30707 WO9943693-A1 Cancer
    HFEAN33 Y30708 WO9943693-A1 Cancer
    HFEAT91 Y30709 WO9943693-A1 Connective/Epithelial,
    Reproductive
    HFPAO71 Y30710 WO9943693-A1 Cancer
    HLWAA17 Y30711 WO9943693-A1 Cancer
    HLYCQ18 Y30712 WO9943693-A1 Immune/Hematopoietic
    HOSFG70 Y30713 WO9943693-A1 Cancer
    HSSAJ29 Y30714 WO9943693-A1 Cancer
    HUSIF44 Y30715 WO9943693-A1 Cancer
    H6EDX46 Y30716 WO9943693-A1 Cancer
    HABAG37 Y30717 WO9943693-A1 Cancer
    HACBD91 Y30718 WO9943693-A1 Cancer
    HADEH21 Y30719 WO9943693-A1 Cancer
    HAGHD57 Y30720 WO9943693-A1 Cancer
    HAGHR69 Y30721 WO9943693-A1 Cancer
    HAHDB16 Y30722 WO9943693-A1 Cardiovascular
    HAHDR32 Y30723 WO9943693-A1 Cancer
    HAJAW93 Y30724 WO9943693-A1 Cancer
    HAJBR69 Y30725 WO9943693-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HAMGO32 Y30726 WO9943693-A1 Reproductive
    HATBR65 Y30727 WO9943693-A1 Cancer
    HBJLD29 Y30728 WO9943693-A1 Immune/Hematopoietic
    HBJNB13 Y30729 WO9943693-A1 Immune/Hematopoietic
    HCE2F54 Y30730 WO9943693-A1 Cancer
    HCE3C52 Y30731 WO9943693-A1 Cancer
    HCEEA88 Y30732 WO9943693-A1 Cancer
    HCEFE96 Y30733 WO9943693-A1 Cancer
    HCEIF12 Y30734 WO9943693-A1 Cancer
    HCEOR67 Y30735 WO9943693-A1 Neural/Sensory
    HCEVB76 Y30736 WO9943693-A1 Cancer
    HNTOA17 Y30737 WO9943693-A1 Cancer
    HDPOW86 Y30811 WO9940100-A1 Cancer
    HSYAG26 Y30812 WO9940100-A1 Cancer
    HLHCH40 Y30813 WO9940100-A1 Cancer
    HPLBM85 Y30814 WO9940100-A1 Cancer
    HLMBO76 Y30815 WO9940100-A1 Excretory,
    Immune/Hematopoietic,
    Reproductive
    HLQDR48 Y30816 WO9940100-A1 Digestive
    HOHBY12 Y30817 WO9940100-A1 Musculoskeletal
    HOSEK86 Y30818 WO9940100-A1 Cancer
    HAJBZ75 Y30819 WO9940100-A1 Cancer
    HAGCH75 Y30820 WO9940100-A1 Neural/Sensory
    HE8MH91 Y30821 WO9940100-A1 Cancer
    HISCJ55 Y30822 WO9940100-A1 Digestive
    HKISB57 Y30823 WO9940100-A1 Cancer
    HTEBJ71 Y30824 WO9940100-A1 Cancer
    HCWGA40 Y30825 WO9940100-A1 Cancer
    HFCEW05 Y30826 WO9940100-A1 Cardiovascular,
    Neural/Sensory
    HCEPF19 Y30827 WO9940100-A1 Cancer
    HTACZ01 Y30828 WO9940100-A1 Immune/Hematopoietic
    HUDAM89 Y30829 WO9940100-A1 Reproductive
    HSAXF60 Y30830 WO9940100-A1 Immune/Hematopoietic
    HTOGR42 Y30831 WO9940100-A1 Immune/Hematopoietic
    HMVBN46 Y30832 WO9940100-A1 Immune/Hematopoietic,
    Neural/Sensory
    HUVEB53 Y30833 WO9940100-A1 Cancer
    HSVBU91 Y30834 WO9940100-A1 Cancer
    HTXFL30 Y30835 WO9940100-A1 Cancer
    HAGAM64 Y30836 WO9940100-A1 Neural/Sensory
    HE2PH36 Y30837 WO9940100-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HGBDY06 Y30838 WO9940100-A1 Cancer
    HWBAO62 Y30839 WO9940100-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HBAFJ33 Y30840 WO9940100-A1 Cancer
    HFXDJ75 Y30841 WO9940100-A1 Neural/Sensory
    HFPCY04 Y30842 WO9940100-A1 Neural/Sensory
    HSNBG78 Y30843 WO9940100-A1 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HBQAB27 Y30844 WO9940100-A1 Endocrine,
    Neural/Sensory
    HTOJY21 Y30845 WO9940100-A1 Cancer
    HHTMM30 Y30846 WO9940100-A1 Cancer
    HLTAF58 Y30847 WO9940100-A1 Digestive,
    Immune/Hematopoietic
    HELFG13 Y30848 WO9940100-A1 Cancer
    HHFDM48 Y30849 WO9940100-A1 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HKABI84 Y30850 WO9940100-A1 Cancer
    HMVAX72 Y30851 WO9940100-A1 Cancer
    HODDN60 Y30852 WO9940100-A1 Cancer
    HPMEI44 Y30853 WO9940100-A1 Cancer
    HNGJP69 Y30854 WO9940100-A1 Immune/Hematopoietic
    HPWBA10 Y30855 WO9940100-A1 Immune/Hematopoietic,
    Reproductive
    HLHCH40 Y30856 WO9940100-A1 Cancer
    HTACZ01 Y30857 WO9940100-A1 Immune/Hematopoietic
    HTOGR42 Y30858 WO9940100-A1 Immune/Hematopoietic
    HTAEK53 Y31811 WO9947538-A1 Cancer
    HFCCQ50 Y36224 WO9931117-A1 Cancer
    HTLAI54 Y36225 WO9931117-A1 Reproductive
    HLWBF94 Y36227 WO9931117-A1 Endocrine,
    Neural/Sensory,
    Reproductive
    HFKFF78 Y36228 WO9931117-A1 Excretory
    HSYBG37 Y36229 WO9931117-A1 Cancer
    HTHCA77 Y36230 WO9931117-A1 Immune/Hematopoietic
    HNHEZ51 Y36231 WO9931117-A1 Immune/Hematopoietic
    HFIAX46 Y36232 WO9931117-A1 Cardiovascular,
    Musculoskeletal
    HFOXO72 Y36233 WO9931117-A1 Cancer
    HODDW40 Y36234 WO9931117-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HSAWG42 Y36235 WO9931117-A1 Immune/Hematopoietic
    HBMSK09 Y36236 WO9931117-A1 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HDPAU16 Y36237 WO9931117-A1 Cancer
    HFEBE12 Y36238 WO9931117-A1 Cancer
    HFLNB64 Y36239 WO9931117-A1 Cancer
    HSAWZ41 Y36240 WO9931117-A1 Immune/Hematopoietic
    HNFJF07 Y36241 WO9931117-A1 Immune/Hematopoietic,
    Neural/Sensory
    HNGJO57 Y36242 WO9931117-A1 Immune/Hematopoietic
    HE7TM22 Y36243 WO9931117-A1 Mixed Fetal
    HFRBR70 Y36244 WO9931117-A1 Cancer
    HTHBK35 Y36245 WO9931117-A1 Immune/Hematopoietic
    HWABA81 Y36246 WO9931117-A1 Immune/Hematopoietic
    HKGAA73 Y36247 WO9931117-A1 Cancer
    HKIYP40 Y36248 WO9931117-A1 Cancer
    HKMMW74 Y36249 WO9931117-A1 Excretory
    HLFBI27 Y36250 WO9931117-A1 Respiratory
    HLQCWS4 Y36251 WO9931117-A1 Digestive
    HBNAV22 Y36252 WO9931117-A1 Digestive,
    Reproductive
    HTEAM34 Y36253 WO9931117-A1 Reproductive
    HTHDK34 Y36254 WO9931117-A1 Digestive,
    Immune/Hematopoietic
    H6BSG32 Y36255 WO9931117-A1 Cardiovascular,
    Immune/Hematopoietic,
    Musculoskeletal
    HAECA01 Y36256 WO9931117-A1 Cancer
    HDTEL03 Y36257 WO9931117-A1 Cancer
    HFXDT43 Y36258 WO9931117-A1 Neural/Sensory
    HNGHQ09 Y36259 WO9931117-A1 Immune/Hematopoietic
    HHGDF16 Y36260 WO9931117-A1 Cancer
    HJBCG12 Y36261 WO9931117-A1 Cancer
    HOGAW62 Y36262 WO9931117-A1 Immune/Hematopoietic,
    Reproductive
    HSWBJ74 Y36263 WO9931117-A1 Cancer
    HGBHR26 Y36264 WO9931117-A1 Digestive
    HKDBF34 Y36265 WO9931117-A1 Cancer
    H6EAB28 Y36266 WO9931117-A1 Cancer
    HLWAO22 Y36267 WO9931117-A1 Cancer
    HAGFH53 Y36268 WO9931117-A1 Cancer
    HHENQ22 Y36269 WO9931117-A1 Immune/Hematopoietic
    HKMLK53 Y36270 WO9931117-A1 Excretory,
    Mixed Fetal
    HSKGQ58 Y36271 WO9931117-A1 Cancer
    HADXB45 Y36272 WO9931117-A1 Cancer
    HAIBZ39 Y36273 WO9931117-A1 Cancer
    HBXFP23 Y36274 WO9931117-A1 Cancer
    HEQBF32 Y36275 WO9931117-A1 Cancer
    HETHE81 Y36276 WO9931117-A1 Cancer
    HFPAC12 Y36277 WO9931117-A1 Cancer
    H6EFA77 Y36278 WO9931117-A1 Cancer
    HFXHD88 Y36279 WO9931117-A1 Neural/Sensory
    HFOXV65 Y36280 WO9931117-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HKADX21 Y36281 WO9931117-A1 Cancer
    HPZAB47 Y36282 WO9931117-A1 Cancer
    HAGFE79 Y36283 WO9931117-A1 Cancer
    HCE1X60 Y36284 WO9931117-A1 Neural/Sensory
    HFXKD36 Y36285 WO9931117-A1 Digestive,
    Musculoskeletal,
    Neural/Sensory
    HBMCU71 Y36286 WO9931117-A1 Immune/Hematopoietic
    HTEIV80 Y36287 WO9931117-A1 Reproductive
    HFIAP16 Y36288 WO9931117-A1 Musculoskeletal
    HODAV86 Y36289 WO9931117-A1 Reproductive
    HTEDF80 Y36290 WO9931117-A1 Reproductive
    HTODJ69 Y36291 WO9931117-A1 Immune/Hematopoietic
    HE6GR02 Y36292 WO9931117-A1 Immune/Hematopoietic,
    Mixed Fetal
    HAPNY86 Y36293 WO9931117-A1 Cancer
    HTLDR33 Y36294 WO9931117-A1 Immune/Hematopoietic,
    Reproductive
    HACBI61 Y36295 WO9931117-A1 Cancer
    HMEIK34 Y36296 WO9931117-A1 Cancer
    HKAAK02 Y36297 WO9931117-A1 Cancer
    HEPAA46 Y36298 WO9931117-A1 Reproductive
    HFPCX09 Y36299 WO9931117-A1 Mixed Fetal,
    Neural/Sensory
    HLWAA88 Y36300 WO9931117-A1 Cancer
    HOHBV89 Y36301 WO9931117-A1 Musculoskeletal,
    Reproductive
    HCEFL57 Y36302 WO9931117-A1 Cancer
    HMEKU83 Y36303 WO9931117-A1 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HOSBY40 Y36304 WO9931117-A1 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HKFBH93 Y36305 WO9931117-A1 Digestive,
    Reproductive
    HMTAD67 Y36306 WO9931117-A1 Cancer
    HTEBP77 Y36307 WO9931117-A1 Immune/Hematopoietic,
    Reproductive
    HE9CO69 Y36308 WO9931117-A1 Cancer
    HCACV51 Y36309 WO9931117-A1 Cancer
    HHPBI45 Y36310 WO9931117-A1 Cardiovascular,
    Neural/Sensory
    HLQDH79 Y36311 WO9931117-A1 Cancer
    HNGFJ67 Y36312 WO9931117-A1 Immune/Hematopoietic
    HEIAC52 Y36313 WO9931117-A1 Cancer
    HFXKL58 Y36314 WO9931117-A1 Cancer
    HMVAM60 Y36315 WO9931117-A1 Cancer
    HMVBR22 Y36316 WO9931117-A1 Cancer
    HPJCW04 Y36317 WO9931117-A1 Reproductive
    HSIDJ81 Y36318 WO9931117-A1 Digestive
    HSLFU05 Y36319 WO9931117-A1 Cancer
    HEQAK71 Y36320 WO9931117-A1 Cancer
    HOSEQ49 Y36321 WO9931117-A1 Cancer
    HRAAM50 Y36322 WO9931117-A1 Excretory,
    Immune/Hematopoietic,
    Mixed Fetal
    HSDFW45 Y36323 WO9931117-A1 Neural/Sensory
    HSLCQ82 Y36324 WO9931117-A1 Cancer
    HSSFT08 Y36325 WO9931117-A1 Musculoskeletal
    HTOIW31 Y36326 WO9931117-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTXKQ85 Y36327 WO9931117-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HUFBK08 Y36328 WO9931117-A1 Digestive,
    Musculoskeletal
    HBJEE48 Y36330 WO9931117-A1 Cancer
    HBXGH74 Y36331 WO9931117-A1 Neural/Sensory
    HISBM03 Y36332 WO9931117-A1 Cancer
    HETCH46 Y36333 WO9931117-A1 Cancer
    HFPCX09 Y36335 WO9931117-A1 Mixed Fetal,
    Neural/Sensory
    HLWAA88 Y36336 WO9931117-A1 Cancer
    HCEFL57 Y36337 WO9931117-A1 Cancer
    HETHE81 Y36650 WO9931117-A1 Cancer
    HTGAU75 Y38386 WO9935158-A1 Immune/Hematopoietic
    HTTDP47 Y38387 WO9935158-A1 Cancer
    HTXJQ11 Y38388 WO9935158-A1 Cancer
    HADCO45 Y38389 WO9935158-A1 Cancer
    HMIAL37 Y38390 WO9935158-A1 Cancer
    HNGDU40 Y38391 WO9935158-A1 Immune/Hematopoietic
    HFXBO84 Y38392 WO9935158-A1 Neural/Sensory
    HLLAX19 Y38393 WO9935158-A1 Cancer
    HPMAG94 Y38394 WO9935158-A1 Cancer
    HSVAK93 Y38395 WO9935158-A1 Cancer
    HMQBO88 Y38396 WO9935158-A1 Cancer
    HMQBU45 Y38397 WO9935158-A1 Immune/Hematopoietic
    HMWAJ53 Y38398 WO9935158-A1 Immune/Hematopoietic
    HCUGO12 Y38401 WO9935158-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HPFCX44 Y38402 WO9935158-A1 Cancer
    HCUBV79 Y38403 WO9935158-A1 Immune/Hematopoietic,
    Neural/Sensory
    HLQBV04 Y38404 WO9935158-A1 Cancer
    HMADW66 Y38405 WO9935158-A1 Cancer
    HLDBE54 Y38406 WO9935158-A1 Digestive,
    Reproductive
    HFTAB66 Y38407 WO9935158-A1 Digestive,
    Neural/Sensory
    HEOMQ63 Y38408 WO9935158-A1 Digestive,
    Immune/Hematopoietic
    HDPJM30 Y38409 WO9935158-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCFMG62 Y38410 WO9935158-A1 Cancer
    HJMAG88 Y38411 WO9935158-A1 Cancer
    HKAAH36 Y38412 WO9935158-A1 Connective/Epithelial,
    Reproductive
    HMADS41 Y38413 WO9935158-A1 Cancer
    HMEFT85 Y38414 WO9935158-A1 Cancer
    HMSBX80 Y38415 WO9935158-A1 Immune/Hematopoietic,
    Reproductive
    HNGCL23 Y38416 WO9935158-A1 Immune/Hematopoietic
    HPIBO15 Y38418 WO9935158-A1 Cancer
    HCYBG92 Y38419 WO9935158-A1 Cancer
    HMDAQ29 Y38420 WO9935158-A1 Neural/Sensory,
    Reproductive
    HSYBI49 Y38421 WO9935158-A1 Cancer
    HDTAB58 Y38422 WO9935158-A1 Cancer
    HFTAB66 Y38423 WO9935158-A1 Digestive,
    Neural/Sensory
    HDPBX23 Y38424 WO9935158-A1 Immune/Hematopoietic,
    Neural/Sensory
    HCFMG62 Y38425 WO9935158-A1 Cancer
    HKAAH36 Y38426 WO9935158-A1 Connective/Epithelial,
    Reproductive
    HKAAH36 Y38427 WO9935158-A1 Connective/Epithelial,
    Reproductive
    HMADS41 Y38428 WO9935158-A1 Cancer
    HNTBI26 Y38429 WO9935158-A1 Cancer
    HCYBI36 Y38430 WO9935158-A1 Cancer
    HTHBJ48 Y41161 US5981231-A Digestive,
    Immune/Hematopoietic
    HDQAC88 Y41164 US5981230-A Cancer
    HKGCR51 Y41308 WO9947540-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HPMDK28 Y41309 WO9947540-A1 Cancer
    HLDCD04 Y41310 WO9947540-A1 Cancer
    HLDON23 Y41311 WO9947540-A1 Cancer
    HLDRM43 Y41312 WO9947540-A1 Digestive,
    Reproductive
    HLQAM28 Y41313 WO9947540-A1 Digestive,
    Reproductive
    HLTDE74 Y41314 WO9947540-A1 Cancer
    HLTFA64 Y41315 WO9947540-A1 Cancer
    HMCFY13 Y41316 WO9947540-A1 Immune/Hematopoietic
    HMMBD35 Y41317 WO9947540-A1 Cancer
    HMQCY03 Y41318 WO9947540-A1 Digestive,
    Immune/Hematopoietic
    HMSBX84 Y41319 WO9947540-A1 Immune/Hematopoietic
    HMSKI86 Y41320 WO9947540-A1 Cancer
    HMVBS81 Y41321 WO9947540-A1 Cancer
    HMWEB02 Y41322 WO9947540-A1 Cancer
    HMZAD77 Y41323 WO9947540-A1 Cancer
    HNFIY77 Y41324 WO9947540-A1 Cancer
    HNHEK85 Y41325 WO9947540-A1 Immune/Hematopoietic,
    Mixed Fetal
    HNHEU93 Y41326 WO9947540-A1 Immune/Hematopoietic
    HODAH74 Y41327 WO9947540-A1 Connective/Epithelial,
    Reproductive,
    Respiratory
    HODCU34 Y41328 WO9947540-A1 Cancer
    HODCZ09 Y41329 WO9947540-A1 Reproductive
    HISCF16 Y41330 WO9947540-A1 Cancer
    HOGAG15 Y41331 WO9947540-A1 Cancer
    HPIBO48 Y41332 WO9947540-A1 Cancer
    HPMFP40 Y41333 WO9947540-A1 Reproductive
    HPRCU95 Y41334 WO9947540-A1 Musculoskeletal,
    Reproductive
    HPTTG19 Y41335 WO9947540-A1 Endocrine,
    Immune/Hematopoietic
    HRDDV47 Y41337 WO9947540-A1 Cancer
    HRDEN56 Y41338 WO9947540-A1 Musculoskeletal
    HSFAN12 Y41339 WO9947540-A1 Cardiovascular
    HSQCM10 Y41340 WO9947540-A1 Cancer
    HSVAT68 Y41341 WO9947540-A1 Excretory,
    Reproductive
    HSXEC75 Y41342 WO9947540-A1 Cancer
    HTDAI54 Y41343 WO9947540-A1 Cancer
    HTEIT45 Y41344 WO9947540-A1 Reproductive
    HTGBE48 Y41345 WO9947540-A1 Immune/Hematopoietic,
    Reproductive
    HTLEP53 Y41346 WO9947540-A1 Neural/Sensory,
    Reproductive
    HTTBI76 Y41347 WO9947540-A1 Cancer
    HTWKG71 Y41348 WO9947540-A1 Immune/Hematopoietic
    HTXDN32 Y41349 WO9947540-A1 Cancer
    HTSGX80 Y41350 WO9947540-A1 Cancer
    HTXEY51 Y41351 WO9947540-A1 Endocrine,
    Immune/Hematopoietic,
    Mixed Fetal
    HTXFH55 Y41352 WO9947540-A1 Cardiovascular,
    Immune/Hematopoietic
    HTXJW17 Y41353 WO9947540-A1 Digestive,
    Immune/Hematopoietic
    HUFCJ30 Y41354 WO9947540-A1 Cancer
    HWAAP70 Y41355 WO9947540-A1 Immune/Hematopoietic
    HWABW49 Y41356 WO9947540-A1 Immune/Hematopoietic
    HWBDP28 Y41357 WO9947540-A1 Cancer
    HWDAC39 Y41358 WO9947540-A1 Connective/Epithelial
    HWHGQ49 Y41359 WO9947540-A1 Cancer
    HJPAD75 Y41360 WO9947540-A1 Cancer
    HLDRP33 Y41361 WO9947540-A1 Digestive,
    Neural/Sensory
    HMSIE02 Y41362 WO9947540-A1 Cancer
    HNGFE55 Y41363 WO9947540-A1 Immune/Hematopoietic
    HRAAJ19 Y41365 WO9947540-A1 Cancer
    HSAWV96 Y41366 WO9947540-A1 Immune/Hematopoietic,
    Neural/Sensory
    HSBBT37 Y41367 WO9947540-A1 Cancer
    HSDZR57 Y41368 WO9947540-A1 Cancer
    HCECQ07 Y41369 WO9947540-A1 Cancer
    HWBCP79 Y41370 WO9947540-A1 Immune/Hematopoietic,
    Reproductive
    HYAAL70 Y41371 WO9947540-A1 Cancer
    HYAAY86 Y41372 WO9947540-A1 Immune/Hematopoietic
    HAPBS03 Y41373 WO9947540-A1 Cancer
    HBJLC01 Y41374 WO9947540-A1 Immune/Hematopoietic
    HBLKD56 Y41375 WO9947540-A1 Musculoskeletal
    HCENK38 Y41376 WO9947540-A1 Cancer
    HE6GA29 Y41379 WO9947540-A1 Mixed Fetal
    HETHO95 Y41381 WO9947540-A1 Digestive,
    Reproductive
    HFCFJ18 Y41382 WO9947540-A1 Cancer
    HFPBM30 Y41383 WO9947540-A1 Neural/Sensory
    HFXKT05 Y41384 WO9947540-A1 Cancer
    HKB1E57 Y41385 WO9947540-A1 Cancer
    HLWAD77 Y41386 WO9947540-A1 Cancer
    HLWAY54 Y41387 WO9947540-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HNGBU28 Y41388 WO9947540-A1 Immune/Hematopoietic
    HOUHH51 Y41389 WO9947540-A1 Cancer
    HRAAB15 Y41390 WO9947540-A1 Digestive,
    Excretory
    HSAVH65 Y41391 WO9947540-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HSDGN55 Y41392 WO9947540-A1 Cancer
    HSXAH81 Y41393 WO9947540-A1 Cancer
    HSXBX80 Y41394 WO9947540-A1 Cancer
    HTEHV08 Y41395 WO9947540-A1 Cancer
    HUFAK67 Y41396 WO9947540-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HUSXS50 Y41397 WO9947540-A1 Cancer
    HAPON17 Y41398 WO9947540-A1 Cancer
    HATAC53 Y41399 WO9947540-A1 Cancer
    HAMFK58 Y41400 WO9947540-A1 Cancer
    HLYCH68 Y41401 WO9947540-A1 Cancer
    HCUHK65 Y41402 WO9947540-A1 Cancer
    HLDCD04 Y41403 WO9947540-A1 Cancer
    HOUHH51 Y41404 WO9947540-A1 Cancer
    HSLCQ82 Y41571 WO9947540-A1 Cancer
    HCGMD59 Y45257 WO9946289-A1 Cancer
    HCNSD76 Y45258 WO9946289-A1 Digestive
    HCNSD93 Y45259 WO9946289-A1 Digestive
    HCWBE22 Y45260 WO9946289-A1 Immune/Hematopoietic,
    Neural/Sensory
    HFEAN33 Y45261 WO9946289-A1 Cancer
    HCWUM50 Y45262 WO9946289-A1 Cancer
    HDHIA94 Y45263 WO9946289-A1 Excretory,
    Neural/Sensory
    HDPAE76 Y45264 WO9946289-A1 Cancer
    HDPIO54 Y45265 WO9946289-A1 Immune/Hematopoietic,
    Reproductive
    HDPNC61 Y45266 WO9946289-A1 Cancer
    HDPND46 Y45267 WO9946289-A1 Immune/Hematopoietic
    HDPSU13 Y45268 WO9946289-A1 Immune/Hematopoietic
    HDTGC73 Y45269 WO9946289-A1 Cancer
    HE2PD49 Y45270 WO9946289-A1 Cancer
    HEEAJ02 Y45271 WO9946289-A1 Cancer
    HELHD64 Y45272 WO9946289-A1 Cancer
    HEPAD91 Y45273 WO9946289-A1 Digestive,
    Reproductive
    HEQBH65 Y45274 WO9946289-A1 Immune/Hematopoietic,
    Reproductive
    HETCO02 Y45275 WO9946289-A1 Cancer
    HFAUO78 Y45276 WO9946289-A1 Cancer
    HFKEE48 Y45277 WO9946289-A1 Cancer
    HFKFG02 Y45278 WO9946289-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    H2CBN14 Y45279 WO9946289-A1 Cancer
    HHFFJ48 Y45280 WO9946289-A1 Cardiovascular,
    Immune/Hematopoietic
    HILCF66 Y45281 WO9946289-A1 Cancer
    HKABN45 Y45282 WO9946289-A1 Cancer
    HKDBK22 Y45284 WO9946289-A1 Excretory
    HKGAZ06 Y45286 WO9946289-A1 Immune/Hematopoietic
    HKGCK61 Y45287 WO9946289-A1 Cancer
    HFEAN33 Y45288 WO9946289-A1 Cancer
    HDHIA94 Y45289 WO9946289-A1 Excretory,
    Neural/Sensory
    HDPJO39 Y52479 WO9940184-A1 Cancer
    HNTCF82 Y58185 US6004780-A Cardiovascular,
    Connective/Epithelial,
    Reproductive
    HETAB62 Y59285 WO200004183-A1 Cancer
    HSYAE36 Y59286 WO200004183-A1 Cancer
    HKAPI15 Y68800 WO200005371-A1 Connective/Epithelial
    HUJCT9C Y72090 WO200068247-A2 Cancer
    HMGBM65 Y72091 WO200068247-A2 Cancer
    HATEE38 Y72092 WO200068247-A2 Cancer
    HCHAK72 Y72093 WO200068247-A2 Cancer
    HHFBJ67 Y72094 WO200068247-A2 Cardiovascular,
    Neural/Sensory
    HTTJK5C Y72095 WO200068247-A2 Cancer
    HWLGJ11 Y72096 WO200068247-A2 Digestive
    HTLEG15 Y72097 WO200068247-A2 Cancer
    HAGAS16 Y72098 WO200068247-A2 Neural/Sensory
    HATEE38 Y72108 WO200068247-A2 Cancer
    HKABZ65 Y76124 WO9958660-A1 Connective/Epithelial
    HNGIC80 Y76125 WO9958660-A1 Immune/Hematopoietic
    HDPUG50 Y76126 WO9958660-A1 Cancer
    HAEAB66 Y76127 WO9958660-A1 Cancer
    HHEPF59 Y76128 WO9958660-A1 Cancer
    HE9BK23 Y76129 WO9958660-A1 Digestive,
    Mixed Fetal
    HCYBI36 Y76130 WO9958660-A1 Cancer
    HSSDX51 Y76131 WO9958660-A1 Cancer
    HSDAJ46 Y76132 WO9958660-A1 Cancer
    HRACG45 Y76133 WO9958660-A1 Cancer
    HAPPW30 Y76134 WO9958660-A1 Cancer
    HE2ES51 Y76135 WO9958660-A1 Cancer
    HTXDW56 Y76136 WO9958660-A1 Cancer
    HDPKI93 Y76138 WO9958660-A1 Cancer
    HDLAC10 Y76139 WO9958660-A1 Cancer
    HDPOH06 Y76140 WO9958660-A1 Cancer
    HCE4G61 Y76141 WO9958660-A1 Cancer
    HCWUI13 Y76142 WO9958660-A1 Immune/Hematopoietic
    HDPSP01 Y76143 WO9958660-A1 Cancer
    HHPEN62 Y76144 WO9958660-A1 Cancer
    HUKBT29 Y76145 WO9958660-A1 Cancer
    HARAP48 Y76146 WO9958660-A1 Cancer
    HBIMB51 Y76147 WO9958660-A1 Connective/Epithelial,
    Reproductive
    HE8DX88 Y76148 WO9958660-A1 Mixed Fetal
    HNGHT03 Y76149 WO9958660-A1 Immune/Hematopoietic
    HWABU17 Y76150 WO9958660-A1 Cancer
    HCE5F84 Y76151 WO9958660-A1 Cancer
    HBXCD55 Y76152 WO9958660-A1 Cancer
    HOVCB25 Y76153 WO9958660-A1 Reproductive
    HSYAV66 Y76154 WO9958660-A1 Digestive,
    Immune/Hematopoietic
    HFPCT29 Y76155 WO9958660-A1 Neural/Sensory
    HAWAT25 Y76156 WO9958660-A1 Cancer
    HNHFR04 Y76157 WO9958660-A1 Immune/Hematopoietic
    HOSFT61 Y76158 WO9958660-A1 Cancer
    HBJIO81 Y76159 WO9958660-A1 Immune/Hematopoietic
    HADCL55 Y76160 WO9958660-A1 Cancer
    HAGGJ80 Y76161 WO9958660-A1 Cancer
    HAIBO81 Y76162 WO9958660-A1 Neural/Sensory
    HBBBC37 Y76163 WO9958660-A1 Cancer
    HBJMX85 Y76164 WO9958660-A1 Cancer
    HCEES66 Y76165 WO9958660-A1 Digestive,
    Neural/Sensory
    HCEMP62 Y76166 WO9958660-A1 Cancer
    HE2FB90 Y76167 WO9958660-A1 Cancer
    HE9DS56 Y76168 WO9958660-A1 Cancer
    HTOHJ89 Y76169 WO9958660-A1 Immune/Hematopoietic
    HASCE69 Y76171 WO9958660-A1 Cancer
    HHTLH52 Y76172 WO9958660-A1 Neural/Sensory,
    Reproductive
    HOUCT90 Y76174 WO9958660-A1 Connective/Epithelial
    HCFLR78 Y76175 WO9958660-A1 Cancer
    HTOHT18 Y76176 WO9958660-A1 Cancer
    HKPMB11 Y76177 WO9958660-A1 Digestive,
    Excretory,
    Musculoskeletal
    HNFHS38 Y76178 WO9958660-A1 Cancer
    HAIBU10 Y76179 WO9958660-A1 Cancer
    HAPOK30 Y76180 WO9958660-A1 Cancer
    HCWUA22 Y76182 WO9958660-A1 Immune/Hematopoietic
    HDSAG91 Y76183 WO9958660-A1 Immune/Hematopoietic
    HNEDJ35 Y76184 WO9958660-A1 Immune/Hematopoietic,
    Reproductive
    HTHBH29 Y76185 WO9958660-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    H7TBA62 Y76186 WO9958660-A1 Cancer
    HNGIO50 Y76187 WO9958660-A1 Immune/Hematopoietic
    HMIAW81 Y76188 WO9958660-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HMMCJ60 Y76189 WO9958660-A1 Immune/Hematopoietic,
    Musculoskeletal
    HDPIO09 Y76190 WO9958660-A1 Cancer
    HHFHH34 Y76191 WO9958660-A1 Cardiovascular
    HISCL83 Y76192 WO9958660-A1 Digestive
    HTOAI70 Y76193 WO9958660-A1 Immune/Hematopoietic
    HSDER95 Y76194 WO9958660-A1 Digestive,
    Neural/Sensory
    HNECL25 Y76195 WO9958660-A1 Immune/Hematopoietic
    HNFGZ45 Y76196 WO9958660-A1 Cardiovascular,
    Digestive,
    Immune/Hematopoietic
    HHGCU49 Y76197 WO9958660-A1 Cancer
    HETDT81 Y76199 WO9958660-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HHLBA14 Y76200 WO9958660-A1 Cancer
    HLTBU43 Y76201 WO9958660-A1 Immune/Hematopoietic
    HNTSJ84 Y76202 WO9958660-A1 Cancer
    HOHCG16 Y76203 WO9958660-A1 Digestive,
    Musculoskeletal
    HTHCB31 Y76204 WO9958660-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HUKAM16 Y76205 WO9958660-A1 Cancer
    HLDOJ66 Y76206 WO9958660-A1 Digestive
    HTXKF10 Y76207 WO9958660-A1 Immune/Hematopoietic
    HPMAI22 Y76208 WO9958660-A1 Reproductive
    HL2AG57 Y76209 WO9958660-A1 Cancer
    HUSAM59 Y76210 WO9958660-A1 Cancer
    HNGGR26 Y76211 WO9958660-A1 Immune/Hematopoietic
    HTLCX30 Y76212 WO9958660-A1 Reproductive
    HCEBC87 Y76213 WO9958660-A1 Cancer
    HATCB92 Y76214 WO9958660-A1 Endocrine
    HLHAL68 Y76216 WO9958660-A1 Respiratory
    HEOMR73 Y76217 WO9958660-A1 Immune/Hematopoietic
    HETIB83 Y76218 WO9958660-A1 Cancer
    HJPDD28 Y76219 WO9958660-A1 Cancer
    HBAMB15 Y76220 WO9958660-A1 Cardiovascular,
    Excretory,
    Reproductive
    HBAFQ33 Y76221 WO9958660-A1 Cancer
    HTOAI70 Y76222 WO9958660-A1 Immune/Hematopoietic
    HJPDD28 Y76223 WO9958660-A1 Cancer
    HRACG45 Y76266 WO9958660-A1 Cancer
    HBXCD55 Y76303 WO9958660-A1 Cancer
    HOSFT61 Y76325 WO9958660-A1 Cancer
    HWBBP10 Y86215 WO9966041-A1 Immune/Hematopoietic,
    Neural/Sensory
    HWBDO80 Y86216 WO9966041-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Reproductive
    HWHGU54 Y86217 WO9966041-A1 Connective/Epithelial
    HYACI76 Y86218 WO9966041-A1 Cancer
    HBHMA23 Y86219 WO9966041-A1 Cancer
    HCE3G20 Y86220 WO9966041-A1 Cancer
    HCEJP80 Y86221 WO9966041-A1 Cardiovascular,
    Neural/Sensory
    HCUDD24 Y86222 WO9966041-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPTD15 Y86223 WO9966041-A1 Immune/Hematopoietic
    HDPWU34 Y86224 WO9966041-A1 Cancer
    HEOOV79 Y86225 WO9966041-A1 Cancer
    HFKET93 Y86226 WO9966041-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HFTDL56 Y86227 WO9966041-A1 Cancer
    HFXJX44 Y86228 WO9966041-A1 Cancer
    HKACU58 Y86229 WO9966041-A1 Cancer
    HKFBC53 Y86230 WO9966041-A1 Cancer
    HLTHR66 Y86231 WO9966041-A1 Cancer
    HLYBA69 Y86232 WO9966041-A1 Cancer
    HNTMX29 Y86233 WO9966041-A1 Cancer
    HNTNC20 Y86234 WO9966041-A1 Cancer
    HNTNI01 Y86235 WO9966041-A1 Cancer
    HPIBW65 Y86236 WO9966041-A1 Cancer
    HSMBE69 Y86237 WO9966041-A1 Cancer
    HT4FW61 Y86238 WO9966041-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HYABK95 Y86239 WO9966041-A1 Cancer
    HYACE88 Y86240 WO9966041-A1 Cancer
    HOABR60 Y86241 WO9966041-A1 Cancer
    HAPOM45 Y86243 WO9966041-A1 Cardiovascular,
    Digestive
    HCEJQ69 Y86244 WO9966041-A1 Cancer
    HAGFI62 Y86245 WO9966041-A1 Cancer
    HAGGS43 Y86246 WO9966041-A1 Neural/Sensory
    HBJHP03 Y86247 WO9966041-A1 Immune/Hematopoietic,
    Reproductive
    HCHPF68 Y86248 WO9966041-A1 Reproductive
    HDPJF37 Y86249 WO9966041-A1 Cancer
    HSDEZ20 Y86250 WO9966041-A1 Neural/Sensory
    HTEKU58 Y86251 WO9966041-A1 Cancer
    HLTBL58 Y86252 WO9966041-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HPWDJ42 Y86253 WO9966041-A1 Digestive,
    Reproductive
    HRACD15 Y86254 WO9966041-A1 Cancer
    HSIAC80 Y86255 WO9966041-A1 Cancer
    HAGFD18 Y86256 WO9966041-A1 Cancer
    HAJAP76 Y86257 WO9966041-A1 Cancer
    HDTGC86 Y86258 WO9966041-A1 Digestive,
    Immune/Heinatopoietic,
    Reproductive
    HAGDI35 Y86259 WO9966041-A1 Cancer
    HELHN47 Y86260 WO9966041-A1 Cancer
    HPRBC80 Y86261 WO9966041-A1 Cancer
    HAQAR23 Y86262 WO9966041-A1 Cancer
    HAIFL18 Y86263 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HJPAY76 Y86264 WO9966041-A1 Cancer
    HUSXE77 Y86265 WO9966041-A1 Cancer
    HUFEF62 Y86266 WO9966041-A1 Digestive
    HTWJK32 Y86267 WO9966041-A1 Cancer
    HTWDF76 Y86268 WO9966041-A1 Immune/Hematopoietic
    HTPBN68 Y86269 WO9966041-A1 Digestive
    HTOIY21 Y86270 WO9966041-A1 Immune/Hematopoietic
    HTLDD53 Y86271 WO9966041-A1 Connective/Epithelial,
    Digestive,
    Reproductive
    HTLFG05 Y86272 WO9966041-A1 Cancer
    HDPXR23 Y86273 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HSIAC45 Y86274 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HSRGW16 Y86275 WO9966041-A1 Cancer
    HSSJC35 Y86276 WO9966041-A1 Cancer
    HTEAX23 Y86277 WO9966041-A1 Reproductive
    HTGCH22 Y86278 WO9966041-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HTJMA95 Y86279 WO9966041-A1 Cancer
    HHEAA08 Y86280 WO9966041-A1 Immune/Hematopoietic
    HBQAA49 Y86281 WO9966041-A1 Neural/Sensory
    HDPBI32 Y86282 WO9966041-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HBIBF16 Y86283 WO9966041-A1 Neural/Sensory
    HBCAY05 Y86284 WO9966041-A1 Cancer
    HCUCK44 Y86285 WO9966041-A1 Cancer
    HCE2W56 Y86286 WO9966041-A1 Cancer
    HCWAG01 Y86287 WO9966041-A1 Immune/Hematopoietic
    HDRMI82 Y86289 WO9966041-A1 Cancer
    HEPCU48 Y86290 WO9966041-A1 Cancer
    HDPRK33 Y86291 WO9966041-A1 Immune/Hematopoietic,
    Mixed Fetal
    HKGAX42 Y86292 WO9966041-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HLMAZ95 Y86293 WO9966041-A1 Cancer
    HLMFC07 Y86294 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HL2AG87 Y86295 WO9966041-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HKGCO27 Y86296 WO9966041-A1 Cancer
    HLDCE79 Y86297 WO9966041-A1 Digestive
    HERAD40 Y86298 WO9966041-A1 Connective/Epithelial
    HFOXB55 Y86299 WO9966041-A1 Cancer
    HFVGZ42 Y86300 WO9966041-A1 Cancer
    HNHAF39 Y86301 WO9966041-A1 Immune/Hematopoietic
    HNTSW57 Y86302 WO9966041-A1 Cancer
    HOGCK20 Y86303 WO9966041-A1 Cancer
    HLYES38 Y86305 WO9966041-A1 Immune/Hematopoietic,
    Reproductive
    HMECK83 Y86306 WO9966041-A1 Cardiovascular
    HMQAG66 Y86308 WO9966041-A1 Immune/Hematopoietic
    HWBBP10 Y86309 WO9966041-A1 Immune/Hematopoietic,
    Neural/Sensory
    HAPAK52 Y86310 WO9966041-A1 Cancer
    HDPWU34 Y86311 WO9966041-A1 Cancer
    HKACU58 Y86312 WO9966041-A1 Cancer
    HLDBQ19 Y86314 WO9966041-A1 Cancer
    HNTMX29 Y86315 WO9966041-A1 Cancer
    HOABR60 Y86316 WO9966041-A1 Cancer
    HPWDJ42 Y86317 WO9966041-A1 Digestive,
    Reproductive
    HPWDJ42 Y86318 WO9966041-A1 Digestive,
    Reproductive
    HRACD15 Y86319 WO9966041-A1 Cancer
    HPRBC80 Y86320 WO9966041-A1 Cancer
    HUFEF62 Y86321 WO9966041-A1 Digestive
    HTLFG05 Y86322 WO9966041-A1 Cancer
    HDPXR23 Y86323 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HSRGW16 Y86324 WO9966041-A1 Cancer
    HDPBI32 Y86327 WO9966041-A1 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDRMI82 Y86328 WO9966041-A1 Cancer
    HKGCO27 Y86330 WO9966041-A1 Cancer
    HNTSW57 Y86332 WO9966041-A1 Cancer
    HOGCK20 Y86333 WO9966041-A1 Cancer
    HNTMX29 Y86388 WO9966041-A1 Cancer
    HPRBC80 Y86463 WO9966041-A1 Cancer
    HTLFG05 Y86488 WO9966041-A1 Cancer
    HDPXR23 Y86489 WO9966041-A1 Digestive,
    Immune/Hematopoietic
    HSRGW16 Y86496 WO9966041-A1 Cancer
    HDRMI82 Y86532 WO9966041-A1 Cancer
    HNTSW57 Y86571 WO9966041-A1 Cancer
    HISCN02 Y87064 WO200004140-A1 Digestive
    HHGDM70 Y87065 WO200004140-A1 Immune/Hematopoietic
    HHPGO40 Y87066 WO200004140-A1 Cancer
    HAMGG68 Y87067 WO200004140-A1 Cancer
    HAPOM49 Y87068 WO200004140-A1 Cancer
    HBGBA69 Y87069 WO200004140-A1 Cancer
    HBJFJ26 Y87070 WO200004140-A1 Cancer
    HCEDH38 Y87071 WO200004140-A1 Mixed Fetal,
    Neural/Sensory
    HDPOJ08 Y87072 WO200004140-A1 Cancer
    HDPRX82 Y87073 WO200004140-A1 Cancer
    HELGK31 Y87074 WO200004140-A1 Cancer
    HFPCX64 Y87075 WO200004140-A1 Mixed Fetal,
    Neural/Sensory
    HFXDO60 Y87076 WO200004140-A1 Neural/Sensory
    HAUAI83 Y87077 WO200004140-A1 Reproductive
    HKGAH42 Y87078 WO200004140-A1 Neural/Sensory
    HMIAP86 Y87079 WO200004140-A1 Cancer
    HMUAP70 Y87080 WO200004140-A1 Cancer
    HRACJ35 Y87081 WO200004140-A1 Cancer
    HTWDE26 Y87082 WO200004140-A1 Cancer
    HBGBB44 Y87083 WO200004140-A1 Cancer
    HBAFA02 Y87084 WO200004140-A1 Cancer
    H2CBT75 Y87085 WO200004140-A1 Cancer
    HAGDQ42 Y87086 WO200004140-A1 Cancer
    HBMCJ42 Y87087 WO200004140-A1 Immune/Hematopoietic,
    Reproductive
    HLCDA16 Y87089 WO200004140-A1 Cancer
    HELHL48 Y87090 WO200004140-A1 Cancer
    HISAQ04 Y87091 WO200004140-A1 Digestive,
    Neural/Sensory,
    Reproductive
    HJACB89 Y87092 WO200004140-A1 Cancer
    HTECC05 Y87093 WO200004140-A1 Cancer
    HBJLF01 Y87094 WO200004140-A1 Cancer
    HBXGP60 Y87095 WO200004140-A1 Cancer
    HCE5B20 Y87096 WO200004140-A1 Mixed Fetal,
    Neural/Sensory
    HCMSQ56 Y87097 WO200004140-A1 Cancer
    HCNAH57 Y87098 WO200004140-A1 Digestive
    HCUEP91 Y87099 WO200004140-A1 Immune/Hematopoietic
    HDPCJ91 Y87100 WO200004140-A1 Cancer
    HDPGK25 Y87101 WO200004140-A1 Cancer
    HE2DY70 Y87102 WO200004140-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Musculoskeletal
    HE2NV57 Y87103 WO200004140-A1 Cancer
    HETBR16 Y87104 WO200004140-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HFXDG13 Y87105 WO200004140-A1 Cancer
    HFXKY27 Y87106 WO200004140-A1 Neural/Sensory
    HHPEC09 Y87107 WO200004140-A1 Cancer
    HISAD54 Y87108 WO200004140-A1 Cancer
    HJBCY35 Y87109 WO200004140-A1 Cancer
    HKAEA19 Y87110 WO200004140-A1 Cancer
    HKGDL36 Y87111 WO200004140-A1 Cancer
    HLDBS43 Y87112 WO200004140-A1 Cancer
    HLWAD92 Y87113 WO200004140-A1 Cancer
    HLYBI15 Y87114 WO200004140-A1 Immune/Hematopoietic
    HMEJE05 Y87115 WO200004140-A1 Cancer
    HNGIX55 Y87116 WO200004140-A1 Immune/Hematopoietic
    HNHEX30 Y87117 WO200004140-A1 Immune/Hematopoietic
    HPJBI33 Y87118 WO200004140-A1 Reproductive
    HRABA80 Y87119 WO200004140-A1 Excretory
    HRACD80 Y87120 WO200004140-A1 Excretory,
    Reproductive
    HSLCX03 Y87121 WO200004140-A1 Cancer
    HT5GJ57 Y87122 WO200004140-A1 Cancer
    HTACS42 Y87123 WO200004140-A1 Cancer
    HTEKE40 Y87124 WO200004140-A1 Cancer
    HTOBX69 Y87125 WO200004140-A1 Cancer
    HUVEO77 Y87126 WO200004140-A1 Reproductive
    H2CBG48 Y87127 WO200004140-A1 Cancer
    H2CBU83 Y87128 WO200004140-A1 Cancer
    HAPNY94 Y87129 WO200004140-A1 Cancer
    HBJHZ58 Y87130 WO200004140-A1 Immune/Hematopoietic,
    Reproductive
    HCE2B33 Y87131 WO200004140-A1 Cancer
    HDPBQ02 Y87132 WO200004140-A1 Immune/Hematopoietic
    HFIYI70 Y87133 WO200004140-A1 Cancer
    HDPOZ56 Y87134 WO200004140-A1 Cancer
    HAPOM49 Y87136 WO200004140-A1 Cancer
    HBJFJ26 Y87137 WO200004140-A1 Cancer
    HCNUA40 Y87138 WO200004140-A1 Cancer
    HCEBW71 Y87139 WO200004140-A1 Mixed Fetal,
    Neural/Sensory
    HCEBW71 Y87140 WO200004140-A1 Mixed Fetal,
    Neural/Sensory
    HAUAI83 Y87141 WO200004140-A1 Reproductive
    HFLQB16 Y87143 WO200004140-A1 Cancer
    HAGFY16 Y87144 WO200004140-A1 Cancer
    HFLQB16 Y87146 WO200004140-A1 Cancer
    HAGFY16 Y87147 WO200004140-A1 Cancer
    HMHBN40 Y87149 WO200004140-A1 Cancer
    HDPBQ71 Y87150 WO200004140-A1 Cancer
    HSKCT36 Y87151 WO200004140-A1 Cancer
    HRACD80 Y87152 WO200004140-A1 Excretory,
    Reproductive
    HSLCX03 Y87153 WO200004140-A1 Cancer
    H2CBU83 Y87154 WO200004140-A1 Cancer
    HFLQB16 Y87180 WO200004140-A1 Cancer
    HAGFY16 Y87181 WO200004140-A1 Cancer
    HFLQB16 Y87183 WO200004140-A1 Cancer
    HAGFY16 Y87184 WO200004140-A1 Cancer
    HMHBN40 Y87187 WO200004140-A1 Cancer
    HDPBQ71 Y87188 WO200004140-A1 Cancer
    HSKCT36 Y87192 WO200004140-A1 Cancer
    HRACD80 Y87205 WO200004140-A1 Excretory,
    Reproductive
    HSLCX03 Y87208 WO200004140-A1 Cancer
    H2CBU83 Y87215 WO200004140-A1 Cancer
    HISCH47 Y87784 US6054289-A Cancer
    HADCD24 Y87789 US6054289-A Cancer
    HDTEA84 Y90357 WO200052028-A1 Cancer
    HKGCN17 Y91346 WO200011014-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HETAD68 Y91347 WO200011014-A1 Cancer
    HPIAT78 Y91348 WO200011014-A1 Cancer
    HHFHG52 Y91349 WO200011014-A1 Cancer
    HDTAB58 Y91350 WO200011014-A1 Cancer
    HEOMQ62 Y91351 WO200011014-A1 Cancer
    HWLJQ88 Y91352 WO200011014-A1 Digestive
    HMICP03 Y91353 WO200011014-A1 Cancer
    HAJAB01 Y91354 WO200011014-A1 Cancer
    HE2AT09 Y91355 WO200011014-A1 Cancer
    HSDJA15 Y91356 WO200011014-A1 Cancer
    HAMGW29 Y91357 WO200011014-A1 Cancer
    HAPSR85 Y91358 WO200011014-A1 Digestive,
    Endocrine
    HTOHD42 Y91359 WO200011014-A1 Immune/Hematopoietic
    HWLIH65 Y91360 WO200011014-A1 Cancer
    HTOJA73 Y91361 WO200011014-A1 Immune/Hematopoietic
    HPMGJ45 Y91362 WO200011014-A1 Cancer
    HFVIC62 Y91363 WO200011014-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HHENW77 Y91364 WO200011014-A1 Cancer
    HMSIV91 Y91365 WO200011014-A1 Cancer
    HMSKC04 Y91366 WO200011014-A1 Immune/Hematopoietic
    HSAZG33 Y91367 WO200011014-A1 Immune/Hematopoietic
    HTEBC92 Y91368 WO200011014-A1 Cancer
    HTXEL29 Y91369 WO200011014-A1 Immune/Hematopoietic
    HDPAW44 Y91370 WO200011014-A1 Cancer
    HMACS20 Y91371 WO200011014-A1 Cancer
    HAJAY88 Y91372 WO200011014-A1 Immune/Hematopoietic
    HBOEG69 Y91373 WO200011014-A1 Cancer
    HWLEQ37 Y91374 WO200011014-A1 Cancer
    HE9CS37 Y91375 WO200011014-A1 Cancer
    HNGEI34 Y91376 WO200011014-A1 Immune/Hematopoietic
    HTOAT76 Y91377 WO200011014-A1 Excretory,
    Immune/Hematopoietic
    HDPVH60 Y91378 WO200011014-A1 Cancer
    HLYCR65 Y91379 WO200011014-A1 Cancer
    HARAY91 Y91380 WO200011014-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HCHNT03 Y91381 WO200011014-A1 Digestive,
    Reproductive
    HCUBW95 Y91382 WO200011014-A1 Immune/Hematopoietic,
    Neural/Sensory
    HDPLV95 Y91383 WO200011014-A1 Immune/Hematopoietic,
    Reproductive
    HEMGB12 Y91384 WO200011014-A1 Cancer
    HHENP27 Y91385 WO200011014-A1 Cancer
    HSPBF70 Y91386 WO200011014-A1 Cancer
    HTXKB57 Y91387 WO200011014-A1 Cancer
    HUKAA55 Y91388 WO200011014-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HFXGT58 Y91389 WO200011014-A1 Neural/Sensory
    HUSFF19 Y91391 WO200011014-A1 Cancer
    HUVDJ43 Y91393 WO200011014-A1 Cardiovascular,
    Reproductive
    HTLCU49 Y91394 WO200011014-A1 Cancer
    HDTAB58 Y91395 WO200011014-A1 Cancer
    HWLJQ88 Y91396 WO200011014-A1 Digestive
    HUVDJ43 Y91399 WO200011014-A1 Cardiovascular,
    Reproductive
    HUVDJ43 Y91400 WO200011014-A1 Cardiovascular,
    Reproductive
    HTLCU49 Y91401 WO200011014-A1 Cancer
    HUVDJ43 Y91446 WO200011014-A1 Cardiovascular,
    Reproductive
    HUVDJ43 Y91448 WO200011014-A1 Cardiovascular,
    Reproductive
    HFKIB49 Y91449 WO200011014-A1 Cancer
    HDPTK41 Y91451 WO200006698-A1 Cancer
    HFXGT26 Y91452 WO200006698-A1 Cancer
    HLTGX30 Y91453 WO200006698-A1 Immune/Hematopoietic
    HLTHG37 Y91454 WO200006698-A1 Cancer
    HNTMZ90 Y91455 WO200006698-A1 Digestive,
    Reproductive
    HPIBX03 Y91456 WO200006698-A1 Cancer
    H6EDY30 Y91457 WO200006698-A1 Cancer
    HAMGR28 Y91458 WO200006698-A1 Cancer
    HAPNZ94 Y91459 WO200006698-A1 Cancer
    HATCP77 Y91460 WO200006698-A1 Cancer
    HDABR72 Y91461 WO200006698-A1 Cancer
    HDPKB18 Y91462 WO200006698-A1 Immune/Hematopoietic
    HEQCC55 Y91463 WO200006698-A1 Cancer
    HETDE26 Y91464 WO200006698-A1 Cancer
    HOEDH84 Y91465 WO200006698-A1 Cancer
    HPIBT55 Y91466 WO200006698-A1 Cancer
    HSLCS05 Y91467 WO200006698-A1 Cancer
    HDPDD03 Y91468 WO200006698-A1 Cancer
    HDTDQ23 Y91470 WO200006698-A1 Cancer
    HE2PY40 Y91471 WO200006698-A1 Mixed Fetal
    HEONM66 Y91472 WO200006698-A1 Immune/Hematopoietic
    HKAEG43 Y91473 WO200006698-A1 Cancer
    HLHDP65 Y91474 WO200006698-A1 Cancer
    HLMDO03 Y91475 WO200006698-A1 Cancer
    HMAGK93 Y91476 WO200006698-A1 Cancer
    HMEAL02 Y91477 WO200006698-A1 Cardiovascular
    HMKCH52 Y91478 WO200006698-A1 Neural/Sensory
    HCEFB69 Y91479 WO200006698-A1 Cancer
    HKADM92 Y91480 WO200006698-A1 Cancer
    HSPMG77 Y91481 WO200006698-A1 Digestive
    HSQAC69 Y91482 WO200006698-A1 Cancer
    HSTBJ86 Y91483 WO200006698-A1 Connective/Epithelial
    HUVDJ43 Y91485 WO200006698-A1 Cardiovascular,
    Reproductive
    HADCP14 Y91486 WO200006698-A1 Connective/Epithelial
    HBXCF95 Y91487 WO200006698-A1 Cancer
    HEQBU15 Y91488 WO200006698-A1 Cancer
    HL1BD22 Y91489 WO200006698-A1 Cancer
    HOEEU24 Y91490 WO200006698-A1 Cancer
    HTTBR96 Y91491 WO200006698-A1 Reproductive
    HWHQS55 Y91492 WO200006698-A1 Cancer
    HCEEK50 Y91493 WO200006698-A1 Cancer
    HCWBU94 Y91494 WO200006698-A1 Immune/Hematopoietic
    HE2NR62 Y91495 WO200006698-A1 Cancer
    HHSGH19 Y91496 WO200006698-A1 Neural/Sensory
    HDPGT01 Y91497 WO200006698-A1 Cancer
    HOHCA35 Y91499 WO200006698-A1 Cancer
    HPMGP24 Y91500 WO200006698-A1 Mixed Fetal,
    Reproductive
    HSDIE16 Y91501 WO200006698-A1 Neural/Sensory
    HSOBK48 Y91502 WO200006698-A1 Digestive
    HTADH39 Y91503 WO200006698-A1 Cancer
    HUSGT36 Y91504 WO200006698-A1 Cardiovascular
    HVAAE95 Y91505 WO200006698-A1 Digestive
    HHEAH25 Y91506 WO200006698-A1 Cancer
    HBJIY92 Y91507 WO200006698-A1 Cancer
    HCLCW50 Y91508 WO200006698-A1 Respiratory
    HDRMF68 Y91509 WO200006698-A1 Digestive,
    Respiratory
    HOUGG12 Y91510 WO200006698-A1 Cancer
    HEEAQ11 Y91511 WO200006698-A1 Reproductive
    HEEAZ65 Y91512 WO200006698-A1 Musculoskeletal,
    Reproductive
    HEGAN94 Y91513 WO200006698-A1 Reproductive
    HFXBL33 Y91514 WO200006698-A1 Cancer
    HLIBD68 Y91515 WO200006698-A1 Cancer
    HLTCO33 Y91516 WO200006698-A1 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HLYAC95 Y91517 WO200006698-A1 Immune/Hematopoietic
    HNHKS18 Y91519 WO200006698-A1 Immune/Hematopoietic
    HSLJW78 Y91520 WO200006698-A1 Musculoskeletal
    HHFHD01 Y91521 WO200006698-A1 Cardiovascular,
    Musculoskeletal,
    Neural/Sensory
    HLWAE11 Y91522 WO200006698-A1 Cancer
    HCYBN55 Y91523 WO200006698-A1 Cancer
    HEONX38 Y91524 WO200006698-A1 Cancer
    HLDQU79 Y91525 WO200006698-A1 Cancer
    HSYBK21 Y91526 WO200006698-A1 Cancer
    HTHDS25 Y91528 WO200006698-A1 Endocrine,
    Immune/Hematopoietic
    HFIHO70 Y91529 WO200006698-A1 Cancer
    HPMEI86 Y91530 WO200006698-A1 Cancer
    HSOBV29 Y91531 WO200006698-A1 Cancer
    HWABY10 Y91532 WO200006698-A1 Cancer
    HACCI17 Y91533 WO200006698-A1 Cancer
    HAPQT22 Y91534 WO200006698-A1 Immune/Hematopoietic
    HDPBO81 Y91535 WO200006698-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPGI49 Y91536 WO200006698-A1 Cancer
    HDTBV77 Y91537 WO200006698-A1 Cancer
    HFIUE82 Y91538 WO200006698-A1 Cancer
    HHEND31 Y91539 WO200006698-A1 Cancer
    HKMND01 Y91540 WO200006698-A1 Excretory
    HLDBI84 Y91541 WO200006698-A1 Cancer
    HLTEK17 Y91542 WO200006698-A1 Cancer
    HEBEJ18 Y91543 WO200006698-A1 Cancer
    HMEAI48 Y91544 WO200006698-A1 Cardiovascular
    HNHGN91 Y91545 WO200006698-A1 Digestive,
    Endocrine
    Immune/Hematopoietic
    HODAE92 Y91546 WO200006698-A1 Cancer
    HODDF13 Y91547 WO200006698-A1 Reproductive
    HATEF60 Y91548 WO200006698-A1 Cancer
    HLTHG37 Y91549 WO200006698-A1 Cancer
    HAMGR28 Y91550 WO200006698-A1 Cancer
    HDPKB18 Y91551 WO200006698-A1 Immune/Hematopoietic
    HEQCC55 Y91552 WO200006698-A1 Cancer
    HEONM66 Y91554 WO200006698-A1 Immune/Hematopoietic
    HKAEG43 Y91555 WO200006698-A1 Cancer
    HLHDP65 Y91556 WO200006698-A1 Cancer
    HOEEU24 Y91557 WO200006698-A1 Cancer
    HHEAH25 Y91558 WO200006698-A1 Cancer
    HCYBN55 Y91559 WO200006698-A1 Cancer
    HEONX38 Y91560 WO200006698-A1 Cancer
    HFIHO70 Y91561 WO200006698-A1 Cancer
    HACCI17 Y91562 WO200006698-A1 Cancer
    HDPBO81 Y91563 WO200006698-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HAMGR28 Y91599 WO200006698-A1 Cancer
    HDPKB18 Y91603 WO200006698-A1 Immune/Hematopoietic
    HEQCC55 Y91604 WO200006698-A1 Cancer
    HLHDP65 Y91631 WO200006698-A1 Cancer
    HOEEU24 Y91643 WO200006698-A1 Cancer
    HHEAH25 Y91647 WO200006698-A1 Cancer
    HHEAH25 Y91648 WO200006698-A1 Cancer
    HLIBD68 Y91656 WO200006698-A1 Cancer
    HCYBN55 Y91670 WO200006698-A1 Cancer
    HEONX38 Y91672 WO200006698-A1 Cancer
    HFIHO70 Y91679 WO200006698-A1 Cancer
    HMKBA64 Y91681 WO200006698-A1 Cancer
    HACCI17 Y91683 WO200006698-A1 Cancer
    HMKEA94 Y93650 WO200036105-A1 Cancer
    HE9SF68 Y93973 WO200042189-A1 Cancer
    HTSGS30 Y93974 WO200042189-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HDQAC88 Y95534 WO200040726-A1 Cancer
    HDPMM34 Y96280 WO200028035-A1 Cancer
    HKABZ65 Y96962 WO200039327-A1 Connective/Epithelial
    HWHGB15 Y96963 WO200039327-A1 Connective/Epithelial
    HCDDP40 Y96964 WO200039327-A1 Immune/Hematopoietic,
    Musculoskeletal
    HETBE01 B03767 US6066724-A Cancer
    HETGI70 B03768 US6066724-A Cancer
    HETDK42 B03769 US6066724-A Cancer
    HTEMZ33 B07705 WO200043493-A2 Cancer
    HE8AW20 B07941 US6103871-A Cancer
    HNEDU15 B08659 WO200050597-A2 Cancer
    HLTBT71 B08661 WO200050597-A2 Cancer
    HBICD95 B08785 WO200050620-A2 Cancer
    HE9CC44 B08786 US6110893-A Cancer
    HPRCC57 B10293 US6077692-A Cancer
    HPRCC57 B10304 US6077692-A Cancer
    HPRCC57 B10310 US6077692-A Cancer
    HPRCC57 B10311 US6077692-A Cancer
    HPRCC57 B10312 US6077692-A Cancer
    HPRCC57 B10313 US6077692-A Cancer
    HPRCC57 B10316 US6077692-A Cancer
    HPRCC57 B10320 US6077692-A Cancer
    HILBX90 B11125 US6133422-A Cancer
    HCQAS17 B12900 US6080722-A Digestive,
    Mixed Fetal,
    Reproductive
    HBMSE33 B15366 WO200042165-A2 Cancer
    HE2BG16 B15413 US6090575-A Cancer
    HT4CC72 B18618 WO200053223-A1 Immune/Hematopoietic
    HAPOR40 B18750 WO200055204-A1 Cancer
    HTSGS30 B18755 WO200055204-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HFGAM58 B18803 WO200053210-A1 Cancer
    HSDFB55 B19550 WO200053793-A1 Cancer
    HUVEO91 B19863 WO200066608-A1 Cancer
    HCDDP40 B25583 WO200029435-A1 Immune/Hematopoietic,
    Musculoskeletal
    HMELK96 B26981 WO200056862-A1 Cancer
    HMELK96 B26987 WO200056862-A1 Cancer
    HTTBN61 B26990 WO200056862-A1 Cancer
    HCUDS60 B26991 WO200056862-A1 Cancer
    HLYBX88 B26992 WO200056862-A1 Cancer
    HILBI36 B28524 US6130051-A Cancer
    HLYBX88 B29790 WO200066156-A1 Cancer
    HCEMP60 B29923 US6130061-A Cancer
    HE8AE45 B33821 WO200056753-A1 Cancer
    HE2OA95 B33822 WO200056753-A1 Cancer
    HJACE54 B35705 WO200063221-A2 Cancer
    HTTBN61 B36265 WO200064465-A1 Cancer
    HPRCB54 B36696 WO200071150-A1 Cancer
    HSDME38 B39392 WO200057903-A2 Cancer
    HSDME38 B39393 WO200057903-A2 Cancer
    HPDDY64 B43604 WO200055350-A1 Cancer
    HPABA51 B44685 WO200058339-A2 Cancer
    HPMSM24 B45376 WO200061628-A1 Cancer
    HOUCQ17 B50002 WO200071577-A1 Cancer
    HODAH63 B50272 WO200071567-A2 Neural/Sensory,
    Reproductive
    HODAH63 B50282 WO200071567-A2 Neural/Sensory,
    Reproductive
    HODAH63 B50283 WO200071567-A2 Neural/Sensory,
    Reproductive
    HCEGY95 B50289 WO200071582-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HE9CC44 B50293 WO200071715-A1 Cancer
    HAGAT55 B50294 WO200071715-A1 Cancer
    HAGAT55 B50704 WO200071152-A1 Cancer
    HKABO35 B50892 WO200073321-A1 Cancer
    HETJY78 B51152 US6153739-A Cancer
    HPRCC57 B58248 WO200055180-A2 Cancer
    HHFCU19 B58276 WO200055180-A2 Cancer
    HNFAG09 B58319 WO200055180-A2 Cancer
    HBZSD43 B58925 WO200055173-A1 Cancer
    HHFHJ57 B58970 WO200055173-A1 Cancer
    HPRCC57 B60201 WO200072872-A1 Cancer
    HPRCC57 B60204 WO200072872-A1 Cancer
    HPRCC57 B60206 WO200072872-A1 Cancer
    HPRCC57 B60207 WO200072872-A1 Cancer
    HPRCC57 B60208 WO200072872-A1 Cancer
    HPRCC57 B60209 WO200072872-A1 Cancer
    HPRCC57 B60210 WO200072872-A1 Cancer
    HPRCC57 B60212 WO200072872-A1 Cancer
    HPRCC57 B60214 WO200072872-A1 Cancer
    HRGBQ38 B64643 WO200077197-A1 Cancer
    HRGBQ38 B64644 WO200077197-A1 Cancer
    HRGBQ38 B64645 WO200077197-A1 Cancer
    HRGBQ38 B64646 WO200077197-A1 Cancer
    HLTBT71 B64873 WO200077256-A1 Cancer
    HTEIX55 B64953 WO200076530-A1 Cancer
    HPABA51 R75085 ZA9403789-A Cancer
    HAPAT57 R76127 WO9517092-A Cancer
    HWFBD68 R76128 WO9517092-A Cancer
    HMPSA79 R77649 WO9532282-A1 Cancer
    HGBAB73 R79008 WO9520678-A1 Cancer
    HLTAW73 R79009 WO9520678-A1 Cancer
    HFCAW19 R80095 WO9527781-A1 Cancer
    HHFBT80 R80575 WO9524474-A1 Cancer
    HFKCU96 R81309 WO9519985-A1 Cancer
    HSRAW34 R81461 WO9605226-A1 Cancer
    HOSBD47 R82686 WO9524473-A1 Cancer
    HOSBH74 R82720 WO9524182-A1 Cancer
    HE8AE45 R82987 WO9524466-A1 Cancer
    HLFBE10 R84522 WO9524411-A1 Cancer
    HAGAT55 R85650 WO9524414-A1 Cancer
    HIBEC52 R87954 WO9530428-A1 Cancer
    HTEAH87 R88390 WO9531539-A1 Cancer
    HFBEH64 R88405 WO9531538-A1 Cancer
    HAFAK86 R88419 WO9535372-A1 Cancer
    HASSB35 R88452 WO9600242-A1 Cancer
    HPAAA47 R88481 WO9601270-A1 Cancer
    HJPAH22 R90703 WO9600297-A1 Cancer
    HIBCL76 R90764 WO9603415-A1 Cancer
    HIBEJ89 R90765 WO9603415-A1 Neural/Sensory
    HLFBE49 R90919 WO9601896-A Cancer
    HIBCL22 R90989 WO9605225-A1 Cancer
    HSSAW84 R91929 WO9612791-A1 Cancer
    HSNME29 R92220 WO9604928-A1 Cancer
    HSNME29 R92753 WO9605221-A Cancer
    HGBAN46 R93086 WO9605856-A1 Cancer
    HE9DR66 R93087 WO9605856-A1 Cancer
    HTPAN40 R93118 WO9606862-A Cancer
    HILBI36 R93156 WO9608557-A1 Cancer
    HJBAQ29 R94350 WO9609311-A1 Cancer
    HASAC73 R94601 WO9611259-A1 Cancer
    HPLAP22 R94602 WO9611259-A1 Cancer
    HT2SA16 R95634 WO9614394-A1 Cancer
    HLHAC42 R95692 WO9615806-A1 Cancer
    HE2CA82 R95830 WO9613603-A1 Cancer
    HTOBA30 R95831 WO9613603-A1 Cancer
    HFSBE16 R97222 WO9616087-A1 Cancer
    HHFCU19 R97565 WO9621736-A1 Cancer
    HE8AW20 R97739 WO9615222-A1 Cancer
    HSBBC75 R97978 WO9615147-A1 Cancer
    HLFBG09 R98224 WO9612501-A1 Cancer
    HHPEC49 R98261 WO9611946-A1 Cancer
    HFGAM58 R98265 WO9618725-A1 Cancer
    HUVCT01 R98994 WO9617931-A1 Cancer
    HFSAG79 R99329 WO9624668-A1 Cancer
    HATBG78 R99353 WO9627009-A1 Endocrine
    HUVEO91 R99453 WO9614328-A1 Cancer
    HPRCC57 W00176 WO9625422-A1 Cancer
    HCAAA02 W00482 WO9621724-A1 Cancer
    HETAN67 W01097 WO9629401-A1 Cancer
    HSSNB01 W01098 WO9629401-A1 Cancer
    HETJY78 W01619 WO9635778-A1 Cancer
    HPRAJ70 W01730 WO9639435-A1 Cancer
    HNFAG09 W02151 WO9625432-A1 Cancer
    HFSBC65 W02613 WO9618730-A1 Cancer
    HE2BG16 W04247 WO9630406-A1 Cancer
    HTECE68 W05295 WO9630524-A1 Cancer
    HRGBQ38 W05313 WO9623410-A1 Cancer
    HFCCE09 W05314 WO9623410-A1 Cancer
    HGOCA18 W05315 WO9623410-A1 Cancer
    HT1SB52 W05809 WO9634095-A1 Cancer
    HFGAN72 W06124 WO9634877-A1 Cancer
    HHFBT80 W06539 WO9639431-A1 Cancer
    HCNAY46 W06545 WO9639419-A1 Cancer
    HCQDM23 W06546 WO9639419-A1 Digestive,
    Reproductive
    HCNUB65 W06548 WO9639419-A1 Cancer
    HCNSE58 W06550 WO9639419-A1 Cancer
    HCNBB33 W06551 WO9639419-A1 Cancer
    HKLSA58 W06552 WO9639419-A1 Cancer
    HCNSD13 W06553 WO9639419-A1 Cancer
    HLQBI14 W06575 WO9639520-A1 Cancer
    HAECD08 W07202 WO9634891-A1 Cancer
    HWFBD68 W07203 WO9634891-A1 Cancer
    HAPAT57 W07204 WO9634891-A1 Cancer
    HDGNR10 W07602 WO9639437-A1 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMSDB49 W07604 WO9639521-A1 Immune/Hematopoietic,
    Reproductive
    HFSAG79 W07605 WO9639522-A1 Cancer
    HTOEX74 W07606 WO9639522-A1 Cancer
    HMWCF06 W07611 WO9639421-A1 Cancer
    HIBEB69 W07617 WO9639438-A1 Cancer
    HGBER32 W07618 WO9639434-A1 Digestive
    HETGQ23 W07619 WO9639436-A1 Cancer
    HE2OA95 W07663 WO9636709-A1 Cancer
    HCEGY95 W08079 WO9639506-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HIBEF51 W08101 WO9639441-A1 Neural/Sensory
    HTNAD29 W08141 WO9639442-A1 Cancer
    HE9CC44 W08142 WO9639507-A1 Cancer
    HDGRC02 W09110 WO9639440-A1 Cancer
    HPTTT24 W09111 WO9639420-A1 Digestive,
    Endocrine
    HUVDR03 W09404 WO9639485-A1 Cancer
    HPBCB95 W09405 WO9639158-A1 Cancer
    HE9NG77 W09408 WO9639486-A1 Cancer
    HATCK89 W09432 WO9639509-A1 Cancer
    HTOEX74 W10574 WO9624668-A1 Cancer
    HOSBD47 W11478 WO9639515-A1 Cancer
    HCQAS17 W12691 WO9639541-A1 Digestive,
    Mixed Fetal,
    Reproductive
    HCACU62 W12692 WO9639424-A1 Cancer
    HAQBM60 W12693 WO9639418-A1 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HLTDG74 W12695 WO9639433-A1 Immune/Hematopoietic
    HODAH63 W12696 WO9639508-A1 Neural/Sensory,
    Reproductive
    HSSAW84 W17043 US5618717-A Cancer
    HHSAN40 W17838 WO9717358-A1 Cancer
    HTPBS22 W19632 WO9722623-A1 Cancer
    HSATU68 W19780 WO9725340-A1 Cancer
    HFCBS02 W22408 WO9711970-A1 Cancer
    HGBAN46 W22669 WO9731098-A1 Cancer
    HE9DR66 W22670 WO9731098-A1 Cancer
    HE9DR66 W22671 WO9731098-A1 Cancer
    HE9DR66 W22672 WO9731098-A1 Cancer
    HE9DR66 W22673 WO9731098-A1 Cancer
    HE9DR66 W22674 WO9731098-A1 Cancer
    HE9DR66 W22675 WO9731098-A1 Cancer
    HPMSM24 W22732 WO9724929-A1 Cancer
    HPABA51 W22882 US5635616-A Cancer
    HESAJ20 W23663 WO9729189-A1 Cancer
    HALTA54 W24137 WO9723640-A1 Cancer
    HCEMP60 W24847 WO9718224-A1 Cancer
    HFCCE09 W25112 US5650313-A Cancer
    HGOCA18 W25113 US5650313-A Cancer
    HRGBQ38 W25114 US5650313-A Cancer
    HTPAN40 W26464 US5654172-A Cancer
    HTECD31 W27087 WO9725349-A1 Cancer
    HLHDC84 W27118 WO9725338-A1 Cancer
    HTPAN08 W27134 WO9733899-A1 Cancer
    HFKET35 W27152 WO9734013-A1 Cancer
    HTTER36 W27224 WO9735870-A1 Cardiovascular,
    Connective/Epithelial,
    Reproductive
    HTOBH93 W27561 WO9727747-A1 Cancer
    HE6BK61 W29291 WO9735010-A1 Cancer
    HE6BK61 W29292 WO9735010-A1 Cancer
    HTOJK64 W30193 WO9735976-A2 Cancer
    HLMBA70 W30891 WO9735028-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HMEIP65 W31512 WO9732993-A1 Cancer
    HTTBN61 W31517 WO9733904-A1 Cancer
    HFLQA68 W31527 WO9737022-A1 Cancer
    HE8AW20 W31692 US5695980-A Cancer
    HTXEI33 W31759 WO9733898-A1 Cancer
    HCUDE60 W31902 WO9737021-A1 Cancer
    HCABA58 W32110 WO9738012-A1 Cancer
    HMEAN51 W32112 WO9734998-A1 Cancer
    HT4CC72 W32255 WO9734911-A1 Immune/Hematopoietic
    HCUDE60 W32323 WO9736915-A1 Cancer
    HSAAU35 W33603 WO9747742-A1 Connective/Epithelial,
    Musculoskeletal,
    Reproductive
    HETBE01 W35802 WO9734997-A1 Cancer
    HETGI70 W35803 WO9734997-A1 Cancer
    HETDK42 W35804 WO9734997-A1 Cancer
    HBJEL88 W35904 WO9738003-A1 Cancer
    HSHCL68 W36449 WO9735027-A1 Cancer
    HLTBT71 W37002 WO9733902-A1 Cancer
    HPDDO12 W37003 WO9733902-A1 Cancer
    HHPEC49 W37799 US5750370-A Cancer
    HCQAJ72 W37844 WO9807749-A1 Cancer
    HMECG71 W37845 WO9807749-A1 Cancer
    HSIEH63 W37846 WO9807749-A1 Digestive
    HBICD95 W37847 WO9807880-A1 Cancer
    HMQBM23 W37935 WO9808870-A1 Cancer
    HOEBG39 W37946 WO9821236-A1 Cancer
    HOSBH74 W39216 EP812916-A2 Cancer
    HOSBH74 W39264 EP812916-A2 Cancer
    HOSBH74 W39265 EP812916-A2 Cancer
    HOSBH74 W39266 EP812916-A2 Cancer
    HOSBH74 W39267 EP812916-A2 Cancer
    HOSBH74 W39268 EP812916-A2 Cancer
    HODAH63 W40077 US5728546-A Neural/Sensory,
    Reproductive
    HFBEH64 W41362 US5723311-A Cancer
    HSAAU35 W41502 EP812913-A2 Connective/Epithelial,
    Musculoskeletal,
    Reproductive
    HSAAU35 W41520 WO9747741-A1 Connective/Epithelial,
    Musculoskeletal,
    Reproductive
    HOSBH74 W41645 WO9747642-A1 Cancer
    HTSEX82 W41938 WO9748807-A1 Digestive,
    Immune/Hematopoietic
    HIBCL76 W42995 US5710019-A Cancer
    HIBEJ89 W42996 US5710019-A Neural/Sensory
    HILBI36 W46518 US5716806-A Cancer
    HCNAY46 W46876 US5733748-A Cancer
    HCQDM23 W46877 US5733748-A Digestive,
    Reproductive
    HCNUB65 W46879 US5733748-A Cancer
    HCNSE58 W46882 US5733748-A Cancer
    HCNBB33 W46883 US5733748-A Cancer
    HKLSA58 W46884 US5733748-A Cancer
    HCNSD13 W46885 US5733748-A Cancer
    HEMEM90 W48334 WO9807881-A1 Cancer
    HE9BK24 W48335 WO9807754-A1 Cancer
    HPASD50 W48391 WO9807735-A1 Cancer
    HETAN67 W48762 WO9812204-A1 Cancer
    HHFHJ57 W49032 WO9825957-A2 Cancer
    HGBER32 W49807 US5776729-A Digestive
    HATCK89 W49826 US5773252-A Cancer
    HCEPR64 W51244 WO9821242-A1 Cancer
    HPRCC57 W52581 WO9806844-A1 Cancer
    HPRCC57 W52582 WO9806844-A1 Cancer
    HPRCC57 W52583 WO9806844-A1 Cancer
    HPRCC57 W52584 WO9806844-A1 Cancer
    HPRCC57 W52585 WO9806844-A1 Cancer
    HPRCC57 W52586 WO9806844-A1 Cancer
    HPRCC57 W52587 WO9806844-A1 Cancer
    HPRCC57 W52588 WO9806844-A1 Cancer
    HPRCC57 W52590 WO9806844-A1 Cancer
    HPRCC57 W52591 WO9806844-A1 Cancer
    HPRCC57 W52592 WO9806844-A1 Cancer
    HPRCC57 W52593 WO9806844-A1 Cancer
    HPRCC57 W52594 WO9806844-A1 Cancer
    HPRCC57 W52595 WO9806844-A1 Cancer
    HPRCC57 W52596 WO9806844-A1 Cancer
    HPRCC57 W52597 WO9806844-A1 Cancer
    HPRCC57 W52598 WO9806844-A1 Cancer
    HPRCC57 W52599 WO9806844-A1 Cancer
    HDQMB53 W52842 WO9807862-A2 Cancer
    HWFBD68 W52843 WO9807862-A2 Cancer
    HPMFW51 W53121 WO9806859-A1 Cancer
    HPMFW51 W53122 WO9806859-A1 Cancer
    HPRCC57 W53787 WO9806844-A1 Cancer
    HPRCC57 W53792 WO9806844-A1 Cancer
    HPRCC57 W53793 WO9806844-A1 Cancer
    HMEEJ22 W53897 WO9808969-A1 Cancer
    HE9CC44 W54036 US5763214-A Cancer
    HMQCD14 W55884 WO9806733-A1 Cancer
    HUVDE75 W56249 WO9806839-A1 Cancer
    HCNBB33 W56503 WO9815624-A1 Cancer
    HTPBR22 W56504 WO9815624-A1 Cancer
    HETAS87 W56505 WO9815624-A1 Cancer
    HETAS87 W56506 WO9815624-A1 Cancer
    HPRAJ70 W56641 US5756309-A Cancer
    HAICL46 W57044 WO9811138-A1 Cancer
    HAECD08 W57688 WO9814582-A1 Cancer
    HAECD08 W57691 WO9814582-A1 Cancer
    HAECD08 W57692 WO9814582-A1 Cancer
    HAECD08 W57693 WO9814582-A1 Cancer
    HAECD08 W57694 WO9814582-A1 Cancer
    HAECD08 W57695 WO9814582-A1 Cancer
    HWFBD68 W57697 WO9814582-A1 Cancer
    HAPAT57 W57698 WO9814582-A1 Cancer
    HAECD08 W57699 WO9814582-A1 Cancer
    HAECD08 W57701 WO9814582-A1 Cancer
    HMSDB49 W57881 WO9824908-A1 Immune/Hematopoietic,
    Reproductive
    HNEDU15 W58391 WO9818921-A1 Cancer
    HE9NG77 W58704 US5780263-A Cancer
    HFSAG79 W58900 WO9814477-A1 Cancer
    HTOEX74 W58901 WO9814477-A1 Cancer
    HTOEX74 W58902 WO9814477-A1 Cancer
    HTOEX74 W58903 WO9814477-A1 Cancer
    HTOEX74 W58904 WO9814477-A1 Cancer
    HTOEX74 W58905 WO9814477-A1 Cancer
    HTOEX74 W58906 WO9814477-A1 Cancer
    HTOEX74 W58907 WO9814477-A1 Cancer
    HTOEX74 W58908 WO9814477-A1 Cancer
    HTOEX74 W58909 WO9814477-A1 Cancer
    HTOEX74 W58910 WO9814477-A1 Cancer
    HTOEX74 W58911 WO9814477-A1 Cancer
    HTOEX74 W58912 WO9814477-A1 Cancer
    HTOEX74 W58913 WO9814477-A1 Cancer
    HTOEX74 W58914 WO9814477-A1 Cancer
    HTOEX74 W58915 WO9814477-A1 Cancer
    HTOEX74 W58916 WO9814477-A1 Cancer
    HTOEX74 W58917 WO9814477-A1 Cancer
    HTOEX74 W58918 WO9814477-A1 Cancer
    HTOEX74 W58919 WO9814477-A1 Cancer
    HTOEX74 W58920 WO9814477-A1 Cancer
    HTOEX74 W58921 WO9814477-A1 Cancer
    HTOEX74 W58922 WO9814477-A1 Cancer
    HTOEX74 W58923 WO9814477-A1 Cancer
    HTOEX74 W58924 WO9814477-A1 Cancer
    HTOEX74 W58925 WO9814477-A1 Cancer
    HTOEX74 W58926 WO9814477-A1 Cancer
    HTOEX74 W58927 WO9814477-A1 Cancer
    HTOEX74 W58928 WO9814477-A1 Cancer
    HTOEX74 W58929 WO9814477-A1 Cancer
    HTOEX74 W58930 WO9814477-A1 Cancer
    HTOEX74 W58931 WO9814477-A1 Cancer
    HTOEX74 W58932 WO9814477-A1 Cancer
    HTOEX74 W58933 WO9814477-A1 Cancer
    HTOEX74 W58934 WO9814477-A1 Cancer
    HTOEX74 W58935 WO9814477-A1 Cancer
    HTOEX74 W58936 WO9814477-A1 Cancer
    HTOEX74 W58937 WO9814477-A1 Cancer
    HTOEX74 W58938 WO9814477-A1 Cancer
    HTOEX74 W58939 WO9814477-A1 Cancer
    HTOEX74 W58940 WO9814477-A1 Cancer
    HTOEX74 W58941 WO9814477-A1 Cancer
    HFSAG79 W58942 WO9814477-A1 Cancer
    HFSAG79 W58943 WO9814477-A1 Cancer
    HFSAG79 W58944 WO9814477-A1 Cancer
    HFSAG79 W58945 WO9814477-A1 Cancer
    HFSAG79 W58946 WO9814477-A1 Cancer
    HFSAG79 W58947 WO9814477-A1 Cancer
    HFSAG79 W58948 WO9814477-A1 Cancer
    HFSAG79 W58949 WO9814477-A1 Cancer
    HFSAG79 W58950 WO9814477-A1 Cancer
    HFSAG79 W58951 WO9814477-A1 Cancer
    HFSAG79 W58952 WO9814477-A1 Cancer
    HFSAG79 W58953 WO9814477-A1 Cancer
    HFSAG79 W58954 WO9814477-A1 Cancer
    HFSAG79 W58955 WO9814477-A1 Cancer
    HFSAG79 W58956 WO9814477-A1 Cancer
    HFSAG79 W58957 WO9814477-A1 Cancer
    HFSAG79 W58958 WO9814477-A1 Cancer
    HFSAG79 W58959 WO9814477-A1 Cancer
    HFSAG79 W58960 WO9814477-A1 Cancer
    HFSAG79 W58961 WO9814477-A1 Cancer
    HFSAG79 W58962 WO9814477-A1 Cancer
    HFSAG79 W58963 WO9814477-A1 Cancer
    HFSAG79 W58964 WO9814477-A1 Cancer
    HFSAG79 W58965 WO9814477-A1 Cancer
    HFSAG79 W58966 WO9814477-A1 Cancer
    HFSAG79 W58967 WO9814477-A1 Cancer
    HFSAG79 W58968 WO9814477-A1 Cancer
    HFSAG79 W58969 WO9814477-A1 Cancer
    HFSAG79 W58970 WO9814477-A1 Cancer
    HFSAG79 W58971 WO9814477-A1 Cancer
    HFSAG79 W58972 WO9814477-A1 Cancer
    HFSAG79 W58973 WO9814477-A1 Cancer
    HFSAG79 W58974 WO9814477-A1 Cancer
    HFSAG79 W58975 WO9814477-A1 Cancer
    HCEGH45 W59666 WO9824900-A1 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HHFCU19 W59753 US5786193-A Cancer
    HLMBP36 W59872 WO9831792-A1 Cancer
    HEMFI85 W59873 WO9831800-A2 Cancer
    HTXET53 W59874 WO9831800-A2 Cancer
    HBZAK03 W59876 WO9831800-A2 Cancer
    HLFBD44 W59877 WO9831800-A2 Cancer
    HEBGM49 W59878 WO9831800-A2 Cancer
    HNGBH54 W59879 WO9831800-A2 Cancer
    HSAAL25 W59880 WO9831800-A2 Cancer
    HSXCK41 W59882 WO9831800-A2 Cancer
    HFKFY79 W59883 WO9831800-A2 Cancer
    HAICH28 W59884 WO9831800-A2 Cancer
    HT1SB52 W60045 WO9818824-A1 Cancer
    HSDFB55 W60054 WO9816643-A1 Cancer
    HEBBC23 W60607 WO9820110-A1 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HTPBS22 W61600 WO9831798-A1 Cancer
    HMACR70 W61616 WO9831799-A2 Cancer
    HTEDK48 W61617 WO9831799-A2 Cancer
    HTPEF86 W61619 WO9831799-A2 Cancer
    HSBBF02 W61620 WO9831799-A2 Cancer
    HLTAH80 W61621 WO9831799-A2 Cancer
    HTPBA27 W61622 WO9831799-A2 Cancer
    HAIDQ59 W61623 WO9831799-A2 Cancer
    HHFEK40 W61624 WO9831799-A2 Cancer
    HGBGV89 W61625 WO9831799-A2 Digestive
    HUVBB80 W61626 WO9831799-A2 Cancer
    HJACE54 W61627 WO9831799-A2 Cancer
    HROAD63 W61628 WO9831799-A2 Connective/Epithelial,
    Digestive
    HMWGS46 W61629 WO9831799-A2 Cancer
    HNFGW06 W61630 WO9831799-A2 Cancer
    HFCAR05 W61912 WO9820042-A1 Cancer
    HHFHG78 W62595 WO9827932-A2 Cancer
    HBGBA67 W63123 WO9833915-A1 Cancer
    HPHAE52 W63622 WO9830694-A2 Cancer
    HTPCH84 W63623 WO9830694-A2 Cancer
    HEBCI67 W64433 WO9829438-A2 Cancer
    HCUDS60 W64483 WO9832856-A1 Cancer
    HPRCB54 W64668 WO9830693-A2 Cancer
    HTOCD71 W69220 WO9828421-A1 Cancer
    HSGSA61 W69221 WO9828420-A1 Cancer
    HSLAZ11 W69229 WO9831801-A1 Cancer
    HCEBJ50 W69230 WO9831801-A1 Cancer
    HMQDO20 W69231 WO9831806-A2 Cancer
    HDPMK33 W69232 WO9831806-A2 Cancer
    HMPAP73 W69233 WO9831806-A2 Immune/Hematopoietic
    HMSHH46 W69234 WO9831806-A2 Cancer
    HMAAB68 W69235 WO9831806-A2 Digestive,
    Immune/Hematopoietic
    HSDME38 W69508 WO9828422-A1 Cancer
    HOEBN05 W70286 WO9833920-A2 Cancer
    HDPMJ44 W70287 WO9835039-A1 Cancer
    HODAH63 W70330 WO9823749-A1 Neural/Sensory,
    Reproductive
    HETDW91 W70458 WO9838311-A1 Cancer
    HE8CV92 W70459 WO9838311-A1 Cancer
    HIBCL22 W70501 US5817477-A Cancer
    HKFBA76 W70525 WO9844111-A1 Cancer
    HKFBA76 W70526 WO9844111-A1 Cancer
    HMSAF34 W70594 WO9844118-A1 Cancer
    HMSAF34 W70596 WO9844118-A1 Cancer
    HMSAF34 W70597 WO9844118-A1 Cancer
    HRDCD54 W71592 WO9833912-A1 Cancer
    HIBEC52 W73130 US5830744-A Cancer
    HSRAW34 W73635 US5861272-A Cancer
    HBWAL95 W76212 WO9837194-A1 Cancer
    HTEJQ70 W76251 WO9831818-A2 Cancer
    HETBW05 W76253 WO9831818-A2 Digestive,
    Reproductive
    HATBG78 W77493 US5798223-A Endocrine
    HMWGS46 W78168 WO9856804-A1 Cancer
    HOUCQ17 W78189 WO9856804-A1 Cancer
    HMWGS46 W78295 WO9856804-A1 Cancer
    HLYBX88 W79083 WO9841629-A2 Cancer
    HTAAW41 W80212 WO9844112-A1 Cancer
    HOUCQ17 W80285 EP874050-A2 Cancer
    HMELK96 W81059 WO9856892-A1 Cancer
    HLJBI75 W81071 WO9851794-A1 Cancer
    HFCBS02 W81106 WO9844109-A1 Cancer
    HHPGS02 W81576 WO9850549-A2 Cancer
    HTOBH93 W83929 US5844081-A Cancer
    HSSAE30 W84184 WO9853069-A2 Cancer
    HCQAS17 W84274 US5861494-A Digestive,
    Mixed Fetal,
    Reproductive
    HRGBQ38 W85561 US5849286-A Cancer
    HFCCE09 W85562 US5849286-A Cancer
    HGOCA18 W85563 US5849286-A Cancer
    HMSIB42 W87769 WO9854199-A1 Cancer
    HTECE68 W89575 US5858705-A Cancer
    HESAJ20 W92460 US5871969-A Cancer
    HESAJ20 W92469 US5871969-A Cancer
    HTXEI33 W92523 US5874240-A Cancer
    HTXEI33 W92524 US5874240-A Cancer
    HKABO35 W92792 WO9854202-A1 Cancer
    HCEGH45 W94074 US5869632-A Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HNFIR05 W94466 WO9900415-A1 Cancer
    HTTBN61 W95538 JP11000170-A Cancer
    HPFCA19 W96192 WO9900498-A1 Cancer
    HPFCA19 W96193 WO9900498-A1 Cancer
    HTSGS30 W97350 WO9903982-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HDTAH85 Y01098 WO9910364-A1 Cancer
    HFJAB36 Y02608 WO9923106-A1 Cancer
    HDTBS70 Y03231 WO9909152-A1 Cancer
    HNGEF08 Y03849 WO9909198-A1 Immune/Hematopoietic,
    Reproductive
    HUKEJ46 Y03850 WO9909198-A1 Digestive,
    Reproductive
    HPASD50 Y04120 WO9909161-A1 Cancer
    HPASD50 Y04121 WO9909161-A1 Cancer
    HAGFE38 Y05451 WO9857989-A1 Cancer
    HFVIF40 Y06461 WO9931116-A1 Cancer
    HFCCQ50 Y06462 WO9931116-A1 Cancer
    HT4CC72 Y06473 WO9935262-A2 Immune/Hematopoietic
    HDPIE88 Y06511 WO9936565-A1 Cancer
    HWFBG79 Y10797 WO9907891-A1 Cancer
    HDGRC02 Y13736 US5928890-A Cancer
    HFCET92 Y14078 WO9921575-A1 Cancer
    HUVEO91 Y14132 WO9923105-A1 Cancer
    HUVEO91 Y14133 WO9923105-A1 Cancer
    HCABA58 Y16587 US5916769-A Cancer
    HOSBD47 Y22320 US5932540-A Cancer
    HOSBD47 Y22321 US5932540-A Cancer
    HPRCC57 Y23761 WO9932135-A1 Cancer
    HMEAA94 Y23884 WO9935160-A1 Cancer
    HL1AP03 Y23885 WO9935160-A1 Cancer
    HSYBM46 Y23886 WO9935160-A1 Cancer
    HFKBC47 Y23887 WO9935160-A1 Cancer
    HSSAWS4 Y24249 US5929225-A Cancer
    HCUDE60 Y25708 WO9938882-A1 Cancer
    HHFCU19 Y27005 US5928924-A Cancer
    HMWJH67 Y28640 WO9940183-A1 Cancer
    HKAFV61 Y28642 WO9940183-A1 Cancer
    HETDK50 Y28643 WO9940183-A1 Cancer
    HKAEF09 Y28644 WO9940183-A1 Cancer
    HOSBD47 Y30518 WO9946364-A1 Cancer
    HOSBD47 Y30519 WO9946364-A1 Cancer
    HILBI36 Y31242 US5955339-A Cancer
    HTAEK53 Y31810 WO9947538-A1 Cancer
    HT4CC72 Y31885 WO9942584-A1 lmmune/Hematopoietic
    HLMBA70 Y32504 US5945309-A Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HPRCC57 Y32888 WO9941282-A1 Cancer
    HPRCC57 Y32895 WO9941282-A1 Cancer
    HPRCC57 Y32896 WO9941282-A1 Cancer
    HPRCC57 Y32897 WO9941282-A1 Cancer
    HPRCC57 Y32898 WO9941282-A1 Cancer
    HPRCC57 Y32901 WO9941282-A1 Cancer
    HPRCC57 Y32905 WO9941282-A1 Cancer
    HPRCC57 Y32916 WO9941282-A1 Cancer
    HMEIP65 Y33847 US5952197-A Cancer
    HFCCQ50 Y36339 WO9931117-A1 Cancer
    HFCCQ50 Y36342 WO9931117-A1 Cancer
    HRDCD54 Y36648 WO9931117-A1 Cancer
    HRDCD54 Y36650 WO9931117-A1 Cancer
    HRDCD54 Y36673 WO9931117-A1 Cancer
    HTSEX82 Y41161 US5981231-A Digestive,
    Immune/Hematopoietic
    HGBAN46 Y41163 US5981230-A Cancer
    HE9DR66 Y41164 US5981230-A Cancer
    HRGBQ38 Y42150 US5968797-A Cancer
    HFCCE09 Y42151 US5968797-A Cancer
    HGOCA18 Y42152 US5968797-A Cancer
    HNFEM05 Y42165 WO9927078-A1 Cancer
    HJACE54 Y44510 WO200001728-A1 Cancer
    HKAEF92 Y44664 WO9962934-A1 Cancer
    HBZSD43 Y45003 WO200006589-A1 Cancer
    HUVEO91 Y45032 WO200008139-A1 Cancer
    HTOBH93 Y49535 US5977309-A Cancer
    HAPOR40 Y49946 WO9914240-A1 Cancer
    HHEAC71 Y52158 WO9920758-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HCFAZ22 Y52159 WO9920758-A1 Cancer
    HT5EA78 Y52160 WO9920758-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HDPJO39 Y52479 WO9940184-A1 Cancer
    HBICD95 Y53061 US5998171-A Cancer
    HTGED19 Y53890 WO9961617-A1 Immune/Hematopoietic
    HFPBX96 Y53891 WO9961617-A1 Cancer
    HFKCU96 Y54900 US5986069-A Cancer
    HSBBC75 Y55748 US5994103-A Cancer
    HLFBE10 Y55750 US5994103-A Cancer
    HLFBE10 Y57166 US5994301-A Cancer
    HIBCL22 Y57167 US5994506-A Cancer
    HTTER36 Y58185 US6004780-A Cardiovascular,
    Connective/Epithelial,
    Reproductive
    HWHGU74 Y59247 WO9962927-A1 Cancer
    HSDFB55 Y67239 US6008020-A Cancer
    HE2BG16 Y67356 US5998164-A Cancer
    HKAPI15 Y68800 WO200005371-A1 Connective/Epithelial
    HTWAF38 Y69674 US6013483-A Cancer
    HATCK89 Y69675 US6013477-A Cancer
    HAPOR40 Y70591 WO200015759-A1 Cancer
    HMUAN45 Y70785 WO200023572-A1 Cancer
    HATCK89 Y71884 WO200067775-A1 Cancer
    HKGDL36 Y71959 WO200066778-A1 Cancer
    HCUDS60 Y72022 WO200067793-A1 Cancer
    HCUDS60 Y72023 WO200067793-A1 Cancer
    HETAN67 Y78790 US6013469-A Cancer
    HDGNR10 Y80128 US6025154-A Digestive,
    Immune/Hematopoietic,
    Reproductive
    HBGBA67 Y87779 US6054289-A Cancer
    HE2CB95 Y87780 US6054289-A Immune/Hematopoietic,
    Mixed Fetal
    HPTTK55 Y87782 US6054289-A Cancer
    HARAO63 Y87783 US6054289-A Cancer
    HLHAR55 Y87787 US6054289-A Cancer
    HSRDG78 Y87788 US6054289-A Cancer
    HCCAA03 Y87789 US6054289-A Cancer
    HWLLM34 Y90351 WO200052136-A2 Cancer
    HA5AA37 Y90352 WO200052136-A2 Cancer
    HDPAK85 Y90353 WO200052136-A2 Cancer
    HPHAE52 Y90357 WO200052028-A1 Cancer
    HTPCH84 Y90358 WO200052028-A1 Cancer
    HMKEA94 Y93650 WO200036105-A1 Cancer
    HOEDH76 Y93912 WO200039166-A1 Cancer
    HOGCC45 Y93951 WO200039136-A2 Cancer
    HTSGS30 Y93973 WO200042189-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HTSGS30 Y93975 WO200042189-A1 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HMWCF06 Y94802 WO200009148-A1 Cancer
    HE9DR66 Y95534 WO200040726-A1 Cancer
    HGBAN46 Y95535 WO200040726-A1 Cancer
    HE9DR66 Y95563 WO200040726-A1 Cancer
    HE9DR66 Y95565 WO200040726-A1 Cancer
    HE9DR66 Y95566 WO200040726-A1 Cancer
    HE9DR66 Y95567 WO200040726-A1 Cancer
    HE9DR66 Y95568 WO200040726-A1 Cancer
    HE9DR66 Y95569 WO200040726-A1 Cancer
    HE9DR66 Y95570 WO200040726-A1 Cancer
    HE9DR66 Y95571 WO200040726-A1 Cancer
    HE9DR66 Y95572 WO200040726-A1 Cancer
    HE9DR66 Y95573 WO200040726-A1 Cancer
    HE9DR66 Y95574 WO200040726-A1 Cancer
    HE9DR66 Y95575 WO200040726-A1 Cancer
    HE9DR66 Y95576 WO200040726-A1 Cancer
    HE9DR66 Y95577 WO200040726-A1 Cancer
    HE9DR66 Y95578 WO200040726-A1 Cancer
    HHEAC71 Y95879 WO200050459-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HCFAZ22 Y95880 WO200050459-A1 Cancer
    HT5EA78 Y95881 WO200050459-A1 Connective/Epithelial,
    Immune/Hematopoietic
    HDPAK85 Y96099 WO200052135-A2 Cancer
    HWLLM34 Y96100 WO200052135-A2 Cancer
    HA5AA37 Y96101 WO200052135-A2 Cancer
    HAPAT57 Y96280 WO200028035-A1 Cancer
    HAPAT57 Y96282 WO200028035-A1 Cancer
    HKABZ65 Y96962 WO200039327-A1 Connective/Epithelial
    HWHGB15 Y96963 WO200039327-A1 Connective/Epithelial
    HCDDP40 Y96964 WO200039327-A1 Immune/Hematopoietic,
    Musculoskeletal
    HOSBD47 Y97144 WO200045835-A1 Cancer
    HOSBD47 Y97145 WO200045835-A1 Cancer
    HFITF82 SEQ ID NO: 73 Immune/Hematopoietic,
    Musculoskeletal
    HFITF82 SEQ ID NO: 74 Immune/Hematopoietic,
    Musculoskeletal
    HFITF82 SEQ ID NO: 75 Immune/Hematopoietic,
    Musculoskeletal
    HFITF82 SEQ ID NO: 76 Immune/Hematopoietic,
    Musculoskeletal
    HBZAI19 SEQ ID NO: 77 Immune/Hematopoietic,
    Reproductive
    HBZAI19 SEQ ID NO: 78 Immune/Hematopoietic,
    Reproductive
    HBZAI19 SEQ ID NO: 79 Immune/Hematopoietic,
    Reproductive
    HDPDI45 SEQ ID NO: 80 Cancer
    HDPDI45 SEQ ID NO: 81 Cancer
    HETHW90 SEQ ID NO: 82 Cancer
    HETHW90 SEQ ID NO: 83 Cancer
    HETHW90 SEQ ID NO: 84 Cancer
    HIBEB47 SEQ ID NO: 85 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HIBEB47 SEQ ID NO: 86 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HIBEB47 SEQ ID NO: 87 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HIBEB47 SEQ ID NO: 88 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HLHFR58 SEQ ID NO: 89 Cancer
    HLHFR58 SEQ ID NO: 90 Cancer
    HLHFR58 SEQ ID NO: 91 Cancer
    HLHFR58 SEQ ID NO: 92 Cancer
    HNGGK54 SEQ ID NO: 93 Cancer
    HNGGK54 SEQ ID NO: 94 Cancer
    HNGGK54 SEQ ID NO: 95 Cancer
    HNGGK54 SEQ ID NO: 96 Cancer
    HUSIE23 SEQ ID NO: 97 Cancer
    HUSIE23 SEQ ID NO: 98 Cancer
    HARMB79 SEQ ID NO: 99 Cancer
    HARMB79 SEQ ID NO: 100 Cancer
    HJBCY84 SEQ ID NO: 101 Cancer
    HJBCY84 SEQ ID NO: 102 Cancer
    HJBCY84 SEQ ID NO: 103 Cancer
    HCMSC92 SEQ ID NO: 104 Cancer
    HCMSC92 SEQ ID NO: 105 Cancer
    HE2AX96 SEQ ID NO: 106 Mixed Fetal
    HE2AX96 SEQ ID NO: 107 Mixed Fetal
    HE2AX96 SEQ ID NO: 108 Mixed Fetal
    HHPDV90 SEQ ID NO: 109 Cancer
    HHPDV90 SEQ ID NO: 110 Cancer
    HHPDV90 SEQ ID NO: 111 Cancer
    HT2SG64 SEQ ID NO: 112 Digestive,
    Immune/Hematopoietic
    HT2SG64 SEQ ID NO: 113 Digestive,
    Immune/Hematopoietic
    HT2SG64 SEQ ID NO: 114 Digestive,
    Immune/Hematopoietic
    HAGAN21 SEQ ID NO: 115 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HAGAN21 SEQ ID NO: 116 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HAGAN21 SEQ ID NO: 117 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HAGAN21 SEQ ID NO: 118 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HAGAN21 SEQ ID NO: 119 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HEBAH57 SEQ ID NO: 120 Neural/Sensory
    HEBAH57 SEQ ID NO: 121 Neural/Sensory
    HEBAH57 SEQ ID NO: 122 Neural/Sensory
    HETDB76 SEQ ID NO: 123 Musculoskeletal,
    Reproductive
    HETDB76 SEQ ID NO: 124 Musculoskeletal,
    Reproductive
    HETDB76 SEQ ID NO: 125 Musculoskeletal,
    Reproductive
    HETDB76 SEQ ID NO: 126 Musculoskeletal,
    Reproductive
    HE8SE91 SEQ ID NO: 127 Cancer
    HE8SE91 SEQ ID NO: 128 Cancer
    HE8SE91 SEQ ID NO: 129 Cancer
    HRGBL78 SEQ ID NO: 130 Cancer
    HRGBL78 SEQ ID NO: 131 Cancer
    HRGBL78 SEQ ID NO: 132 Cancer
    HRGBL78 SEQ ID NO: 133 Cancer
    HHFUC40 SEQ ID NO: 134 Cardiovascular
    HHFUC40 SEQ ID NO: 135 Cardiovascular
    HETCP58 SEQ ID NO: 136 Immune/Hematopoietic,
    Reproductive
    HETCP58 SEQ ID NO: 137 Immune/Hematopoietic,
    Reproductive
    HETCP58 SEQ ID NO: 138 Immune/Hematopoietic,
    Reproductive
    HTTBM40 SEQ ID NO: 139 Cancer
    HTTBM40 SEQ ID NO: 140 Cancer
    HTTBS64 SEQ ID NO: 141 Reproductive
    HTTBS64 SEQ ID NO: 142 Reproductive
    HTTBS64 SEQ ID NO: 143 Reproductive
    HCEVB32 SEQ ID NO: 144 Cancer
    HCEVB32 SEQ ID NO: 145 Cancer
    HCEVB32 SEQ ID NO: 146 Cancer
    HCEVB32 SEQ ID NO: 147 Cancer
    HHPFU18 SEQ ID NO: 148 Cancer
    HHPFU18 SEQ ID NO: 149 Cancer
    HPRCA90 SEQ ID NO: 150 Cancer
    HPRCA90 SEQ ID NO: 151 Cancer
    HPRCA90 SEQ ID NO: 152 Cancer
    HPRCA90 SEQ ID NO: 153 Cancer
    HPRCE33 SEQ ID NO: 154 Cancer
    HPRCE33 SEQ ID NO: 155 Cancer
    HHFFU55 SEQ ID NO: 156 Cardiovascular,
    Immune/Hematopoietic
    HHFFU55 SEQ ID NO: 157 Cardiovascular,
    Immune/Hematopoietic
    HUVDP63 SEQ ID NO: 158 Cancer
    HUVDP63 SEQ ID NO: 159 Cancer
    HUVDP63 SEQ ID NO: 160 Cancer
    HUVDP63 SEQ ID NO: 161 Cancer
    HUVDP63 SEQ ID NO: 162 Cancer
    HCEFI77 SEQ ID NO: 163 Neural/Sensory
    HCEFI77 SEQ ID NO: 164 Neural/Sensory
    HCEFI77 SEQ ID NO: 165 Neural/Sensory
    HHFDH56 SEQ ID NO: 166 Cancer
    HHFDN48 SEQ ID NO: 167 Cancer
    HHFDN48 SEQ ID NO: 168 Cancer
    HHFDN48 SEQ ID NO: 169 Cancer
    HHFDN48 SEQ ID NO: 170 Cancer
    HHFDN48 SEQ ID NO: 171 Cancer
    HHFDN67 SEQ ID NO: 172 Cardiovascular
    HHFDN67 SEQ ID NO: 173 Cardiovascular
    HHFDG51 SEQ ID NO: 174 Connective/Epithelial,
    Musculoskeletal
    HHFDG51 SEQ ID NO: 175 Connective/Epithelial,
    Musculoskeletal
    HHFDG51 SEQ ID NO: 176 Connective/Epithelial,
    Musculoskeletal
    HE8AO36 SEQ ID NO: 177 Cancer
    HE8AO36 SEQ ID NO: 178 Cancer
    HE8AO36 SEQ ID NO: 179 Cancer
    HTPAB57 SEQ ID NO: 180 Cancer
    HTPAB57 SEQ ID NO: 181 Cancer
    HTPAB57 SEQ ID NO: 182 Cancer
    HTPAB57 SEQ ID NO: 183 Cancer
    HFXAX45 SEQ ID NO: 184 Neural/Sensory
    HFXAX45 SEQ ID NO: 185 Neural/Sensory
    HFXAX45 SEQ ID NO: 186 Neural/Sensory
    HTLBE23 SEQ ID NO: 187 Reproductive
    HTLBE23 SEQ ID NO: 188 Reproductive
    HCQAM33 SEQ ID NO: 189 Musculoskeletal,
    Reproductive
    HCQAM33 SEQ ID NO: 190 Musculoskeletal,
    Reproductive
    HCQAM33 SEQ ID NO: 191 Musculoskeletal,
    Reproductive
    HCEWE17 SEQ ID NO: 192 Digestive,
    Neural/Sensory
    HCEWE17 SEQ ID NO: 193 Digestive,
    Neural/Sensory
    HCEWE17 SEQ ID NO: 194 Digestive,
    Neural/Sensory
    HTEGI42 SEQ ID NO: 195 Cancer
    HTEGI42 SEQ ID NO: 196 Cancer
    HTEGI42 SEQ ID NO: 197 Cancer
    HTEGI42 SEQ ID NO: 198 Cancer
    HTEGI42 SEQ ID NO: 199 Cancer
    HCEIE80 SEQ ID NO: 200 Cancer
    HCEIE80 SEQ ID NO: 201 Cancer
    HCEIE80 SEQ ID NO: 202 Cancer
    HCEIE80 SEQ ID NO: 203 Cancer
    HLMCA92 SEQ ID NO: 204 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HLMCA92 SEQ ID NO: 205 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HLMCA92 SEQ ID NO: 206 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HLMCA92 SEQ ID NO: 207 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HLHCF36 SEQ ID NO: 208 Respiratory
    HLHCF36 SEQ ID NO: 209 Respiratory
    HLHCF36 SEQ ID NO: 210 Respiratory
    HCEZR26 SEQ ID NO: 211 Cancer
    HCEZR26 SEQ ID NO: 212 Cancer
    HGBCO51 SEQ ID NO: 213 Cancer
    HGBCO51 SEQ ID NO: 214 Cancer
    HGBCO51 SEQ ID NO: 215 Cancer
    HGBCO51 SEQ ID NO: 216 Cancer
    HTABP30 SEQ ID NO: 217 Cancer
    HTABP30 SEQ ID NO: 218 Cancer
    HUKCD10 SEQ ID NO: 219 Cancer
    HUKCD10 SEQ ID NO: 220 Cancer
    HUKCD10 SEQ ID NO: 221 Cancer
    HOUHT39 SEQ ID NO: 222 Cancer
    HOUHT39 SEQ ID NO: 223 Cancer
    HOUHT39 SEQ ID NO: 224 Cancer
    HTXBN56 SEQ ID NO: 225 Cancer
    HTXBN56 SEQ ID NO: 226 Cancer
    HTXBN56 SEQ ID NO: 227 Cancer
    HETEU28 SEQ ID NO: 228 Cancer
    HETEU28 SEQ ID NO: 229 Cancer
    HODDD43 SEQ ID NO: 230 Cancer
    HODDD43 SEQ ID NO: 231 Cancer
    HODDD43 SEQ ID NO: 232 Cancer
    HPWAL61 SEQ ID NO: 233 Musculoskeletal,
    Reproductive
    HPWAL61 SEQ ID NO: 234 Musculoskeletal,
    Reproductive
    HPWAL61 SEQ ID NO: 235 Musculoskeletal,
    Reproductive
    HPWAL61 SEQ ID NO: 236 Musculoskeletal,
    Reproductive
    HTSER67 SEQ ID NO: 237 Cancer
    HTSER67 SEQ ID NO: 238 Cancer
    HMSDL37 SEQ ID NO: 239 Cancer
    HMSDL37 SEQ ID NO: 240 Cancer
    HMSDL37 SEQ ID NO: 241 Cancer
    HMSDL37 SEQ ID NO: 242 Cancer
    HSDAJ53 SEQ ID NO: 243 Cancer
    HSDAJ53 SEQ ID NO: 244 Cancer
    HSDAJ53 SEQ ID NO: 245 Cancer
    HSDAJ53 SEQ ID NO: 246 Cancer
    HEBDF05 SEQ ID NO: 247 Neural/Sensory
    HEBDF05 SEQ ID NO: 248 Neural/Sensory
    HEBDF05 SEQ ID NO: 249 Neural/Sensory
    HSQFT30 SEQ ID NO: 250 Cancer
    HSQFT30 SEQ ID NO: 251 Cancer
    HSIDX71 SEQ ID NO: 252 Digestive,
    Neural/Sensory
    HSIDX71 SEQ ID NO: 253 Digestive,
    Neural/Sensory
    HSAUA82 SEQ ID NO: 254 Immune/Hematopoietic,
    Reproductive
    HSAUA82 SEQ ID NO: 255 Immune/Hematopoietic,
    Reproductive
    HPWAY46 SEQ ID NO: 256 Cancer
    HPWAY46 SEQ ID NO: 257 Cancer
    HPWAY46 SEQ ID NO: 258 Cancer
    HSSEN70 SEQ ID NO: 259 Cancer
    HSSEN70 SEQ ID NO: 260 Cancer
    HTOHB55 SEQ ID NO: 261 Cancer
    HTOHB55 SEQ ID NO: 262 Cancer
    HTOHM15 SEQ ID NO: 263 Cancer
    HTOHM15 SEQ ID NO: 264 Cancer
    HTOHM15 SEQ ID NO: 265 Cancer
    HTOHM15 SEQ ID NO: 266 Cancer
    HHNAB56 SEQ ID NO: 267 Digestive
    HHNAB56 SEQ ID NO: 268 Digestive
    HHNAB56 SEQ ID NO: 269 Digestive
    HJABL02 SEQ ID NO: 270 Cancer
    HJABL02 SEQ ID NO: 271 Cancer
    HJACG30 SEQ ID NO: 272 Immune/Hematopoietic
    HJACG30 SEQ ID NO: 273 Immune/Hematopoietic
    HJACG30 SEQ ID NO: 274 Immune/Hematopoietic
    HTAEE28 SEQ ID NO: 275 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HTAEE28 SEQ ID NO: 276 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HTAEE28 SEQ ID NO: 277 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HTHBG43 SEQ ID NO: 278 Immune/Hematopoietic
    HTHBG43 SEQ ID NO: 279 Immune/Hematopoietic
    HJPCE80 SEQ ID NO: 280 Cancer
    HJPCE80 SEQ ID NO: 281 Cancer
    HJPCE80 SEQ ID NO: 282 Cancer
    HTOIZ02 SEQ ID NO: 283 Cancer
    HTOIZ02 SEQ ID NO: 284 Cancer
    HJPCR70 SEQ ID NO: 285 Cancer
    HJPCR70 SEQ ID NO: 286 Cancer
    HJPCR70 SEQ ID NO: 287 Cancer
    HJPCR70 SEQ ID NO: 288 Cancer
    HJPCP42 SEQ ID NO: 289 Digestive,
    Immune/Hematopoietic
    HJPCP42 SEQ ID NO: 290 Digestive,
    Immune/Hematopoietic
    HJPCP42 SEQ ID NO: 291 Digestive,
    Immune/Hematopoietic
    HJPCP42 SEQ ID NO: 292 Digestive,
    Immune/Hematopoietic
    HNFFD47 SEQ ID NO: 293 Immune/Hematopoietic
    HNFFD47 SEQ ID NO: 294 Immune/Hematopoietic
    HNFFD47 SEQ ID NO: 295 Immune/Hematopoietic
    HNFFI46 SEQ ID NO: 296 Cancer
    HNFFI46 SEQ ID NO: 297 Cancer
    HNFFI46 SEQ ID NO: 298 Cancer
    HNFFI46 SEQ ID NO: 299 Cancer
    HNFFI46 SEQ ID NO: 300 Cancer
    HTOIQ42 SEQ ID NO: 301 Cancer
    HTOIQ42 SEQ ID NO: 302 Cancer
    HLTDW13 SEQ ID NO: 303 Cancer
    HLTDW13 SEQ ID NO: 304 Cancer
    HLTDW13 SEQ ID NO: 305 Cancer
    HLTDW13 SEQ ID NO: 306 Cancer
    HLTDW13 SEQ ID NO: 307 Cancer
    HLTDY51 SEQ ID NO: 308 Cancer
    HLTDY51 SEQ ID NO: 309 Cancer
    HNFFZ56 SEQ ID NO: 310 Cancer
    HNFFZ56 SEQ ID NO: 311 Cancer
    HNGAV54 SEQ ID NO: 312 Immune/Hematopoietic
    HNGAV54 SEQ ID NO: 313 Immune/Hematopoietic
    HSLCA15 SEQ ID NO: 314 Cancer
    HSLCA15 SEQ ID NO: 315 Cancer
    HSLCA15 SEQ ID NO: 316 Cancer
    HSLCA15 SEQ ID NO: 317 Cancer
    HSLCA15 SEQ ID NO: 318 Cancer
    HSLCA15 SEQ ID NO: 319 Cancer
    HSLCP57 SEQ ID NO: 320 Cancer
    HSLCP57 SEQ ID NO: 321 Cancer
    HTOJP95 SEQ ID NO: 322 Immune/Hematopoietic
    HTOJP95 SEQ ID NO: 323 Immune/Hematopoietic
    HBMVI55 SEQ ID NO: 324 Cancer
    HBMVI55 SEQ ID NO: 325 Cancer
    HBMVI55 SEQ ID NO: 326 Cancer
    HBMVI55 SEQ ID NO: 327 Cancer
    HBMVI55 SEQ ID NO: 328 Cancer
    HFXBS68 SEQ ID NO: 329 Neural/Sensory
    HFXBS68 SEQ ID NO: 330 Neural/Sensory
    HFXBS68 SEQ ID NO: 331 Neural/Sensory
    HFXBS68 SEQ ID NO: 332 Neural/Sensory
    HNGBC07 SEQ ID NO: 333 Immune/Hematopoietic
    HNGBC07 SEQ ID NO: 334 Immune/Hematopoietic
    HNGBC07 SEQ ID NO: 335 Immune/Hematopoietic
    HMSFK67 SEQ ID NO: 336 Cancer
    HMSFK67 SEQ ID NO: 337 Cancer
    HMSFK67 SEQ ID NO: 338 Cancer
    HCE1P80 SEQ ID NO: 339 Cancer
    HCE1P80 SEQ ID NO: 340 Cancer
    HCE1P80 SEQ ID NO: 341 Cancer
    HOUDU29 SEQ ID NO: 342 Cancer
    HOUDU29 SEQ ID NO: 343 Cancer
    HOUDU29 SEQ ID NO: 344 Cancer
    HOUDU29 SEQ ID NO: 345 Cancer
    HOUDU29 SEQ ID NO: 346 Cancer
    HHFEC49 SEQ ID NO: 347 Cancer
    HCE3T57 SEQ ID NO: 348 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCE3T57 SEQ ID NO: 349 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCE3T57 SEQ ID NO: 350 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCE3T57 SEQ ID NO: 351 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCE3T57 SEQ ID NO: 352 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCE4Y07 SEQ ID NO: 353 Cancer
    HCE4Y07 SEQ ID NO: 354 Cancer
    HCE5G23 SEQ ID NO: 355 Cancer
    HCE5G23 SEQ ID NO: 356 Cancer
    HCE5G23 SEQ ID NO: 357 Cancer
    HFCEP45 SEQ ID NO: 358 Neural/Sensory
    HFCEP45 SEQ ID NO: 359 Neural/Sensory
    HFCEP45 SEQ ID NO: 360 Neural/Sensory
    HFCEP45 SEQ ID NO: 361 Neural/Sensory
    HMWEJ52 SEQ ID NO: 362 Immune/Hematopoietic
    HMWEJ52 SEQ ID NO: 363 Immune/Hematopoietic
    HMWEY26 SEQ ID NO: 364 Cancer
    HMWEY26 SEQ ID NO: 365 Cancer
    HMWEY26 SEQ ID NO: 366 Cancer
    HMWEY26 SEQ ID NO: 367 Cancer
    HMWEY26 SEQ ID NO: 368 Cancer
    HATDM46 SEQ ID NO: 369 Cancer
    HATDM46 SEQ ID NO: 370 Cancer
    HATDM46 SEQ ID NO: 371 Cancer
    HATDM46 SEQ ID NO: 372 Cancer
    HATDM46 SEQ ID NO: 373 Cancer
    HATDM46 SEQ ID NO: 374 Cancer
    HHFHD37 SEQ ID NO: 375 Cardiovascular,
    Immune/Hematopoietic,
    Respiratory
    HHFHD37 SEQ ID NO: 376 Cardiovascular,
    Immune/Hematopoietic,
    Respiratory
    HHFHI76 SEQ ID NO: 377 Cancer
    HHFHI76 SEQ ID NO: 378 Cancer
    HATDZ29 SEQ ID NO: 379 Endocrine,
    Immune/Hematopoietic
    HATDZ29 SEQ ID NO: 380 Endocrine,
    Immune/Hematopoietic
    HFVGE32 SEQ ID NO: 381 Digestive,
    Immune/Hematopoietic
    HFVGE32 SEQ ID NO: 382 Digestive,
    Immune/Hematopoietic
    HLHFE92 SEQ ID NO: 383 Cancer
    HLHFE92 SEQ ID NO: 384 Cancer
    HLHFE92 SEQ ID NO: 385 Cancer
    HMKAI25 SEQ ID NO: 386 Cancer
    HMKAI25 SEQ ID NO: 387 Cancer
    HMKAI25 SEQ ID NO: 388 Cancer
    HMKAI25 SEQ ID NO: 389 Cancer
    HMKAI25 SEQ ID NO: 390 Cancer
    HNHEI42 SEQ ID NO: 391 Endocrine,
    Immune/Hematopoietic
    HNHEI42 SEQ ID NO: 392 Endocrine,
    Immune/Hematopoietic
    HNHEI42 SEQ ID NO: 393 Endocrine,
    Immune/Hematopoietic
    HNHEI42 SEQ ID NO: 394 Endocrine,
    Immune/Hematopoietic
    HNHEI85 SEQ ID NO: 395 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HNHEI85 SEQ ID NO: 396 Digestive,
    Immune/Hematopoietic,
    Musculoskeletal
    HOEDE28 SEQ ID NO: 397 Cancer
    HOEDE28 SEQ ID NO: 398 Cancer
    H2CBH03 SEQ ID NO: 399 Cancer
    HTHCA18 SEQ ID NO: 400 Immune/Hematopoietic
    HTHCA18 SEQ ID NO: 401 Immune/Hematopoietic
    HTHCO79 SEQ ID NO: 402 Cancer
    HTHCO79 SEQ ID NO: 403 Cancer
    HNGFB76 SEQ ID NO: 404 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HNGFB76 SEQ ID NO: 405 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HNGFB76 SEQ ID NO: 406 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HNGFB76 SEQ ID NO: 407 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HOQBJ82 SEQ ID NO: 408 Cancer
    HOQBJ82 SEQ ID NO: 409 Cancer
    HNFHY51 SEQ ID NO: 410 Immune/Hematopoietic,
    Reproductive
    HNFHY51 SEQ ID NO: 411 Immune/Hematopoietic,
    Reproductive
    HNFHY51 SEQ ID NO: 412 Immune/Hematopoietic,
    Reproductive
    HNFHY51 SEQ ID NO: 413 Immune/Hematopoietic,
    Reproductive
    HNEEB45 SEQ ID NO: 414 Immune/Hematopoietic,
    Mixed Fetal
    HNEEB45 SEQ ID NO: 415 Immune/Hematopoietic,
    Mixed Fetal
    HSDFA44 SEQ ID NO: 416 Neural/Sensory
    HSDFA44 SEQ ID NO: 417 Neural/Sensory
    HSDFA44 SEQ ID NO: 418 Neural/Sensory
    HAGEB14 SEQ ID NO: 419 Cancer
    HAGEB14 SEQ ID NO: 420 Cancer
    HCGBE81 SEQ ID NO: 421 Neural/Sensory,
    Reproductive
    HCGBE81 SEQ ID NO: 422 Neural/Sensory,
    Reproductive
    HEOMX53 SEQ ID NO: 423 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HEOMX53 SEQ ID NO: 424 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HEOMX53 SEQ ID NO: 425 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HEONC95 SEQ ID NO: 426 Cancer
    HEONC95 SEQ ID NO: 427 Cancer
    HKMLP68 SEQ ID NO: 428 Excretory,
    Mixed Fetal,
    Reproductive
    HKMLP68 SEQ ID NO: 429 Excretory,
    Mixed Fetal,
    Reproductive
    HKMLP68 SEQ ID NO: 430 Excretory,
    Mixed Fetal,
    Reproductive
    HMWIG83 SEQ ID NO: 431 Cancer
    HMWIG83 SEQ ID NO: 432 Cancer
    HMSKH19 SEQ ID NO: 433 Cancer
    HMSKH19 SEQ ID NO: 434 Cancer
    HMSKH19 SEQ ID NO: 435 Cancer
    HFAME37 SEQ ID NO: 436 Neural/Sensory
    HFAME37 SEQ ID NO: 437 Neural/Sensory
    HFAME37 SEQ ID NO: 438 Neural/Sensory
    HFXFG45 SEQ ID NO: 439 Immune/Hematopoietic,
    Neural/Sensory
    HFXFG45 SEQ ID NO: 440 Immune/Hematopoietic,
    Neural/Sensory
    HFXFG45 SEQ ID NO: 441 Immune/Hematopoietic,
    Neural/Sensory
    HFXFG45 SEQ ID NO: 442 Immune/Hematopoietic,
    Neural/Sensory
    HFXFH04 SEQ ID NO: 443 Immune/Hematopoietic,
    Neural/Sensory
    HFXFH04 SEQ ID NO: 444 Immune/Hematopoietic,
    Neural/Sensory
    HFXFH04 SEQ ID NO: 445 Immune/Hematopoietic,
    Neural/Sensory
    HFXFH04 SEQ ID NO: 446 Immune/Hematopoietic,
    Neural/Sensory
    HGCAC66 SEQ ID NO: 447 Cancer
    HGCAC66 SEQ ID NO: 448 Cancer
    HSSJF55 SEQ ID NO: 449 Musculoskeletal
    HSSJF55 SEQ ID NO: 450 Musculoskeletal
    HFXHM17 SEQ ID NO: 451 Cancer
    HFXHM17 SEQ ID NO: 452 Cancer
    HFXHM17 SEQ ID NO: 453 Cancer
    HFXHM17 SEQ ID NO: 454 Cancer
    HOSFQ65 SEQ ID NO: 455 Cancer
    HOSFQ65 SEQ ID NO: 456 Cancer
    HOSFQ65 SEQ ID NO: 457 Cancer
    HOSFQ65 SEQ ID NO: 458 Cancer
    HOSFQ65 SEQ ID NO: 459 Cancer
    HKGAS32 SEQ ID NO: 460 Connective/Epithelial,
    Neural/Sensory
    HKGAS32 SEQ ID NO: 461 Connective/Epithelial,
    Neural/Sensory
    HKGAU45 SEQ ID NO: 462 Immune/Hematopoietic
    HKGAU45 SEQ ID NO: 463 Immune/Hematopoietic
    HKGAU45 SEQ ID NO: 464 Immune/Hematopoietic
    HKGBH24 SEQ ID NO: 465 Cancer
    HKGBH24 SEQ ID NO: 466 Cancer
    HKGBH24 SEQ ID NO: 467 Cancer
    HKGBS01 SEQ ID NO: 468 Cancer
    HKGBS01 SEQ ID NO: 469 Cancer
    HKGBS01 SEQ ID NO: 470 Cancer
    HACCL63 SEQ ID NO: 471 Cancer
    HACCL63 SEQ ID NO: 472 Cancer
    HACCL63 SEQ ID NO: 473 Cancer
    HACCL63 SEQ ID NO: 474 Cancer
    HFIIN69 SEQ ID NO: 475 Musculoskeletal,
    Neural/Sensory,
    Reproductive
    HFIIN69 SEQ ID NO: 476 Musculoskeletal,
    Neural/Sensory,
    Reproductive
    HFIIN69 SEQ ID NO: 477 Musculoskeletal,
    Neural/Sensory,
    Reproductive
    HFIIZ70 SEQ ID NO: 478 Cancer
    HFIIZ70 SEQ ID NO: 479 Cancer
    HMIAJ30 SEQ ID NO: 480 Cancer
    HMIAJ30 SEQ ID NO: 481 Cancer
    HMIAJ30 SEQ ID NO: 482 Cancer
    HMIAJ30 SEQ ID NO: 483 Cancer
    HMIAV73 SEQ ID NO: 484 Cancer
    HMIAV73 SEQ ID NO: 485 Cancer
    HMIAV73 SEQ ID NO: 486 Cancer
    HMIAV73 SEQ ID NO: 487 Cancer
    HAPOD80 SEQ ID NO: 488 Cancer
    HISBL03 SEQ ID NO: 489 Cancer
    HISBL03 SEQ ID NO: 490 Cancer
    HISBL03 SEQ ID NO: 491 Cancer
    HISBL03 SEQ ID NO: 492 Cancer
    HISBL03 SEQ ID NO: 493 Cancer
    HISBL03 SEQ ID NO: 494 Cancer
    HISBL03 SEQ ID NO: 495 Cancer
    HMICK94 SEQ ID NO: 496 Cancer
    HMICK94 SEQ ID NO: 497 Cancer
    HMICK94 SEQ ID NO: 498 Cancer
    HISBF60 SEQ ID NO: 499 Cancer
    HISBF60 SEQ ID NO: 500 Cancer
    HISBF60 SEQ ID NO: 501 Cancer
    HISBF60 SEQ ID NO: 502 Cancer
    HISBF60 SEQ ID NO: 503 Cancer
    HMVAV54 SEQ ID NO: 504 Immune/Hematopoietic
    HMVAV54 SEQ ID NO: 505 Immune/Hematopoietic
    HMVAV54 SEQ ID NO: 506 Immune/Hematopoietic
    HMVAV54 SEQ ID NO: 507 Immune/Hematopoietic
    HMVAV54 SEQ ID NO: 508 Immune/Hematopoietic
    HPICB53 SEQ ID NO: 509 Cancer
    HPICB53 SEQ ID NO: 510 Cancer
    HPICC86 SEQ ID NO: 511 Reproductive
    HPICC86 SEQ ID NO: 512 Reproductive
    HPICC86 SEQ ID NO: 513 Reproductive
    HPICC86 SEQ ID NO: 514 Reproductive
    HPICC86 SEQ ID NO: 515 Reproductive
    HPJAP43 SEQ ID NO: 516 Cancer
    HPJAP43 SEQ ID NO: 517 Cancer
    HPJAP43 SEQ ID NO: 518 Cancer
    HPJCG42 SEQ ID NO: 519 Immune/Hematopoietic,
    Reproductive
    HPJCG42 SEQ ID NO: 520 Immune/Hematopoietic,
    Reproductive
    HPJCG42 SEQ ID NO: 521 Immune/Hematopoietic,
    Reproductive
    HPJCG42 SEQ ID NO: 522 Immune/Hematopoietic,
    Reproductive
    HPJCG42 SEQ ID NO: 523 Immune/Hematopoietic,
    Reproductive
    HPJBK11 SEQ ID NO: 524 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HPJBK11 SEQ ID NO: 525 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HPJBK11 SEQ ID NO: 526 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HPJBK12 SEQ ID NO: 527 Reproductive
    HPJBK12 SEQ ID NO: 528 Reproductive
    HPJBK12 SEQ ID NO: 529 Reproductive
    HPJBK12 SEQ ID NO: 530 Reproductive
    HPJCT08 SEQ ID NO: 531 Connective/Epithelial,
    Reproductive
    HPJCT08 SEQ ID NO: 532 Connective/Epithelial,
    Reproductive
    HPJCT08 SEQ ID NO: 533 Connective/Epithelial,
    Reproductive
    HT4ES80 SEQ ID NO: 534 Cancer
    HT4ES80 SEQ ID NO: 535 Cancer
    HT4ES80 SEQ ID NO: 536 Cancer
    HNTNB49 SEQ ID NO: 537 Cancer
    HNTNB49 SEQ ID NO: 538 Cancer
    HNTRS57 SEQ ID NO: 539 Cancer
    HNTRS57 SEQ ID NO: 540 Cancer
    HNTRS57 SEQ ID NO: 541 Cancer
    HNTRS57 SEQ ID NO: 542 Cancer
    HNTRS57 SEQ ID NO: 543 Cancer
    HNTSL47 SEQ ID NO: 544 Cardiovascular,
    Digestive
    HNTSL47 SEQ ID NO: 545 Cardiovascular,
    Digestive
    HNTSL47 SEQ ID NO: 546 Cardiovascular,
    Digestive
    HBJLR70 SEQ ID NO: 547 Immune/Hematopoietic,
    Neural/Sensory
    HBJLR70 SEQ ID NO: 548 Immune/Hematopoietic,
    Neural/Sensory
    HNTSY18 SEQ ID NO: 549 Cardiovascular,
    Reproductive
    HNTSY18 SEQ ID NO: 550 Cardiovascular,
    Reproductive
    HBHME51 SEQ ID NO: 551 Reproductive,
    Respiratory
    HBHME51 SEQ ID NO: 552 Reproductive,
    Respiratory
    HBHME51 SEQ ID NO: 553 Reproductive,
    Respiratory
    HMCHR48 SEQ ID NO: 554 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HMCHR48 SEQ ID NO: 555 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HMCHR48 SEQ ID NO: 556 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HMCIJ07 SEQ ID NO: 557 Immune/Hematopoietic
    HMCIJ07 SEQ ID NO: 558 Immune/Hematopoietic
    HSIFL06 SEQ ID NO: 559 Cancer
    HSIFL06 SEQ ID NO: 560 Cancer
    HMZME33 SEQ ID NO: 561 Connective/Epithelial,
    Digestive
    HMZME33 SEQ ID NO: 562 Connective/Epithelial,
    Digestive
    HMZMF54 SEQ ID NO: 563 Digestive
    HMZMF54 SEQ ID NO: 564 Digestive
    HMZMF54 SEQ ID NO: 565 Digestive
    HMVCQ82 SEQ ID NO: 566 Immune/Hematopoietic
    HMVCQ82 SEQ ID NO: 567 Immune/Hematopoietic
    HMVCQ82 SEQ ID NO: 568 Immune/Hematopoietic
    HMVDP35 SEQ ID NO: 569 Immune/Hematopoietic,
    Reproductive
    HMVDP35 SEQ ID NO: 570 Immune/Hematopoietic,
    Reproductive
    HMVDP35 SEQ ID NO: 571 Immune/Hematopoietic,
    Reproductive
    HMVDF54 SEQ ID NO: 572 Cancer
    HMVDF54 SEQ ID NO: 573 Cancer
    HMVDF54 SEQ ID NO: 574 Cancer
    HROBM46 SEQ ID NO: 575 Connective/Epithelial,
    Digestive
    HROBM46 SEQ ID NO: 576 Connective/Epithelial,
    Digestive
    HCNDR47 SEQ ID NO: 577 Cancer
    HCNDR47 SEQ ID NO: 578 Cancer
    HCNDR47 SEQ ID NO: 579 Cancer
    HCNDV12 SEQ ID NO: 580 Digestive,
    Reproductive
    HCNDV12 SEQ ID NO: 581 Digestive,
    Reproductive
    HCNDV12 SEQ ID NO: 582 Digestive,
    Reproductive
    HSODE04 SEQ ID NO: 583 Digestive
    HSODE04 SEQ ID NO: 584 Digestive
    HBFMC03 SEQ ID NO: 585 Digestive,
    Musculoskeletal,
    Reproductive
    HBFMC03 SEQ ID NO: 586 Digestive,
    Musculoskeletal,
    Reproductive
    HHSFB67 SEQ ID NO: 587 Neural/Sensory
    HHSFB67 SEQ ID NO: 588 Neural/Sensory
    HHSFB67 SEQ ID NO: 589 Neural/Sensory
    HHSFB67 SEQ ID NO: 590 Neural/Sensory
    HHSGW69 SEQ ID NO: 591 Cancer
    HHSGW69 SEQ ID NO: 592 Cancer
    HHSGW69 SEQ ID NO: 593 Cancer
    HCLCJ15 SEQ ID NO: 594 Cancer
    HCLCJ15 SEQ ID NO: 595 Cancer
    HCLCJ15 SEQ ID NO: 596 Cancer
    HCLCJ15 SEQ ID NO: 597 Cancer
    HSLJG37 SEQ ID NO: 598 Cancer
    HSLJG37 SEQ ID NO: 599 Cancer
    HSLJG37 SEQ ID NO: 600 Cancer
    HWLEC41 SEQ ID NO: 601 Cancer
    HWLEC41 SEQ ID NO: 602 Cancer
    HWLEC41 SEQ ID NO: 603 Cancer
    HSXEQ06 SEQ ID NO: 604 Cancer
    HSXEQ06 SEQ ID NO: 605 Cancer
    HSXEQ06 SEQ ID NO: 606 Cancer
    HEEAA16 SEQ ID NO: 607 Cancer
    HEEAA16 SEQ ID NO: 608 Cancer
    HEEAA16 SEQ ID NO: 609 Cancer
    HEEAM62 SEQ ID NO: 610 Reproductive
    HEEAM62 SEQ ID NO: 611 Reproductive
    HEEAM62 SEQ ID NO: 612 Reproductive
    HEEAM62 SEQ ID NO: 613 Reproductive
    HNHKL90 SEQ ID NO: 614 Immune/Hematopoietic
    HNHKL90 SEQ ID NO: 615 Immune/Hematopoietic
    HNHKL90 SEQ ID NO: 616 Immune/Hematopoietic
    HWLFQ64 SEQ ID NO: 617 Digestive
    HWLFQ64 SEQ ID NO: 618 Digestive
    HWLFR02 SEQ ID NO: 619 Cancer
    HWLFR02 SEQ ID NO: 620 Cancer
    HWLFR02 SEQ ID NO: 621 Cancer
    HBKED12 SEQ ID NO: 622 Cancer
    HBKED12 SEQ ID NO: 623 Cancer
    HBKED12 SEQ ID NO: 624 Cancer
    HBKED12 SEQ ID NO: 625 Cancer
    HBKED12 SEQ ID NO: 626 Cancer
    HWLFJ10 SEQ ID NO: 627 Cancer
    HWLFJ10 SEQ ID NO: 628 Cancer
    HCRNO87 SEQ ID NO: 629 Cancer
    HCRNO87 SEQ ID NO: 630 Cancer
    HCRNO87 SEQ ID NO: 631 Cancer
    HCRNO87 SEQ ID NO: 632 Cancer
    HWLJX42 SEQ ID NO: 633 Cancer
    HWLJX42 SEQ ID NO: 634 Cancer
    HWLJX42 SEQ ID NO: 635 Cancer
    HSPBY63 SEQ ID NO: 636 Digestive
    HSPBY63 SEQ ID NO: 637 Digestive
    HSPBY63 SEQ ID NO: 638 Digestive
    HAPSO15 SEQ ID NO: 639 Cancer
    HAPSO15 SEQ ID NO: 640 Cancer
    HAPSO15 SEQ ID NO: 641 Cancer
    HE8QG24 SEQ ID NO: 642 Mixed Fetal
    HE8QG24 SEQ ID NO: 643 Mixed Fetal
    HE8QG24 SEQ ID NO: 644 Mixed Fetal
    HE8QV43 SEQ ID NO: 645 Cancer
    HE8QV43 SEQ ID NO: 646 Cancer
    HE8QV43 SEQ ID NO: 647 Cancer
    HE8QV43 SEQ ID NO: 648 Cancer
    HE9QN39 SEQ ID NO: 649 Cancer
    HE9QN39 SEQ ID NO: 650 Cancer
    HE9RO44 SEQ ID NO: 651 Immune/Hematopoietic,
    Mixed Fetal
    HE9RO44 SEQ ID NO: 652 Immune/Hematopoietic,
    Mixed Fetal
    HE9RO44 SEQ ID NO: 653 Immune/Hematopoietic,
    Mixed Fetal
    HE9SE18 SEQ ID NO: 654 Digestive,
    Mixed Fetal
    HE9SE18 SEQ ID NO: 655 Digestive,
    Mixed Fetal
    HE9SE18 SEQ ID NO: 656 Digestive,
    Mixed Fetal
    HISCV60 SEQ ID NO: 657 Digestive
    HISCV60 SEQ ID NO: 658 Digestive
    HE8UT25 SEQ ID NO: 659 Mixed Fetal
    HE8UT25 SEQ ID NO: 660 Mixed Fetal
    HE8UT25 SEQ ID NO: 661 Mixed Fetal
    HE8UY36 SEQ ID NO: 662 Cancer
    HE8UY36 SEQ ID NO: 663 Cancer
    HNHNT13 SEQ ID NO: 664 Immune/Hematopoietic
    HNHNT13 SEQ ID NO: 665 Immune/Hematopoietic
    HNHNT13 SEQ ID NO: 666 Immune/Hematopoietic
    HODEB50 SEQ ID NO: 667 Reproductive
    HODEB50 SEQ ID NO: 668 Reproductive
    HODEB50 SEQ ID NO: 669 Reproductive
    HNGMJ91 SEQ ID NO: 670 Immune/Hematopoietic
    HNGMJ91 SEQ ID NO: 671 Immune/Hematopoietic
    HNGMJ91 SEQ ID NO: 672 Immune/Hematopoietic
    HNGNB69 SEQ ID NO: 673 Immune/Hematopoietic
    HODFW41 SEQ ID NO: 674 Reproductive
    HODFW41 SEQ ID NO: 675 Reproductive
    HNGOI12 SEQ ID NO: 676 Immune/Hematopoietic
    HNGOI12 SEQ ID NO: 677 Immune/Hematopoietic
    HNGOI12 SEQ ID NO: 678 Immune/Hematopoietic
    HNGPM78 SEQ ID NO: 679 Immune/Hematopoietic,
    Neural/Sensory
    HNGPM78 SEQ ID NO: 680 Immune/Hematopoietic,
    Neural/Sensory
    HYASC80 SEQ ID NO: 681 Cancer
    HYASC80 SEQ ID NO: 682 Cancer
    HWLHM66 SEQ ID NO: 683 Cancer
    HWLHM66 SEQ ID NO: 684 Cancer
    HWLHM66 SEQ ID NO: 685 Cancer
    HWLHM66 SEQ ID NO: 686 Cancer
    HBBBC71 SEQ ID NO: 687 Cancer
    HBBBC71 SEQ ID NO: 688 Cancer
    HBBBC71 SEQ ID NO: 689 Cancer
    HLJBF86 SEQ ID NO: 690 Cancer
    HLJBF86 SEQ ID NO: 691 Cancer
    HLJBF86 SEQ ID NO: 692 Cancer
    HLJBJ61 SEQ ID NO: 693 Cancer
    HLJBJ61 SEQ ID NO: 694 Cancer
    HHBCS39 SEQ ID NO: 695 Cancer
    HHBCS39 SEQ ID NO: 696 Cancer
    HHBCS39 SEQ ID NO: 697 Cancer
    HLJEA01 SEQ ID NO: 698 Respiratory
    HLJEA01 SEQ ID NO: 699 Respiratory
    HLEDB16 SEQ ID NO: 700 Cancer
    HOGCK63 SEQ ID NO: 701 Cancer
    HOGCK63 SEQ ID NO: 702 Cancer
    HOFMQ33 SEQ ID NO: 703 Reproductive
    HOFMQ33 SEQ ID NO: 704 Reproductive
    HOFMQ33 SEQ ID NO: 705 Reproductive
    HOFMQ33 SEQ ID NO: 706 Reproductive
    HOFMT75 SEQ ID NO: 707 Reproductive
    HOFMT75 SEQ ID NO: 708 Reproductive
    HOFMT75 SEQ ID NO: 709 Reproductive
    HOFMT75 SEQ ID NO: 710 Reproductive
    HOGCS52 SEQ ID NO: 711 Cancer
    HOGCS52 SEQ ID NO: 712 Cancer
    HOGCS52 SEQ ID NO: 713 Cancer
    HOFNM53 SEQ ID NO: 714 Reproductive
    HOFNM53 SEQ ID NO: 715 Reproductive
    HOFNM53 SEQ ID NO: 716 Reproductive
    HOFNM53 SEQ ID NO: 717 Reproductive
    HOFOB27 SEQ ID NO: 718 Cancer
    HOFOB27 SEQ ID NO: 719 Cancer
    HOFOB27 SEQ ID NO: 720 Cancer
    HOFOB27 SEQ ID NO: 721 Cancer
    HOFOC33 SEQ ID NO: 722 Reproductive
    HOFOC33 SEQ ID NO: 723 Reproductive
    HOFOC33 SEQ ID NO: 724 Reproductive
    HOFOC33 SEQ ID NO: 725 Reproductive
    HOFOC33 SEQ ID NO: 726 Reproductive
    HOFOC33 SEQ ID NO: 727 Reproductive
    HOFOC73 SEQ ID NO: 728 Cancer
    HOFOC73 SEQ ID NO: 729 Cancer
    HOFOC73 SEQ ID NO: 730 Cancer
    HOFOC73 SEQ ID NO: 731 Cancer
    HNTAC64 SEQ ID NO: 732 Cancer
    HNTAC64 SEQ ID NO: 733 Cancer
    HNTAC64 SEQ ID NO: 734 Cancer
    HNTAC64 SEQ ID NO: 735 Cancer
    HDTBD53 SEQ ID NO: 736 Cancer
    HDTBD53 SEQ ID NO: 737 Cancer
    HDTAQ57 SEQ ID NO: 738 Cancer
    HDTAQ57 SEQ ID NO: 739 Cancer
    HDTAR06 SEQ ID NO: 740 Cancer
    HDTAR06 SEQ ID NO: 741 Cancer
    HDPML23 SEQ ID NO: 742 Immune/Hematopoietic,
    Neural/Sensory
    HDPML23 SEQ ID NO: 743 Immune/Hematopoietic,
    Neural/Sensory
    HDPML23 SEQ ID NO: 744 Immune/Hematopoietic,
    Neural/Sensory
    HDPML23 SEQ ID NO: 745 Immune/Hematopoietic,
    Neural/Sensory
    HDPML23 SEQ ID NO: 746 Immune/Hematopoietic,
    Neural/Sensory
    HDPMM88 SEQ ID NO: 747 Cancer
    HDPMM88 SEQ ID NO: 748 Cancer
    HDPMM88 SEQ ID NO: 749 Cancer
    HDPMM88 SEQ ID NO: 750 Cancer
    HDPMM88 SEQ ID NO: 751 Cancer
    HDPMM88 SEQ ID NO: 752 Cancer
    HDPMM88 SEQ ID NO: 753 Cancer
    HDPMS12 SEQ ID NO: 754 Cancer
    HDPMS12 SEQ ID NO: 755 Cancer
    HDPMS12 SEQ ID NO: 756 Cancer
    HDPMS12 SEQ ID NO: 757 Cancer
    HDPMS12 SEQ ID NO: 758 Cancer
    HDPMS12 SEQ ID NO: 759 Cancer
    HDPAP35 SEQ ID NO: 760 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPAP35 SEQ ID NO: 761 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPAP35 SEQ ID NO: 762 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPAP35 SEQ ID NO: 763 Excretory,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPAQ55 SEQ ID NO: 764 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPAQ55 SEQ ID NO: 765 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPAQ55 SEQ ID NO: 766 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HDPAQ55 SEQ ID NO: 767 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HKAAV61 SEQ ID NO: 768 Connective/Epithelial
    HKAAV61 SEQ ID NO: 769 Connective/Epithelial
    HKAAV61 SEQ ID NO: 770 Connective/Epithelial
    HDPCJ43 SEQ ID NO: 771 Cancer
    HDPCJ43 SEQ ID NO: 772 Cancer
    HDPCJ43 SEQ ID NO: 773 Cancer
    HDPCJ43 SEQ ID NO: 774 Cancer
    HKACM93 SEQ ID NO: 775 Cancer
    HKACM93 SEQ ID NO: 776 Cancer
    HKACM93 SEQ ID NO: 777 Cancer
    HKACM93 SEQ ID NO: 778 Cancer
    HKAFT66 SEQ ID NO: 779 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HKAFT66 SEQ ID NO: 780 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HKAFT66 SEQ ID NO: 781 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HHEMM74 SEQ ID NO: 782 Cancer
    HHEMM74 SEQ ID NO: 783 Cancer
    HHEMM74 SEQ ID NO: 784 Cancer
    HHEMM74 SEQ ID NO: 785 Cancer
    HAMFC93 SEQ ID NO: 786 Cancer
    HAMFC93 SEQ ID NO: 787 Cancer
    HAMFC93 SEQ ID NO: 788 Cancer
    HSYAZ50 SEQ ID NO: 789 Cancer
    HSYAZ50 SEQ ID NO: 790 Cancer
    HSYAZ50 SEQ ID NO: 791 Cancer
    HSYAZ50 SEQ ID NO: 792 Cancer
    HLWAX42 SEQ ID NO: 793 Cancer
    HLWAX42 SEQ ID NO: 794 Cancer
    HLWAX42 SEQ ID NO: 795 Cancer
    HLWAZ70 SEQ ID NO: 796 Cancer
    HLWAZ70 SEQ ID NO: 797 Cancer
    HLWAZ70 SEQ ID NO: 798 Cancer
    HLWAZ70 SEQ ID NO: 799 Cancer
    HLWBG83 SEQ ID NO: 800 Cancer
    HLWBG83 SEQ ID NO: 801 Cancer
    HLWBG83 SEQ ID NO: 802 Cancer
    HLWBG83 SEQ ID NO: 803 Cancer
    HLWBG83 SEQ ID NO: 804 Cancer
    HLWBH18 SEQ ID NO: 805 Reproductive
    HLWBH18 SEQ ID NO: 806 Reproductive
    HRABS65 SEQ ID NO: 807 Cancer
    HRABV43 SEQ ID NO: 808 Cancer
    HRABV43 SEQ ID NO: 809 Cancer
    HRABV43 SEQ ID NO: 810 Cancer
    HHEPG23 SEQ ID NO: 811 Cancer
    HHEPG23 SEQ ID NO: 812 Cancer
    HHEPG23 SEQ ID NO: 813 Cancer
    HHEPJ23 SEQ ID NO: 814 Cancer
    HHEPJ23 SEQ ID NO: 815 Cancer
    HDPIW06 SEQ ID NO: 816 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPIW06 SEQ ID NO: 817 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPIW06 SEQ ID NO: 818 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPIW06 SEQ ID NO: 819 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPIW06 SEQ ID NO: 820 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HDPPA04 SEQ ID NO: 821 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    HDPPA04 SEQ ID NO: 822 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    HDPPA04 SEQ ID NO: 823 Cardiovascular,
    Connective/Epithelial,
    Immune/Hematopoietic
    HDPPN86 SEQ ID NO: 824 Cancer
    HDPPN86 SEQ ID NO: 825 Cancer
    HDTEK44 SEQ ID NO: 826 Connective/Epithelial,
    Immune/Hematopoietic,
    Rrproductive
    HDTEK44 SEQ ID NO: 827 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HDTEK44 SEQ ID NO: 828 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HDTEK44 SEQ ID NO: 829 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HOHBL42 SEQ ID NO: 830 Cancer
    HOHBL42 SEQ ID NO: 831 Cancer
    HOHBL42 SEQ ID NO: 832 Cancer
    HOHBL42 SEQ ID NO: 833 Cancer
    HOHBP82 SEQ ID NO: 834 Musculoskeletal
    HOHBP82 SEQ ID NO: 835 Musculoskeletal
    HOHBP82 SEQ ID NO: 836 Musculoskeletal
    HOHBP82 SEQ ID NO: 837 Musculoskeletal
    HOHBY44 SEQ ID NO: 838 Cancer
    HOHBY44 SEQ ID NO: 839 Cancer
    HOHBY44 SEQ ID NO: 840 Cancer
    HWBAD01 SEQ ID NO: 841 Immune/Hematopoietic
    HWBAD01 SEQ ID NO: 842 Immune/Hematopoietic
    HWBAD01 SEQ ID NO: 843 Immune/Hematopoietic
    HOHCJ90 SEQ ID NO: 844 Cancer
    HOHCJ90 SEQ ID NO: 845 Cancer
    HWABE12 SEQ ID NO: 846 Cancer
    HWABE12 SEQ ID NO: 847 Cancer
    HWABE12 SEQ ID NO: 848 Cancer
    HWBAR14 SEQ ID NO: 849 Cancer
    HWBAR14 SEQ ID NO: 850 Cancer
    HWBAR14 SEQ ID NO: 851 Cancer
    HWBAR14 SEQ ID NO: 852 Cancer
    HWBAR88 SEQ ID NO: 853 Cancer
    HWBCH13 SEQ ID NO: 854 Immune/Hematopoietic
    HWBCH13 SEQ ID NO: 855 Immune/Hematopoietic
    HWBCH13 SEQ ID NO: 856 Immune/Hematopoietic
    HWBCH13 SEQ ID NO: 857 Immune/Hematopoietic
    HWBCM79 SEQ ID NO: 858 Immune/Hematopoietic
    HWBCV72 SEQ ID NO: 859 Cancer
    HWBCV72 SEQ ID NO: 860 Cancer
    HWBCV72 SEQ ID NO: 861 Cancer
    HWBCV72 SEQ ID NO: 862 Cancer
    HWBDM62 SEQ ID NO: 863 Endocrine,
    Immune/Hematopoietic
    HWBDM62 SEQ ID NO: 864 Endocrine,
    Immune/Hematopoietic
    HWBDM62 SEQ ID NO: 865 Endocrine,
    Immune/Hematopoietic
    HWBDM62 SEQ ID NO: 866 Endocrine,
    Immune/Hematopoietic
    HMTAL77 SEQ ID NO: 867 Cancer
    HMTAL77 SEQ ID NO: 868 Cancer
    HDPRH52 SEQ ID NO: 869 Cancer
    HDPRH52 SEQ ID NO: 870 Cancer
    HDPSB18 SEQ ID NO: 871 Cancer
    HDPSB18 SEQ ID NO: 872 Cancer
    HDPSB18 SEQ ID NO: 873 Cancer
    HDPSB18 SEQ ID NO: 874 Cancer
    HDPSH53 SEQ ID NO: 875 Immune/Hematopoietic,
    Reproductive
    HDPSH53 SEQ ID NO: 876 Immune/Hematopoietic,
    Reproductive
    HDPLO25 SEQ ID NO: 877 Cancer
    HDPLO25 SEQ ID NO: 878 Cancer
    HDPLO25 SEQ ID NO: 879 Cancer
    HDPRN70 SEQ ID NO: 880 Immune/Hematopoietic
    HDPRN70 SEQ ID NO: 881 Immune/Hematopoietic
    HDPTW24 SEQ ID NO: 882 Immune/Hematopoietic
    HDPTW65 SEQ ID NO: 883 Excretory
    HDPTW65 SEQ ID NO: 884 Excretory
    HDPTW65 SEQ ID NO: 885 Excretory
    HDPWN93 SEQ ID NO: 886 Cancer
    HDPWN93 SEQ ID NO: 887 Cancer
    HDPWN93 SEQ ID NO: 888 Cancer
    HDPXY01 SEQ ID NO: 889 Cancer
    HDPXY01 SEQ ID NO: 890 Cancer
    HDPXY01 SEQ ID NO: 891 Cancer
    HDPXY01 SEQ ID NO: 892 Cancer
    HWHPM16 SEQ ID NO: 893 Cancer
    HWHPM16 SEQ ID NO: 894 Cancer
    HLDQA07 SEQ ID NO: 895 Digestive
    HLDQA07 SEQ ID NO: 896 Digestive
    HDTFE17 SEQ ID NO: 897 Cancer
    HDTFE17 SEQ ID NO: 898 Cancer
    HDTFE17 SEQ ID NO: 899 Cancer
    HWDAD17 SEQ ID NO: 900 Cancer
    HWDAD17 SEQ ID NO: 901 Cancer
    HWEAC77 SEQ ID NO: 902 Connective/Epithelial
    HWEAC77 SEQ ID NO: 903 Connective/Epithelial
    HWBEM18 SEQ ID NO: 904 Cancer
    HWBEM18 SEQ ID NO: 905 Cancer
    HWBEM18 SEQ ID NO: 906 Cancer
    HWBFE57 SEQ ID NO: 907 Cancer
    HWBFE57 SEQ ID NO: 908 Cancer
    HWBFE57 SEQ ID NO: 909 Cancer
    HOHDF66 SEQ ID NO: 910 Musculoskeletal
    HOHDF66 SEQ ID NO: 911 Musculoskeletal
    HOHDF66 SEQ ID NO: 912 Musculoskeletal
    HOHDC86 SEQ ID NO: 913 Musculoskeletal
    HOHDC86 SEQ ID NO: 914 Musculoskeletal
    HOHDC86 SEQ ID NO: 915 Musculoskeletal
    HRADO01 SEQ ID NO: 916 Excretory
    HRADO01 SEQ ID NO: 917 Excretory
    HRADO01 SEQ ID NO: 918 Excretory
    HRAEE45 SEQ ID NO: 919 Connective/Epithelial,
    Excretory,
    Immune/Hematopoietic
    HRAEE45 SEQ ID NO: 920 Connective/Epithelial,
    Excretory,
    Immune/Hematopoietic
    HRAEE45 SEQ ID NO: 921 Connective/Epithelial,
    Excretory,
    Immune/Hematopoietic
    HRAEH37 SEQ ID NO: 922 Cancer
    HRAEH37 SEQ ID NO: 923 Cancer
    HRAEH37 SEQ ID NO: 924 Cancer
    HWDAH38 SEQ ID NO: 925 Cancer
    HWDAH38 SEQ ID NO: 926 Cancer
    HLWCP78 SEQ ID NO: 927 Cancer
    HLWCP78 SEQ ID NO: 928 Cancer
    HLWCP78 SEQ ID NO: 929 Cancer
    HLWCP78 SEQ ID NO: 930 Cancer
    HTJML75 SEQ ID NO: 931 Cancer
    HTJML75 SEQ ID NO: 932 Cancer
    HTJNX29 SEQ ID NO: 933 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HTJNX29 SEQ ID NO: 934 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HTJNX29 SEQ ID NO: 935 Connective/Epithelial,
    Digestive,
    Immune/Hematopoietic
    HHESQ62 SEQ ID NO: 936 Immune/Hematopoietic
    HHESQ62 SEQ ID NO: 937 Immune/Hematopoietic
    HHESQ62 SEQ ID NO: 938 Immune/Hematopoietic
    HHESQ62 SEQ ID NO: 939 Immune/Hematopoietic
    HHESQ62 SEQ ID NO: 940 Immune/Hematopoietic
    HDQGO29 SEQ ID NO: 941 Immune/Hematopoietic
    HDQGO29 SEQ ID NO: 942 Immune/Hematopoietic
    HDQGO29 SEQ ID NO: 943 Immune/Hematopoietic
    HDQGO29 SEQ ID NO: 944 Immune/Hematopoietic
    HDQHY04 SEQ ID NO: 945 Cancer
    HDQHY04 SEQ ID NO: 946 Cancer
    HDQHY04 SEQ ID NO: 947 Cancer
    HBXAB02 SEQ ID NO: 948 Cancer
    HBXAB02 SEQ ID NO: 949 Cancer
    HBXAB02 SEQ ID NO: 950 Cancer
    HCWAU23 SEQ ID NO: 951 Immune/Hematopoietic
    HCWAU23 SEQ ID NO: 952 Immune/Hematopoietic
    HCWAU23 SEQ ID NO: 953 Immune/Hematopoietic
    HBXAM53 SEQ ID NO: 954 Cancer
    HBXAM53 SEQ ID NO: 955 Cancer
    HBXAM53 SEQ ID NO: 956 Cancer
    HCWBP34 SEQ ID NO: 957 Immune/Hematopoietic
    HCWBP34 SEQ ID NO: 958 Immune/Hematopoietic
    HCWBP34 SEQ ID NO: 959 Immune/Hematopoietic
    HBXCT44 SEQ ID NO: 960 Cancer
    HBXCT44 SEQ ID NO: 961 Cancer
    HBXCT44 SEQ ID NO: 962 Cancer
    HBXCT44 SEQ ID NO: 963 Cancer
    HCWDY64 SEQ ID NO: 964 Excretory,
    Immune/Hematopoietic
    HCWDY64 SEQ ID NO: 965 Excretory,
    Immune/Hematopoietic
    HCWDY64 SEQ ID NO: 966 Excretory,
    Immune/Hematopoietic
    HCWEB58 SEQ ID NO: 967 Cancer
    HCWEB58 SEQ ID NO: 968 Cancer
    HBXED80 SEQ ID NO: 969 Immune/Hematopoietic,
    Neural/Sensory
    HBXED80 SEQ ID NO: 970 Immune/Hematopoietic,
    Neural/Sensory
    HBXED80 SEQ ID NO: 971 Immune/Hematopoietic,
    Neural/Sensory
    HBXED80 SEQ ID NO: 972 Immune/Hematopoietic,
    Neural/Sensory
    HCWFT79 SEQ ID NO: 973 Immune/Hematopoietic
    HCWFT79 SEQ ID NO: 974 Immune/Hematopoietic
    HCWFT79 SEQ ID NO: 975 Immune/Hematopoietic
    HCWFU77 SEQ ID NO: 976 Cancer
    HCWFU77 SEQ ID NO: 977 Cancer
    HCWFU77 SEQ ID NO: 978 Cancer
    HBXFZ38 SEQ ID NO: 979 Cancer
    HBXFZ38 SEQ ID NO: 980 Cancer
    HBXFZ38 SEQ ID NO: 981 Cancer
    HCUGC55 SEQ ID NO: 982 Immune/Hematopoietic
    HCUGC55 SEQ ID NO: 983 Immune/Hematopoietic
    HCUGC55 SEQ ID NO: 984 Immune/Hematopoietic
    HCWGU37 SEQ ID NO: 985 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCWGU37 SEQ ID NO: 986 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HCWHV88 SEQ ID NO: 987 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HCWHV88 SEQ ID NO: 988 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HCWHX82 SEQ ID NO: 989 Immune/Hematopoietic,
    Neural/Sensory
    HCWHX82 SEQ ID NO: 990 Immune/Hematopoietic,
    Neural/Sensory
    HCWHX82 SEQ ID NO: 991 Immune/Hematopoietic,
    Neural/Sensory
    HCWFZ59 SEQ ID NO: 992 Immune/Hematopoietic
    HCWFZ59 SEQ ID NO: 993 Immune/Hematopoietic
    HCWFZ59 SEQ ID NO: 994 Immune/Hematopoietic
    HCWFZ59 SEQ ID NO: 995 Immune/Hematopoietic
    HBWCB95 SEQ ID NO: 996 Neural/Sensory
    HBWCB95 SEQ ID NO: 997 Neural/Sensory
    HBWCB95 SEQ ID NO: 998 Neural/Sensory
    HBWBR94 SEQ ID NO: 999 Neural/Sensory
    HBWBR94 SEQ ID NO: 1000 Neural/Sensory
    HBWBR94 SEQ ID NO: 1001 Neural/Sensory
    HBWCF75 SEQ ID NO: 1002 Neural/Sensory
    HBWCF75 SEQ ID NO: 1003 Neural/Sensory
    HBWCF75 SEQ ID NO: 1004 Neural/Sensory
    HBWCM83 SEQ ID NO: 1005 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HBWCM83 SEQ ID NO: 1006 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HBWCM83 SEQ ID NO: 1007 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HBWCM83 SEQ ID NO: 1008 Digestive,
    Immune/Hematopoietic,
    Neural/Sensory
    HRSMQ86 SEQ ID NO: 1009 Cancer
    HRSMQ86 SEQ ID NO: 1010 Cancer
    HFCAA91 SEQ ID NO: 1011 Neural/Sensory
    HFCAA91 SEQ ID NO: 1012 Neural/Sensory
    HFCAA91 SEQ ID NO: 1013 Neural/Sensory
    HFCAL39 SEQ ID NO: 1014 Cancer
    HFCAL39 SEQ ID NO: 1015 Cancer
    HFCAL39 SEQ ID NO: 1016 Cancer
    HCEBN44 SEQ ID NO: 1017 Neural/Sensory
    HCEBN44 SEQ ID NO: 1018 Neural/Sensory
    HHFCP32 SEQ ID NO: 1019 Cancer
    HGBAJ60 SEQ ID NO: 1020 Cancer
    HGBAJ60 SEQ ID NO: 1021 Cancer
    HHFCW75 SEQ ID NO: 1022 Cardiovascular
    HHFCW75 SEQ ID NO: 1023 Cardiovascular
    HHFCW75 SEQ ID NO: 1024 Cardiovascular
    HHFCZ67 SEQ ID NO: 1025 Cancer
    HHFCZ67 SEQ ID NO: 1026 Cancer
    HHFCZ67 SEQ ID NO: 1027 Cancer
    HHFCZ67 SEQ ID NO: 1028 Cancer
    HJBAR01 SEQ ID NO: 1029 Cancer
    HJBAR01 SEQ ID NO: 1030 Cancer
    HETAR42 SEQ ID NO: 1031 Cancer
    HETAR42 SEQ ID NO: 1032 Cancer
    HETAR42 SEQ ID NO: 1033 Cancer
    HETAR42 SEQ ID NO: 1034 Cancer
    HETAM53 SEQ ID NO: 1035 Cancer
    HETAM53 SEQ ID NO: 1036 Cancer
    HETAM53 SEQ ID NO: 1037 Cancer
    HETAM53 SEQ ID NO: 1038 Cancer
    HETAM53 SEQ ID NO: 1039 Cancer
    HTPBG16 SEQ ID NO: 1040 Digestive,
    Immune/Hematopoietic
    HTPBG16 SEQ ID NO: 1041 Digestive,
    Immune/Hematopoietic
    HTPBG16 SEQ ID NO: 1042 Digestive,
    Immune/Hematopoietic
    HJAAJ58 SEQ ID NO: 1043 Immune/Hematopoietic
    HJAAJ58 SEQ ID NO: 1044 Immune/Hematopoietic
    HJAAJ58 SEQ ID NO: 1045 Immune/Hematopoietic
    HJAAJ58 SEQ ID NO: 1046 Immune/Hematopoietic
    HSBBT12 SEQ ID NO: 1047 Cancer
    HSBBT12 SEQ ID NO: 1048 Cancer
    HSBBTI2 SEQ ID NO: 1049 Cancer
    HE8MH77 SEQ ID NO: 1050 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HE8MH77 SEQ ID NO: 1051 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HE8MH77 SEQ ID NO: 1052 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HTEDJ85 SEQ ID NO: 1053 Cancer
    HTEDJ85 SEQ ID NO: 1054 Cancer
    HTEDJ85 SEQ ID NO: 1055 Cancer
    HTEDJ85 SEQ ID NO: 1056 Cancer
    HOVAF78 SEQ ID NO: 1057 Cancer
    HOVAF78 SEQ ID NO: 1058 Cancer
    HOVAF78 SEQ ID NO: 1059 Cancer
    HOVAF78 SEQ ID NO: 1060 Cancer
    HOVAF78 SEQ ID NO: 1061 Cancer
    HHGDE24 SEQ ID NO: 1062 Cancer
    HHGDE24 SEQ ID NO: 1063 Cancer
    HHGDE24 SEQ ID NO: 1064 Cancer
    HOUFU35 SEQ ID NO: 1065 Connective/Epithelial
    HOUFU35 SEQ ID NO: 1066 Connective/Epithelial
    HOUFU35 SEQ ID NO: 1067 Connective/Epithelial
    HOUFU35 SEQ ID NO: 1068 Connective/Epithelial
    HSIGD79 SEQ ID NO: 1069 Cancer
    HSIGD79 SEQ ID NO: 1070 Cancer
    HCQCT05 SEQ ID NO: 1071 Digestive,
    Endocrine,
    Reproductive
    HCQCT05 SEQ ID NO: 1072 Digestive,
    Endocrine,
    Reproductive
    HMVDL30 SEQ ID NO: 1073 Cancer
    HMVDL30 SEQ ID NO: 1074 Cancer
    HMVDL30 SEQ ID NO: 1075 Cancer
    HMVDL30 SEQ ID NO: 1076 Cancer
    HTGGO35 SEQ ID NO: 1077 Cancer
    HTGGO35 SEQ ID NO: 1078 Cancer
    HTGGO35 SEQ ID NO: 1079 Cancer
    HCLBW50 SEQ ID NO: 1080 Cancer
    HCLBW50 SEQ ID NO: 1081 Cancer
    HCLBW50 SEQ ID NO: 1082 Cancer
    HCLBW50 SEQ ID NO: 1083 Cancer
    HWLEV32 SEQ ID NO: 1084 Cancer
    HWLEV32 SEQ ID NO: 1085 Cancer
    HWLEV32 SEQ ID NO: 1086 Cancer
    HWLEV32 SEQ ID NO: 1087 Cancer
    HWLFE89 SEQ ID NO: 1088 Cancer
    HWLFE89 SEQ ID NO: 1089 Cancer
    HWLFE89 SEQ ID NO: 1090 Cancer
    HE8PW38 SEQ ID NO: 1091 Neural/Sensory
    HE8PW38 SEQ ID NO: 1092 Neural/Sensory
    HE8PW38 SEQ ID NO: 1093 Neural/Sensory
    HE9RO27 SEQ ID NO: 1094 Connective/Epithelial,
    Mixed Fetal
    HE9RO27 SEQ ID NO: 1095 Connective/Epithelial,
    Mixed Fetal
    HE9RO27 SEQ ID NO: 1096 Connective/Epithelial,
    Mixed Fetal
    HCRPV17 SEQ ID NO: 1097 Cancer
    HCRPV17 SEQ ID NO: 1098 Cancer
    HCRPV17 SEQ ID NO: 1099 Cancer
    HCRPV17 SEQ ID NO: 1100 Cancer
    HHBGF77 SEQ ID NO: 1101 Cancer
    HHBGF77 SEQ ID NO: 1102 Cancer
    HLUDB47 SEQ ID NO: 1103 Cancer
    HLUDB47 SEQ ID NO: 1104 Cancer
    HLUDB47 SEQ ID NO: 1105 Cancer
    HHENZ16 SEQ ID NO: 1106 Cancer
    HHENZ16 SEQ ID NO: 1107 Cancer
    HHENZ16 SEQ ID NO: 1108 Cancer
    HSYBZ44 SEQ ID NO: 1109 Cancer
    HARNB17 SEQ ID NO: 1110 Cancer
    HARNB17 SEQ ID NO: 1111 Cancer
    HARNB17 SEQ ID NO: 1112 Cancer
    HARNB92 SEQ ID NO: 1113 Cancer
    HARNB92 SEQ ID NO: 1114 Cancer
    HARNB92 SEQ ID NO: 1115 Cancer
    HAMGV47 SEQ ID NO: 1116 Cancer
    HAMGV47 SEQ ID NO: 1117 Cancer
    HAMGV47 SEQ ID NO: 1118 Cancer
    HDTMK50 SEQ ID NO: 1119 Cancer
    HDTMK50 SEQ ID NO: 1120 Cancer
    HDTMK50 SEQ ID NO: 1121 Cancer
    HARBA09 SEQ ID NO: 1122 Cancer
    HARBA09 SEQ ID NO: 1123 Cancer
    HARBA09 SEQ ID NO: 1124 Cancer
    HARBA09 SEQ ID NO: 1125 Cancer
    HE8OK73 SEQ ID NO: 1126 Mixed Fetal,
    Neural/Sensory
    HE8OK73 SEQ ID NO: 1127 Mixed Fetal,
    Neural/Sensory
    HE8OK73 SEQ ID NO: 1128 Mixed Fetal,
    Neural/Sensory
    HSDJL42 SEQ ID NO: 1129 Cancer
    HSDJL42 SEQ ID NO: 1130 Cancer
    HSDJL42 SEQ ID NO: 1131 Cancer
    HCE2P86 SEQ ID NO: 1132 Cancer
    HCE2P86 SEQ ID NO: 1133 Cancer
    HCE2P86 SEQ ID NO: 1134 Cancer
    HNGNN78 SEQ ID NO: 1135 Cancer
    HNGNN78 SEQ ID NO: 1136 Cancer
    HNGNN78 SEQ ID NO: 1137 Cancer
    HTLHC59 SEQ ID NO: 1138 Digestive,
    Reproductive
    HTLHC59 SEQ ID NO: 1139 Digestive,
    Reproductive
    HTLJF15 SEQ ID NO: 1140 Immune/Hematopoietic,
    Reproductive
    HTLJF15 SEQ ID NO: 1141 Immune/Hematopoietic,
    Reproductive
    HTLJF15 SEQ ID NO: 1142 Immune/Hematopoietic,
    Reproductive
    HPJCC05 SEQ ID NO: 1143 Reproductive
    HPJCC05 SEQ ID NO: 1144 Reproductive
    HPJCC05 SEQ ID NO: 1145 Reproductive
    HDPVW11 SEQ ID NO: 1146 Cancer
    HDPVW11 SEQ ID NO: 1147 Cancer
    HDPWP69 SEQ ID NO: 1148 Cancer
    HDPWP69 SEQ ID NO: 1149 Cancer
    HDPWP69 SEQ ID NO: 1150 Cancer
    HWHHD11 SEQ ID NO: 1151 Cancer
    HWHHD11 SEQ ID NO: 1152 Cancer
    HWHHD11 SEQ ID NO: 1153 Cancer
    HBIMT93 SEQ ID NO: 1154 Cancer
    HBIMT93 SEQ ID NO: 1155 Cancer
    HBIMT93 SEQ ID NO: 1156 Cancer
    HHATA33 SEQ ID NO: 1157 Cancer
    HHATA33 SEQ ID NO: 1158 Cancer
    HNTDL21 SEQ ID NO: 1159 Cancer
    HNTDL21 SEQ ID NO: 1160 Cancer
    HNTNK95 SEQ ID NO: 1161 Cancer
    HNTNK95 SEQ ID NO: 1162 Cancer
    HNTNK95 SEQ ID NO: 1163 Cancer
    HWEAD64 SEQ ID NO: 1164 Cancer
    HWEAD64 SEQ ID NO: 1165 Cancer
    HWLHZ28 SEQ ID NO: 1166 Cancer
    HWLHZ28 SEQ ID NO: 1167 Cancer
    HWLHZ28 SEQ ID NO: 1168 Cancer
    HWLHZ28 SEQ ID NO: 1169 Cancer
    HWLJE21 SEQ ID NO: 1170 Cancer
    HWLJE21 SEQ ID NO: 1171 Cancer
    HWLJE21 SEQ ID NO: 1172 Cancer
    HPASD51 SEQ ID NO: 1173 Digestive,
    Excretory,
    Reproductive
    HPASD51 SEQ ID NO: 1174 Digestive,
    Excretory,
    Reproductive
    HSICQ15 SEQ ID NO: 1175 Cancer
    HSICQ15 SEQ ID NO: 1176 Cancer
    HFEBP27 SEQ ID NO: 1177 Cancer
    HFEBP27 SEQ ID NO: 1178 Cancer
    HFEBP27 SEQ ID NO: 1179 Cancer
    HTOIZ28 SEQ ID NO: 1180 Cancer
    HTOIZ28 SEQ ID NO: 1181 Cancer
    HTOIZ28 SEQ ID NO: 1182 Cancer
    HTOIZ28 SEQ ID NO: 1183 Cancer
    HCE4L28 SEQ ID NO: 1184 Cancer
    HCE4L28 SEQ ID NO: 1185 Cancer
    HCE4L28 SEQ ID NO: 1186 Cancer
    HCE4L28 SEQ ID NO: 1187 Cancer
    HFVGM16 SEQ ID NO: 1188 Cancer
    HFVGM16 SEQ ID NO: 1189 Cancer
    HPMGR66 SEQ ID NO: 1190 Cancer
    HPMGR66 SEQ ID NO: 1191 Cancer
    HLYDU43 SEQ ID NO: 1192 Cancer
    HLYDU43 SEQ ID NO: 1193 Cancer
    HPJCK10 SEQ ID NO: 1194 Cancer
    HPJCK10 SEQ ID NO: 1195 Cancer
    HT5EK75 SEQ ID NO: 1196 Cancer
    HT5EK75 SEQ ID NO: 1197 Cancer
    HT5EK75 SEQ ID NO: 1198 Cancer
    HWLEZ82 SEQ ID NO: 1199 Cancer
    HWLEZ82 SEQ ID NO: 1200 Cancer
    HWLEZ82 SEQ ID NO: 1201 Cancer
    HWLEZ82 SEQ ID NO: 1202 Cancer
    HDRMB11 SEQ ID NO: 1203 Digestive
    HDRMB11 SEQ ID NO: 1204 Digestive
    HDRMB11 SEQ ID NO: 1205 Digestive
    HCRNC80 SEQ ID NO: 1206 Cancer
    HCRNC80 SEQ ID NO: 1207 Cancer
    HCRNC80 SEQ ID NO: 1208 Cancer
    HCRNF14 SEQ ID NO: 1209 Cancer
    HCRNF14 SEQ ID NO: 1210 Cancer
    HCRNF14 SEQ ID NO: 1211 Cancer
    HE9PF45 SEQ ID NO: 1212 Cancer
    HE9PF45 SEQ ID NO: 1213 Cancer
    HE9PF45 SEQ ID NO: 1214 Cancer
    HISEN93 SEQ ID NO: 1215 Cancer
    HISEN93 SEQ ID NO: 1216 Cancer
    HISEN93 SEQ ID NO: 1217 Cancer
    HODEA51 SEQ ID NO: 1218 Cancer
    HODEA51 SEQ ID NO: 1219 Cancer
    HODEA51 SEQ ID NO: 1220 Cancer
    HODEA51 SEQ ID NO: 1221 Cancer
    HUSJN32 SEQ ID NO: 1222 Cancer
    HUSJN32 SEQ ID NO: 1223 Cancer
    HUSJN32 SEQ ID NO: 1224 Cancer
    HNGNW50 SEQ ID NO: 1225 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HNGNW50 SEQ ID NO: 1226 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HNGNW50 SEQ ID NO: 1227 Immune/Hematopoietic,
    Mixed Fetal,
    Reproductive
    HUVFB80 SEQ ID NO: 1228 Cancer
    HUVFB80 SEQ ID NO: 1229 Cancer
    HFIDQ92 SEQ ID NO: 1230 Cancer
    HFIDQ92 SEQ ID NO: 1231 Cancer
    HFIDQ92 SEQ ID NO: 1232 Cancer
    HTLJC07 SEQ ID NO: 1233 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTLJC07 SEQ ID NO: 1234 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTLJC07 SEQ ID NO: 1235 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HMSOW51 SEQ ID NO: 1236 Cancer
    HMSOW51 SEQ ID NO: 1237 Cancer
    HPJEZ38 SEQ ID NO: 1238 Cancer
    HPJEZ38 SEQ ID NO: 1239 Cancer
    HPJEZ38 SEQ ID NO: 1240 Cancer
    HTAGN51 SEQ ID NO: 1241 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HTAGN51 SEQ ID NO: 1242 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HHFLH45 SEQ ID NO: 1243 Cardiovascular,
    Reproductive
    HHFLH45 SEQ ID NO: 1244 Cardiovascular,
    Reproductive
    HHFLH45 SEQ ID NO: 1245 Cardiovascular,
    Reproductive
    HFKLE15 SEQ ID NO: 1246 Cancer
    HFKLE15 SEQ ID NO: 1247 Cancer
    HNSAA27 SEQ ID NO: 1248 Digestive
    HNSAA27 SEQ ID NO: 1249 Digestive
    HUVFY29 SEQ ID NO: 1250 Cancer
    HUVFY29 SEQ ID NO: 1251 Cancer
    HAVUR23 SEQ ID NO: 1252 Neural/Sensory
    HAVUR23 SEQ ID NO: 1253 Neural/Sensory
    HTPIH83 SEQ ID NO: 1254 Digestive,
    Reproductive
    HTPIH83 SEQ ID NO: 1255 Digestive,
    Reproductive
    HTPIH83 SEQ ID NO: 1256 Digestive,
    Reproductive
    HUCNC61 SEQ ID NO: 1257 Cancer
    HIDAF73 SEQ ID NO: 1258 Cancer
    HIDAF73 SEQ ID NO: 1259 Cancer
    HIDAF73 SEQ ID NO: 1260 Cancer
    HOFMA42 SEQ ID NO: 1261 Reproductive
    HOFMA42 SEQ ID NO: 1262 Reproductive
    HKABW11 SEQ ID NO: 1263 Cancer
    HKABW11 SEQ ID NO: 1264 Cancer
    HWBAO29 SEQ ID NO: 1265 Immune/Hematopoietic,
    Reproductive
    HWBAO29 SEQ ID NO: 1266 Immune/Hematopoietic,
    Reproductive
    HWBAO29 SEQ ID NO: 1267 Immune/Hematopoietic,
    Reproductive
    HDPTM61 SEQ ID NO: 1268 Digestive,
    Immune/Hematopoietic
    HDPTM61 SEQ ID NO: 1269 Digestive,
    Immune/Hematopoietic
    HKAHL26 SEQ ID NO: 1270 Cancer
    HKAHL26 SEQ ID NO: 1271 Cancer
    HDQHC29 SEQ ID NO: 1272 Cancer
    HDQHQ91 SEQ ID NO: 1273 Cancer
    HDQHQ91 SEQ ID NO: 1274 Cancer
    HDQHQ91 SEQ ID NO: 1275 Cancer
    HDTLR06 SEQ ID NO: 1276 Cancer
    HDTLR06 SEQ ID NO: 1277 Cancer
    HNTDE84 SEQ ID NO: 1278 Cancer
    HNTDE84 SEQ ID NO: 1279 Cancer
    HWAFT87 SEQ ID NO: 1280 Cardiovascular,
    Immune/Hematopoietic
    HWAFT87 SEQ ID NO: 1281 Cardiovascular,
    Immune/Hematopoietic
    HWAFT87 SEQ ID NO: 1282 Cardiovascular,
    Immune/Hematopoietic
    HOGCE48 SEQ ID NO: 1283 Cancer
    HOGCE48 SEQ ID NO: 1284 Cancer
    HBINS58 SEQ ID NO: 1285 Connective/Epithelial,
    Reproductive
    HBINS58 SEQ ID NO: 1286 Connective/Epithelial,
    Reproductive
    HHAUQ28 SEQ ID NO: 1287 Cancer
    HHAUQ28 SEQ ID NO: 1288 Cancer
    HBIOH81 SEQ ID NO: 1289 Cancer
    HBIOH81 SEQ ID NO: 1290 Cancer
    HOGDP46 SEQ ID NO: 1291 Cancer
    HOGDP46 SEQ ID NO: 1292 Cancer
    HWHIH10 SEQ ID NO: 1293 Cancer
    HWHIH10 SEQ ID NO: 1294 Cancer
    HCWCT62 SEQ ID NO: 1295 Immune/Hematopoietic
    HCWCT62 SEQ ID NO: 1296 Immune/Hematopoietic
    HCWCT62 SEQ ID NO: 1297 Immune/Hematopoietic
    HBXCL50 SEQ ID NO: 1298 Digestive,
    Excretory,
    Neural/Sensory
    HBXCL50 SEQ ID NO: 1299 Digestive,
    Excretory,
    Neural/Sensory
    HACAA29 SEQ ID NO: 1300 Cancer
    HACAA29 SEQ ID NO: 1301 Cancer
    HAJAR23 SEQ ID NO: 1302 Cancer
    HAJAR23 SEQ ID NO: 1303 Cancer
    HAJAR23 SEQ ID NO: 1304 Cancer
    HDPQN12 SEQ ID NO: 1305 Cancer
    HDPQN12 SEQ ID NO: 1306 Cancer
    HDQFN31 SEQ ID NO: 1307 Cancer
    HDQFN31 SEQ ID NO: 1308 Cancer
    HDQIH54 SEQ ID NO: 1309 Immune/Hematopoietic
    HDQIH54 SEQ ID NO: 1310 Immune/Hematopoietic
    HETKL27 SEQ ID NO: 1311 Cancer
    HETKL27 SEQ ID NO: 1312 Cancer
    HETKL27 SEQ ID NO: 1313 Cancer
    HETKL27 SEQ ID NO: 1314 Cancer
    HFIHQ89 SEQ ID NO: 1315 Cancer
    HFIHQ89 SEQ ID NO: 1316 Cancer
    HFKHW50 SEQ ID NO: 1317 Cancer
    HFKHW50 SEQ ID NO: 1318 Cancer
    HFKHW50 SEQ ID NO: 1319 Cancer
    HMEJL08 SEQ ID NO: 1320 Cancer
    HMEJL08 SEQ ID NO: 1321 Cancer
    HMEJL08 SEQ ID NO: 1322 Cancer
    HMSCT72 SEQ ID NO: 1323 Connective/Epithelial,
    Immune/Hematopoietic
    HMSCT72 SEQ ID NO: 1324 Connective/Epithelial,
    Immune/Hematopoietic
    HMSCT72 SEQ ID NO: 1325 Connective/Epithelial,
    Immune/Hematopoietic
    HPJEX20 SEQ ID NO: 1326 Immune/Hematopoietic,
    Reproductive
    HPJEX20 SEQ ID NO: 1327 Immune/Hematopoietic,
    Reproductive
    HPJEX20 SEQ ID NO: 1328 Immune/Hematopoietic,
    Reproductive
    HPJEX20 SEQ ID NO: 1329 Immune/Hematopoietic,
    Reproductive
    HSLGM21 SEQ ID NO: 1330 Cancer
    HSLGM21 SEQ ID NO: 1331 Cancer
    HSLHI86 SEQ ID NO: 1332 Cancer
    HSLHI86 SEQ ID NO: 1333 Cancer
    HSLHI86 SEQ ID NO: 1334 Cancer
    HSLHI86 SEQ ID NO: 1335 Cancer
    HUCNP80 SEQ ID NO: 1336 Cancer
    HUCNP80 SEQ ID NO: 1337 Cancer
    HBINK72 SEQ ID NO: 1338 Cancer
    HBINK72 SEQ ID NO: 1339 Cancer
    HBINK72 SEQ ID NO: 1340 Cancer
    HIABC55 SEQ ID NO: 1341 Cancer
    HIABC55 SEQ ID NO: 1342 Cancer
    HIABC55 SEQ ID NO: 1343 Cancer
    HIABC55 SEQ ID NO: 1344 Cancer
    HGBAR55 SEQ ID NO: 1345 Cancer
    HGBAR55 SEQ ID NO: 1346 Cancer
    HGBAR55 SEQ ID NO: 1347 Cancer
    HE2FE45 SEQ ID NO: 1348 Cancer
    HE2FE45 SEQ ID NO: 1349 Cancer
    HE2FE45 SEQ ID NO: 1350 Cancer
    HMRAD54 SEQ ID NO: 1351 Cancer
    HMRAD54 SEQ ID NO: 1352 Cancer
    HMRAD54 SEQ ID NO: 1353 Cancer
    HCEFB80 SEQ ID NO: 1354 Cancer
    HCEFB80 SEQ ID NO: 1355 Cancer
    HFTBN23 SEQ ID NO: 1356 Cancer
    HFTBN23 SEQ ID NO: 1357 Cancer
    HFTBN23 SEQ ID NO: 1358 Cancer
    HFTBQ52 SEQ ID NO: 1359 Cancer
    HFTBQ52 SEQ ID NO: 1360 Cancer
    HMEEJ79 SEQ ID NO: 1361 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HMEEJ79 SEQ ID NO: 1362 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HROAJ39 SEQ ID NO: 1363 Cancer
    HROAJ39 SEQ ID NO: 1364 Cancer
    HROAJ39 SEQ ID NO: 1365 Cancer
    HFEBV76 SEQ ID NO: 1366 Cancer
    HFEBV76 SEQ ID NO: 1367 Cancer
    HTADC09 SEQ ID NO: 1368 Cancer
    HTADC09 SEQ ID NO: 1369 Cancer
    HFXBJ12 SEQ ID NO: 1370 Neural/Sensory
    HFXBJ12 SEQ ID NO: 1371 Neural/Sensory
    HFXBJ12 SEQ ID NO: 1372 Neural/Sensory
    HMHBN86 SEQ ID NO: 1373 Cancer
    HMHBN86 SEQ ID NO: 1374 Cancer
    HMHBN86 SEQ ID NO: 1375 Cancer
    HFKFL92 SEQ ID NO: 1376 Cancer
    HFKFL92 SEQ ID NO: 1377 Cancer
    HFKFL92 SEQ ID NO: 1378 Cancer
    HASAW52 SEQ ID NO: 1379 Cancer
    HTLDT76 SEQ ID NO: 1380 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HTLDT76 SEQ ID NO: 1381 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HTLDT76 SEQ ID NO: 1382 Cardiovascular,
    Neural/Sensory,
    Reproductive
    HTLEC34 SEQ ID NO: 1383 Immune/Hematopoiefic,
    Neural/Sensory,
    Reproductive
    HTLEC34 SEQ ID NO: 1384 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HNHFB60 SEQ ID NO: 1385 Immune/Hemaropoietic
    HNHFB60 SEQ ID NO: 1386 Immune/Hematopoietic
    HNHFB60 SEQ ID NO: 1387 Immune/Hematopoietic
    H2CBK33 SEQ ID NO: 1388 Cancer
    H2CBK33 SEQ ID NO: 1389 Cancer
    H2CBK33 SEQ ID NO: 1390 Cancer
    HNGEY29 SEQ ID NO: 1391 Cancer
    HNGEY29 SEQ ID NO: 1392 Cancer
    HUSFE58 SEQ ID NO: 1393 Cancer
    HUSFE58 SEQ ID NO: 1394 Cancer
    HMSHS36 SEQ ID NO: 1395 Immune/Hematopoietic
    HMSHS36 SEQ ID NO: 1396 Immune/Hematopoietic
    HMSKC10 SEQ ID NO: 1397 Immune/Hematopoietic
    HMSKC10 SEQ ID NO: 1398 Immune/Hematopoietic
    HMSKC10 SEQ ID NO: 1399 Immune/Hematopoietic
    HSLGU75 SEQ ID NO: 1400 Cancer
    HSLGU75 SEQ ID NO: 1401 Cancer
    HSLGU75 SEQ ID NO: 1402 Cancer
    HDABU01 SEQ ID NO: 1403 Cancer
    HDABU01 SEQ ID NO: 1404 Cancer
    HDABU01 SEQ ID NO: 1405 Cancer
    HADGD17 SEQ ID NO: 1406 Connective/Epithelial
    HADGD17 SEQ ID NO: 1407 Connective/Epithelial
    HADGD17 SEQ ID NO: 1408 Connective/Epithelial
    HFIUE67 SEQ ID NO: 1409 Cancer
    HKGAM29 SEQ ID NO: 1410 Cancer
    HACBD86 SEQ ID NO: 1411 Cancer
    HACBD86 SEQ ID NO: 1412 Cancer
    HACBD86 SEQ ID NO: 1413 Cancer
    HEGAK23 SEQ ID NO: 1414 Cancer
    HEGAK23 SEQ ID NO: 1415 Cancer
    HEGAK23 SEQ ID NO: 1416 Cancer
    HEGAK23 SEQ ID NO: 1417 Cancer
    HCHAR90 SEQ ID NO: 1418 Cancer
    HCHAR90 SEQ ID NO: 1419 Cancer
    HCHAR90 SEQ ID NO: 1420 Cancer
    HLYCK27 SEQ ID NO: 1421 Immune/Hematopoietic
    HMVBP38 SEQ ID NO: 1422 Cancer
    HMVBP38 SEQ ID NO: 1423 Cancer
    HMVBP38 SEQ ID NO: 1424 Cancer
    HFACI31 SEQ ID NO: 1425 Neural/Sensory
    HFACI31 SEQ ID NO: 1426 Neural/Sensory
    HFACI31 SEQ ID NO: 1427 Neural/Sensory
    HBJKC04 SEQ ID NO: 1428 Immune/Hematopoietic
    HBJKC04 SEQ ID NO: 1429 Immune/Hematopoietic
    HBJKC04 SEQ ID NO: 1430 Immune/Hematopoietic
    HBJIT60 SEQ ID NO: 1431 Immune/Hematopoietic
    HBJIT60 SEQ ID NO: 1432 Immune/Hematopoietic
    HBJIT60 SEQ ID NO: 1433 Immune/Hematopoietic
    HPJBK03 SEQ ID NO: 1434 Cancer
    HPJBK03 SEQ ID NO: 1435 Cancer
    HPJCL22 SEQ ID NO: 1436 Cancer
    HPJCL22 SEQ ID NO: 1437 Cancer
    HPJCL22 SEQ ID NO: 1438 Cancer
    HTWJB71 SEQ ID NO: 1439 Immune/Hematopoietic,
    Neural/Sensory
    HNTOE45 SEQ ID NO: 1440 Cancer
    HNTOE45 SEQ ID NO: 1441 Cancer
    HNTRW30 SEQ ID NO: 1442 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HNTRW30 SEQ ID NO: 1443 Digestive,
    Immune/Hematopoietic,
    Mixed Fetal
    HCHPU32 SEQ ID NO: 1444 Cancer
    HCHPU32 SEQ ID NO: 1445 Cancer
    HCHPU32 SEQ ID NO: 1446 Cancer
    HGCNC48 SEQ ID NO: 1447 Reproductive
    HGCNC48 SEQ ID NO: 1448 Reproductive
    HLTHO84 SEQ ID NO: 1449 Cancer
    HSLIA81 SEQ ID NO: 1450 Cancer
    HSLIA81 SEQ ID NO: 1451 Cancer
    HSLIA81 SEQ ID NO: 1452 Cancer
    HSLIA81 SEQ ID NO: 1453 Cancer
    HBFMA07 SEQ ID NO: 1454 Cancer
    HBODE48 SEQ ID NO: 1455 Digestive,
    Excretory,
    Immune/Hematopoietic
    HBODE48 SEQ ID NO: 1456 Digestive,
    Excretory,
    Immune/Hematopoietic
    HBODE48 SEQ ID NO: 1457 Digestive,
    Excretory,
    Immune/Hematopoietic
    HBODE48 SEQ ID NO: 1458 Digestive,
    Excretory,
    Immune/Hematopoietic
    HCRME12 SEQ ID NO: 1459 Cancer
    HCRME12 SEQ ID NO: 1460 Cancer
    HBODQ16 SEQ ID NO: 1461 Cancer
    HBODQ16 SEQ ID NO: 1462 Cancer
    HASMB80 SEQ ID NO: 1463 Cancer
    HASMB80 SEQ ID NO: 1464 Cancer
    HBOEG11 SEQ ID NO: 1465 Cancer
    HBOEG11 SEQ ID NO: 1466 Cancer
    HCRNU76 SEQ ID NO: 1467 Cancer
    HCRNU76 SEQ ID NO: 1468 Cancer
    HAPSQ21 SEQ ID NO: 1469 Reproductive,
    Respiratory
    HAPSQ21 SEQ ID NO: 1470 Reproductive,
    Respiratory
    HAPSQ21 SEQ ID NO: 1471 Reproductive,
    Respiratory
    HWLNF33 SEQ ID NO: 1472 Cancer
    HWLNF33 SEQ ID NO: 1473 Cancer
    HE8QO53 SEQ ID NO: 1474 Cancer
    HE8QO53 SEQ ID NO: 1475 Cancer
    HE8QV67 SEQ ID NO: 1476 Cancer
    HE8QV67 SEQ ID NO: 1477 Cancer
    HE8TB68 SEQ ID NO: 1478 Cancer
    HE8TY90 SEQ ID NO: 1479 Cancer
    HE8TY90 SEQ ID NO: 1480 Cancer
    HE8TY90 SEQ ID NO: 1481 Cancer
    HE8TY90 SEQ ID NO: 1482 Cancer
    HETLM70 SEQ ID NO: 1483 Digestive,
    Excretory,
    Reproductive
    HETLM70 SEQ ID NO: 1484 Digestive,
    Excretory,
    Reproductive
    HETLM70 SEQ ID NO: 1485 Digestive,
    Excretory,
    Reproductive
    HISES66 SEQ ID NO: 1486 Digestive,
    Reproductive
    HISES66 SEQ ID NO: 1487 Digestive,
    Reproductive
    HISES66 SEQ ID NO: 1488 Digestive,
    Reproductive
    HTXKV29 SEQ ID NO: 1489 Cancer
    HTXKV29 SEQ ID NO: 1490 Cancer
    HTXKV29 SEQ ID NO: 1491 Cancer
    HTXLH48 SEQ ID NO: 1492 Immune/Hematopoietic
    HTXLH48 SEQ ID NO: 1493 Immune/Hematopoietic
    HTXLH48 SEQ ID NO: 1494 Immune/Hematopoietic
    HTEMD27 SEQ ID NO: 1495 Cancer
    HTEMD27 SEQ ID NO: 1496 Cancer
    HTEME02 SEQ ID NO: 149T Cancer
    HTEME02 SEQ ID NO: 1498 Cancer
    HTEME02 SEQ ID NO: 1499 Cancer
    HNHLD23 SEQ ID NO: 1500 Immune/Hematopoietic
    HETLT82 SEQ ID NO: 1501 Immune/Hematopoietic,
    Reproductive
    HETLT82 SEQ ID NO: 1502 Immune/Hematopoietic,
    Reproductive
    HETLT82 SEQ ID NO: 1503 Immune/Hematopoietic,
    Reproductive
    HNGLH60 SEQ ID NO: 1504 Immune/Hematopoietic,
    Musculoskeletal
    HNGLH60 SEQ ID NO: 1505 Immune/Hematopoietic,
    Musculoskeletal
    HNOLH60 SEQ ID NO: 1506 Immune/Hematopoietic,
    Musculoskeletal
    HNHPG05 SEQ ID NO: 1507 Immune/Hematopoietic
    HNHPG05 SEQ ID NO: 1508 Immune/Hematopoietic
    HNHPG05 SEQ ID NO: 1509 Immune/Hematopoietic
    HUSIY89 SEQ ID NO: 1510 Cardiovascular,
    Immune/Hematopoietic
    HUSIY89 SEQ ID NO: 1511 Cardiovascular,
    Immune/Hematopoietic
    HUSJM25 SEQ ID NO: 1512 Cancer
    HUSJM25 SEQ ID NO: 1513 Cancer
    HTXNL31 SEQ ID NO: 1514 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HTXNL31 SEQ ID NO: 1515 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HBGNQ12 SEQ ID NO: 1516 Cancer
    HBGNQ12 SEQ ID NO: 1517 Cancer
    HNGNS74 SEQ ID NO: 1518 Cancer
    HNGNS74 SEQ ID NO: 1519 Cancer
    HNGOD80 SEQ ID NO: 1520 Cancer
    HNGOD80 SEQ ID NO: 1521 Cancer
    HODHK19 SEQ ID NO: 1522 Reproductive
    HODHK19 SEQ ID NO: 1523 Reproductive
    HODHK19 SEQ ID NO: 1524 Reproductive
    HTLHR26 SEQ ID NO: 1525 Immune/Hematopoieric,
    Reproductive
    HTLHR26 SEQ ID NO: 1526 Immune/Hematopoietic,
    Reproductive
    HTLHR26 SEQ ID NO: 1527 Immune/Hematopoietic,
    Reproductive
    HUSZS75 SEQ ID NO: 1528 Cancer
    HUSZS75 SEQ ID NO: 1529 Cancer
    HLQDY81 SEQ ID NO: 1530 Cardiovascular,
    Digestive,
    Musculoskeletal
    HBGNU56 SEQ ID NO: 1531 Cancer
    HBGNU56 SEQ ID NO: 1532 Cancer
    HODGL52 SEQ ID NO: 1533 Cancer
    HODGL52 SEQ ID NO: 1534 Cancer
    HTXNV67 SEQ ID NO: 1535 Cancer
    HTXNV67 SEQ ID NO: 1536 Cancer
    HTXNV67 SEQ ID NO: 1537 Cancer
    HOCNE30 SEQ ID NO: 1538 Digestive,
    Musculoskeletal,
    Neural/Sensory
    HOCNE30 SEQ ID NO: 1539 Digestive,
    Musculoskeletal,
    Neural/Sensory
    HOCNE30 SEQ ID NO: 1540 Digestive,
    Musculoskeletal,
    Neural/Sensory
    HMSOC30 SEQ ID NO: 1541 Cancer
    HMSOC30 SEQ ID NO: 1542 Cancer
    HWMAF61 SEQ ID NO: 1543 Digestive
    HWMAF61 SEQ ID NO: 1544 Digestive
    HWMAF61 SEQ ID NO: 1545 Digestive
    HWMAF61 SEQ ID NO: 1546 Digestive
    HWMAF61 SEQ ID NO: 1547 Digestive
    HWMAH36 SEQ ID NO: 1548 Immune/Hematopoietic
    HWMAH36 SEQ ID NO: 1549 Immune/Hematopoietic
    HXOAC69 SEQ ID NO: 1550 Cancer
    HXOAC69 SEQ ID NO: 1551 Cancer
    HPJDA23 SEQ ID NO: 1552 Mixed Fetal,
    Neural/Sensory,
    Reproductive
    HPJDA23 SEQ ID NO: 1553 Mixed Fetal,
    Neural/Sensory,
    Reproductive
    HPJEE14 SEQ ID NO: 1554 Reproductive
    HPJEE14 SEQ ID NO: 1555 Reproductive
    HPJEG57 SEQ ID NO: 1556 Reproductive
    HPJEG57 SEQ ID NO: 1557 Reproductive
    HPJEG57 SEQ ID NO: 1558 Reproductive
    HPJEV11 SEQ ID NO: 1559 Cancer
    HTTKT43 SEQ ID NO: 1560 Cancer
    HTTKT43 SEQ ID NO: 1561 Cancer
    HTTKT43 SEQ ID NO: 1562 Cancer
    HHFKM76 SEQ ID NO: 1563 Cancer
    HHFKM76 SEQ ID NO: 1564 Cancer
    HHFKM76 SEQ ID NO: 1565 Cancer
    HHFML08 SEQ ID NO: 1566 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HHFML08 SEQ ID NO: 1567 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HHFML08 SEQ ID NO: 1568 Cardiovascular,
    Immune/Hematopoietic,
    Mixed Fetal
    HTPFX69 SEQ ID NO: 1569 Cancer
    HTPFX69 SEQ ID NO: 1570 Cancer
    HTPFX69 SEQ ID NO: 1571 Cancer
    HTPFX69 SEQ ID NO: 1572 Cancer
    HFKLX38 SEQ ID NO: 1573 Excretory,
    Respiratory
    HFKLX38 SEQ ID NO: 1574 Excretory,
    Respiratory
    HFKLX38 SEQ ID NO: 1575 Excretory,
    Respiratory
    HFKME15 SEQ ID NO: 1576 Excretory
    HFKME15 SEQ ID NO: 1577 Excretory
    HUVFH14 SEQ ID NO: 1578 Cancer
    HUVFH14 SEQ ID NO: 1579 Cancer
    HUVFH14 SEQ ID NO: 1580 Cancer
    HE2KK74 SEQ ID NO: 1581 Cancer
    HE2KK74 SEQ ID NO: 1582 Cancer
    HE2KK74 SEQ ID NO: 1583 Cancer
    HMALI42 SEQ ID NO: 1584 Immune/Hematopoietic
    HE2LW65 SEQ ID NO: 1585 Cancer
    HE2LW65 SEQ ID NO: 1586 Cancer
    HE2LW65 SEQ ID NO: 1587 Cancer
    HTFOS57 SEQ ID NO: 1588 Cancer
    HTFOS57 SEQ ID NO: 1589 Cancer
    HTFOS57 SEQ ID NO: 1590 Cancer
    HUVHI35 SEQ ID NO: 1591 Cancer
    HUVHI35 SEQ ID NO: 1592 Cancer
    HUVHI35 SEQ ID NO: 1593 Cancer
    HUVHI35 SEQ ID NO: 1594 Cancer
    HTPHS66 SEQ ID NO: 1595 Cancer
    HTPHS66 SEQ ID NO: 1596 Cancer
    HTPHS66 SEQ ID NO: 1597 Cancer
    HHFOJ29 SEQ ID NO: 1598 Cancer
    HHFOJ29 SEQ ID NO: 1599 Cancer
    HHFOJ29 SEQ ID NO: 1600 Cancer
    HMAMI15 SEQ ID NO: 1601 Cancer
    HTXQM57 SEQ ID NO: 1602 Immune/Hematopoietic,
    Mixed Fetal
    HE2RO22 SEQ ID NO: 1603 Mixed Fetal
    HE2RO22 SEQ ID NO: 1604 Mixed Fetal
    HE2SI26 SEQ ID NO: 1605 Cancer
    HTXRE15 SEQ ID NO: 1606 Cancer
    HTXRE15 SEQ ID NO: 1607 Cancer
    HUCPD31 SEQ ID NO: 1608 Cancer
    HUCPD31 SEQ ID NO: 1609 Cancer
    HFPHA80 SEQ ID NO: 1610 Neural/Sensory
    HFPHA80 SEQ ID NO: 1611 Neural/Sensory
    HFPHA80 SEQ ID NO: 1612 Neural/Sensory
    HFPHA80 SEQ ID NO: 1613 Neural/Sensory
    HFPHB92 SEQ ID NO: 1614 Excretory,
    Neural/Sensory
    HFPHS77 SEQ ID NO: 1615 Cancer
    HFPHS77 SEQ ID NO: 1616 Cancer
    HFPHS77 SEQ ID NO: 1617 Cancer
    HIPAJ43 SEQ ID NO: 1618 Cancer
    HIPAJ43 SEQ ID NO: 1619 Cancer
    HDDMW90 SEQ ID NO: 1620 Cancer
    HDDMW90 SEQ ID NO: 1621 Cancer
    HBCPB32 SEQ ID NO: 1622 Neural/Sensory,
    Reproductive
    HFVKC95 SEQ ID NO: 1623 Cancer
    HFVKC95 SEQ ID NO: 1624 Cancer
    HFVKC95 SEQ ID NO: 1625 Cancer
    HCOMM91 SEQ ID NO: 1626 Cancer
    HCOMM91 SEQ ID NO: 1627 Cancer
    HVVAM64 SEQ ID NO: 1628 Cancer
    HVVAM64 SEQ ID NO: 1629 Cancer
    HVVAM64 SEQ ID NO: 1630 Cancer
    HNBUC50 SEQ ID NO: 1631 Cancer
    HNBUC50 SEQ ID NO: 1632 Cancer
    HNBUC50 SEQ ID NO: 1633 Cancer
    HNBUC50 SEQ ID NO: 1634 Cancer
    HUUDF48 SEQ ID NO: 1635 Immune/Hematopoietic
    HUUDF48 SEQ ID NO: 1636 Immune/Hematopoietic
    HBCQL32 SEQ ID NO: 1637 Cancer
    HBCQL32 SEQ ID NO: 1638 Cancer
    HCBND16 SEQ ID NO: 1639 Cancer
    HCBND16 SEQ ID NO: 1640 Cancer
    HNNBM45 SEQ ID NO: 1641 Immune/Hematopoietic,
    Reproductive
    HNNBM45 SEQ ID NO: 1642 Immune/Hematopoietic,
    Reproductive
    HWMGN33 SEQ ID NO: 1643 Digestive
    HWMGN33 SEQ ID NO: 1644 Digestive
    HWMLN52 SEQ ID NO: 1645 Digestive,
    Immune/Hematopoietic
    HWMLN52 SEQ ID NO: 1646 Digestive,
    Immune/Hematopoietic
    HWMLN52 SEQ ID NO: 1647 Digestive,
    Immune/Hematopoietic
    HVARW53 SEQ ID NO: 1648 Digestive
    HVARW53 SEQ ID NO: 1649 Digestive
    HAHFU44 SEQ ID NO: 1650 Cardiovascular,
    Digestive,
    Musculoskeletal
    HAHFU44 SEQ ID NO: 1651 Cardiovascular,
    Digestive,
    Musculoskeletal
    HAHFU44 SEQ ID NO: 1652 Cardiovascular,
    Digestive,
    Musculoskeletal
    HCOOS80 SEQ ID NO: 1653 Cancer
    HCOOS80 SEQ ID NO: 1654 Cancer
    HCOOS80 SEQ ID NO: 1655 Cancer
    HNKCO80 SEQ ID NO: 1656 Cancer
    HNKCO80 SEQ ID NO: 1657 Cancer
    HLTIP27 SEQ ID NO: 1658 Immune/Hematopoietic
    HLTIP27 SEQ ID NO: 1659 Immune/Hematopoietic
    HLTIP94 SEQ ID NO: 1660 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HLTIP94 SEQ ID NO: 1661 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HLTIP94 SEQ ID NO: 1662 Immune/Hematopoietic,
    Mixed Fetal,
    Neural/Sensory
    HOCPM23 SEQ ID NO: 1663 Reproductive
    HOCPM23 SEQ ID NO: 1664 Reproductive
    HPDWP28 SEQ ID NO: 1665 Reproductive
    HPDWP28 SEQ ID NO: 1666 Reproductive
    HLCND09 SEQ ID NO: 1667 Cancer
    HLCND09 SEQ ID NO: 1668 Cancer
    HEEBI05 SEQ ID NO: 1669 Digestive,
    Reproductive
    HEEBB55 SEQ ID NO: 1670 Cancer
    HEEBB55 SEQ ID NO: 1671 Cancer
    HEEBB55 SEQ ID NO: 1672 Cancer
    HEGCL11 SEQ ID NO: 1673 Cancer
    HEGCL11 SEQ ID NO: 1674 Cancer
    HNTPB82 SEQ ID NO: 1675 Cancer
    HNTPB82 SEQ ID NO: 1676 Cancer
    HOFMM69 SEQ ID NO: 1677 Reproductive
    HOFMM69 SEQ ID NO: 1678 Reproductive
    HLDAB75 SEQ ID NO: 1679 Cancer
    HLDAB75 SEQ ID NO: 1680 Cancer
    HKACC80 SEQ ID NO: 1681 Cancer
    HKACC80 SEQ ID NO: 1682 Cancer
    HKACC80 SEQ ID NO: 1683 Cancer
    HKAEL28 SEQ ID NO: 1684 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HKAEL28 SEQ ID NO: 1685 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HDPGT25 SEQ ID NO: 1686 Cancer
    HDPGT25 SEQ ID NO: 1687 Cancer
    HLWBT09 SEQ ID NO: 1688 Excretory,
    Reproductive
    HLWBT09 SEQ ID NO: 1689 Excretory,
    Reproductive
    HHEDN80 SEQ ID NO: 1690 Cancer
    HHEDN80 SEQ ID NO: 1691 Cancer
    HHEDN80 SEQ ID NO: 1692 Cancer
    HDFQB14 SEQ ID NO: 1693 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HWAAW33 SEQ ID NO: 1694 Cardiovascular,
    Immune/Hematopoietic,
    Musculoskeletal
    HWAAW33 SEQ ID NO: 1695 Cardiovascular,
    Immune/Hematopoietic,
    Musculoskeletal
    HWABF47 SEQ ID NO: 1696 Cancer
    HWABF47 SEQ ID NO: 1697 Cancer
    HWABI12 SEQ ID NO: 1698 Immune/Hematopoietic
    HWABI12 SEQ ID NO: 1699 Immune/Hematopoietic
    HWBBT49 SEQ ID NO: 1700 Cancer
    HWBBT49 SEQ ID NO: 1701 Cancer
    HWBBT49 SEQ ID NO: 1702 Cancer
    HAMGG89 SEQ ID NO: 1703 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HAMGG89 SEQ ID NO: 1704 Immune/Hematopoietic,
    Neural/Sensory,
    Reproductive
    HAJBW16 SEQ ID NO: 1705 Neural/Sensory
    HAJBW16 SEQ ID NO: 1706 Neural/Sensory
    HNTAI35 SEQ ID NO: 1707 Cancer
    HNTAI35 SEQ ID NO: 1708 Cancer
    HNTAI35 SEQ ID NO: 1709 Cancer
    HNTAI35 SEQ ID NO: 1710 Cancer
    HNTAI35 SEQ ID NO: 1711 Cancer
    HNTBN41 SEQ ID NO: 1712 Immune/Hematopoietic
    HNTBN41 SEQ ID NO: 1713 Immune/Hematopoietic
    HNTBN41 SEQ ID NO: 1714 Immune/Hematopoietic
    HNTBN41 SEQ ID NO: 1715 Immune/Hematopoietic
    HDPRJ60 SEQ ID NO: 1716 Cancer
    HDPRJ60 SEQ ID NO: 1717 Cancer
    HDPRJ60 SEQ ID NO: 1718 Cancer
    HDPSB01 SEQ ID NO: 1719 Cancer
    HDPSB01 SEQ ID NO: 1720 Cancer
    HDPSB01 SEQ ID NO: 1721 Cancer
    HDPSB01 SEQ ID NO: 1722 Cancer
    HDPSB01 SEQ ID NO: 1723 Cancer
    HDPTC31 SEQ ID NO: 1724 Immune/Hematopoietic
    HDPTC31 SEQ ID NO: 1725 Immune/Hematopoietic
    HDPTC31 SEQ ID NO: 1726 Immune/Hematopoietic
    HDPXL05 SEQ ID NO: 1727 Immune/Hematopoietic,
    Reproductive
    HDPXL05 SEQ ID NO: 1728 Immune/Hematopoietic,
    Reproductive
    HDPXL05 SEQ ID NO: 1729 Immune/Hematopoietic,
    Reproductive
    HDPXY88 SEQ ID NO: 1730 Cancer
    HDPXY88 SEQ ID NO: 1731 Cancer
    HDPXY88 SEQ ID NO: 1732 Cancer
    HLDQZ72 SEQ ID NO: 1733 Cancer
    HLDQZ72 SEQ ID NO: 1734 Cancer
    HLDQZ72 SEQ ID NO: 1735 Cancer
    HWBEV57 SEQ ID NO: 1736 Immune/Hematopoietic
    HWBEV57 SEQ ID NO: 1737 Immune/Hematopoietic
    HWBEV57 SEQ ID NO: 1738 Immune/Hematopoietic
    HAMHH20 SEQ ID NO: 1739 Cancer
    HAMHH20 SEQ ID NO: 1740 Cancer
    HDLAY18 SEQ ID NO: 1741 Cancer
    HDLAY18 SEQ ID NO: 1742 Cancer
    HKAHN23 SEQ ID NO: 1743 Connective/Epithelial,
    Digestive,
    Mixed Fetal
    HKAHN23 SEQ ID NO: 1744 Connective/Epithelial,
    Digestive,
    Mixed Fetal
    HKAJW28 SEQ ID NO: 1745 Cancer
    HKAJW28 SEQ ID NO: 1746 Cancer
    HDQFU73 SEQ ID NO: 1747 Digestive,
    Immune/Hematopoietic
    HDQFU73 SEQ ID NO: 1748 Digestive,
    Immune/Hematopoietic
    HDQFU73 SEQ ID NO: 1749 Digestive,
    Immune/Hematopoietic
    HDTKS69 SEQ ID NO: 1750 Cancer
    HSYDT06 SEQ ID NO: 1751 Cancer
    HSYDT06 SEQ ID NO: 1752 Cancer
    HSYDT06 SEQ ID NO: 1753 Cancer
    HSYDT06 SEQ ID NO: 1754 Cancer
    HNTEF28 SEQ ID NO: 1755 Cancer
    HNTEF28 SEQ ID NO: 1756 Cancer
    HNTEF53 SEQ ID NO: 1757 Cancer
    HNTEF53 SEQ ID NO: 1758 Cancer
    HNTEF53 SEQ ID NO: 1759 Cancer
    HNTEF53 SEQ ID NO: 1760 Cancer
    HDQFN60 SEQ ID NO: 1761 Cancer
    HDQFN60 SEQ ID NO: 1762 Cancer
    HHEXM06 SEQ ID NO: 1763 Immune/Hematopoietic
    HHEXM06 SEQ ID NO: 1764 Immune/Hematopoietic
    HBINU36 SEQ ID NO: 1765 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HBINU36 SEQ ID NO: 1766 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HBINU36 SEQ ID NO: 1767 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HUJCQ39 SEQ ID NO: 1768 Cancer
    HUJCQ39 SEQ ID NO: 1769 Cancer
    HUJCQ39 SEQ ID NO: 1770 Cancer
    HCCCG83 SEQ ID NO: 1771 Cancer
    HCCCG83 SEQ ID NO: 1772 Cancer
    HCCCG83 SEQ ID NO: 1773 Cancer
    HWHIM26 SEQ ID NO: 1774 Connective/Epithelial,
    Immune/Hematopoietic
    HWHIM26 SEQ ID NO: 1775 Connective/Epithelial,
    Immune/Hematopoietic
    HWHKC09 SEQ ID NO: 1776 Cancer
    HWHKC09 SEQ ID NO: 1777 Cancer
    HWHKC09 SEQ ID NO: 1778 Cancer
    HWHKC09 SEQ ID NO: 1779 Cancer
    HWHKR51 SEQ ID NO: 1780 Cancer
    HWHKR51 SEQ ID NO: 1781 Cancer
    HWHKR51 SEQ ID NO: 1782 Cancer
    HWHRL06 SEQ ID NO: 1783 Cancer
    HWHRL06 SEQ ID NO: 1784 Cancer
    HAZAD32 SEQ ID NO: 1785 Cancer
    HAZAD32 SEQ ID NO: 1786 Cancer
    HPAMY60 SEQ ID NO: 1787 Excretory
    HPAMY60 SEQ ID NO: 1788 Excretory
    HAOTS04 SEQ ID NO: 1789 Reproductive
    HAOTS04 SEQ ID NO: 1790 Reproductive
    HAZAP37 SEQ ID NO: 1791 Reproductive
    HKZAS29 SEQ ID NO: 1792 Cancer
    HKZAS29 SEQ ID NO: 1793 Cancer
    HOVJP29 SEQ ID NO: 1794 Reproductive
    HOVJP29 SEQ ID NO: 1795 Reproductive
    HWHSB53 SEQ ID NO: 1796 Cancer
    HWHSB53 SEQ ID NO: 1797 Cancer
    HKZBS01 SEQ ID NO: 1798 Cancer
    HKZBS01 SEQ ID NO: 1799 Cancer
    HWHSO13 SEQ ID NO: 1800 Connective/Epithelial
    HWHSO13 SEQ ID NO: 1801 Connective/Epithelial
    HKZCK47 SEQ ID NO: 1802 Immune/Hematopoietic,
    Reproductive
    HCUHQ40 SEQ ID NO: 1803 Cancer
    HCUHQ40 SEQ ID NO: 1804 Cancer
    HCUHQ40 SEQ ID NO: 1805 Cancer
    HPJCP79 SEQ ID NO: 1806 Cancer
    HPJCP79 SEQ ID NO: 1807 Cancer
    HPJCP79 SEQ ID NO: 1808 Cancer
    HPJCP79 SEQ ID NO: 1809 Cancer
    HFXDI56 SEQ ID NO: 1810 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HFXDI56 SEQ ID NO: 1811 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HFXDI56 SEQ ID NO: 1812 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HFXDI56 SEQ ID NO: 1813 Immune/Hematopoietic,
    Musculoskeletal,
    Neural/Sensory
    HRDEP41 SEQ ID NO: 1814 Cancer
    HRDEP41 SEQ ID NO: 1815 Cancer
    HTEGF16 SEQ ID NO: 1816 Cancer
    HTEGF16 SEQ ID NO: 1817 Cancer
    HTEGF16 SEQ ID NO: 1818 Cancer
    HSUMA53 SEQ ID NO: 1819 Cancer
    HSUMA53 SEQ ID NO: 1820 Cancer
    HSUMA53 SEQ ID NO: 1821 Cancer
    HSUMA53 SEQ ID NO: 1822 Cancer
    HISET33 SEQ ID NO: 1823 Digestive
    HISET33 SEQ ID NO: 1824 Digestive
    HTTIJ31 SEQ ID NO: 1825 Reproductive
    HTTIJ31 SEQ ID NO: 1826 Reproductive
    HTPFX16 SEQ ID NO: 1827 Digestive,
    Reproductive,
    Respiratory
    HTPFX16 SEQ ID NO: 1828 Digestive,
    Reproductive,
    Respiratory
    HTFMX90 SEQ ID NO: 1829 Cancer
    HTFMX90 SEQ ID NO: 1830 Cancer
    HTFMX90 SEQ ID NO: 1831 Cancer
    HE8FD93 SEQ ID NO: 1832 Cancer
    HE8FD93 SEQ ID NO: 1833 Cancer
    HE8FD93 SEQ ID NO: 1834 Cancer
    HE8FD93 SEQ ID NO: 1835 Cancer
    HKGBJ74 SEQ ID NO: 1836 Cancer
    HKGBJ74 SEQ ID NO: 1837 Cancer
    HKGBJ74 SEQ ID NO: 1838 Cancer
    HKGBJ74 SEQ ID NO: 1839 Cancer
    HEEAG84 SEQ ID NO: 1840 Reproductive
    HEEAG84 SEQ ID NO: 1841 Reproductive
    HEOQX60 SEQ ID NO: 1842 Cancer
    HEOQX60 SEQ ID NO: 1843 Cancer
    HNGGB09 SEQ ID NO: 1844 Immune/Hematopoietic
    HNGGB09 SEQ ID NO: 1845 Immune/Hematopoietic
    HKIYI48 SEQ ID NO: 1846 Cancer
    HKIYI48 SEQ ID NO: 1847 Cancer
    HKIYI48 SEQ ID NO: 1848 Cancer
    HKIYI48 SEQ ID NO: 1849 Cancer
    HSYAB05 SEQ ID NO: 1850 Cancer
    HSYAB05 SEQ ID NO: 1851 Cancer
    HARMJ38 SEQ ID NO: 1852 Cancer
    HARMJ38 SEQ ID NO: 1853 Cancer
    HARMJ38 SEQ ID NO: 1854 Cancer
    HARMJ38 SEQ ID NO: 1855 Cancer
    HDTJG33 SEQ ID NO: 1856 Cancer
    HWAGJ85 SEQ ID NO: 1857 Cardiovascular,
    Immune/Hematopoietic
    HWAGJ85 SEQ ID NO: 1858 Cardiovascular,
    Immune/Hematopoietic
    HE2OW03 SEQ ID NO: 1859 Mixed Fetal
    HE2OW03 SEQ ID NO: 1860 Mixed Fetal
    HBQAE92 SEQ ID NO: 1861 Digestive,
    Neural/Sensory
    HBQAE92 SEQ ID NO: 1862 Digestive,
    Neural/Sensory
    HBQAE92 SEQ ID NO: 1863 Digestive,
    Neural/Sensory
    HTODL92 SEQ ID NO: 1864 Cancer
    HTODL92 SEQ ID NO: 1865 Cancer
    HTODL92 SEQ ID NO: 1866 Cancer
    HLQBR41 SEQ ID NO: 1867 Cancer
    HLQBR41 SEQ ID NO: 1868 Cancer
    HDSAP92 SEQ ID NO: 1869 Cancer
    HDSAP92 SEQ ID NO: 1870 Cancer
    HTAEC92 SEQ ID NO: 1871 Cancer
    HTAEC92 SEQ ID NO: 1872 Cancer
    HSLCK11 SEQ ID NO: 1873 Cancer
    HSLCK11 SEQ ID NO: 1874 Cancer
    HSLCK11 SEQ ID NO: 1875 Cancer
    HFCDR13 SEQ ID NO: 1876 Neural/Sensory
    HSLDS06 SEQ ID NO: 1877 Musculoskeletal
    HSLEF58 SEQ ID NO: 1878 Cardiovascular,
    Digestive,
    Musculoskeletal
    HPCAO10 SEQ ID NO: 1879 Cancer
    HMEJL61 SEQ ID NO: 1880 Cancer
    HMEJL61 SEQ ID NO: 1881 Cancer
    HMEJL61 SEQ ID NO: 1882 Cancer
    HUSHH92 SEQ ID NO: 1883 Cancer
    HUSHH92 SEQ ID NO: 1884 Cancer
    HUSHH92 SEQ ID NO: 1885 Cancer
    HUSHH92 SEQ ID NO: 1886 Cancer
    HBZAI90 SEQ ID NO: 1887 Immune/Hematopoietic,
    Reproductive
    HBZAI90 SEQ ID NO: 1888 Immune/Hematopoietic,
    Reproductive
    HNGIQ57 SEQ ID NO: 1889 Immune/Hematopoietic
    HNGIQ57 SEQ ID NO: 1890 Immune/Hematopoietic
    HNGJF62 SEQ ID NO: 1891 Immune/Hematopoietic
    HNGJF62 SEQ ID NO: 1892 Immune/Hematopoietic
    HFXJY38 SEQ ID NO: 1893 Neural/Sensory
    HFXJY38 SEQ ID NO: 1894 Neural/Sensory
    HFXKR54 SEQ ID NO: 1895 Endocrine,
    Immune/Hematopoietic,
    Neural/Sensory
    HFXKR54 SEQ ID NO: 1896 Endocrine,
    Immune/Hematopoietic,
    Neural/Sensory
    HFXKR54 SEQ ID NO: 1897 Endocrine,
    Immune/Hematopoietic,
    Neural/Sensory
    HAPOB80 SEQ ID NO: 1898 Immune/Hematopoietic,
    Musculoskeletal
    HAPOB80 SEQ ID NO: 1899 Immune/Hematopoietic,
    Musculoskeletal
    HAPOB80 SEQ ID NO: 1900 Immune/Hematopoietic,
    Musculoskeletal
    HAPOB80 SEQ ID NO: 1901 Immune/Hematopoietic,
    Musculoskeletal
    HBJHJ80 SEQ ID NO: 1902 Connective/Epithelial,
    Immune/Hematopoietic,
    Reproductive
    HFADF37 SEQ ID NO: 1903 Cancer
    HFADF37 SEQ ID NO: 1904 Cancer
    HNTSS75 SEQ ID NO: 1905 Cancer
    HCQDE22 SEQ ID NO: 1906 Digestive
    HCQDE22 SEQ ID NO: 1907 Digestive
    HE8NQ42 SEQ ID NO: 1908 Mixed Fetal
    HE8NQ42 SEQ ID NO: 1909 Mixed Fetal
    HE8QD31 SEQ ID NO: 1910 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HE8QD31 SEQ ID NO: 1911 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HE9PR39 SEQ ID NO: 1912 Digestive,
    Mixed Fetal,
    Musculoskeletal
    HE9PR39 SEQ ID NO: 1913 Digestive,
    Mixed Fetal,
    Musculoskeletal
    HE9PR39 SEQ ID NO: 1914 Digestive,
    Mixed Fetal,
    Musculoskeletal
    HE9PR39 SEQ ID NO: 1915 Digestive,
    Mixed Fetal,
    Musculoskeletal
    HNHLA36 SEQ ID NO: 1916 Immune/Hematopoietic,
    Reproductive
    HNHLA36 SEQ ID NO: 1917 Immune/Hematopoietic,
    Reproductive
    HNHOD23 SEQ ID NO: 1918 Cancer
    HNHOD23 SEQ ID NO: 1919 Cancer
    HNHOD23 SEQ ID NO: 1920 Cancer
    HNGNI25 SEQ ID NO: 1921 Immune/Hematopoietic
    HNGNI25 SEQ ID NO: 1922 Immune/Hematopoietic
    HNGNI25 SEQ ID NO: 1923 Immune/Hematopoietic
    HNGNI25 SEQ ID NO: 1924 Immune/Hematopoietic
    HNGOQ44 SEQ ID NO: 1925 Immune/Hematopoietic
    HNGOQ44 SEQ ID NO: 1926 Immune/Hematopoietic
    HTLGE31 SEQ ID NO: 1927 Immune/Hematopoietic,
    HODHE60 SEQ ID NO: 1928 Reproductive
    HODHE60 SEQ ID NO: 1929 Reproductive
    HTLIV19 SEQ ID NO: 1930 Reproductive
    HOSDW58 SEQ ID NO: 1931 Cancer
    HOSDW58 SEQ ID NO: 1932 Cancer
    HOSDW58 SEQ ID NO: 1933 Cancer
    HPJDM47 SEQ ID NO: 1934 Reproductive
    HPJDM47 SEQ ID NO: 1935 Reproductive
    HPJEC20 SEQ ID NO: 1936 Cancer
    HPJEC20 SEQ ID NO: 1937 Cancer
    HTTJK27 SEQ ID NO: 1938 Reproductive
    HTTJK27 SEQ ID NO: 1939 Reproductive
    HTFOE85 SEQ ID NO: 1940 Immune/Hematopoietic
    HTFOE85 SEQ ID NO: 1941 Immune/Hematopoietic
    HTFOE85 SEQ ID NO: 1942 Immune/Hematopoietic
    HIPBA31 SEQ ID NO: 1943 Cancer
    HIPBA31 SEQ ID NO: 1944 Cancer
    HFVJY02 SEQ ID NO: 1945 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HFVJY02 SEQ ID NO: 1946 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HFVJY02 SEQ ID NO: 1947 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HFVJY02 SEQ ID NO: 1948 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HFVJY02 SEQ ID NO: 1949 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HOCOO19 SEQ ID NO: 1950 Cancer
    HOCOO19 SEQ ID NO: 1951 Cancer
    HOCOO19 SEQ ID NO: 1952 Cancer
    HWMKQ25 SEQ ID NO: 1953 Digestive,
    Reproductive
    HWMKQ25 SEQ ID NO: 1954 Digestive,
    Reproductive
    HWMKQ25 SEQ ID NO: 1955 Digestive,
    Reproductive
    HCOPG62 SEQ ID NO: 1956 Cancer
    HCOPG62 SEQ ID NO: 1957 Cancer
    HNKEL47 SEQ ID NO: 1958 Cardiovascular,
    Connective/Epithelial,
    Digestive
    HNKEL47 SEQ ID NO: 1959 Cardiovascular,
    Connective/Epithelial,
    Digestive
    HTPIY88 SEQ ID NO: 1960 Digestive
    HTPIY88 SEQ ID NO: 1961 Digestive
    HTPIY88 SEQ ID NO: 1962 Digestive
    HTPIY88 SEQ ID NO: 1963 Digestive
    HEGBS69 SEQ ID NO: 1964 Neural/Sensory,
    Reproductive
    HEGBS69 SEQ ID NO: 1965 Neural/Sensory,
    Reproductive
    HOFMU07 SEQ ID NO: 1966 Reproductive
    HOFMU07 SEQ ID NO: 1967 Reproductive
    HLWBM40 SEQ ID NO: 1968 Neural/Sensory,
    Reproductive
    HLWBM40 SEQ ID NO: 1969 Neural/Sensory,
    Reproductive
    HLWBM40 SEQ ID NO: 1970 Neural/Sensory,
    Reproductive
    HAMFT10 SEQ ID NO: 1971 Cancer
    HAMFT10 SEQ ID NO: 1972 Cancer
    HNTBP17 SEQ ID NO: 1973 Cancer
    HNTBP17 SEQ ID NO: 1974 Cancer
    HWDAO40 SEQ ID NO: 1975 Cancer
    HWDAO40 SEQ ID NO: 1976 Cancer
    HWDAO40 SEQ ID NO: 1977 Cancer
    HAJCL25 SEQ ID NO: 1978 Immune/Hematopoietic
    HAJCL25 SEQ ID NO: 1979 Immune/Hematopoietic
    HAJCL25 SEQ ID NO: 1980 Immune/Hematopoietic
    HNTEO95 SEQ ID NO: 1981 Immune/Hematopoietic
    HNTEO95 SEQ ID NO: 1982 Immune/Hematopoietic
    HNTEO95 SEQ ID NO: 1983 Immune/Hematopoietic
    HWAFG52 SEQ ID NO: 1984 Cancer
    HWAFG52 SEQ ID NO: 1985 Cancer
    HWAFG52 SEQ ID NO: 1986 Cancer
    HWAFG52 SEQ ID NO: 1987 Cancer
    HWAHE17 SEQ ID NO: 1988 Digestive,
    Immune/Hematopoietic
    HWAHE17 SEQ ID NO: 1989 Digestive,
    Immune/Hematopoietic
    HWAHE17 SEQ ID NO: 1990 Digestive,
    Immune/Hematopoietic
    HUJBK19 SEQ ID NO: 1991 Cancer
    HUJBK19 SEQ ID NO: 1992 Cancer
    HUJBK19 SEQ ID NO: 1993 Cancer
    HWHJD93 SEQ ID NO: 1994 Cancer
    HWHJD93 SEQ ID NO: 1995 Cancer
    HAOST94 SEQ ID NO: 1996 Cancer
    HAOST94 SEQ ID NO: 1997 Cancer
    HKZAH22 SEQ ID NO: 1998 Reproductive
    HKZAH22 SEQ ID NO: 1999 Reproductive
    HKZAH22 SEQ ID NO: 2000 Reproductive
    HKZAO35 SEQ ID NO: 2001 Reproductive
    HKZAO35 SEQ ID NO: 2002 Reproductive
    HWHSK19 SEQ ID NO: 2003 Cancer
    HWHSK19 SEQ ID NO: 2004 Cancer
    HWHSK19 SEQ ID NO: 2005 Cancer
    HMWFG79 SEQ ID NO: 2006 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMWFG79 SEQ ID NO: 2007 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMWFG79 SEQ ID NO: 2008 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMWFG79 SEQ ID NO: 2009 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMWFG79 SEQ ID NO: 2010 Digestive,
    Immune/Hematopoietic,
    Reproductive
    HMTAE85 SEQ ID NO: 2011 Cancer
    HMTBI36 SEQ ID NO: 2012 Cancer
    HSUME76 SEQ ID NO: 2013 Cancer
    HSUME76 SEQ ID NO: 2014 Cancer
    HTEAF65 SEQ ID NO: 2015 Excretory,
    Reproductive
    HTEAT31 SEQ ID NO: 2016 Cancer
    HAJAN23 SEQ ID NO: 2017 Cancer
    HAPRJ16 SEQ ID NO: 2018 Cancer
    HDTDT55 SEQ ID NO: 2019 Cancer
    HSKDA27 SEQ ID NO: 2020 Cancer
    HSKDA27 SEQ ID NO: 2021 Cancer
    HWLED11 SEQ ID NO: 2022 Cancer
    HADGD33 SEQ ID NO: 2023 Connective/Epithelial,
    Neural/Sensory,
    Reproductive
    HCEBF19 SEQ ID NO: 2024 Cancer
    HCEBF19 SEQ ID NO: 2025 Cancer
    HDPHH40 SEQ ID NO: 2026 Cancer
    HHEPM33 SEQ ID NO: 2027 Cancer
    HJBAF16 SEQ ID NO: 2028 Cancer
    HJBCU04 SEQ ID NO: 2029 Cancer
    HWABY10 SEQ ID NO: 2030 Cancer
    HWABY10 SEQ ID NO: 2031 Cancer
    HWABY10 SEQ ID NO: 2032 Cancer
    HWABY10 SEQ ID NO: 2033 Cancer
    HDPQN11 SEQ ID NO: 2034 Cancer
    HDPQN11 SEQ ID NO: 2035 Cancer
    HMSAW68 SEQ ID NO: 2036 Cancer
    HMSGP80 SEQ ID NO: 2037 Cancer
    HPJBZ76 SEQ ID NO: 2038 Cancer
    HSIGM62 SEQ ID NO: 2039 Cancer
    HSLHS22 SEQ ID NO: 2040 Cancer
    HTXOZ19 SEQ ID NO: 2041 Cancer
    HTXOZ19 SEQ ID NO: 2042 Cancer
    HAPQQ94 SEQ ID NO: 2043 Immune/Hematopoietic,
    Reproductive
    HAPQQ94 SEQ ID NO: 2044 Immune/Hematopoietic,
    Reproductive
    HAPSA79 SEQ ID NO: 2045 Cancer
    HAPSA79 SEQ ID NO: 2046 Cancer
    HAPSA79 SEQ ID NO: 2047 Cancer
    HDPAJ93 SEQ ID NO: 2048 Cancer
    HELGF34 SEQ ID NO: 2049 Cancer
    HETEQ88 SEQ ID NO: 2050 Cancer
    HMSAC18 SEQ ID NO: 2051 Cancer
    HMSAC18 SEQ ID NO: 2052 Cancer
    HPQSH59 SEQ ID NO: 2053 Cancer
    HSIFV30 SEQ ID NO: 2054 Cancer
    HSVCB08 SEQ ID NO: 2055 Cancer
    HT3SF53 SEQ ID NO: 2056 Cancer
    HARMS04 SEQ ID NO: 2057 Connective/Epithelial,
    Digestive
    HCDBP36 SEQ ID NO: 2058 Musculoskeletal
    HCEPE30 SEQ ID NO: 2059 Excretory,
    Neural/Sensory
    HE9RM63 SEQ ID NO: 2060 Cancer
    HKAJF71 SEQ ID NO: 2061 Cancer
    HNBAF49 SEQ ID NO: 2062 Cancer
    HSLDJ89 SEQ ID NO: 2063 Cancer
    HSXGI47 SEQ ID NO: 2064 Cancer
    HTEAJ18 SEQ ID NO: 2065 Reproductive
    HTTEV40 SEQ ID NO: 2066 Cancer
    HWBCB89 SEQ ID NO: 2067 Cancer
    HWHGZ51 SEQ ID NO: 2068 Cancer
    HADDH60 SEQ ID NO: 2069 Connective/Epithelial,
    Immune/Hematopoietic,
    Neural/Sensory
    HBXCL93 SEQ ID NO: 2070 Neural/Sensory,
    Reproductive
    HPTRH66 SEQ ID NO: 2071 Cancer
    HNFHD58 SEQ ID NO: 2072 Cancer
    HACAB58 SEQ ID NO: 2073 Cancer
    HCE3Z39 SEQ ID NO: 2074 Cancer
    HCFCU69 SEQ ID NO: 2075 Cancer
    HCE3Z39 SEQ ID NO: 2076 Cancer
    HCELE47 SEQ ID NO: 2077 Cancer
    HCWHP79 SEQ ID NO: 2078 Immune/Hematopoietic
    HDLAG89 SEQ ID NO: 2079 Cancer
    HDLAO28 SEQ ID NO: 2080 Cancer
    HDQGY41 SEQ ID NO: 2081 Cancer
    HE8FK78 SEQ ID NO: 2082 Cancer
    HE8FK78 SEQ ID NO: 2083 Cancer
    HETHR73 SEQ ID NO: 2084 Cancer
    HFIUW36 SEQ ID NO: 2085 Cancer
    HFKKS66 SEQ ID NO: 2086 Cancer
    HFPFK57 SEQ ID NO: 2087 Neural/Sensory,
    Reproductive
    HFVJP07 SEQ ID NO: 2088 Digestive,
    Immune/Hematopoietic
    HLQEM64 SEQ ID NO: 2089 Cancer
    HSSDG41 SEQ ID NO: 2090 Cancer
    HLQGP82 SEQ ID NO: 2091 Connective/Epithelial,
    Digestive,
    Musculoskeletal
    HMSMD07 SEQ ID NO: 2092 Cancer
    HNGIR58 SEQ ID NO: 2093 Immune/Hematopoietic
    HMAMI21 SEQ ID NO: 2094 Cancer
    HNHNB29 SEQ ID NO: 2095 Immune/Hematopoietic
    HNTEO78 SEQ ID NO: 2096 Digestive,
    Immune/Hematopoietic
    HJPAY76 SEQ ID NO: 2097 Cancer
    HOEEK12 SEQ ID NO: 2098 Cancer
    HOFNC14 SEQ ID NO: 2099 Reproductive
    HOSNU69 SEQ ID NO: 2100 Cancer
    HPJCL28 SEQ ID NO: 2101 Neural/Sensory,
    Reproductive
    HRACI26 SEQ ID NO: 2102 Digestive,
    Excretory
    HTLIT63 SEQ ID NO: 2103 Reproductive
    HTEAM34 SEQ ID NO: 2104 Reproductive
    HTEAM34 SEQ ID NO: 2105 Reproductive
    HUFGH53 SEQ ID NO: 2106 Cancer
    HUSBA88 SEQ ID NO: 2107 Cancer
    HELHN47 SEQ ID NO: 2108 Cancer
    HELHN47 SEQ ID NO: 2109 Cancer
    HELHN47 SEQ ID NO: 2110 Cancer
    HETAY39 SEQ ID NO: 2111 Cancer
    HFICR14 SEQ ID NO: 2112 Cancer
    HFICR14 SEQ ID NO: 2113 Cancer
    HFKET18 SEQ ID NO: 2114 Cancer
    HFXDK20 SEQ ID NO: 2115 Immune/Hematopoietic,
    Neural/Sensory
    HKMLX18 SEQ ID NO: 2116 Cancer
    HMSCM88 SEQ ID NO: 2117 Immune/Hematopoietic
    HMABG70 SEQ ID NO: 2118 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HMADJ74 SEQ ID NO: 2119 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HMADJ14 SEQ ID NO: 2120 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HMADJ14 SEQ ID NO: 2121 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HMADJ14 SEQ ID NO: 2122 Connective/Epithelial,
    Immune/Hematopoietic,
    Musculoskeletal
    HNEBY54 SEQ ID NO: 2123 Cancer
    HNEDD37 SEQ ID NO: 2124 Cancer
    HNGOU82 SEQ ID NO: 2125 Immune/Hematopoietic,
    Reproductive
    HNGOW62 SEQ ID NO: 2126 Immune/Hematopoietic
    HSIC066 SEQ ID NO: 2127 Cancer
    HSIDQ93 SEQ ID NO: 2128 Cancer
    HSLGM81 SEQ ID NO: 2129 Cancer
    HSYBM41 SEQ ID NO: 2130 Cancer
    HSODB85 SEQ ID NO: 2131 Cancer
    HSRFZ57 SEQ ID NO: 2132 Excretory,
    Musculoskeletal
    HSXAZ05 SEQ ID NO: 2133 Neural/Sensory,
    Respiratory
    HTPCW21 SEQ ID NO: 2134 Digestive,
    Neural/Sensory
    HTPCW21 SEQ ID NO: 2135 Digestive,
    Neural/Sensory
    HTXKF95 SEQ ID NO: 2136 Cancer
    HTXKF95 SEQ ID NO: 2137 Cancer
    HUFBC44 SEQ ID NO: 2138 Digestive,
    Mixed Fetal,
    Neural/Sensory
    HAAAI67 SEQ ID NO: 2139 Cancer
    HFKIA71 SEQ ID NO: 2140 Cancer
    HAMFP32 SEQ ID NO: 2141 Cancer
    HAPQU71 SEQ ID NO: 2142 Cancer
    HAPQU71 SEQ ID NO: 2143 Cancer
    HLHDL42 SEQ ID NO: 2144 Cancer
    HAVVG36 SEQ ID NO: 2145 Cancer
    HBGNP63 SEQ ID NO: 2146 Reproductive
    HBJNC59 SEQ ID NO: 2147 Cancer
    HAPQT56 SEQ ID NO: 2148 Cancer
    HCABW07 SEQ ID NO: 2149 Cancer
    HDPFB02 SEQ ID NO: 2150 Cancer
    HMWDB84 SEQ ID NO: 2151 Cancer
    HDPFB02 SEQ ID NO: 2152 Cancer
    HDPFY41 SEQ ID NO: 2153 Cancer
    HDPIE85 SEQ ID NO: 2154 Cancer
    HDPOE32 SEQ ID NO: 2155 Cancer
    HWABL61 SEQ ID NO: 2156 Cancer
    HWABW88 SEQ ID NO: 2157 Cancer
    HWDAQ83 SEQ ID NO: 2158 Cancer
    HWDAQ83 SEQ ID NO: 2159 Cancer
    HWLHZ79 SEQ ID NO: 2160 Connective/Epithelial,
    Digestive,
    Reproductive
    HTXJM94 SEQ ID NO: 2161 Cancer
    HDPQG01 SEQ ID NO: 2162 Cancer
    HJPAD80 SEQ ID NO: 2163 Cancer
    HDPQG01 SEQ ID NO: 2164 Cancer
    HFXLF67 SEQ ID NO: 2165 Neural/Sensory
    HE2IO57 SEQ ID NO: 2166 Cancer
    HKGDP17 SEQ ID NO: 2167 Respiratory
    HLQFB12 SEQ ID NO: 2168 Digestive,
    Reproductive
    HLQFT18 SEQ ID NO: 2169 Digestive,
    Reproductive
    HOFNX30 SEQ ID NO: 2170 Reproductive
    HSSDM23 SEQ ID NO: 2171 Cancer
    HSSDM23 SEQ ID NO: 2172 Cancer
    HSVBD67 SEQ ID NO: 2173 Cancer
    HSVBD67 SEQ ID NO: 2174 Cancer
    HTGAT51 SEQ ID NO: 2175 Cardiovascular,
    Immune/Hematopoietic,
    Reproductive
    HTLGV19 SEQ ID NO: 2176 Excretory,
    Reproductive
    HTPHH74 SEQ ID NO: 2177 Cancer
    HTFOB75 SEQ ID NO: 2178 Cancer
    HTPHH74 SEQ ID NO: 2179 Cancer
    HWHGK36 SEQ ID NO: 2180 Cancer
    HLWAD77 SEQ ID NO: 2181 Cancer
    HDTGF15 SEQ ID NO: 2182 Cancer
    HWMBB68 SEQ ID NO: 2183 Cancer
    HWMBB68 SEQ ID NO: 2184 Cancer
    HAGDA35 SEQ ID NO: 2185 Cancer
    HAGDA35 SEQ ID NO: 2186 Cancer
    HAGDA35 SEQ ID NO: 2187 Cancer
    HRODQ04 SEQ ID NO: 2188 Cancer
    HTOJV86 SEQ ID NO: 2189 Cancer
    HCEFZ82 SEQ ID NO: 2190 Cancer
    HNGFW58 SEQ ID NO: 2191 Cancer
    HHBGE77 SEQ ID NO: 2192 Cancer
    HADFW77 SEQ ID NO: 2193 Cancer
    HSIED48 SEQ ID NO: 2194 Cancer
    HCEFZ82 SEQ ID NO: 2195 Cancer
    HTTCT46 SEQ ID NO: 2196 Cancer
    HSDEE58 SEQ ID NO: 2197 Cancer
    HEBCV31 SEQ ID NO: 2198 Cancer
    HDPOL27 SEQ ID NO: 2199 Cancer
    HDPOL27 SEQ ID NO: 2200 Cancer
    HE6DI14 SEQ ID NO: 2201 Cancer
    HLYAN43 SEQ ID NO: 2202 Cancer
    HDPUM13 SEQ ID NO: 2203 Cancer
    HPLAT62 SEQ ID NO: 2204 Cancer
    HAPQT56 SEQ ID NO: 2205 Cancer
    HACBG19 SEQ ID NO: 2206 Cancer
    HACBG19 SEQ ID NO: 2207 Cancer
    HLYAV34 SEQ ID NO: 2208 Cancer
    HCNSM85 SEQ ID NO: 2209 Cancer
    HTOCG60 SEQ ID NO: 2210 Cancer
    HLYAV34 SEQ ID NO: 2211 Cancer
    HDPWX42 SEQ ID NO: 2212 Cancer
    HOFNF53 SEQ ID NO: 2213 Reproductive
    HOFNF53 SEQ ID NO: 2214 Reproductive
    HMSEO15 SEQ ID NO: 2215 Cancer
    HBXFT65 SEQ ID NO: 2216 Cancer
    HFCEQ37 SEQ ID NO: 2217 Cancer
    HWNFG66 SEQ ID NO: 2218 Digestive
    HOHCA60 SEQ ID NO: 2219 Cancer
    HOHCA60 SEQ ID NO: 2220 Cancer
    HOHCA60 SEQ ID NO: 2221 Cancer
    HOHCA60 SEQ ID NO: 2222 Cancer
    HOHCA60 SEQ ID NO: 2223 Cancer
    HLDRR08 SEQ ID NO: 2224 Digestive
    HSKNP59 SEQ ID NO: 2225 Musculoskeletal
    HSKNP59 SEQ ID NO: 2226 Musculoskeletal
    HAMHE82 SEQ ID NO: 2227 Cancer
    HBIOO68 SEQ ID NO: 2228 Cancer
    HCE3C63 SEQ ID NO: 2229 Mixed Fetal,
    Neural/Sensory
    HCNDV12 SEQ ID NO: 2230 Digestive,
    Reproductive
    HMWDW68 SEQ ID NO: 2231 Cancer
    HE2BC57 SEQ ID NO: 2232 Cancer
    HSDEE58 SEQ ID NO: 2233 Cancer
    HE9OW91 SEQ ID NO: 2234 Cancer
    HFCFI20 SEQ ID NO: 2235 Cancer
    HELEN05 SEQ ID NO: 2236 Cancer
    HISEL50 SEQ ID NO: 2237 Cancer
    HLHDL62 SEQ ID NO: 2238 Cancer
    HDFQB93 SEQ ID NO: 2239 Cancer
    HLHDQ86 SEQ ID NO: 2240 Cancer
    HLNAB24 SEQ ID NO: 2241 Immune/Hematopoietic
    HLYBQ90 SEQ ID NO: 2242 Cancer
    HLYBQ90 SEQ ID NO: 2243 Cancer
    HNHDP39 SEQ ID NO: 2244 Endocrine,
    Immune/Hematopoietic,
    Reproductive
    HNTAC64 SEQ ID NO: 2245 Cancer
    HNTMY29 SEQ ID NO: 2246 Connective/Epithelial,
    Reproductive
    HOFOC33 SEQ ID NO: 2247 Reproductive
    HOFOC33 SEQ ID NO: 2248 Reproductive
    HTWFK18 SEQ ID NO: 2249 Connective/Epithelial,
    Immune/Hematopoietic
    HAPNJ39 SEQ ID NO: 2250 Cancer
    HDQFU27 SEQ ID NO: 2251 Cancer
    HETJZ45 SEQ ID NO: 2252 Cancer
    HTEMX36 SEQ ID NO: 2253 Cancer
    HNTCH90 SEQ ID NO: 2254 Cancer
    HWLBP46 SEQ ID NO: 2255 Cancer
    HA5BM53 SEQ ID NO: 2256 Cancer
    HMCEH49 SEQ ID NO: 2257 Cancer
    HKBAL25 SEQ ID NO: 2258 Digestive,
    Musculoskeletal
    HE8EF43 SEQ ID NO: 2259 Cancer
    HE2RN91 SEQ ID NO: 2260 Cancer
    HTLIO20 SEQ ID NO: 2261 Immune/Hematopoietic,
    Neural/Sensory
    HBIMF63 SEQ ID NO: 2262 Reproductive
    HE9PM90 SEQ ID NO: 2263 Cancer
    HNTDX22 SEQ ID NO: 2264 Reproductive
    HHFCE59 SEQ ID NO: 2265 Cancer
    HCGAD44 SEQ ID NO: 2266 Cancer
    HSSJJ51 SEQ ID NO: 2267 Cancer
  • In preferred embodiments, the albumin fusion proteins of the invention are capable of a therapeutic activity and/or biologic activity corresponding to the therapeutic activity and/or biologic activity of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein listed in the corresponding row of Table 1. In further preferred embodiments, the therapeutically active protein portions of the albumin fusion proteins of the invention are fragments or variants of the reference sequence cited in the “Exemplary Identifier” column of Table 1, and are capable of the therapeutic activity and/or biologic activity of the corresponding Therapeutic protein. [0073]
  • Polypeptide and Polynucleotide Fragments and Variants [0074]
  • Fragments [0075]
  • The present invention is further directed to fragments of the Therapeutic proteins described in Table 1, albumin proteins, and/or albumin fusion proteins of the invention. [0076]
  • Even if deletion of one or more amino acids from the N-terminus of a protein results in modification or loss of one or more biological functions of the Therapeutic protein, albumin protein, and/or albumin fusion protein, other Therapeutic activities and/or functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) may still be retained. For example, the ability of polypeptides with N-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptides generally will be retained when less than the majority of the residues of the complete polypeptide are removed from the N-terminus. Whether a particular polypeptide lacking N-terminal residues of a complete polypeptide retains such immunologic activities can readily be determined by routine methods described herein and otherwise known in the art. It is not unlikely that a mutein with a large number of deleted N-terminal amino acid residues may retain some biological or immunogenic activities. In fact, peptides composed of as few as six amino acid residues may often evoke an immune response. [0077]
  • Accordingly, fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., a Therapeutic protein as disclosed in Table 1). In particular, N-terminal deletions may be described by the general formula m−q, where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1), and m is defined as any integer ranging from 2 to q−6. Polynucleotides encoding these polypeptides are also encompassed by the invention. [0078]
  • In addition, fragments of serum albumin polypeptides corresponding to an albumin protein portion of an albumin fusion protein of the invention, include the full length protein as well as polypeptides having one or more residues deleted from the amino terminus of the amino acid sequence of the reference polypeptide (i.e., serum albumin). In particular, N-terminal deletions may be described by the general formula m[0079] −585, where 585 is a whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO:18), and m is defined as any integer ranging from 2 to 579. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • Moreover, fragments of albumin fusion proteins of the invention, include the full length albumin fusion protein as well as polypeptides having one or more residues deleted from the amino terminus of the albumin fusion protein. In particular, N-terminal deletions may be described by the general formula m−q, where q is a whole integer representing the total number of amino acid residues in the albumin fusion protein, and m is defined as any integer ranging from 2 to q−6. Polynucleotides encoding these polypeptides are also encompassed by the invention. [0080]
  • Also as mentioned above, even if deletion of one or more amino acids from the N-terminus or C-terminus of a reference polypeptide (e.g., a Therapeutic protein and/or serum albumin protein) results in modification or loss of one or more biological functions of the protein, other functional activities (e.g., biological activities, ability to multimerize, ability to bind a ligand) and/or Therapeutic activities may still be retained. For example the ability of polypeptides with C-terminal deletions to induce and/or bind to antibodies which recognize the complete or mature forms of the polypeptide generally will be retained when less than the majority of the residues of the complete or mature polypeptide are removed from the C-terminus. Whether a particular polypeptide lacking the N-terminal and/or C-terminal residues of a reference polypeptide retains Therapeutic activity can readily be determined by routine methods described herein and/or otherwise known in the art. [0081]
  • The present invention further provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., a Therapeutic protein referred to in Table 1). In particular, C-terminal deletions may be described by the [0082] general formula 1−n, where n is any whole integer ranging from 6 to q−1, and where q is a whole integer representing the total number of amino acid residues in a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1). Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • In addition, the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of the amino acid sequence of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention (e.g., serum albumin). In particular, C-terminal deletions may be described by the [0083] general formula 1−n, where n is any whole integer ranging from 6 to 584, where 584 is the whole integer representing the total number of amino acid residues in serum albumin (SEQ ID NO: 18) minus 1. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • Moreover, the present invention provides polypeptides having one or more residues deleted from the carboxy terminus of an albumin fusion protein of the invention. In particular, C-terminal deletions may be described by the [0084] general formula 1−n, where n is any whole integer ranging from 6 to q−1, and where q is a whole integer representing the total number of amino acid residues in an albumin fusion protein of the invention. Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • In addition, any of the above described N- or C-terminal deletions can be combined to produce a N- and C-terminal deleted reference polypeptide. The invention also provides polypeptides having one or more amino acids deleted from both the amino and the carboxyl termini, which may be described generally as having residues m-n of a reference polypeptide (e.g., a Therapeutic protein referred to in Table 1, or serum albumin (e.g., SEQ ID NO: 18), or an albumin fusion protein of the invention) where n and m are integers as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention. [0085]
  • The present application is also directed to proteins containing polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to a reference polypeptide sequence (e.g., a Therapeutic protein, serum albumin protein or an albumin fusion protein of the invention) set forth herein, or fragments thereof. In preferred embodiments, the application is directed to proteins comprising polypeptides at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to reference polypeptides having the amino acid sequence of N- and C-terminal deletions as described above. Polynucleotides encoding these polypeptides are also encompassed by the invention. [0086]
  • Preferred polypeptide fragments of the invention are fragments comprising, or alternatively, consisting of, an amino acid sequence that displays a Therapeutic activity and/or functional activity (e.g. biological activity) of the polypeptide sequence of the Therapeutic protein or serum albumin protein of which the amino acid sequence is a fragment. [0087]
  • Other preferred polypeptide fragments are biologically active fragments. Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of the polypeptide of the present invention. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity. [0088]
  • Variants [0089]
  • “Variant” refers to a polynucleotide or nucleic acid differing from a reference nucleic acid or polypeptide, but retaining essential properties thereof. Generally, variants are overall closely similar, and, in many regions, identical to the reference nucleic acid or polypeptide. [0090]
  • As used herein, “variant”, refers to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin portion of an albumin fusion protein of the invention, or albumin fusion protein differing in sequence from a Therapeutic protein (e.g. see “therapeutic” column of Table 1), albumin protein, and/or albumin fusion protein of the invention, respectively, but retaining at least one functional and/or therapeutic property thereof as described elsewhere herein or otherwise known in the art. Generally, variants are overall very similar, and, in many regions, identical to the amino acid sequence of the Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion protein of the invention. Nucleic acids encoding these variants are also encompassed by the invention. [0091]
  • The present invention is also directed to proteins which comprise, or alternatively consist of, an amino acid sequence which is at least 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100%, identical to, for example, the amino acid sequence of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention (e.g., an amino acid sequence disclosed in the “Exemplary Identifier” column of Table 1, or fragments or variants thereof), albumin proteins (e.g., SEQ ID NO:18 or fragments or variants thereof) corresponding to an albumin protein portion of an albumin fusion protein of the invention, and/or albumin fusion proteins of the invention. Fragments of these polypeptides are also provided (e.g., those fragments described herein). Further polypeptides encompassed by the invention are polypeptides encoded by polynucleotides which hybridize to the complement of a nucleic acid molecule encoding an amino acid sequence of the invention under stringent hybridization conditions (e.g., hybridization to filter bound DNA in 6×Sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.2×SSC, 0.1% SDS at about 50-65 degrees Celsius), under highly stringent conditions (e.g., hybridization to filter bound DNA in 6×sodium chloride/Sodium citrate (SSC) at about 45 degrees Celsius, followed by one or more washes in 0.1×SSC, 0.2% SDS at about 68 degrees Celsius), or under other stringent hybridization conditions which are known to those of skill in the art (see, for example, Ausubel, F. M. et al., eds., 1989 [0092] Current protocol in Molecular Biology, Green publishing associates, Inc., and John Wiley & Sons Inc., New York, at pages 6.3.1-6.3.6 and 2.10.3). Polynucleotides encoding these polypeptides are also encompassed by the invention.
  • By a polypeptide having an amino acid sequence at least, for example, 95% “identical” to a query amino acid sequence of the present invention, it is intended that the amino acid sequence of the subject polypeptide is identical to the query sequence except that the subject polypeptide sequence may include up to five amino acid alterations per each 100 amino acids of the query amino acid sequence. In other words, to obtain a polypeptide having an amino acid sequence at least 95% identical to a query amino acid sequence, up to 5% of the amino acid residues in the subject sequence may be inserted, deleted, or substituted with another amino acid. These alterations of the reference sequence may occur at the amino- or carboxy-terminal positions of the reference amino acid sequence or anywhere between those terminal positions, interspersed either individually among residues in the reference sequence or in one or more contiguous groups within the reference sequence. [0093]
  • As a practical matter, whether any particular polypeptide is at least 80%, 85%, 90%, 95%, 96%, 97%, 98% or 99% identical to, for instance, the amino acid sequence of an albumin fusion protein of the invention or a fragment thereof (such as the Therapeutic protein portion of the albumin fusion protein or the albumin portion of the albumin fusion protein), can be determined conventionally using known computer programs. A preferred method for determining the best overall match between a query sequence (a sequence of the present invention) and a subject sequence, also referred to as a global sequence alignment, can be determined using the FASTDB computer program based on the algorithm of Brutlag et al. (Comp. App. Biosci. 6:237-245 (1990)). In a sequence alignment the query and subject sequences are either both nucleotide sequences or both amino acid sequences. The result of said global sequence alignment is expressed as percent identity. Preferred parameters used in a FASTDB amino acid alignment are: Matrix=[0094] PAM 0, k-tuple=2, Mismatch Penalty=1, Joining Penalty=20, Randomization Group Length=0, Cutoff Score=1, Window Size=sequence length, Gap Penalty=5, Gap Size Penalty=0.05, Window Size=500 or the length of the subject amino acid sequence, whichever is shorter.
  • If the subject sequence is shorter than the query sequence due to N- or C-terminal deletions, not because of internal deletions, a manual correction must be made to the results. This is because the FASTDB program does not account for N- and C-terminal truncations of the subject sequence when calculating global percent identity. For subject sequences truncated at the N- and C-termini, relative to the query sequence, the percent identity is corrected by calculating the number of residues of the query sequence that are N- and C-terminal of the subject sequence, which are not matched/aligned with a corresponding subject residue, as a percent of the total bases of the query sequence. Whether a residue is matched/aligned is determined by results of the FASTDB sequence alignment. This percentage is then subtracted from the percent identity, calculated by the above FASTDB program using the specified parameters, to arrive at a final percent identity score. This final percent identity score is what is used for the purposes of the present invention. Only residues to the N- and C-termini of the subject sequence, which are not matched/aligned with the query sequence, are considered for the purposes of manually adjusting the percent identity score. That is, only query residue positions outside the farthest N- and C-terminal residues of the subject sequence. [0095]
  • For example, a 90 amino acid residue subject sequence is aligned with a 100 residue query sequence to determine percent identity. The deletion occurs at the N-terminus of the subject sequence and therefore, the FASTDB alignment does not show a matching/alignment of the first 10 residues at the N-terminus. The 10 unpaired residues represent 10% of the sequence (number of residues at the N- and C-termini not matched/total number of residues in the query sequence) so 10% is subtracted from the percent identity score calculated by the FASTDB program. If the remaining 90 residues were perfectly matched the final percent identity would be 90%. In another example, a 90 residue subject sequence is compared with a 100 residue query sequence. This time the deletions are internal deletions so there are no residues at the N- or C-termini of the subject sequence which are not matched/aligned with the query. In this case the percent identity calculated by FASTDB is not manually corrected. Once again, only residue positions outside the N- and C-terminal ends of the subject sequence, as displayed in the FASTDB alignment, which are not matched/aligned with the query sequence are manually corrected for. No other manual corrections are to made for the purposes of the present invention. [0096]
  • The variant will usually have at least 75% (preferably at least about 80%, 90%, 95% or 99%) sequence identity with a length of normal HA or Therapeutic protein which is the same length as the variant. Homology or identity at the nucleotide or amino acid sequence level is determined by BLAST (Basic Local Alignment Search Tool) analysis using the algorithm employed by the programs blastp, blastn, blastx, tblastn and tblastx (Karlin et al., Proc. Natl. Acad. Sci. USA 87: 2264-2268 (1990) and Altschul, J. Mol. Evol. 36: 290-300 (1993), fully incorporated by reference) which are tailored for sequence similarity searching. [0097]
  • The approach used by the BLAST program is to first consider similar segments between a query sequence and a database sequence, then to evaluate the statistical significance of all matches that are identified and finally to summarize only those matches which satisfy a preselected threshold of significance. For a discussion of basic issues in similarity searching of sequence databases, see Altschul et al., (Nature Genetics 6: 119-129 (1994)) which is fully incorporated by reference. The search parameters for histogram, descriptions, alignments, expect (i.e., the statistical significance threshold for reporting matches against database sequences), cutoff, matrix and filter are at the default settings. The default scoring matrix used by blastp, blastx, tblastn, and tblastx is the BLOSUM62 matrix (Henikoff et al., Proc. Natl. Acad. Sci. USA 89: 10915-10919 (1992), fully incorporated by reference). For blastn, the scoring matrix is set by the ratios of M (i.e., the reward score for a pair of matching residues) to N (i.e., the penalty score for mismatching residues), wherein the default values for M and N are 5 and -4, respectively. Four blastn parameters may be adjusted as follows: Q=10 (gap creation penalty); R=10 (gap extension penalty); wink=1 (generates word hits at every wink[0098] th position along the query); and gapw—16 (sets the window width within which gapped alignments are generated). The equivalent Blastp parameter settings were Q=9; R=2; wink=1; and gapw=32. A Bestfit comparison between sequences, available in the GCG package version 10.0, uses DNA parameters GAP=50 (gap creation penalty) and LEN=3 (gap extension penalty) and the equivalent settings in protein comparisons are GAP=8 and LEN=2.
  • The polynucleotide variants of the invention may contain alterations in the coding regions, non-coding regions, or both. Especially preferred are polynucleotide variants containing alterations which produce silent substitutions, additions, or deletions, but do not alter the properties or activities of the encoded polypeptide. Nucleotide variants produced by silent substitutions due to the degeneracy of the genetic code are preferred. Moreover, polypeptide variants in which less than 50, less than 40, less than 30, less than 20, less than 10, or 5-50, 5-25, 5-10, 1-5, or 1-2 amino acids are substituted, deleted, or added in any combination are also preferred. Polynucleotide variants can be produced for a variety of reasons, e.g., to optimize codon expression for a particular host (change codons in the human mRNA to those preferred by a bacterial host, such as, yeast or [0099] E. coli).
  • In a preferred embodiment, a polynucleotide encoding an albumin portion of an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells. In further preferred embodiment, a polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells. In a still further preferred embodiment, a polynucleotide encoding an albumin fusion protein of the invention is optimized for expression in yeast or mammalian cells. [0100]
  • In an alternative embodiment, a codon optimized polynucleotide encoding a Therapeutic protein portion of an albumin fusion protein of the invention does not hybridize to the wild type polynucleotide encoding the Therapeutic protein under stringent hybridization conditions as described herein. In a further embodiment, a codon optimized polynucleotide encoding an albumin portion of an albumin fusion protein of the invention do not hybridize to the wild type polynucleotide encoding the albumin protein under stringent hybridization conditions as described herein. In another embodiment, a codon optimized polynucleotide encoding an albumin fusion protein of the invention do not hybridize to the wild type polynucleotide encoding the Therapeutic protein portin or the albumin protein portion under stringent hybridization conditions as described herein. [0101]
  • In an additional embodiment, polynucleotides encoding a Therapeutic protein portion of an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of that Therapeutic protein. In a further embodiment, polynucleotides encoding an albumin protein portion of an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of albumin protein. In an alternative embodiment, polynucleotides encoding an albumin fusion protein of the invention do not comprise, or alternatively consist of, the naturally occurring sequence of a Therapeutic protein portion or the albumin protein portion. [0102]
  • Naturally occurring variants are called “allelic variants,” and refer to one of several alternate forms of a gene occupying a given locus on a chromosome of an organism. (Genes II, Lewin, B., ed., John Wiley & Sons, New York (1985)). These allelic variants can vary at either the polynucleotide and/or polypeptide level and are included in the present invention. Alternatively, non-naturally occurring variants may be produced by mutagenesis techniques or by direct synthesis. [0103]
  • Using known methods of protein engineering and recombinant DNA technology, variants may be generated to improve or alter the characteristics of the polypeptides of the present invention. For instance, one or more amino acids can be deleted from the N-terminus or C-terminus of the polypeptide of the present invention without substantial loss of biological function. As an example, Ron et al. (J. Biol. Chem. 268: 2984-2988 (1993)) reported variant KGF proteins having heparin binding activity even after deleting 3, 8, or 27 amino-terminal amino acid residues. Similarly, Interferon gamma exhibited up to ten times higher activity after deleting 8-10 amino acid residues from the carboxy terminus of this protein. (Dobeli et al., J. Biotechnology 7:199-216 (1988).) [0104]
  • Moreover, ample evidence demonstrates that variants often retain a biological activity similar to that of the naturally occurring protein. For example, Gayle and coworkers (J. Biol. Chem. 268:22105-22111 (1993)) conducted extensive mutational analysis of human cytokine IL-1a. They used random mutagenesis to generate over 3,500 individual IL-1a mutants that averaged 2.5 amino acid changes per variant over the entire length of the molecule. Multiple mutations were examined at every possible amino acid position. The investigators found that “[m]ost of the molecule could be altered with little effect on either [binding or biological activity].” In fact, only 23 unique amino acid sequences, out of more than 3,500 nucleotide sequences examined, produced a protein that significantly differed in activity from wild-type. [0105]
  • Furthermore, even if deleting one or more amino acids from the N-terminus or C-terminus of a polypeptide results in modification or loss of one or more biological functions, other biological activities may still be retained. For example, the ability of a deletion variant to induce and/or to bind antibodies which recognize the secreted form will likely be retained when less than the majority of the residues of the secreted form are removed from the N-terminus or C-terminus. Whether a particular polypeptide lacking N- or C-terminal residues of a protein retains such immunogenic activities can readily be determined by routine methods described herein and otherwise known in the art. [0106]
  • Thus, the invention further includes polypeptide variants which have a functional activity (e.g., biological activity and/or therapeutic activity). In highly preferred embodiments the invention provides variants of albumin fusion proteins that have a functional activity (e.g., biological activity and/or therapeutic activity) that corresponds to one or more biological and/or therapeutic activities of the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein. Such variants include deletions, insertions, inversions, repeats, and substitutions selected according to general rules known in the art so as have little effect on activity. [0107]
  • In preferred embodiments, the variants of the invention have conservative substitutions. By “conservative substitutions” is intended swaps within groups such as replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gln, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly. [0108]
  • Guidance concerning how to make phenotypically silent amino acid substitutions is provided, for example, in Bowie et al., “Deciphering the Message in Protein Sequences: Tolerance to Amino Acid Substitutions,” Science 247:1306-1310 (1990), wherein the authors indicate that there are two main strategies for studying the tolerance of an amino acid sequence to change. [0109]
  • The first strategy exploits the tolerance of amino acid substitutions by natural selection during the process of evolution. By comparing amino acid sequences in different species, conserved amino acids can be identified. These conserved amino acids are likely important for protein function. In contrast, the amino acid positions where substitutions have been tolerated by natural selection indicates that these positions are not critical for protein function. Thus, positions tolerating amino acid substitution could be modified while still maintaining biological activity of the protein. [0110]
  • The second strategy uses genetic engineering to introduce amino acid changes at specific positions of a cloned gene to identify regions critical for protein function. For example, site directed mutagenesis or alanine-scanning mutagenesis (introduction of single alanine mutations at every residue in the molecule) can be used. See Cunningham and Wells, Science 244:1081-1085 (1989). The resulting mutant molecules can then be tested for biological activity. [0111]
  • As the authors state, these two strategies have revealed that proteins are surprisingly tolerant of amino acid substitutions. The authors further indicate which amino acid changes are likely to be permissive at certain amino acid positions in the protein. For example, most buried (within the tertiary structure of the protein) amino acid residues require nonpolar side chains, whereas few features of surface side chains are generally conserved. Moreover, tolerated conservative amino acid substitutions involve replacement of the aliphatic or hydrophobic amino acids Ala, Val, Leu and Ile; replacement of the hydroxyl residues Ser and Thr; replacement of the acidic residues Asp and Glu; replacement of the amide residues Asn and Gln, replacement of the basic residues Lys, Arg, and His; replacement of the aromatic residues Phe, Tyr, and Trp, and replacement of the small-sized amino acids Ala, Ser, Thr, Met, and Gly. Besides conservative amino acid substitution, variants of the present invention include (i) polypeptides containing substitutions of one or more of the non-conserved amino acid residues, where the substituted amino acid residues may or may not be one encoded by the genetic code, or (ii) polypeptides containing substitutions of one or more of the amino acid residues having a substituent group, or (iii) polypeptides which have been fused with or chemically conjugated to another compound, such as a compound to increase the stability and/or solubility of the polypeptide (for example, polyethylene glycol), (iv) polypeptide containing additional amino acids, such as, for example, an IgG Fc fusion region peptide. Such variant polypeptides are deemed to be within the scope of those skilled in the art from the teachings herein. [0112]
  • For example, polypeptide variants containing amino acid substitutions of charged amino acids with other charged or neutral amino acids may produce proteins with improved characteristics, such as less aggregation. Aggregation of pharmaceutical formulations both reduces activity and increases clearance due to the aggregate's immunogenic activity. See Pinckard et al., Clin. Exp. Immunol. 2:331-340 (1967); Robbins et al., Diabetes 36: 838-845 (1987); Cleland et al., Crit. Rev. Therapeutic Drug Carrier Systems 10:307-377 (1993). [0113]
  • In specific embodiments, the polypeptides of the invention comprise, or alternatively, consist of, fragments or variants of the amino acid sequence of a Therapeutic protein described herein and/or human serum albumin, and/or albumin fusion protein of the invention, wherein the fragments or variants have 1-5, 5-10, 5-25, 5-50, 10-50 or 50-150, amino acid residue additions, substitutions, and/or deletions when compared to the reference amino acid sequence. In preferred embodiments, the amino acid substitutions are conservative. Nucleic acids encoding these polypeptides are also encompassed by the invention. [0114]
  • The polypeptide of the present invention can be composed of amino acids joined to each other by peptide bonds or modified peptide bonds, i.e., peptide isosteres, and may contain amino acids other than the 20 gene-encoded amino acids. The polypeptides may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. Such modifications are well described in basic texts and in more detailed monographs, as well as in a voluminous research literature. Modifications can occur anywhere in a polypeptide, including the peptide backbone, the amino acid side-chains and the amino or carboxyl termini. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Also, a given polypeptide may contain many types of modifications. Polypeptides may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. (See, for instance, PROTEINS—STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)). [0115]
  • Functional Activity [0116]
  • “A polypeptide having functional activity” refers to a polypeptide capable of displaying one or more known functional activities associated with the full-length, pro-protein, and/or mature form of a Therapeutic protein. Such functional activities include, but are not limited to, biological activity, antigenicity [ability to bind (or compete with a polypeptide for binding) to an anti-polypeptide antibody], immunogenicity (ability to generate antibody which binds to a specific polypeptide of the invention), ability to form multimers with polypeptides of the invention, and ability to bind to a receptor or ligand for a polypeptide. [0117]
  • “A polypeptide having biological activity” refers to a polypeptide exhibiting activity similar to, but not necessarily identical to, an activity of a Therapeutic protein of the present invention, including mature forms, as measured in a particular biological assay, with or without dose dependency. In the case where dose dependency does exist, it need not be identical to that of the polypeptide, but rather substantially similar to the dose-dependence in a given activity as compared to the polypeptide of the present invention (i.e., the candidate polypeptide will exhibit greater activity or not more than about 25-fold less and, preferably, not more than about tenfold less activity, and most preferably, not more than about three-fold less activity relative to the polypeptide of the present invention). [0118]
  • In preferred embodiments, an albumin fusion protein of the invention has at least one biological and/or therapeutic activity associated with the Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin. [0119]
  • The albumin fusion proteins of the invention can be assayed for functional activity (e.g., biological activity) using or routinely modifying assays known in the art, as well as assays described herein. Additionally, one of skill in the art may routinely assay fragments of a Therapeutic protein corresponding to a Therapeutic protein portion of an albumin fusion protein of the invention, for activity using assays referenced in its corresponding row of Table 1. Further, one of skill in the art may routinely assay fragments of an albumin protein corresponding to an albumin protein portion of an albumin fusion protein of the invention, for activity using assays known in the art and/or as described in the Examples section below. [0120]
  • For example, in one embodiment where one is assaying for the ability of an albumin fusion protein of the invention to bind or compete with a Therapeutic protein for binding to an anti-Therapeutic polypeptide antibody and/or anti-albumin antibody, various immunoassays known in the art can be used, including but not limited to, competitive and non-competitive assay systems using techniques such as radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoradiometric assays, gel diffusion precipitation reactions, immunodiffusion assays, in situ immunoassays (using colloidal gold, enzyme or radioisotope labels, for example), western blots, precipitation reactions, agglutination assays (e.g., gel agglutination assays, hemagglutination assays), complement fixation assays, immunofluorescence assays, protein A assays, and immunoelectrophoresis assays, etc. In one embodiment, antibody binding is detected by detecting a label on the primary antibody. In another embodiment, the primary antibody is detected by detecting binding of a secondary antibody or reagent to the primary antibody. In a further embodiment, the secondary antibody is labeled. Many means are known in the art for detecting binding in an immunoassay and are within the scope of the present invention. [0121]
  • In a preferred embodiment, where a binding partner (e.g., a receptor or a ligand) of a Therapeutic protein is identified, binding to that binding partner by an albumin fusion protein containing that Therapeutic protein as the Therapeutic protein portion of the fusion can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., Microbiol. Rev. 59:94-123 (1995). In another embodiment, the ability of physiological correlates of an albumin fusion protein of the present invention to bind to a substrate(s) of the Therapeutic polypeptide corresponding to the Therapeutic portion of the albumin fusion protein of the invention can be routinely assayed using techniques known in the art. [0122]
  • In an alternative embodiment, where the ability of an albumin fusion protein of the invention to multimerize is being evaluated, association with other components of the multimer can be assayed, e.g., by means well-known in the art, such as, for example, reducing and non-reducing gel chromatography, protein affinity chromatography, and affinity blotting. See generally, Phizicky et al., supra. [0123]
  • In preferred embodiments, an albumin fusion protein of the invention comprising all or a portion of an antibody that binds a Therapeutic protein, has at least one biological and/or therapeutic activity (e.g., to specifically bind a polypeptide or epitope) associated with the antibody that binds a Therapeutic protein (or fragment or variant thereof) when it is not fused to albumin. In other preferred embodiments, the biological activity and/or therapeutic activity of an albumin fusion protein of the invention comprising all or a portion of an antibody that binds a Therapeutic protein is the inhibition (i.e. antagonism) or activation (i.e., agonism) of one or more of the biological activities and/or therapeutic activities associated with the polypeptide that is specifically bound by antibody that binds a Therapeutic protein. [0124]
  • Albumin fusion proteins of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) may be characterized in a variety of ways. In particular, albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art. [0125]
  • Assays for the ability of the albumin fusion proteins of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Pat. No. 5,223,409), on spores (e.g., Patent Nos. 5,571,698; 5,403,484; and 5,223,409), on plasmids (e.g., Cull et al., Proc. Natl. Acad. Sci. USA 89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990); Devlin, Science 249:404-406 (1990); Cwirla et al., Proc. Natl. Acad. Sci. USA 87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)) (each of these references is incorporated herein in its entirety by reference). Albumin fusion proteins of the invention comprising at least a fragment or variant of a Therapeutic antibody may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art. [0126]
  • The albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art. [0127]
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation). [0128]
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding the albumin fusion protein of the invention (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C., adding sepharose beads coupled to an anti-albumin antibody, for example, to the cell lysate, incubating for about an hour or more at 40 degrees C., washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the albumin fusion protein of the invention to immunoprecipitate a particular antigen can be assessed by, e.g. western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10. 16.1. [0129]
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), applying the albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., [0130] 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the albumin fusion protein (e.g., comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the albumin fusion proteins specifically bound to the antigen coating the well. In ELISAs the albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes albumin fusion protein) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, the albumin fusion protein may be coated to the well. In this case, the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase). One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1. [0131]
  • The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., [0132] 3H or 125I) with the albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen. The affinity of the albumin fusion protein of the present invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the albumin fusion protein, can also be determined using radioimmunoassays. In this case, the protein, antigen or epitope is incubated with an albumin fusion protein of the present invention conjugated to a labeled compound (e.g., 3H or 125I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • In a preferred embodiment, BIAcore kinetic analysis is used to determine the binding on and off rates of albumin fusion proteins of the invention to a protein, antigen or epitope. BIAcore kinetic analysis comprises analyzing the binding and dissociation of albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes or albumin fusion proteins, respectively, on their surface. [0133]
  • Antibodies that bind a Therapeutic protein corresponding to the Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity for a given protein or antigen, preferably the antigen which they specifically bind. Preferred binding affinities include those with a dissociation constant or Kd less than 5×10[0134] −2 M, 10−2 M, 5×10−3 M, 10−3 M, 5×10−4 M, 10−4 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5×10−5 M, 10−5M, 5×10−6 M, 10−6M, 5×10−7 M, 107 M, 5×10−8 M or 10−8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5×10−9 M, 10−9 M, 5×10− M, 10−10 M, 5×10−11 M, 10−11 M, 5×10−12 M, 10−12 M, 5×10−13 M, 10−13 M, 5×10−14 M, 10−14 M, 5×10−15 M, or 10−15 M. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody. In addition, assays described herein (see Examples and Table 1) and otherwise known in the art may routinely be applied to measure the ability of albumin fusion proteins of the present invention and fragments, variants and derivatives thereof to elicit biological activity and/or Therapeutic activity (either in vitro or in vivo) related to either the Therapeutic protein portion and/or albumin portion of the albumin fusion protein of the present invention. Other methods will be known to the skilled artisan and are within the scope of the invention.
  • Albumin [0135]
  • As described above, an albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion or chemical conjugation. [0136]
  • The terms, human serum albumin (HSA) and human albumin (HA) are used interchangeably herein. The terms, “albumin and “serum albumin” are broader, and encompass human serum albumin (and fragments and variants thereof) as well as albumin from other species (and fragments and variants thereof). [0137]
  • As used herein, “albumin” refers collectively to albumin protein or amino acid sequence, or an albumin fragment or variant, having one or more functional activities (e.g., biological activities) of albumin. In particular, “albumin” refers to human albumin or fragments thereof (see [0138] EP 201 239, EP 322 094 WO 97/24445, WO95/23857) especially the mature form of human albumin as shown in FIG. 15 and SEQ ID NO: 18, or albumin from other vertebrates or fragments thereof, or analogs or variants of these molecules or fragments thereof.
  • In preferred embodiments, the human serum albumin protein used in the albumin fusion proteins of the invention contains one or both of the following sets of point mutations with reference to SEQ ID NO:18: Leu-407 to Ala, Leu-408 to Val, Val-409 to Ala, and Arg-410 to Ala; or Arg-410 to A, Lys-413 to Gln, and Lys-414 to Gln (see, e.g., International Publication No. WO95/23857, hereby incorporated in its entirety by reference herein). In even more preferred embodiments, albumin fusion proteins of the invention that contain one or both of above-described sets of point mutations have improved stability/resistance to yeast Yap3p proteolytic cleavage, allowing increased production of recombinant albumin fusion proteins expressed in yeast host cells. [0139]
  • As used herein, a portion of albumin sufficient to prolong the therapeutic activity or shelf-life of the Therapeutic protein refers to a portion of albumin sufficient in length or structure to stabilize or prolong the therapeutic activity of the protein so that the shelf life of the Therapeutic protein portion of the albumin fusion protein is prolonged or extended compared to the shelf-life in the non-fusion state. The albumin portion of the albumin fusion proteins may comprise the full length of the HA sequence as described above or as shown in FIG. 15, or may include one or more fragments thereof that are capable of stabilizing or prolonging the therapeutic activity. Such fragments may be of 10 or more amino acids in length or may include about 15, 20, 25, 30, 50, or more contiguous amino acids from the HA sequence or may include part or all of specific domains of HA. For instance, one or more fragments of HA spanning the first two immunoglobulin-like domains may be used. [0140]
  • The albumin portion of the albumin fusion proteins of the invention may be a variant of normal HA. The Therapeutic protein portion of the albumin fusion proteins of the invention may also be variants of the Therapeutic proteins as described herein. The term “variants” includes insertions, deletions and substitutions, either conservative or non conservative, where such changes do not substantially alter one or more of the oncotic, useful ligand-binding and non-immunogenic properties of albumin, or the active site, or active domain which confers the therapeutic activities of the Therapeutic proteins. [0141]
  • In particular, the albumin fusion proteins of the invention may include naturally occurring polymorphic variants of human albumin and fragments of human albumin, for example those fragments disclosed in EP 322 094 (namely HA (Pn), where n is 369 to 419). The albumin may be derived from any vertebrate, especially any mammal, for example human, cow, sheep, or pig. Non-mammalian albumins include, but are not limited to, hen and salmon. The albumin portion of the albumin fusion protein may be from a different animal than the Therapeutic protein portion. [0142]
  • Generally speaking, an HA fragment or variant will be at least 100 amino acids long, preferably at least 150 amino acids long. The HA variant may consist of or alternatively comprise at least one whole domain of HA, for example domains 1 (amino acids 1-194 of SEQ ID NO:18), 2 (amino acids 195-387 of SEQ ID NO:18), 3 (amino acids 388-585 of SEQ ID NO:18), 1+2 (1-387 of SEQ ID NO:18), 2+3 (195-585 of SEQ ID NO:18) or 1+3 (amino acids 1-194 of SEQ ID NO:18+amino acids 388-585 of SEQ ID NO:18). Each domain is itself made up of two homologous subdomains namely 1-105, 120-194, 195-291, 316-387, 388-491 and 512-585, with flexible inter-subdomain linker regions comprising residues Lys106 to Glu119, Glu292 to Val315 and Glu492 to Ala511. [0143]
  • Preferably, the albumin portion of an albumin fusion protein of the invention comprises at least one subdomain or domain of HA or conservative modifications thereof. If the fusion is based on subdomains, some or all of the adjacent linker is preferably used to link to the Therapeutic protein moiety. [0144]
  • Antibodies that Specifically Bind Therapeutic Proteins are also Therapeutic Proteins [0145]
  • The present invention also encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that specifically binds a Therapeutic protein disclosed in Table 1. It is specifically contemplated that the term “Therapeutic protein” encompasses antibodies that bind a Therapeutic protein (e.g., as Described in column I of Table 1) and fragments and variants thereof. Thus an albumin fusion protein of the invention may contain at least a fragment or variant of a Therapeutic protein, and/or at least a fragment or variant of an an antibody that binds a Therapeutic protein. [0146]
  • Antibody Structure and Background [0147]
  • The basic antibody structural unit is known to comprise a tetramer. Each tetramer is composed of two identical pairs of polypeptide chains, each pair having one “light” (about 25 kDa) and one “heavy” chain (about 50-70 kDa). The amino-terminal portion of each chain includes a variable region of about 100 to 110 or more amino acids primarily responsible for antigen recognition. The carboxy-terminal portion of each chain defines a constant region primarily responsible for effector function. Human light chains are classified as kappa and lambda light chains. Heavy chains are classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, IgD, 1gG, IgA, and IgE, respectively. See generally, [0148] Fundamental Immunology Chapters 3-5 (Paul, W., ed., 4th ed. Raven Press, N.Y. (1998)) (incorporated by reference in its entirety for all purposes). The variable regions of each light/heavy chain pair form the antibody binding site.
  • Thus, an intact IgG antibody has two binding sites. Except in bifunctional or bispecific antibodies, the two binding sites are the same. [0149]
  • The chains all exhibit the same general structure of relatively conserved framework regions (FR) joined by three hypervariable regions, also called complementarity determining regions or CDRs. The CDR regions, in general, are the portions of the antibody which make contact with the antigen and determine its specificity. The CDRs from the heavy and the light chains of each pair are aligned by the framework regions, enabling binding to a specific epitope. From N-terminal to C-terminal, both light and heavy chains variable regions comprise the domains FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. The variable regions are connected to the heavy or light chain constant region. The assignment of amino acids to each domain is in accordance with the definitions of Kabat [0150] Sequences of Proteins of Immunological Interest (National Institutes of Health, Bethesda, Md. (1987 and 1991)), or Chothia & Lesk J. Mol. Biol. 196:901-917 (1987); Chothia et al. Nature 342:878-883 (1989).
  • As used herein, “antibody” refers to immunoglobulin molecules and immunologically active portions of immunoglobulin molecules, i.e., molecules that contain an antigen binding site that specifically binds an antigen (e.g., a molecule containing one or more CDR regions of an antibody). Antibodies that may correspond to a Therapeutic protein portion of an albumin fusion protein include, but are not limited to, monoclonal, multispecific, human, humanized or chimeric antibodies, single chain antibodies (e.g., single chain Fvs), Fab fragments, F(ab′) fragments, fragments produced by a Fab expression library, anti-idiotypic (anti-Id) antibodies (including, e.g., anti-Id antibodies specific to antibodies of the invention), and epitope-binding fragments of any of the above (e.g., VH domains, VL domains, or one or more CDR regions). [0151]
  • Antibodies that Bind Therapeutic Proteins [0152]
  • The present invention encompasses albumin fusion proteins that comprise at least a fragment or variant of an antibody that binds a Therapeutic Protein (e.g., as disclosed in Table 1) or fragment or variant thereof. [0153]
  • Antibodies that bind a Therapeutic protein (or fragment or variant thereof) may be from any animal origin, including birds and mammals. Preferably, the antibodies are human, murine (e.g., mouse and rat), donkey, sheep, rabbit, goat, guinea pig, camel, horse, or chicken antibodies. Most preferably, the antibodies are human antibodies. As used herein, “human” antibodies include antibodies having the amino acid sequence of a human immunoglobulin and include antibodies isolated from human immunoglobulin libraries and xenomice or other organisms that have been genetically engineered to produce human antibodies. [0154]
  • The antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention can be of any type (e.g., IgG, IgE, IgM, IgD, IgA and IgY), class (e.g., IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2) or subclass of immunoglobulin molecule. In preferred embodiments, the antibody molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG1. In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG2. In other preferred embodiments, the immunoglobulin molecules that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are IgG4. [0155]
  • Most preferably the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention are human antigen-binding antibody fragments of the present invention and include, but are not limited to, Fab, Fab′ and F(ab′)[0156] 2, Fd, single-chain Fvs (scFv), single-chain antibodies, disulfide-linked Fvs (sdFv) and fragments comprising either a VL or VH domain. Antigen-binding antibody fragments, including single-chain antibodies, may comprise the variable region(s) alone or in combination with the entirety or a portion of the following: hinge region, CH1, CH2, and CH3 domains.
  • The antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be monospecific, bispecific, trispecific or of greater multispecificity. Multispecific antibodies may be specific for different epitopes of a Therapeutic protein or may be specific for both a Therapeutic protein as well as for a heterologous epitope, such as a heterologous polypeptide or solid support material. See, e.g., PCT publications WO 93/17715; WO 92/08802; WO 91/00360; WO 92/05793; Tutt, et al., J. Immunol. 147:60-69 (1991); U.S. Pat. Nos. 4,474,893; 4,714,681; 4,925,648; 5,573,920; 5,601,819; Kostelny et al., J. Immunol. 148:1547-1553 (1992). [0157]
  • Antibodies that bind a Therapeutic protein (or fragment or variant thereof) may be bispecific or bifunctional which means that the antibody is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. Bispecific antibodies can be produced by a variety of methods including fusion of hybridomas or linking of Fab′ fragments. See, e.g., Songsivilai & Lachmann [0158] Clin. Exp. Immunol. 79: 315-321 (1990), Kostelny et al. J. Immunol. 148:1547 1553 (1992). In addition, bispecific antibodies may be formed as “diabodies” (Holliger et al. “‘Diabodies’: small bivalent and bispecific antibody fragments” PNAS USA 90:6444-6448 (1993)) or “Janusins” (Traunecker et al. “Bispecific single chain molecules (Janusins) target cytotoxic lymphocytes on HIV infected cells” EMBO J 10:3655-3659 (1991) and Traunecker et al. “Janusin: new molecular design for bispecific reagents” Int J Cancer Suppl 7:51-52 (1992)).
  • The present invention also provides albumin fusion proteins that comprise, fragments or variants (including derivatives) of an antibody described herein or known elsewhere in the art. Standard techniques known to those of skill in the art can be used to introduce mutations in the nucleotide sequence encoding a molecule of the invention, including, for example, site-directed mutagenesis and PCR-mediated mutagenesis which result in amino acid substitutions. Preferably, the variants (including derivatives) encode less than 50 amino acid substitutions, less than 40 amino acid subsitutions, less than 30 amino acid substitutions, less than 25 amino acid substitutions, less than 20 amino acid substitutions, less than 15 amino acid substitutions, less than 10 amino acid substitutions, less than 5 amino acid substitutions, less than 4 amino acid substitutions, less than 3 amino acid substitutions, or less than 2 amino acid substitutions relative to the reference VH domain, VHCDR1, VHCDR2, VHCDR3, VL domain, VLCDR1, VLCDR2, or VLCDR3. In specific embodiments, the variants encode substitutions of VHCDR3. In a preferred embodiment, the variants have conservative amino acid substitutions at one or more predicted non-essential amino acid residues. [0159]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be described or specified in terms of the epitope(s) or portion(s) of a Therapeutic protein which they recognize or specifically bind. Antibodies which specifically bind a Therapeutic protein or a specific epitope of a Therapeutic protein may also be excluded. Therefore, the present invention encompasses antibodies that specifically bind Therapeutic proteins, and allows for the exclusion of the same. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, binds the same epitopes as the. [0160]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their cross-reactivity. Antibodies that do not bind any other analog, ortholog, or homolog of a Therapeutic protein are included. Antibodies that bind polypeptides with at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 65%, at least 60%, at least 55%, and at least 50% identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention. In specific embodiments, antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention cross-react with murine, rat and/or rabbit homologs of human proteins and the corresponding epitopes thereof. Antibodies that do not bind polypeptides with less than 95%, less than 90%, less than 85%, less than 80%, less than 75%, less than 70%, less than 65%, less than 60%, less than 55%, and less than 50% identity (as calculated using methods known in the art and described herein) to a Therapeutic protein are also included in the present invention. In a specific embodiment, the above-described cross-reactivity is with respect to any single specific antigenic or immunogenic polypeptide, or combination(s) of 2, 3, 4, 5, or more of the specific antigenic and/or immunogenic polypeptides disclosed herein. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially identical cross reactivity characteristics compared to the. [0161]
  • Further included in the present invention are antibodies which bind polypeptides encoded by polynucleotides which hybridize to a polynucleotide encoding a Therapeutic protein under stringent hybridization conditions (as described herein). Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may also be described or specified in terms of their binding affinity to a polypeptide of the invention. Preferred binding affinities include those with a dissociation constant or Kd less than 5×10[0162] −2 M, 10−2 M, 5×10−3 M, 10−3 M, 5×10−4 M, 10−4 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5×10−5 M, 10−5 M, 5×10−6 M, 10−6M, 5×10−7 M, 107 M, 5×10−8 M or 10−8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5×10−9 M, 10−9 M, 5×10−10 M, 10−10 M, 5×10−11 M, 10−11 M, 5×10−12 M, 10−12 M, 5×10−13 M, 10−13 M, 5×10−14 M, 10−14 M, 5×10−15 M, or 10−15 M. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has an affinity for a given protein or epitope similar to that of the corresponding antibody (not fused to albumin) that binds a Therapeutic protein, taking into account the valency of the albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) and the valency of the corresponding antibody.
  • The invention also provides antibodies that competitively inhibit binding of an antibody to an epitope of a Therapeutic protein as determined by any method known in the art for determining competitive binding, for example, the immunoassays described herein. In preferred embodiments, the antibody competitively inhibits binding to the epitope by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of an antibody to an epitope of a Therapeutic protein as well as the competitively inhibits binding of an antibody to an epitope of a Therapeutic protein. In other preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, competitively inhibits binding of the to an epitope of a Therapeutic protein by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50%. [0163]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may act as agonists or antagonists of the Therapeutic protein. For example, the present invention includes antibodies which disrupt the receptor/ligand interactions with the polypeptides of the invention either partially or fully. The invention features both receptor-specific antibodies and ligand-specific antibodies. The invention also features receptor-specific antibodies which do not prevent ligand binding but prevent receptor activation. Receptor activation (i.e., signaling) may be determined by techniques described herein or otherwise known in the art. For example, receptor activation can be determined by detecting the phosphorylation (e.g., tyrosine or serine/threonine) of the receptor or its substrate by immunoprecipitation followed by western blot analysis (for example, as described supra). In specific embodiments, antibodies are provided that inhibit ligand activity or receptor activity by at least 95%, at least 90%, at least 85%, at least 80%, at least 75%, at least 70%, at least 60%, or at least 50% of the activity in absence of the antibody. In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, has similar or substantially similar characteristics with regard to preventing ligand binding and/or preventing receptor activation compared to the. [0164]
  • The invention also features receptor-specific antibodies which both prevent ligand binding and receptor activation as well as antibodies that recognize the receptor-ligand complex, and, preferably, do not specifically recognize the unbound receptor or the unbound ligand. Likewise, included in the invention are neutralizing antibodies which bind the ligand and prevent binding of the ligand to the receptor, as well as antibodies which bind the ligand, thereby preventing receptor activation, but do not prevent the ligand from binding the receptor. Further included in the invention are antibodies which activate the receptor. These antibodies may act as receptor agonists, i.e., potentiate or activate either all or a subset of the biological activities of the ligand-mediated receptor activation, for example, by inducing dimerization of the receptor. The antibodies may be specified as agonists, antagonists or inverse agonists for biological activities comprising the specific biological activities of the Therapeutic protreins (e.g. as disclosed in Table 1). The above antibody agonists can be made using methods known in the art. See, e.g., PCT publication WO 96/40281; U.S. Pat. No. 5,811,097; Deng et al., Blood 92(6):1981-1988 (1998); Chen et al., Cancer Res. 58(16):3668-3678 (1998); Harrop et al., J. Immunol. 161(4):1786-1794 (1998); Zhu et al., Cancer Res. 58(15):3209-3214 (1998); Yoon et al., J. Immunol. 160(7):3170-3179 (1998); Prat et al., J. Cell. Sci. 111(Pt2):237-247 (1998); Pitard et al., J. Immunol. Methods 205(2):177-190 (1997); Liautard et al., Cytokine 9(4):233-241 (1997); Carlson et al., J. Biol. Chem. 272(17):11295-11301 (1997); Taryrnan et al., Neuron 14(4):755-762 (1995); Muller et al., Structure 6(9):1153-1167 (1998); Bartunek et al., Cytokine 8(1):14-20 (1996) (which are all incorporated by reference herein in their entireties). In preferred embodiments, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, have similar or substantially identical agonist or antagonist properties as the. [0165]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be used, for example, to purify, detect, and target Therapeutic proteins, including both in in vitro and in vivo diagnostic and therapeutic methods. For example, the antibodies have utility in immunoassays for qualitatively and quantitatively measuring levels of the Therapeutic protein in biological samples. See, e.g., Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); incorporated by reference herein in its entirety. Likewise, albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, may be used, for example, to purify, detect, and target Therapeutic proteins, including both in in vitro and in vivo diagnostic and therapeutic methods. [0166]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein include derivatives that are modified, i.e, by the covalent attachment of any type of molecule to the antibody. For example, but not by way of limitation, the antibody derivatives include antibodies that have been modified, e.g., by glycosylation, acetylation, pegylation, phosphylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to a cellular ligand or other protein, etc. Any of numerous chemical modifications may be carried out by known techniques, including, but not limited to specific chemical cleavage, acetylation, formylation, metabolic synthesis of tunicamycin, etc. Additionally, the derivative may contain one or more non-classical amino acids. Albumin fusion proteins of the invention may also be modified as described above. [0167]
  • Methods of Producing Antibodies that Bind Therapeutic Proteins [0168]
  • The antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention may be generated by any suitable method known in the art. Polyclonal antibodies to an antigen-of-interest can be produced by various procedures well known in the art. For example, a Therapeutic protein may be administered to various host animals including, but not limited to, rabbits, mice, rats, etc. to induce the production of sera containing polyclonal antibodies specific for the antigen. Various adjuvants may be used to increase the immunological response, depending on the host species, and include but are not limited to, Freund's (complete and incomplete), mineral gels such as aluminum hydroxide, surface active substances such as lysolecithin, pluronic polyols, polyanions, peptides, oil emulsions, keyhole limpet hemocyanins, dinitrophenol, and potentially useful human adjuvants such as BCG (bacille Calmette-Guerin) and corynebacterium parvum. Such adjuvants are also well known in the art. [0169]
  • Monoclonal antibodies can be prepared using a wide variety of techniques known in the art including the use of hybridoma, recombinant, and phage display technologies, or a combination thereof. For example, monoclonal antibodies can be produced using hybridoma techniques including those known in the art and taught, for example, in Harlow et al., Antibodies: A Laboratory Manual, (Cold Spring Harbor Laboratory Press, 2nd ed. 1988); Hammerling, et al., in: Monoclonal Antibodies and T-Cell Hybridomas 563-681 (Elsevier, N.Y., 1981) (said references incorporated by reference in their entireties). The term “monoclonal antibody” as used herein is not limited to antibodies produced through hybridoma technology. The term “monoclonal antibody” refers to an antibody that is derived from a single clone, including any eukaryotic, prokaryotic, or phage clone, and not the method by which it is produced. [0170]
  • Methods for producing and screening for specific antibodies using hybridoma technology are routine and well known in the art. In a non-limiting example, mice can be immunized with a Therapeutic protein or fragment or variant thereof or a cell expressing such a Therapeutic protein or fragment or variant thereof. Once an immune response is detected, e.g., antibodies specific for the antigen are detected in the mouse serum, the mouse spleen is harvested and splenocytes isolated. The splenocytes are then fused by well known techniques to any suitable myeloma cells, for example cells from cell line SP20 available from the ATCC. Hybridomas are selected and cloned by limited dilution. The hybridoma clones are then assayed by methods known in the art for cells that secrete antibodies capable of binding a polypeptide of the invention. Ascites fluid, which generally contains high levels of antibodies, can be generated by immunizing mice with positive hybridoma clones. [0171]
  • Accordingly, the present invention provides methods of generating monoclonal antibodies as well as antibodies produced by the method comprising culturing a hybridoma cell secreting an antibody wherein, preferably, the hybridoma is generated by fusing splenocytes isolated from a mouse immunized with an antigen of the invention with myeloma cells and then screening the hybridomas resulting from the fusion for hybridoma clones that secrete an antibody able to bind a polypeptide of the invention. [0172]
  • Another well known method for producing both polyclonal and monoclonal human B cell lines is transformation using Epstein Barr Virus (EBV). Protocols for generating EBV-transformed B cell lines are commonly known in the art, such as, for example, the protocol outlined in Chapter 7.22 of Current Protocols in Immunology, Coligan et al., Eds., 1994, John Wiley & Sons, NY, which is hereby incorporated in its entirety by reference. The source of B cells for transformation is commonly human peripheral blood, but B cells for transformation may also be derived from other sources including, but not limited to, lymph nodes, tonsil, spleen, tumor tissue, and infected tissues. Tissues are generally made into single cell suspensions prior to EBV transformation. Additionally, steps may be taken to either physically remove or inactivate T cells (e.g., by treatment with cyclosporin A) in B cell-containing samples, because T cells from individuals seropositive for anti-EBV antibodies can suppress B cell immortalization by EBV. [0173]
  • In general, the sample containing human B cells is innoculated with EBV, and cultured for 3-4 weeks. A typical source of EBV is the culture supernatant of the B95-8 cell line (ATCC #VR-1492). Physical signs of EBV transformation can generally be seen towards the end of the 3-4 week culture period. By phase-contrast microscopy, transformed cells may appear large, clear, hairy and tend to aggregate in tight clusters of cells. Initially, EBV lines are generally polyclonal. However, over prolonged periods of cell cultures, EBV lines may become monoclonal or polyclonal as a result of the selective outgrowth of particular B cell clones. Alternatively, polyclonal EBV transformed lines may be subcloned (e.g., by limiting dilution culture) or fused with a suitable fusion partner and plated at limiting dilution to obtain monoclonal B cell lines. Suitable fusion partners for EBV transformed cell lines include mouse myeloma cell lines (e.g., SP2/0, X63-Ag8.653), heteromyeloma cell lines (human×mouse; e.g, SPAM-8, SBC-H[0174] 2O, and CB-F7), and human cell lines (e.g., GM 1500, SKO-007, RPMI 8226, and KR-4). Thus, the present invention also provides a method of generating polyclonal or monoclonal human antibodies against polypeptides of the invention or fragments thereof, comprising EBV-transformation of human B cells.
  • Antibody fragments which recognize specific epitopes may be generated by known techniques. For example, Fab and F(ab′)[0175] 2 fragments of the invention may be produced by proteolytic cleavage of immunoglobulin molecules, using enzymes such as papain (to produce Fab fragments) or pepsin (to produce F(ab′)2 fragments). F(ab′)2 fragments contain the variable region, the light chain constant region and the CH1 domain of the heavy chain.
  • For example, antibodies that bind to a Therapeutic protein can also be generated using various phage display methods known in the art. In phage display methods, functional antibody domains are displayed on the surface of phage particles which carry the polynucleotide sequences encoding them. In a particular embodiment, such phage can be utilized to display antigen binding domains expressed from a repertoire or combinatorial antibody library (e.g., human or murine). Phage expressing an antigen binding domain that binds the antigen of interest can be selected or identified with antigen, e.g., using labeled antigen or antigen bound or captured to a solid surface or bead. Phage used in these methods are typically filamentous phage including fd and M13 binding domains expressed from phage with Fab, Fv or disulfide stabilized Fv antibody domains recombinantly fused to either the phage gene III or gene VIII protein. Examples of phage display methods that can be used to make antibodies that bind to a Therapeutic protein include those disclosed in Brinkman et al., J. Immunol. Methods 182:41-50 (1995); Ames et al., J. Immunol. Methods 184:177-186 (1995); Kettleborough et al., Eur. J. Immunol. 24:952-958 (1994); Persic et al., Gene 1879-18 (1997); Burton et al., Advances in Immunology 57:191-280 (1994); PCT application No. PCT/GB91/01134; PCT publications WO 90/02809; WO 91/10737; WO 92/01047; WO 92/18619; WO 93/11236; WO 95/15982; WO 95/20401; and U.S. Pat. Nos. 5,698,426; 5,223,409; 5,403,484; 5,580,717; 5,427,908; 5,750,753; 5,821,047; 5,571,698; 5,427,908; 5,516,637; 5,780,225; 5,658,727; 5,733,743 and 5,969,108; each of which is incorporated herein by reference in its entirety. [0176]
  • As described in the above references, after phage selection, the antibody coding regions from the phage can be isolated and used to generate whole antibodies, including human antibodies, or any other desired antigen binding fragment, and expressed in any desired host, including mammalian cells, insect cells, plant cells, yeast, and bacteria, e.g., as described in detail below. For example, techniques to recombinantly produce Fab, Fab′ and F(ab′)2 fragments can also be employed using methods known in the art such as those disclosed in PCT publication WO 92/22324; Mullinax et al., BioTechniques 12(6):864-869 (1992); and Sawai et al., AJRI 34:26-34 (1995); and Better et al., Science 240:1041-1043 (1988) (said references incorporated by reference in their entireties). [0177]
  • Examples of techniques which can be used to produce single-chain Fvs and antibodies include those described in U.S. Pat. Nos. 4,946,778 and 5,258,498; Huston et al., Methods in Enzymology 203:46-88 (1991); Shu et al., PNAS 90:7995-7999 (1993); and Skerra et al., Science 240:1038-1040 (1988). For some uses, including in vivo use of antibodies in humans and in vitro detection assays, it may be preferable to use chimeric, humanized, or human antibodies. A chimeric antibody is a molecule in which different portions of the antibody are derived from different animal species, such as antibodies having a variable region derived from a murine monoclonal antibody and a human immunoglobulin constant region. Methods for producing chimeric antibodies are known in the art. See e.g., Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Gillies et al., (1989) J. Immunol. Methods 125:191-202; U.S. Pat. Nos. 5,807,715; 4,816,567; and 4,816,397, which are incorporated herein by reference in their entirety. Humanized antibodies are antibody molecules from non-human species antibody that binds the desired antigen having one or more complementarity determining regions (CDRs) from the non-human species and a framework regions from a human immunoglobulin molecule. Often, framework residues in the human framework regions will be substituted with the corresponding residue from the CDR donor antibody to alter, preferably improve, antigen binding. These framework substitutions are identified by methods well known in the art, e.g., by modeling of the interactions of the CDR and framework residues to identify framework residues important for antigen binding and sequence comparison to identify unusual framework residues at particular positions. (See, e.g., Queen et al., U.S. Pat. No. 5,585,089; Riechmann et al., Nature 332:323 (1988), which are incorporated herein by reference in their entireties.) Antibodies can be humanized using a variety of techniques known in the art including, for example, CDR-grafting (EP 239,400; PCT publication WO 91/09967; U.S. Pat. Nos. 5,225,539; 5,530,101; and 5,585,089), veneering or resurfacing (EP 592,106; EP 519,596; Padlan, Molecular Immunology 28(4/5):489-498 (1991); Studnicka et al., Protein Engineering 7(6):805-814 (1994); Roguska. et al., PNAS 91:969-973 (1994)), and chain shuffling (U.S. Pat. No. 5,565,332). [0178]
  • Completely human antibodies are particularly desirable for therapeutic treatment of human patients. Human antibodies can be made by a variety of methods known in the art including phage display methods described above using antibody libraries derived from human immunoglobulin sequences. See also, U.S. Pat. Nos. 4,444,887 and 4,716,111; and PCT publications WO 98/46645, WO 98/50433, WO 98/24893, WO 98/16654, WO 96/34096, WO 96/33735, and WO 91/10741; each of which is incorporated herein by reference in its entirety. [0179]
  • Human antibodies can also be produced using transgenic mice which are incapable of expressing functional endogenous immunoglobulins, but which can express human immunoglobulin genes. For example, the human heavy and light chain immunoglobulin gene complexes may be introduced randomly or by homologous recombination into mouse embryonic stem cells. Alternatively, the human variable region, constant region, and diversity region may be introduced into mouse embryonic stem cells in addition to the human heavy and light chain genes. The mouse heavy and light chain immunoglobulin genes may be rendered non-functional separately or simultaneously with the introduction of human immunoglobulin loci by homologous recombination. In particular, homozygous deletion of the JH region prevents endogenous antibody production. The modified embryonic stem cells are expanded and microinjected into blastocysts to produce chimeric mice. The chimeric mice are then bred to produce homozygous offspring which express human antibodies. The transgenic mice are immunized in the normal fashion with a selected antigen, e.g., all or a portion of a polypeptide of the invention. Monoclonal antibodies directed against the antigen can be obtained from the immunized, transgenic mice using conventional hybridoma technology. The human immunoglobulin transgenes harbored by the transgenic mice rearrange during B cell differentiation, and subsequently undergo class switching and somatic mutation. Thus, using such a technique, it is possible to produce therapeutically useful IgG, IgA, IgM and IgE antibodies. For an overview of this technology for producing human antibodies, see Lonberg and Huszar, Int. Rev. Immunol. 13:65-93 (1995). For a detailed discussion of this technology for producing human antibodies and human monoclonal antibodies and protocols for producing such antibodies, see, e.g., PCT publications WO 98/24893; WO 92/01047; WO 96/34096; WO 96/33735; European Patent No. 0 598 877; U.S. Pat. Nos. 5,413,923; 5,625,126; 5,633,425; 5,569,825; 5,661,016; 5,545,806; 5,814,318; 5,885,793; 5,916,771; 5,939,598; 6,075,181; and 6,114,598, which are incorporated by reference herein in their entirety. In addition, companies such as Abgenix, Inc. (Freemont, Calif.) and Genpharm (San Jose, Calif.) can be engaged to provide human antibodies directed against a selected antigen using technology similar to that described above. [0180]
  • Completely human antibodies which recognize a selected epitope can be generated using a technique referred to as “guided selection.” In this approach a selected non-human monoclonal antibody, e.g., a mouse antibody, is used to guide the selection of a completely human antibody recognizing the same epitope. (Jespers et al., Bio/technology 12:899-903 (1988)). [0181]
  • Polynucleotides Encoding Antibodies [0182]
  • The invention further provides polynucleotides comprising a nucleotide sequence encoding an antibody and fragments thereof. The invention also encompasses polynucleotides that hybridize under stringent or alternatively, under lower stringency hybridization conditions, e.g., as defined supra, to polynucleotides that encode an antibody, preferably, that specifically binds to a Therapeutic protein, and more preferably, an antibody that binds to a polypeptide having the amino acid sequence of a “therapeutic protein X as discosed in the “Exemplay Identifier” column of Table 1. [0183]
  • The polynucleotides may be obtained, and the nucleotide sequence of the polynucleotides determined, by any method known in the art. For example, if the nucleotide sequence of the antibody is known, a polynucleotide encoding the antibody may be assembled from chemically synthesized oligonucleotides (e.g., as described in Kutmeier et al., BioTechniques 17:242 (1994)), which, briefly, involves the synthesis of overlapping oligonucleotides containing portions of the sequence encoding the antibody, annealing and ligating of those oligonucleotides, and then amplification of the ligated oligonucleotides by PCR. [0184]
  • Alternatively, a polynucleotide encoding an antibody may be generated from nucleic acid from a suitable source. If a clone containing a nucleic acid encoding a particular antibody is not available, but the sequence of the antibody molecule is known, a nucleic acid encoding the immunoglobulin may be chemically synthesized or obtained from a suitable source (e.g., an antibody cDNA library, or a cDNA library generated from, or nucleic acid, preferably poly A+ RNA, isolated from, any tissue or cells expressing the antibody, such as hybridoma cells selected to express an antibody) by PCR amplification using synthetic primers hybridizable to the 3′ and 5′ ends of the sequence or by cloning using an oligonucleotide probe specific for the particular gene sequence to identify, e.g., a cDNA clone from a cDNA library that encodes the antibody. Amplified nucleic acids generated by PCR may then be cloned into replicable cloning vectors using any method well known in the art (See Example 60). [0185]
  • Once the nucleotide sequence and corresponding amino acid sequence of the antibody is determined, the nucleotide sequence of the antibody may be manipulated using methods well known in the art for the manipulation of nucleotide sequences, e.g., recombinant DNA techniques, site directed mutagenesis, PCR, etc. (see, for example, the techniques described in Sambrook et al., 1990, Molecular Cloning, A Laboratory Manual, 2d Ed., Cold Spring Harbor Laboratory, Cold Spring Harbor, N.Y. and Ausubel et al., eds., 1998, Current Protocols in Molecular Biology, John Wiley & Sons, NY, which are both incorporated by reference herein in their entireties), to generate antibodies having a different amino acid sequence, for example to create amino acid substitutions, deletions, and/or insertions. [0186]
  • In a specific embodiment, the amino acid sequence of the heavy and/or light chain variable domains may be inspected to identify the sequences of the complementarity determining regions (CDRs) by methods that are well know in the art, e.g., by comparison to known amino acid sequences of other heavy and light chain variable regions to determine the regions of sequence hypervariability. Using routine recombinant DNA techniques, one or more of the CDRs may be inserted within framework regions, e.g., into human framework regions to humanize a non-human antibody, as described supra. The framework regions may be naturally occurring or consensus framework regions, and preferably human framework regions (see, e.g., Chothia et al., J. Mol. Biol. 278: 457-479 (1998) for a listing of human framework regions). Preferably, the polynucleotide generated by the combination of the framework regions and CDRs encodes an antibody that specifically binds a polypeptide of the invention. Preferably, as discussed supra, one or more amino acid substitutions may be made within the framework regions, and, preferably, the amino acid substitutions improve binding of the antibody to its antigen. Additionally, such methods may be used to make amino acid substitutions or deletions of one or more variable region cysteine residues participating in an intrachain disulfide bond to generate antibody molecules lacking one or more intrachain disulfide bonds. Other alterations to the polynucleotide are encompassed by the present invention and within the skill of the art. [0187]
  • In addition, techniques developed for the production of “chimeric antibodies” (Morrison et al., Proc. Natl. Acad. Sci. 81:851-855 (1984); Neuberger et al., Nature 312:604-608 (1984); Takeda et al., Nature 314:452-454 (1985)) by splicing genes from a mouse antibody molecule of appropriate antigen specificity together with genes from a human antibody molecule of appropriate biological activity can be used. As described supra, a chimeric antibody is a molecule in which different portions are derived from different animal species, such as those having a variable region derived from a murine mAb and a human immunoglobulin constant region, e.g., humanized antibodies. [0188]
  • Alternatively, techniques described for the production of single chain antibodies (U.S. Pat. No. 4,946,778; Bird, Science 242:423-42 (1988); Huston et al., Proc. Natl. Acad. Sci. USA 85:5879-5883 (1988); and Ward et al., Nature 334:544-54 (1989)) can be adapted to produce single chain antibodies. Single chain antibodies are formed by linking the heavy and light chain fragments of the Fv region via an amino acid bridge, resulting in a single chain polypeptide. Techniques for the assembly of functional Fv fragments in [0189] E. coli may also be used (Skerra et al., Science 242:1038-1041 (1988)).
  • Recombinant Expression of Antibodies [0190]
  • Recombinant expression of an antibody, or fragment, derivative or analog thereof, (e.g., a heavy or light chain of an antibody or a single chain antibody), requires construction of an expression vector containing a polynucleotide that encodes the antibody. Once a polynucleotide encoding an antibody molecule or a heavy or light chain of an antibody, or portion thereof (preferably containing the heavy or light chain variable domain), of the invention has been obtained, the vector for the production of the antibody molecule may be produced by recombinant DNA technology using techniques well known in the art. Thus, methods for preparing a protein by expressing a polynucleotide containing an antibody encoding nucleotide sequence are described herein. Methods which are well known to those skilled in the art can be used to construct expression vectors containing antibody coding sequences and appropriate transcriptional and translational control signals. These methods include, for example, in vitro recombinant DNA techniques, synthetic techniques, and in vivo genetic recombination. The invention, thus, provides replicable vectors comprising a nucleotide sequence encoding an antibody molecule of the invention, or a heavy or light chain thereof, or a heavy or light chain variable domain, operably linked to a promoter. Such vectors may include the nucleotide sequence encoding the constant region of the antibody molecule (see, e.g., PCT Publication WO 86/05807; PCT Publication WO 89/01036; and U.S. Pat. No. 5,122,464) and the variable domain of the antibody may be cloned into such a vector for expression of the entire heavy or light chain. [0191]
  • The expression vector is transferred to a host cell by conventional techniques and the transfected cells are then cultured by conventional techniques to produce an antibody. Thus, the invention includes host cells containing a polynucleotide encoding an antibody of the invention, or a heavy or light chain thereof, or a single chain antibody, operably linked to a heterologous promoter. In preferred embodiments for the expression of double-chained antibodies, vectors encoding both the heavy and light chains may be co-expressed in the host cell for expression of the entire immunoglobulin molecule, as detailed below. [0192]
  • A variety of host-expression vector systems may be utilized to express the antibody molecules of the invention. Such host-expression systems represent vehicles by which the coding sequences of interest may be produced and subsequently purified, but also represent cells which may, when transformed or transfected with the appropriate nucleotide coding sequences, express an antibody molecule of the invention in situ. These include but are not limited to microorganisms such as bacteria (e.g., [0193] E. coli, B. subtilis) transformed with recombinant bacteriophage DNA, plasmid DNA or cosmid DNA expression vectors containing antibody coding sequences; yeast (e.g., Saccharomyces, Pichia) transformed with recombinant yeast expression vectors containing antibody coding sequences; insect cell systems infected with recombinant virus expression vectors (e.g., baculovirus) containing antibody coding sequences; plant cell systems infected with recombinant virus expression vectors (e.g., cauliflower mosaic virus, CAMV; tobacco mosaic virus, TMV) or transformed with recombinant plasmid expression vectors (e.g., Ti plasmid) containing antibody coding sequences; or mammalian cell systems (e.g., COS, CHO, BHK, 293, 3T3 cells) harboring recombinant expression constructs containing promoters derived from the genome of mammalian cells (e.g., metallothionein promoter) or from mammalian viruses (e.g., the adenovirus late promoter; the vaccinia virus 7.5K promoter). Preferably, bacterial cells such as Escherichia coli, and more preferably, eukaryotic cells, especially for the expression of whole recombinant antibody molecule, are used for the expression of a recombinant antibody molecule. For example, mammalian cells such as Chinese hamster ovary cells (CHO), in conjunction with a vector such as the major intermediate early gene promoter element from human cytomegalovirus is an effective expression system for antibodies (Foecking et al., Gene 45:101 (1986); Cockett et al., Bio/Technology 8:2 (1990)).
  • In bacterial systems, a number of expression vectors may be advantageously selected depending upon the use intended for the antibody molecule being expressed. For example, when a large quantity of such a protein is to be produced, for the generation of pharmaceutical compositions of an antibody molecule, vectors which direct the expression of high levels of fusion protein products that are readily purified may be desirable. Such vectors include, but are not limited, to the [0194] E. coli expression vector pUR278 (Ruther et al., EMBO J. 2:1791 (1983)), in which the antibody coding sequence may be ligated individually into the vector in frame with the lac Z coding region so that a fusion protein is produced; pIN vectors (Inouye & Inouye, Nucleic Acids Res. 13:3101-3109 (1985); Van Heeke & Schuster, J. Biol. Chem. 24:5503-5509 (1989)); and the like. pGEX vectors may also be used to express foreign polypeptides as fusion proteins with glutathione S-transferase (GST). In general, such fusion proteins are soluble and can easily be purified from lysed cells by adsorption and binding to matrix glutathione-agarose beads followed by elution in the presence of free glutathione. The pGEX vectors are designed to include thrombin or factor Xa protease cleavage sites so that the cloned target gene product can be released from the GST moiety.
  • In an insect system, [0195] Autographa californica nuclear polyhedrosis virus (AcNPV) is used as a vector to express foreign genes. The virus grows in Spodoptera frugiperda cells. The antibody coding sequence may be cloned individually into non-essential regions (for example the polyhedrin gene) of the virus and placed under control of an AcNPV promoter (for example the polyhedrin promoter).
  • In mammalian host cells, a number of viral-based expression systems may be utilized. In cases where an adenovirus is used as an expression vector, the antibody coding sequence of interest may be ligated to an adenovirus transcription/translation control complex, e.g., the late promoter and tripartite leader sequence. This chimeric gene may then be inserted in the adenovirus genome by in vitro or in vivo recombination. Insertion in a non-essential region of the viral genome (e.g., region E1 or E3) will result in a recombinant virus that is viable and capable of expressing the antibody molecule in infected hosts. (e.g., see Logan & Shenk, Proc. Natl. Acad. Sci. USA 81:355-359 (1984)). Specific initiation signals may also be required for efficient translation of inserted antibody coding sequences. These signals include the ATG initiation codon and adjacent sequences. Furthermore, the initiation codon must be in phase with the reading frame of the desired coding sequence to ensure translation of the entire insert. These exogenous translational control signals and initiation codons can be of a variety of origins, both natural and synthetic. The efficiency of expression may be enhanced by the inclusion of appropriate transcription enhancer elements, transcription terminators, etc. (see Bittner et al., Methods in Enzymol. 153:51-544 (1987)). [0196]
  • In addition, a host cell strain may be chosen which modulates the expression of the inserted sequences, or modifies and processes the gene product in the specific fashion desired. Such modifications (e.g., glycosylation) and processing (e.g., cleavage) of protein products may be important for the function of the protein. Different host cells have characteristic and specific mechanisms for the post-translational processing and modification of proteins and gene products. Appropriate cell lines or host systems can be chosen to ensure the correct modification and processing of the foreign protein expressed. To this end, eukaryotic host cells which possess the cellular machinery for proper processing of the primary transcript, glycosylation, and phosphorylation of the gene product may be used. Such mammalian host cells include but are not limited to CHO, VERY, BHK, Hela, COS, MDCK, 293, 3T3, WI38, and in particular, breast cancer cell lines such as, for example, BT483, Hs578T, HTB2, BT20 and T47D, and normal mammary gland cell line such as, for example, CRL7030 and Hs578Bst. [0197]
  • For long-term, high-yield production of recombinant proteins, stable expression is preferred. For example, cell lines which stably express the antibody molecule may be engineered. Rather than using expression vectors which contain viral origins of replication, host cells can be transformed with DNA controlled by appropriate expression control elements (e.g., promoter, enhancer, sequences, transcription terminators, polyadenylation sites, etc.), and a selectable marker. Following the introduction of the foreign DNA, engineered cells may be allowed to grow for 1-2 days in an enriched media, and then are switched to a selective media. The selectable marker in the recombinant plasmid confers resistance to the selection and allows cells to stably integrate the plasmid into their chromosomes and grow to form foci which in turn can be cloned and expanded into cell lines. This method may advantageously be used to engineer cell lines which express the antibody molecule. Such engineered cell lines may be particularly useful in screening and evaluation of compounds that interact directly or indirectly with the antibody molecule. [0198]
  • A number of selection systems may be used, including but not limited to the herpes simplex virus thymidine kinase (Wigler et al., Cell 11:223 (1977)), hypoxanthine-guanine phosphoribosyltransferase (Szybalska & Szybalski, Proc. Natl. Acad. Sci. USA 48:202 (1992)), and adenine phosphoribosyltransferase (Lowy et al., Cell 22:817 (1980)) genes can be employed in tk-, hgprt- or aprt-cells, respectively. Also, antimetabolite resistance can be used as the basis of selection for the following genes: dhfr, which confers resistance to methotrexate (Wigler et al., Natl. Acad. Sci. USA 77:357 (1980); O'Hare et al., Proc. Natl. Acad. Sci. USA 78:1527 (1981)); gpt, which confers resistance to mycophenolic acid (Mulligan & Berg, Proc. Natl. Acad. Sci. USA 78:2072 (1981)); neo, which confers resistance to the aminoglycoside G-418 Clinical Pharmacy 12:488-505; Wu and Wu, Biotherapy 3:87-95 (1991); Tolstoshev, Ann. Rev. Pharmacol. Toxicol. 32:573-596 (1993); Mulligan, Science 260:926-932 (1993); and Morgan and Anderson, Ann. Rev. Biochem. 62:191-217 (1993); May, 1993, TIB TECH 11(5):155-215 (1993)); and hygro, which confers resistance to hygromycin (Santerre et al., Gene 30:147 (1984)). Methods commonly known in the art of recombinant DNA technology may be routinely applied to select the desired recombinant clone, and such methods are described, for example, in Ausubel et al. (eds.), Current Protocols in Molecular Biology, John Wiley & Sons, NY (1993); Kriegler, Gene Transfer and Expression, A Laboratory Manual, Stockton Press, NY (1990); and in Chapters 12 and 13, Dracopoli et al. (eds), Current Protocols in Human Genetics, John Wiley & Sons, NY (1994); Colberre-Garapin et al., J. Mol. Biol. 150:1 (1981), which are incorporated by reference herein in their entireties. [0199]
  • The expression levels of an antibody molecule can be increased by vector amplification (for a review, see Bebbington and Hentschel, The use of vectors based on gene amplification for the expression of cloned genes in mammalian cells in DNA cloning, Vol.3. (Academic Press, New York, 1987)). When a marker in the vector system expressing antibody is amplifiable, increase in the level of inhibitor present in culture of host cell will increase the number of copies of the marker gene. Since the amplified region is associated with the antibody gene, production of the antibody will also increase (Crouse et al., Mol. Cell. Biol. 3:257 (1983)). [0200]
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NS0) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g. Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657 which are incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors that may be used according to the present invention are commercially available from suppliers, including, for example Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al, [0201] Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are incorporated in their entirities by reference herein.
  • The host cell may be co-transfected with two expression vectors of the invention, the first vector encoding a heavy chain derived polypeptide and the second vector encoding a light chain derived polypeptide. The two vectors may contain identical selectable markers which enable equal expression of heavy and light chain polypeptides. Alternatively, a single vector may be used which encodes, and is capable of expressing, both heavy and light chain polypeptides. In such situations, the light chain should be placed before the heavy chain to avoid an excess of toxic free heavy chain (Proudfoot, Nature 322:52 (1986); Kohler, Proc. Natl. Acad. Sci. USA 77:2197 (1980)). The coding sequences for the heavy and light chains may comprise cDNA or genomic DNA. [0202]
  • Once an antibody molecule of the invention has been produced by an animal, chemically synthesized, or recombinantly expressed, it may be purified by any method known in the art for purification of an immunoglobulin molecule, for example, by chromatography (e.g., ion exchange, affinity, particularly by affinity for the specific antigen after Protein A, and sizing column chromatography), centrifugation, differential solubility, or by any other standard technique for the purification of proteins. In addition, the antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention or fragments thereof can be fused to heterologous polypeptide sequences described herein or otherwise known in the art, to facilitate purification. [0203]
  • Modifications of Antibodies [0204]
  • Antibodies that bind a Therapeutic protein or fragments or variants can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311), among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the “HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the “flag” tag. [0205]
  • The present invention further encompasses antibodies or fragments thereof conjugated to a diagnostic or therapeutic agent. The antibodies can be used diagnostically to, for example, monitor the development or progression of a tumor as part of a clinical testing procedure to, e.g., determine the efficacy of a given treatment regimen. Detection can be facilitated by coupling the antibody to a detectable substance. Examples of detectable substances include various enzymes, prosthetic groups, fluorescent materials, luminescent materials, bioluminescent materials, radioactive materials, positron emitting metals using various positron emission tomographies, and nonradioactive paramagnetic metal ions. The detectable substance may be coupled or conjugated either directly to the antibody (or fragment thereof) or indirectly, through an intermediate (such as, for example, a linker known in the art) using techniques known in the art. See, for example, U.S. Pat. No. 4,741,900 for metal ions which can be conjugated to antibodies for use as diagnostics according to the present invention. Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include 125I, 131I, 111In or 99Tc. Other examples of detectable substances have been described elsewwhere herein. [0206]
  • Further, an antibody of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine). [0207]
  • The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, β-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al., [0208] Int. Immunol., 6:1567-1574 (1994)), VEGI (See, International Publication No. WO 99/23105), a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors.
  • Antibodies may also be attached to solid supports, which are particularly useful for immunoassays or purification of the target antigen. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene. [0209]
  • Techniques for conjugating such therapeutic moiety to antibodies are well known. See, for example, Arnon et al., “Monoclonal Antibodies For Immunotargeting Of Drugs In Cancer Therapy”, in Monoclonal Antibodies And Cancer Therapy, Reisfeld et al. (eds.), pp. 243-56 (Alan R. Liss, Inc. 1985); Hellstrom et al., “Antibodies For Drug Delivery”, in Controlled Drug Delivery (2nd Ed.), Robinson et al. (eds.), pp. 623-53 (Marcel Dekker, Inc. 1987); Thorpe, “Antibody Carriers Of Cytotoxic Agents In Cancer Therapy: A Review”, in Monoclonal Antibodies '84: Biological And Clinical Applications, Pinchera et al. (eds.), pp. 475-506 (1985); “Analysis, Results, And Future Prospective Of The Therapeutic Use Of Radiolabeled Antibody In Cancer Therapy”, in Monoclonal Antibodies For Cancer Detection And Therapy, Baldwin et al. (eds.), pp. 303-16 (Academic Press 1985), and Thorpe et al., “The Preparation And Cytotoxic Properties Of Antibody-Toxin Conjugates”, Immunol. Rev. 62:119-58 (1982). [0210]
  • Alternatively, an antibody can be conjugated to a second antibody to form an antibody heteroconjugate as described by Segal in U.S. Pat. No. 4,676,980, which is incorporated herein by reference in its entirety. [0211]
  • An antibody, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic. [0212]
  • Antibody-Albumin Fusion [0213]
  • Antibodies that bind to a Therapeutic protein and that may correspond to a Therapeutic protein portion of an albumin fusion protein of the invention include, but are not limited to, antibodies that bind a Therapeutic protein disclosed in the “Therapeutic Protein X” column of Table 1, or a fragment or variant thereof. [0214]
  • In specific embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH domain. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VH CDRs. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of the VH CDR1. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VH CDR3. [0215]
  • In specific embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL domain. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two or three VL CDRs. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR1. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR2. In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, the VL CDR3. [0216]
  • In other embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, one, two, three, four, five, or six VH and/or VL CDRs. [0217]
  • In preferred embodiments, the fragment or variant of an antibody that immunospecifcally binds a Therapeutic protein and that corresponds to a Therapeutic protein portion of an albumin fusion protein comprises, or alternatively consists of, an scFv comprising the VH domain of the Therapeutic antibody, linked to the VL domain of the therapeutic antibody by a peptide linker such as (Gly[0218] 4Ser)3 (SEQ ID NO:36).
  • Immunophenotyping [0219]
  • The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be utilized for immunophenotyping of cell lines and biological samples. Therapeutic proteins of the present invention may be useful as cell-specific markers, or more specifically as cellular markers that are differentially expressed at various stages of differentiation and/or maturation of particular cell types. Monoclonal antibodies (or albumin fusion proteins comprsing at least a fragment or variant of an antibody that binds a Therapeutic protein) directed against a specific epitope, or combination of epitopes, will allow for the screening of cellular populations expressing the marker. Various techniques can be utilized using monoclonal antibodies (or albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) to screen for cellular populations expressing the marker(s), and include magnetic separation using antibody-coated magnetic beads, “panning” with antibody attached to a solid matrix (i.e., plate), and flow cytometry (See, e.g., U.S. Pat. No. 5,985,660; and Morrison et al., [0220] Cell, 96:737-49 (1999)).
  • These techniques allow for the screening of particular populations of cells, such as might be found with hematological malignancies (i.e. minimal residual disease (MRD) in acute leukemic patients) and “non-self” cells in transplantations to prevent Graft-versus-Host Disease (GVHD). Alternatively, these techniques allow for the screening of hematopoietic stem and progenitor cells capable of undergoing proliferation and/or differentiation, as might be found in human umbilical cord blood. [0221]
  • Characterizing Antibodies that Bind a Therapeutic Protein and Albumin Fusion Proteins Comprising a Fragment or Variant of an Antibody that Binds a Therapeutic Protein [0222]
  • The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may be characterized in a variety of ways. In particular, Albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for the ability to specifically bind to the same antigens specifically bound by the antibody that binds a Therapeutic protein corresponding to the antibody that binds a Therapeutic protein portion of the albumin fusion protein using techniques described herein or routinely modifying techniques known in the art. [0223]
  • Assays for the ability of the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to (specifically) bind a specific protein or epitope may be performed in solution (e.g., Houghten, Bio/Techniques 13:412-421(1992)), on beads (e.g., Lam, Nature 354:82-84 (1991)), on chips (e.g., Fodor, Nature 364:555-556 (1993)), on bacteria (e.g., U.S. Pat. No. 5,223,409), on spores (e.g., Patent Nos. 5,571,698; 5,403,484; and 5,223,409), on plasmids (e.g., Cull et al., Proc. Natl. Acad. Sci. USA 89:1865-1869 (1992)) or on phage (e.g., Scott and Smith, Science 249:386-390 (1990); Devlin, Science 249:404-406 (1990); Cwirla et al., Proc. Natl. Acad. Sci. USA 87:6378-6382 (1990); and Felici, J. Mol. Biol. 222:301-310 (1991)) (each of these references is incorporated herein in its entirety by reference). The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) may also be assayed for their specificity and affinity for a specific protein or epitope using or routinely modifying techniques described herein or otherwise known in the art. [0224]
  • The albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be assayed for cross-reactivity with other antigens (e.g., molecules that have sequence/structure conservation with the molecule(s) specifically bound by the antibody that binds a Therapeutic protein (or fragment or variant thereof) corresponding to the Therapeutic protein portion of the albumin fusion protein of the invention) by any method known in the art. [0225]
  • Immunoassays which can be used to analyze (immunospecific) binding and cross-reactivity include, but are not limited to, competitive and non-competitive assay systems using techniques such as western blots, radioimmunoassays, ELISA (enzyme linked immunosorbent assay), “sandwich” immunoassays, immunoprecipitation assays, precipitin reactions, gel diffusion precipitin reactions, immunodiffusion assays, agglutination assays, complement-fixation assays, immunoradiometric assays, fluorescent immunoassays, and protein A immunoassays, to name but a few. Such assays are routine and well known in the art (see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York, which is incorporated by reference herein in its entirety). Exemplary immunoassays are described briefly below (but are not intended by way of limitation). [0226]
  • Immunoprecipitation protocols generally comprise lysing a population of cells in a lysis buffer such as RIPA buffer (1% NP-40 or Triton X-100, 1% sodium deoxycholate, 0.1% SDS, 0.15 M NaCl, 0.01 M sodium phosphate at pH 7.2, 1% Trasylol) supplemented with protein phosphatase and/or protease inhibitors (e.g., EDTA, PMSF, aprotinin, sodium vanadate), adding an antibody of the invention or albumin fusion protein of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein (or fragment or variant thereof) to the cell lysate, incubating for a period of time (e.g., 1 to 4 hours) at 40 degrees C., adding protein A and/or protein G sepharose beads (or beads coated with an appropriate anti-iditoypic antibody or anti-albumin antibody in the case when an albumin fusion protein comprising at least a fragment or variant of a Therapeutic antibody) to the cell lysate, incubating for about an hour or more at 40 degrees C., washing the beads in lysis buffer and resuspending the beads in SDS/sample buffer. The ability of the antibody or albumin fusion protein of the invention to immunoprecipitate a particular antigen can be assessed by, e.g., western blot analysis. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the binding of the antibody or albumin fusion protein to an antigen and decrease the background (e.g., pre-clearing the cell lysate with sepharose beads). For further discussion regarding immunoprecipitation protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.16.1. [0227]
  • Western blot analysis generally comprises preparing protein samples, electrophoresis of the protein samples in a polyacrylamide gel (e.g., 8%-20% SDS-PAGE depending on the molecular weight of the antigen), transferring the protein sample from the polyacrylamide gel to a membrane such as nitrocellulose, PVDF or nylon, blocking the membrane in blocking solution (e.g., PBS with 3% BSA or non-fat milk), washing the membrane in washing buffer (e.g., PBS-Tween 20), applying the antibody or albumin fusion protein of the invention (diluted in blocking buffer) to the membrane, washing the membrane in washing buffer, applying a secondary antibody (which recognizes the albumin fusion protein, e.g., an anti-human serum albumin antibody) conjugated to an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) or radioactive molecule (e.g., [0228] 32P or 125I) diluted in blocking buffer, washing the membrane in wash buffer, and detecting the presence of the antigen. One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected and to reduce the background noise. For further discussion regarding western blot protocols see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 10.8.1.
  • ELISAs comprise preparing antigen, coating the well of a 96-well microtiter plate with the antigen, washing away antigen that did not bind the wells, adding the antibody or albumin fusion protein (comprising at least a fragment or variant of an antibody that binds a Therapeutic protein) of the invention conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase) to the wells and incubating for a period of time, washing away unbound or non-specifically bound albumin fusion proteins, and detecting the presence of the antibody or albumin fusion proteins specifically bound to the antigen coating the well. In ELISAs the antibody or albumin fusion protein does not have to be conjugated to a detectable compound; instead, a second antibody (which recognizes the antibody or albumin fusion protein, respectively) conjugated to a detectable compound may be added to the well. Further, instead of coating the well with the antigen, antibody or the albumin fusion protein may be coated to the well. In this case, the detectable molecule could be the antigen conjugated to a detectable compound such as an enzymatic substrate (e.g., horseradish peroxidase or alkaline phosphatase). One of skill in the art would be knowledgeable as to the parameters that can be modified to increase the signal detected as well as other variations of ELISAs known in the art. For further discussion regarding ELISAs see, e.g., Ausubel et al, eds, 1994, Current Protocols in Molecular Biology, Vol. 1, John Wiley & Sons, Inc., New York at 11.2.1. [0229]
  • The binding affinity of an albumin fusion protein to a protein, antigen, or epitope and the off-rate of an antibody- or albumin fusion protein-protein/antigen/epitope interaction can be determined by competitive binding assays. One example of a competitive binding assay is a radioimmunoassay comprising the incubation of labeled antigen (e.g., [0230] 3H or 125I) with the antibody or albumin fusion protein of the invention in the presence of increasing amounts of unlabeled antigen, and the detection of the antibody bound to the labeled antigen. The affinity of the antibody or albumin fusion protein of the present invention for a specific protein, antigen, or epitope and the binding off-rates can be determined from the data by Scatchard plot analysis. Competition with a second protein that binds the same protein, antigen or epitope as the antibody or albumin fusion protein, can also be determined using radioimmunoassays. In this case, the protein, antigen or epitope is incubated with an antibody or albumin fusion protein of the present invention conjugated to a labeled compound (e.g. 3H or 125I) in the presence of increasing amounts of an unlabeled second protein that binds the same protein, antigen, or epitope as the albumin fusion protein of the invention.
  • In a preferred embodiment, BIAcore kinetic analysis is used to determine the binding on and off rates of antibody or albumin fusion proteins of the invention to a protein, antigen or epitope. BIAcore kinetic analysis comprises analyzing the binding and dissociation of antibodies, albumin fusion proteins, or specific polypeptides, antigens or epitopes from chips with immobilized specific polypeptides, antigens or epitopes, antibodies or albumin fusion proteins, respectively, on their surface. [0231]
  • Therapeutic Uses [0232]
  • The present invention is further directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more of the disclosed diseases, disorders, or conditions. Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (including fragments, analogs and derivatives thereof as described herein), nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein), albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein, and nucleic acids encoding such albumin fusion proteins. The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein. [0233]
  • In a specific and preferred embodiment, the present invention is directed to antibody-based therapies which involve administering antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein to an animal, preferably a mammal, and most preferably a human, patient for treating one or more diseases, disorders, or conditions, including but not limited to: neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions., and/or as described elsewhere herein. Therapeutic compounds of the invention include, but are not limited to, antibodies of the invention (e.g., antibodies directed to the full length protein expressed on the cell surface of a mammalian cell; antibodies directed to an epitope of a Therapeutic protein and nucleic acids encoding antibodies of the invention (including fragments, analogs and derivatives thereof and anti-idiotypic antibodies as described herein). The antibodies of the invention can be used to treat, inhibit or prevent diseases, disorders or conditions associated with aberrant expression and/or activity of a Therapeutic protein, including, but not limited to, any one or more of the diseases, disorders, or conditions described herein. The treatment and/or prevention of diseases, disorders, or conditions associated with aberrant expression and/or activity of a Therapeutic protein includes, but is not limited to, alleviating symptoms associated with those diseases, disorders or conditions. Antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be provided in pharmaceutically acceptable compositions as known in the art or as described herein. [0234]
  • A summary of the ways in which the antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be used therapeutically includes binding Therapeutic proteins locally or systemically in the body or by direct cytotoxicity of the antibody, e.g. as mediated by complement (CDC) or by effector cells (ADCC). Some of these approaches are described in more detail below. Armed with the teachings provided herein, one of ordinary skill in the art will know how to use the antibodies of the invention or albumin fusion proteins of the invention comprising at-least a fragment or variant of an antibody that binds a Therapeutic protein for diagnostic, monitoring or therapeutic purposes without undue experimentation. [0235]
  • The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be advantageously utilized in combination with other monoclonal or chimeric antibodies, or with lymphokines or hematopoietic growth factors (such as, e.g., IL-2, IL-3 and IL-7), for example, which serve to increase the number or activity of effector cells which interact with the antibodies. [0236]
  • The antibodies of the invention or albumin fusion proteins of the invention comprising at least a fragment or variant of an antibody that binds a Therapeutic protein may be administered alone or in combination with other types of treatments (e.g., radiation therapy, chemotherapy, hormonal therapy, immunotherapy and anti-tumor agents). Generally, administration of products of a species origin or species reactivity (in the case of antibodies) that is the same species as that of the patient is preferred. Thus, in a preferred embodiment, human antibodies, fragments derivatives, analogs, or nucleic acids, are administered to a human patient for therapy or prophylaxis. [0237]
  • It is preferred to use high affinity and/or potent in vivo inhibiting and/or neutralizing antibodies against Therapeutic proteins, fragments or regions thereof, (or the albumin fusion protein correlate of such an antibody) for both immunoassays directed to and therapy of disorders related to polynucleotides or polypeptides, including fragments thereof, of the present invention. Such antibodies, fragments, or regions, will preferably have an affinity for polynucleotides or polypeptides of the invention, including fragments thereof. Preferred binding affinities include dissociation constants or Kd's less than 5×10[0238] −2 M, 10−2 M, 5×10−3 M, 10−3 M, 5×10−4 M, 10−4 M. More preferred binding affinities include those with a dissociation constant or Kd less than 5×10−5 M, 10−5 M, 5×10−6 M, 10−6 M, 5×10−7 M, 107 M, 5×10−8 M or 10−8 M. Even more preferred binding affinities include those with a dissociation constant or Kd less than 5×10−9 M, 10−9 M, 5×10−10 M, 10−10 M, 5×10−11 M, 10−11 M, 5×10−12 M, 10−12 M, 5×10−13 M, 10−13 M, 5×10−14 M, 10−14M, 5×10−15 M, or 10−15 M.
  • Gene Therapy [0239]
  • In a specific embodiment, nucleic acids comprising sequences encoding antibodies that bind therapeutic proteins or albumin fusion proteins comprising at least a fragment or varaint of an antibody that binds a Therapeutic protein are administered to treat, inhibit or prevent a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein, by way of gene therapy. Gene therapy refers to therapy performed by the administration to a subject of an expressed or expressible nucleic acid. In this embodiment of the invention, the nucleic acids produce their encoded protein that mediates a therapeutic effect. [0240]
  • Any of the methods for gene therapy available in the art can be used according to the present invention. Exemplary methods are described in more detail elsewhere in this application. [0241]
  • Demonstration of Therapeutic or Prophylactic Activity [0242]
  • The compounds or pharmaceutical compositions of the invention are preferably tested in vitro, and then in vivo for the desired therapeutic or prophylactic activity, prior to use in humans. For example, in vitro assays to demonstrate the therapeutic or prophylactic utility of a compound or pharmaceutical composition include, the effect of a compound on a cell line or a patient tissue sample. The effect of the compound or composition on the cell line and/or tissue sample can be determined utilizing techniques known to those of skill in the art including, but not limited to, rosette formation assays and cell lysis assays. In accordance with the invention, in vitro assays which can be used to determine whether administration of a specific compound is indicated, include in vitro cell culture assays in which a patient tissue sample is grown in culture, and exposed to or otherwise administered a compound, and the effect of such compound upon the tissue sample is observed. [0243]
  • Therapeutic/Prophylactic Administration and Composition [0244]
  • The invention provides methods of treatment, inhibition and prophylaxis by administration to a subject of an effective amount of a compound or pharmaceutical composition of the invention, preferably an antibody. In a preferred embodiment, the compound is substantially purified (e.g., substantially free from substances that limit its effect or produce undesired side-effects). The subject is preferably an animal, including but not limited to animals such as cows, pigs, horses, chickens, cats, dogs, etc., and is preferably a mammal, and most preferably human. [0245]
  • Formulations and methods of administration that can be employed when the compound comprises a nucleic acid or an immunoglobulin are described above; additional appropriate formulations and routes of administration can be selected from among those described herein below. [0246]
  • Various delivery systems are known and can be used to administer a compound of the invention, e.g., encapsulation in liposomes, microparticles, microcapsules, recombinant cells capable of expressing the compound, receptor-mediated endocytosis (see, e.g., Wu and Wu, J. Biol. Chem. 262:4429-4432 (1987)), construction of a nucleic acid as part of a retroviral or other vector, etc. Methods of introduction include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, and oral routes. The compounds or compositions may be administered by any convenient route, for example by infusion or bolus injection, by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal and intestinal mucosa, etc.) and may be administered together with other biologically active agents. Administration can be systemic or local. In addition, it may be desirable to introduce the pharmaceutical compounds or compositions of the invention into the central nervous system by any suitable route, including intraventricular and intrathecal injection; intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir, such as an Ommaya reservoir. Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, and formulation with an aerosolizing agent. [0247]
  • In a specific embodiment, it may be desirable to administer the pharmaceutical compounds or compositions of the invention locally to the area in need of treatment; this may be achieved by, for example, and not by way of limitation, local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers. Preferably, when administering a protein, including an antibody, of the invention, care must be taken to use materials to which the protein does not absorb. [0248]
  • In another embodiment, the compound or composition can be delivered in a vesicle, in particular a liposome (see Langer, Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 353-365 (1989); Lopez-Berestein, ibid., pp. 317-327; see generally ibid.) [0249]
  • In yet another embodiment, the compound or composition can be delivered in a controlled release system. In one embodiment, a pump may be used (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). In another embodiment, polymeric materials can be used (see Medical Applications of Controlled Release, Langer and Wise (eds.), CRC Pres., Boca Raton, Fla. (1974); Controlled Drug Bioavailability, Drug Product Design and Performance, Smolen and Ball (eds.), Wiley, New York (1984); Ranger and Peppas, J., Macromol. Sci. Rev. Macromol. Chem. 23:61 (1983); see also Levy et al., Science 228:190 (1985); During et al., Ann. Neurol. 25:351 (1989); Howard et al., J. Neurosurg. 71:105 (1989)). In yet another embodiment, a controlled release system can be placed in proximity of the therapeutic target, e.g., the brain, thus requiring only a fraction of the systemic dose (see, e.g., Goodson, in Medical Applications of Controlled Release, supra, vol. 2, pp. 115-138 (1984)). [0250]
  • Other controlled release systems are discussed in the review by Langer (Science 249:1527-1533 (1990)). [0251]
  • In a specific embodiment where the compound of the invention is a nucleic acid encoding a protein, the nucleic acid can be administered in vivo to promote expression of its encoded protein, by constructing it as part of an appropriate nucleic acid expression vector and administering it so that it becomes intracellular, e.g., by use of a retroviral vector (see U.S. Pat. No. 4,980,286), or by direct injection, or by use of microparticle bombardment (e.g., a gene gun; Biolistic, Dupont), or coating with lipids or cell-surface receptors or transfecting agents, or by administering it in linkage to a homeobox-like peptide which is known to enter the nucleus (see e.g., Joliot et al., Proc. Natl. Acad. Sci. USA 88:1864-1868 (1991)), etc. Alternatively, a nucleic acid can be introduced intracellularly and incorporated within host cell DNA for expression, by homologous recombination. [0252]
  • The present invention also provides pharmaceutical compositions. Such compositions comprise a therapeutically effective amount of a compound, and a pharmaceutically acceptable carrier. In a specific embodiment, the term “pharmaceutically acceptable” means approved by a regulatory agency of the Federal or a state government or listed in the U.S. Pharmacopeia or other generally recognized pharmacopeia for use in animals, and more particularly in humans. The term “carrier” refers to a diluent, adjuvant, excipient, or vehicle with which the therapeutic is administered. Such pharmaceutical carriers can be sterile liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Water is a preferred carrier when the pharmaceutical composition is administered intravenously. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. Suitable pharmaceutical excipients include starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, water, ethanol and the like. The composition, if desired, can also contain minor amounts of wetting or emulsifying agents, or pH buffering agents. These compositions can take the form of solutions, suspensions, emulsion, tablets, pills, capsules, powders, sustained-release formulations and the like. The composition can be formulated as a suppository, with traditional binders and carriers such as triglycerides. Oral formulation can include standard carriers such as pharmaceutical grades of mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, magnesium carbonate, etc. Examples of suitable pharmaceutical carriers are described in “Remington's Pharmaceutical Sciences” by E. W. Martin. Such compositions will contain a therapeutically effective amount of the compound, preferably in purified form, together with a suitable amount of carrier so as to provide the form for proper administration to the patient. The formulation should suit the mode of administration. [0253]
  • In a preferred embodiment, the composition is formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compositions for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the composition may also include a solubilizing agent and a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the composition is to be administered by infusion, it can be dispensed with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the composition is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration. [0254]
  • The compounds of the invention can be formulated as neutral or salt forms. Pharmaceutically acceptable salts include those formed with anions such as those derived from hydrochloric, phosphoric, acetic, oxalic, tartaric acids, etc., and those formed with cations such as those derived from sodium, potassium, ammonium, calcium, ferric hydroxides, isopropylamine, triethylamine, 2-ethylamino ethanol, histidine, procaine, etc. [0255]
  • The amount of the compound of the invention which will be effective in the treatment, inhibition and prevention of a disease or disorder associated with aberrant expression and/or activity of a Therapeutic protein can be determined by standard clinical techniques. In addition, in vitro assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the formulation will also depend on the route of administration, and the seriousness of the disease or disorder, and should be decided according to the judgment of the practitioner and each patient's circumstances. Effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. [0256]
  • For antibodies, the dosage administered to a patient is typically 0.1 mg/kg to 100 mg/kg of the patient's body weight. Preferably, the dosage administered to a patient is between 0.1 mg/kg and 20 mg/kg of the patient's body weight, more preferably 1 mg/kg to 10 mg/kg of the patient's body weight. Generally, human antibodies have a longer half-life within the human body than antibodies from other species due to the immune response to the foreign polypeptides. Thus, lower dosages of human antibodies and less frequent administration is often possible. Further, the dosage and frequency of administration of antibodies of the invention may be reduced by enhancing uptake and tissue penetration (e.g., into the brain) of the antibodies by modifications such as, for example, lipidation. [0257]
  • Diagnosis and Imaging [0258]
  • Labeled antibodies and derivatives and analogs thereof that bind a Therapeutic protein (or fragment or variant thereof) (including albumin fusion proteins comprising at least a fragment or variant of an antibody that binds a Therapeutic protein), can be used for diagnostic purposes to detect, diagnose, or monitor diseases, disorders, and/or conditions associated with the aberrant expression and/or activity of Therapeutic protein. The invention provides for the detection of aberrant expression of a Therapeutic protein, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the polypeptide interest and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein expression level compared to the standard expression level is indicative of aberrant expression. [0259]
  • The invention provides a diagnostic assay for diagnosing a disorder, comprising (a) assaying the expression of the Therapeutic protein in cells or body fluid of an individual using one or more antibodies specific to the Therapeutic protein or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein, and (b) comparing the level of gene expression with a standard gene expression level, whereby an increase or decrease in the assayed Therapeutic protein gene expression level compared to the standard expression level is indicative of a particular disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer. [0260]
  • Antibodies of the invention or albumin fusion proteins comprising at least a fragment of variant of an antibody specific to a Therapeutic protein can be used to assay protein levels in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen et al., J. Cell. Biol. 105:3087-3096 (1987)). Other antibody-based methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine (125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99Tc); luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. [0261]
  • One facet of the invention is the detection and diagnosis of a disease or disorder associated with aberrant expression of a Therapeutic protein in an animal, preferably a mammal and most preferably a human. In one embodiment, diagnosis comprises: a) administering (for example, parenterally, subcutaneously, or intraperitoneally) to a subject an effective amount of a labeled molecule which specifically binds to the polypeptide of interest; b) waiting for a time interval following the administering for permitting the labeled molecule to preferentially concentrate at sites in the subject where the Therapeutic protein is expressed (and for unbound labeled molecule to be cleared to background level); c) determining background level; and d) detecting the labeled molecule in the subject, such that detection of labeled molecule above the background level indicates that the subject has a particular disease or disorder associated with aberrant expression of the therapeutic protein. Background level can be determined by various methods including, comparing the amount of labeled molecule detected to a standard value previously determined for a particular system. [0262]
  • It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99 mTc. The labeled antibody, antibody fragment, or albumin fusion protein comprising at least a fragement or variant of an antibody that binds a Therapeutic protein will then preferentially accumulate at the location of cells which contain the specific Therapeutic protein. In vivo tumor imaging is described in S. W. Burchiel et al., “Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments.” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)). [0263]
  • Depending on several variables, including the type of label used and the mode of administration, the time interval following the administration for permitting the labeled molecule to preferentially concentrate at sites in the subject and for unbound labeled molecule to be cleared to background level is 6 to 48 hours or 6 to 24 hours or 6 to 12 hours. In another embodiment the time interval following administration is 5 to 20 days or 5 to 10 days. [0264]
  • In an embodiment, monitoring of the disease or disorder is carried out by repeating the method for diagnosing the disease or disease, for example, one month after initial diagnosis, six months after initial diagnosis, one year after initial diagnosis, etc. [0265]
  • Presence of the labeled molecule can be detected in the patient using methods known in the art for in vivo scanning. These methods depend upon the type of label used. Skilled artisans will be able to determine the appropriate method for detecting a particular label. Methods and devices that may be used in the diagnostic methods of the invention include, but are not limited to, computed tomography (CT), whole body scan such as position emission tomography (PET), magnetic resonance imaging (MRI), and sonography. [0266]
  • In a specific embodiment, the molecule is labeled with a radioisotope and is detected in the patient using a radiation responsive surgical instrument (Thurston et al., U.S. Pat. No. 5,441,050). In another embodiment, the molecule is labeled with a fluorescent compound and is detected in the patient using a fluorescence responsive scanning instrument. In another embodiment, the molecule is labeled with a positron emitting metal and is detected in the patent using positron emission-tomography. In yet another embodiment, the molecule is labeled with a paramagnetic label and is detected in a patient using magnetic resonance imaging (MRI). [0267]
  • Kits [0268]
  • The present invention provides kits that can be used in the above methods. In one embodiment, a kit comprises an antibody, preferably a purified antibody, in one or more containers. In a specific embodiment, the kits of the present invention contain a substantially isolated polypeptide comprising an epitope which is specifically immunoreactive with an antibody included in the kit. Preferably, the kits of the present invention further comprise a control antibody which does not react with the polypeptide of interest. In another specific embodiment, the kits of the present invention contain a means for detecting the binding of an antibody to a polypeptide of interest (e.g., the antibody may be conjugated to a detectable substrate such as a fluorescent compound, an enzymatic substrate, a radioactive compound or a luminescent compound, or a second antibody which recognizes the first antibody may be conjugated to a detectable substrate). [0269]
  • In another specific embodiment of the present invention, the kit is a diagnostic kit for use in screening serum containing antibodies specific against proliferative and/or cancerous polynucleotides and polypeptides. Such a kit may include a control antibody that does not react with the polypeptide of interest. Such a kit may include a substantially isolated polypeptide antigen comprising an epitope which is specifically immunoreactive with at least one anti-polypeptide antigen antibody. Further, such a kit includes means for detecting the binding of said antibody to the antigen (e.g., the antibody may be conjugated to a fluorescent compound such as fluorescein or rhodamine which can be detected by flow cytometry). In specific embodiments, the kit may include a recombinantly produced or chemically synthesized polypeptide antigen. The polypeptide antigen of the kit may also be attached to a solid support. [0270]
  • In a more specific embodiment the detecting means of the above-described kit includes a solid support to which said polypeptide antigen is attached. Such a kit may also include a non-attached reporter-labeled anti-human antibody. In this embodiment, binding of the antibody to the polypeptide antigen can be detected by binding of the said reporter-labeled antibody. [0271]
  • In an additional embodiment, the invention includes a diagnostic kit for use in screening serum containing antigens of the polypeptide of the invention. The diagnostic kit includes a substantially isolated antibody specifically immunoreactive with polypeptide or polynucleotide antigens, and means for detecting the binding of the polynucleotide or polypeptide antigen to the antibody. In one embodiment, the antibody is attached to a solid support. In a specific embodiment, the antibody may be a monoclonal antibody. The detecting means of the kit may include a second, labeled monoclonal antibody. Alternatively, or in addition, the detecting means may include a labeled, competing antigen. [0272]
  • In one diagnostic configuration, test serum is reacted with a solid phase reagent having a surface-bound antigen obtained by the methods of the present invention. After binding with specific antigen antibody to the reagent and removing unbound serum components by washing, the reagent is reacted with reporter-labeled-anti-human antibody to bind reporter to the reagent in proportion to the amount of bound anti-antigen antibody on the solid support. The reagent is again washed to remove unbound labeled antibody, and the amount of reporter associated with the reagent is determined. Typically, the reporter is an enzyme which is detected by incubating the solid phase in the presence of a suitable fluorometric, luminescent or colorimetric substrate (Sigma, St. Louis, Mo.). [0273]
  • The solid surface reagent in the above assay is prepared by known techniques for attaching protein material to solid support material, such as polymeric beads, dip sticks, 96-well plate or filter material. These attachment methods generally include non-specific adsorption of the protein to the support or covalent attachment of the protein, typically through a free amine group, to a chemically reactive group on the solid support, such as an activated carboxyl, hydroxyl, or aldehyde group. Alternatively, streptavidin coated plates can be used in conjunction with biotinylated antigen(s). [0274]
  • Thus, the invention provides an assay system or kit for carrying out this diagnostic method. The kit generally includes a support with surface-bound recombinant antigens, and a reporter-labeled anti-human antibody for detecting surface-bound anti-antigen antibody. [0275]
  • Albumin Fusion Proteins [0276]
  • The present invention relates generally to albumin fusion proteins and methods of treating, preventing, or ameliorating diseases or disorders. As used herein, “albumin fusion protein” refers to a protein formed by the fusion of at least one molecule of albumin (or a fragment or variant thereof) to at least one molecule of a Therapeutic protein (or fragment or variant thereof). An albumin fusion protein of the invention comprises at least a fragment or variant of a Therapeutic protein and at least a fragment or variant of human serum albumin, which are associated with one another, preferably by genetic fusion (i.e., the albumin fusion protein is generated by translation of a nucleic acid in which a polynucleotide encoding all or a portion of a Therapeutic protein is joined in-frame with a polynucleotide encoding all or a portion of albumin) or chemical conjugation to one another. The Therapeutic protein and albumin protein, once part of the albumin fusion protein, may be referred to as a “portion”, “region” or “moiety” of the albumin fusion protein. [0277]
  • In one embodiment, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein (e.g., as described in Table 1) and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment of a Therapeutic protein and a serum albumin protein. In other embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active variant of a Therapeutic protein and a serum albumin protein. In preferred embodiments, the serum albumin protein component of the albumin fusion protein is the mature portion of serum albumin. [0278]
  • In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein, and a biologically active and/or therapeutically active fragment of serum albumin. In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a Therapeutic protein and a biologically active and/or therapeutically active variant of serum albumin. In preferred embodiments, the Therapeutic protein portion of the albumin fusion protein is the mature portion of the Therapeutic protein. [0279]
  • In further embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, a biologically active and/or therapeutically active fragment or variant of a Therapeutic protein and a biologically active and/or therapeutically active fragment or variant of serum albumin. In preferred embodiments, the invention provides an albumin fusion protein comprising, or alternatively consisting of, the mature portion of a Therapeutic protein and the mature portion of serum albumin. [0280]
  • Preferably, the albumin fusion protein comprises HA as the N-terminal portion, and a Therapeutic protein as the C-terminal portion. Alternatively, an albumin fusion protein comprising HA as the C-terminal portion, and a Therapeutic protein as the N-terminal portion may also be used. [0281]
  • In other embodiments, the albumin fusion protein has a Therapeutic protein fused to both the N-terminus and the C-terminus of albumin. In a preferred embodiment, the Therapeutic proteins fused at the N- and C-termini are the same Therapeutic proteins. In a preferred embodiment, the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins. In another preferred embodiment, the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins which may be used to treat or prevent the same disease, disorder, or condition (e.g. as listed in the “Preferred Indication Y” column of Table 1). In another preferred embodiment, the Therapeutic proteins fused at the N- and C-termini are different Therapeutic proteins which may be used to treat or prevent diseases or disorders (e.g. as listed in the “Preferred Indication Y” column of Table 1) which are known in the art to commonly occur in patients simultaneously. [0282]
  • In addition to albumin fusion protein in which the albumin portion is fused N-terminal and/or C-terminal of the Therapeutic protein portion, albumin fusion proteins of the invention may also be produced by inserting the Therapeutic protein or peptide of interest (e.g., a Therapeutic protein X as diclosed in Table 1, or an antibody that binds a Therapeutic protein or a fragment or variant thereof) into an internal region of HA. For instance, within the protein sequence of the HA molecule a number of loops or turns exist between the end and beginning of α-helices, which are stabilized by disulphide bonds (see FIGS. [0283] 9-11). The loops, as determined from the crystal structure of HA (FIG. 13) (PDB identifiers IAO6, 1BJ5, 1BKE, 1BM0, 1E7E to 1E7I and 1UOR) for the most part extend away from the body of the molecule. These loops are useful for the insertion, or internal fusion, of therapeutically active peptides, particularly those requiring a secondary structure to be functional, or Therapeutic proteins, to essentially generate an albumin molecule with specific biological activity.
  • Loops in human albumin structure into which peptides or polypeptides may be inserted to generate albumin fusion proteins of the invention include: Val54-Asn61, Thr76-Asp89, Ala92-Glu100, Gln170-Ala176, His 247-Glu252, Glu 266-Glu277, Glu 280-His288, Ala362-Glu368, Lys439-Pro447,Val462-Lys475, Thr478-Pro486, and Lys560-Thr566. In more preferred embodiments, peptides or polypeptides are inserted into the Val54-Asn61, Gln170-Ala176, and/or Lys560-Thr566 loops of mature human albumin (SEQ ID NO: 18). [0284]
  • Peptides to be inserted may be derived from either phage display or synthetic peptide libraries screened for specific biological activity or from the active portions of a molecule with the desired function. Additionally, random peptide libraries may be generated within particular loops or by insertions of randomized peptides into particular loops of the HA molecule and in which all possible combinations of amino acids are represented. [0285]
  • Such library(s) could be generated on HA or domain fragments of HA by one of the following methods: [0286]
  • (a) randomized mutation of amino acids within one or more peptide loops of HA or HA domain fragments. Either one, more or all the residues within a loop could be mutated in this manner (for example see FIG. 10[0287] a);
  • (b) replacement of, or insertion into one or more loops of HA or HA domain fragments (i.e., internal fusion) of a randomized peptide(s) of length X[0288] n (where X is an amino acid and n is the number of residues (for example see FIG. 10b);
  • (c) N-, C- or N- and C-terminal peptide/protein fusions in addition to (a) and/or (b). [0289]
  • The HA or HA domain fragment may also be made multifunctional by grafting the peptides derived from different screens of different loops against different targets into the same HA or HA domain fragment. [0290]
  • In preferred embodiments, peptides inserted into a loop of human serum albumin are peptide fragments or peptide variants of the Therapeutic proteins disclosed in Table 1. More particulary, the invention encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids in length inserted into a loop of human serum albumin. The invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the N-terminus of human serum albumin. The invention also encompasses albumin fusion proteins which comprise peptide fragments or peptide variants at least 7 at least 8, at least 9, at least 10, at least 11, at least 12, at least 13, at least 14, at least 15, at least 20, at least 25, at least 30, at least 35, or at least 40 amino acids fused to the C-terminus of human serum albumin. [0291]
  • Generally, the albumin fusion proteins of the invention may have one HA-derived region and one Therapeutic protein-derived region. Multiple regions of each protein, however, may be used to make an albumin fusion protein of the invention. Similarly, more than one Therapeutic protein may be used to make an albumin fusion protein of the invention. For instance, a Therapeutic protein may be fused to both the N- and C-terminal ends of the HA. In such a configuration, the Therapeutic protein portions may be the same or different Therapeutic protein molecules. The structure of bifunctional albumin fusion proteins may be represented as: X-HA-Y or Y-HA-X. [0292]
  • For example, an anti-BLyS™ scFv-HA-IFNα-2b fusion may be prepared to modulate the immune response to IFNα-2b by anti-BLyS™ scFv. An alternative is making a bi (or even multi) functional dose of HA-fusions e.g. HA-IFNα-2b fusion mixed with HA-anti-BLyS™ scFv fusion or other HA-fusions in various ratio's depending on function, half-life etc. [0293]
  • Bi- or multi-functional albumin fusion proteins may also be prepared to target the Therapeutic protein portion of a fusion to a target organ or cell type via protein or peptide at the opposite terminus of HA. [0294]
  • As an alternative to the fusion of known therapeutic molecules, the peptides could be obtained by screening libraries constructed as fusions to the N-, C- or N- and C-termini of HA, or domain fragment of HA, of typically 6, 8, 12, 20 or 25 or X[0295] n (where X is an amino acid (aa) and n equals the number of residues) randomized amino acids, and in which all possible combinations of amino acids were represented. A particular advantage of this approach is that the peptides may be selected in situ on the HA molecule and the properties of the peptide would therefore be as selected for rather than, potentially, modified as might be the case for a peptide derived by any other method then being attached to HA.
  • Additionally, the albumin fusion proteins of the invention may include a linker peptide between the fused portions to provide greater physical separation between the moieties and thus maximize the accessibility of the Therapeutic protein portion, for instance, for binding to its cognate receptor. The linker peptide may consist of amino acids such that it is flexible or more rigid. [0296]
  • The linker sequence may be cleavable by a protease or chemically to yield the growth hormone related moiety. Preferably, the protease is one which is produced naturally by the host, for example the [0297] S. cerevisiae protease kex2 or equivalent proteases.
  • Therefore, as described above, the albumin fusion proteins of the invention may have the following formula R1-L-R2; R2-L-R1; or R1-L-R2-L-R1, wherein R1 is at least one Therapeutic protein, peptide or polypeptide sequence, and not necessarily the same Therapeutic protein, L is a linker and R2 is a serum albumin sequence. [0298]
  • In preferred embodiments, Albumin fusion proteins of the invention comprising a Therapeutic protein have extended shelf life compared to the shelf life the same Therapeutic protein when not fused to albumin. Shelf-life typically refers to the time period over which the therapeutic activity of a Therapeutic protein in solution or in some other storage formulation, is stable without undue loss of therapeutic activity. Many of the Therapeutic proteins are highly labile in their unfused state. As described below, the typical shelf-life of these Therapeutic proteins is markedly prolonged upon incorporation into the albumin fusion protein of the invention. [0299]
  • Albumin fusion proteins of the invention with “prolonged” or “extended” shelf-life exhibit greater therapeutic activity relative to a standard that has been subjected to the same storage and handling conditions. The standard may be the unfused full-length Therapeutic protein. When the Therapeutic protein portion of the albumin fusion protein is an analog, a variant, or is otherwise altered or does not include the complete sequence for that protein, the prolongation of therapeutic activity may alternatively be compared to the unfused equivalent of that analog, variant, altered peptide or incomplete sequence. As an example, an albumin fusion protein of the invention may retain greater than about 100% of the therapeutic activity, or greater than about 105%, 110%, 120%, 130%, 150% or 200% of the therapeutic activity of a standard when subjected to the same storage and handling conditions as the standard when compared at a given time point. [0300]
  • Shelf-life may also be assessed in terms of therapeutic activity remaining after storage, normalized to therapeutic activity when storage began. Albumin fusion proteins of the invention with prolonged or extended shelf-life as exhibited by prolonged or extended therapeutic activity may retain greater than about 50% of the therapeutic activity, about 60%, 70%, 80%, or 90% or more of the therapeutic activity of the equivalent unfused Therapeutic protein when subjected to the same conditions. For example, as discussed in Example 1, an albumin fusion protein of the invention comprising hGH fused to the full length HA sequence may retain about 80% or more of its original activity in solution for periods of up to 5 weeks or more under various temperature conditions. [0301]
  • Expression of Fusion Proteins [0302]
  • The albumin fusion proteins of the invention may be produced as recombinant molecules by secretion from yeast, a microorganism such as a bacterium, or a human or animal cell line. Preferably, the polypeptide is secreted from the host cells. We have found that, by fusing the hGH coding sequence to the HA coding sequence, either to the 5′ end or 3′ end, it is possible to secrete the albumin fusion protein from yeast without the requirement for a yeast-derived pro sequence. This was surprising, as other workers have found that a yeast derived pro sequence was needed for efficient secretion of hGH in yeast. [0303]
  • For example, Hiramatsu et al. (Appl Environ Microbiol 56:2125 (1990); Appl Environ Microbiol 57:2052 (1991)) found that the N-terminal portion of the pro sequence in the [0304] Mucor pusillus rennin pre-pro leader was important. Other authors, using the MF -1 signal, have always included the MF -1 pro sequence when secreting hGH. The pro sequences were believed to assist in the folding of the hGH by acting as an intramolecular chaperone. The present invention shows that HA or fragments of HA can perform a similar function.
  • Hence, a particular embodiment of the invention comprises a DNA construct encoding a signal sequence effective for directing secretion in yeast, particularly a yeast-derived signal sequence (especially one which is homologous to the yeast host), and the fused molecule of the first aspect of the invention, there being no yeast-derived pro sequence between the signal and the mature polypeptide. [0305]
  • The [0306] Saccharomyces cerevisiae invertase signal is a preferred example of a yeast-derived signal sequence.
  • Conjugates of the kind prepared by Poznansky et al, (FEBS Lett. 239:18 (1988)), in which separately-prepared polypeptides are joined by chemical cross-linking, are not contemplated. [0307]
  • The present invention also includes a cell, preferably a yeast cell transformed to express an albumin fusion protein of the invention. In addition to the transformed host cells themselves, the present invention also contemplates a culture of those cells, preferably a monoclonal (clonally homogeneous) culture, or a culture derived from a monoclonal culture, in a nutrient medium. If the polypeptide is secreted, the medium will contain the polypeptide, with the cells, or without the cells if they have been filtered or centrifuged away. Many expression systems are known and may be used, including bacteria (for example [0308] E. coli and Bacillus subtilis), yeasts (for example Saccharomyces cerevisiae, Kluyveromyces lactis and Pichia pastoris, filamentous fungi (for example Aspergillus), plant cells, animal cells and insect cells.
  • Preferred yeast strains to be used in the production of albumin fusion proteins are D88, DXY1 and BXP10. D88 [leu2-3, leu2-122, canI, praI, ubc4] is a derivative of parent strain AH22his[0309] + (also known as DB1; see, e.g., Sleep et al. Biotechnology 8:42-46 (1990)). The strain contains a leu2 mutation which allows for auxotropic selection of 2 micron-based plasmids that contain the LEU2 gene. D88 also exhibits a derepression of PRB1 in glucose excess. The PRB1 promoter is normally controlled by two checkpoints that monitor glucose levels and growth stage. The promoter is activated in wild type yeast upon glucose depletion and entry into stationary phase. Strain D88 exhibits the repression by glucose but maintains the induction upon entry into stationary phase. The PRA1 gene encodes a yeast vacuolar protease, YscA endoprotease A, that is localized in the ER. The UBC4 gene is in the ubiquitination pathway and is involved in targeting short lived and abnormal proteins for ubiquitin dependant degradation. Isolation of this ubc4 mutation was found to increase the copy number of an expression plasmid in the cell and cause an increased level of expression of a desired protein expressed from the plasmid (see, e.g., International Publication No. WO99/00504, hereby incorporated in its entirety by reference herein).
  • DXY1, a derivative of D88, has the following genotype: [leu2-3, leu2-122, canI, pra1, ubc4, ura3::yap3]. In addition to the mutations isolated in D88, this strain also has a knockout of the YAP3 protease. This protease causes cleavage of mostly di-basic residues (RR, RK, KR, KK) but can also promote cleavage at single basic residues in proteins. Isolation of this yap3 mutation resulted in higher levels of full length HSA production (see, e.g., U.S. Pat. No. 5,965,386 and Kerry-Williams et al., Yeast 14:161-169 (1998), hereby incorporated in their entireties by reference herein). [0310]
  • BXP10 has the following genotype: leu2-3, leu2-122, can1, pra1, ubc4, ura3, yap3::URA3, lys2, hsp150::LYS2, pmt1::URA3. In addition to the mutations isolated in DXY1, this strain also has a knockout of the PMT1 gene and the HSP150 gene. The PMT1 gene is a member of the evolutionarily conserved family of dolichyl-phosphate-D-mannose protein O-mannosyltransferases (Pmts). The transmembrane topology of Pmt1p suggests that it is an integral membrane protein of the endoplasmic reticulum with a role in O-linked glycosylation. This mutation serves to reduce/eliminate O-linked glycosylation of HSA fusions (see, e.g., International Publication No. WO00/44772, hereby incorporated in its entirety by reference herein). Studies revealed that the Hsp150 protein is inefficiently separated from rHA by ion exchange chromatography. The mutation in the HSP150 gene removes a potential contaminant that has proven difficult to remove by standard purification techniques. See, e.g., U.S. Pat. No. 5,783,423, hereby incorporated in its entirety by reference herein. [0311]
  • The desired protein is produced in conventional ways, for example from a coding sequence inserted in the host chromosome or on a free plasmid. The yeasts are transformed with a coding sequence for the desired protein in any of the usual ways, for example electroporation. Methods for transformation of yeast by electroporation are disclosed in Becker & Guarente (1990) [0312] Methods Enzymol. 194, 182.
  • Successfully transformed cells, i.e., cells that contain a DNA construct of the present invention, can be identified by well known techniques. For example, cells resulting from the introduction of an expression construct can be grown to produce the desired polypeptide. Cells can be harvested and lysed and their DNA content examined for the presence of the DNA using a method such as that described by Southern (1975) [0313] J. Mol. Biol. 98, 503 or Berent et al. (1985) Biotech. 3, 208. Alternatively, the presence of the protein in the supernatant can be detected using antibodies.
  • Useful yeast plasmid vectors include pRS403-406 and pRS413-416 and are generally available from Stratagene Cloning Systems, La Jolla, Calif. 92037, USA. Plasmids pRS403, pRS404, pRS405 and pRS406 are Yeast Integrating plasmids (YIps) and incorporate the yeast selectable markers HIS3, 7RP1, LEU2 and URA3. Plasmids pRS413-416 are Yeast Centromere plasmids (Ycps). [0314]
  • Preferred vectors for making albumin fusion proteins for expression in yeast include pPPC0005, pScCHSA, pScNHSA, and pC4:HSA which are described in detail in Example 2. FIG. 4 shows a map of the pPPC0005 plasmid that can be used as the base vector into which polynucleotides encoding Therapeutic proteins may be cloned to form HA-fusions. It contains a PRB1 [0315] S. cerevisiae promoter (PRB1p), a Fusion leader sequence (FL), DNA encoding HA (rHA) and an ADH1 S. cerevisiae terminator sequence. The sequence of the fusion leader sequence consists of the first 19 amino acids of the signal peptide of human serum albumin (SEQ ID NO:29) and the last five amino acids of the mating factor alpha 1 promoter (SLDKR, see EP-A-387 319 which is hereby incorporated by reference in its entirety.
  • The plasmids, pPPC0005, pScCHSA, pScNHSA, and pC4:HSA were deposited on Apr. 11, 2001 at the American Type Culture Collection, 10801 University Boulevard, Manassas, Va. 20110-2209 and given accession numbers ATCC ______, ______, ______, and ______, respectively. Another vector useful for expressing an albumin fusion protein in yeast the pSAC35 vector which is described in Sleep et al., BioTechnology 8:42 (1990) which is hereby incorporated by reference in its entirety. [0316]
  • A variety of methods have been developed to operably link DNA to vectors via complementary cohesive termini. For instance, complementary homopolymer tracts can be added to the DNA segment to be inserted to the vector DNA. The vector and DNA segment are then joined by hydrogen bonding between the complementary homopolymeric tails to form recombinant DNA molecules. [0317]
  • Synthetic linkers containing one or more restriction sites provide an alternative method of joining the DNA segment to vectors. The DNA segment, generated by endonuclease restriction digestion, is treated with bacteriophage T4 DNA polymerase or [0318] E. coli DNA polymerase I, enzymes that remove protruding, -single-stranded termini with their 3′ 5′-exonucleolytic activities, and fill in recessed 3′-ends with their polymerizing activities.
  • The combination of these activities therefore generates blunt-ended DNA segments. The blunt-ended segments are then incubated with a large molar excess of linker molecules in the presence of an enzyme that is able to catalyze the ligation of blunt-ended DNA molecules, such as bacteriophage T4 DNA ligase. Thus, the products of the reaction are DNA segments carrying polymeric linker sequences at their ends. These DNA segments are then cleaved with the appropriate restriction enzyme and ligated to an expression vector that has been cleaved with an enzyme that produces termini compatible with those of the DNA segment. [0319]
  • Synthetic linkers containing a variety of restriction endonuclease sites are commercially available from a number of sources including International Biotechnologies Inc, New Haven, Conn., USA. [0320]
  • A desirable way to modify the DNA in accordance with the invention, if, for example, HA variants are to be prepared, is to use the polymerase chain reaction as disclosed by Saiki et al. (1988) [0321] Science 239, 487-491. In this method the DNA to be enzymatically amplified is flanked by two specific oligonucleotide primers which themselves become incorporated into the amplified DNA. The specific primers may contain restriction endonuclease recognition sites which can be used for cloning into expression vectors using methods known in the art.
  • Exemplary genera of yeast contemplated to be useful in the practice of the present invention as hosts for expressing the albumin fusion proteins are Pichia (Hansenula), Saccharomyces, Kluyveromyces, Candida, Torulopsis, Torulaspora, Schizosaccharomyces, Citeromyces, Pachysolen, Debaromyces, Metschunikowia, Rhodosporidium, Leucosporidium, Botryoascus, Sporidiobolus, Endomycopsis, and the like. Preferred genera are those selected from the group consisting of Saccharomyces, Schizosaccharomyces, Kluyveromyces, Pichia and Torulaspora. Examples of Saccharomyces spp. are [0322] S. cerevisiae, S. italicus and S. rouxii.
  • Examples of Kluyveromyces spp. are [0323] K. fragilis, K. lactis and K. marxianus. A suitable Torulaspora species is T. delbrueckii. Examples of Pichia (Hansenula) spp. are P. angusta (formerly H. polymorpha), P. anomala (formerly H. anomala) and P. pastoris. Methods for the transformation of S. cerevisiae are taught generally in EP 251 744, EP 258 067 and WO 90/01063, all of which are incorporated herein by reference.
  • Preferred exemplary species of Saccharomyces include [0324] S. cerevisiae, S. italicus, S. diastaticus, and Zygosaccharomyces rouxii. Preferred exemplary species of Kluyveromyces include K. fragilis and K. lactis. Preferred exemplary species of Hansenula include H. polymorpha (now Pichia angusta), H. anomala (now Pichia anomala), and Pichia capsulata. Additional preferred exemplary species of Pichia include P. pastoris. Preferred exemplary species of Aspergillus include A. niger and A. nidulans. Preferred exemplary species of Yarrowia include Y. lipolytica. Many preferred yeast species are available from the ATCC. For example, the following preferred yeast species are available from the ATCC and are useful in the expression of albumin fusion proteins: Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 yap3 mutant (ATCC Accession No. 4022731); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 hsp150 mutant (ATCC Accession No. 4021266); Saccharomyces cerevisiae Hansen, teleomorph strain BY4743 pmt1 mutant (ATCC Accession No. 4023792); Saccharomyces cerevisiae Hansen, teleomorph (ATCC Accession Nos. 20626; 44773; 44774; and 62995); Saccharomyces diastaticus Andrews et Gilliland ex van der Walt, teleomorph (ATCC Accession No. 62987); Kluyveromyces lactis (Dombrowski) van der Walt, teleomorph (ATCC Accession No. 76492); Pichia angusta (Teunisson et al.) Kurtzman, teleomorph deposited as Hansenula polymorpha de Morais et Maia, teleomorph (ATCC Accession No. 26012); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 9029); Aspergillus niger van Tieghem, anamorph (ATCC Accession No. 16404); Aspergillus nidulans (Eidam) Winter, anamorph (ATCC Accession No. 48756); and Yarrowia lipolytica (Wickerham et al.) van der Walt et von Arx, teleomorph (ATCC Accession No. 201847).
  • Suitable promoters for [0325] S. cerevisiae include those associated with the PGKI gene, GAL1 or GAL10 genes, CYCI, PHO5, TRPI, ADHI, ADH2, the genes for glyceraldehyde-3-phosphate dehydrogenase, hexokinase, pyruvate decarboxylase, phosphofructokinase, triose phosphate isomerase, phosphoglucose isomerase, glucokinase, alpha-mating factor pheromone, [a mating factor pheromone], the PRBI promoter, the GUT2 promoter, the GPDI promoter, and hybrid promoters involving hybrids of parts of 5′ regulatory regions with parts of 5′ regulatory regions of other promoters or with upstream activation sites (e.g. the promoter of EP-A-258 067).
  • Convenient regulatable promoters for use in [0326] Schizosaccharomyces pombe are the thiamine-repressible promoter from the nmt gene as described by Maundrell (1990) J. Biol. Chem. 265, 10857-10864 and the glucose repressible jbp1 gene promoter as described by Hoffman & Winston (1990) Genetics 124, 807-816.
  • Methods of transforming Pichia for expression of foreign genes are taught in, for example, Cregg et al. (1993), and various Phillips patents (e.g. U.S. Pat. No. 4,857,467, incorporated herein by reference), and Pichia expression kits are commercially available from Invitrogen BV, Leek, Netherlands, and Invitrogen Corp., San Diego, Calif. Suitable promoters include AOXI and AOX2. Gleeson et al. (1986) J. Gen. Microbiol. 132, 3459-3465 include information on Hansenula vectors and transformation, suitable promoters being MOX1 and FMD1; whilst [0327] EP 361 991, Fleer et al. (1991) and other-publications from Rhone-Poulenc Rorer teach how to express foreign proteins in Kluyveromyces spp., a suitable promoter being PGKI.
  • The transcription termination signal is preferably the 3′ flanking sequence of a eukaryotic gene which contains proper signals for transcription termination and polyadenylation. Suitable 3′ flanking sequences may, for example, be those of the gene naturally linked to the expression control sequence used, i.e. may correspond to the promoter. Alternatively, they may be different in which case the termination signal of the [0328] S. cerevisiae ADHI gene is preferred.
  • The desired albumin fusion protein may be initially expressed with a secretion leader sequence, which may be any leader effective in the yeast chosen. Leaders useful in [0329] S. cerevisiae include that from the mating factor polypeptide (MF -1) and the hybrid leaders of EP-A-387 319. Such leaders (or signals) are cleaved by the yeast before the mature albumin is released into the surrounding medium. Further such leaders include those of S. cerevisiae invertase (SUC2) disclosed in JP 62-096086 (granted as 911036516), acid phosphatase (PH05), the pre-sequence of MFoz-1, 0 glucanase (BGL2) and killer toxin; S. diastaticus glucoamylase II; S. carlsbergensis-galactosidase (MEL1); K. lactis killer toxin; and Candida glucoarnylase.
  • Additional Methods of Recombinant and Synthetic Production of Albumin Fusion Proteins [0330]
  • The present invention also relates to vectors containing a polynucleotide encoding an albumin fusion protein of the present invention, host cells, and the production of albumin fusion proteins by synthetic and recombinant techniques. The vector may be, for example, a phage, plasmid, viral, or retroviral vector. Retroviral vectors may be replication competent or replication defective. In the latter case, viral propagation generally will occur only in complementing host cells. [0331]
  • The polynucleotides encoding albumin fusion proteins of the invention may be joined to a vector containing a selectable marker for propagation in a host. Generally, a plasmid vector is introduced in a precipitate, such as a calcium phosphate precipitate, or in a complex with a charged lipid. If the vector is a virus, it may be packaged in vitro using an appropriate packaging cell line and then transduced into host cells. [0332]
  • The polynucleotide insert should be operatively linked to an appropriate promoter, such as the phage lambda PL promoter, the [0333] E. coli lac, trp, phoA and tac promoters, the SV40 early and late promoters and promoters of retroviral LTRs, to name a few. Other suitable promoters will be known to the skilled artisan. The expression constructs will further contain sites for transcription initiation, termination, and, in the transcribed region, a ribosome binding site for translation. The coding portion of the transcripts expressed by the constructs will preferably include a translation initiating codon at the beginning and a termination codon (UAA, UGA or UAG) appropriately positioned at the end of the polypeptide to be translated.
  • As indicated, the expression vectors will preferably include at least one selectable marker. Such markers include dihydrofolate reductase, G418, glutamine synthase, or neomycin resistance for eukaryotic cell culture, and tetracycline, kanamycin or ampicillin resistance genes for culturing in [0334] E. coli and other bacteria. Representative examples of appropriate hosts include, but are not limited to, bacterial cells, such as E. coli, Streptomyces and Salmonella typhimurium cells; fungal cells, such as yeast cells (e.g., Saccharomyces cerevisiae or Pichia pastoris (ATCC Accession No. 201178)); insect cells such as Drosophila S2 and Spodoptera Sf9 cells; animal cells such as CHO, COS, NSO, 293, and Bowes melanoma cells; and plant cells. Appropriate culture mediums and conditions for the above-described host cells are known in the art.
  • Among vectors preferred for use in bacteria include pQE70, pQE60 and pQE-9, available from QIAGEN, Inc.; pBluescript vectors, Phagescript vectors, pNH8A, pNH16a, pNH18A, pNH46A, available from Stratagene Cloning Systems, Inc.; and ptrc99a, pKK223-3, pKK233-3, pDR540, pRIT5 available from Pharmacia Biotech, Inc. Among preferred eukaryotic vectors are pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; and pSVK3, pBPV, pMSG and pSVL available from Pharmacia. Preferred expression vectors for use in yeast systems include, but are not limited to pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalph, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, pPIC9K, and PAO815 (all available from Invitrogen, Carlbad, Calif.). Other suitable vectors will be readily apparent to the skilled artisan. [0335]
  • In one embodiment, polynucleotides encoding an albumin fusion protein of the invention may be fused to signal sequences which will direct the localization of a protein of the invention to particular compartments of a prokaryotic or eukaryotic cell and/or direct the secretion of a protein of the invention from a prokaryotic or eukaryotic cell. For example, in [0336] E. coli, one may wish to direct the expression of the protein to the periplasmic space. Examples of signal sequences or proteins (or fragments thereof) to which the albumin fusion proteins of the invention may be fused in order to direct the expression of the polypeptide to the periplasmic space of bacteria include, but are not limited to, the pelB signal sequence, the maltose binding protein (MBP) signal sequence, MBP, the ompA signal sequence, the signal sequence of the periplasmic E. coli heat-labile enterotoxin B-subunit, and the signal sequence of alkaline phosphatase. Several vectors are commercially available for the construction of fusion proteins which will direct the localization of a protein, such as the pMAL series of vectors (particularly the pMAL-p series) available from New England Biolabs. In a specific embodiment, polynucleotides albumin fusion proteins of the invention may be fused to the pelB pectate lyase signal sequence to increase the efficiency of expression and purification of such polypeptides in Gram-negative bacteria. See, U.S. Pat. Nos. 5,576,195 and 5,846,818, the contents of which are herein incorporated by reference in their entireties.
  • Examples of signal peptides that may be fused to an albumin fusion protein of the invention in order to direct its secretion in mammalian cells include, but are not limited to, the MPIF-1 signal sequence (e.g., amino acids 1-21 of GenBank Accession number AAB51134), the stanniocalcin signal sequence (MLQNSAVLLLLVISASA, SEQ ID NO:34), and a consensus signal sequence (MPTWAWWLFLVLLLALWAPARG, SEQ ID NO:35). A suitable signal sequence that may be used in conjunction with baculoviral expression systems is the gp67 signal sequence (e.g., amino acids 1-19 of GenBank Accession Number AAA72759). [0337]
  • Vectors which use glutamine synthase (GS) or DHFR as the selectable markers can be amplified in the presence of the drugs methionine sulphoximine or methotrexate, respectively. An advantage of glutamine synthase based vectors are the availabilty of cell lines (e.g., the murine myeloma cell line, NSO) which are glutamine synthase negative. Glutamine synthase expression systems can also function in glutamine synthase expressing cells (e.g., Chinese Hamster Ovary (CHO) cells) by providing additional inhibitor to prevent the functioning of the endogenous gene. A glutamine synthase expression system and components thereof are detailed in PCT publications: WO87/04462; WO86/05807; WO89/01036; WO89/10404; and WO91/06657, which are hereby incorporated in their entireties by reference herein. Additionally, glutamine synthase expression vectors can be obtained from Lonza Biologics, Inc. (Portsmouth, N.H.). Expression and production of monoclonal antibodies using a GS expression system in murine myeloma cells is described in Bebbington et al., [0338] Bio/technology 10:169(1992) and in Biblia and Robinson Biotechnol. Prog. 11:1 (1995) which are herein incorporated by reference.
  • The present invention also relates to host cells containing the above-described vector constructs described herein, and additionally encompasses host cells containing nucleotide sequences of the invention that are operably associated with one or more heterologous control regions (e.g., promoter and/or enhancer) using techniques known of in the art. The host cell can be a higher eukaryotic cell, such as a mammalian cell (e.g., a human derived cell), or a lower eukaryotic cell, such as a yeast cell, or the host cell can be a prokaryotic cell, such as a bacterial cell. A host strain may be chosen which modulates the expression of the inserted gene sequences, or modifies and processes the gene product in the specific fashion desired. Expression from certain promoters can be elevated in the presence of certain inducers; thus expression of the genetically engineered polypeptide may be controlled. Furthermore, different host cells have characteristics and specific mechanisms for the translational and post-translational processing and modification (e.g., phosphorylation, cleavage) of proteins. Appropriate cell lines can be chosen to ensure the desired modifications and processing of the foreign protein expressed. [0339]
  • Introduction of the nucleic acids and nucleic acid constructs of the invention into the host cell can be effected by calcium phosphate transfection, DEAE-dextran mediated transfection, cationic lipid-mediated transfection, electroporation, transduction, infection, or other methods. Such methods are described in many standard laboratory manuals, such as Davis et al., Basic Methods In Molecular Biology (1986). It is specifically contemplated that the polypeptides of the present invention may in fact be expressed by a host cell lacking a recombinant vector. [0340]
  • In addition to encompassing host cells containing the vector constructs discussed herein, the invention also encompasses primary, secondary, and immortalized host cells of vertebrate origin, particularly mammalian origin, that have been engineered to delete or replace endogenous genetic material (e.g., the coding sequence corresponding to a Therapeutic protein may be replaced with an albumin fusion protein corresponding to the Therapeutic protein), and/or to include genetic material (e.g., heterologous polynucleotide sequences such as for example, an albumin fusion protein of the invention corresponding to the Therapeutic protein may be included). The genetic material operably associated with the endogenous polynucleotide may activate, alter, and/or amplify endogenous polynucleotides. [0341]
  • In addition, techniques known in the art may be used to operably associate heterologous polynucleotides (e.g., polynucleotides encoding an albumin protein, or a fragment or variant thereof) and/or heterologous control regions (e.g., promoter and/or enhancer) with endogenous polynucleotide sequences encoding a Therapeutic protein via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication Number WO 96/29411; International Publication Number WO 94/12650; Koller et al., [0342] Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), the disclosures of each of which are incorporated by reference in their entireties).
  • Albumin fusion proteins of the invention can be recovered and purified from recombinant cell cultures by well-known methods including ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography, hydrophobic charge interaction chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification. [0343]
  • In preferred embodiments the albumin fusion proteins of the invention are purified using Anion Exchange Chromatography including, but not limited to, chromatography on Q-sepharose, DEAE sepharose, poros HQ, poros DEAE, Toyopearl Q, Toyopearl QAE, Toyopearl DEAE, Resource/Source Q and DEAE, Fractogel Q and DEAE columns. [0344]
  • In specific embodiments the albumin fusion proteins of the invention are purified using Cation Exchange Chromatography including, but not limited to, SP-sepharose, CM sepharose, poros HS, poros CM, Toyopearl SP, Toyopearl CM, Resource/Source S and CM, Fractogel S and CM columns and their equivalents and comparables. [0345]
  • In specific embodiments the albumin fusion proteins of the invention are purified using Hydrophobic Interaction Chromatography including, but not limited to, Phenyl, Butyl, Methyl, Octyl, Hexyl-sepharose, poros Phenyl, Butyl, Methyl, Octyl, Hexyl Toyopearl Phenyl, Butyl, Methyl, Octyl, Hexyl Resource/Source Phenyl, Butyl, Methyl, Octyl, Hexyl, Fractogel Phenyl, Butyl, Methyl, Octyl, Hexyl columns and their equivalents and comparables. [0346]
  • In specific embodiments the albumin fusion proteins of the invention are purified using Size Exclusion Chromatography including, but not limited to, sepharose S100, S200, S300, superdex resin columns and their equivalents and comparables. [0347]
  • In specific embodiments the albumin fusion proteins of the invention are purified using Affinity Chromatography including, but not limited to, Mimetic Dye affinity, peptide affinity and antibody affinity columns that are selective for either the HSA or the “fusion target” molecules. [0348]
  • In preferred embodiments albumin fusion proteins of the invention are purified using one or more Chromatography methods listed above. In other preferred embodiments, albumin fusion proteins of the invention are purified using one or more of the following Chromatography columns, Q sepharose FF column, SP Sepharose FF column, Q Sepharose High Performance Column, Blue Sepharose FF column, Blue Column, Phenyl Sepharose FF column, DEAE Sepharose FF, or Methyl Column. [0349]
  • Additionally, albumin fusion proteins of the invention may be purified using the process described in PCT International Publication WO 00/44772 which is herein incorporated by reference in its entirety. One of skill in the art could easily modify the process described therein for use in the purification of albumin fusion proteins of the invention. [0350]
  • Albumin fusion proteins of the present invention may be recovered from: products of chemical synthetic procedures; and products produced by recombinant techniques from a prokaryotic or eukaryotic host, including, for example, bacterial, yeast, higher plant, insect, and mammalian cells. Depending upon the host employed in a recombinant production procedure, the polypeptides of the present invention may be glycosylated or may be non-glycosylated. In addition, albumin fusion proteins of the invention may also include an initial modified methionine residue, in some cases as a result of host-mediated processes. Thus, it is well known in the art that the N-terminal methionine encoded by the translation initiation codon generally is removed with high efficiency from any protein after translation in all eukaryotic cells. While the N-terminal methionine on most proteins also is efficiently removed in most prokaryotes, for some proteins, this prokaryotic removal process is inefficient, depending on the nature of the amino acid to which the N-terminal methionine is covalently linked. [0351]
  • In one embodiment, the yeast [0352] Pichia pastoris is used to express albumin fusion proteins of the invention in a eukaryotic system. Pichia pastoris is a methylotrophic yeast which can metabolize methanol as its sole carbon source. A main step in the methanol metabolization pathway is the oxidation of methanol to formaldehyde using O2. This reaction is catalyzed by the enzyme alcohol oxidase. In order to metabolize methanol as its sole carbon source, Pichia pastoris must generate high levels of alcohol oxidase due, in part, to the relatively low affinity of alcohol oxidase for O2. Consequently, in a growth medium depending on methanol as a main carbon source, the promoter region of one of the two alcohol oxidase genes (AOX1) is highly active. In the presence of methanol, alcohol oxidase produced from the AOX1 gene comprises up to approximately 30% of the total soluble protein in Pichia pastoris. See Ellis, S. B., et al., Mol. Cell. Biol. 5:1111-21 (1985); Koutz, P. J, et al., Yeast 5:167-77 (1989); Tschopp, J. F., et al., Nucl. Acids Res. 15:3859-76 (1987). Thus, a heterologous coding sequence, such as, for example, a polynucleotide of the present invention, under the transcriptional regulation of all or part of the AOX1 regulatory sequence is expressed at exceptionally high levels in Pichia yeast grown in the presence of methanol.
  • In one example, the plasmid vector pPIC9K is used to express DNA encoding an albumin fusion protein of the invention, as set forth herein, in a Pichea yeast system essentially as described in “Pichia Protocols: Methods in Molecular Biology,” D. R. Higgins and J. Cregg, eds. The Humana Press, Totowa, N.J., 1998. This expression vector allows expression and secretion of a polypeptide of the invention by virtue of the strong AOX1 promoter linked to the [0353] Pichia pastoris alkaline phosphatase (PHO) secretory signal peptide (i.e., leader) located upstream of a multiple cloning site.
  • Many other yeast vectors could be used in place of pPIC9K, such as, pYES2, pYD1, pTEF1/Zeo, pYES2/GS, pPICZ, pGAPZ, pGAPZalpha, pPIC9, pPIC3.5, pHIL-D2, pHIL-S1, pPIC3.5K, and PAO815, as one skilled in the art would readily appreciate, as long as the proposed expression construct provides appropriately located signals for transcription, translation, secretion (if desired), and the like, including an in-frame AUG as required. [0354]
  • In another embodiment, high-level expression of a heterologous coding sequence, such as, for example, a polynucleotide encoding an albumin fusion protein of the present invention, may be achieved by cloning the heterologous polynucleotide of the invention into an expression vector such as, for example, pGAPZ or pGAPZalpha, and growing the yeast culture in the absence of methanol. [0355]
  • In addition, albumin fusion proteins of the invention can be chemically synthesized using techniques known in the art (e.g., see Creighton, 1983, Proteins: Structures and Molecular Principles, W. H. Freeman & Co., N.Y., and Hunkapiller et al., [0356] Nature, 310:105-111 (1984)). For example, a polypeptide corresponding to a fragment of a polypeptide can be synthesized by use of a peptide synthesizer. Furthermore, if desired, nonclassical amino acids or chemical amino acid analogs can be introduced as a substitution or addition into the polypeptide sequence. Non-classical amino acids include, but are not limited to, to the D-isomers of the common amino acids, 2,4-diaminobutyric acid, a-amino isobutyric acid, 4-aminobutyric acid, Abu, 2-amino butyric acid, g-Abu, e-Ahx, 6-amino hexanoic acid, Aib, 2-amino isobutyric acid, 3-amino propionic acid, ornithine, norleucine, norvaline, hydroxyproline, sarcosine, citrulline, homocitrulline, cysteic acid, t-butylglycine, t-butylalanine, phenylglycine, cyclohexylalanine, b-alanine, fluoro-amino acids, designer amino acids such as b-methyl amino acids, Ca-methyl amino acids, Na-methyl amino acids, and amino acid analogs in general. Furthermore, the amino acid can be D (dextrorotary) or L (levorotary).
  • The invention encompasses albumin fusion proteins of the present invention which are differentially modified during or after translation, e.g., by glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, linkage to an antibody molecule or other cellular ligand, etc. Any of numerous chemical modifications may be carried out by known techniques, including but not limited, to specific chemical cleavage by cyanogen bromide, trypsin, chymotrypsin, papain, V8 protease, NaBH[0357] 4; acetylation, formylation, oxidation, reduction; metabolic synthesis in the presence of tunicamycin; etc.
  • Additional post-translational modifications encompassed by the invention include, for example, e.g., N-linked or O-linked carbohydrate chains, processing of N-terminal or C-terminal ends), attachment of chemical moieties to the amino acid backbone, chemical modifications of N-linked or O-linked carbohydrate chains, and addition or deletion of an N-terminal methionine residue as a result of procaryotic host cell expression. The albumin fusion proteins may also be modified with a detectable label, such as an enzymatic, fluorescent, isotopic or affinity label to allow for detection and isolation of the protein. [0358]
  • Examples of suitable enzymes include horseradish peroxidase, alkaline phosphatase, beta-galactosidase, or acetylcholinesterase; examples of suitable prosthetic group complexes include streptavidin/biotin and avidin/biotin; examples of suitable fluorescent materials include umbelliferone, fluorescein, fluorescein isothiocyanate, rhodamine, dichlorotriazinylamine fluorescein, dansyl chloride or phycoerythrin; an example of a luminescent material includes luminol; examples of bioluminescent materials include luciferase, luciferin, and aequorin; and examples of suitable radioactive material include iodine ([0359] 121I, 123I, 125I, 131I), carbon (14C), sulfur (35S), tritium (3H), indium (111In, 112In, 113mIn, 115mIn), technetium (99Tc,99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F), 153Sm, 177Lu, 159Gd, 149Pm, 140La, 175Yb, 166Ho, 90Y, 47Sc, 186Re, 188Re, 142Pr, 105Rh, and 97Ru.
  • In specific embodiments, albumin fusion proteins of the present invention or fragments or variants thereof are attached to macrocyclic chelators that associate with radiometal ions, including but not limited to, [0360] 177Lu, 90Y, 166Ho, and 153Sm, to polypeptides. In a preferred embodiment, the radiometal ion associated with the macrocyclic chelators is 111In. In another preferred embodiment, the radiometal ion associated with the macrocyclic chelator is 90Y. In specific embodiments, the macrocyclic chelator is 1,4,7,10-tetraazacyclododecane-N,N′,N″,N′″-tetraacetic acid (DOTA). In other specific embodiments, DOTA is attached to an antibody of the invention or fragment thereof via linker molecule. Examples of linker molecules useful for conjugating DOTA to a polypeptide are commonly known in the art—see, for example, DeNardo et al., Clin Cancer Res. 4(10):2483-90 (1998); Peterson et al., Bioconjug. Chem. 10(4):553-7 (1999); and Zimmerman et al, Nucl. Med. Biol. 26(8):943-50 (1999); which are hereby incorporated by reference in their entirety.
  • As mentioned, the albumin fusion proteins of the invention may be modified by either natural processes, such as post-translational processing, or by chemical modification techniques which are well known in the art. It will be appreciated that the same type of modification may be present in the same or varying degrees at several sites in a given polypeptide. Polypeptides of the invention may be branched, for example, as a result of ubiquitination, and they may be cyclic, with or without branching. Cyclic, branched, and branched cyclic polypeptides may result from posttranslation natural processes or may be made by synthetic methods. Modifications include acetylation, acylation, ADP-ribosylation, amidation, covalent attachment of flavin, covalent attachment of a heme moiety, covalent attachment of a nucleotide or nucleotide derivative, covalent attachment of a lipid or lipid derivative, covalent attachment of phosphotidylinositol, cross-linking, cyclization, disulfide bond formation, demethylation, formation of covalent cross-links, formation of cysteine, formation of pyroglutamate, formylation, gamma-carboxylation, glycosylation, GPI anchor formation, hydroxylation, iodination, methylation, myristylation, oxidation, pegylation, proteolytic processing, phosphorylation, prenylation, racemization, selenoylation, sulfation, transfer-RNA mediated addition of amino acids to proteins such as arginylation, and ubiquitination. (See, for instance, PROTEINS—STRUCTURE AND MOLECULAR PROPERTIES, 2nd Ed., T. E. Creighton, W. H. Freeman and Company, New York (1993); POST-TRANSLATIONAL COVALENT MODIFICATION OF PROTEINS, B. C. Johnson, Ed., Academic Press, New York, pgs. 1-12 (1983); Seifter et al., Meth. Enzymol. 182:626-646 (1990); Rattan et al., Ann. N.Y. Acad. Sci. 663:48-62 (1992)). [0361]
  • Albumin fusion proteins of the invention and antibodies that bind a Therapeutic protein or fragments or variants thereof can be fused to marker sequences, such as a peptide to facilitate purification. In preferred embodiments, the marker amino acid sequence is a hexa-histidine peptide, such as the tag provided in a pQE vector (QIAGEN, Inc., 9259 Eton Avenue, Chatsworth, Calif., 91311), among others, many of which are commercially available. As described in Gentz et al., Proc. Natl. Acad. Sci. USA 86:821-824 (1989), for instance, hexa-histidine provides for convenient purification of the fusion protein. Other peptide tags useful for purification include, but are not limited to, the “HA” tag, which corresponds to an epitope derived from the influenza hemagglutinin protein (Wilson et al., Cell 37:767 (1984)) and the “flag” tag. [0362]
  • Further, an albumin fusion protein of the invention may be conjugated to a therapeutic moiety such as a cytotoxin, e.g., a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, 213Bi. A cytotoxin or cytotoxic agent includes any agent that is detrimental to cells. Examples include paclitaxol, cytochalasin B, gramicidin D, ethidium bromide, emetine, mitomycin, etoposide, tenoposide, vincristine, vinblastine, colchicin, doxorubicin, daunorubicin, dihydroxy anthracin dione, mitoxantrone, mithramycin, actinomycin D, 1-dehydrotestosterone, glucocorticoids, procaine, tetracaine, lidocaine, propranolol, and puromycin and analogs or homologs thereof. Therapeutic agents include, but are not limited to, antimetabolites (e.g., methotrexate, 6-mercaptopurine, 6-thioguanine, cytarabine, 5-fluorouracil decarbazine), alkylating agents (e.g., mechlorethamine, thioepa chlorambucil, melphalan, carmustine (BSNU) and lomustine (CCNU), cyclothosphamide, busulfan, dibromomannitol, streptozotocin, mitomycin C, and cis-dichlorodiamine platinum (II) (DDP) cisplatin), anthracyclines (e.g., daunorubicin (formerly daunomycin) and doxorubicin), antibiotics (e.g., dactinomycin (formerly actinomycin), bleomycin, mithramycin, and anthramycin (AMC)), and anti-mitotic agents (e.g., vincristine and vinblastine). [0363]
  • The conjugates of the invention can be used for modifying a given biological response, the therapeutic agent or drug moiety is not to be construed as limited to classical chemical therapeutic agents. For example, the drug moiety may be a protein or polypeptide possessing a desired biological activity. Such proteins may include, for example, a toxin such as abrin, ricin A, pseudomonas exotoxin, or diphtheria toxin; a protein such as tumor necrosis factor, alpha-interferon, β-interferon, nerve growth factor, platelet derived growth factor, tissue plasminogen activator, an apoptotic agent, e.g., TNF-alpha, TNF-beta, AIM I (See, International Publication No. WO 97/33899), AIM II (See, International Publication No. WO 97/34911), Fas Ligand (Takahashi et al, Int. Immunol., 6:1567-1574 (1994)), VEGI (See, International Publication No. WO 99/23105), a thrombotic agent or an anti-angiogenic agent, e.g., angiostatin or endostatin; or, biological response modifiers such as, for example, lymphokines, interleukin-1 (“IL-1”), interleukin-2 (“IL-2”), interleukin-6 (“IL-6”), granulocyte macrophage colony stimulating factor (“GM-CSF”), granulocyte colony stimulating factor (“G-CSF”), or other growth factors. Techniques for conjugating such therapeutic moiety to proteins (e.g., albumin fusion proteins) are well known in the art. [0364]
  • Albumin fusion proteins may also be attached to solid supports, which are particularly useful for immunoassays or purification of polypeptides that are bound by, that bind to, or associate with albumin fusion proteins of the invention. Such solid supports include, but are not limited to, glass, cellulose, polyacrylamide, nylon, polystyrene, polyvinyl chloride or polypropylene. [0365]
  • Albumin fusion proteins, with or without a therapeutic moiety conjugated to it, administered alone or in combination with cytotoxic factor(s) and/or cytokine(s) can be used as a therapeutic. [0366]
  • In embodiments where the albumin fusion protein of the invention comprises only the VH domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VL domain of the same antibody that binds a Therapeutic protein, such that the VH-albumin fusion protein and VL protein will associate (either covalently or non-covalently) post-translationally. [0367]
  • In embodiments where the albumin fusion protein of the invention comprises only the VL domain of an antibody that binds a Therapeutic protein, it may be necessary and/or desirable to coexpress the fusion protein with the VH domain of the same antibody that binds a Therapeutic protein, such that the VL-albumin fusion protein and VH protein will associate (either covalently or non-covalently) post-translationally. [0368]
  • Some Therapeutic antibodies are bispecific antibodies, meaning the antibody that binds a Therapeutic protein is an artificial hybrid antibody having two different heavy/light chain pairs and two different binding sites. In order to create an albumin fusion protein corresponding to that Therapeutic protein, it is possible to create an albumin fusion protein which has an scFv fragment fused to both the N- and C-terminus of the albumin protein moiety. More particularly, the scFv fused to the N-terminus of albumin would correspond to one of the heavy/light (VH/VL) pairs of the original antibody that binds a Therapeutic protein and the scFv fused to the C-terminus of albumin would correspond to the other heavy/light (VH/VL) pair of the original antibody that binds a Therapeutic protein. [0369]
  • Also provided by the invention are chemically modified derivatives of the albumin fusion proteins of the invention which may provide additional advantages such as increased solubility, stability and circulating time of the polypeptide, or decreased immunogenicity (see U.S. Pat. No. 4,179,337). The chemical moieties for derivitization may be selected from water soluble polymers such as polyethylene glycol, ethylene glycol/propylene glycol copolymers, carboxymethylcellulose, dextran, polyvinyl alcohol and the like. The albumin fusion proteins may be modified at random positions within the molecule, or at predetermined positions within the molecule and may include one, two, three or more attached chemical moieties. [0370]
  • The polymer may be of any molecular weight, and may be branched or unbranched. For polyethylene glycol, the preferred molecular weight is between about 1 kDa and about 100 kDa (the term “about” indicating that in preparations of polyethylene glycol, some molecules will weigh more, some less, than the stated molecular weight) for ease in handling and manufacturing. Other sizes may be used, depending on the desired therapeutic profile (e.g., the duration of sustained release desired, the effects, if any on biological activity, the ease in handling, the degree or lack of antigenicity and other known effects of the polyethylene glycol to a Therapeutic protein or analog). For example, the polyethylene glycol may have an average molecular weight of about 200, 500, 1000, 1500, 2000, 2500, 3000, 3500, 4000, 4500, 5000, 5500, 6000, 6500, 7000, 7500, 8000, 8500, 9000, 9500, 10,000, 10,500, 11,000, 11,500, 12,000, 12,500, 13,000, 13,500, 14,000, 14,500, 15,000, 15,500, 16,000, 16,500, 17,000, 17,500, 18,000, 18,500, 19,000, 19,500, 20,000, 25,000, 30,000, 35,000, 40,000, 45,000, 50,000, 55,000, 60,000, 65,000, 70,000, 75,000, 80,000, 85,000, 90,000, 95,000, or 100,000 kDa. [0371]
  • As noted above, the polyethylene glycol may have a branched structure. Branched polyethylene glycols are described, for example, in U.S. Pat. No. 5,643,575; Morpurgo et al., [0372] Appl. Biochem. Biotechnol. 56:59-72 (1996); Vorobjev et al., Nucleosides Nucleotides 18:2745-2750 (1999); and Caliceti et al., Bioconjug. Chem. 10:638-646 (1999), the disclosures of each of which are incorporated herein by reference.
  • The polyethylene glycol molecules (or other chemical moieties) should be attached to the protein with consideration of effects on functional or antigenic domains of the protein. There are a number of attachment methods available to those skilled in the art, such as, for example, the method disclosed in [0373] EP 0 401 384 (coupling PEG to G-CSF), herein incorporated by reference; see also Malik et al., Exp. Hematol. 20:1028-1035 (1992), reporting pegylation of GM-CSF using tresyl chloride. For example, polyethylene glycol may be covalently bound through amino acid residues via reactive group, such as a free amino or carboxyl group. Reactive groups are those to which an activated polyethylene glycol molecule may be bound. The amino acid residues having a free amino group may include lysine residues and the N-terminal amino acid residues; those having a free carboxyl group may include aspartic acid residues glutamic acid residues and the C-terminal amino acid residue. Sulfhydryl groups may also be used as a reactive group for attaching the polyethylene glycol molecules. Preferred for therapeutic purposes is attachment at an amino group, such as attachment at the N-terminus or lysine group.
  • As suggested above, polyethylene glycol may be attached to proteins via linkage to any of a number of amino acid residues. For example, polyethylene glycol can be linked to proteins via covalent bonds to lysine, histidine, aspartic acid, glutamic acid, or cysteine residues. One or more reaction chemistries may be employed to attach polyethylene glycol to specific amino acid residues (e.g., lysine, histidine, aspartic acid, glutamic acid, or cysteine) of the protein or to more than one type of amino acid residue (e.g., lysine, histidine, aspartic acid, glutamic acid, cysteine and combinations thereof) of the protein. [0374]
  • One may specifically desire proteins chemically modified at the N-terminus. Using polyethylene glycol as an illustration of the present composition, one may select from a variety of polyethylene glycol molecules (by molecular weight, branching, etc.), the proportion of polyethylene glycol molecules to protein (polypeptide) molecules in the reaction mix, the type of pegylation reaction to be performed, and the method of obtaining the selected N-terminally pegylated protein. The method of obtaining the N-terminally pegylated preparation (i.e., separating this moiety from other monopegylated moieties if necessary) may be by purification of the N-terminally pegylated material from a population of pegylated protein molecules. Selective proteins chemically modified at the N-terminus modification may be accomplished by reductive alkylation which exploits differential reactivity of different types of primary amino groups (lysine versus the N-terminal) available for derivatization in a particular protein. Under the appropriate reaction conditions, substantially selective derivatization of the protein at the N-terminus with a carbonyl group containing polymer is achieved. [0375]
  • As indicated above, pegylation of the albumin fusion proteins of the invention may be accomplished by any number of means. For example, polyethylene glycol may be attached to the albumin fusion protein either directly or by an intervening linker. Linkerless systems for attaching polyethylene glycol to proteins are described in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992); Francis et al., Intern. J. of Hematol. 68:1-18 (1998); U.S. Pat. No. 4,002,531; U.S. Pat. No. 5,349,052; WO 95/06058; and WO 98/32466, the disclosures of each of which are incorporated herein by reference. [0376]
  • One system for attaching polyethylene glycol directly to amino acid residues of proteins without an intervening linker employs tresylated MPEG, which is produced by the modification of monmethoxy polyethylene glycol (MPEG) using tresylchloride (ClSO[0377] 2CH2CF3). Upon reaction of protein with tresylated MPEG, polyethylene glycol is directly attached to amine groups of the protein. Thus, the invention includes protein-polyethylene glycol conjugates produced by reacting proteins of the invention with a polyethylene glycol molecule having a 2,2,2-trifluoreothane sulphonyl group.
  • Polyethylene glycol can also be attached to proteins using a number of different intervening linkers. For example, U.S. Pat. No. 5,612,460, the entire disclosure of which is incorporated herein by reference, discloses urethane linkers for connecting polyethylene glycol to proteins. Protein-polyethylene glycol conjugates wherein the polyethylene glycol is attached to the protein by a linker can also be produced by reaction of proteins with compounds such as MPEG-succinimidylsuccinate, MPEG activated with 1,1′-carbonyldiimidazole, MPEG-2,4,5-trichloropenylcarbonate, MPEG-p-nitrophenolcarbonate, and various MPEG-succinate derivatives. A number of additional polyethylene glycol derivatives and reaction chemistries for attaching polyethylene glycol to proteins are described in International Publication No. WO 98/32466, the entire disclosure of which is incorporated herein by reference. Pegylated protein products produced using the reaction chemistries set out herein are included within the scope of the invention. [0378]
  • The number of polyethylene glycol moieties attached to each albumin fusion protein of the invention (i.e., the degree of substitution) may also vary. For example, the pegylated proteins of the invention may be linked, on average, to 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 15, 17, 20, or more polyethylene glycol molecules. Similarly, the average degree of substitution within ranges such as 1-3, 2-4, 3-5, 4-6, 5-7, 6-8, 7-9, 8-10, 9-11, 10-12, 11-13, 12-14, 13-15, 14-16, 15-17, 16-18, 17-19, or 18-20 polyethylene glycol moieties per protein molecule. Methods for determining the degree of substitution are discussed, for example, in Delgado et al., Crit. Rev. Thera. Drug Carrier Sys. 9:249-304 (1992). [0379]
  • The polypeptides of the invention can be recovered and purified from chemical synthesis and recombinant cell cultures by standard methods which include, but are not limited to, ammonium sulfate or ethanol precipitation, acid extraction, anion or cation exchange chromatography, phosphocellulose chromatography, hydrophobic interaction chromatography, affinity chromatography, hydroxylapatite chromatography and lectin chromatography. Most preferably, high performance liquid chromatography (“HPLC”) is employed for purification. Well known techniques for refolding protein may be employed to regenerate active conformation when the polypeptide is denatured during isolation and/or purification. [0380]
  • The presence and quantity of albumin fusion proteins of the invention may be determined using ELISA, a well known immunoassay known in the art. In one ELISA protocol that would be useful for detecting/quantifying albumin fusion proteins of the invention, comprises the steps of coating an ELISA plate with an anti-human serum albumin antibody, blocking the plate to prevent non-specific binding, washing the ELISA plate, adding a solution containing the albumin fusion protein of the invention (at one or more different concentrations), adding a secondary anti-Therapeutic protein specific antibody coupled to a detectable label (as described herein or otherwise known in the art), and detecting the presence of the secondary antibody. In an alternate version of this protocol, the ELISA plate might be coated with the anti-Therapeutic protein specific antibody and the labeled secondary reagent might be the anti-human albumin specific antibody. [0381]
  • Uses of the Polynucleotides [0382]
  • Each of the polynucleotides identified herein can be used in numerous ways as reagents. The following description should be considered exemplary and utilizes known techniques. [0383]
  • The polynucleotides of the present invention are useful to produce the albumin fusion proteins of the invention. As described in more detail below, polynucleotides of the invention (encoding albumin fusion proteins) may be used in recombinant DNA methods useful in genetic engineering to make cells, cell lines, or tissues that express the albumin fusion protein encoded by the polynucleotides encoding albumin fusion proteins of the invention. [0384]
  • Polynucleotides of the present invention are also useful in gene therapy. One goal of gene therapy is to insert a normal gene into an organism having a defective gene, in an effort to correct the genetic defect. The polynucleotides disclosed in the present invention offer a means of targeting such genetic defects in a highly accurate manner. Another goal is to insert a new gene that was not present in the host genome, thereby producing a new trait in the host cell. Additional non-limiting examples of gene therapy methods encompassed by the present invention are more thoroughly described elsewhere herein (see, e.g., the sections labeled “Gene Therapy”, and Examples 17 and 18). [0385]
  • Uses of the Polypeptides [0386]
  • Each of the polypeptides identified herein can be used in numerous ways. The following description should be considered exemplary and utilizes known techniques. [0387]
  • Albumin fusion proteins of the invention are useful to provide immunological probes for differential identification of the tissue(s) (e.g., immunohistochemistry assays such as, for example, ABC immunoperoxidase (Hsu et al., J. Histochem. Cytochem. 29:577-580 (1981)) or cell type(s) (e.g., immunocytochemistry assays). [0388]
  • Albumin fusion proteins can be used to assay levels of polypeptides in a biological sample using classical immunohistological methods known to those of skill in the art (e.g., see Jalkanen, et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, et al., J. Cell. Biol. 105:3087-3096 (1987)). Other methods useful for detecting protein gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable assay labels are known in the art and include enzyme labels, such as, glucose oxidase; radioisotopes, such as iodine ([0389] 131I, 125I, 123I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (115mIn, 113mIn, 112In, 111In), and technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F), 153Sm, 177Lu, 159Gd, 149Pm, 140La, 175Yb, 166Ho, 90Y, 47Sc, 186Re, 188Re, 142Pr, 105Rh, 97Ru; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • Albumin fusion proteins of the invention can also be detected in vivo by imaging. Labels or markers for in vivo imaging of protein include those detectable by X-radiography, nuclear magnetic resonance (NMR) or electron spin relaxtion (ESR). For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients given to a cell line expressing the albumin fusion protein of the invention. [0390]
  • An albumin fusion protein which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, [0391] 131I, 112In, 99mTc, (131I, 125I, 123I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (115mIn, 113mIn, 112In, 111In), and technetium (99Tc, 99mTc), thallium (201Ti), gallium (68Ga, 67Ga), palladium (103Pd), molybdenum (99Mo), xenon (133Xe), fluorine (18F, 153Sm, 177Lu, 159Gd, 149Pm, 140La, 175Yb, 166Ho, 90Y, 47Sc, 186Re, 188Re, 142Pr, 105Rh, 97Ru), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for immune system disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled albumin fusion protein will then preferentially accumulate at locations in the body (e.g., organs, cells, extracellular spaces or matrices) where one or more receptors, ligands or substrates (corresponding to that of the Therapeutic protein used to make the albumin fusion protein of the invention) are located. Alternatively, in the case where the albumin fusion protein comprises at least a fragment or variant of a Therapeutic antibody, the labeled albumin fusion protein will then preferentially accumulate at the locations in the body (e.g., organs, cells, extracellular spaces or matrices) where the polypeptides/epitopes corresponding to those bound by the Therapeutic antibody (used to make the albumin fusion protein of the invention) are located. In vivo tumor imaging is described in S. W. Burchiel et al., “Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)). The protocols described therein could easily be modified by one of skill in the art for use with the albumin fusion proteins of the invention.
  • In one embodiment, the invention provides a method for the specific delivery of albumin fusion proteins of the invention to cells by administering albumin fusion proteins of the invention (e.g., polypeptides encoded by polynucleotides encoding albumin fusion proteins of the invention and/or antibodies) that are associated with heterologous polypeptides or nucleic acids. In one example, the invention provides a method for delivering a Therapeutic protein into the targeted cell. In another example, the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell. [0392]
  • In another embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering albumin fusion proteins of the invention in association with toxins or cytotoxic prodrugs. [0393]
  • By “toxin” is meant one or more compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or on the surface of a cell that under defined conditions cause the cell's death. Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin. “Toxin” also includes a cytostatic or cytocidal agent, a therapeutic agent or a radioactive metal ion, e.g., alpha-emitters such as, for example, [0394] 213Bi, or other radioisotopes such as, for example, 103Pd, 133Xe, 131I, 68Ge, 57Co, 65Zn, 85Sr, 32P, 35S, 90Y, 153Sm, 153Gd, 169Yb, 51Cr, 54Mn, 75Se, 113Sn, 90Yttrium, 117Tin, 186Rhenium, 166Holmium, and 188Rhenium; luminescent labels, such as luminol; and fluorescent labels, such as fluorescein and rhodamine, and biotin. In a specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 90Y. In another specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 111In. In a further specific embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering polypeptides of the invention or antibodies of the invention in association with the radioisotope 131I.
  • Techniques known in the art may be applied to label polypeptides of the invention. Such techniques include, but are not limited to, the use of bifunctional conjugating agents (see e.g., U.S. Pat. Nos. 5,756,065; 5,714,631; 5,696,239; 5,652,361; 5,505,931; 5,489,425; 5,435,990; 5,428,139; 5,342,604; 5,274,119; 4,994,560; and 5,808,003; the contents of each of which are hereby incorporated by reference in its entirety). [0395]
  • The albumin fusion proteins of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans. Such disorders include, but are not limited to, those described herein under the section heading “Biological Activities,” below. [0396]
  • Thus, the invention provides a diagnostic method of a disorder, which involves (a) assaying the expression level of a certain polypeptide in cells or body fluid of an individual using an albumin fusion protein of the invention; and (b) comparing the assayed polypeptide expression level with a standard polypeptide expression level, whereby an increase or decrease in the assayed polypeptide expression level compared to the standard expression level is indicative of a disorder. With respect to cancer, the presence of a relatively high amount of transcript in biopsied tissue from an individual may indicate a predisposition for the development of the disease, or may provide a means for detecting the disease prior to the appearance of actual clinical symptoms. A more definitive diagnosis of this type may allow health professionals to employ preventative measures or aggressive treatment earlier thereby preventing the development or further progression of the cancer. [0397]
  • Moreover, albumin fusion proteins of the present invention can be used to treat or prevent diseases or conditions such as, for example, neural disorders, immune system disorders, muscular disorders, reproductive disorders, gastrointestinal disorders, pulmonary disorders, cardiovascular disorders, renal disorders, proliferative disorders, and/or cancerous diseases and conditions. For example, patients can be administered a polypeptide of the present invention in an effort to replace absent or decreased levels of the polypeptide (e.g., insulin), to supplement absent or decreased levels of a different polypeptide (e.g., hemoglobin S for hemoglobin B, SOD, catalase, DNA repair proteins), to inhibit the activity of a polypeptide (e.g., an oncogene or tumor supressor), to activate the activity of a polypeptide (e.g., by binding to a receptor), to reduce the activity of a membrane bound receptor by competing with it for free ligand (e.g., soluble TNF receptors used in reducing inflammation), or to bring about a desired response (e.g., blood vessel growth inhibition, enhancement of the immune response to proliferative cells or tissues). [0398]
  • In particular, albumin fusion proteins comprising of at least a fragment or variant of a Therapeutic antibody can also be used to treat disease (as described supra, and elsewhere herein). For example, administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can bind, and/or neutralize the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, and/or reduce overproduction of the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds. Similarly, administration of an albumin fusion protein comprising of at least a fragment or variant of a Therapeutic antibody can activate the polypeptide to which the Therapeutic antibody used to make the albumin fusion protein specifically binds, by binding to the polypeptide bound to a membrane (receptor). [0399]
  • At the very least, the albumin fusion proteins of the invention of the present invention can be used as molecular weight markers on SDS-PAGE gels or on molecular sieve gel filtration columns using methods well known to those of skill in the art. Albumin fusion proteins of the invention can also be used to raise antibodies, which in turn may be used to measure protein expression of the Therapeutic protein, albumin protein, and/or the albumin fusion protein of the invention from a recombinant cell, as a way of assessing transformation of the host cell, or in a biological sample. Moreover, the albumin fusion proteins of the present invention can be used to test the biological activities described herein. [0400]
  • Diagnostic Assays [0401]
  • The compounds of the present invention are useful for diagnosis, treatment, prevention and/or prognosis of various disorders in mammals, preferably humans. Such disorders include, but are not limited to, those described for each Therapeutic protein in the corresponding row of Table 1 and herein under the section headings “Immune Activity,” “Blood Related Disorders,” “Hyperproliferative Disorders,” “Renal Disorders,” “Cardiovascular Disorders,” “Respiratory Disorders,” “Anti-Angiogenesis Activity,” “Diseases at the Cellular Level,” “Wound Healing and Epithelial Cell Proliferation,” “Neural Activity and Neurological Diseases,” “Endocrine Disorders,” “Reproductive System Disorders,” “Infectious Disease,” “Regeneration,” and/or “Gastrointestinal Disorders,” infra. [0402]
  • For a number of disorders, substantially altered (increased or decreased) levels of gene expression can be detected in tissues, cells or bodily fluids (e.g., sera, plasma, urine, semen, synovial fluid or spinal fluid) taken from an individual having such a disorder, relative to a “standard” gene expression level, that is, the expression level in tissues or bodily fluids from an individual not having the disorder. Thus, the invention provides a diagnostic method useful during diagnosis of a disorder, which involves measuring the expression level of the gene encoding a polypeptide in tissues, cells or body fluid from an individual and comparing the measured gene expression level with a standard gene expression level, whereby an increase or decrease in the gene expression level(s) compared to the standard is indicative of a disorder. These diagnostic assays may be performed in vivo or in vitro, such as, for example, on blood samples, biopsy tissue or autopsy tissue. [0403]
  • The present invention is also useful as a prognostic indicator, whereby patients exhibiting enhanced or depressed gene expression will experience a worse clinical outcome By “assaying the expression level of the gene encoding a polypeptide” is intended qualitatively or quantitatively measuring or estimating the level of a particular polypeptide (e.g. a polypeptide corresponding to a Therapeutic protein disclosed in Table 1) or the level of the mRNA encoding the polypeptide of the invention in a first biological sample either directly (e.g., by determining or estimating absolute protein level or mRNA level) or relatively (e.g., by comparing to the polypeptide level or mRNA level in a second biological sample). Preferably, the polypeptide expression level or mRNA level in the first biological sample is measured or estimated and compared to a standard polypeptide level or mRNA level, the standard being taken from a second biological sample obtained from an individual not having the disorder or being determined by averaging levels from a population of individuals not having the disorder. As will be appreciated in the art, once a standard polypeptide level or mRNA level is known, it can be used repeatedly as a standard for comparison. [0404]
  • By “biological sample” is intended any biological sample obtained from an individual, cell line, tissue culture, or other source containing polypeptides of the invention (including portions thereof) or mRNA. As indicated, biological samples include body fluids (such as sera, plasma, urine, synovial fluid and spinal fluid) and tissue sources found to express the full length or fragments thereof of a polypeptide or mRNA. Methods for obtaining tissue biopsies and body fluids from mammals are well known in the art. Where the biological sample is to include mRNA, a tissue biopsy is the preferred source. [0405]
  • Total cellular RNA can be isolated from a biological sample using any suitable technique such as the single-step guanidinium-thiocyanate-phenol-chloroform method described in Chomczynski and Sacchi, Anal. Biochem. 162:156-159 (1987). Levels of mRNA encoding the polypeptides of the invention are then assayed using any appropriate method. These include Northern blot analysis, S1 nuclease mapping, the polymerase chain reaction (PCR), reverse transcription in combination with the polymerase chain reaction (RT-PCR), and reverse transcription in combination with the ligase chain reaction (RT-LCR). [0406]
  • The present invention also relates to diagnostic assays such as quantitative and diagnostic assays for detecting levels of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the invention, in a biological sample (e.g., cells and tissues), including determination of normal and abnormal levels of polypeptides. Thus, for instance, a diagnostic assay in accordance with the invention for detecting abnormal expression of polypeptides that bind to, are bound by, or associate with albumin fusion proteins compared to normal control tissue samples may be used to detect the presence of tumors. Assay techniques that can be used to determine levels of a polypeptide that bind to, are bound by, or associate with albumin fusion proteins of the present invention in a sample derived from a host are well-known to those of skill in the art. Such assay methods include radioimmunoassays, competitive-binding assays, Western Blot analysis and ELISA assays. Assaying polypeptide levels in a biological sample can occur using any art-known method. [0407]
  • Assaying polypeptide levels in a biological sample can occur using a variety of techniques. For example, polypeptide expression in tissues can be studied with classical immunohistological methods (Jalkanen et al., J. Cell. Biol. 101:976-985 (1985); Jalkanen, M., et al., J. Cell. Biol. 105:3087-3096 (1987)). Other methods useful for detecting polypeptide gene expression include immunoassays, such as the enzyme linked immunosorbent assay (ELISA) and the radioimmunoassay (RIA). Suitable antibody assay labels are known in the art and include enzyme labels, such as, glucose oxidase, and radioisotopes, such as iodine ([0408] 125I, 121I), carbon (14C), sulfur (35S), tritium (3H), indium (112In), and technetium (99mTc), and fluorescent labels, such as fluorescein and rhodamine, and biotin.
  • The tissue or cell type to be analyzed will generally include those which are known, or suspected, to express the gene of interest (such as, for example, cancer). The protein isolation methods employed herein may, for example, be such as those described in Harlow and Lane (Harlow, E. and Lane, D., 1988, “Antibodies: A Laboratory Manual”, Cold Spring Harbor Laboratory Press, Cold Spring Harbor, N.Y.), which is incorporated herein by reference in its entirety. The isolated cells can be derived from cell culture or from a patient. The analysis of cells taken from culture may be a necessary step in the assessment of cells that could be used as part of a cell-based gene therapy technique or, alternatively, to test the effect of compounds on the expression of the gene. [0409]
  • For example, albumin fusion proteins may be used to quantitatively or qualitatively detect the presence of polypeptides that bind to, are bound by, or associate with albumin fusion proteins of the present invention. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled albumin fusion protein coupled with light microscopic, flow cytometric, or fluorimetric detection. [0410]
  • In a preferred embodiment, albumin fusion proteins comprising at least a fragment or variant of an antibody that specifically binds at least a Therapeutic protein disclosed herein (e.g., the Therapeutic proteins disclosed in Table 1) or otherwise known in the art may be used to quantitatively or qualitatively detect the presence of gene products or conserved variants or peptide fragments thereof. This can be accomplished, for example, by immunofluorescence techniques employing a fluorescently labeled antibody coupled with light microscopic, flow cytometric, or fluorimetric detection. [0411]
  • The albumin fusion proteins of the present invention may, additionally, be employed histologically, as in immunofluorescence, immunoelectron microscopy or non-immunological assays, for in situ detection of polypeptides that bind to, are bound by, or associate with an albumin fusion protein of the present invention. In situ detection may be accomplished by removing a histological specimen from a patient, and applying thereto a labeled antibody or polypeptide of the present invention. The albumin fusion proteins are preferably applied by overlaying the labeled albumin fusion proteins onto a biological sample. Through the use of such a procedure, it is possible to determine not only the presence of the polypeptides that bind to, are bound by, or associate with albumin fusion proteins, but also its distribution in the examined tissue. Using the present invention, those of ordinary skill will readily perceive that any of a wide variety of histological methods (such as staining procedures) can be modified in order to achieve such in situ detection. [0412]
  • Immunoassays and non-immunoassays that detect polypeptides that bind to, are bound by, or associate with albumin fusion proteins will typically comprise incubating a sample, such as a biological fluid, a tissue extract, freshly harvested cells, or lysates of cells which have been incubated in cell culture, in the presence of a detectably labeled antibody capable of binding gene products or conserved variants or peptide fragments thereof, and detecting the bound antibody by any of a number of techniques well-known in the art. [0413]
  • The biological sample may be brought in contact with and immobilized onto a solid phase support or carrier such as nitrocellulose, or other solid support which is capable of immobilizing cells, cell particles or soluble proteins. The support may then be washed with suitable buffers followed by treatment with the detectably labeled albumin fusion protein of the invention. The solid phase support may then be washed with the buffer a second time to remove unbound antibody or polypeptide. Optionally the antibody is subsequently labeled. The amount of bound label on solid support may then be detected by conventional means. [0414]
  • By “solid phase support or carrier” is intended any support capable of binding a polypeptide (e.g., an albumin fusion protein, or polypeptide that binds, is bound by, or associates with an albumin fusion protein of the invention.) Well-known supports or carriers include glass, polystyrene, polypropylene, polyethylene, dextran, nylon, amylases, natural and modified celluloses, polyacrylamides, gabbros, and magnetite. The nature of the carrier can be either soluble to some extent or insoluble for the purposes of the present invention. The support material may have virtually any possible structural configuration so long as the coupled molecule is capable of binding to a polypeptide. Thus, the support configuration may be spherical, as in a bead, or cylindrical, as in the inside surface of a test tube, or the external surface of a rod. Alternatively, the surface may be flat such as a sheet, test strip, etc. Preferred supports include polystyrene beads. Those skilled in the art will know many other suitable carriers for binding antibody or antigen, or will be able to ascertain the same by use of routine experimentation. [0415]
  • The binding activity of a given lot of albumin fusion protein may be determined according to well known methods. Those skilled in the art will be able to determine operative and optimal assay conditions for each determination by employing routine experimentation. [0416]
  • In addition to assaying polypeptide levels in a biological sample obtained from an individual, polypeptide can also be detected in vivo by imaging. For example, in one embodiment of the invention, albumin fusion proteins of the invention are used to image diseased or neoplastic cells. [0417]
  • Labels or markers for in vivo imaging of albumin fusion proteins of the invention include those detectable by X-radiography, NMR, MRI, CAT-scans or ESR. For X-radiography, suitable labels include radioisotopes such as barium or cesium, which emit detectable radiation but are not overtly harmful to the subject. Suitable markers for NMR and ESR include those with a detectable characteristic spin, such as deuterium, which may be incorporated into the albumin fusion protein by labeling of nutrients of a cell line (or bacterial or yeast strain) engineered. [0418]
  • Additionally, albumin fusion proteins of the invention whose presence can be detected, can be administered. For example, albumin fusion proteins of the invention labeled with a radio-opaque or other appropriate compound can be administered and visualized in vivo, as discussed, above for labeled antibodies. Further, such polypeptides can be utilized for in vitro diagnostic procedures. [0419]
  • A polypeptide-specific antibody or antibody fragment which has been labeled with an appropriate detectable imaging moiety, such as a radioisotope (for example, [0420] 131I, 112In, 99mTc), a radio-opaque substance, or a material detectable by nuclear magnetic resonance, is introduced (for example, parenterally, subcutaneously or intraperitoneally) into the mammal to be examined for a disorder. It will be understood in the art that the size of the subject and the imaging system used will determine the quantity of imaging moiety needed to produce diagnostic images. In the case of a radioisotope moiety, for a human subject, the quantity of radioactivity injected will normally range from about 5 to 20 millicuries of 99mTc. The labeled albumin fusion protein will then preferentially accumulate at the locations in the body which contain a polypeptide or other substance that binds to, is bound by or associates with an albumin fusion protein of the present invention. In vivo tumor imaging is described in S. W. Burchiel et al., “Immunopharmacokinetics of Radiolabeled Antibodies and Their Fragments” (Chapter 13 in Tumor Imaging: The Radiochemical Detection of Cancer, S. W. Burchiel and B. A. Rhodes, eds., Masson Publishing Inc. (1982)).
  • One of the ways in which an albumin fusion protein of the present invention can be detectably labeled is by linking the same to a reporter enzyme and using the linked product in an enzyme immunoassay (EIA) (Voller, A., “The Enzyme Linked Immunosorbent Assay (ELISA)”, 1978, Diagnostic Horizons 2:1-7, Microbiological Associates Quarterly Publication, Walkersville, Md.); Voller et al., [0421] J. Clin. Pathol. 31:507-520 (1978); Butler, J. E., Meth. Enzymol. 73:482-523 (1981); Maggio, E. (ed.), 1980, Enzyme Immunoassay, CRC Press, Boca Raton, Fla.,; Ishikawa, E. et al., (eds.), 1981, Enzyme Immunoassay, Kgaku Shoin, Tokyo). The reporter enzyme which is bound to the antibody will react with an appropriate substrate, preferably a chromogenic substrate, in such a manner as to produce a chemical moiety which can be detected, for example, by spectrophotometric, fluorimetric or by visual means. Reporter enzymes which can be used to detectably label the antibody include, but are not limited to, malate dehydrogenase, staphylococcal nuclease, delta-5-steroid isomerase, yeast alcohol dehydrogenase, alpha-glycerophosphate, dehydrogenase, triose phosphate isomerase, horseradish peroxidase, alkaline phosphatase, asparaginase, glucose oxidase, beta-galactosidase, ribonuclease, urease, catalase, glucose-6-phosphate dehydrogenase, glucoamylase and acetylcholinesterase. Additionally, the detection can be accomplished by colorimetric methods which employ a chromogenic substrate for the reporter enzyme. Detection may also be accomplished by visual comparison of the extent of enzymatic reaction of a substrate in comparison with similarly prepared standards.
  • Albumin fusion proteins may also be radiolabelled and used in any of a variety of other immunoassays. For example, by radioactively labeling the albumin fusion proteins, it is possible to the use the albumin fusion proteins in a radioimmunoassay (RIA) (see, for example, Weintraub, B., Principles of Radioimmunoassays, Seventh Training Course on Radioligand Assay Techniques, The Endocrine Society, March, 1986, which is incorporated by reference herein). The radioactive isotope can be detected by means including, but not limited to, a gamma counter, a scintillation counter, or autoradiography. [0422]
  • It is also possible to label the albumin fusion proteins with a fluorescent compound. When the fluorescently labeled antibody is exposed to light of the proper wave length, its presence can then be detected due to fluorescence. Among the most commonly used fluorescent labeling compounds are fluorescein isothiocyanate, rhodamine, phycoerythrin, phycocyanin, allophycocyanin, ophthaldehyde and fluorescamine. [0423]
  • The albumin fusion protein can also be detectably labeled using fluorescence emitting metals such as [0424] 152Eu, or others of the lanthamide series. These metals can be attached to the antibody using such metal chelating groups as diethylenetriaminepentacetic acid (DTPA) or ethylenediaminetetraacetic acid (EDTA).
  • The albumin fusion proteins can also can be detectably labeled by coupling it to a chemiluminescent compound. The presence of the chemiluminescent-tagged albumin fusion protein is then determined by detecting the presence of luminescence that arises during the course of a chemical reaction. Examples of particularly useful chemiluminescent labeling compounds are luminol, isoluminol, theromatic acridinium ester, imidazole, acridinium salt and oxalate ester. [0425]
  • Likewise, a bioluminescent compound may be used to label albumin fusion proteins of the present invention. Bioluminescence is a type of chemiluminescence found in biological systems in, which a catalytic protein increases the efficiency of the chemiluminescent reaction. The presence of a bioluminescent protein is determined by detecting the presence of luminescence. Important bioluminescent compounds for purposes of labeling are luciferin, luciferase and aequorin. [0426]
  • Transgenic Organisms [0427]
  • Transgenic organisms that express the albumin fusion proteins of the invention are also included in the invention. Transgenic organisms are genetically modified organisms into which recombinant, exogenous or cloned genetic material has been transferred. Such genetic material is often referred to as a transgene. The nucleic acid sequence of the transgene may include one or more transcriptional regulatory sequences and other nucleic acid sequences such as introns, that may be necessary for optimal expression and secretion of the encoded protein. The transgene may be designed to direct the expression of the encoded protein in a manner that facilitates its recovery from the organism or from a product produced by the organism, e.g. from the milk, blood, urine, eggs, hair or seeds of the organism. The transgene may consist of nucleic acid sequences derived from the genome of the same species or of a different species than the species of the target animal. The transgene may be integrated either at a locus of a genome where that particular nucleic acid sequence is not otherwise normally found or at the normal locus for the transgene. [0428]
  • The term “germ cell line transgenic organism” refers to a transgenic organism in which the genetic alteration or genetic information was introduced into a germ line cell, thereby conferring the ability of the transgenic organism to transfer the genetic information to offspring. If such offspring in fact possess some or all of that alteration or genetic information, then they too are transgenic organisms. The alteration or genetic information may be foreign to the species of organism to which the recipient belongs, foreign only to the particular individual recipient, or may be genetic information already possessed by the recipient. In the last case, the altered or introduced gene may be expressed differently than the native gene. [0429]
  • A transgenic organism may be a transgenic animal or a transgenic plant. Transgenic animals can be produced by a variety of different methods including transfection, electroporation, microinjection, gene targeting in embryonic stem cells and recombinant viral and retroviral infection (see, e.g., U.S. Pat. No. 4,736,866; U.S. Pat. No. 5,602,307; Mullins et al. (1993) Hypertension 22(4):630-633; Brenin et al. (1997) Surg. Oncol. 6(2)99-110; Tuan (ed.), [0430] Recombinant Gene Expression Protocols, Methods in Molecular Biology No. 62, Humana Press (1997)). The method of introduction of nucleic acid fragments into recombination competent mammalian cells can be by any method which favors co-transformation of multiple nucleic acid molecules. Detailed procedures for producing transgenic animals are readily available to one skilled in the art, including the disclosures in U.S. Pat. No. 5,489,743 and U.S. Pat. No. 5,602,307.
  • A number of recombinant or transgenic mice have been produced, including those which express an activated oncogene sequence (U.S. Pat. No. 4,736,866); express simian SV40 T-antigen (U.S. Pat. No. 5,728,915); lack the expression of interferon regulatory factor 1 (IRF-1) (U.S. Pat. No. 5,731,490); exhibit dopaminergic dysfunction (U.S. Pat. No. 5,723,719); express at least one human gene which participates in blood pressure control (U.S. Pat. No. 5,731,489); display greater similarity to the conditions existing in naturally occurring Alzheimer's disease (U.S. Pat. No. 5,720,936); have a reduced capacity to mediate cellular adhesion (U.S. Pat. No. 5,602,307); possess a bovine growth hormone gene (Clutter et al. (1996) Genetics 143(4):1753-1760); or, are capable of generating a fully human antibody response (McCarthy (1997) The Lancet 349(9049):405). [0431]
  • While mice and rats remain the animals of choice for most transgenic experimentation, in some instances it is preferable or even necessary to use alternative animal species. Transgenic procedures have been successfully utilized in a variety of non-murine animals, including sheep, goats, pigs, dogs, cats, monkeys, chimpanzees, hamsters, rabbits, cows and guinea pigs (see, e.g., Kim et al. (1997) Mol. Reprod. Dev. 46(4):515-526; Houdebine (1995) Reprod. Nutr. Dev. 35(6):609-617; Petters (1994) Reprod. Fertil. Dev. 6(5):643-645; Schnieke et al. (1997) Science 278(5346):2130-2133; and Amoah (1997) J. Animal Science 75(2):578-585). [0432]
  • To direct the secretion of the transgene-encoded protein of the invention into the milk of transgenic mammals, it may be put under the control of a promoter that is preferentially activated in mammary epithelial cells. Promoters that control the genes encoding milk proteins are preferred, for example the promoter for casein, beta lactoglobulin, whey acid protein, or lactalbumin (see, e.g., DiTullio (1992) BioTechnology 10:74-77; Clark et al. (1989) BioTechnology 7:487-492; Gorton et al. (1987) BioTechnology 5:1183-1187; and Soulier et al. (1992) FEBS Letts. 297:13). The transgenic mammals of choice would produce large volumes of milk and have long lactating periods, for example goats, cows, camels or sheep. [0433]
  • An albumin fusion protein of the invention can also be expressed in a transgenic plant, e.g. a plant in which the DNA transgene is inserted into the nuclear or plastidic genome. Plant transformation procedures used to introduce foreign nucleic acids into plant cells or protoplasts are known in the art (e.g., see Example 19). See, in general, Methods in Enzymology Vol. 153 (“Recombinant DNA Part D”) 1987, Wu and Grossman Eds., Academic Press and European Patent Application EP 693554. Methods for generation of genetically engineered plants are further described in U.S. Pat. No. 5,283,184, U.S. Pat. No. 5, 482,852, and European Patent Application EP 693 554, all of which are hereby incorporated by reference. [0434]
  • Pharmaceutical or Therapeutic Compositions [0435]
  • The albumin fusion proteins of the invention or formulations thereof may be administered by any conventional method including parenteral (e.g. subcutaneous or intramuscular) injection or intravenous infusion. The treatment may consist of a single dose or a plurality of doses over a period of time. [0436]
  • While it is possible for an albumin fusion protein of the invention to be administered alone, it is preferable to present it as a pharmaceutical formulation, together with one or more acceptable carriers. The carrier(s) must be “acceptable” in the sense of being compatible with the albumin fusion protein and not deleterious to the recipients thereof. Typically, the carriers will be water or saline which will be sterile and pyrogen free. Albumin fusion proteins of the invention are particularly well suited to formulation in aqueous carriers such as sterile pyrogen free water, saline or other isotonic solutions because of their extended shelf-life in solution. For instance, pharmaceutical compositions of the invention may be formulated well in advance in aqueous form, for instance, weeks or months or longer time periods before being dispensed. [0437]
  • For example, wherein the Therapeutic protein is hGH, EPO, alpha-IFN or beta-IFN, formulations containing the albumin fusion protein may be prepared taking into account the extended shelf-life of the albumin fusion protein in aqueous formulations. As exhibited in Table 2, most Therapeutic proteins are unstable with short shelf-lives after formulation with an aqueous carrier. As discussed above, the shelf-life of many of these Therapeutic proteins are markedly increased or prolonged after fusion to HA. [0438]
    TABLE 2
    Storage Conditions
    Tradename, of Non-Fusion
    Protein Manufacturer Route Formulation Protein
    Interferon, Roferon-A, sc sol n 4-8° C.
    alpha-2a Hoffmann- im (vial or pre-filled
    LaRoche syringe)
    Interferon, Intron-A, iv sc sol n; 4-8° C.
    alpha-2b Schering Plough im powder + dil. (all preps, before and
    after dilution)
    COMBO Rebetron po + Rebetol capsule +
    Interferon (Intron-A + sc Intron-A injection
    alpha-2b + Rebetol)
    Ribavirin Schering Plough
    Interferon, Infergen sc sol n 4-8° C.
    Alphacon-1 Amgen
    Interferon, Wellferon, sc sol n 4-8° C.
    alpha-n1, Wellcome im (with albumin as
    Lymphoblastoid stablizer)
    Interferon, Avonex, im powder + dil. 4-8° C.
    beta-1a Biogen (with albumin) (before and after
    dilution)
    (Use within 3-6 h of
    reconstitution)
    Rebif, sc sol n,
    Ares-Serono in pre-filled syringe
    (Europe only)
    Interferon, Betaseron, sc powder + dil. 4-8° C.
    beta-1b Chiron (with albumin) (before and after
    (Europe: dilution)
    Betaferon) (Use within 3 h of
    reconstitution)
    Single use vials.
    Interferon, Actimmune, sc 4-8° C.
    Gamma-1b InterMune (before and after
    Pharmaceuticals dilution)
    (Use within 3 h of
    reconstitution).
    Growth Genotropin, powder/dil 4-8° C.
    Hormone Pharmacia cartridges (single or (before and after
    (somatropin) Upjohn multi-use); dilution);
    single use single use MiniQuick
    MiniQuick injector Delivery Device
    should be
    refrigerated until use.
    Humatrope, sc powder + dil. 4-8° C.
    Eli Lilly im (Vial or pen (before and after
    cartridge) dilution)
    (Use vials within
    25 h, cartridges
    within 28 d, of
    reconstitution).
    Norditropin,
    Novo Nordisk
    Pharmaceuticals
    Nutropin, sc powder + dil. 4-8° C.
    Genentech (stable for 14 d after
    dil n)
    (all preps, before and
    after dilution)
    Nutropin AQ, sc sol n 4-8° C.
    Genentech (Stable for 28 d after
    1st use)
    Nutropin Depot, sc microsphere 4-8° C.
    Genentech suspension as Single use pkges.
    powder + dil. Dose 1-2x/month
    (ProLease micro-
    encapsulation
    technol.)
    Saizen, sc powder + dil. Powder should be
    (Serono) im stored at Rm
    Temp. After
    reconstitution store
    4-8° C. for up to 14 d.
    Serostim, Powder should be
    Serono stored at Rm
    Temp. After
    reconstitution store
    in 4-8° C. for up to
    14 d.
    hGH, with Protropin, sc powder + dil. 4-8° C.
    N-term. Met Genentech im (all preps, before
    (somatrem) and after dilution)
    Erythropoietin Epogen, iv sol n 4-8° C.
    (Epoetin alfa) Amgen sc (use within 21 d of
    first use)
    (Single & multi-dose
    vials)
    Procrit, iv sol n 4-8° C.
    Amgen sc (use within 21 d of
    first use)
    (Single & multi-dose
    vials)
  • In instances where aerosol administration is appropriate, the albumin fusion proteins of the invention can be formulated as aerosols using standard procedures. The term “aerosol” includes any gas-borne suspended phase of an albumin fusion protein of the instant invention which is capable of being inhaled into the bronchioles or nasal passages. Specifically, aerosol includes a gas-borne suspension of droplets of an albumin fusion protein of the instant invention, as may be produced in a metered dose inhaler or nebulizer, or in a mist sprayer. Aerosol also includes a dry powder composition of a compound of the instant invention suspended in air or other carrier gas, which may be delivered by insufflation from an inhaler device, for example. See Ganderton & Jones, [0439] Drug Delivery to the Respiratory Tract, Ellis Horwood (19 87); Gonda (1990) Critical Reviews in Therapeutic Drug Carrier Systems 6:273-313; and Raeburn et al, (1992) Pharmacol. Toxicol. Methods 27:143-159.
  • The formulations of the invention are also typically non-immunogenic, in part, because of the use of the components of the albumin fusion protein being derived from the proper species. For instance, for human use, both the Therapeutic protein and albumin portions of the albumin fusion protein will typically be human. In some cases, wherein either component is non human-derived, that component may be humanized by substitution of key amino acids so that specific epitopes appear to the human immune system to be human in nature rather than foreign. [0440]
  • The formulations may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy. Such methods include the step of bringing into association the albumin fusion protein with the carrier that constitutes one or more accessory ingredients. In general the formulations are prepared by uniformly and intimately bringing into association the active ingredient with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product. [0441]
  • Formulations suitable for parenteral administration include aqueous and non-aqueous sterile injection solutions which may contain anti-oxidants, buffers, bacteriostats and solutes which render the formulation appropriate for the intended recipient; and aqueous and non-aqueous sterile suspensions which may include suspending agents and thickening agents. The formulations may be presented in unit-dose or multi-dose containers, for example sealed ampules, vials or syringes, and may be stored in a freeze-dried (lyophilised) condition requiring only the addition of the sterile liquid carrier, for example water for injections, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders. Dosage formulations may contain the Therapeutic protein portion at a lower molar concentration or lower dosage compared to the non-fused standard formulation for the Therapeutic protein given the extended serum half-life exhibited by many of the albumin fusion proteins of the invention. [0442]
  • As an example, when an albumin fusion protein of the invention comprises growth hormone as one or more of the Therapeutic protein regions, the dosage form can be calculated on the basis of the potency of the albumin fusion protein relative to the potency of hGH, while taking into account the prolonged serum half-life and shelf-life of the albumin fusion proteins compared to that of native hGH. Growth hormone is typically administered at 0.3 to 30.0 IU/kg/week, for example 0.9 to 12.0 IU/kg/week, given in three or seven divided doses for a year or more. In an albumin fusion protein consisting of full length HA fused to full length GH, an equivalent dose in terms of units would represent a greater weight of agent but the dosage frequency can be reduced, for example to twice a week, once a week or less. [0443]
  • Formulations or compositions of the invention may be packaged together with, or included in a kit with, instructions or a package insert referring to the extended shelf-life of the albumin fusion protein component. For instance, such instructions or package inserts may address recommended storage conditions, such as time, temperature and light, taking into account the extended or prolonged shelf-life of the albumin fusion proteins of the invention. Such instructions or package inserts may also address the particular advantages of the albumin fusion proteins of the inventions, such as the ease of storage for formulations that may require use in the field, outside of controlled hospital, clinic or office conditions. As described above, formulations of the invention may be in aqueous form and may be stored under less than ideal circumstances without significant loss of therapeutic activity. [0444]
  • Albumin fusion proteins of the invention can also be included in nutraceuticals. For instance, certain albumin fusion proteins of the invention may be administered in natural products, including milk or milk product obtained from a transgenic mammal which expresses albumin fusion protein. Such compositions can also include plant or plant products obtained from a transgenic plant which expresses the albumin fusion protein. The albumin fusion protein can also be provided in powder or tablet form, with or without other known additives, carriers, fillers and diluents. Nutraceuticals are described in Scott Hegenhart, [0445] Food Product Design, December 1993.
  • The invention also provides methods of treatment and/or prevention of diseases or disorders (such as, for example, any one or more of the diseases or disorders disclosed herein) by administration to a subject of an effective amount of an albumin fusion protein of the invention or a polynucleotide encoding an albumin fusion protein of the invention (“albumin fusion polynucleotide”) in a pharmaceutically acceptable carrier. [0446]
  • The albumin fusion protein and/or polynucleotide will be formulated and dosed in a fashion consistent with good medical practice, taking into account the clinical condition of the individual patient (especially the side effects of treatment with the albumin fusion protein and/or polynucleotide alone), the site of delivery, the method of administration, the scheduling of administration, and other factors known to practitioners. The “effective amount” for purposes herein is thus determined by such considerations. [0447]
  • As a general proposition, the total pharmaceutically effective amount of the albumin fusion protein administered parenterally per dose will be in the range of about 1 ug/kg/day to 10 mg/kg/day of patient body weight, although, as noted above, this will be subject to therapeutic discretion. More preferably, this dose is at least 0.01 mg/kg/day, and most preferably for humans between about 0.01 and 1 mg/kg/day for the hormone. If given continuously, the albumin fusion protein is typically administered at a dose rate of about 1 ug/kg/hour to about 50 ug/kg/hour, either by 1-4 injections per day or by continuous subcutaneous infusions, for example, using a mini-pump. An intravenous bag solution may also be employed. The length of treatment needed to observe changes and the interval following treatment for responses to occur appears to vary depending on the desired effect. [0448]
  • Albumin fusion proteins and/or polynucleotides can be are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray. “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any. The term “parenteral” as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion. [0449]
  • Albumin fusion proteins and/or polynucleotides of the invention are also suitably administered by sustained-release systems. Examples of sustained-release albumin fusion proteins and/or polynucleotides are administered orally, rectally, parenterally, intracisternally, intravaginally, intraperitoneally, topically (as by powders, ointments, gels, drops or transdermal patch), bucally, or as an oral or nasal spray. “Pharmaceutically acceptable carrier” refers to a non-toxic solid, semisolid or liquid filler, diluent, encapsulating material or formulation auxiliary of any type. The term “parenteral” as used herein refers to modes of administration which include intravenous, intramuscular, intraperitoneal, intrasternal, subcutaneous and intraarticular injection and infusion. Additional examples of sustained-release albumin fusion proteins and/or polynucleotides include suitable polymeric materials (such as, for example, semi-permeable polymer matrices in the form of shaped articles, e.g., films, or mirocapsules), suitable hydrophobic materials (for example as an emulsion in an acceptable oil) or ion exchange resins, and sparingly soluble derivatives (such as, for example, a sparingly soluble salt). [0450]
  • Sustained-release matrices include polylactides (U.S. Pat. No. 3,773,919, EP 58,481), copolymers of L-glutamic acid and gamma-ethyl-L-glutamate (Sidman et al., Biopolymers 22:547-556 (1983)), poly (2-hydroxyethyl methacrylate) (Langer et al., J. Biomed. Mater. Res. 15:167-277 (1981), and Langer, Chem. Tech. 12:98-105 (1982)), ethylene vinyl acetate (Langer et al., Id.) or poly-D-(−)-3-hydroxybutyric acid (EP 133,988). [0451]
  • Sustained-release albumin fusion proteins and/or polynucleotides also include liposomally entrapped albumin fusion proteins and/or polynucleotides of the invention (see generally, Langer, [0452] Science 249:1527-1533 (1990); Treat et al., in Liposomes in the Therapy of Infectious Disease and Cancer, Lopez-Berestein and Fidler (eds.), Liss, New York, pp. 317-327 and 353-365 (1989)). Liposomes containing the albumin fusion protein and/or polynucleotide are prepared by methods known per se: DE 3,218,121; Epstein et al., Proc. Natl. Acad. Sci. (USA) 82:3688-3692 (1985); Hwang et al., Proc. Natl. Acad. Sci.(USA) 77:4030-4034 (1980); EP 52,322; EP 36,676; EP 88,046; EP 143,949; EP 142,641; Japanese Pat. Appl. 83-118008; U.S. Pat. Nos. 4,485,045 and 4,544,545; and EP 102,324. Ordinarily, the liposomes are of the small (about 200-800 Angstroms) unilamellar type in which the lipid content is greater than about 30 mol. percent cholesterol, the selected proportion being adjusted for the optimal Therapeutic.
  • In yet an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are delivered by way of a pump (see Langer, supra; Sefton, CRC Crit. Ref. Biomed. Eng. 14:201 (1987); Buchwald et al., Surgery 88:507 (1980); Saudek et al., N. Engl. J. Med. 321:574 (1989)). [0453]
  • Other controlled release systems are discussed in the review by Langer ([0454] Science 249:1527-1533 (1990)).
  • For parenteral administration, in one embodiment, the albumin fusion protein and/or polynucleotide is formulated generally by mixing it at the desired degree of purity, in a unit dosage injectable form (solution, suspension, or emulsion), with a pharmaceutically acceptable carrier, i.e., one that is non-toxic to recipients at the dosages and concentrations employed and is compatible with other ingredients of the formulation. For example, the formulation preferably does not include oxidizing agents and other compounds that are known to be deleterious to the Therapeutic. [0455]
  • Generally, the formulations are prepared by contacting the albumin fusion protein and/or polynucleotide uniformly and intimately with liquid carriers or finely divided solid carriers or both. Then, if necessary, the product is shaped into the desired formulation. Preferably the carrier is a parenteral carrier, more preferably a solution that is isotonic with the blood of the recipient. Examples of such carrier vehicles include water, saline, Ringer's solution, and dextrose solution. Non-aqueous vehicles such as fixed oils and ethyl oleate are also useful herein, as well as liposomes. [0456]
  • The carrier suitably contains minor amounts of additives such as substances that enhance isotonicity and chemical stability. Such materials are non-toxic to recipients at the dosages and concentrations employed, and include buffers such as phosphate, citrate, succinate, acetic acid, and other organic acids or their salts; antioxidants such as ascorbic acid; low molecular weight (less than about ten residues) polypeptides, e.g., polyarginine or tripeptides; proteins, such as serum albumin, gelatin, or immunoglobulins; hydrophilic polymers such as polyvinylpyrrolidone; amino acids, such as glycine, glutamic acid, aspartic acid, or arginine; monosaccharides, disaccharides, and other carbohydrates including cellulose or its derivatives, glucose, manose, or dextrins; chelating agents such as EDTA; sugar alcohols such as mannitol or sorbitol; counterions such as sodium; and/or nonionic surfactants such as polysorbates, poloxamers, or PEG. [0457]
  • The albumin fusion protein is typically formulated in such vehicles at a concentration of about 0.1 mg/ml to 100 mg/ml, preferably 1-10 mg/ml, at a pH of about 3 to 8. It will be understood that the use of certain of the foregoing excipients, carriers, or stabilizers will result in the formation of polypeptide salts. [0458]
  • Any pharmaceutical used for therapeutic administration can be sterile. Sterility is readily accomplished by filtration through sterile filtration membranes (e.g., 0.2 micron membranes). Albumin fusion proteins and/or polynucleotides generally are placed into a container having a sterile access port, for example, an intravenous solution bag or vial having a stopper pierceable by a hypodermic injection needle. [0459]
  • Albumin fusion proteins and/or polynucleotides ordinarily will be stored in unit or multi-dose containers, for example, sealed ampoules or vials, as an aqueous solution or as a lyophilized formulation for reconstitution. As an example of a lyophilized formulation, 10-ml vials are filled with 5 ml of sterile-filtered 1% (w/v) aqueous albumin fusion protein and/or polynucleotide solution, and the resulting mixture is lyophilized. The infusion solution is prepared by reconstituting the lyophilized albumin fusion protein and/or polynucleotide using bacteriostatic Water-for-Injection. [0460]
  • In a specific and preferred embodiment, the Albumin fusion protein formulations comprises 0.01 M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/[0461] milliliter polysorbate 80, pH 7.2. In another specific and preferred embodiment, the Albumin fusion protein formulations consists 0.01 M sodium phosphate, 0.15 mM sodium chloride, 0.16 micromole sodium octanoate/milligram of fusion protein, 15 micrograms/milliliter polysorbate 80, pH 7.2. The pH and buffer are chosen to match physiological conditions and the salt is added as a tonicifier. Sodium octanoate has been chosen due to its reported ability to increase the thermal stability of the protein in solution. Finally, polysorbate has been added as a generic surfactant, which lowers the surface tension of the solution and lowers non-specific adsorption of the albumin fusion protein to the container closure system.
  • The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the albumin fusion proteins and/or polynucleotides of the invention. Associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In addition, the albumin fusion proteins and/or polynucleotides may be employed in conjunction with other therapeutic compounds. [0462]
  • The albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with adjuvants. Adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, alum, alum plus deoxycholate (ImmunoAg), MTP-PE (Biocine Corp.), QS21 (Genentech, Inc.), BCG (e.g., THERACYS®), MPL and nonviable preparations of [0463] Corynebacterium parvum. In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with alum. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with QS-21. Further adjuvants that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Monophosphoryl lipid immunomodulator, AdjuVax 100a, QS-21, QS-18, CRL1005, Aluminum salts, MF-59, and Virosomal adjuvant technology. Vaccines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, vaccines directed toward protection against MMR (measles, mumps, rubella), polio, varicella, tetanus/diptheria, hepatitis A, hepatitis B, Haemophilus influenzae B, whooping cough, pneumonia, influenza, Lyme's Disease, rotavirus, cholera, yellow fever, Japanese encephalitis, poliomyelitis, rabies, typhoid fever, and pertussis. Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially. This includes presentations in which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g., as through separate intravenous lines into the same individual. Administration “in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second.
  • The albumin fusion proteins and/or polynucleotides of the invention may be administered alone or in combination with other therapeutic agents. Albumin fusion protein and/or polynucleotide agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include but not limited to, chemotherapeutic agents, antibiotics, steroidal and non-steroidal anti-inflammatories, conventional immunotherapeutic agents, and/or therapeutic treatments described below. Combinations may be administered either concomitantly, e.g., as an admixture, separately but simultaneously or concurrently; or sequentially. This includes presentations in which the combined agents are administered together as a therapeutic mixture, and also procedures in which the combined agents are administered separately but simultaneously, e.g., as through separate intravenous lines into the same individual. Administration “in combination” further includes the separate administration of one of the compounds or agents given first, followed by the second. [0464]
  • In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an anticoagulant. Anticoagulants that may be administered with the compositions of the invention include, but are not limited to, heparin, low molecular weight heparin, warfarin sodium (e.g., COUMADIN®), dicumarol, 4-hydroxycoumarin, anisindione (e.g., MIRADON™), acenocoumarol (e.g., nicoumalone, SINTHROME™), indan-1,3-dione, phenprocoumon (e.g., MARCUMAR™), ethyl biscoumacetate (e.g., TROMEXAN™), and aspirin. In a specific embodiment, compositions of the invention are administered in combination with heparin and/or warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin. In another specific embodiment, compositions of the invention are administered in combination with warfarin and aspirin. In another specific embodiment, compositions of the invention are administered in combination with heparin. In another specific embodiment, compositions of the invention are administered in combination with heparin and aspirin. [0465]
  • In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with thrombolytic drugs. Thrombolytic drugs that may be administered with the compositions of the invention include, but are not limited to, plasminogen, lys-plasminogen, alpha2-antiplasmin, streptokinae (e.g., KABIKINASE™), antiresplace (e.g., EMINASE™), tissue plasminogen activator (t-PA, altevase, ACTIVASE™), urokinase (e.g., ABBOKINASE™), sauruplase, (Prourokinase, single chain urokinase), and aminocaproic acid (e.g., AMICAR™). In a specific embodiment, compositions of the invention are administered in combination with tissue plasminogen activator and aspirin. [0466]
  • In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiplatelet drugs. Antiplatelet drugs that may be administered with the compositions of the invention include, but are not limited to, aspirin, dipyridamole (e.g., PERSANTINE™), and ticlopidine (e.g., TICLID™). [0467]
  • In specific embodiments, the use of anti-coagulants, thrombolytic and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention, diagnosis, and/or treatment of thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina. In specific embodiments, the use of anticoagulants, thrombolytic drugs and/or antiplatelet drugs in combination with albumin fusion proteins and/or polynucleotides of the invention is contemplated for the prevention of occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease. Other uses for the therapeutics of the invention, alone or in combination with antiplatelet, anticoagulant, and/or thrombolytic drugs, include, but are not limited to, the prevention of occlusions in extracorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines). [0468]
  • In certain embodiments, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with antiretroviral agents, nucleoside/nucleotide reverse transcriptase inhibitors (NRTIs), non-nucleoside reverse transcriptase inhibitors (NNRTIs), and/or protease inhibitors (PIs). NRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, RETROVIR™ (zidovudine/AZT), VIDEX™ (didanosine/ddI), HVID™ (zalcitabine/ddC), ZERIT™ (stavudine/d4T), EPIVIR™ (lamivudine/3TC), and COMBIVIR™ (zidovudine/lamivudine). NNRTIs that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, VIRAMUNE™ (nevirapine), RESCRIPTOR™ (delavirdine), and SUSTIVA™ (efavirenz). Protease inhibitors that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, CRIXIVAN™ (indinavir), NORVIR™ (ritonavir), INVRASE™ (saquinavir), and VIRACEPT™ (nelfinavir). In a specific embodiment, antiretroviral agents, nucleoside reverse transcriptase inhibitors, non-nucleoside reverse transcriptase inhibitors, and/or protease inhibitors may be used in any combination with albumin fusion proteins and/or polynucleotides of the invention to treat AIDS and/or to prevent or treat HIV infection. [0469]
  • Additional NRTIs include LODENOSINE™ (F-ddA; an acid-stable adenosine NRTI; Triangle/Abbott; COVIRACIL™ (emtricitabine/FTC; structurally related to lamivudine (3TC) but with 3- to 10-fold greater activity in vitro; Triangle/Abbott); dOTC (BCH-10652, also structurally related to lamivudine but retains activity against a substantial proportion of lamivudine-resistant isolates; Biochem Pharma); Adefovir (refused approval for anti-HIV therapy by FDA; Gilead Sciences); PREVEON® (Adefovir Dipivoxil, the active prodrug of adefovir; its active form is PMEA-pp); TENOFOVIR™ (bis-POC PMPA, a PMPA prodrug; Gilead); DAPD/DXG (active metabolite of DAPD; Triangle/Abbott); D-D4FC (related to 3TC, with activity against AZT/3TC-resistant virus); GW420867X (Glaxo Wellcome); ZIAGEN™ (abacavir/159U89; Glaxo Wellcome Inc.); CS-87 (3′azido-2′,3′-dideoxyuridine; WO 99/66936); and S-acyl-2-thioethyl (SATE)-bearing prodrug forms of β-L-FD4C and β-L-FddC (WO 98/17281). [0470]
  • Additional NNRTIs include COACTINON™ (Emivirine/MKC-442, potent NNRTI of the HEPT class; Triangle/Abbott); CAPRAVIRINE™ (AG-1549/S-1153, a next generation NNRTI with activity against viruses containing the K103N mutation; Agouron); PNU-142721 (has 20- to 50-fold greater activity than its predecessor delavirdine and is active against K103N mutants; Pharmacia & Upjohn); DPC-961 and DPC-963 (second-generation derivatives of efavirenz, designed to be active against viruses with the K103N mutation; DuPont); GW-420867×(has 25-fold greater activity than HBY097 and is active against K103N mutants; Glaxo Wellcome); CALANOLIDE A (naturally occurring agent from the latex tree; active against viruses containing either or both the Y181C and K103N mutations); and Propolis (WO 99/49830). [0471]
  • Additional protease inhibitors include LOPINAVIR™ (ABT378/r; Abbott Laboratories); BMS-232632 (an azapeptide; Bristol-Myres Squibb); TIPRANAVIR™ (PNU-140690, a non-peptic dihydropyrone; Pharmacia & Upjohn); PD-178390 (a nonpeptidic dihydropyrone; Parke-Davis); BMS 232632 (an azapeptide; Bristol-Myers Squibb); L-756,423 (an indinavir analog; Merck); DMP-450 (a cyclic urea compound; Avid & DuPont); AG-1776 (a peptidomimetic with in vitro activity against protease inhibitor-resistant viruses; Agouron); VX-175/GW-433908 (phosphate prodrug of amprenavir; Vertex & Glaxo Welcome); CGP61755 (Ciba); and AGENERASE™ (amprenavir; Glaxo Wellcome Inc.). [0472]
  • Additional antiretroviral agents include fusion inhibitors/gp41 binders. Fusion inhibitors/gp41 binders include T-20 (a peptide from residues 643-678 of the HIV gp41 transmembrane protein ectodomain which binds to gp41 in its resting state and prevents transformation to the fusogenic state; Trimeris) and T-1249 (a second-generation fusion inhibitor; Trimeris). [0473]
  • Additional antiretroviral agents include fusion inhibitors/chemokine receptor antagonists. Fusion inhibitors/chemokine receptor antagonists include CXCR4 antagonists such as AMD 3100 (a bicyclam), SDF-1 and its analogs, and ALX40-4C (a cationic peptide), T22 (an 18 amino acid peptide; Trimeris) and the T22 analogs T134 and T140; CCR5 antagonists such as RANTES (9-68), AOP-RANTES, NNY-RANTES, and TAK-779; and CCR5/CXCR4 antagonists such as NSC 651016 (a distamycin analog). Also included are CCR2B, CCR3, and CCR6 antagonists. Chemokine recpetor agonists such as RANTES, SDF-1, MIP-1α, MIP-1β, etc., may also inhibit fusion. [0474]
  • Additional antiretroviral agents include integrase inhibitors. Integrase inhibitors include dicaffeoylquinic (DFQA) acids; L-chicoric acid (a dicaffeoyltartaric (DCTA) acid); quinalizarin (QLC) and related anthraquinones; ZINTEVIR™ ([0475] AR 177, an oligonucleotide that probably acts at cell surface rather than being a true integrase inhibitor; Arondex); and naphthols such as those disclosed in WO 98/50347.
  • Additional antiretroviral agents include hydroxyurea-like compunds such as BCX-34 (a purine nucleoside phosphorylase inhibitor; Biocryst); ribonucleotide reductase inhibitors such as DIDOX™ (Molecules for Health); inosine monophosphate dehydrogenase (IMPDH) inhibitors sucha as VX-497 (Vertex); and mycopholic acids such as CellCept (mycophenolate mofetil; Roche). [0476]
  • Additional antiretroviral agents include inhibitors of viral integrase, inhibitors of viral genome nuclear translocation such as arylene bis(methylketone) compounds; inhibitors of HIV entry such as AOP-RANTES, NNY-RANTES, RANTES-IgG fusion protein, soluble complexes of RANTES and glycosaminoglycans (GAG), and AMD-3100; nucleocapsid zinc finger inhibitors such as dithiane compounds; targets of HIV Tat and Rev; and pharmacoenhancers such as ABT-378. [0477]
  • Other antiretroviral therapies and adjunct therapies include cytokines and lymphokines such as MIP-1α, MIP-1β, SDF-1α, IL-2, PROLEUKIN™ (aldesleukin/L2-7001; Chiron), 1L-4, IL-10, IL-12, and IL-13; interferons such as IFN-α2a; antagonists of TNFs, NFκB, GM-CSF, M-CSF, and IL-10; agents that modulate immune activation such as cyclosporin and prednisone; vaccines such as Remune™ (HIV Immunogen), APL 400-003 (Apollon), recombinant gp120 and fragments, bivalent (B/E) recombinant envelope glycoprotein, rgp120CM235, MN rgp120, SF-2 rgp120, gp120/soluble CD4 complex, Delta JR-FL protein, branched synthetic peptide derived from discontinuous gp120 C3/C4 domain, fusion-competent immunogens, and Gag, Pol, Nef, and Tat vaccines; gene-based therapies such as genetic suppressor elements (GSEs; WO 98/54366), and intrakines (genetically modified CC chemokines targetted to the ER to block surface expression of newly synthesized CCR5 (Yang et al., [0478] PNAS 94:11567-72 (1997); Chen et al., Nat. Med. 3:1110-16 (1997)); antibodies such as the anti-CXCR4 antibody 12G5, the anti-CCR5 antibodies 2D7, 5C7, PA8, PA9, PA10, PA11, PA12, and PA14, the anti-CD4 antibodies Q4120 and RPA-T4, the anti-CCR3 antibody 7B11, the anti-gp120 antibodies 17b, 48d, 447-52D, 257-D, 268-D and 50.1, anti-Tat antibodies, anti-TNF-α antibodies, and monoclonal antibody 33A; aryl hydrocarbon (AH) receptor agonists and antagonists such as TCDD, 3,3′,4,4′,5-pentachlorobiphenyl, 3,3′,4,4′-tetrachlorobiphenyl, and α-naphthoflavone (WO 98/30213); and antioxidants such as γ-L-glutamyl-L-cysteine ethyl ester (γ-GCE; WO 99/56764).
  • In a further embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiviral agent. Antiviral agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, acyclovir, ribavirin, amantadine, and remantidine. [0479]
  • In other embodiments, albumin fusion proteins and/or polynucleotides of the invention may be administered in combination with anti-opportunistic infection agents. Anti-opportunistic agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention, include, but are not limited to, TRIMETHOPRIM-SULFAMETHOXAZOLE™, DAPSONE™, PENTAMIDNE™, ATOVAQUONE™, ISONIAZID™, RIFAMPIN™, PYRAZINAMIDE™, ETHAMBUTOL™, RIFABUTIN™, CLARITHROMYCIN™, AZITHROMYCIN™, GANCICLOVIR™, FOSCARNET™, CIDOFOVIR™, FLUCONAZOLE™, ITRACONAZOLE™, KETOCONAZOLE™, ACYCLOVIR™, FAMCICOLVIR™, PYRIMETHAMINE™, LEUCOVORIN™, NEUPOGEN™ (filgrastim/G-CSF), and LEUKINE™ (sargramostim/GM-CSF). In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with TRIMETHOPRIM-SULFAMETHOXAZOLE™, DAPSONE™, PENTAMIDINE™, and/or ATOVAQUONE™ to prophylactically treat or prevent an opportunistic [0480] Pneumocystis carinii pneumonia infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ISONIAZID™, RIFAMPIN™, PYRAZINAMIDE™, and/or ETHAMBUTOL™ to prophylactically treat or prevent an opportunistic Mycobacterium avium complex infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with RIFABUTIN™, CLARITHROMYCIN™, and/or AZITHROMYCIN™ to prophylactically treat or prevent an opportunistic Mycobacterium tuberculosis infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with GANCICLOVIR™, FOSCARNET™, and/or CIDOFOVIR™ to prophylactically treat or prevent an opportunistic cytomegalovirus infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with FLUCONAZOLE™, ITRACONAZOLE™, and/or KETOCONAZOLE™ to prophylactically treat or prevent an opportunistic fungal infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with ACYCLOVIR™ and/or FAMCICOLVIR™ to prophylactically treat or prevent an opportunistic herpes simplex virus type I and/or type II infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with PYRIMETHAMINE™ and/or LEUCOVORIN™ to prophylactically treat or prevent an opportunistic Toxoplasma gondii infection. In another specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are used in any combination with LEUCOVORIN™ and/or NEUPOGEN™ to prophylactically treat or prevent an opportunistic bacterial infection.
  • In a further embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antibiotic agent. Antibiotic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, amoxicillin, beta-lactamases, aminoglycosides, beta-lactam (glycopeptide), beta-lactamases, Clindamycin, chloramphenicol, cephalosporins, ciprofloxacin, erythromycin, fluoroquinolones, macrolides, metronidazole, penicillins, quinolones, rapamycin, rifampin, streptomycin, sulfonamide, tetracyclines, trimethoprim, trimethoprim-sulfamethoxazole, and vancomycin. [0481]
  • In other embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunostimulants. Immunostimulants that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, levamisole (e.g., ERGAMISOL™), isoprinosine (e.g. INOSIPLEX™), interferons (e.g. interferon alpha), and interleukins (e.g., IL-2). [0482]
  • In other embodiments, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with immunosuppressive agents. Immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, steroids, cyclosporine, cyclosporine analogs, cyclophosphamide methylprednisone, prednisone, azathioprine, FK-506, 15-deoxyspergualin, and other immunosuppressive agents that act by suppressing the function of responding T cells. Other immunosuppressive agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, prednisolone, methotrexate, thalidomide, methoxsalen, rapamycin, leflunomide, mizoribine (BREDININ™), brequinar, deoxyspergualin, and azaspirane (SKF 105685), ORTHOCLONE OKT® 3 (muromonab-CD3), SANDIMMUNE™, NEORAL™, SANGDYA™ (cyclosporine), PROGRAF® (FK506, tacrolimus), CELLCEPT® (mycophenolate motefil, of which the active metabolite is mycophenolic acid), IMURAN™ (azathioprine), glucocorticosteroids, adrenocortical steroids such as DELTASONE™ (prednisone) and HYDELTRASOL™ (prednisolone), FOLEX™ and MEXATE™ (methotrxate), OXSORALEN-ULTRA™ (methoxsalen) and RAPAMUNE™ (sirolimus). In a specific embodiment, immunosuppressants may be used to prevent rejection of organ or bone marrow transplantation. [0483]
  • In an additional embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with one or more intravenous immune globulin preparations. Intravenous immune globulin preparations that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but not limited to, GAMMAR™, IVEEGAM™, SANDOGLOBULIN™, GAMMAGARD S/D™, ATGAM™ (antithymocyte glubulin), and GAMIMUNE™. In a specific embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with intravenous immune globulin preparations in transplantation therapy (e.g., bone marrow transplant). [0484]
  • In certain embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered alone or in combination with an anti-inflammatory agent. Anti-inflammatory agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, corticosteroids (e.g. betamethasone, budesonide, cortisone, dexamethasone, hydrocortisone, methylprednisolone, prednisolone, prednisone, and triamcinolone), nonsteroidal anti-inflammatory drugs (e.g., diclofenac, diflunisal, etodolac, fenoprofen, floctafenine, flurbiprofen, ibuprofen, indomethacin, ketoprofen, meclofenamate, mefenamic acid, meloxicam, nabumetone, naproxen, oxaprozin, phenylbutazone, piroxicam, sulindac, tenoxicam, tiaprofenic acid, and tolmetin.), as well as antihistamines, aminoarylcarboxylic acid derivatives, arylacetic acid derivatives, arylbutyric acid derivatives, arylcarboxylic acids, arylpropionic acid derivatives, pyrazoles, pyrazolones, salicylic acid derivatives, thiazinecarboxamides, e-acetamidocaproic acid, S-adenosylmethionine, 3-amino-4-hydroxybutyric acid, amixetrine, bendazac, benzydamine, bucolome, difenpiramide, ditazol, emorfazone, guaiazulene, nabumetone, nimesulide, orgotein, oxaceprol, paranyline, perisoxal, pifoxime, proquazone, proxazole, and tenidap. [0485]
  • In an additional embodiment, the compositions of the invention are administered alone or in combination with an anti-angiogenic agent. Anti-angiogenic agents that may be administered with the compositions of the invention include, but are not limited to, Angiostatin (Entremed, Rockville, Md.), Troponin-1 (Boston Life Sciences, Boston, Mass.), anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel (Taxol), Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, VEGI, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter “d group” transition metals. [0486]
  • Lighter “d group” transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition metal complexes. [0487]
  • Representative examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanadate complexes include metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodium metavanadate, and sodium orthovanadate. Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates. [0488]
  • Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid. Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide. Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars. [0489]
  • A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include, but are not limited to, [0490] platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, (1991)); Sulphated Polysaccharide Peptidoglycan Complex (SP-PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine; modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloffet al., J. Bio. Chem. 267:17321-17326, (1992)); Chymostatin (Tomkinson et al., Biochem J. 286:475-480, (1992)); Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, (1990)); Gold Sodium Thiomalate (“GST”; Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, (1987)); anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J. Biol. Chem. 262(4):1659-1664, (1987)); Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-carboxyphenyl-4-chloroanthronilic acid disodium or “CCA”; (Takeuchi et al., Agents Actions 36:312-316, (1992)); and metalloproteinase inhibitors such as BB94.
  • Additional anti-angiogenic factors that may also be utilized within the context of the present invention include Thalidomide, (Celgene, Warren, N.J.); Angiostatic steroid; AGM-1470 (H. Brem and J. Folkman [0491] J Pediatr. Surg. 28:445-51 (1993)); an integrin alpha v beta 3 antagonist (C. Storgard et al., J Clin. Invest. 103:47-54 (1999)); carboxynaminolmidazole; Carboxyamidotriazole (CAI) (National Cancer Institute, Bethesda, Md.); Conbretastatin A-4 (CA4P) (OXiGENE, Boston, Mass.); Squalamine (Magainin Pharmaceuticals, Plymouth Meeting, Pa.); TNP-470, (Tap Pharmaceuticals, Deerfield, Ill.); ZD-0101 AstraZeneca (London, UK); APRA (CT2584); Benefin, Byrostatin-1 (SC339555); CGP-41251 (PKC 412); CM101; Dexrazoxane (ICRF187); DMXAA; Endostatin; Flavopridiol; Genestein; GTE; ImmTher; Iressa (ZD1839); Octreotide (Somatostatin); Panretin; Penacillamine; Photopoint; PI-88; Prinomastat (AG-3340) Purlytin; Suradista (FCE26644); Tamoxifen (Nolvadex); Tazarotene; Tetrathiomolybdate; Xeloda (Capecitabine); and 5-Fluorouracil.
  • Anti-angiogenic agents that may be administed in combination with the compounds of the invention may work through a variety of mechanisms including, but not limited to, inhibiting proteolysis of the extracellular matrix, blocking the function of endothelial cell-extracellular matrix adhesion molecules, by antagonizing the function of angiogenesis inducers such as growth factors, and inhibiting integrin receptors expressed on proliferating endothelial cells. Examples of anti-angiogenic inhibitors that interfere with extracellular matrix proteolysis and which may be administered in combination with the compositons of the invention include, but are not lmited to, AG-3340 (Agouron, La Jolla, Calif.), BAY-12-9566 (Bayer, West Haven, Conn.), BMS-275291 (Bristol Myers Squibb, Princeton, N.J.), CGS-27032A (Novartis, East Hanover, N.J.), Marimastat (British Biotech, Oxford, UK), and Metastat (Aeterna, St-Foy, Quebec). Examples of anti-angiogenic inhibitors that act by blocking the function of endothelial cell-extracellular matrix adhesion molecules and which may be administered in combination with the compositons of the invention include, but are not lmited to, EMD-121974 (Merck KcgaA Darmstadt, Germany) and Vitaxin (Ixsys, La Jolla, Calif./Medimmune, Gaithersburg, Md.). Examples of anti-angiogenic agents that act by directly antagonizing or inhibiting angiogenesis inducers and which may be administered in combination with the compositons of the invention include, but are not lmited to, Angiozyme (Ribozyme, Boulder, Colo.), Anti-VEGF antibody (Genentech, S. San Francisco, Calif.), PTK-787/ZK-225846 (Novartis, Basel, Switzerland), SU-101 (Sugen, S. San Francisco, Calif.), SU-5416 (Sugen/Pharmacia Upjohn, Bridgewater, N.J.), and SU-6668 (Sugen). Other anti-angiogenic agents act to indirectly inhibit angiogenesis. Examples of indirect inhibitors of angiogenesis which may be administered in combination with the compositons of the invention include, but are not limited to, IM-862 (Cytran, Kirkland, Wash.), Interferon-alpha, IL-12 (Roche, Nutley, N.J.), and Pentosan polysulfate (Georgetown University, Washington, D.C.). [0492]
  • In particular embodiments, the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of an autoimmune disease, such as for example, an autoimmune disease described herein. [0493]
  • In a particular embodiment, the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of arthritis. In a more particular embodiment, the use of compositions of the invention in combination with anti-angiogenic agents is contemplated for the treatment, prevention, and/or amelioration of rheumatoid arthritis. [0494]
  • In another embodiment, the polynucleotides encoding a polypeptide of the present invention are administered in combination with an angiogenic protein, or polynucleotides encoding an angiogenic protein. Examples of angiogenic proteins that may be administered with the compositions of the invention include, but are not limited to, acidic and basic fibroblast growth factors, VEGF-1, VEGF-2, VEGF-3, epidermal growth factor alpha and beta, platelet-derived endothelial cell growth factor, platelet-derived growth factor, tumor necrosis factor alpha, hepatocyte growth factor, insulin-like growth factor, colony stimulating factor, macrophage colony stimulating factor, granulocyte/macrophage colony stimulating factor, and nitric oxide synthase. [0495]
  • In additional embodiments, compositions of the invention are administered in combination with a chemotherapeutic agent. Chemotherapeutic agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to alkylating agents such as nitrogen mustards (for example, Mechlorethamine, cyclophosphamide, Cyclophosphamide Ifosfamide, Melphalan (L-sarcolysin), and Chlorambucil), ethylenimines and methylmelamines (for example, Hexamethylmelamine and Thiotepa), alkyl sulfonates (for example, Busulfan), nitrosoureas (for example, Carmustine (BCNU), Lomustine (CCNU), Semustine (methyl-CCNU), and Streptozocin (streptozotocin)), triazenes (for example, Dacarbazine (DTIC; dimethyltriazenoimidazolecarboxamide)), folic acid analogs (for example, Methotrexate (amethopterin)), pyrimidine analogs (for example, Fluorouacil (5-fluorouracil; 5-FU), Floxuridine (fluorodeoxyuridine; FudR), and Cytarabine (cytosine arabinoside)), purine analogs and related inhibitors (for example, Mercaptopurine (6-mercaptopurine; 6-MP), Thioguanine (6-thioguanine; TG), and Pentostatin (2′-deoxycoformycin)), vinca alkaloids (for example, Vinblastine (VLB, vinblastine sulfate)) and Vincristine (vincristine sulfate)), epipodophyllotoxins (for example, Etoposide and Teniposide), antibiotics (for example, Dactinomycin (actinomycin D), Daunorubicin (daunomycin; rubidomycin), Doxorubicin, Bleomycin, Plicamycin (mithramycin), and Mitomycin (mitomycin C), enzymes (for example, L-Asparaginase), biological response modifiers (for example, Interferon-alpha and interferon-alpha-2b), platinum coordination compounds (for example, Cisplatin (cis-DDP) and Carboplatin), anthracenedione (Mitoxantrone), substituted ureas (for example, Hydroxyurea), methylhydrazine derivatives (for example, Procarbazine (N-methylhydrazine; MIH), adrenocorticosteroids (for example, Prednisone), progestins (for example, Hydroxyprogesterone caproate, Medroxyprogesterone, Medroxyprogesterone acetate, and Megestrol acetate), estrogens (for example, Diethylstilbestrol (DES), Diethylstilbestrol diphosphate, Estradiol, and Ethinyl estradiol), antiestrogens (for example, Tamoxifen), androgens (Testosterone proprionate, and Fluoxymesterone), antiandrogens (for example, Flutamide), gonadotropin-releasing horomone analogs (for example, Leuprolide), other hormones and hormone analogs (for example, methyltestosterone, estramustine, estramustine phosphate sodium, chlorotrianisene, and testolactone), and others (for example, dicarbazine, glutamic acid, and mitotane). [0496]
  • In one embodiment, the compositions of the invention are administered in combination with one or more of the following drugs: infliximab (also known as Remicade™ Centocor, Inc.), Trocade (Roche, RO-32-3555), Leflunomide (also known as Arava™ from Hoechst Marion Roussel), Kineret™ (an IL-1 Receptor antagonist also known as Anakinra from Amgen, Inc.) [0497]
  • In a specific embodiment, compositions of the invention are administered in combination with CHOP (cyclophosphamide, doxorubicin, vincristine, and prednisone) or combination of one or more of the components of CHOP. In one embodiment, the compositions of the invention are administered in combination with anti-CD20 antibodies, human monoclonal anti-CD20 antibodies. In another embodiment, the compositions of the invention are administered in combination with anti-CD20 antibodies and CHOP, or anti-CD20 antibodies and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. In a specific embodiment, compositions of the invention are administered in combination with Rituximab. In a further embodiment, compositions of the invention are administered with Rituximab and CHOP, or Rituximab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. In a specific embodiment, compositions of the invention are administered in combination with tositumomab. In a further embodiment, compositions of the invention are administered with tositumomab and CHOP, or tositumomab and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. The anti-CD20 antibodies may optionally be associated with radioisotopes, toxins or cytotoxic prodrugs. [0498]
  • In another specific embodiment, the compositions of the invention are administered in combination Zevalin™. In a further embodiment, compositions of the invention are administered with Zevalin™ and CHOP, or Zevalin™ and any combination of one or more of the components of CHOP, particularly cyclophosphamide and/or prednisone. Zevalin™ may be associated with one or more radisotopes. Particularly preferred isotopes are [0499] 90Y and 111In.
  • In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with cytokines. Cytokines that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, IL2, IL3, IL4, IL5, IL6, IL7, IL10, IL12, IL13, IL15, anti-CD40, CD40L, IFN-gamma and TNF-alpha. In another embodiment, albumin fusion proteins and/or polynucleotides of the invention may be administered with any interleukin, including, but not limited to, IL-1alpha, IL-1beta, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-16, IL-17, IL-18, IL-19, IL-20, and IL-21. [0500]
  • In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with members of the TNF family. TNF, TNF-related or TNF-like molecules that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, soluble forms of TNF-alpha, lymphotoxin-alpha (LT-alpha, also known as TNF-beta), LT-beta (found in complex heterotrimer LT-alpha2-beta), OPGL, FasL, CD27L, CD30L, CD40L, 4-1BBL, DcR3, OX40L, TNF-gamma (International Publication No. WO 96/14328), AIM-I (International Publication No. WO 97/33899), endokine-alpha (International Publication No. WO 98/07880), OPG, and netitrokine-alpha (International Publication No. WO 98/18921, OX40, and nerve growth factor (NGF), and soluble forms of Fas, CD30, CD27, CD40 and 4-IBB, TR2 (International Publication No. WO 96/34095), DR3 (International Publication No. WO 97/33904), DR4 (International Publication No. WO 98/32856), TR5 (International Publication No. WO 98/30693), TRANK, TR9 (International Publication No. WO 98/56892),TR10 (International Publication No. WO 98/54202), 312C2 (International Publication No. WO 98/06842), and TR12, and soluble forms CD154, CD70, and CD153. [0501]
  • In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with angiogenic proteins. Angiogenic proteins that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Glioma Derived Growth Factor (GDGF), as disclosed in European Patent Number EP-399816; Platelet Derived Growth Factor-A (PDGF-A), as disclosed in European Patent Number EP-682110; Platelet Derived Growth Factor-B (PDGF-B), as disclosed in European Patent Number EP-282317; Placental Growth Factor (PIGF), as disclosed in International Publication Number WO 92/06194; Placental Growth Factor-2 (PIGF-2), as disclosed in Hauser et al., Growth Factors, 4:259-268 (1993); Vascular Endothelial Growth Factor (VEGF), as disclosed in International Publication Number WO 90/13649; Vascular Endothelial Growth Factor-A (VEGF-A), as disclosed in European Patent Number EP-506477; Vascular Endothelial Growth Factor-2 (VEGF-2), as disclosed in International Publication Number WO 96/39515; Vascular Endothelial Growth Factor B (VEGF-3); Vascular Endothelial Growth Factor B-186 (VEGF-B186), as disclosed in International Publication Number WO 96/26736; Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO 98/02543; Vascular Endothelial Growth Factor-D (VEGF-D), as disclosed in International Publication Number WO 98/07832; and Vascular Endothelial Growth Factor-E (VEGF-E), as disclosed in German Patent Number DE19639601. The above mentioned references are herein incorporated by reference in their entireties. [0502]
  • In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with Fibroblast Growth Factors. Fibroblast Growth Factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, FGF-1, FGF-2, FGF-3, FGF-4, FGF-5, FGF-6, FGF-7, FGF-8, FGF-9, FGF-10, FGF-11, FGF-12, FGF-13, FGF-14, and FGF-15. [0503]
  • In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with hematopoietic growth factors. Hematopoietic growth factors that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, granulocyte macrophage colony stimulating factor (GM-CSF) (sargramostim, LEUKINE™, PROKINE™), granulocyte colony stimulating factor (G-CSF) (filgrastim, NEUPOGEN™), macrophage colony stimulating factor (M-CSF, CSF-1) erythropoietin (epoetin alfa, EPOGEN™, PROCRIT™), stem cell factor (SCF, c-kit ligand, steel factor), megakaryocyte colony stimulating factor, PIXY321 (a GMCSF/IL-3 fusion protein), interleukins, especially any one or more of IL-1 through IL-12, interferon-gamma, or thrombopoietin. [0504]
  • In certain embodiments, albumin fusion proteins and/or polynucleotides of the present invention are administered in combination with adrenergic blockers, such as, for example, acebutolol, atenolol, betaxolol, bisoprolol, carteolol, labetalol, metoprolol, nadolol, oxprenolol, penbutolol, pindolol, propranolol, sotalol, and timolol. [0505]
  • In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with an antiarrhythmic drug (e.g., adenosine, amidoarone, bretylium, digitalis, digoxin, digitoxin, diliazem, disopyramide, esmolol, flecainide, lidocaine, mexiletine, moricizine, phenytoin, procainamide, N-acetyl procainamide, propafenone, propranolol, quinidine, sotalol, tocainide, and verapamil). [0506]
  • In another embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with diuretic agents, such as carbonic anhydrase-inhibiting agents (e.g., acetazolamide, dichlorphenamide, and methazolamide), osmotic diuretics (e.g., glycerin, isosorbide, mannitol, and urea), diuretics that inhibit Na[0507] +-K+-2Cl symport (e.g., furosemide, bumetamide, azosemide, piretamide, tripamide, ethacrynic acid, muzolimine, and torsemide), thiazide and thiazide-like diuretics (e.g., bendroflumethiazide, benzthiazide, chlorothiazide, hydrochlorothiazide, hydroflumethiazide, methyclothiazide, polythiazide, trichormethiazide, chlorthalidone, indapamide, metolazone, and quinethazone), potassium sparing diuretics (e.g., amiloride and triamterene), and mineralcorticoid receptor antagonists (e.g., spironolactone, canrenone, and potassium canrenoate).
  • In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for endocrine and/or hormone imbalance disorders. Treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, [0508] 127I, radioactive isotopes of iodine such as 131I and 123I; recombinant growth hormone, such as HUMATROPE™ (recombinant somatropin); growth hormone analogs such as PROTROPIN™ (somatrem); dopamine agonists such as PARLODEL™ (bromocriptine); somatostatin analogs such as SANDOSTATIN™ (octreotide); gonadotropin preparations such as PREGNYL™, A.P.L.™ and PROFASI™ (chorionic gonadotropin (CG)), PERGONAL™ (menotropins), and METRODIN™ (urofollitropin (uFSH)); synthetic human gonadotropin releasing hormone preparations such as FACTREL™ and LUTREPULSE™ (gonadorelin hydrochloride); synthetic gonadotropin agonists such as LUPRON™ (leuprolide acetate), SUPPRELIN™ (histrelin acetate), SYNAREL™ (nafarelin acetate), and ZOLADEX™ (goserelin acetate); synthetic preparations of thyrotropin-releasing hormone such as RELEFACT TRH™ and THYPINONE™ (protirelin); recombinant human TSH such as THYROGEN™; synthetic preparations of the sodium salts of the natural isomers of thyroid hormones such as L-T4™, SYNTHROID™ and LEVOTHROID™ (levothyroxine sodium), L-T3™, CYTOMEL™ and TRIOSTAT™ (liothyroine sodium), and THYROLAR™ (liotrix); antithyroid compounds such as 6-n-propylthiouracil (propylthiouracil), 1-methyl-2-mercaptoimidazole and TAPAZOLE™ (methimazole), NEO-MERCAZOLE™ (carbimazole); beta-adrenergic receptor antagonists such as propranolol and esmolol; Ca2+ channel blockers; dexamethasone and iodinated radiological contrast agents such as TELEPAQUE™ (iopanoic acid) and ORAGRAFIN™ (sodium ipodate).
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, estrogens or congugated estrogens such as ESTRACE™ (estradiol), ESTINYL™ (ethinyl estradiol), PREMARIN™, ESTRATAB™, ORTHO-EST™, OGEN™ and estropipate (estrone), ESTROVIS™ (quinestrol), ESTRADERM™ (estradiol), DELESTROGEN™ and VALERGEN™ (estradiol valerate), DEPO-ESTRADIOL CYPIONATE™ and ESTROJECT LA™ (estradiol cypionate); antiestrogens such as NOLVADEX™ (tamoxifen), SEROPHENE™ and CLOMID™ (clomiphene); progestins such as DURALUTIN™ (hydroxyprogesterone caproate), MPA™ and DEPO-PROVERA™ (medroxyprogesterone acetate), PROVERA™ and CYCRIN™ (MPA), MEGACE™ (megestrol acetate), NORLUTIN™ (norethindrone), and NORLUTATE™ and AYGESTIN™ (norethindrone acetate); progesterone implants such as NORPLANT SYSTEM™ (subdermal implants of norgestrel); antiprogestins such as RU 486™ (mifepristone); hormonal contraceptives such as ENOVID™ (norethynodrel plus mestranol), PROGESTASERT™ (intrauterine device that releases progesterone), LOESTRIN™, BREVICON™, MODICON™, GENORA™, NELONA™, NORINYL™, OVACON-35™ and OVACON-50™ (ethinyl estradiol/norethindrone), LEVLEN™, NORDETTE™, TRI-LEVLEN™ and TRIPHASIL-21™ (ethinyl estradiol/levonorgestrel) LO/OVRAL™ and OVRAL™ (ethinyl estradiol/norgestrel), DEMULEN™ (ethinyl estradiol/ethynodiol diacetate), NORINYL™, ORTHO-NOVUM™, NORETHIN™, GENORA™, and NELOVA™ (norethindrone/mestranol), DESOGEN™ and ORTHO-CEPT™ (ethinyl estradiol/desogestrel), ORTHO-CYCLEN™ and ORTHO-TRICYCLEN™ (ethinyl estradiol/norgestimate), MICRONOR™ and NOR-QD™ (norethindrone), and OVRETTE™ (norgestrel). [0509]
  • Additional treatments for endocrine and/or hormone imbalance disorders include, but are not limited to, testosterone esters such as methenolone acetate and testosterone undecanoate; parenteral and oral androgens such as TESTOJECT-50™ (testosterone), TESTEX™ (testosterone propionate), DELATESTRYL™ (testosterone enanthate), DEPO-TESTOSTERONE™ (testosterone cypionate), DANOCRINE™ (danazol), HALOTESTIN™ (fluoxymesterone), ORETON METHYL™, TESTRED™ and VIRILON™ (methyltestosterone), and OXANDRIN™ (oxandrolone); testosterone transdermal systems such as TESTODERM™; androgen receptor antagonist and 5-alpha-reductase inhibitors such as ANDROCUR™ (cyproterone acetate), EULEXN™ (flutamide), and PROSCAR™ (finasteride); adrenocorticotropic hormone preparations such as CORTROSYN™ (cosyntropin); adrenocortical steroids and their synthetic analogs such as ACLOVATE™ (alclometasone dipropionate), CYCLOCORT™ (amcinonide), BECLOVENT™ and VANCERIL™ (beclomethasone dipropionate), CELESTONE™ (betamethasone), BENISONE™ and UTICORT™ (betamethasone benzoate), DIPROSONE™ (betamethasone dipropionate), CELESTONE PHOSPHATE™ (betamethasone sodium phosphate), CELESTONE SOLUSPAN™ (betamethasone sodium phosphate and acetate), BETA-VAL™ and VALISONE™ (betamethasone valerate), TEMOVATE™ (clobetasol propionate), CLODERM™ (clocortolone pivalate), CORTEF™ and HYDROCORTONE™ (cortisol (hydrocortisone)), HYDROCORTONE ACETATE™ (cortisol (hydrocortisone) acetate), LOCOID™ (cortisol (hydrocortisone) butyrate), HYDROCORTONE PHOSPHATE™ (cortisol (hydrocortisone) sodium phosphate), A-HYDROCORT™ and SOLU CORTEF™ (cortisol (hydrocortisone) sodium succinate), WESTCORT™ (cortisol (hydrocortisone) valerate), CORTISONE ACETATE™ (cortisone acetate), DESOWEN™ and TRIDESILON™ (desonide), TOPICORT™ (desoximetasone), DECADRON™ (dexamethasone), DECADRON LA™ (dexamethasone acetate), DECADRON PHOSPHATE™ and HEXADROL PHOSPHATE™ (dexamethasone sodium phosphate), FLORONE™ and MAXIFLOR™ (diflorasone diacetate), FLORINEF ACETATE™ (fludrocortisone acetate), AEROBID™ and NASALIDE™ (flunisolide), FLUONID™ and SYNALAR™ (fluocinolone acetonide), LIDEX™ (fluocinonide), FLUOR-OP™ and FML™ (fluorometholone), CORDRAN™ (flurandrenolide), HALOG™ (halcinonide), HMS LIZUIFILM™ (medrysone), MEDROL™ (methylprednisolone), DEPO-MEDROL™ and MEDROL ACETATE™ (methylprednisone acetate), A-METHAPRED™ and SOLUMEDROL™ (methylprednisolone sodium succinate), ELOCON™ (mometasone furoate), HALDRONE™ (paramethasone acetate), DELTA-CORTEF™ (prednisolone), ECONOPRED™ (prednisolone acetate), HYDELTRASOL™ (prednisolone sodium phosphate), HYDELTRA-T.B.A™ (prednisolone tebutate), DELTASONE™ (prednisone), ARISTOCORT™ and KENACORT™ (triamcinolone), KENALOG™ (triamcinolone acetonide), ARISTOCORT™ and KENACORT DIACETATE™ (triamcinolone diacetate), and ARISTOSPAN™ (triamcinolone hexacetonide); inhibitors of biosynthesis and action of adrenocortical steroids such as CYTADREN™ (aminoglutethimide), NIZORAL™ (ketoconazole), MODRASTANE™ (trilostane), and METOPIRONE™ (metyrapone); bovine, porcine or human insulin or mixtures thereof; insulin analogs; recombinant human insulin such as HUMULIN™ and NOVOLIN™; oral hypoglycemic agents such as ORAIMIDE™ and ORINASE™ (tolbutamide), DIABINESE™ (chlorpropamide), TOLAMIDE™ and TOLINASE™ (tolazamide), DYMELOR™ (acetohexamide), glibenclamide, MICRONASE™, DIBETA™ and GLYNASE™ (glyburide), GLUCOTROL™ (glipizide), and DIAMICRON™ (gliclazide), GLUCOPHAGE™ (metformin), ciglitazone, pioglitazone, and alpha-glucosidase inhibitors; bovine or porcine glucagon; somatostatins such as SANDOSTATIN™ (octreotide); and diazoxides such as PROGLYCEM™ (diazoxide). [0510]
  • In one embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for uterine motility disorders. Treatments for uterine motility disorders include, but are not limited to, estrogen drugs such as conjugated estrogens (e.g., PREMARIN® and ESTRATAB®), estradiols (e.g., CLIMARA® and ALORA®), estropipate, and chlorotrianisene; progestin drugs (e.g., AMEN® (medroxyprogesterone), MICRONOR® (norethidrone acetate), PROMETRIUM® progesterone, and megestrol acetate); and estrogen/progesterone combination therapies such as, for example, conjugated estrogens/medroxyprogesterone (e.g., PREMPRO™ and PREMPHASE®) and norethindrone acetate/ethinyl estsradiol (e.g., FEMHRT™). [0511]
  • In an additional embodiment, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with drugs effective in treating iron deficiency and hypochromic anemias, including but not limited to, ferrous sulfate (iron sulfate, FEOSOL™), ferrous fumarate (e.g., FEOSTAT™), ferrous gluconate (e.g., FERGON™), polysaccharide-iron complex (e.g., NIFEREX™), iron dextran injection (e.g., INFED™), cupric sulfate, pyroxidine, riboflavin, Vitamin B[0512] 12, cyancobalamin injection (e.g., REDISOL™, RUBRAMIN PC™), hydroxocobalamin, folic acid (e.g., FOLVITE™), leucovorin (folinic acid, 5-CHOH4PteGlu, citrovorum factor) or WELLCOVORIN (Calcium salt of leucovorin), transferrin or ferritin.
  • In certain embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat psychiatric disorders. Psychiatric drugs that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antipsychotic agents (e.g., chlorpromazine, chlorprothixene, clozapine, fluphenazine, haloperidol, loxapine, mesoridazine, molindone, olanzapine, perphenazine, pimozide, quetiapine, risperidone, thioridazine, thiothixene, trifluoperazine, and triflupromazine), antimanic agents (e.g., carbamazepine, divalproex sodium, lithium carbonate, and lithium citrate), antidepressants (e.g., amitriptyline, amoxapine, bupropion, citalopram, clomipramine, desipramine, doxepin, fluvoxamine, fluoxetine, imipramine, isocarboxazid, maprotiline, mirtazapine, nefazodone, nortriptyline, paroxetine, phenelzine, protriptyline, sertraline, tranylcypromine, trazodone, trimipramine, and venlafaxine), antianxiety agents (e.g., alprazolam, buspirone, chlordiazepoxide, clorazepate, diazepam, halazepam, lorazepam, oxazepam, and prazepam), and stimulants (e.g., d-amphetamine, methylphenidate, and pemoline). [0513]
  • In other embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with agents used to treat neurological disorders. Neurological agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, antiepileptic agents (e.g., carbamazepine, clonazepam, ethosuximide, phenobarbital, phenytoin, primidone, valproic acid, divalproex sodium, felbamate, gabapentin, lamotrigine, levetiracetam, oxcarbazepine, tiagabine, topiramate, zonisamide, diazepam, lorazepam, and clonazepam), antiparkinsonian agents (e.g., levodopa/carbidopa, selegiline, amantidine, bromocriptine, pergolide, ropinirole, pramipexole, benztropine; biperiden; ethopropazine; procyclidine; trihexyphenidyl, tolcapone), and ALS therapeutics (e.g. riluzole). In another embodiment, albumin fusion proteins and/or polynucleotides of the invention are administered in combination with vasodilating agents and/or calcium channel blocking agents. Vasodilating agents that may be administered with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to, Angiotensin Converting Enzyme (ACE) inhibitors (e.g., papaverine, isoxsuprine, benazepril, captopril, cilazapril, enalapril, enalaprilat, fosinopril, lisinopril, moexipril, perindopril, quinapril, ramipril, spirapril, trandolapril, and nylidrin), and nitrates (e.g., isosorbide dinitrate, isosorbide mononitrate, and nitroglycerin). Examples of calcium channel blocking agents that may be administered in combination with the albumin fusion proteins and/or polynucleotides of the invention include, but are not limited to amlodipine, bepridil, diltiazem, felodipine, flunarizine, isradipine, nicardipine, nifedipine, nimodipine, and verapamil. [0514]
  • In certain embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with treatments for gastrointestinal disorders. Treatments for gastrointestinal disorders that may be administered with the albumin fusion protein and/or polynucleotide of the invention include, but are not limited to, H[0515] 2 histamine receptor antagonists (e.g., TAGAMET™ (cimetidine), ZANTAC™ (ranitidine), PEPCID™ (famotidine), and AXID™ (nizatidine)); inhibitors of H+, K+ ATPase (e.g., PREVACID™ (lansoprazole) and PRILOSEC™ (omeprazole)); Bismuth compounds (e.g., PEPTO-BISMOL™ (bismuth subsalicylate) and DE-NOL™ (bismuth subcitrate)); various antacids; sucralfate; prostaglandin analogs (e.g. CYTOTEC™ (misoprostol)); muscarinic cholinergic antagonists; laxatives (e.g., surfactant laxatives, stimulant laxatives, saline and osmotic laxatives); antidiarrheal agents (e.g., LOMOTIL™ (diphenoxylate), MOTOFEN™ (diphenoxin), and IMODIUM™ (loperamide hydrochloride)), synthetic analogs of somatostatin such as SANDOSTATIN™ (octreotide), antiemetic agents (e.g., ZOFRAN™ (ondansetron), KYTRIL™ (granisetron hydrochloride), tropisetron, dolasetron, metoclopramide, chlorpromazine, perphenazine, prochlorperazine, promethazine, thiethylperazine, triflupromazine, domperidone, haloperidol, droperidol, trimethobenzamide, dexamethasone, methylprednisolone, dronabinol, and nabilone); D2 antagonists (e.g., metoclopramide, trimethobenzamide and chlorpromazine); bile salts; chenodeoxycholic acid; ursodeoxycholic acid; and pancreatic enzyme preparations such as pancreatin and pancrelipase.
  • In additional embodiments, the albumin fusion proteins and/or polynucleotides of the invention are administered in combination with other therapeutic or prophylactic regimens, such as, for example, radiation therapy. [0516]
  • The invention also provides a pharmaceutical pack or kit comprising one or more containers filled with one or more of the ingredients of the pharmaceutical compositions comprising albumin fusion proteins of the invention. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. [0517]
  • Gene Therapy [0518]
  • Constructs encoding albumin fusion proteins of the invention can be used as a part of a gene therapy protocol to deliver therapeutically effective doses of the albumin fusion protein. A preferred approach for in vivo introduction of nucleic acid into a cell is by use of a viral vector containing nucleic acid, encoding an albumin fusion protein of the invention. Infection of cells with a viral vector has the advantage that a large proportion of the targeted cells can receive the nucleic acid. Additionally, molecules encoded within the viral vector, e.g., by a cDNA contained in the viral vector, are expressed efficiently in cells which have taken up viral vector nucleic acid. [0519]
  • Retrovirus vectors and adeno-associated virus vectors can be used as a recombinant gene delivery system for the transfer of exogenous nucleic acid molecules encoding albumin fusion proteins in vivo. These vectors provide efficient delivery of nucleic acids into cells, and the transferred nucleic acids are stably integrated into the chromosomal DNA of the host. The development of specialized cell lines (termed “packaging cells”) which produce only replication-defective retroviruses has increased the utility of retroviruses for gene therapy, and defective retroviruses are characterized for use in gene transfer for gene therapy purposes (for a review see Miller, A. D. (1990) [0520] Blood 76:271). A replication defective retrovirus can be packaged into virions which can be used to infect a target cell through the use of a helper virus by standard techniques. Protocols for producing recombinant retroviruses and for infecting cells in vitro or in vivo with such viruses can be found in Current Protocols in Molecular Biology, Ausubel, F. M. et al., (eds.) Greene Publishing Associates, (1989), Sections 9.10-9.14 and other standard laboratory manuals.
  • Another viral gene delivery system useful in the present invention uses adenovirus-derived vectors. The genome of an adenovirus can be manipulated such that it encodes and expresses a gene product of interest but is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. See, for example, Berkner et al., [0521] BioTechniques 6:616 (1988); Rosenfeld et al., Science 252:431-434 (1991); and Rosenfeld et al., Cell 68:143-155 (1992). Suitable adenoviral vectors derived from the adenovirus strain Ad type 5 d1324 or other strains of adenovirus (e.g., Ad2, Ad3, Ad7 etc.) are known to those skilled in the art. Recombinant adenoviruses can be advantageous in certain circumstances in that they are not capable of infecting nondividing cells and can be used to infect a wide variety of cell types, including epithelial cells (Rosenfeld et al., (1992) cited supra). Furthermore, the virus particle is relatively stable and amenable to purification and concentration, and as above, can be modified so as to affect the spectrum of infectivity. Additionally, introduced adenoviral DNA (and foreign DNA contained therein) is not integrated into the genome of a host cell but remains episomal, thereby avoiding potential problems that can occur as a result of insertional mutagenesis in situations where introduced DNA becomes integrated into the host genome (e.g., retroviral DNA). Moreover, the carrying capacity of the adenoviral genome for foreign DNA is large (up to 8 kilobases) relative to other gene delivery vectors (Berkner et al., cited supra; Haj-Ahmand et al., J. Virol. 57:267 (1986)).
  • In another embodiment, non-viral gene delivery systems of the present invention rely on endocytic pathways for the uptake of the subject nucleotide molecule by the targeted cell. Exemplary gene delivery systems of this type include liposomal derived systems, poly-lysine conjugates, and artificial viral envelopes. In a representative embodiment, a nucleic acid molecule encoding an albumin fusion protein of the invention can be entrapped in liposomes bearing positive charges on their surface (e.g., lipofectins) and (optionally) which are tagged with antibodies against cell surface antigens of the target tissue (Mizuno et al. (1992) [0522] No Shinkei Geka 20:547-551; PCT publication WO91/06309; Japanese patent application 1047381; and European patent publication EP-A-43075).
  • Gene delivery systems for a gene encoding an albumin fusion protein of the invention can be introduced into a patient by any of a number of methods. For instance, a pharmaceutical preparation of the gene delivery system can be introduced systemically, e.g. by intravenous injection, and specific transduction of the protein in the target cells occurs predominantly from specificity of transfection provided by the gene delivery vehicle, cell-type or tissue-type expression due to the transcriptional regulatory sequences controlling expression of the receptor gene, or a combination thereof. In other embodiments, initial delivery of the recombinant gene is more limited with introduction into the animal being quite localized. For example, the gene delivery vehicle can be introduced by catheter (see U.S. Pat. No. 5,328,470) or by Stereotactic injection (e.g. Chen et al. (1994) [0523] PNAS 91: 3054-3057). The pharmaceutical preparation of the gene therapy construct can consist essentially of the gene delivery system in an acceptable diluent, or can comprise a slow release matrix in which the gene delivery vehicle is imbedded. Where the albumin fusion protein can be produced intact from recombinant cells, e.g. retroviral vectors, the pharmaceutical preparation can comprise one or more cells which produce the albumin fusion protein.
  • Additional Gene Therapy Methods [0524]
  • Also encompassed by the invention are gene therapy methods for treating or preventing disorders, diseases and conditions. The gene therapy methods relate to the introduction of nucleic acid (DNA, RNA and antisense DNA or RNA) sequences into an animal to achieve expression of an albumin fusion protein of the invention. This method requires a polynucleotide which codes for an albumin fusion protein of the present invention operatively linked to a promoter and any other genetic elements necessary for the expression of the fusion protein by the target tissue. Such gene therapy and delivery techniques are known in the art, see, for example, WO90/11092, which is herein incorporated by reference. [0525]
  • Thus, for example, cells from a patient may be engineered with a polynucleotide (DNA or RNA) comprising a promoter operably linked to a polynucleotide encoding an albumin fusion protein of the present invention ex vivo, with the engineered cells then being provided to a patient to be treated with the fusion protein of the present invention. Such methods are well-known in the art. For example, see Belldegrun, A., et al., J. Natl. Cancer Inst. 85: 207-216 (1993); Ferrantini, M. et al., Cancer Research 53: 1107-1112 (1993); Ferrantini, M. et al., J. Immunology 153: 4604-4615 (1994); Kaido, T., et al., Int. J. Cancer 60: 221-229 (1995); Ogura, H., et al., Cancer Research 50: 5102-5106 (1990); Santodonato, L., et al., Human Gene Therapy 7:1-10 (1996); Santodonato, L., et al., Gene Therapy 4:1246-1255 (1997); and Zhang, J.-F. et al., Cancer Gene Therapy 3: 31-38 (1996)), which are herein incorporated by reference. In one embodiment, the cells which are engineered are arterial cells. The arterial cells may be reintroduced into the patient through direct injection to the artery, the tissues surrounding the artery, or through catheter injection. [0526]
  • As discussed in more detail below, the polynucleotide constructs can be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, and the like). The polynucleotide constructs may be delivered in a pharmaceutically acceptable liquid or aqueous carrier. [0527]
  • In one embodiment, polynucleotides encoding the albumin fusion proteins of the present invention is delivered as a naked polynucleotide. The term “naked” polynucleotide, DNA or RNA refers to sequences that are free from any delivery vehicle that acts to assist, promote or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like. However, polynucleotides encoding the albumin fusion proteins of the present invention can also be delivered in liposome formulations and lipofectin formulations and the like can be prepared by methods well known to those skilled in the art. Such methods are described, for example, in U.S. Pat. Nos. 5,593,972, 5,589,466, and 5,580,859, which are herein incorporated by reference. [0528]
  • The polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Appropriate vectors include pWLNEO, pSV2CAT, pOG44, pXT1 and pSG available from Stratagene; pSVK3, pBPV, pMSG and pSVL available from Pharmacia; and pEF1/V5, pcDNA3.1, and pRc/CMV2 available from Invitrogen. Other suitable vectors will be readily apparent to the skilled artisan. [0529]
  • Any strong promoter known to those skilled in the art can be used for driving the expression of the polynucleotide sequence. Suitable promoters include adenoviral promoters, such as the adenoviral major late promoter; or heterologous promoters, such as the cytomegalovirus (CMV) promoter; the respiratory syncytial virus (RSV) promoter; inducible promoters, such as the MMT promoter, the metallothionein promoter; heat shock promoters; the albumin promoter; the ApoAI promoter; human globin promoters; viral thymidine kinase promoters, such as the Herpes Simplex thymidine kinase promoter; retroviral LTRs; the b-actin promoter; and human growth hormone promoters. The promoter also may be the native promoter for the gene corresponding to the Therapeutic protein portion of the albumin fusion proteins of the invention. [0530]
  • Unlike other gene therapy techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months. [0531]
  • The polynucleotide construct can be delivered to the interstitial space of tissues within the an animal, including of muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue. Interstitial space of the tissues comprises the intercellular, fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides. [0532]
  • For the naked nucleic acid sequence injection, an effective dosage amount of DNA or RNA will be in the range of from about 0.05 mg/kg body weight to about 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration. [0533]
  • The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, other parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose. In addition, naked DNA constructs can be delivered to arteries during angioplasty by the catheter used in the procedure. [0534]
  • The naked polynucleotides are delivered by any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, and so-called “gene guns”. These delivery methods are known in the art. [0535]
  • The constructs may also be delivered with delivery vehicles such as viral sequences, viral particles, liposome formulations, lipofectin, precipitating agents, etc. Such methods of delivery are known in the art. [0536]
  • In certain embodiments, the polynucleotide constructs are complexed in a liposome preparation. Liposomal preparations for use in the instant invention include cationic (positively charged), anionic (negatively charged) and neutral preparations. However, cationic liposomes are particularly preferred because a tight charge complex can be formed between the cationic liposome and the polyanionic nucleic acid. Cationic liposomes have been shown to mediate intracellular delivery of plasmid DNA (Felgner et al., Proc. Natl. Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference); mRNA (Malone et al., Proc. Natl. Acad. Sci. USA (1989) 86:6077-6081, which is herein incorporated by reference); and purified transcription factors (Debs et al., J. Biol. Chem. (1990) 265:10189-10192, which is herein incorporated by reference), in functional form. [0537]
  • Cationic liposomes are readily available. For example, N[1-2,3-dioleyloxy)propyl]-N,N,N-triethylammonium (DOTMA) liposomes are particularly useful and are available under the trademark Lipofectin, from GIBCO BRL, Grand Island, N.Y. (See, also, Felgner et al., Proc. Natl. Acad. Sci. USA (1987) 84:7413-7416, which is herein incorporated by reference). Other commercially available liposomes include transfectace (DDAB/DOPE) and DOTAP/DOPE (Boehringer). [0538]
  • Other cationic liposomes can be prepared from readily available materials using techniques well known in the art. See, e.g. PCT Publication No. WO 90/11092 (which is herein incorporated by reference) for a description of the synthesis of DOTAP (1,2-bis(oleoyloxy)-3-(trimethylammonio)propane) liposomes. Preparation of DOTMA liposomes is explained in the literature, see, e.g., P. Felgner et al., Proc. Natl. Acad. Sci. USA 84:7413-7417, which is herein incorporated by reference. Similar methods can be used to prepare liposomes from other cationic lipid materials. [0539]
  • Similarly, anionic and neutral liposomes are readily available, such as from Avanti Polar Lipids (Birmingham, Ala.), or can be easily prepared using readily available materials. Such materials include phosphatidyl, choline, cholesterol, phosphatidyl ethanolamine, dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), dioleoylphoshatidyl ethanolamine (DOPE), among others. These materials can also be mixed with the DOTMA and DOTAP starting materials in appropriate ratios. Methods for making liposomes using these materials are well known in the art. [0540]
  • For example, commercially dioleoylphosphatidyl choline (DOPC), dioleoylphosphatidyl glycerol (DOPG), and dioleoylphosphatidyl ethanolamine (DOPE) can be used in various combinations to make conventional liposomes, with or without the addition of cholesterol. Thus, for example, DOPG/DOPC vesicles can be prepared by drying 50 mg each of DOPG and DOPC under a stream of nitrogen gas into a sonication vial. The sample is placed under a vacuum pump overnight and is hydrated the following day with deionized water. The sample is then sonicated for 2 hours in a capped vial, using a Heat Systems model 350 sonicator equipped with an inverted cup (bath type) probe at the maximum setting while the bath is circulated at 15EC. Alternatively, negatively charged vesicles can be prepared without sonication to produce multilamellar vesicles or by extrusion through nucleopore membranes to produce unilamellar vesicles of discrete size. Other methods are known and available to those of skill in the art. [0541]
  • The liposomes can comprise multilamellar vesicles (MLVs), small unilamellar vesicles (SUVs), or large unilamellar vesicles (LUVs), with SUVs being preferred. The various liposome-nucleic acid complexes are prepared using methods well known in the art. See, e.g., Straubinger et al., Methods of Immunology (1983), 101:512-527, which is herein incorporated by reference. For example, MLVs containing nucleic acid can be prepared by depositing a thin film of phospholipid on the walls of a glass tube and subsequently hydrating with a solution of the material to be encapsulated. SUVs are prepared by extended sonication of MLVs to produce a homogeneous population of unilamellar liposomes. The material to be entrapped is added to a suspension of preformed MLVs and then sonicated. When using liposomes containing cationic lipids, the dried lipid film is resuspended in an appropriate solution such as sterile water or an isotonic buffer solution such as 10 mM Tris/NaCl, sonicated, and then the preformed liposomes are mixed directly with the DNA. The liposome and DNA form a very stable complex due to binding of the positively charged liposomes to the cationic DNA. SUVs find use with small nucleic acid fragments. LUVs are prepared by a number of methods, well known in the art. Commonly used methods include Ca[0542] 2+-EDTA chelation (Papahadjopoulos et al., Biochim. Biophys. Acta (1975) 394:483; Wilson et al., Cell 17:77 (1979)); ether injection (Deamer, D. and Bangham, A., Biochim. Biophys. Acta 443:629 (1976); Ostro et al., Biochem. Biophys. Res. Commun. 76:836 (1977); Fraley et al., Proc. Natl. Acad. Sci. USA 76:3348 (1979)); detergent dialysis (Enoch, H. and Strittmatter, P., Proc. Natl. Acad. Sci. USA 76:145 (1979)); and reverse-phase evaporation (REV) (Fraley et al., J. Biol. Chem. 255:10431 (1980); Szoka, F. and Papahadjopoulos, D., Proc. Natl. Acad. Sci. USA 75:145 (1978); Schaefer-Ridder et al., Science 215:166 (1982)), which are herein incorporated by reference.
  • Generally, the ratio of DNA to liposomes will be from about 10:1 to about 1:10. Preferably, the ration will be from about 5:1 to about 1:5. More preferably, the ration will be about 3:1 to about 1:3. Still more preferably, the ratio will be about 1:1. [0543]
  • U.S. Pat. No. 5,676,954 (which is herein incorporated by reference) reports on the injection of genetic material, complexed with cationic liposomes carriers, into mice. U.S. Pat. Nos. 4,897,355, 4,946,787, 5,049,386, 5,459,127, 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 (which are herein incorporated by reference) provide cationic lipids for use in transfecting DNA into cells and mammals. U.S. Pat. Nos. 5,589,466, 5,693,622, 5,580,859, 5,703,055, and international publication no. WO 94/9469 provide methods for delivering DNA-cationic lipid complexes to mammals. [0544]
  • In certain embodiments, cells are engineered, ex vivo or in vivo, using a retroviral particle containing RNA which comprises a sequence encoding an albumin fusion protein of the present invention. Retroviruses from which the retroviral plasmid vectors may be derived include, but are not limited to, Moloney Murine Leukemia Virus, spleen necrosis virus, Rous sarcoma Virus, Harvey Sarcoma Virus, avian leukosis virus, gibbon ape leukemia virus, human immunodeficiency virus, Myeloproliferative Sarcoma Virus, and mammary tumor virus. [0545]
  • The retroviral plasmid vector is employed to transduce packaging cell lines to form producer cell lines. Examples of packaging cells which may be transfected include, but are not limited to, the PE501, PA317, R-2, R-AM, PA12, T19-14X, VT-19-17-H2, RCRE, RCRIP, GP+E-86, GP+envAm12, and DAN cell lines as described in Miller, Human Gene Therapy 1:5-14 (1990), which is incorporated herein by reference in its entirety. The vector may transduce the packaging cells through any means known in the art. Such means include, but are not limited to, electroporation, the use of liposomes, and CaPO[0546] 4 precipitation. In one alternative, the retroviral plasmid vector may be encapsulated into a liposome, or coupled to a lipid, and then administered to a host.
  • The producer cell line generates infectious retroviral vector particles which include polynucleotide encoding an albumin fusion protein of the present invention. Such retroviral vector particles then may be employed, to transduce eukaryotic cells, either in vitro or in vivo. The transduced eukaryotic cells will express a fusion protin of the present invention. [0547]
  • In certain other embodiments, cells are engineered, ex vivo or in vivo, with polynucleotide contained in an adenovirus vector. Adenovirus can be manipulated such that it encodes and expresses fusion protein of the present invention, and at the same time is inactivated in terms of its ability to replicate in a normal lytic viral life cycle. Adenovirus expression is achieved without integration of the viral DNA into the host cell chromosome, thereby alleviating concerns about insertional mutagenesis. Furthermore, adenoviruses have been used as live enteric vaccines for many years with an excellent safety profile (Schwartz et al. Am. Rev. Respir. Dis. 109:233-238 (1974)). Finally, adenovirus mediated gene transfer has been demonstrated in a number of instances including transfer of alpha-1-antitrypsin and CFTR to the lungs of cotton rats (Rosenfeld, M. A. et al. (1991) Science 252:431-434; Rosenfeld et al., (1992) Cell 68:143-155). Furthermore, extensive studies to attempt to establish adenovirus as a causative agent in human cancer were uniformly negative (Green, M. et al. (1979) Proc. Natl. Acad. Sci. USA 76:6606). [0548]
  • Suitable adenoviral vectors useful in the present invention are described, for example, in Kozarsky and Wilson, Curr. Opin. Genet. Devel. 3:499-503 (1993); Rosenfeld et al., Cell 68:143-155 (1992); Engelhardt et al., Human Genet. Ther. 4:759-769 (1993); Yang et al., Nature Genet. 7:362-369 (1994); Wilson et al., Nature 365:691-692 (1993); and U.S. Pat. No. 5,652,224, which are herein incorporated by reference. For example, the adenovirus vector Ad2 is useful and can be grown in human 293 cells. These cells contain the E1 region of adenovirus and constitutively express E1a and E1b, which complement the defective adenoviruses by providing the products of the genes deleted from the vector. In addition to Ad2, other varieties of adenovirus (e.g., Ad3, Ad5, and Ad7) are also useful in the present invention. [0549]
  • Preferably, the adenoviruses used in the present invention are replication deficient. Replication deficient adenoviruses require the aid of a helper virus and/or packaging cell line to form infectious particles. The resulting virus is capable of infecting cells and can express a polynucleotide of interest which is operably linked to a promoter, but cannot replicate in most cells. Replication deficient adenoviruses may be deleted in one or more of all or a portion of the following genes: E1a, E1b, E3, E4, E2a, or L1 through L5. [0550]
  • In certain other embodiments, the cells are engineered, ex vivo or in vivo, using an adeno-associated virus (AAV). AAVs are naturally occurring defective viruses that require helper viruses to produce infectious particles (Muzyczka, N., Curr. Topics in Microbiol. Immunol. 158:97 (1992)). It is also one of the few viruses that may integrate its DNA into non-dividing cells. Vectors containing as little as 300 base pairs of AAV can be packaged and can integrate, but space for exogenous DNA is limited to about 4.5 kb. Methods for producing and using such AAVs are known in the art. See, for example, U.S. Pat. Nos. 5,139,941, 5,173,414, 5,354,678, 5,436,146, 5,474,935, 5,478,745, and 5,589,377. [0551]
  • For example, an appropriate AAV vector for use in the present invention will include all the sequences necessary for DNA replication, encapsidation, and host-cell integration. The polynucleotide construct is inserted into the AAV vector using standard cloning methods, such as those found in Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Press (1989). The recombinant AAV vector is then transfected into packaging cells which are infected with a helper virus, using any standard technique, including lipofection, electroporation, calcium phosphate precipitation, etc. Appropriate helper viruses include adenoviruses, cytomegaloviruses, vaccinia viruses, or herpes viruses. Once the packaging cells are transfected and infected, they will produce infectious AAV viral particles which contain the polynucleotide construct. These viral particles are then used to transduce eukaryotic cells, either ex vivo or in vivo. The transduced cells will contain the polynucleotide construct integrated into its genome, and will express a fsuion protein of the invention. [0552]
  • Another method of gene therapy involves operably associating heterologous control regions and endogenous polynucleotide sequences (e.g. encoding a polypeptide of the present invention) via homologous recombination (see, e.g., U.S. Pat. No. 5,641,670, issued Jun. 24, 1997; International Publication No. WO 96/29411, published Sep. 26, 1996; International Publication No. WO 94/12650, published Aug. 4, 1994; Koller et al., Proc. Natl. Acad. Sci. USA 86:8932-8935 (1989); and Zijlstra et al., Nature 342:435-438 (1989), which are herein encorporated by reference. This method involves the activation of a gene which is present in the target cells, but which is not normally expressed in the cells, or is expressed at a lower level than desired. [0553]
  • Polynucleotide constructs are made, using standard techniques known in the art, which contain the promoter with targeting sequences flanking the promoter. Suitable promoters are described herein. The targeting sequence is sufficiently complementary to an endogenous sequence to permit homologous recombination of the promoter-targeting sequence with the endogenous sequence. The targeting sequence will be sufficiently near the 5′ end of the desired endogenous polynucleotide sequence so the promoter will be operably linked to the endogenous sequence upon homologous recombination. [0554]
  • The promoter and the targeting sequences can be amplified using PCR. Preferably, the amplified promoter contains distinct restriction enzyme sites on the 5′ and 3′ ends. Preferably, the 3′ end of the first targeting sequence contains the same restriction enzyme site as the 5′ end of the amplified promoter and the 5′ end of the second targeting sequence contains the same restriction site as the 3′ end of the amplified promoter. The amplified promoter and targeting sequences are digested and ligated together. [0555]
  • The promoter-targeting sequence construct is delivered to the cells, either as naked polynucleotide, or in conjunction with transfection-facilitating agents, such as liposomes, viral sequences, viral particles, whole viruses, lipofection, precipitating agents, etc., described in more detail above. The P promoter-targeting sequence can be delivered by any method, included direct needle injection, intravenous injection, topical administration, catheter infusion, particle accelerators, etc. The methods are described in more detail below. [0556]
  • The promoter-targeting sequence construct is taken up by cells. Homologous recombination between the construct and the endogenous sequence takes place, such that an endogenous sequence is placed under the control of the promoter. The promoter then drives the expression of the endogenous sequence. [0557]
  • The polynucleotide encoding an albumin fusion protein of the present invention may contain a secretory signal sequence that facilitates secretion of the protein. Typically, the signal sequence is positioned in the coding region of the polynucleotide to be expressed towards or at the 5′ end of the coding region. The signal sequence may be homologous or heterologous to the polynucleotide of interest and may be homologous or heterologous to the cells to be transfected. Additionally, the signal sequence may be chemically synthesized using methods known in the art. [0558]
  • Any mode of administration of any of the above-described polynucleotides constructs can be used so long as the mode results in the expression of one or more molecules in an amount sufficient to provide a therapeutic effect. This includes direct needle injection, systemic injection, catheter infusion, biolistic injectors, particle accelerators (i.e., “gene guns”), gelfoam sponge depots, other commercially available depot materials, osmotic pumps (e.g., Alza minipumps), oral or suppositorial solid (tablet or pill) pharmaceutical formulations, and decanting or topical applications during surgery. For example, direct injection of naked calcium phosphate-precipitated plasmid into rat liver and rat spleen or a protein-coated plasmid into the portal vein has resulted in gene expression of the foreign gene in the rat livers (Kaneda et al., Science 243:375 (1989)). [0559]
  • A preferred method of local administration is by direct injection. Preferably, an albumin fusion protein of the present invention complexed with a delivery vehicle is administered by direct injection into or locally within the area of arteries. Administration of a composition locally within the area of arteries refers to injecting the composition centimeters and preferably, millimeters within arteries. [0560]
  • Another method of local administration is to contact a polynucleotide construct of the present invention in or around a surgical wound. For example, a patient can undergo surgery and the polynucleotide construct can be coated on the surface of tissue inside the wound or the construct can be injected into areas of tissue inside the wound. [0561]
  • Therapeutic compositions useful in systemic administration, include fusion proteins of the present invention complexed to a targeted delivery vehicle of the present invention. Suitable delivery vehicles for use with systemic administration comprise liposomes comprising ligands for targeting the vehicle to a particular site. In specific embodiments, suitable delivery vehicles for use with systemic administration comprise liposomes comprising albumin fusion proteins of the invention for targeting the vehicle to a particular site. [0562]
  • Preferred methods of systemic administration, include intravenous injection, aerosol, oral and percutaneous (topical) delivery. Intravenous injections can be performed using methods standard in the art. Aerosol delivery can also be performed using methods standard in the art (see, for example, Stribling et al., Proc. Natl. Acad. Sci. USA 189:11277-11281, 1992, which is incorporated herein by reference). Oral delivery can be performed by complexing a polynucleotide construct of the present invention to a carrier capable of withstanding degradation by digestive enzymes in the gut of an animal. Examples of such carriers, include plastic capsules or tablets, such as those known in the art. Topical delivery can be performed by mixing a polynucleotide construct of the present invention with a lipophilic reagent (e.g., DMSO) that is capable of passing into the skin. [0563]
  • Determining an effective amount of substance to be delivered can depend upon a number of factors including, for example, the chemical structure and biological activity of the substance, the age and weight of the animal, the precise condition requiring treatment and its severity, and the route of administration. The frequency of treatments depends upon a number of factors, such as the amount of polynucleotide constructs administered per dose, as well as the health and history of the subject. The precise amount, number of doses, and timing of doses will be determined by the attending physician or veterinarian. [0564]
  • Albumin fusion proteins of the present invention can be administered to any animal, preferably to mammals and birds. Preferred mammals include humans, dogs, cats, mice, rats, rabbits sheep, cattle, horses and pigs, with humans being particularly preferred. [0565]
  • Biological Activities [0566]
  • Albumin fusion proteins and/or polynucleotides encoding albumin fusion proteins of the present invention, can be used in assays to test for one or more biological activities. If an albumin fusion protein and/or polynucleotide exhibits an activity in a particular assay, it is likely that the Therapeutic protein corresponding to the fusion portein may be involved in the diseases associated with the biological activity. Thus, the fusion protein could be used to treat the associated disease. [0567]
  • Members of the secreted family of proteins are believed to be involved in biological activities associated with, for example, cellular signaling. Accordingly, albumin fusion proteins of the invention and polynucleotides encoding these protiens, may be used in diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with aberrant activity of secreted polypeptides. [0568]
  • In preferred embodiments, fusion proteins of the present invention may be used in the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders relating to diseases and disorders of the endocrine system, the nervous system (See, for example, “Neurological Disorders” section below), and the immune system (See, for example, “Immune Activity” section below), respiratory system (See, for example, “Respiratory Disorders” section below), cardiovascular system (See, for example, “Cardiovascular Disorders” section below), reproductive system (See, for example, “Reproductive System Disorders” section below) digestive system (See, for example, “Gastrointestinal Disorders” section below), diseases and/or disorders relating to cell proliferation (See, for example, “Hyperproliferative Disorders” section below), and/or diseases or disorders relating to the blood ((See, for example, “Blood-Related Disorders” section below). [0569]
  • In preferred embodiments, the present invention encompasses a method of treating a disease or disorder listed in the “Preferred Indication Y” column of Table 1 comprising administering to a patient in which such treatment, prevention or amelioration is desired an albumin fusion protein of the invention that comprises a Therapeutic protein portion corresponding to a Therapeutic protein disclosed in the “Therapeutic Protein X” column of Table 1 (in the same row as the disease or disorder to be treated is listed in the “Preferred Indication Y” column of Table 1) in an amount effective to treat, prevent or ameliorate the disease or disorder. [0570]
  • In certain embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to a Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., leukemia, cancer, and/or as described below under “Hyperproliferative Disorders”). [0571]
  • In additional embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a neoplasm located in a tissue selected from the group consisting of: colon, abdomen, bone, breast, digestive system, liver, pancreas, prostate, peritoneum, lung, blood (e.g., leukemia), endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), uterus, eye, head and neck, nervous (central and peripheral), lymphatic system, pelvic, skin, soft tissue, spleen, thoracic, and urogenital. [0572]
  • In other embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a pre-neoplastic condition, selected from the group consisting of: hyperplasia (e.g., endometrial hyperplasia and/or as described in the section entitled “Hyperproliferative Disorders”), metaplasia (e.g., connective tissue metaplasia, a typical metaplasia, and/or as described in the section entitled “Hyperproliferative Disorders”), and dysplasia (e.g., cervical dysplasia, and bronchopulmonary dysplasia). [0573]
  • In additional embodiments, a Therapeutic protein having a “Cancer” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a benign dysproliferative disorder selected from the group consisting of: benign tumors, fibrocystic conditions, tissue hypertrophy, and/or as described in the section entitled “Hyperproliferative Disorders”. [0574]
  • In certain embodiments, a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a blood disorder (e.g., as described below under “Immune Activity”, “Cardiovascular Disorders” and/or “Blood-Related Disorders”), and/or an infection (e.g., as described below under “Infectious Disease”). [0575]
  • In additional embodiments, a Therapeutic protein having a “Immune/Hematopoietic” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: anemia, pancytopenia, leukopenia, thrombocytopenia, leukemias, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL), plasmacytomas, multiple myeloma, Burkitt's lymphoma, arthritis, asthma, AIDS, autoimmune disease, rheumatoid arthritis, granulomatous disease, immune deficiency, inflammatory bowel disease, sepsis, neutropenia, neutrophilia, psoriasis, an immune reaction to a transplanted organ and/or tissue, systemic lupus erythematosis, hemophilia, hypercoagulation, diabetes mellitus, endocarditis, meningitis, Lyme Disease, and allergies. [0576]
  • In other embodiments, a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the reproductive system (e.g., as described below under “Reproductive System Disorders”). [0577]
  • In additional embodiments, a Therapeutic protein having a “Reproductive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: cryptorchism, prostatitis, inguinal hernia, varicocele, a leydig cell tumor, verrucous carcinoma, prostatitis, malacoplakia, Peyronie's disease, penile carcinoma, squamous cell hyperplasia, dysmenorrhea, ovarian adenocarcinoma, Turner's syndrome, mucopurulent cervicitis, a Sertoli-leydig tumor, ovarian cancer, uterine cancer, pelvic inflammatory disease, testicular cancer, prostate cancer, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes mellitus, cystic fibrosis, Kartagener's syndrome, testicular atrophy, testicular feminization, anorchia, ectopic testis, epididymitis, orchitis, gonorrhea, syphilis, testicular torsion, vasitis nodosa, a germ cell tumor, a stromal tumor, dysmenorrhea, retroverted uterus, endometriosis, fibroids, adenomyosis, anovulatory bleeding, amenorrhea, Cushing's syndrome, a hydatidiform mole, Asherman's syndrome, premature menopause, precocious puberty, uterine polyps, dysfunctional uterine bleeding, cervicitis, chronic cervicitis, mucopurulent cervicitis, cervical dysplasia, cervical polyps, Nabothian cysts, cervical erosion, cervical incompetence, a cervical neoplasm, pseudohermaphroditism, and premenstrual syndrome. [0578]
  • In other embodiments, a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the immune system (e.g., as described below under “Immune Activity”). [0579]
  • In further embodiments, a Therapeutic protein having a “Musculoskeletal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of bone cancer (e.g., osteochondroma, benign chondroma, chondroblastoma, chondromyxoid fibroma, osteoid osteoma, giant cell tumor, multiple myeloma, and osteosarcoma), Paget's Disease, rheumatoid arthritis, systemic lupus erythematosus, osteomyelitis, Lyme Disease, gout, bursitis, tendonitis, osteoporosis, osteoarthritis, muscular dystrophy, mitochondrial myopathy, cachexia, and multiple sclerosis. [0580]
  • In other embodiments, a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), and/or a disorder of the cardiovascular system (e.g., as described below under “Cardiovascular Disorders”). [0581]
  • In additional embodiments, a Therapeutic protein having a “Cardiovascular” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: myxoma, fibroma, rhabdomyoma, cardiovascular abnormality (e.g., a congenital heart defect, cerebral arterioyenous malformation, septal defect), heart disease (e.g., heart failure, congestive heart disease, arrhythmia, tachycardia, fibrillation, pericardial Disease, endocarditis), cardiac arrest, heart valve disease (e.g., stenosis, regurgitation, prolapse), vascular disease (e.g., hypertension, coronary artery disease, angina, aneurysm, arteriosclerosis, peripheral vascular disease), hyponatremia, hypernatremia, hypokalemia, and hyperkalemia. [0582]
  • In other embodiments, a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”). [0583]
  • In further embodiments, a Therapeutic protein having a “Mixed Fetal” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: spina bifida, hydranencephaly, neurofibromatosis, fetal alcohol syndrome, diabetes mellitus, PKU, Down's syndrome, Patau syndrome, Edwards syndrome, Turner syndrome, Apert syndrome, Carpenter syndrome, Conradi syndrome, Crouzon syndrome, cutis laxa, Cornelia de Lange syndrome, Ellis-van Creveld syndrome, Holt-Oram syndrome, Kartagener syndrome, Meckel-Gruber syndrome, Noonan syndrome, Pallister-Hall syndrome, Rubinstein-Taybi syndrome, Scimitar syndrome, Smith-Lemli-Opitz syndrome, thromocytopenia-absent radius (TAR) syndrome, Treacher Collins syndrome, Williams syndrome, Hirschsprung's disease, Meckel's diverticulum, polycystic kidney disease, Turner's syndrome, and gonadal dysgenesis, Klippel-Feil syndrome, Ostogenesis imperfecta, muscular dystrophy, Tay-Sachs disease, Wilm's tumor, neuroblastoma, and retinoblastoma. [0584]
  • In further embodiments, a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a renal disorder (e.g., as described below under “Renal Disorders”). [0585]
  • In other embodiments, a Therapeutic protein having a “Excretory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: bladder cancer, prostate cancer, benign prostatic hyperplasia, bladder disorders (e.g., urinary incontinence, urinary retention, urinary obstruction, urinary tract Infections, interstitial cystitis, prostatitis, neurogenic bladder, hematuria), a renal disorder (e.g., hydronephrosis, proteinuria, renal failure, pyelonephritis, urolithiasis, reflux nephropathy, and unilateral obstructive uropathy). [0586]
  • In further embodiments, a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the nervous system (e.g., as described below under “Neural Activity and Neurological Diseases”). [0587]
  • In other embodiments, a Therapeutic protein having a “Neural/Sensory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: brain cancer (e.g., brain stem glioma, brain tumor, central nervous system (Primary) lymphoma, central nervous system lymphoma, cerebellar astrocytoma, and cerebral astrocytoma, a neurodegenerative disorder (e.g., Alzheimer's Disease, Creutzfeldt-Jakob Disease, Parkinson's Disease, and Idiopathic Presenile Dementia), encephalomyelitis, cerebral malaria, meningitis, a metabolic brain disease (e.g., phenylketonuria and pyruvate carboxylase deficiency), cerebellar ataxia, ataxia telangiectasia, and AIDS Dementia Complex, schizophrenia, attention deficit disorder, hyperactive attention deficit disorder, autism, and an obsessive compulsive disorder. [0588]
  • In other embodiments, a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the respiratory system (e.g., as described below under “Respiratory Disorders”). [0589]
  • In other embodiments, a Therapeutic protein having a “Respiratory” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: a cancer of the respiratory system (such as larynx cancer, pharynx cancer, trachea cancer, epiglottis cancer, lung cancer, squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcinoma), an allergic reaction, cystic fibrosis, sarcoidosis, histiocytosis X, an infiltrative lung disease (e.g., pulmonary fibrosis and lymphoid interstitial pneumonia), an obstructive airway disease (e.g., asthma, emphysema, chronic or acute bronchitis), an occupational lung disease (e.g., silicosis and asbestosis), pneumonia, and pleurisy. [0590]
  • In other embodiments, a Therapeutic protein having an “Endocrine” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a disease or disorder of the respiratory system (e.g., as described below under “Respiratory Disorders”), a renal disorder (e.g., as described below under “Renal Disorders”), and/or a disorder of the endocrine system (e.g., as described below under “Endocrine Disorders”). [0591]
  • In other embodiments, a Therapeutic protein having a “Endocrine” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: a cancer of endocrine tissues and/or organs (e.g., cancer of the hypothalamus, pituitary gland, thyroid gland, parathyroid glands, pancreas, adrenal glands, ovaries, and testes), diabetes (e.g., diabetes insipidus, type I and type II diabetes mellitus), obesity, a disorder related to pituitary glands (e.g., hyperpituitarism, hypopituitarism, and pituitary dwarfism), hypothyroidism, hyperthyroidism, goiter, reproductive disorders (e.g. male and female infertility), a disorder related to adrenal glands (e.g., Addison's Disease, corticosteroid deficiency, and Cushing's Syndrome), kidney cancer (e.g., hypemephroma, transitional cell cancer, and Wilm's tumor), diabetic nephropathy, interstitial nephritis, polycystic kidney disease, glomerulonephritis (e.g., IgM mesangial proliferative glomerulonephritis and glomerulonephritis caused by an autoimmune disorder; such as Goodpasture's syndrome), and nephrocalcinosis. [0592]
  • In additional embodiments, a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to, for example, a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”) and/or a disease or disorder of the gastrointestinal system (e.g., as described below under “Gastrointestinal Disorders”). [0593]
  • In other embodiments, a Therapeutic protein having a “Digestive” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: ulcerative colitis, appendicitis, Crohn's disease, hepatitis, hepatic encephalopathy, portal hypertension, cholelithiasis, cancer of the digestive system (e.g., biliary tract cancer, stomach cancer, colon cancer, gastric cancer, pancreatic cancer, cancer of the bile duct, a tumor of the colon (e.g., polyps or cancers), and cirrhosis), pancreatitis, ulcerative disease, pyloric stenosis, gastroenteritis, gastritis, gastric atropy, a benign tumor of the duodenum, distension, irritable bowel syndrome, malabsorption, a congenital disorder of the small intestine, bacterial and parasitic infection, megacolon, Hirschsprung's disease, aganglionic megacolon, acquired megacolon, colitis, a anorectal disorder (e.g., anal fistulas, hemorrhoids), a congenital disorder of the liver (e.g., Wilson's disease, hemochromatosis, cystic fibrosis, biliary atresia, and alpha1-antitrypsin deficiency), portal hypertension, cholelithiasis, and jaundice. [0594]
  • In further embodiments, a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder relating to a neoplastic disease (e.g., as described below under “Hyperproliferative Disorders”), a cellular and/or genetic abnormality (e.g., as described below under “Diseases at the Cellular Level”), angiogenesis (e.g., as described below under “Anti-Angiogenesis Activity”), and/or to promote or inhibit regeneration (e.g., as described below under “Regeneration”), and/or wound healing (e.g., as described below under “Wound Healing and Epithelial Cell Proliferation”). [0595]
  • In certain embodiments, a Therapeutic protein having a “Connective/Epithelial” recitation in the “Preferred Indication” column of Table 1, an albumin fusion protein that comprises a Therapeutic protein portion corresponding to this Therapeutic protein, and fragments and variants thereof, may be used to treat a disease and/or disorder selected from the group consisting of: connective tissue metaplasia, mixed connective tissue disease, focal epithelial hyperplasia, epithelial metaplasia, mucoepithelial dysplasia, graft v. host disease, polymyositis, cystic hyperplasia, cerebral dysplasia, tissue hypertrophy, Alzheimer's disease, lymphoproliferative disorder, Waldenstron's macroglobulinemia, Crohn's disease, pernicious anemia, idiopathic Addison's disease, glomerulonephritis, bullous pemphigoid, Sjogren's syndrome, diabetes mellitus, cystic fibrosis, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, osteoporosis, osteocarthritis, periodontal disease, wound healing, relapsing polychondritis, vasculitis, polyarteritis nodosa, Wegener's granulomatosis, cellulitis, rheumatoid arthritis, psoriatic arthritis, discoid lupus erythematosus, systemic lupus erythematosus, scleroderma, CREST syndrome, Sjogren's syndrome, polymyositis, dermatomyositis, mixed connective tissue disease, relapsing polychondritis, vasculitis, Henoch-Schonlein syndrome, erythema nodosum, polyarteritis nodosa, temporal (giant cell) arteritis, Takayasu's arteritis, Wegener's granulomatosis, Reiter's syndrome, Behcet's syndrome, ankylosing spondylitis, cellulitis, keloids, Ehler Danlos syndrome, Marfan syndrome, pseudoxantoma elasticum, osteogenese imperfecta, chondrodysplasias, epidermolysis bullosa, Alport syndrome, and cutis laxa. [0596]
  • In certain embodiments, an albumin fusion protein of the present invention may be used to diagnose and/or prognose diseases and/or disorders associated with the tissue(s) in which the gene corresponding to the Therapeutic protein portion of the fusion portien of the invention is expressed. [0597]
  • Thus, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention are useful in the diagnosis, detection and/or treatment of diseases and/or disorders associated with activities that include, but are not limited to, prohormone activation, neurotransmitter activity, cellular signaling, cellular proliferation, cellular differentiation, and cell migration. [0598]
  • More generally, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful for the diagnosis, prognosis, prevention and/or treatment of diseases and/or disorders associated with the following systems. [0599]
  • Immune Activity [0600]
  • Albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing diseases, disorders, and/or conditions of the immune system, by, for example, activating or inhibiting the proliferation, differentiation, or mobilization (chemotaxis) of immune cells. Immune cells develop through a process called hematopoiesis, producing myeloid (platelets, red blood cells, neutrophils, and macrophages) and lymphoid (B and T lymphocytes) cells from pluripotent stem cells. The etiology of these immune diseases, disorders, and/or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy or toxins), or infectious. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as a marker or detector of a particular immune system disease or disorder. [0601]
  • In another embodiment, a fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention, may be used to treat diseases and disorders of the immune system and/or to inhibit or enhance an immune response generated by cells associated with the tissue(s) in which the polypeptide of the invention is expressed. [0602]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing, and/or prognosing immunodeficiencies, including both congenital and acquired immunodeficiencies. Examples of B cell immunodeficiencies in which immunoglobulin levels B cell function and/or B cell numbers are decreased include: X-linked agammaglobulinemia (Bruton's disease), X-linked infantile agammaglobulinemia, X-linked immunodeficiency with hyper IgM, non X-linked immunodeficiency with hyper IgM, X-linked lymphoproliferative syndrome (XLP), agammaglobulinemia including congenital and acquired agammaglobulinemia, adult onset agammaglobulinemia, late-onset agammaglobulinemia, dysgammaglobulinemia, hypogammaglobulinemia, unspecified hypogammaglobulinemia, recessive agammaglobulinemia (Swiss type), Selective IgM deficiency, selective IgA deficiency, selective IgG subclass deficiencies, IgG subclass deficiency (with or without IgA deficiency), Ig deficiency with increased IgM, IgG and IgA deficiency with increased IgM, antibody deficiency with normal or elevated Igs, Ig heavy chain deletions, kappa chain deficiency, B cell lymphoproliferative disorder (BLPD), common variable immunodeficiency (CVID), common variable immunodeficiency (CVI) (acquired), and transient hypogammaglobulinemia of infancy. [0603]
  • In specific embodiments, ataxia-telangiectasia or conditions associated with ataxia-telangiectasia are treated, prevented, diagnosed, and/or prognosing using the, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0604]
  • Examples of congenital immunodeficiencies in which T cell and/or B cell function and/or number is decreased include, but are not limited to: DiGeorge anomaly, severe combined immunodeficiencies (SCID) (including, but not limited to, X-linked SCID, autosomal recessive SCID, adenosine deaminase deficiency, purine nucleoside phosphorylase (PNP) deficiency, Class II MHC deficiency (Bare lymphocyte syndrome), Wiskott-Aldrich syndrome, and ataxia telangiectasia), thymic hypoplasia, third and fourth pharyngeal pouch syndrome, 22q11.2 deletion, chronic mucocutaneous candidiasis, natural killer cell deficiency (NK), idiopathic CD4+ T-lymphocytopenia, immunodeficiency with predominant T cell defect (unspecified), and unspecified immunodeficiency of cell mediated immunity. [0605]
  • In specific embodiments, DiGeorge anomaly or conditions associated with DiGeorge anomaly are treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0606]
  • Other immunodeficiencies that may be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, chronic granulomatous disease, Chédiak-Higashi syndrome, myeloperoxidase deficiency, leukocyte glucose-6-phosphate dehydrogenase deficiency, X-linked lymphoproliferative syndrome (XLP), leukocyte adhesion deficiency, complement component deficiencies (including C1, C2, C3, C4, C5, C6, C7, C8 and/or C9 deficiencies), reticular dysgenesis, thymic alymphoplasia-aplasia, immunodeficiency with thymoma, severe congenital leukopenia, dysplasia with immunodeficiency, neonatal neutropenia, short limbed dwarfism, and Nezelof syndrome-combined immunodeficiency with Igs. [0607]
  • In a preferred embodiment, the immunodeficiencies and/or conditions associated with the immunodeficiencies recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0608]
  • In a preferred embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among immunodeficient individuals. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an agent to boost immunoresponsiveness among B cell and/or T cell immunodeficient individuals. [0609]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, diagnosing and/or prognosing autoimmune disorders. Many autoimmune disorders result from inappropriate recognition of self as foreign material by immune cells. This inappropriate recognition results in an immune response leading to the destruction of the host tissue. Therefore, the administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can inhibit an immune response, particularly the proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing autoimmune disorders. [0610]
  • Autoimmune diseases or disorders that may be treated, prevented, diagnosed and/or prognosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, one or more of the following: systemic lupus erythematosus, rheumatoid arthritis, ankylosing spondylitis, multiple sclerosis, autoimmune thyroiditis, Hashimoto's thyroiditis, autoimmune hemolytic anemia, hemolytic anemia, thrombocytopenia, autoimmune thrombocytopenia purpura, autoimmune neonatal thrombocytopenia, idiopathic thrombocytopenia purpura, purpura (e.g., Henloch-Scoenlein purpura), autoimmunocytopenia, Goodpasture's syndrome, Pemphigus vulgaris, myasthenia gravis, Grave's disease (hyperthyroidism), and insulin-resistant diabetes mellitus. [0611]
  • Additional disorders that are likely to have an autoimmune component that may be treated, prevented, and/or diagnosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, type II collagen-induced arthritis, antiphospholipid syndrome, dermatitis, allergic encephalomyelitis, myocarditis, relapsing polychondritis, rheumatic heart disease, neuritis, uveitis ophthalmia, polyendocrinopathies, Reiter's Disease, Stiff-Man Syndrome, autoimmune pulmonary inflammation, autism, Guillain-Barre Syndrome, insulin dependent diabetes mellitus, and autoimmune inflammatory eye disorders. [0612]
  • Additional disorders that are likely to have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, scleroderma with anti-collagen antibodies (often characterized, e.g., by nucleolar and other nuclear antibodies), mixed connective tissue disease (often characterized, e.g., by antibodies to extractable nuclear antigens (e.g., ribonucleoprotein)), polymyositis (often characterized, e.g., by nonhistone ANA), pernicious anemia (often characterized, e.g., by antiparietal cell, microsomes, and intrinsic factor antibodies), idiopathic Addison's disease (often characterized, e.g., by humoral and cell-mediated adrenal cytotoxicity, infertility (often characterized, e.g., by antispermatozoal antibodies), glomerulonephritis (often characterized, e.g., by glomerular basement membrane antibodies or immune complexes), bullous pemphigoid (often characterized, e.g., by IgG and complement in basement membrane), Sjogren's syndrome (often characterized, e.g., by multiple tissue antibodies, and/or a specific nonhistone ANA (SS-B)), diabetes mellitus (often characterized, e.g., by cell-mediated and humoral islet cell antibodies), and adrenergic drug resistance (including adrenergic drug resistance with asthma or cystic fibrosis) (often characterized, e.g., by beta-adrenergic receptor antibodies). [0613]
  • Additional disorders that may have an autoimmune component that may be treated, prevented, diagnosed and/or prognosed with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, chronic active hepatitis (often characterized, e.g., by smooth muscle antibodies), primary biliary cirrhosis (often characterized, e.g., by mitochondria antibodies), other endocrine gland failure (often characterized, e.g., by specific tissue antibodies in some cases), vitiligo (often characterized, e.g., by melanocyte antibodies), vasculitis (often characterized, e.g., by Ig and complement in vessel walls and/or low serum complement), post-MI (often characterized, e.g., by myocardial antibodies), cardiotomy syndrome (often characterized, e.g., by myocardial antibodies), urticaria (often characterized, e.g., by IgG and IgM antibodies to IgE), atopic dermatitis (often characterized, e.g., by IgG and IgM antibodies to IgE), asthma (often characterized, e.g., by IgG and IgM antibodies to IgE), and many other inflammatory, granulomatous, degenerative, and atrophic disorders. [0614]
  • In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using for example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. In a specific preferred embodiment, rheumatoid arthritis is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0615]
  • In another specific preferred embodiment, systemic lupus erythematosus is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. In another specific preferred embodiment, idiopathic thrombocytopenia purpura is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0616]
  • In another specific preferred embodiment IgA nephropathy is treated, prevented, and/or diagnosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0617]
  • In a preferred embodiment, the autoimmune diseases and disorders and/or conditions associated with the diseases and disorders recited above are treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0618]
  • In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a immunosuppressive agent(s). [0619]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, prognosing, and/or diagnosing diseases, disorders, and/or conditions of hematopoietic cells. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with a decrease in certain (or many) types hematopoietic cells, including but not limited to, leukopenia, neutropenia, anemia, and thrombocytopenia. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to increase differentiation and proliferation of hematopoietic cells, including the pluripotent stem cells, in an effort to treat or prevent those diseases, disorders, and/or conditions associated with an increase in certain (or many) types of hematopoietic cells, including but not limited to, histiocytosis. [0620]
  • Allergic reactions and conditions, such as asthma (particularly allergic asthma) or other respiratory problems, may also be treated, prevented, diagnosed and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Moreover, these molecules can be used to treat, prevent, prognose, and/or diagnose anaphylaxis, hypersensitivity to an antigenic molecule, or blood group incompatibility. [0621]
  • Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose and/or prognose IgE-mediated allergic reactions. Such allergic reactions include, but are not limited to, asthma, rhinitis, and eczema. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate IgE concentrations in vitro or in vivo. [0622]
  • Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention have uses in the diagnosis, prognosis, prevention, and/or treatment of inflammatory conditions. For example, since fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the activation, proliferation and/or differentiation of cells involved in an inflammatory response, these molecules can be used to prevent and/or treat chronic and acute inflammatory conditions. Such inflammatory conditions include, but are not limited to, for example, inflammation associated with infection (e.g., septic shock, sepsis, or systemic inflammatory response syndrome), ischemia-reperfusion injury, endotoxin lethality, complement-mediated hyperacute rejection, nephritis, cytokine or chemokine induced lung injury, inflammatory bowel disease, Crohn's disease, over production of cytokines (e.g., TNF or IL-1.), respiratory disorders (e.g., asthma and allergy); gastrointestinal disorders (e.g., inflammatory bowel disease); cancers (e.g., gastric, ovarian, lung, bladder, liver, and breast); CNS disorders (e.g., multiple sclerosis; ischemic brain injury and/or stroke, traumatic brain injury, neurodegenerative disorders (e.g., Parkinson's disease and Alzheimer's disease); AIDS-related dementia; and prion disease); cardiovascular disorders (e.g., atherosclerosis, myocarditis, cardiovascular disease, and cardiopulmonary bypass complications); as well as many additional diseases, conditions, and disorders that are characterized by inflammation (e.g., hepatitis, rheumatoid arthritis, gout, trauma, pancreatitis, sarcoidosis, dermatitis, renal ischemia-reperfusion injury, Grave's disease, systemic lupus erythematosus, diabetes mellitus, and allogenic transplant rejection). [0623]
  • Because inflammation is a fundamental defense mechanism, inflammatory disorders can effect virtually any tissue of the body. Accordingly, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, have uses in the treatment of tissue-specific inflammatory disorders, including, but not limited to, adrenalitis, alveolitis, angiocholecystitis, appendicitis, balanitis, blepharitis, bronchitis, bursitis, carditis, cellulitis, cervicitis, cholecystitis, chorditis, cochlitis, colitis, conjunctivitis, cystitis, dermatitis, diverticulitis, encephalitis, endocarditis, esophagitis, eustachitis, fibrositis, folliculitis, gastritis, gastroenteritis, gingivitis, glossitis, hepatosplenitis, keratitis, labyrinthitis, laryngitis, lymphangitis, mastitis, media otitis, meningitis, metritis, mucitis, myocarditis, myosititis, myringitis, nephritis, neuritis, orchitis, osteochondritis, otitis, pericarditis, peritendonitis, peritonitis, pharyngitis, phlebitis, poliomyelitis, prostatitis, pulpitis, retinitis, rhinitis, salpingitis, scleritis, sclerochoroiditis, scrotitis, sinusitis, spondylitis, steatitis, stomatitis, synovitis, syringitis, tendonitis, tonsillitis, urethritis, and vaginitis. [0624]
  • In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, prevent, and/or treat organ transplant rejections and graft-versus-host disease. Organ rejection occurs by host immune cell destruction of the transplanted tissue through an immune response. Similarly, an immune response is also involved in GVHD, but, in this case, the foreign transplanted immune cells destroy the host tissues. Polypeptides, antibodies, or polynucleotides of the invention, and/or agonists or antagonists thereof, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing organ rejection or GVHD. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, that inhibit an immune response, particularly the activation, proliferation, differentiation, or chemotaxis of T-cells, may be an effective therapy in preventing experimental allergic and hyperacute xenograft rejection. [0625]
  • In other embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are useful to diagnose, prognose, prevent, and/or treat immune complex diseases, including, but not limited to, serum sickness, post streptococcal glomerulonephritis, polyarteritis nodosa, and immune complex-induced vasculitis. [0626]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat, detect, and/or prevent infectious agents. For example, by increasing the immune response, particularly increasing the proliferation activation and/or differentiation of B and/or T cells, infectious diseases may be treated, detected, and/or prevented. The immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also directly inhibit the infectious agent (refer to section of application listing infectious agents, etc), without necessarily eliciting an immune response. [0627]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a vaccine adjuvant that enhances immune responsiveness to an antigen. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance tumor-specific immune responses. [0628]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-viral immune responses. Anti-viral immune responses that may be enhanced using the compositions of the invention as an adjuvant, include virus and virus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of: AIDS, meningitis, Dengue, EBV, and hepatitis (e.g., hepatitis B). In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a virus, disease, or symptom selected from the group consisting of: HIV/AIDS, respiratory syncytial virus, Dengue, rotavirus, Japanese B encephalitis, influenza A and B, parainfluenza, measles, cytomegalovirus, rabies, Junin, Chikungunya, Rift Valley Fever, herpes simplex, and yellow fever. [0629]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-bacterial or anti-fungal immune responses. Anti-bacterial or anti-fungal immune responses that may be enhanced using the compositions of the invention as an adjuvant, include bacteria or fungus and bacteria or fungus associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: tetanus, Diphtheria, botulism, and meningitis type B. [0630]
  • In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to a bacteria or fungus, disease, or symptom selected from the group consisting of: [0631] Vibrio cholerae, Mycobacterium leprae, Salmonella typhi, Salmonella paratyphi, Meisseria meningitidis, Streptococcus pneumoniae, Group B streptococcus, Shigella spp., Enterotoxigenic Escherichia coli, Enterohemorrhagic E. coli, and Borrelia burgdorferi.
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an adjuvant to enhance anti-parasitic immune responses. Anti-parasitic immune responses that may be enhanced using the compositions of the invention as an adjuvant, include parasite and parasite associated diseases or symptoms described herein or otherwise known in the art. In specific embodiments, the compositions of the invention are used as an adjuvant to enhance an immune response to a parasite. In another specific embodiment, the compositions of the invention are used as an adjuvant to enhance an immune response to Plasmodium (malaria) or Leishmania. [0632]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat infectious diseases including silicosis, sarcoidosis, and idiopathic pulmonary fibrosis; for example, by preventing the recruitment and activation of mononuclear phagocytes. [0633]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an antigen for the generation of antibodies to inhibit or enhance immune mediated responses against polypeptides of the invention. [0634]
  • In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are administered to an animal (e.g., mouse, rat, rabbit, hamster, guinea pig, pigs, micro-pig, chicken, camel, goat, horse, cow, sheep, dog, cat, non-human primate, and human, most preferably human) to boost the immune system to produce increased quantities of one or more antibodies (e.g., IgG, IgA, IgM, and IgE), to induce higher affinity antibody production and immunoglobulin class switching (e.g., IgG, IgA, IgM, and IgE), and/or to increase an immune response. [0635]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell responsiveness to pathogens. [0636]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an activator of T cells. [0637]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent that elevates the immune status of an individual prior to their receipt of immunosuppressive therapies. [0638]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to induce higher affinity antibodies. [0639]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to increase serum immunoglobulin concentrations. [0640]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to accelerate recovery of immunocompromised individuals. [0641]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among aged populations and/or neonates. [0642]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an immune system enhancer prior to, during, or after bone marrow transplant and/or other transplants (e.g., allogeneic or xenogeneic organ transplantation). With respect to transplantation, compositions of the invention may be administered prior to, concomitant with, and/or after transplantation. In a specific embodiment, compositions of the invention are administered after transplantation, prior to the beginning of recovery of T-cell populations. In another specific embodiment, compositions of the invention are first administered after transplantation after the beginning of recovery of T cell populations, but prior to full recovery of B cell populations. [0643]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having an acquired loss of B cell function. Conditions resulting in an acquired loss of B cell function that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, HIV Infection, AIDS, bone marrow transplant, and B cell chronic lymphocytic leukemia (CLL). [0644]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to boost immunoresponsiveness among individuals having a temporary immune deficiency. Conditions resulting in a temporary immune deficiency that may be ameliorated or treated by administering the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, recovery from viral infections (e.g., influenza), conditions associated with malnutrition, recovery from infectious mononucleosis, or conditions associated with stress, recovery from measles, recovery from blood transfusion, and recovery from surgery. [0645]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a regulator of antigen presentation by monocytes, dendritic cells, and/or B-cells. In one embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention enhance antigen presentation or antagonizes antigen presentation in vitro or in vivo. Moreover, in related embodiments, this enhancement or antagonism of antigen presentation may be useful as an anti-tumor treatment or to modulate the immune system. [0646]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as an agent to direct an individual's immune system towards development of a humoral response (i.e. TH2) as opposed to a TH1 cellular response. [0647]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means to induce tumor proliferation and thus make it more susceptible to anti-neoplastic agents. For example, multiple myeloma is a slowly dividing disease and is thus refractory to virtually all anti-neoplastic regimens. If these cells were forced to proliferate more rapidly their susceptibility profile would likely change. [0648]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a stimulator of B cell production in pathologies such as AIDS, chronic lymphocyte disorder and/or Common Variable Immunodificiency. [0649]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for generation and/or regeneration of lymphoid tissues following surgery, trauma or genetic defect. In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in the pretreatment of bone marrow samples prior to transplant. [0650]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a gene-based therapy for genetically inherited disorders resulting in immuno-incompetence/immunodeficiency such as observed among SCID patients. [0651]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of activating monocytes/macrophages to defend against parasitic diseases that effect monocytes such as Leishmania. [0652]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of regulating secreted cytokines that are elicited by polypeptides of the invention. [0653]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used in one or more of the applications decribed herein, as they may apply to veterinary medicine. [0654]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of blocking various aspects of immune responses to foreign agents or self. Examples of diseases or conditions in which blocking of certain aspects of immune responses may be desired include autoimmune disorders such as lupus, and arthritis, as well as immunoresponsiveness to skin allergies, inflammation, bowel disease, injury and diseases/disorders associated with pathogens. [0655]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for preventing the B cell proliferation and Ig secretion associated with autoimmune diseases such as idiopathic thrombocytopenic purpura, systemic lupus erythematosus and multiple sclerosis. [0656]
  • In another specific embodiment, polypeptides, antibodies, polynucleotides and/or agonists or antagonists of the present fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention invention are used as a inhibitor of B and/or T cell migration in endothelial cells. This activity disrupts tissue architecture or cognate responses and is useful, for example in disrupting immune responses, and blocking sepsis. [0657]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for chronic hypergammaglobulinemia evident in such diseases as monoclonal gammopathy of undetermined significance (MGUS), Waldenstrom's disease, related idiopathic monoclonal gammopathies, and plasmacytomas. [0658]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed for instance to inhibit polypeptide chemotaxis and activation of macrophages and their precursors, and of neutrophils, basophils, B lymphocytes and some T-cell subsets, e.g., activated and CD8 cytotoxic T cells and natural killer cells, in certain autoimmune and chronic inflammatory and infective diseases. Examples of autoimmune diseases are described herein and include multiple sclerosis, and insulin-dependent diabetes. [0659]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to treat idiopathic hyper-eosinophilic syndrome by, for example, preventing eosinophil production and migration. [0660]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit complement mediated cell lysis. [0661]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to enhance or inhibit antibody dependent cellular cytotoxicity. [0662]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed for treating atherosclerosis, for example, by preventing monocyte infiltration in the artery wall. [0663]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be employed to treat adult respiratory distress syndrome (ARDS). [0664]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful for stimulating wound and tissue repair, stimulating angiogenesis, and/or stimulating the repair of vascular or lymphatic diseases or disorders. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to stimulate the regeneration of mucosal surfaces. [0665]
  • In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, treat, and/or prevent a disorder characterized by primary or acquired immunodeficiency, deficient serum immunoglobulin production, recurrent infections, and/or immune system dysfunction. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat or prevent infections of the joints, bones, skin, and/or parotid glands, blood-borne infections (e.g., sepsis, meningitis, septic arthritis, and/or osteomyelitis), autoimmune diseases (e.g., those disclosed herein), inflammatory disorders, and malignancies, and/or any disease or disorder or condition associated with these infections, diseases, disorders and/or malignancies) including, but not limited to, CVID, other primary immune deficiencies, HIV disease, CLL, recurrent bronchitis, sinusitis, otitis media, conjunctivitis, pneumonia, hepatitis, meningitis, herpes zoster (e.g., severe herpes zoster), and/or pneumocystis carnii. Other diseases and disorders that may be prevented, diagnosed, prognosed, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, HIV infection, HTLV-BLV infection, lymphopenia, phagocyte bactericidal dysfunction anemia, thrombocytopenia, and hemoglobinuria. [0666]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, and/or diagnose an individual having common variable immunodeficiency disease (“CVID”; also known as “acquired agammaglobulinemia” and “acquired hypogammaglobulinemia”) or a subset of this disease. [0667]
  • In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to diagnose, prognose, prevent, and/or treat cancers or neoplasms including immune cell or immune tissue-related cancers or neoplasms. Examples of cancers or neoplasms that may be prevented, diagnosed, or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, acute myelogenous leukemia, chronic myelogenous leukemia, Hodgkin's disease, non-Hodgkin's lymphoma, acute lymphocytic anemia (ALL) Chronic lymphocyte leukemia, plasmacytomas, multiple myeloma, Burkitt's lymphoma, EBV-transformed diseases, and/or diseases and disorders described in the section entitled “Hyperproliferative Disorders” elsewhere herein. [0668]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a therapy for decreasing cellular proliferation of Large B-cell Lymphomas. [0669]
  • In another specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used as a means of decreasing the involvement of B cells and Ig associated with Chronic Myelogenous Leukemia. [0670]
  • In specific embodiments, the compositions of the invention are used as an agent to boost immunoresponsiveness among B cell immunodeficient individuals, such as, for example, an individual who has undergone a partial or complete splenectomy. [0671]
  • Blood-Related Disorders [0672]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hemostatic (the stopping of bleeding) or thrombolytic (clot dissolving) activity. For example, by increasing hemostatic or thrombolytic activity, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used to treat or prevent blood coagulation diseases, disorders, and/or conditions (e.g., afibrinogenemia, factor deficiencies, hemophilia), blood platelet diseases, disorders, and/or conditions (e.g., thrombocytopenia), or wounds resulting from trauma, surgery, or other causes. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention that can decrease hemostatic or thrombolytic activity could be used to inhibit or dissolve clotting. These molecules could be important in the treatment or prevention of heart attacks (infarction), strokes, or scarring. [0673]
  • In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, diagnose, prognose, and/or treat thrombosis, arterial thrombosis, venous thrombosis, thromboembolism, pulmonary embolism, atherosclerosis, myocardial infarction, transient ischemic attack, unstable angina. In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for the prevention of occulsion of saphenous grafts, for reducing the risk of periprocedural thrombosis as might accompany angioplasty procedures, for reducing the risk of stroke in patients with atrial fibrillation including nonrheumatic atrial fibrillation, for reducing the risk of embolism associated with mechanical heart valves and or mitral valves disease. Other uses for the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, the prevention of occlusions in extrcorporeal devices (e.g., intravascular canulas, vascular access shunts in hemodialysis patients, hemodialysis machines, and cardiopulmonary bypass machines). [0674]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to prevent, diagnose, prognose, and/or treat diseases and disorders of the blood and/or blood forming organs associated with the tissue(s) in which the polypeptide of the invention is expressed. [0675]
  • The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to modulate hematopoietic activity (the formation of blood cells). For example, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to increase the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets. The ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of anemias and leukopenias described below. Alternatively, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to decrease the quantity of all or subsets of blood cells, such as, for example, erythrocytes, lymphocytes (B or T cells), myeloid cells (e.g., basophils, eosinophils, neutrophils, mast cells, macrophages) and platelets. The ability to decrease the quantity of blood cells or subsets of blood cells may be useful in the prevention, detection, diagnosis and/or treatment of leukocytoses, such as, for example eosinophilia. [0676]
  • The fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to prevent, treat, or diagnose blood dyscrasia. [0677]
  • Anemias are conditions in which the number of red blood cells or amount of hemoglobin (the protein that carries oxygen) in them is below normal. Anemia may be caused by excessive bleeding, decreased red blood cell production, or increased red blood cell destruction (hemolysis). The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias. Anemias that may be treated prevented or diagnosed by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include iron deficiency anemia, hypochromic anemia, microcytic anemia, chlorosis, hereditary siderob;astic anemia, idiopathic acquired sideroblastic anemia, red cell aplasia, megaloblastic anemia (e.g., pernicious anemia, (vitamin B12 deficiency) and folic acid deficiency anemia), aplastic anemia, hemolytic anemias (e.g., autoimmune helolytic anemia, microangiopathic hemolytic anemia, and paroxysmal nocturnal hemoglobinuria). The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with diseases including but not limited to, anemias associated with systemic lupus erythematosus, cancers, lymphomas, chronic renal disease, and enlarged spleens. The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias arising from drug treatments such as anemias associated with methyldopa, dapsone, and/or sulfadrugs. Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing anemias associated with abnormal red blood cell architecture including but not limited to, hereditary spherocytosis, hereditary elliptocytosis, glucose-6-phosphate dehydrogenase deficiency, and sickle cell anemia. [0678]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing hemoglobin abnormalities, (e.g., those associated with sickle cell anemia, hemoglobin C disease, hemoglobin S-C disease, and hemoglobin E disease). Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating thalassemias, including, but not limited to, major and minor forms of alpha-thalassemia and beta-thalassemia. [0679]
  • In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating bleeding disorders including, but not limited to, thrombocytopenia (e.g., idiopathic thrombocytopenic purpura, and thrombotic thrombocytopenic purpura), Von Willebrand's disease, hereditary platelet disorders (e.g., storage pool disease such as Chediak-Higashi and Hermansky-Pudlak syndromes, thromboxane A2 dysfunction, thromboasthenia, and Bernard-Soulier syndrome), hemolytic-uremic syndrome, hemophelias such as hemophelia A or Factor VII deficiency and Christmas disease or Factor IX deficiency, Hereditary Hemorhhagic Telangiectsia, also known as Rendu-Osler-Weber syndrome, allergic purpura (Henoch Schonlein purpura) and disseminated intravascular coagulation. [0680]
  • The effect of the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention on the clotting time of blood may be monitored using any of the clotting tests known in the art including, but not limited to, whole blood partial thromboplastin time (PTT), the activated partial thromboplastin time (aPTT), the activated clotting time (ACT), the recalcified activated clotting time, or the Lee-White Clotting time. [0681]
  • Several diseases and a variety of drugs can cause platelet dysfunction. Thus, in a specific embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating acquired platelet dysfunction such as platelet dysfunction accompanying kidney failure, leukemia, multiple myeloma, cirrhosis of the liver, and systemic lupus erythematosus as well as platelet dysfunction associated with drug treatments, including treatment with aspirin, ticlopidine, nonsteroidal anti-inflammatory drugs (used for arthritis, pain, and sprains), and penicillin in high doses. [0682]
  • In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders characterized by or associated with increased or decreased numbers of white blood cells. Leukopenia occurs when the number of white blood cells decreases below normal. Leukopenias include, but are not limited to, neutropenia and lymphocytopenia. An increase in the number of white blood cells compared to normal is known as leukocytosis. The body generates increased numbers of white blood cells during infection. Thus, leukocytosis may simply be a normal physiological parameter that reflects infection. Alternatively, leukocytosis may be an indicator of injury or other disease such as cancer. Leokocytoses, include but are not limited to, eosinophilia, and accumulations of macrophages. In specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukopenia. In other specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukocytosis. [0683]
  • Leukopenia may be a generalized decreased in all types of white blood cells, or may be a specific depletion of particular types of white blood cells. Thus, in specific embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating decreases in neutrophil numbers, known as neutropenia. Neutropenias that may be diagnosed, prognosed, prevented, and/or treated by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, infantile genetic agranulocytosis, familial neutropenia, cyclic neutropenia, neutropenias resulting from or associated with dietary deficiencies (e.g., vitamin B 12 deficiency or folic acid deficiency), neutropenias resulting from or associated with drug treatments (e.g., antibiotic regimens such as penicillin treatment, sulfonamide treatment, anticoagulant treatment, anticonvulsant drugs, anti-thyroid drugs, and cancer chemotherapy), and neutropenias resulting from increased neutrophil destruction that may occur in association with some bacterial or viral infections, allergic disorders, autoimmune diseases, conditions in which an individual has an enlarged spleen (e.g., Felty syndrome, malaria and sarcoidosis), and some drug treatment regimens. [0684]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating lymphocytopenias (decreased numbers of B and/or T lymphocytes), including, but not limited to, lymphocytopenias resulting from or associated with stress, drug treatments (e.g., drug treatment with corticosteroids, cancer chemotherapies, and/or radiation therapies), AIDS infection and/or other diseases such as, for example, cancer, rheumatoid arthritis, systemic lupus erythematosus, chronic infections, some viral infections and/or hereditary disorders (e.g., DiGeorge syndrome, Wiskott-Aldrich Syndome, severe combined immunodeficiency, ataxia telangiectsia). [0685]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with macrophage numbers and/or macrophage function including, but not limited to, Gaucher's disease, Niemann-Pick disease, Letterer-Siwe disease and Hand-Schuller-Christian disease. [0686]
  • In another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders associated with eosinophil numbers and/or eosinophil function including, but not limited to, idiopathic hypereosinophilic syndrome, eosinophilia-myalgia syndrome, and Hand-Schuller-Christian disease. [0687]
  • In yet another embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating leukemias and lymphomas including, but not limited to, acute lymphocytic (lymphpblastic) leukemia (ALL), acute myeloid (myelocytic, myelogenous, myeloblastic, or myelomonocytic) leukemia, chronic lymphocytic leukemia (e.g., B cell leukemias, T cell leukemias, Sezary syndrome, and Hairy cell leukenia), chronic myelocytic (myeloid, myelogenous, or granulocytic) leukemia, Hodgkin's lymphoma, non-hodgkin's lymphoma, Burkitt's lymphoma, and mycosis fungoides. [0688]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in diagnosing, prognosing, preventing, and/or treating diseases and disorders of plasma cells including, but not limited to, plasma cell dyscrasias, monoclonal gammaopathies, monoclonal gammopathies of undetermined significance, multiple myeloma, macroglobulinemia, Waldenstrom's macroglobulinemia, cryoglobulinemia, and Raynaud's phenomenon. [0689]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating, preventing, and/or diagnosing myeloproliferative disorders, including but not limited to, polycythemia vera, relative polycythemia, secondary polycythemia, myelofibrosis, acute myelofibrosis, agnogenic myelod metaplasia, thrombocythemia, (including both primary and seconday thrombocythemia) and chronic myelocytic leukemia. [0690]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a treatment prior to surgery, to increase blood cell production. [0691]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to enhance the migration, phagocytosis, superoxide production, antibody dependent cellular cytotoxicity of neutrophils, eosionophils and macrophages. [0692]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to stem cells pheresis. In another specific embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase the number of stem cells in circulation prior to platelet pheresis. [0693]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as an agent to increase cytokine production. [0694]
  • In other embodiments, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in preventing, diagnosing, and/or treating primary hematopoietic disorders. [0695]
  • Hyperproliferative Disorders [0696]
  • In certain embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect hyperproliferative disorders, including neoplasms. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit the proliferation of the disorder through direct or indirect interactions. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may proliferate other cells which can inhibit the hyperproliferative disorder. [0697]
  • For example, by increasing an immune response, particularly increasing antigenic qualities of the hyperproliferative disorder or by proliferating, differentiating, or mobilizing T-cells, hyperproliferative disorders can be treated. This immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, decreasing an immune response may also be a method of treating hyperproliferative disorders, such as a chemotherapeutic agent. [0698]
  • Examples of hyperproliferative disorders that can be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to neoplasms located in the: colon, abdomen, bone, breast, digestive system, liver, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract. [0699]
  • Similarly, other hyperproliferative disorders can also be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to: Acute Childhood Lymphoblastic Leukemia, Acute Lymphoblastic Leukemia, Acute Lymphocytic Leukemia, Acute Myeloid Leukemia, Adrenocortical Carcinoma, Adult (Primary) Hepatocellular Cancer, Adult (Primary) Liver Cancer, Adult Acute Lymphocytic Leukemia, Adult Acute Myeloid Leukemia, Adult Hodgkin's Disease, Adult Hodgkin's Lymphoma, Adult Lymphocytic Leukemia, Adult Non-Hodgkin's Lymphoma, Adult Primary Liver Cancer, Adult Soft Tissue Sarcoma, AIDS-Related Lymphoma, AIDS-Related Malignancies, Anal Cancer, Astrocytoma, Bile Duct Cancer, Bladder Cancer, Bone Cancer, Brain Stem Glioma, Brain Tumors, Breast Cancer, Cancer of the Renal Pelvis and Ureter, Central Nervous System (Primary) Lymphoma, Central Nervous System Lymphoma, Cerebellar Astrocytoma, Cerebral Astrocytoma, Cervical Cancer, Childhood (Primary) Hepatocellular Cancer, Childhood (Primary) Liver Cancer, Childhood Acute Lymphoblastic Leukemia, Childhood Acute Myeloid Leukemia, Childhood Brain Stem Glioma, Childhood Cerebellar Astrocytoma, Childhood Cerebral Astrocytoma, Childhood Extracranial Germ Cell Tumors, Childhood Hodgkin's Disease, Childhood Hodgkin's Lymphoma, Childhood Hypothalamic and Visual Pathway Glioma, Childhood Lymphoblastic Leukemia, Childhood Medulloblastoma, Childhood Non-Hodgkin's Lymphoma, Childhood Pineal and Supratentorial Primitive Neuroectodermal Tumors, Childhood Primary Liver Cancer, Childhood Rhabdomyosarcoma, Childhood Soft Tissue Sarcoma, Childhood Visual Pathway and Hypothalamic Glioma, Chronic Lymphocytic Leukemia, Chronic Myelogenous Leukemia, Colon Cancer, Cutaneous T-Cell Lymphoma, Endocrine Pancreas Islet Cell Carcinoma, Endometrial Cancer, Ependymoma, Epithelial Cancer, Esophageal Cancer, Ewing's Sarcoma and Related Tumors, Exocrine Pancreatic Cancer, Extracranial Germ Cell Tumor, Extragonadal Germ Cell Tumor, Extrahepatic Bile Duct Cancer, Eye Cancer, Female Breast Cancer, Gaucher's Disease, Gallbladder Cancer, Gastric Cancer, Gastrointestinal Carcinoid Tumor, Gastrointestinal Tumors, Germ Cell Tumors, Gestational Trophoblastic Tumor, Hairy Cell Leukemia, Head and Neck Cancer, Hepatocellular Cancer, Hodgkin's Disease, Hodgkin's Lymphoma, Hypergammaglobulinemia, Hypopharyngeal Cancer, Intestinal Cancers, Intraocular Melanoma, Islet Cell Carcinoma, Islet Cell Pancreatic Cancer, Kaposi's Sarcoma, Kidney Cancer, Laryngeal Cancer, Lip and Oral Cavity Cancer, Liver Cancer, Lung Cancer, Lymphoproliferative Disorders, Macroglobulinemia, Male Breast Cancer, Malignant Mesothelioma, Malignant Thymoma, Medulloblastoma, Melanoma, Mesothelioma, Metastatic Occult Primary Squamous Neck Cancer, Metastatic Primary Squamous Neck Cancer, Metastatic Squamous Neck Cancer, Multiple Myeloma, Multiple Myeloma/Plasma Cell Neoplasm, Myelodysplastic Syndrome, Myelogenous Leukemia, Myeloid Leukemia, Myeloproliferative Disorders, Nasal Cavity and Paranasal Sinus Cancer, Nasopharyngeal Cancer, Neuroblastoma, Non-Hodgkin's Lymphoma During Pregnancy, Nonmelanoma Skin Cancer, Non-Small Cell Lung Cancer, Occult Primary Metastatic Squamous Neck Cancer, Oropharyngeal Cancer, Osteo-/Malignant Fibrous Sarcoma, Osteosarcoma/Malignant Fibrous Histiocytoma, Osteosarcoma/Malignant Fibrous Histiocytoma of Bone, Ovarian Epithelial Cancer, Ovarian Germ Cell Tumor, Ovarian Low Malignant Potential Tumor, Pancreatic Cancer, Paraproteinemias, Purpura, Parathyroid Cancer, Penile Cancer, Pheochromocytoma, Pituitary Tumor, Plasma Cell Neoplasm/Multiple Myeloma, Primary Central Nervous System Lymphoma, Primary Liver Cancer, Prostate Cancer, Rectal Cancer, Renal Cell Cancer, Renal Pelvis and Ureter Cancer, Retinoblastoma, Rhabdomyosarcoma, Salivary Gland Cancer, Sarcoidosis Sarcomas, Sezary Syndrome, Skin Cancer, Small Cell Lung Cancer, Small Intestine Cancer, Soft Tissue Sarcoma, Squamous Neck Cancer, Stomach Cancer, Supratentorial Primitive Neuroectodermal and Pineal Tumors, T-Cell Lymphoma, Testicular Cancer, Thymoma, Thyroid Cancer, Transitional Cell Cancer of the Renal Pelvis and Ureter, Transitional Renal Pelvis and Ureter Cancer, Trophoblastic Tumors, Ureter and Renal Pelvis Cell Cancer, Urethral Cancer, Uterine Cancer, Uterine Sarcoma, Vaginal Cancer, Visual Pathway and Hypothalamic Glioma, Vulvar Cancer, Waldenstrom's Macroglobulinemia, Wilms' Tumor, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above. [0700]
  • In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to diagnose, prognose, prevent, and/or treat premalignant conditions and to prevent progression to a neoplastic or malignant state, including but not limited to those disorders described above. Such uses are indicated in conditions known or suspected of preceding progression to neoplasia or cancer, in particular, where non-neoplastic cell growth consisting of hyperplasia, metaplasia, or most particularly, dysplasia has occurred (for review of such abnormal growth conditions, see Robbins and Angell, 1976, Basic Pathology, 2d Ed., W. B. Saunders Co., Philadelphia, pp. 68-79.) [0701]
  • Hyperplasia is a form of controlled cell proliferation, involving an increase in cell number in a tissue or organ, without significant alteration in structure or function. Hyperplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, angiofollicular mediastinal lymph node hyperplasia, angiolymphoid hyperplasia with eosinophilia, a typical melanocytic hyperplasia, basal cell hyperplasia, benign giant lymph node hyperplasia, cementum hyperplasia, congenital adrenal hyperplasia, congenital sebaceous hyperplasia, cystic hyperplasia, cystic hyperplasia of the breast, denture hyperplasia, ductal hyperplasia, endometrial hyperplasia, fibromuscular hyperplasia, focal epithelial hyperplasia, gingival hyperplasia, inflammatory fibrous hyperplasia, inflammatory papillary hyperplasia, intravascular papillary endothelial hyperplasia, nodular hyperplasia of prostate, nodular regenerative hyperplasia, pseudoepitheliomatous hyperplasia, senile sebaceous hyperplasia, and verrucous hyperplasia. [0702]
  • Metaplasia is a form of controlled cell growth in which one type of adult or fully differentiated cell substitutes for another type of adult cell. Metaplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, agnogenic myeloid metaplasia, apocrine metaplasia, a typical metaplasia, autoparenchymatous metaplasia, connective tissue metaplasia, epithelial metaplasia, intestinal metaplasia, metaplastic anemia, metaplastic ossification, metaplastic polyps, myeloid metaplasia, primary myeloid metaplasia, secondary myeloid metaplasia, squamous metaplasia, squamous metaplasia of amnion, and symptomatic myeloid metaplasia. [0703]
  • Dysplasia is frequently a forerunner of cancer, and is found mainly in the epithelia; it is the most disorderly form of non-neoplastic cell growth, involving a loss in individual cell uniformity and in the architectural orientation of cells. Dysplastic cells often have abnormally large, deeply stained nuclei, and exhibit pleomorphism. Dysplasia characteristically occurs where there exists chronic irritation or inflammation. Dysplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, anhidrotic ectodermal dysplasia, anterofacial dysplasia, asphyxiating thoracic dysplasia, atriodigital dysplasia, bronchopulmonary dysplasia, cerebral dysplasia, cervical dysplasia, chondroectodermal dysplasia, cleidocranial dysplasia, congenital ectodermal dysplasia, craniodiaphysial dysplasia, craniocarpotarsal dysplasia, craniometaphysial dysplasia, dentin dysplasia, diaphysial dysplasia, ectodermal dysplasia, enamel dysplasia, encephalo-ophthalmic dysplasia, dysplasia epiphysialis hemimelia, dysplasia epiphysialis multiplex, dysplasia epiphysialis punctata, epithelial dysplasia, faciodigitogenital dysplasia, familial fibrous dysplasia of jaws, familial white folded dysplasia, fibromuscular dysplasia, fibrous dysplasia of bone, florid osseous dysplasia, hereditary renal-retinal dysplasia, hidrotic ectodermal dysplasia, hypohidrotic ectodermal dysplasia, lymphopenic thymic dysplasia, mammary dysplasia, mandibulofacial dysplasia, metaphysial dysplasia, Mondini dysplasia, monostotic fibrous dysplasia, mucoepithelial dysplasia, multiple epiphysial dysplasia, oculoauriculovertebral dysplasia, oculodentodigital dysplasia, oculovertebral dysplasia, odontogenic dysplasia, ophthalmomandibulomelic dysplasia, periapical cemental dysplasia, polyostotic fibrous dysplasia, pseudoachondroplastic spondyloepiphysial dysplasia, retinal dysplasia, septo-optic dysplasia, spondyloepiphysial dysplasia, and ventriculoradial dysplasia. [0704]
  • Additional pre-neoplastic disorders which can be diagnosed, prognosed, prevented, and/or treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, benign dysproliferative disorders (e.g., benign tumors, fibrocystic conditions, tissue hypertrophy, intestinal polyps, colon polyps, and esophageal dysplasia), leukoplakia, keratoses, Bowen's disease, Farmer's Skin, solar cheilitis, and solar keratosis. [0705]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to diagnose and/or prognose disorders associated with the tissue(s) in which the polypeptide of the invention is expressed. [0706]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat cancers and neoplasms, including, but not limited to, those described herein. In a further preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention conjugated to a toxin or a radioactive isotope, as described herein, may be used to treat acute myelogenous leukemia. [0707]
  • Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may affect apoptosis, and therefore, would be useful in treating a number of diseases associated with increased cell survival or the inhibition of apoptosis. For example, diseases associated with increased cell survival or the inhibition of apoptosis that could be diagnosed, prognosed, prevented, and/or treated by polynucleotides, polypeptides, and/or agonists or antagonists of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. [0708]
  • In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metastasis of cancers, in particular those listed above. [0709]
  • Additional diseases or conditions associated with increased cell survival that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, emangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma. [0710]
  • Diseases associated with increased apoptosis that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include AIDS; neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, retinitis pigmentosa, cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia. [0711]
  • Hyperproliferative diseases and/or disorders that could be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, neoplasms located in the liver, abdomen, bone, breast, digestive system, pancreas, peritoneum, endocrine glands (adrenal, parathyroid, pituitary, testicles, ovary, thymus, thyroid), eye, head and neck, nervous system (central and peripheral), lymphatic system, pelvis, skin, soft tissue, spleen, thorax, and urogenital tract. [0712]
  • Similarly, other hyperproliferative disorders can also be diagnosed, prognosed, prevented, and/or treated by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of such hyperproliferative disorders include, but are not limited to: hypergammaglobulinemia, lymphoproliferative disorders, paraproteinemias, purpura, sarcoidosis, Sezary Syndrome, Waldenstron's macroglobulinemia, Gaucher's Disease, histiocytosis, and any other hyperproliferative disease, besides neoplasia, located in an organ system listed above. [0713]
  • Another preferred embodiment utilizes polynucleotides encoding albumin fusion proteins of the invention to inhibit aberrant cellular division, by gene therapy using the present invention, and/or protein fusions or fragments thereof. [0714]
  • Thus, the present invention provides a method for treating cell proliferative disorders by inserting into an abnormally proliferating cell a polynucleotide encoding an albumin fusion protein of the present invention, wherein said polynucleotide represses said expression. [0715]
  • Another embodiment of the present invention provides a method of treating cell-proliferative disorders in individuals comprising administration of one or more active gene copies of the present invention to an abnormally proliferating cell or cells. In a preferred embodiment, polynucleotides of the present invention is a DNA construct comprising a recombinant expression vector effective in expressing a DNA sequence encoding said polynucleotides. In another preferred embodiment of the present invention, the DNA construct encoding the fusion protein of the present invention is inserted into cells to be treated utilizing a retrovirus, or more preferably an adenoviral vector (See G J. Nabel, et. al., PNAS 1999 96: 324-326, which is hereby incorporated by reference). In a most preferred embodiment, the viral vector is defective and will not transform non-proliferating cells, only proliferating cells. Moreover, in a preferred embodiment, the polynucleotides of the present invention inserted into proliferating cells either alone, or in combination with or fused to other polynucleotides, can then be modulated via an external stimulus (i.e. magnetic, specific small molecule, chemical, or drug administration, etc.), which acts upon the promoter upstream of said polynucleotides to induce expression of the encoded protein product. As such the beneficial therapeutic affect of the present invention may be expressly modulated (i.e. to increase, decrease, or inhibit expression of the present invention) based upon said external stimulus. [0716]
  • Polynucleotides of the present invention may be useful in repressing expression of oncogenic genes or antigens. By “repressing expression of the oncogenic genes” is intended the suppression of the transcription of the gene, the degradation of the gene transcript (pre-message RNA), the inhibition of splicing, the destruction of the messenger RNA, the prevention of the post-translational modifications of the protein, the destruction of the protein, or the inhibition of the normal function of the protein. [0717]
  • For local administration to abnormally proliferating cells, polynucleotides of the present invention may be administered by any method known to those of skill in the art including, but not limited to transfection, electroporation, microinjection of cells, or in vehicles such as liposomes, lipofectin, or as naked polynucleotides, or any other method described throughout the specification. The polynucleotide of the present invention may be delivered by known gene delivery systems such as, but not limited to, retroviral vectors (Gilboa, J. Virology 44:845 (1982); Hocke, Nature 320:275 (1986); Wilson, et al., Proc. Natl. Acad. Sci. U.S.A. 85:3014), vaccinia virus system (Chakrabarty et al., Mol. Cell Biol. 5:3403 (1985) or other efficient DNA delivery systems (Yates et al., Nature 313:812 (1985)) known to those skilled in the art. These references are exemplary only and are hereby incorporated by reference. In order to specifically deliver or transfect cells which are abnormally proliferating and spare non-dividing cells, it is preferable to utilize a retrovirus, or adenoviral (as described in the art and elsewhere herein) delivery system known to those of skill in the art. Since host DNA replication is required for retroviral DNA to integrate and the retrovirus will be unable to self replicate due to the lack of the retrovirus genes needed for its life cycle. Utilizing such a retroviral delivery system for polynucleotides of the present invention will target said gene and constructs to abnormally proliferating cells and will spare the non-dividing normal cells. [0718]
  • The polynucleotides of the present invention may be delivered directly to cell proliferative disorder/disease sites in internal organs, body cavities and the like by use of imaging devices used to guide an injecting needle directly to the disease site. The polynucleotides of the present invention may also be administered to disease sites at the time of surgical intervention. [0719]
  • By “cell proliferative disease” is meant any human or animal disease or disorder, affecting any one or any combination of organs, cavities, or body parts, which is characterized by single or multiple local abnormal proliferations of cells, groups of cells, or tissues, whether benign or malignant. [0720]
  • Any amount of the polynucleotides of the present invention may be administered as long as it has a biologically inhibiting effect on the proliferation of the treated cells. Moreover, it is possible to administer more than one of the polynucleotide of the present invention simultaneously to the same site. By “biologically inhibiting” is meant partial or total growth inhibition as well as decreases in the rate of proliferation or growth of the cells. The biologically inhibitory dose may be determined by assessing the effects of the polynucleotides of the present invention on target malignant or abnormally proliferating cell growth in tissue culture, tumor growth in animals and cell cultures, or any other method known to one of ordinary skill in the art. [0721]
  • Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention of the present invention are useful in inhibiting the angiogenesis of proliferative cells or tissues, either alone, as a protein fusion, or in combination with other polypeptides directly or indirectly, as described elsewhere herein. In a most preferred embodiment, said anti-angiogenesis effect may be achieved indirectly, for example, through the inhibition of hematopoietic, tumor-specific cells, such as tumor-associated macrophages (See Joseph I B, et al. J Natl Cancer Inst, 90(21):1648-53 (1998), which is hereby incorporated by reference). [0722]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in inhibiting proliferative cells or tissues through the induction of apoptosis. These fusion protieins and/or polynucleotides may act either directly, or indirectly to induce apoptosis of proliferative cells and tissues, for example in the activation of a death-domain receptor, such as tumor necrosis factor (TNF) receptor-1, CD95 (Fas/APO-1), TNF-receptor-related apoptosis-mediated protein (TRAMP) and TNF-related apoptosis-inducing ligand (TRAIL) receptor-1 and -2 (See Schulze-Osthoff K, et.al., Eur J Biochem 254(3):439-59 (1998), which is hereby incorporated by reference). Moreover, in another preferred embodiment of the present invention, these fusion proteins and/or polynucleotides may induce apoptosis through other mechanisms, such as in the activation of other proteins which will activate apoptosis, or through stimulating the expression of these proteins, either alone or in combination with small molecule drugs or adjuviants, such as apoptonin, galectins, thioredoxins, anti-inflammatory proteins (See for example, Mutat Res 400(1-2):447-55 (1998), Med Hypotheses. 50(5):423-33 (1998), Chem Biol Interact. Apr 24;111-112:23-34 (1998), J Mol Med. 76(6):402-12 (1998), Int J Tissue React;20(1):3-15 (1998), which are all hereby incorporated by reference). [0723]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are useful in inhibiting the metastasis of proliferative cells or tissues. Inhibition may occur as a direct result of administering these albumin fusion proteins and/or polynucleotides, or indirectly, such as activating the expression of proteins known to inhibit metastasis, for [0724] example alpha 4 integrins, (See, e.g., Curr Top Microbiol Immunol 1998;231:125-41, which is hereby incorporated by reference). Such thereapeutic affects of the present invention may be achieved either alone, or in combination with small molecule drugs or adjuvants.
  • In another embodiment, the invention provides a method of delivering compositions containing the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to targeted cells expressing the a polypeptide bound by, that binds to, or associates with an albumin fuison protein of the invention. Albumin fusion proteins of the invention may be associated with with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalent interactions. [0725]
  • Albumin fusion proteins of the invention are useful in enhancing the immunogenicity and/or antigenicity of proliferating cells or tissues, either directly, such as would occur if the albumin fusion proteins of the invention ‘vaccinated’ the immune response to respond to proliferative antigens and immunogens, or indirectly, such as in activating the expression of proteins known to enhance the immune response (e.g. chemokines), to said antigens and immunogens. [0726]
  • Renal Disorders [0727]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose disorders of the renal system. Renal disorders which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, kidney failure, nephritis, blood vessel disorders of kidney, metabolic and congenital kidney disorders, urinary disorders of the kidney, autoimmune disorders, sclerosis and necrosis, electrolyte imbalance, and kidney cancers. [0728]
  • Kidney diseases which can be diagnosed, prognosed, prevented, and/or treated with compositions of the invention include, but are not limited to, acute kidney failure, chronic kidney failure, atheroembolic renal failure, end-stage renal disease, inflammatory diseases of the kidney (e.g., acute glomerulonephritis, postinfectious glomerulonephritis, rapidly progressive glomerulonephritis, nephrotic syndrome, membranous glomerulonephritis, familial nephrotic syndrome, membranoproliferative glomerulonephritis I and II, mesangial proliferative glomerulonephritis, chronic glomerulonephritis, acute tubulointerstitial nephritis, chronic tubulointerstitial nephritis, acute post-streptococcal glomerulonephritis (PSGN), pyelonephritis, lupus nephritis, chronic nephritis, interstitial nephritis, and post-streptococcal glomerulonephritis), blood vessel disorders of the kidneys (e.g., kidney infarction, atheroembolic kidney disease, cortical necrosis, malignant nephrosclerosis, renal vein thrombosis, renal underperfusion, renal retinopathy, renal ischemia-reperfusion, renal artery embolism, and renal artery stenosis), and kidney disorders resulting form urinary tract disease (e.g., pyelonephritis, hydronephrosis, urolithiasis (renal lithiasis, nephrolithiasis), reflux nephropathy, urinary tract infections, urinary retention, and acute or chronic unilateral obstructive uropathy.) [0729]
  • In addition, compositions of the invention can be used to diagnose, prognose, prevent, and/or treat metabolic and congenital disorders of the kidney (e.g., uremia, renal amyloidosis, renal osteodystrophy, renal tubular acidosis, renal glycosuria, nephrogenic diabetes insipidus, cystinuria, Fanconi's syndrome, renal fibrocystic osteosis (renal rickets), Hartnup disease, Bartter's syndrome, Liddle's syndrome, polycystic kidney disease, medullary cystic disease, medullary sponge kidney, Alport's syndrome, nail-patella syndrome, congenital nephrotic syndrome, CRUSH syndrome, horseshoe kidney, diabetic nephropathy, nephrogenic diabetes insipidus, analgesic nephropathy, kidney stones, and membranous nephropathy), and autoimmune disorders of the kidney (e.g., systemic lupus erythematosus (SLE), Goodpasture syndrome, IgA nephropathy, and IgM mesangial proliferative glomerulonephritis). [0730]
  • Compositions of the invention can also be used to diagnose, prognose, prevent, and/or treat sclerotic or necrotic disorders of the kidney (e.g., glomerulosclerosis, diabetic nephropathy, focal segmental glomerulosclerosis (FSGS), necrotizing glomerulonephritis, and renal papillary necrosis), cancers of the kidney (e.g., nephroma, hypemephroma, nephroblastoma, renal cell cancer, transitional cell cancer, renal adenocarcinoma, squamous cell cancer, and Wilm's tumor), and electrolyte imbalances (e.g., nephrocalcinosis, pyuria, edema, hydronephritis, proteinuria, hyponatremia, hypernatremia, hypokalemia, hyperkalemia, hypocalcemia, hypercalcemia, hypophosphatemia, and hyperphosphatemia). [0731]
  • Compositions of the invention may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art. Compositions of the invention may be administered as part of a Therapeutic, described in more detail below. Methods of delivering polynucleotides of the invention are described in more detail herein. [0732]
  • Cardiovascular Disorders [0733]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose cardiovascular disorders, including, but not limited to, peripheral artery disease, such as limb ischemia. [0734]
  • Cardiovascular disorders include, but are not limited to, cardiovascular abnormalities, such as arterio-arterial fistula, arterioyenous fistula, cerebral arterioyenous malformations, congenital heart defects, pulmonary atresia, and Scimitar Syndrome. Congenital heart defects include, but are not limited to, aortic coarctation, cor triatriatum, coronary vessel anomalies, crisscross heart, dextrocardia, patent ductus arteriosus, Ebstein's anomaly, Eisenmenger complex, hypoplastic left heart syndrome, levocardia, tetralogy of fallot, transposition of great vessels, double outlet right ventricle, tricuspid atresia, persistent truncus arteriosus, and heart septal defects, such as aortopulmonary septal defect, endocardial cushion defects, Lutembacher's Syndrome, trilogy of Fallot, ventricular heart septal defects. [0735]
  • Cardiovascular disorders also include, but are not limited to, heart disease, such as arrhythmias, carcinoid heart disease, high cardiac output, low cardiac output, cardiac tamponade, endocarditis (including bacterial), heart aneurysm, cardiac arrest, congestive heart failure, congestive cardiomyopathy, paroxysmal dyspnea, cardiac edema, heart hypertrophy, congestive cardiomyopathy, left ventricular hypertrophy, right ventricular hypertrophy, post-infarction heart rupture, ventricular septal rupture, heart valve diseases, myocardial diseases, myocardial ischemia, pericardial effusion, pericarditis (including constrictive and tuberculous), pneumopericardium, postpericardiotomy syndrome, pulmonary heart disease, rheumatic heart disease, ventricular dysfunction, hyperemia, cardiovascular pregnancy complications, Scimitar Syndrome, cardiovascular syphilis, and cardiovascular tuberculosis. [0736]
  • Arrhythmias include, but are not limited to, sinus arrhythmia, atrial fibrillation, atrial flutter, bradycardia, extrasystole, Adams-Stokes Syndrome, bundle-branch block, sinoatrial block, long QT syndrome, parasystole, Lown-Ganong-Levine Syndrome, Mahaim-type pre-excitation syndrome, Wolff-Parkinson-White syndrome, sick sinus syndrome, tachycardias, and ventricular fibrillation. Tachycardias include paroxysmal tachycardia, supraventricular tachycardia, accelerated idioventricular rhythm, atrioventricular nodal reentry tachycardia, ectopic atrial tachycardia, ectopic junctional tachycardia, sinoatrial nodal reentry tachycardia, sinus tachycardia, Torsades de Pointes, and ventricular tachycardia. [0737]
  • Heart valve diseases include, but are not limited to, aortic valve insufficiency, aortic valve stenosis, hear murmurs, aortic valve prolapse, mitral valve prolapse, tricuspid valve prolapse, mitral valve insufficiency, mitral valve stenosis, pulmonary atresia, pulmonary valve insufficiency, pulmonary valve stenosis, tricuspid atresia, tricuspid valve insufficiency, and tricuspid valve stenosis. [0738]
  • Myocardial diseases include, but are not limited to, alcoholic cardiomyopathy, congestive cardiomyopathy, hypertrophic cardiomyopathy, aortic subvalvular stenosis, pulmonary subvalvular stenosis, restrictive cardiomyopathy, Chagas cardiomyopathy, endocardial fibroelastosis, endomyocardial fibrosis, Kearns Syndrome, myocardial reperfusion injury, and myocarditis. [0739]
  • Myocardial ischemias include, but are not limited to, coronary disease, such as angina pectoris, coronary aneurysm, coronary arteriosclerosis, coronary thrombosis, coronary vasospasm, myocardial infarction and myocardial stunning. [0740]
  • Cardiovascular diseases also include vascular diseases such as aneurysms, angiodysplasia, angiomatosis, bacillary angiomatosis, Hippel-Lindau Disease, Klippel-Trenaunay-Weber Syndrome, Sturge-Weber Syndrome, angioneurotic edema, aortic diseases, Takayasu's Arteritis, aortitis, Leriche's Syndrome, arterial occlusive diseases, arteritis, enarteritis, polyarteritis nodosa, cerebrovascular disorders, diabetic angiopathies, diabetic retinopathy, embolisms, thrombosis, erythromelalgia, hemorrhoids, hepatic veno-occlusive disease, hypertension, hypotension, ischemia, peripheral vascular diseases, phlebitis, pulmonary veno-occlusive disease, Raynaud's disease, CREST syndrome, retinal vein occlusion, Scimitar syndrome, superior vena cava syndrome, telangiectasia, atacia telangiectasia, hereditary hemorrhagic telangiectasia, varicocele, varicose veins, varicose ulcer, vasculitis, and venous insufficiency. [0741]
  • Aneurysms include, but are not limited to, dissecting aneurysms, false aneurysms, infected aneurysms, ruptured aneurysms, aortic aneurysms, cerebral aneurysms, coronary aneurysms, heart aneurysms, and iliac aneurysms. [0742]
  • Arterial occlusive diseases include, but are not limited to, arteriosclerosis, intermittent claudication, carotid stenosis, fibromuscular dysplasias, mesenteric vascular occlusion, Moyamoya disease, renal artery obstruction, retinal artery occlusion, and thromboangiitis obliterans. [0743]
  • Cerebrovascular disorders include, but are not limited to, carotid artery diseases, cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arterioyenous malformation, cerebral artery diseases, cerebral embolism and thrombosis, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, cerebral hemorrhage, epidural hematoma, subdural hematoma, subaraxhnoid hemorrhage, cerebral infarction, cerebral ischemia (including transient), subclavian steal syndrome, periventricular leukomalacia, vascular headache, cluster headache, migraine, and vertebrobasilar insufficiency. [0744]
  • Embolisms include, but are not limited to, air embolisms, amniotic fluid embolisms, cholesterol embolisms, blue toe syndrome, fat embolisms, pulmonary embolisms, and thromoboembolisms. Thrombosis include, but are not limited to, coronary thrombosis, hepatic vein thrombosis, retinal vein occlusion, carotid artery thrombosis, sinus thrombosis, Wallenberg's syndrome, and thrombophlebitis. [0745]
  • Ischemic disorders include, but are not limited to, cerebral ischemia, ischemic colitis, compartment syndromes, anterior compartment syndrome, myocardial ischemia, reperfusion injuries, and peripheral limb ischemia. Vasculitis includes, but is not limited to, aortitis, arteritis, Behcet's Syndrome, Churg-Strauss Syndrome, mucocutaneous lymph node syndrome, thromboangiitis obliterans, hypersensitivity vasculitis, Schoenlein-Henoch purpura, allergic cutaneous vasculitis, and Wegener's granulomatosis. [0746]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered using any method known in the art, including, but not limited to, direct needle injection at the delivery site, intravenous injection, topical administration, catheter infusion, biolistic injectors, particle accelerators, gelfoam sponge depots, other commercially available depot materials, osmotic pumps, oral or suppositorial solid pharmaceutical formulations, decanting or topical applications during surgery, aerosol delivery. Such methods are known in the art. Methods of delivering polynucleotides are described in more detail herein. [0747]
  • Respiratory Disorders [0748]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat, prevent, diagnose, and/or prognose diseases and/or disorders of the respiratory system. [0749]
  • Diseases and disorders of the respiratory system include, but are not limited to, nasal vestibulitis, nonallergic rhinitis (e.g., acute rhinitis, chronic rhinitis, atrophic rhinitis, vasomotor rhinitis), nasal polyps, and sinusitis, juvenile angiofibromas, cancer of the nose and juvenile papillomas, vocal cord polyps, nodules (singer's nodules), contact ulcers, vocal cord paralysis, laryngoceles, pharyngitis (e.g., viral and bacterial), tonsillitis, tonsillar cellulitis, parapharyngeal abscess, laryngitis, laryngoceles, and throat cancers (e.g., cancer of the nasopharynx, tonsil cancer, larynx cancer), lung cancer (e.g., squamous cell carcinoma, small cell (oat cell) carcinoma, large cell carcinoma, and adenocarcinoma), allergic disorders (eosinophilic pneumonia, hypersensitivity pneumonitis (e.g., extrinsic allergic alveolitis, allergic interstitial pneumonitis, organic dust pneumoconiosis, allergic bronchopulmonary aspergillosis, asthma, Wegener's granulomatosis (granulomatous vasculitis), Goodpasture's syndrome)), pneumonia (e.g., bacterial pneumonia (e.g., [0750] Streptococcus pneumoniae (pneumoncoccal pneumonia), Staphylococcus aureus (staphylococcal pneumonia), Gram-negative bacterial pneumonia (caused by, e.g., Klebsiella and Pseudomas spp.), Mycoplasma pneumoniae pneumonia, Hemophilus influenzae pneumonia, Legionella pneumophila (Legionnaires' disease), and Chlamydia psittaci (Psittacosis)), and viral pneumonia (e.g., influenza, chickenpox (varicella).
  • Additional diseases and disorders of the respiratory system include, but are not limited to bronchiolitis, polio (poliomyelitis), croup, respiratory syncytial viral infection, mumps, erythema infectiosum (fifth disease), roseola infantum, progressive rubella panencephalitis, german measles, and subacute sclerosing panencephalitis), fungal pneumonia (e.g., Histoplasmosis, Coccidioidomycosis, Blastomycosis, fungal infections in people with severely suppressed immune systems (e.g., cryptococcosis, caused by [0751] Cryptococcus neoformans; aspergillosis, caused by Aspergillus spp.; candidiasis, caused by Candida; and mucormycosis)), Pneumocystis carinii (pneumocystis pneumonia), a typical pneumonias (e.g., Mycoplasma and Chlamydia spp.), opportunistic infection pneumonia, nosocomial pneumonia, chemical pneumonitis, and aspiration pneumonia, pleural disorders (e.g., pleurisy, pleural effusion, and pneumothorax (e.g., simple spontaneous pneumothorax, complicated spontaneous pneumothorax, tension pneumothorax)), obstructive airway diseases (e.g., asthma, chronic obstructive pulmonary disease (COPD), emphysema, chronic or acute bronchitis), occupational lung diseases (e.g., silicosis, black lung (coal workers' pneumoconiosis), asbestosis, berylliosis, occupational asthsma, byssinosis, and benign pneumoconioses), Infiltrative Lung Disease (e.g., pulmonary fibrosis (e.g., fibrosing alveolitis, usual interstitial pneumonia), idiopathic pulmonary fibrosis, desquamative interstitial pneumonia, lymphoid interstitial pneumonia, histiocytosis X (e.g., Letterer-Siwe disease, Hand-Schüller-Christian disease, eosinophilic granuloma), idiopathic pulmonary hemosiderosis, sarcoidosis and pulmonary alveolar proteinosis), Acute respiratory distress syndrome (also called, e.g., adult respiratory distress syndrome), edema, pulmonary embolism, bronchitis (e.g., viral, bacterial), bronchiectasis, atelectasis, lung abscess (caused by, e.g., Staphylococcus aureus or Legionella pneumophila), and cystic fibrosis.
  • Anti-Angiogenesis Activity [0752]
  • The naturally occurring balance between endogenous stimulators and inhibitors of angiogenesis is one in which inhibitory influences predominate. Rastinejad et al., [0753] Cell 56:345-355 (1989). In those rare instances in which neovascularization occurs under normal physiological conditions, such as wound healing, organ regeneration, embryonic development, and female reproductive processes, angiogenesis is stringently regulated and spatially and temporally delimited. Under conditions of pathological angiogenesis such as that characterizing solid tumor growth, these regulatory controls fail. Unregulated angiogenesis becomes pathologic and sustains progression of many neoplastic and non-neoplastic diseases. A number of serious diseases are dominated by abnormal neovascularization including solid tumor growth and metastases, arthritis, some types of eye disorders, and psoriasis. See, e.g., reviews by Moses et al., Biotech. 9:630-634 (1991); Folkman et al., N. Engl. J. Med., 333:1757-1763 (1995); Auerbach et al., J. Microvasc. Res. 29:401-411 (1985); Folkman, Advances in Cancer Research, eds. Klein and Weinhouse, Academic Press, New York, pp. 175-203 (1985); Patz, Am. J. Opthalmol. 94:715-743 (1982); and Folkman et al., Science 221:719-725 (1983). In a number of pathological conditions, the process of angiogenesis contributes to the disease state. For example, significant data have accumulated which suggest that the growth of solid tumors is dependent on angiogenesis. Folkman and Klagsbrun, Science 235:442-447 (1987).
  • The present invention provides for treatment of diseases or disorders associated with neovascularization by administration of fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Malignant and metastatic conditions which can be treated with the polynucleotides and polypeptides, or agonists or antagonists of the invention include, but are not limited to, malignancies, solid tumors, and cancers described herein and otherwise known in the art (for a review of such disorders, see Fishman et al., Medicine, 2d Ed., J. B. Lippincott Co., Philadelphia (1985)).Thus, the present invention provides a method of treating an angiogenesis-related disease and/or disorder, comprising administering to an individual in need thereof a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention. For example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a variety of additional methods in order to therapeutically treat a cancer or tumor. Cancers which may be treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to solid tumors, including prostate, lung, breast, ovarian, stomach, pancreas, larynx, esophagus, testes, liver, parotid, biliary tract, colon, rectum, cervix, uterus, endometrium, kidney, bladder, thyroid cancer; primary tumors and metastases; melanomas; glioblastoma; Kaposi's sarcoma; leiomyosarcoma; non-small cell lung cancer; colorectal cancer; advanced malignancies; and blood born tumors such as leukemias. For example, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be delivered topically, in order to treat cancers such as skin cancer, head and neck tumors, breast tumors, and Kaposi's sarcoma. [0754]
  • Within yet other aspects, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized to treat superficial forms of bladder cancer by, for example, intravesical administration. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be delivered directly into the tumor, or near the tumor site, via injection or a catheter. Of course, as the artisan of ordinary skill will appreciate, the appropriate mode of administration will vary according to the cancer to be treated. Other modes of delivery are discussed herein. [0755]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful in treating other disorders, besides cancers, which involve angiogenesis. These disorders include, but are not limited to: benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; artheroscleric plaques; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions; myocardial angiogenesis; coronary collaterals; cerebral collaterals; arterioyenous malformations; ischemic limb angiogenesis; Osler-Webber Syndrome; plaque neovascularization; telangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia; wound granulation; Crohn's disease; and atherosclerosis. [0756]
  • For example, within one aspect of the present invention methods are provided for treating hypertrophic scars and keloids, comprising the step of administering albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to a hypertrophic scar or keloid. [0757]
  • Within one embodiment of the present invention fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are directly injected into a hypertrophic scar or keloid, in order to prevent the progression of these lesions. This therapy is of particular value in the prophylactic treatment of conditions which are known to result in the development of hypertrophic scars and keloids (e.g., bums), and is preferably initiated after the proliferative phase has had time to progress (approximately 14 days after the initial injury), but before hypertrophic scar or keloid development. As noted above, the present invention also provides methods for treating neovascular diseases of the eye, including for example, corneal neovascularization, neovascular glaucoma, proliferative diabetic retinopathy, retrolental fibroplasia and macular degeneration. [0758]
  • Moreover, Ocular disorders associated with neovascularization which can be treated with the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to: neovascular glaucoma, diabetic retinopathy, retinoblastoma, retrolental fibroplasia, uveitis, retinopathy of prematurity macular degeneration, corneal graft neovascularization, as well as other eye inflammatory diseases, ocular tumors and diseases associated with choroidal or iris neovascularization. See, e.g., reviews by Waltman et al., [0759] Am. J. Ophthal. 85:704-710 (1978) and Gartner et al., Surv. Ophthal. 22:291-312 (1978).
  • Thus, within one aspect of the present invention methods are provided for treating neovascular diseases of the eye such as corneal neovascularization (including corneal graft neovascularization), comprising the step of administering to a patient a therapeutically effective amount of a compound (e.g., fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention) to the cornea, such that the formation of blood vessels is inhibited. Briefly, the cornea is a tissue which normally lacks blood vessels. In certain pathological conditions however, capillaries may extend into the cornea from the pericorneal vascular plexus of the limbus. When the cornea becomes vascularized, it also becomes clouded, resulting in a decline in the patient's visual acuity. Visual loss may become complete if the cornea completely opacitates. A wide variety of disorders can result in corneal neovascularization, including for example, corneal infections (e.g., trachoma, herpes simplex keratitis, leishmaniasis and onchocerciasis), immunological processes (e.g., graft rejection and Stevens-Johnson's syndrome), alkali bums, trauma, inflammation (of any cause), toxic and nutritional deficiency states, and as a complication of wearing contact lenses. [0760]
  • Within particularly preferred embodiments of the invention, may be prepared for topical administration in saline (combined with any of the preservatives and antimicrobial agents commonly used in ocular preparations), and administered in eyedrop form. The solution or suspension may be prepared in its pure form and administered several times daily. Alternatively, anti-angiogenic compositions, prepared as described above, may also be administered directly to the cornea. Within preferred embodiments, the anti-angiogenic composition is prepared with a muco-adhesive polymer which binds to cornea. Within further embodiments, the anti-angiogenic factors or anti-angiogenic compositions may be utilized as an adjunct to conventional steroid therapy. Topical therapy may also be useful prophylactically in corneal lesions which are known to have a high probability of inducing an angiogenic response (such as chemical burns). In these instances the treatment, likely in combination with steroids, may be instituted immediately to help prevent subsequent complications. [0761]
  • Within other embodiments, the compounds described above may be injected directly into the corneal stroma by an ophthalmologist under microscopic guidance. The preferred site of injection may vary with the morphology of the individual lesion, but the goal of the administration would be to place the composition at the advancing front of the vasculature (i.e., interspersed between the blood vessels and the normal cornea). In most cases this would involve perilimbic corneal injection to “protect” the cornea from the advancing blood vessels. This method may also be utilized shortly after a corneal insult in order to prophylactically prevent corneal neovascularization. In this situation the material could be injected in the perilimbic cornea interspersed between the corneal lesion and its undesired potential limbic blood supply. Such methods may also be utilized in a similar fashion to prevent capillary invasion of transplanted corneas. In a sustained-release form injections might only be required 2-3 times per year. A steroid could also be added to the injection solution to reduce inflammation resulting from the injection itself. [0762]
  • Within another aspect of the present invention, methods are provided for treating neovascular glaucoma, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eye, such that the formation of blood vessels is inhibited. In one embodiment, the compound may be administered topically to the eye in order to treat early forms of neovascular glaucoma. Within other embodiments, the compound may be implanted by injection into the region of the anterior chamber angle. Within other embodiments, the compound may also be placed in any location such that the compound is continuously released into the aqueous humor. Within another aspect of the present invention, methods are provided for treating proliferative diabetic retinopathy, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eyes, such that the formation of blood vessels is inhibited. [0763]
  • Within particularly preferred embodiments of the invention, proliferative diabetic retinopathy may be treated by injection into the aqueous humor or the vitreous, in order to increase the local concentration of the polynucleotide, polypeptide, antagonist and/or agonist in the retina. Preferably, this treatment should be initiated prior to the acquisition of severe disease requiring photocoagulation. [0764]
  • Within another aspect of the present invention, methods are provided for treating retrolental fibroplasia, comprising the step of administering to a patient a therapeutically effective amount of an albumin fusion protein of the invention and/or polynucleotides encoding an albumin fusion protein of the invention to the eye, such that the formation of blood vessels is inhibited. The compound may be administered topically, via intravitreous injection and/or via intraocular implants. [0765]
  • Additionally, disorders which can be treated with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, hemangioma, arthritis, psoriasis, angiofibroma, atherosclerotic plaques, delayed wound healing, granulations, hemophilic joints, hypertrophic scars, nonunion fractures, Osler-Weber syndrome, pyogenic granuloma, scleroderma, trachoma, and vascular adhesions. [0766]
  • Moreover, disorders and/or states, which can be treated, prevented, diagnosed, and/or prognosed with the the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention of the invention include, but are not limited to, solid tumors, blood born tumors such as leukemias, tumor metastasis, Kaposi's sarcoma, benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas, rheumatoid arthritis, psoriasis, ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, and uvietis, delayed wound healing, endometriosis, vascluogenesis, granulations, hypertrophic scars (keloids), nonunion fractures, scleroderma, trachoma, vascular adhesions, myocardial angiogenesis, coronary collaterals, cerebral collaterals, arterioyenous malformations, ischemic limb angiogenesis, Osler-Webber Syndrome, plaque neovascularization, telangiectasia, hemophiliac joints, angiofibroma fibromuscular dysplasia, wound granulation, Crohn's disease, atherosclerosis, birth control agent by preventing vascularization required for embryo implantation controlling menstruation, diseases that have angiogenesis as a pathologic consequence such as cat scratch disease (Rochele minalia quintosa), ulcers ([0767] Helicobacter pylori), Bartonellosis and bacillary angiomatosis.
  • In one aspect of the birth control method, an amount of the compound sufficient to block embryo implantation is administered before or after intercourse and fertilization have occurred, thus providing an effective method of birth control, possibly a “morning after” method. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be used in controlling menstruation or administered as either a peritoneal lavage fluid or for peritoneal implantation in the treatment of endometriosis. [0768]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be incorporated into surgical sutures in order to prevent stitch granulomas. [0769]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be utilized in a wide variety of surgical procedures. For example, within one aspect of the present invention a compositions (in the form of, for example, a spray or film) may be utilized to coat or spray an area prior to removal of a tumor, in order to isolate normal surrounding tissues from malignant tissue, and/or to prevent the spread of disease to surrounding tissues. Within other aspects of the present invention, compositions (e.g., in the form of a spray) may be delivered via endoscopic procedures in order to coat tumors, or inhibit angiogenesis in a desired locale. Within yet other aspects of the present invention, surgical meshes which have been coated with anti-angiogenic compositions of the present invention may be utilized in any procedure wherein a surgical mesh might be utilized. For example, within one embodiment of the invention a surgical mesh laden with an anti-angiogenic composition may be utilized during abdominal cancer resection surgery (e.g., subsequent to colon resection) in order to provide support to the structure, and to release an amount of the anti-angiogenic factor. [0770]
  • Within further aspects of the present invention, methods are provided for treating tumor excision sites, comprising administering albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention to the resection margins of a tumor subsequent to excision, such that the local recurrence of cancer and the formation of new blood vessels at the site is inhibited. Within one embodiment of the invention, the anti-angiogenic compound is administered directly to the tumor excision site (e.g., applied by swabbing, brushing or otherwise coating the resection margins of the tumor with the anti-angiogenic compound). Alternatively, the anti-angiogenic compounds may be incorporated into known surgical pastes prior to administration. Within particularly preferred embodiments of the invention, the anti-angiogenic compounds are applied after hepatic resections for malignancy, and after neurosurgical operations. [0771]
  • Within one aspect of the present invention, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be administered to the resection margin of a wide variety of tumors, including for example, breast, colon, brain and hepatic tumors. For example, within one embodiment of the invention, anti-angiogenic compounds may be administered to the site of a neurological tumor subsequent to excision, such that the formation of new blood vessels at the site are inhibited. [0772]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be administered along with other anti-angiogenic factors. Representative examples of other anti-angiogenic factors include: Anti-Invasive Factor, retinoic acid and derivatives thereof, paclitaxel, Suramin, Tissue Inhibitor of Metalloproteinase-1, Tissue Inhibitor of Metalloproteinase-2, Plasminogen Activator Inhibitor-1, Plasminogen Activator Inhibitor-2, and various forms of the lighter “d group” transition metals. [0773]
  • Lighter “d group” transition metals include, for example, vanadium, molybdenum, tungsten, titanium, niobium, and tantalum species. Such transition metal species may form transition metal complexes. Suitable complexes of the above-mentioned transition metal species include oxo transition metal complexes. [0774]
  • Representative examples of vanadium complexes include oxo vanadium complexes such as vanadate and vanadyl complexes. Suitable vanadate complexes include metavanadate and orthovanadate complexes such as, for example, ammonium metavanadate, sodium metavanadate, and sodium orthovanadate. Suitable vanadyl complexes include, for example, vanadyl acetylacetonate and vanadyl sulfate including vanadyl sulfate hydrates such as vanadyl sulfate mono- and trihydrates. [0775]
  • Representative examples of tungsten and molybdenum complexes also include oxo complexes. Suitable oxo tungsten complexes include tungstate and tungsten oxide complexes. Suitable tungstate complexes include ammonium tungstate, calcium tungstate, sodium tungstate dihydrate, and tungstic acid. Suitable tungsten oxides include tungsten (IV) oxide and tungsten (VI) oxide. Suitable oxo molybdenum complexes include molybdate, molybdenum oxide, and molybdenyl complexes. Suitable molybdate complexes include ammonium molybdate and its hydrates, sodium molybdate and its hydrates, and potassium molybdate and its hydrates. Suitable molybdenum oxides include molybdenum (VI) oxide, molybdenum (VI) oxide, and molybdic acid. Suitable molybdenyl complexes include, for example, molybdenyl acetylacetonate. Other suitable tungsten and molybdenum complexes include hydroxo derivatives derived from, for example, glycerol, tartaric acid, and sugars. [0776]
  • A wide variety of other anti-angiogenic factors may also be utilized within the context of the present invention. Representative examples include [0777] platelet factor 4; protamine sulphate; sulphated chitin derivatives (prepared from queen crab shells), (Murata et al., Cancer Res. 51:22-26, 1991); Sulphated Polysaccharide Peptidoglycan Complex (SP-PG) (the function of this compound may be enhanced by the presence of steroids such as estrogen, and tamoxifen citrate); Staurosporine; modulators of matrix metabolism, including for example, proline analogs, cishydroxyproline, d,L-3,4-dehydroproline, Thiaproline, alpha,alpha-dipyridyl, aminopropionitrile fumarate; 4-propyl-5-(4-pyridinyl)-2(3H)-oxazolone; Methotrexate; Mitoxantrone; Heparin; Interferons; 2 Macroglobulin-serum; ChIMP-3 (Pavloff et al., J. Bio. Chem. 267:17321-17326, (1992)); Chymostatin (Tomkinson et al., Biochem J. 286:475-480, (1992)); Cyclodextrin Tetradecasulfate; Eponemycin; Camptothecin; Fumagillin (Ingber et al., Nature 348:555-557, 1990); Gold Sodium Thiomalate (“GST”; Matsubara and Ziff, J. Clin. Invest. 79:1440-1446, (1987)); anticollagenase-serum; alpha2-antiplasmin (Holmes et al., J. Biol. Chem. 262(4):1659-1664, (1987)); Bisantrene (National Cancer Institute); Lobenzarit disodium (N-(2)-carboxyphenyl-4-chloroanthronilic acid disodium or “CCA”; Takeuchi et al., Agents Actions 36:312-316, (1992)); Thalidomide; Angostatic steroid; AGM-1470; carboxynaminolmidazole; and metalloproteinase inhibitors such as BB94.
  • Diseases at the Cellular Level [0778]
  • Diseases associated with increased cell survival or the inhibition of apoptosis that could be treated, prevented, diagnosed, and/or prognosed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include cancers (such as follicular lymphomas, carcinomas with p53 mutations, and hormone-dependent tumors, including, but not limited to colon cancer, cardiac tumors, pancreatic cancer, melanoma, retinoblastoma, glioblastoma, lung cancer, intestinal cancer, testicular cancer, stomach cancer, neuroblastoma, myxoma, myoma, lymphoma, endothelioma, osteoblastoma, osteoclastoma, osteosarcoma, chondrosarcoma, adenoma, breast cancer, prostate cancer, Kaposi's sarcoma and ovarian cancer); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) and viral infections (such as herpes viruses, pox viruses and adenoviruses), inflammation, graft v. host disease, acute graft rejection, and chronic graft rejection. [0779]
  • In preferred embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to inhibit growth, progression, and/or metasis of cancers, in particular those listed above. [0780]
  • Additional diseases or conditions associated with increased cell survival that could be treated or detected by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but are not limited to, progression, and/or metastases of malignancies and related disorders such as leukemia (including acute leukemias (e.g., acute lymphocytic leukemia, acute myelocytic leukemia (including myeloblastic, promyelocytic, myelomonocytic, monocytic, and erythroleukemia)) and chronic leukemias (e.g., chronic myelocytic (granulocytic) leukemia and chronic lymphocytic leukemia)), polycythemia vera, lymphomas (e.g., Hodgkin's disease and non-Hodgkin's disease), multiple myeloma, Waldenstrom's macroglobulinemia, heavy chain disease, and solid tumors including, but not limited to, sarcomas and carcinomas such as fibrosarcoma, myxosarcoma, liposarcoma, chondrosarcoma, osteogenic sarcoma, chordoma, angiosarcoma, endotheliosarcoma, lymphangiosarcoma, lymphangioendotheliosarcoma, synovioma, mesothelioma, Ewing's tumor, leiomyosarcoma, rhabdomyosarcoma, colon carcinoma, pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squamous cell carcinoma, basal cell carcinoma, adenocarcinoma, sweat gland carcinoma, sebaceous gland carcinoma, papillary carcinoma, papillary adenocarcinomas, cystadenocarcinoma, medullary carcinoma, bronchogenic carcinoma, renal cell carcinoma, hepatoma, bile duct carcinoma, choriocarcinoma, seminoma, embryonal carcinoma, Wilm's tumor, cervical cancer, testicular tumor, lung carcinoma, small cell lung carcinoma, bladder carcinoma, epithelial carcinoma, glioma, astrocytoma, medulloblastoma, craniopharyngioma, ependymoma, pinealoma, hemangioblastoma, acoustic neuroma, oligodendroglioma, menangioma, melanoma, neuroblastoma, and retinoblastoma. [0781]
  • Diseases associated with increased apoptosis that could be treated, prevented, diagnosed, and/or prognesed using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, include, but are not limited to, AIDS; neurodegenerative disorders (such as Alzheimer's disease, Parkinson's disease, Amyotrophic lateral sclerosis, Retinitis pigmentosa, Cerebellar degeneration and brain tumor or prior associated disease); autoimmune disorders (such as, multiple sclerosis, Sjogren's syndrome, Hashimoto's thyroiditis, biliary cirrhosis, Behcet's disease, Crohn's disease, polymyositis, systemic lupus erythematosus and immune-related glomerulonephritis and rheumatoid arthritis) myelodysplastic syndromes (such as aplastic anemia), graft v. host disease, ischemic injury (such as that caused by myocardial infarction, stroke and reperfusion injury), liver injury (e.g., hepatitis related liver injury, ischemia/reperfusion injury, cholestosis (bile duct injury) and liver cancer); toxin-induced liver disease (such as that caused by alcohol), septic shock, cachexia and anorexia. [0782]
  • Wound Healing and Epithelial Cell Proliferation [0783]
  • In accordance with yet a further aspect of the present invention, there is provided a process for utilizing fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, for therapeutic purposes, for example, to stimulate epithelial cell proliferation and basal keratinocytes for the purpose of wound healing, and to stimulate hair follicle production and healing of dermal wounds. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be clinically useful in stimulating wound healing including surgical wounds, excisional wounds, deep wounds involving damage of the dermis and epidermis, eye tissue wounds, dental tissue wounds, oral cavity wounds, diabetic ulcers, dermal ulcers, cubitus ulcers, arterial ulcers, venous stasis ulcers, burns resulting from heat exposure or chemicals, and other abnormal wound healing conditions such as uremia, malnutrition, vitamin deficiencies and complications associated with systemic treatment with steroids, radiation therapy and antineoplastic drugs and antimetabolites. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to promote dermal reestablishment subsequent to dermal loss [0784]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to increase the adherence of skin grafts to a wound bed and to stimulate re-epithelialization from the wound bed. The following are types of grafts that fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to increase adherence to a wound bed: autografts, artificial skin, allografts, autodermic graft, autoepdermic grafts, avacular grafts, Blair-Brown grafts, bone graft, brephoplastic grafts, cutis graft, delayed graft, dermic graft, epidermic graft, fascia graft, full thickness graft, heterologous graft, xenograft, homologous graft, hyperplastic graft, lamellar graft, mesh graft, mucosal graft, Ollier-Thiersch graft, omenpal graft, patch graft, pedicle graft, penetrating graft, split skin graft, thick split graft. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to promote skin strength and to improve the appearance of aged skin. [0785]
  • It is believed that fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, will also produce changes in hepatocyte proliferation, and epithelial cell proliferation in the lung, breast, pancreas, stomach, small intestine, and large intestine. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could promote proliferation of epithelial cells such as sebocytes, hair follicles, hepatocytes, type II pneumocytes, mucin-producing goblet cells, and other epithelial cells and their progenitors contained within the skin, lung, liver, and gastrointestinal tract. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may promote proliferation of endothelial cells, keratinocytes, and basal keratinocytes. [0786]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could also be used to reduce the side effects of gut toxicity that result from radiation, chemotherapy treatments or viral infections. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may have a cytoprotective effect on the small intestine mucosa. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may also stimulate healing of mucositis (mouth ulcers) that result from chemotherapy and viral infections. [0787]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could further be used in full regeneration of skin in full and partial thickness skin defects, including bums, (i.e., repopulation of hair follicles, sweat glands, and sebaceous glands), treatment of other skin defects such as psoriasis. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to treat epidermolysis bullosa, a defect in adherence of the epidermis to the underlying dermis which results in frequent, open and painful blisters by accelerating reepithelialization of these lesions. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could also be used to treat gastric and doudenal ulcers and help heal by scar formation of the mucosal lining and regeneration of glandular mucosa and duodenal mucosal lining more rapidly. Inflammatory bowel diseases, such as Crohn's disease and ulcerative colitis, are diseases which result in destruction of the mucosal surface of the small or large intestine, respectively. Thus, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to promote the resurfacing of the mucosal surface to aid more rapid healing and to prevent progression of inflammatory bowel disease. Treatment with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, is expected to have a significant effect on the production of mucus throughout the gastrointestinal tract and could be used to protect the intestinal mucosa from injurious substances that are ingested or following surgery. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to treat diseases associate with the under expression. [0788]
  • Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to prevent and heal damage to the lungs due to various pathological states. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, which could stimulate proliferation and differentiation and promote the repair of alveoli and brochiolar epithelium to prevent or treat acute or chronic lung damage. For example, emphysema, which results in the progressive loss of aveoli, and inhalation injuries, i.e., resulting from smoke inhalation and bums, that cause necrosis of the bronchiolar epithelium and alveoli could be effectively treated using polynucleotides or polypeptides, agonists or antagonists of the present invention. Also fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to stimulate the proliferation of and differentiation of type II pneumocytes, which may help treat or prevent disease such as hyaline membrane diseases, such as infant respiratory distress syndrome and bronchopulmonary displasia, in premature infants. [0789]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could stimulate the proliferation and differentiation of hepatocytes and, thus, could be used to alleviate or treat liver diseases and pathologies such as fulminant liver failure caused by cirrhosis, liver damage caused by viral hepatitis and toxic substances (i.e., acetaminophen, carbon tetraholoride and other hepatotoxins known in the art). [0790]
  • In addition, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used treat or prevent the onset of diabetes mellitus. In patients with newly diagnosed Types I and II diabetes, where some islet cell function remains, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used to maintain the islet function so as to alleviate, delay or prevent permanent manifestation of the disease. Also, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, could be used as an auxiliary in islet cell transplantation to improve or promote islet cell function. [0791]
  • Neural Activity and Neurological Diseases [0792]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for the diagnosis and/or treatment of diseases, disorders, damage or injury of the brain and/or nervous system. Nervous system disorders that can be treated with the compositions of the invention (e.g., fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention), include, but are not limited to, nervous system injuries, and diseases or disorders which result in either a disconnection of axons, a diminution or degeneration of neurons, or demyelination. Nervous system lesions which may be treated in a patient (including human and non-human mammalian patients) according to the methods of the invention, include but are not limited to, the following lesions of either the central (including spinal cord, brain) or peripheral nervous systems: (1) ischemic lesions, in which a lack of oxygen in a portion of the nervous system results in neuronal injury or death, including cerebral infarction or ischemia, or spinal cord infarction or ischemia; (2) traumatic lesions, including lesions caused by physical injury or associated with surgery, for example, lesions which sever a portion of the nervous system, or compression injuries; (3) malignant lesions, in which a portion of the nervous system is destroyed or injured by malignant tissue which is either a nervous system associated malignancy or a malignancy derived from non-nervous system tissue; (4) infectious lesions, in which a portion of the nervous system is destroyed or injured as a result of infection, for example, by an abscess or associated with infection by human immunodeficiency virus, herpes zoster, or herpes simplex virus or with Lyme disease, tuberculosis, or syphilis; (5) degenerative lesions, in which a portion of the nervous system is destroyed or injured as a result of a degenerative process including but not limited to, degeneration associated with Parkinson's disease, Alzheimer's disease, Huntington's chorea, or amyotrophic lateral sclerosis (ALS); (6) lesions associated with nutritional diseases or disorders, in which a portion of the nervous system is destroyed or injured by a nutritional disorder or disorder of metabolism including, but not limited to, vitamin B12 deficiency, folic acid deficiency, Wernicke disease, tobacco-alcohol amblyopia, Marchiafava-Bignami disease (primary degeneration of the corpus callosum), and alcoholic cerebellar degeneration; (7) neurological lesions associated with systemic diseases including, but not limited to, diabetes (diabetic neuropathy, Bell's palsy), systemic lupus erythematosus, carcinoma, or sarcoidosis; (8) lesions caused by toxic substances including alcohol, lead, or particular neurotoxins; and (9) demyelinated lesions in which a portion of the nervous system is destroyed or injured by a demyelinating disease including, but not limited to, multiple sclerosis, human immunodeficiency virus-associated myelopathy, transverse myelopathy or various etiologies, progressive multifocal leukoencephalopathy, and central pontine myelinolysis. [0793]
  • In one embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to protect neural cells from the damaging effects of hypoxia. In a further preferred embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to protect neural cells from the damaging effects of cerebral hypoxia. According to this embodiment, the compositions of the invention are used to treat or prevent neural cell injury associated with cerebral hypoxia. In one non-exclusive aspect of this embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, are used to treat or prevent neural cell injury associated with cerebral ischemia. In another non-exclusive aspect of this embodiment, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with cerebral infarction. [0794]
  • In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with a stroke. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent cerebral neural cell injury associated with a stroke. [0795]
  • In another preferred embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent neural cell injury associated with a heart attack. In a specific embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat or prevent cerebral neural cell injury associated with a heart attack. [0796]
  • The compositions of the invention which are useful for treating or preventing a nervous system disorder may be selected by testing for biological activity in promoting the survival or differentiation of neurons. For example, and not by way of limitation, compositions of the invention which elicit any of the following effects may be useful according to the invention: (1) increased survival time of neurons in culture either in the presence or absence of hypoxia or hypoxic conditions; (2) increased sprouting of neurons in culture or in vivo; (3) increased production of a neuron-associated molecule in culture or in vivo, e.g., choline acetyltransferase or acetylcholinesterase with respect to motor neurons; or (4) decreased symptoms of neuron dysfunction in vivo. Such effects may be measured by any method known in the art. In preferred, non-limiting embodiments, increased survival of neurons may routinely be measured using a method set forth herein or otherwise known in the art, such as, for example, in Zhang et al., [0797] Proc Natl Acad Sci USA 97:3637-42 (2000) or in Arakawa et al., J. Neurosci., 10:3507-15 (1990); increased sprouting of neurons may be detected by methods known in the art, such as, for example, the methods set forth in Pestronk et al., Exp. Neurol., 70:65-82 (1980), or Brown et al., Ann. Rev. Neurosci., 4:17-42 (1981); increased production of neuron-associated molecules may be measured by bioassay, enzymatic assay, antibody binding, Northern blot assay, etc., using techniques known in the art and depending on the molecule to be measured; and motor neuron dysfunction may be measured by assessing the physical manifestation of motor neuron disorder, e.g., weakness, motor neuron conduction velocity, or functional disability.
  • In specific embodiments, motor neuron disorders that may be treated according to the invention include, but are not limited to, disorders such as infarction, infection, exposure to toxin, trauma, surgical damage, degenerative disease or malignancy that may affect motor neurons as well as other components of the nervous system, as well as disorders that selectively affect neurons such as amyotrophic lateral sclerosis, and including, but not limited to, progressive spinal muscular atrophy, progressive bulbar palsy, primary lateral sclerosis, infantile and juvenile muscular atrophy, progressive bulbar paralysis of childhood (Fazio-Londe syndrome), poliomyelitis and the post polio syndrome, and Hereditary Motorsensory Neuropathy (Charcot-Marie-Tooth Disease). [0798]
  • Further, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may play a role in neuronal survival; synapse formation; conductance; neural differentiation, etc. Thus, compositions of the invention (including fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention) may be used to diagnose and/or treat or prevent diseases or disorders associated with these roles, including, but not limited to, learning and/or cognition disorders. The compositions of the invention may also be useful in the treatment or prevention of neurodegenerative disease states and/or behavioural disorders. Such neurodegenerative disease states and/or behavioral disorders include, but are not limited to, Alzheimer's Disease, Parkinson's Disease, Huntington's Disease, Tourette Syndrome, schizophrenia, mania, dementia, paranoia, obsessive compulsive disorder, panic disorder, learning disabilities, ALS, psychoses, autism, and altered behaviors, including disorders in feeding, sleep patterns, balance, and perception. In addition, compositions of the invention may also play a role in the treatment, prevention and/or detection of developmental disorders associated with the developing embryo, or sexually-linked disorders. [0799]
  • Additionally, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be useful in protecting neural cells from diseases, damage, disorders, or injury, associated with cerebrovascular disorders including, but not limited to, carotid artery diseases (e.g., carotid artery thrombosis, carotid stenosis, or Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arterioyenous malformations, cerebral artery diseases, cerebral embolism and thrombosis (e.g., carotid artery thrombosis, sinus thrombosis, or Wallenberg's Syndrome), cerebral hemorrhage (e.g., epidural or subdural hematoma, or subarachnoid hemorrhage), cerebral infarction, cerebral ischemia (e.g., transient cerebral ischemia, Subclavian Steal Syndrome, or vertebrobasilar insufficiency), vascular dementia (e.g., multi-infarct), leukomalacia, periventricular, and vascular headache (e.g., cluster headache or migraines). [0800]
  • In accordance with yet a further aspect of the present invention, there is provided a process for utilizing fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, for therapeutic purposes, for example, to stimulate neurological cell proliferation and/or differentiation. Therefore, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used to treat and/or detect neurologic diseases. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used as a marker or detector of a particular nervous system disease or disorder. [0801]
  • Examples of neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, brain diseases, such as metabolic brain diseases which includes phenylketonuria such as maternal phenylketonuria, pyruvate carboxylase deficiency, pyruvate dehydrogenase complex deficiency, Wernicke's Encephalopathy, brain edema, brain neoplasms such as cerebellar neoplasms which include infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothalamic neoplasms, supratentorial neoplasms, canavan disease, cerebellar diseases such as cerebellar ataxia which include spinocerebellar degeneration such as ataxia telangiectasia, cerebellar dyssynergia, Friederich's Ataxia, Machado-Joseph Disease, olivopontocerebellar atrophy, cerebellar neoplasms such as infratentorial neoplasms, diffuse cerebral sclerosis such as encephalitis periaxialis, globoid cell leukodystrophy, metachromatic leukodystrophy and subacute sclerosing panencephalitis. [0802]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include cerebrovascular disorders (such as carotid artery diseases which include carotid artery thrombosis, carotid stenosis and Moyamoya Disease), cerebral amyloid angiopathy, cerebral aneurysm, cerebral anoxia, cerebral arteriosclerosis, cerebral arterioyenous malformations, cerebral artery diseases, cerebral embolism and thrombosis such as carotid artery thrombosis, sinus thrombosis and Wallenberg's Syndrome, cerebral hemorrhage such as epidural hematoma, subdural hematoma and subarachnoid hemorrhage, cerebral infarction, cerebral ischemia such as transient cerebral ischemia, Subclavian Steal Syndrome and vertebrobasilar insufficiency, vascular dementia such as multi-infarct dementia, periventricular leukomalacia, vascular headache such as cluster headache and migraine. [0803]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include dementia such as AIDS Dementia Complex, presenile dementia such as Alzheimer's Disease and Creutzfeldt-Jakob Syndrome, senile dementia such as Alzheimer's Disease and progressive supranuclear palsy, vascular dementia such as multi-infarct dementia, encephalitis which include encephalitis periaxialis, viral encephalitis such as epidemic encephalitis, Japanese Encephalitis, St. Louis Encephalitis, tick-borne encephalitis and West Nile Fever, acute disseminated encephalomyelitis, meningoencephalitis such as uveomeningoencephalitic syndrome, Postencephalitic Parkinson Disease and subacute sclerosing panencephalitis, encephalomalacia such as periventricular leukomalacia, epilepsy such as generalized epilepsy which includes infantile spasms, absence epilepsy, myoclonic epilepsy which includes MERRF Syndrome, tonic-clonic epilepsy, partial epilepsy such as complex partial epilepsy, frontal lobe epilepsy and temporal lobe epilepsy, post-traumatic epilepsy, status epilepticus such as Epilepsia Partialis Continua, and Hallervorden-Spatz Syndrome. [0804]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include hydrocephalus such as Dandy-Walker Syndrome and normal pressure hydrocephalus, hypothalamic diseases such as hypothalamic neoplasms, cerebral malaria, narcolepsy which includes cataplexy, bulbar poliomyelitis, cerebri pseudotumor, Rett Syndrome, Reye's Syndrome, thalamic diseases, cerebral toxoplasmosis, intracranial tuberculoma and Zellweger Syndrome, central nervous system infections such as AIDS Dementia Complex, Brain Abscess, subdural empyema, encephalomyelitis such as Equine Encephalomyelitis, Venezuelan Equine Encephalomyelitis, Necrotizing Hemorrhagic Encephalomyelitis, Visna, and cerebral malaria. [0805]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include meningitis such as arachnoiditis, aseptic meningtitis such as viral meningtitis which includes lymphocytic choriomeningitis, Bacterial meningtitis which includes Haemophilus Meningtitis, Listeria Meningtitis, Meningococcal Meningtitis such as Waterhouse-Friderichsen Syndrome, Pneumococcal Meningtitis and meningeal tuberculosis, fungal meningitis such as Cryptococcal Meningtitis, subdural effusion, meningoencephalitis such as uvemeningoencephalitic syndrome, myelitis such as transverse myelitis, neurosyphilis such as tabes dorsalis, poliomyelitis which includes bulbar poliomyelitis and postpoliomyelitis syndrome, prion diseases (such as Creutzfeldt-Jakob Syndrome, Bovine Spongiform Encephalopathy, Gerstmann-Straussler Syndrome, Kuru, Scrapie), and cerebral toxoplasmosis. [0806]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include central nervous system neoplasms such as brain neoplasms that include cerebellar neoplasms such as infratentorial neoplasms, cerebral ventricle neoplasms such as choroid plexus neoplasms, hypothalamic neoplasms and supratentorial neoplasms, meningeal neoplasms, spinal cord neoplasms which include epidural neoplasms, demyelinating diseases such as Canavan Diseases, diffuse cerebral sceloris which includes adrenoleukodystrophy, encephalitis periaxialis, globoid cell leukodystrophy, diffuse cerebral sclerosis such as metachromatic leukodystrophy, allergic encephalomyelitis, necrotizing hemorrhagic encephalomyelitis, progressive multifocal leukoencephalopathy, multiple sclerosis, central pontine myelinolysis, transverse myelitis, neuromyelitis optica, Scrapie, Swayback, Chronic Fatigue Syndrome, Visna, High Pressure Nervous Syndrome, Meningism, spinal cord diseases such as amyotonia congenita, amyotrophic lateral sclerosis, spinal muscular atrophy such as Werdnig-Hoffmann Disease, spinal cord compression, spinal cord neoplasms such as epidural neoplasms, syringomyelia, Tabes Dorsalis, Stiff-Man Syndrome, mental retardation such as Angelman Syndrome, Cri-du-Chat Syndrome, De Lange's Syndrome, Down Syndrome, Gangliosidoses such as gangliosidoses G(M1), Sandhoff Disease, Tay-Sachs Disease, Hartnup Disease, homocystinuria, Laurence-Moon-Biedl Syndrome, Lesch-Nyhan Syndrome, Maple Syrup Urine Disease, mucolipidosis such as fucosidosis, neuronal ceroid-lipofuscinosis, oculocerebrorenal syndrome, phenylketonuria such as maternal phenylketonuria, Prader-Willi Syndrome, Rett Syndrome, Rubinstein-Taybi Syndrome, Tuberous Sclerosis, WAGR Syndrome, nervous system abnormalities such as holoprosencephaly, neural tube defects such as anencephaly which includes hydrangencephaly, Arnold-Chairi Deformity, encephalocele, meningocele, meningomyelocele, spinal dysraphism such as spina bifida cystica and spina bifida occulta. [0807]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include hereditary motor and sensory neuropathies which include Charcot-Marie Disease, Hereditary optic atrophy, Refsum's Disease, hereditary spastic paraplegia, Werdnig-Hoffiann Disease, Hereditary Sensory and Autonomic Neuropathies such as Congenital Analgesia and Familial Dysautonomia, Neurologic manifestations (such as agnosia that include Gerstmann's Syndrome, Amnesia such as retrograde amnesia, apraxia, neurogenic bladder, cataplexy, communicative disorders such as hearing disorders that includes deafness, partial hearing loss, loudness recruitment and tinnitus, language disorders such as aphasia which include agraphia, anomia, broca aphasia, and Wernicke Aphasia, Dyslexia such as Acquired Dyslexia, language development disorders, speech disorders such as aphasia which includes anomia, broca aphasia and Wernicke Aphasia, articulation disorders, communicative disorders such as speech disorders which include dysarthria, echolalia, mutism and stuttering, voice disorders such as aphonia and hoarseness, decerebrate state, delirium, fasciculation, hallucinations, meningism, movement disorders such as angelman syndrome, ataxia, athetosis, chorea, dystonia, hypokinesia, muscle hypotonia, myoclonus, tic, torticollis and tremor, muscle hypertonia such as muscle rigidity such as stiff-man syndrome, muscle spasticity, paralysis such as facial paralysis which includes Herpes Zoster Oticus, Gastroparesis, Hemiplegia, ophthalmoplegia such as diplopia, Duane's Syndrome, Horner's Syndrome, Chronic progressive external ophthalmoplegia such as Kearns Syndrome, Bulbar Paralysis, Tropical Spastic Paraparesis, Paraplegia such as Brown-Sequard Syndrome, quadriplegia, respiratory paralysis and vocal cord paralysis, paresis, phantom limb, taste disorders such as ageusia and dysgeusia, vision disorders such as amblyopia, blindness, color vision defects, diplopia, hemianopsia, scotoma and subnormal vision, sleep disorders such as hypersomnia which includes Kleine-Levin Syndrome, insomnia, and somnambulism, spasm such as trismus, unconsciousness such as coma, persistent vegetative state and syncope and vertigo, neuromuscular diseases such as amyotonia congenita, amyotrophic lateral sclerosis, Lambert-Eaton Myasthenic Syndrome, motor neuron disease, muscular atrophy such as spinal muscular atrophy, Charcot-Marie Disease and Werdnig-Hoffmann Disease, Postpoliomyelitis Syndrome, Muscular Dystrophy, Myasthenia Gravis, Myotonia Atrophica, Myotonia Confenita, Nemaline Myopathy, Familial Periodic Paralysis, Multiplex Paramyloclonus, Tropical Spastic Paraparesis and Stiff-Man Syndrome, peripheral nervous system diseases such as acrodynia, amyloid neuropathies, autonomic nervous system diseases such as Adie's Syndrome, Barre-Lieou Syndrome, Familial Dysautonomia, Horner's Syndrome, Reflex Sympathetic Dystrophy and Shy-Drager Syndrome, Cranial Nerve Diseases such as Acoustic Nerve Diseases such as Acoustic Neuroma which includes Neurofibromatosis 2, Facial Nerve Diseases such as Facial Neuralgia, Melkersson-Rosenthal Syndrome, ocular motility disorders which includes amblyopia, nystagmus, oculomotor nerve paralysis, ophthalmoplegia such as Duane's Syndrome, Horner's Syndrome, Chronic Progressive External Ophthalmoplegia which includes Kearns Syndrome, Strabismus such as Esotropia and Exotropia, Oculomotor Nerve Paralysis, Optic Nerve Diseases such as Optic Atrophy which includes Hereditary Optic Atrophy, Optic Disk Drusen, Optic Neuritis such as Neuromyelitis Optica, Papilledema, Trigeminal Neuralgia, Vocal Cord Paralysis, Demyelinating Diseases such as Neuromyelitis Optica and Swayback, and Diabetic neuropathies such as diabetic foot. [0808]
  • Additional neurologic diseases which can be treated or detected with fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include nerve compression syndromes such as carpal tunnel syndrome, tarsal tunnel syndrome, thoracic outlet syndrome such as cervical rib syndrome, ulnar nerve compression syndrome, neuralgia such as causalgia, cervico-brachial neuralgia, facial neuralgia and trigeminal neuralgia, neuritis such as experimental allergic neuritis, optic neuritis, polyneuritis, polyradiculoneuritis and radiculities such as polyradiculitis, hereditary motor and sensory neuropathies such as Charcot-Marie Disease, Hereditary Optic Atrophy, Refsum's Disease, Hereditary Spastic Paraplegia and Werdnig-Hoffmann Disease, Hereditary Sensory and Autonomic Neuropathies which include Congenital Analgesia and Familial Dysautonomia, POEMS Syndrome, Sciatica, Gustatory Sweating and Tetany). [0809]
  • Endocrine Disorders [0810]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose disorders and/or diseases related to hormone imbalance, and/or disorders or diseases of the endocrine system. [0811]
  • Hormones secreted by the glands of the endocrine system control physical growth, sexual function, metabolism, and other functions. Disorders may be classified in two ways: disturbances in the production of hormones, and the inability of tissues to respond to hormones. The etiology of these hormone imbalance or endocrine system diseases, disorders or conditions may be genetic, somatic, such as cancer and some autoimmune diseases, acquired (e.g., by chemotherapy, injury or toxins), or infectious. Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as a marker or detector of a particular disease or disorder related to the endocrine system and/or hormone imbalance. [0812]
  • Endocrine system and/or hormone imbalance and/or diseases encompass disorders of uterine motility including, but not limited to: complications with pregnancy and labor (e.g., pre-term labor, post-term pregnancy, spontaneous abortion, and slow or stopped labor); and disorders and/or diseases of the menstrual cycle (e.g., dysmenorrhea and endometriosis). [0813]
  • Endocrine system and/or hormone imbalance disorders and/or diseases include disorders and/or diseases of the pancreas, such as, for example, diabetes mellitus, diabetes insipidus, congenital pancreatic agenesis, pheochromocytoma—islet cell tumor syndrome; disorders and/or diseases of the adrenal glands such as, for example, Addison's Disease, corticosteroid deficiency, virilizing disease, hirsutism, Cushing's Syndrome, hyperaldosteronism, pheochromocytoma; disorders and/or diseases of the pituitary gland, such as, for example, hyperpituitarism, hypopituitarism, pituitary dwarfism, pituitary adenoma, panhypopituitarism, acromegaly, gigantism; disorders and/or diseases of the thyroid, including but not limited to, hyperthyroidism, hypothyroidism, Plummer's disease, Graves' disease (toxic diffuse goiter), toxic nodular goiter, thyroiditis (Hashimoto's thyroiditis, subacute granulomatous thyroiditis, and silent lymphocytic thyroiditis), Pendred's syndrome, myxedema, cretinism, thyrotoxicosis, thyroid hormone coupling defect, thymic aplasia, Hurthle cell tumours of the thyroid, thyroid cancer, thyroid carcinoma, Medullary thyroid carcinoma; disorders and/or diseases of the parathyroid, such as, for example, hyperparathyroidism, hypoparathyroidism; disorders and/or diseases of the hypothalamus. [0814]
  • In addition, endocrine system and/or hormone imbalance disorders and/or diseases may also include disorders and/or diseases of the testes or ovaries, including cancer. Other disorders and/or diseases of the testes or ovaries further include, for example, ovarian cancer, polycystic ovary syndrome, Klinefelter's syndrome, vanishing testes syndrome (bilateral anorchia), congenital absence of Leydig's cells, cryptorchidism, Noonan's syndrome, myotonic dystrophy, capillary haemangioma of the testis (benign), neoplasias of the testis and neo-testis. [0815]
  • Moreover, endocrine system and/or hormone imbalance disorders and/or diseases may also include disorders and/or diseases such as, for example, polyglandular deficiency syndromes, pheochromocytoma, neuroblastoma, multiple Endocrine neoplasia, and disorders and/or cancers of endocrine tissues. [0816]
  • In another embodiment, albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to diagnose; prognose, prevent, and/or treat endocrine diseases and/or disorders associated with the tissue(s) in which the Therapeutic protein corresponding to the Therapeutic protein portion of the albumin protein of the invention is expressed, [0817]
  • Reproductive System Disorders [0818]
  • The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used for the diagnosis, treatment, or prevention of diseases and/or disorders of the reproductive system. Reproductive system disorders that can be treated by the compositions of the invention, include, but are not limited to, reproductive system injuries, infections, neoplastic disorders, congenital defects, and diseases or disorders which result in infertility, complications with pregnancy, labor, or parturition, and postpartum difficulties. [0819]
  • Reproductive system disorders and/or diseases include diseases and/or disorders of the testes, including testicular atrophy, testicular feminization, cryptorchism (unilateral and bilateral), anorchia, ectopic testis, epididymitis and orchitis (typically resulting from infections such as, for example, gonorrhea, mumps, tuberculosis, and syphilis), testicular torsion, vasitis nodosa, germ cell tumors (e.g., seminomas, embryonal cell carcinomas, teratocarcinomas, choriocarcinomas, yolk sac tumors, and teratomas), stromal tumors (e.g., Leydig cell tumors), hydrocele, hematocele, varicocele, spermatocele, inguinal hernia, and disorders of sperm production (e.g., immotile cilia syndrome, aspermia, asthenozoospermia, azoospermia, oligospermia, and teratozoospermia). [0820]
  • Reproductive system disorders also include disorders of the prostate gland, such as acute non-bacterial prostatitis, chronic non-bacterial prostatitis, acute bacterial prostatitis, chronic bacterial prostatitis, prostatodystonia, prostatosis, granulomatous prostatitis, malacoplakia, benign prostatic hypertrophy or hyperplasia, and prostate neoplastic disorders, including adenocarcinomas, transitional cell carcinomas, ductal carcinomas, and squamous cell carcinomas. [0821]
  • Additionally, the compositions of the invention may be useful in the diagnosis, treatment, and/or prevention of disorders or diseases of the penis and urethra, including inflammatory disorders, such as balanoposthitis, balanitis xerotica obliterans, phimosis, paraphimosis, syphilis, herpes simplex virus, gonorrhea, non-gonococcal urethritis, chlamydia, mycoplasma, trichomonas, HIV, AIDS, Reiter's syndrome, condyloma acuminatum, condyloma latum, and pearly penile papules; urethral abnormalities, such as hypospadias, epispadias, and phimosis; premalignant lesions, including Erythroplasia of Queyrat, Bowen's disease, Bowenoid paplosis, giant condyloma of Buscke-Lowenstein, and varrucous carcinoma; penile cancers, including squamous cell carcinomas, carcinoma in situ, verrucous carcinoma, and disseminated penile carcinoma; urethral neoplastic disorders, including penile urethral carcinoma, bulbomembranous urethral carcinoma, and prostatic urethral carcinoma; and erectile disorders, such as priapism, Peyronie's disease, erectile dysfunction, and impotence. [0822]
  • Moreover, diseases and/or disorders of the vas deferens include vasculititis and CBAVD (congenital bilateral absence of the vas deferens); additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases and/or disorders of the seminal vesicles, including hydatid disease, congenital chloride diarrhea, and polycystic kidney disease. [0823]
  • Other disorders and/or diseases of the male reproductive system include, for example, Klinefelter's syndrome, Young's syndrome, premature ejaculation, diabetes mellitus, cystic fibrosis, Kartagener's syndrome, high fever, multiple sclerosis, and gynecomastia. [0824]
  • Further, the polynucleotides, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases and/or disorders of the vagina and vulva, including bacterial vaginosis, candida vaginitis, herpes simplex virus, chancroid, granuloma inguinale, lymphogranuloma venereum, scabies, human papillomavirus, vaginal trauma, vulvar trauma, adenosis, chlamydia vaginitis, gonorrhea, trichomonas vaginitis, condyloma acuminatum, syphilis, molluscum contagiosum, atrophic vaginitis, Paget's disease, lichen sclerosus, lichen planus, vulvodynia, toxic shock syndrome, vaginismus, vulvovaginitis, vulvar vestibulitis, and neoplastic disorders, such as squamous cell hyperplasia, clear cell carcinoma, basal cell carcinoma, melanomas, cancer of Bartholin's gland, and vulvar intraepithelial neoplasia. [0825]
  • Disorders and/or diseases of the uterus include dysmenorrhea, retroverted uterus, endometriosis, fibroids, adenomyosis, anovulatory bleeding, amenorrhea, Cushing's syndrome, hydatidiform moles, Asherman's syndrome, premature menopause, precocious puberty, uterine polyps, dysfunctional uterine bleeding (e.g., due to aberrant hormonal signals), and neoplastic disorders, such as adenocarcinomas, keiomyosarcomas, and sarcomas. Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be useful as a marker or detector of, as well as in the diagnosis, treatment, and/or prevention of congenital uterine abnormalities, such as bicornuate uterus, septate uterus, simple unicornuate uterus, unicornuate uterus with a noncavitary rudimentary horn, unicornuate uterus with a non-communicating cavitary rudimentary horn, unicornuate uterus with a communicating cavitary horn, arcuate uterus, uterine didelfus, and T-shaped uterus. [0826]
  • Ovarian diseases and/or disorders include anovulation, polycystic ovary syndrome (Stein-Leventhal syndrome), ovarian cysts, ovarian hypofunction, ovarian insensitivity to gonadotropins, ovarian overproduction of androgens, right ovarian vein syndrome, amenorrhea, hirutism, and ovarian cancer (including, but not limited to, primary and secondary cancerous growth, Sertoli-Leydig tumors, endometriod carcinoma of the ovary, ovarian papillary serous adenocarcinoma, ovarian mucinous adenocarcinoma, and Ovarian Krukenberg tumors). [0827]
  • Cervical diseases and/or disorders include cervicitis, chronic cervicitis, mucopurulent cervicitis, cervical dysplasia, cervical polyps, Nabothian cysts, cervical erosion, cervical incompetence, and cervical neoplasms (including, for example, cervical carcinoma, squamous metaplasia, squamous cell carcinoma, adenosquamous cell neoplasia, and columnar cell neoplasia). Additionally, diseases and/or disorders of the reproductive system include disorders and/or diseases of pregnancy, including miscarriage and stillbirth, such as early abortion, late abortion, spontaneous abortion, induced abortion, therapeutic abortion, threatened abortion, missed abortion, incomplete abortion, complete abortion, habitual abortion, missed abortion, and septic abortion; ectopic pregnancy, anemia, Rh incompatibility, vaginal bleeding during pregnancy, gestational diabetes, intrauterine growth retardation, polyhydramnios, HELLP syndrome, abruptio placentae, placenta previa, hyperemesis, preeclampsia, eclampsia, herpes gestationis, and urticaria of pregnancy. Additionally, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may be used in the diagnosis, treatment, and/or prevention of diseases that can complicate pregnancy, including heart disease, heart failure, rheumatic heart disease, congenital heart disease, mitral valve prolapse, high blood pressure, anemia, kidney disease, infectious disease (e.g., rubella, cytomegalovirus, toxoplasmosis, infectious hepatitis, chlamydia, HIV, AIDS, and genital herpes), diabetes mellitus, Graves' disease, thyroiditis, hypothyroidism, Hashimoto's thyroiditis, chronic active hepatitis, cirrhosis of the liver, primary biliary cirrhosis, asthma, systemic lupus eryematosis, rheumatoid arthritis, myasthenia gravis, idiopathic thrombocytopenic purpura, appendicitis, ovarian cysts, gallbladder disorders, and obstruction of the intestine. [0828]
  • Complications associated with labor and parturition include premature rupture of the membranes, pre-term labor, post-term pregnancy, postmaturity, labor that progresses too slowly, fetal distress (e.g., abnormal heart rate (fetal or maternal), breathing problems, and abnormal fetal position), shoulder dystocia, prolapsed umbilical cord, amniotic fluid embolism, and aberrant uterine bleeding. [0829]
  • Further, diseases and/or disorders of the postdelivery period, including endometritis, myometritis, parametritis, peritonitis, pelvic thrombophlebitis, pulmonary embolism, endotoxemia, pyelonephritis, saphenous thrombophlebitis, mastitis, cystitis, postpartum hemorrhage, and inverted uterus. [0830]
  • Other disorders and/or diseases of the female reproductive system that may be diagnosed, treated, and/or prevented by the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, for example, Turner's syndrome, pseudohermaphroditism, premenstrual syndrome, pelvic inflammatory disease, pelvic congestion (vascular engorgement), frigidity, anorgasmia, dyspareunia, ruptured fallopian tube, and Mittelschmerz. [0831]
  • Infectious Disease [0832]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to treat or detect infectious agents. For example, by increasing the immune response, particularly increasing the proliferation and differentiation of B and/or T cells, infectious diseases may be treated. The immune response may be increased by either enhancing an existing immune response, or by initiating a new immune response. Alternatively, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also directly inhibit the infectious agent, without necessarily eliciting an immune response. [0833]
  • Viruses are one example of an infectious agent that can cause disease or symptoms that can be treated or detected by albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. Examples of viruses, include, but are not limited to Examples of viruses, include, but are not limited to the following DNA and RNA viruses and viral families: Arbovirus, Adenoviridae, Arenaviridae, Arterivirus, Birnaviridae, Bunyaviridae, Caliciviridae, Circoviridae, Coronaviridae, Dengue, EBV, HIV, Flaviviridae, Hepadnaviridae (Hepatitis), Herpesviridae (such as, Cytomegalovirus, Herpes Simplex, Herpes Zoster), Mononegavirus (e.g., Paramyxoviridae, Morbillivirus, Rhabdoviridae), Orthomyxoviridae (e.g., Influenza A, Influenza B, and parainfluenza), Papiloma virus, Papovaviridae, Parvoviridae, Picomaviridae, Poxyiridae (such as Smallpox or Vaccinia), Reoviridae (e.g., Rotavirus), Retroviridae (HTLV-I, HTLV-II, Lentivirus), and Togaviridae (e.g., Rubivirus). Viruses falling within these families can cause a variety of diseases or symptoms, including, but not limited to: arthritis, bronchiollitis, respiratory syncytial virus, encephalitis, eye infections (e.g., conjunctivitis, keratitis), chronic fatigue syndrome, hepatitis (A, B, C, E, Chronic Active, Delta), Japanese B encephalitis, Junin, Chikungunya, Rift Valley fever, yellow fever, meningitis, opportunistic infections (e.g., AIDS), pneumonia, Burkitt's Lymphoma, chickenpox, hemorrhagic fever, Measles, Mumps, Parainfluenza, Rabies, the common cold, Polio, leukemia, Rubella, sexually transmitted diseases, skin diseases (e.g., Kaposi's, warts), and viremia. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat or detect any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat: meningitis, Dengue, EBV, and/or hepatitis (e.g., hepatitis B). In an additional specific embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat patients nonresponsive to one or more other commercially available hepatitis vaccines. In a further specific embodiment fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat AIDS. [0834]
  • Similarly, bacterial and fungal agents that can cause disease or symptoms and that can be treated or detected by albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but not limited to, the following Gram-Negative and Gram-positive bacteria, bacterial families, and fungi: Actinomyces (e.g., Norcardia), Acinetobacter, [0835] Cryptococcus neoformans, Aspergillus, Bacillaceae (e.g., Bacillus anthrasis), Bacteroides (e.g., Bacteroides fragilis), Blastomycosis, Bordetella, Borrelia (e.g., Borrelia burgdorferi), Brucella, Candidia, Campylobacter, Chlamydia, Clostridium (e.g., Clostridium botulinum, Clostridium dificile, Clostridium perfringens, Clostridium tetani), Coccidioides, Corynebacterium (e.g., Corynebacterium diptheriae), Cryptococcus, Dermatocycoses, E. coli (e.g., Enterotoxigenic E. coli and Enterohemorrhagic E. coli), Enterobacter (e.g. Enterobacter aerogenes), Enterobacteriaceae (Klebsiella, Salmonella (e.g., Salmonella typhi, Salmonella enteritidis, Salmonella typhi), Serratia, Yersinia, Shigella), Erysipelothrix, Haemophilus (e.g., Haemophilus influenza type B), Helicobacter, Legionella (e.g., Legionella pneumophila), Leptospira, Listeria (e.g., Listeria monocytogenes), Mycoplasma, Mycobacterium (e.g., Mycobacterium leprae and Mycobacterium tuberculosis), Vibrio (e.g., Vibrio cholerae), Neisseriaceae (e.g., Neisseria gonorrhea, Neisseria meningitidis), Pasteurellacea, Proteus, Pseudomonas (e.g., Pseudomonas aeruginosa), Rickettsiaceae, Spirochetes (e.g., Treponema spp., Leptospira spp., Borrelia spp.), Shigella spp., Staphylococcus (e.g., Staphylococcus aureus), Meningiococcus, Pneumococcus and Streptococcus (e.g., Streptococcus pneumoniae and Groups A, B, and C Streptococci), and Ureaplasmas. These bacterial, parasitic, and fungal families can cause diseases or symptoms, including, but not limited to: antibiotic-resistant infections, bacteremia, endocarditis, septicemia, eye infections (e.g., conjunctivitis), uveitis, tuberculosis, gingivitis, bacterial diarrhea, opportunistic infections (e.g., AIDS related infections), paronychia, prosthesis-related infections, dental caries, Reiter's Disease, respiratory tract infections, such as Whooping Cough or Empyema, sepsis, Lyme Disease, Cat-Scratch Disease, dysentery, paratyphoid fever, food poisoning, Legionella disease, chronic and acute inflammation, erythema, yeast infections, typhoid, pneumonia, gonorrhea, meningitis (e.g., mengitis types A and B), chlamydia, syphillis, diphtheria, leprosy, brucellosis, peptic ulcers, anthrax, spontaneous abortions, birth defects, pneumonia, lung infections, ear infections, deafness, blindness, lethargy, malaise, vomiting, chronic diarrhea, Crohn's disease, colitis, vaginosis, sterility, pelvic inflammatory diseases, candidiasis, paratuberculosis, tuberculosis, lupus, botulism, gangrene, tetanus, impetigo, Rheumatic Fever, Scarlet Fever, sexually transmitted diseases, skin diseases (e.g., cellulitis, dermatocycoses), toxemia, urinary tract infections, wound infections, noscomial infections. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat or detect any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat: tetanus, diptheria, botulism, and/or meningitis type B.
  • Moreover, parasitic agents causing disease or symptoms that can be treated, prevented, and/or diagnosed by fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention include, but not limited to, the following families or class: Amebiasis, Babesiosis, Coccidiosis, Cryptosporidiosis, Dientamoebiasis, Dourine, Ectoparasitic, Giardias, Helminthiasis, Leishmaniasis, Schistisoma, Theileriasis, Toxoplasmosis, Trypanosomiasis, and Trichomonas and Sporozoans (e.g., [0836] Plasmodium virax, Plasmodium falciparium, Plasmodium malariae and Plasmodium ovale). These parasites can cause a variety of diseases or symptoms, including, but not limited to: Scabies, Trombiculiasis, eye infections, intestinal disease (e.g., dysentery, giardiasis), liver disease, lung disease, opportunistic infections (e.g., AIDS related), malaria, pregnancy complications, and toxoplasmosis. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, can be used to treat, prevent, and/or diagnose any of these symptoms or diseases. In specific embodiments, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention are used to treat, prevent, and/or diagnose malaria.
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could either be by administering an effective amount of an albumin fusion protein of the invnetion to the patient, or by removing cells from the patient, supplying the cells with a polynucleotide of the present invention, and returning the engineered cells to the patient (ex vivo therapy). Moreover, the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used as an antigen in a vaccine to raise an immune response against infectious disease. [0837]
  • Regeneration [0838]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention can be used to differentiate, proliferate, and attract cells, leading to the regeneration of tissues. (See, Science 276:59-87 (1997)). The regeneration of tissues could be used to repair, replace, or protect tissue damaged by congenital defects, trauma (wounds, burns, incisions, or ulcers), age, disease (e.g. osteoporosis, osteocarthritis, periodontal disease, liver failure), surgery, including cosmetic plastic surgery, fibrosis, reperfusion injury, or systemic cytokine damage. [0839]
  • Tissues that could be regenerated using the present invention include organs (e.g., pancreas, liver, intestine, kidney, skin, endothelium), muscle (smooth, skeletal or cardiac), vasculature (including vascular and lymphatics), nervous, hematopoietic, and skeletal (bone, cartilage, tendon, and ligament) tissue. Preferably, regeneration occurs without or decreased scarring. Regeneration also may include angiogenesis. [0840]
  • Moreover, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may increase regeneration of tissues difficult to heal. For example, increased tendon/ligament regeneration would quicken recovery time after damage. Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could also be used prophylactically in an effort to avoid damage. Specific diseases that could be treated include of tendinitis, carpal tunnel syndrome, and other tendon or ligament defects. A further example of tissue regeneration of non-healing wounds includes pressure ulcers, ulcers associated with vascular insufficiency, surgical, and traumatic wounds. [0841]
  • Similarly, nerve and brain tissue could also be regenerated by using fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, to proliferate and differentiate nerve cells. Diseases that could be treated using this method include central and peripheral nervous system diseases, neuropathies, or mechanical and traumatic disorders (e.g., spinal cord disorders, head trauma, cerebrovascular disease, and stoke). Specifically, diseases associated with peripheral nerve injuries, peripheral neuropathy (e.g., resulting from chemotherapy or other medical therapies), localized neuropathies, and central nervous system diseases (e.g., Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, and Shy-Drager syndrome), could all be treated using the albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention. [0842]
  • Gastrointestinal Disorders [0843]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention, may be used to treat, prevent, diagnose, and/or prognose gastrointestinal disorders, including inflammatory diseases and/or conditions, infections, cancers (e.g., intestinal neoplasms (carcinoid tumor of the small intestine, non-Hodgkin's lymphoma of the small intestine, small bowl lymphoma)), and ulcers, such as peptic ulcers. [0844]
  • Gastrointestinal disorders include dysphagia, odynophagia, inflammation of the esophagus, peptic esophagitis, gastric reflux, submucosal fibrosis and stricturing, Mallory-Weiss lesions, leiomyomas, lipomas, epidermal cancers, adeoncarcinomas, gastric retention disorders, gastroenteritis, gastric atrophy, gastric/stomach cancers, polyps of the stomach, autoimmune disorders such as pernicious anemia, pyloric stenosis, gastritis (bacterial, viral, eosinophilic, stress-induced, chronic erosive, atrophic, plasma cell, and Menetrier's), and peritoneal diseases (e.g., chyloperioneum, hemoperitoneum, mesenteric cyst, mesenteric lymphadenitis, mesenteric vascular occlusion, panniculitis, neoplasms, peritonitis, pneumoperitoneum, bubphrenic abscess,). [0845]
  • Gastrointestinal disorders also include disorders associated with the small intestine, such as malabsorption syndromes, distension, irritable bowel syndrome, sugar intolerance, celiac disease, duodenal ulcers, duodenitis, tropical sprue, Whipple's disease, intestinal lymphangiectasia, Crohn's disease, appendicitis, obstructions of the ileum, Meckel's diverticulum, multiple diverticula, failure of complete rotation of the small and large intestine, lymphoma, and bacterial and parasitic diseases (such as Traveler's diarrhea, typhoid and paratyphoid, cholera, infection by Roundworms ([0846] Ascariasis lumbricoides), Hookworms (Ancylostoma duodenale), Threadworms (Enterobius vermicularis), Tapeworms (Taenia saginata, Echinococcus granulosus, Diphyllobothrium spp., and T. solium).
  • Liver diseases and/or disorders include intrahepatic cholestasis (alagille syndrome, biliary liver cirrhosis), fatty liver (alcoholic fatty liver, reye syndrome), hepatic vein thrombosis, hepatolentricular degeneration, hepatomegaly, hepatopulmonary syndrome, hepatorenal syndrome, portal hypertension (esophageal and gastric varices), liver abscess (amebic liver abscess), liver cirrhosis (alcoholic, biliary and experimental), alcoholic liver diseases (fatty liver, hepatitis, cirrhosis), parasitic (hepatic echinococcosis, fascioliasis, amebic liver abscess), jaundice (hemolytic, hepatocellular, and cholestatic), cholestasis, portal hypertension, liver enlargement, ascites, hepatitis (alcoholic hepatitis, animal hepatitis, chronic hepatitis (autoimmune, hepatitis B, hepatitis C, hepatitis D, drug induced), toxic hepatitis, viral human hepatitis (hepatitis A, hepatitis B, hepatitis C, hepatitis D, hepatitis E), Wilson's disease, granulomatous hepatitis, secondary biliary cirrhosis, hepatic encephalopathy, portal hypertension, varices, hepatic encephalopathy, primary biliary cirrhosis, primary sclerosing cholangitis, hepatocellular adenoma, hemangiomas, bile stones, liver failure (hepatic encephalopathy, acute liver failure), and liver neoplasms (angiomyolipoma, calcified liver metastases, cystic liver metastases, epithelial tumors, fibrolamellar hepatocarcinoma, focal nodular hyperplasia, hepatic adenoma, hepatobiliary cystadenoma, hepatoblastoma, hepatocellular carcinoma, hepatoma, liver cancer, liver hemangioendothelioma, mesenchymal hamartoma, mesenchymal tumors of liver, nodular regenerative hyperplasia, benign liver tumors (Hepatic cysts [Simple cysts, Polycystic liver disease, Hepatobiliary cystadenoma, Choledochal cyst], Mesenchymal tumors [Mesenchymal hamartoma, Infantile hemangioendothelioma, Hemangioma, Peliosis hepatis, Lipomas, Inflammatory pseudotumor, Miscellaneous], Epithelial tumors [Bile duct epithelium (Bile duct hamartoma, Bile duct adenoma), Hepatocyte (Adenoma, Focal nodular hyperplasia, Nodular regenerative hyperplasia)], malignant liver tumors [hepatocellular, hepatoblastoma, hepatocellular carcinoma, cholangiocellular, cholangiocarcinoma, cystadenocarcinoma, tumors of blood vessels, angiosarcoma, Karposi's sarcoma, hemangioendothelioma, other tumors, embryonal sarcoma, fibrosarcoma, leiomyosarcoma, rhabdomyosarcoma, carcinosarcoma, teratoma, carcinoid, squamous carcinoma, primary lymphoma]), peliosis hepatis, erythrohepatic porphyria, hepatic porphyria (acute intermittent porphyria, porphyria cutanea tarda), Zellweger syndrome). [0847]
  • Pancreatic diseases and/or disorders include acute pancreatitis, chronic pancreatitis (acute necrotizing pancreatitis, alcoholic pancreatitis), neoplasms (adenocarcinoma of the pancreas, cystadenocarcinoma, insulinoma, gastrinoma, and glucagonoma, cystic neoplasms, islet-cell tumors, pancreoblastoma), and other pancreatic diseases (e.g., cystic fibrosis, cyst (pancreatic pseudocyst, pancreatic fistula, insufficiency)). [0848]
  • Gallbladder diseases include gallstones (cholelithiasis and choledocholithiasis), postcholecystectomy syndrome, diverticulosis of the gallbladder, acute cholecystitis, chronic cholecystitis, bile duct tumors, and mucocele. [0849]
  • Diseases and/or disorders of the large intestine include antibiotic-associated colitis, diverticulitis, ulcerative colitis, acquired megacolon, abscesses, fungal and bacterial infections, anorectal disorders (e.g., fissures, hemorrhoids), colonic diseases (colitis, colonic neoplasms [colon cancer, adenomatous colon polyps (e.g., villous adenoma), colon carcinoma, colorectal cancer], colonic diverticulitis, colonic diverticulosis, megacolon [Hirschsprung disease, toxic megacolon]; sigmoid diseases [proctocolitis, sigmoin neoplasms]), constipation, Crohn's disease, diarrhea (infantile diarrhea, dysentery), duodenal diseases (duodenal neoplasms, duodenal obstruction, duodenal ulcer, duodenitis), enteritis (enterocolitis), HIV enteropathy, ileal diseases (ileal neoplasms, ileitis), immunoproliferative small intestinal disease, inflammatory bowel disease (ulcerative colitis, Crohn's disease), intestinal atresia, parasitic diseases (anisakiasis, balantidiasis, blastocystis infections, cryptosporidiosis, dientamoebiasis, amebic dysentery, giardiasis), intestinal fistula (rectal fistula), intestinal neoplasms (cecal neoplasms, colonic neoplasms, duodenal neoplasms, ileal neoplasms, intestinal polyps, jejunal neoplasms, rectal neoplasms), intestinal obstruction (afferent loop syndrome, duodenal obstruction, impacted feces, intestinal pseudo-obstruction [cecal volvulus], intussusception), intestinal perforation, intestinal polyps (colonic polyps, gardner syndrome, peutz-jeghers syndrome), jejunal diseases (jejunal neoplasms), malabsorption syndromes (blind loop syndrome, celiac disease, lactose intolerance, short bowl syndrome, tropical sprue, whipple's disease), mesenteric vascular occlusion, pneumatosis cystoides intestinalis, protein-losing enteropathies (intestinal lymphagiectasis), rectal diseases (anus diseases, fecal incontinence, hemorrhoids, proctitis, rectal fistula, rectal prolapse, rectocele), peptic ulcer (duodenal ulcer, peptic esophagitis, hemorrhage, perforation, stomach ulcer, Zollinger-Ellison syndrome), postgastrectomy syndromes (dumping syndrome), stomach diseases (e.g., achlorhydria, duodenogastric reflux (bile reflux), gastric antral vascular ectasia, gastric fistula, gastric outlet obstruction, gastritis (atrophic or hypertrophic), gastroparesis, stomach dilatation, stomach diverticulum, stomach neoplasms (gastric cancer, gastric polyps, gastric adenocarcinoma, hyperplastic gastric polyp), stomach rupture, stomach ulcer, stomach volvulus), tuberculosis, visceroptosis, vomiting (e.g., hematemesis, hyperemesis gravidarum, postoperative nausea and vomiting) and hemorrhagic colitis. [0850]
  • Further diseases and/or disorders of the gastrointestinal system include biliary tract diseases, such as, gastroschisis, fistula (e.g., biliary fistula, esophageal fistula, gastric fistula, intestinal fistula, pancreatic fistula), neoplasms (e.g., biliary tract neoplasms, esophageal neoplasms, such as adenocarcinoma of the esophagus, esophageal squamous cell carcinoma, gastrointestinal neoplasms, pancreatic neoplasms, such as adenocarcinoma of the pancreas, mucinous cystic neoplasm of the pancreas, pancreatic cystic neoplasms, pancreatoblastoma, and peritoneal neoplasms), esophageal disease (e.g., bullous diseases, candidiasis, glycogenic acanthosis, ulceration, barrett esophagus varices, atresia, cyst, diverticulum (e.g., Zenker's diverticulum), fistula (e.g., tracheoesophageal fistula), motility disorders (e.g., CREST syndrome, deglutition disorders, achalasia, spasm, gastroesophageal reflux), neoplasms, perforation (e.g., Boerhaave syndrome, Mallory-Weiss syndrome), stenosis, esophagitis, diaphragmatic hernia (e.g., hiatal hernia); gastrointestinal diseases, such as, gastroenteritis (e.g., cholera morbus, norwalk virus infection), hemorrhage (e.g., hematemesis, melena, peptic ulcer hemorrhage), stomach neoplasms (gastric cancer, gastric polyps, gastric adenocarcinoma, stomach cancer)), hernia (e.g., congenital diaphragmatic hernia, femoral hernia, inguinal hernia, obturator hernia, umbilical hernia, ventral hernia), and intestinal diseases (e.g., cecal diseases (appendicitis, cecal neoplasms)). [0851]
  • Chemotaxis [0852]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may have chemotaxis activity. A chemotaxic molecule attracts or mobilizes cells (e.g., monocytes, fibroblasts, neutrophils, T-cells, mast cells, eosinophils, epithelial and/or endothelial cells) to a particular site in the body, such as inflammation, infection, or site of hyperproliferation. The mobilized cells can then fight off and/or heal the particular trauma or abnormality. [0853]
  • Albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may increase chemotaxic activity of particular cells. These chemotactic molecules can then be used to treat inflammation, infection, hyperproliferative disorders, or any immune system disorder by increasing the number of cells targeted to a particular location in the body. For example, chemotaxic molecules can be used to treat wounds and other trauma to tissues by attracting immune cells to the injured location. Chemotactic molecules of the present invention can also attract fibroblasts, which can be used to treat wounds. [0854]
  • It is also contemplated that fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may inhibit chemotactic activity. These molecules could also be used to treat disorders. Thus, fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention could be used as an inhibitor of chemotaxis. [0855]
  • Binding Activity [0856]
  • Albumin fusion proteins of the invention may be used to screen for molecules that bind to the Therapeutic protein portion of the fusion protein or for molecules to which the Therapeutic protein portion of the fusion protein binds. The binding of the fusion protein and the molecule may activate (agonist), increase, inhibit (antagonist), or decrease activity of the fusion protein or the molecule bound. Examples of such molecules include antibodies, oligonucleotides, proteins (e.g., receptors), or small molecules. [0857]
  • Preferably, the molecule is closely related to the natural ligand of the Therapeutic protein portion of the fusion protein of the invention, e.g., a fragment of the ligand, or a natural substrate, a ligand, a structural or functional mimetic. (See, Coligan et al., Current Protocols in Imnunology 1(2):Chapter 5 (1991)). Similarly, the molecule can be closely related to the natural receptor to which the Therapeutic protein portion of an albumin fusion protein of the invention binds, or at least, a fragment of the receptor capable of being bound by the Therapeutic protein portion of an albumin fusion protein of the invention (e.g., active site). In either case, the molecule can be rationally designed using known techniques. [0858]
  • Preferably, the screening for these molecules involves producing appropriate cells which express the albumin fusion proteins of the invention. Preferred cells include cells from mammals, yeast, Drosophila, or [0859] E. coli.
  • The assay may simply test binding of a candidate compound to an albumin fusion protein of the invention, wherein binding is detected by a label, or in an assay involving competition with a labeled competitor. Further, the assay may test whether the candidate compound results in a signal generated by binding to the fusion protein. [0860]
  • Alternatively, the assay can be carried out using cell-free preparations, fusion protein/molecule affixed to a solid support, chemical libraries, or natural product mixtures. The assay may also simply comprise the steps of mixing a candidate compound with a solution containing an albumin fusion protein, measuring fusion protein/molecule activity or binding, and comparing the fusion protein/molecule activity or binding to a standard. [0861]
  • Preferably, an ELISA assay can measure fusion protein level or activity in a sample (e.g., biological sample) using a monoclonal or polyclonal antibody. The antibody can measure fusion protein level or activity by either binding, directly or indirectly, to the albumin fusion protein or by competing with the albumin fusion protein for a substrate. [0862]
  • Additionally, the receptor to which a Therapeutic protein portion of an albumin fusion protein of the invention binds can be identified by numerous methods known to those of skill in the art, for example, ligand panning and FACS sorting (Coligan, et al., Current Protocols in Immun., 1(2), [0863] Chapter 5, (1991)). For example, in cases wherein the Therapeutic protein portion of the fusion protein corresponds to FGF, expression cloning may be employed wherein polyadenylated RNA is prepared from a cell responsive to the albumin fusion protein, for example, NIH3T3 cells which are known to contain multiple receptors for the FGF family proteins, and SC-3 cells, and a cDNA library created from this RNA is divided into pools and used to transfect COS cells or other cells that are not responsive to the albumin fusion protein. Transfected cells which are grown on glass slides are exposed to the albumin fusion protein of the present invention, after they have been labeled. The albumin fusion proteins can be labeled by a variety of means including iodination or inclusion of a recognition site for a site-specific protein kinase.
  • Following fixation and incubation, the slides are subjected to auto-radiographic analysis. Positive pools are identified and sub-pools are prepared and re-transfected using an iterative sub-pooling and re-screening process, eventually yielding a single clones that encodes the putative receptor. [0864]
  • As an alternative approach for receptor identification, a labeled albumin fusion protein can be photoaffinity linked with cell membrane or extract preparations that express the receptor molecule for the Therapeutoc protein component of an albumin fusion protein of the invention, the linked material may be resolved by PAGE analysis and exposed to X-ray film. The labeled complex containing the receptors of the fusion protein can be excised, resolved into peptide fragments, and subjected to protein microsequencing. The amino acid sequence obtained from microsequencing would be used to design a set of degenerate oligonucleotide probes to screen a cDNA library to identify the genes encoding the putative receptors. [0865]
  • Moreover, the techniques of gene-shuffling, motif-shuffling, exon-shuffling, and/or codon-shuffling (collectively referred to as “DNA shuffling”) may be employed to modulate the activities of the fusion protein, and/or Therapeutic protein portion or albumin component of an albumin fusion protein of the present invention, thereby effectively generating agonists and antagonists of an albumin fusion protein of the present invention. See generally, U.S. Pat. Nos. 5,605,793, 5,811,238, 5,830,721, 5,834,252, and 5,837,458, and Patten, P. A., et al., [0866] Curr. Opinion Biotechnol. 8:724-33 (1997); Harayama, S. Trends Biotechnol. 16(2):76-82 (1998); Hansson, L. O., et al., J. Mol. Biol. 287:265-76 (1999); and Lorenzo, M. M. and Blasco, R. Biotechniques 24(2):308-13 (1998); each of these patents and publications are hereby incorporated by reference). In one embodiment, alteration of polynucleotides encoding albumin fusion proteins of the invention and thus, the albumin fusion proteins encoded thereby, may be achieved by DNA shuffling. DNA shuffling involves the assembly of two or more DNA segments into a desired molecule by homologous, or site-specific, recombination. In another embodiment, polynucleotides encoding albumin fusion proteins of the invention and thus, the albumin fusion proteins encoded thereby, may be altered by being subjected to random mutagenesis by error-prone PCR, random nucleotide insertion or other methods prior to recombination. In another embodiment, one or more components, motifs, sections, parts, domains, fragments, etc., of an albumin fusion protein of the present invention may be recombined with one or more components, motifs, sections, parts, domains, fragments, etc. of one or more heterologous molecules. In preferred embodiments, the heterologous molecules are family members. In further preferred embodiments, the heterologous molecule is a growth factor such as, for example, platelet-derived growth factor (PDGF), insulin-like growth factor (IGF-I), transforming growth factor (TGF)-alpha, epidermal growth factor (EGF), fibroblast growth factor (FGF), TGF-beta, bone morphogenetic protein (BMP)-2, BMP-4, BMP-5, BMP-6, BMP-7, activins A and B, decapentaplegic(dpp), 60A, OP-2, dorsalin, growth differentiation factors (GDFs), nodal, MIS, inhibin-alpha, TGF-beta1, TGF-beta2, TGF-beta3, TGF-beta5, and glial-derived neurotrophic factor (GDNF).
  • Other preferred fragments are biologically active fragments of the Therapeutic protein portion and/or albumin component of the albumin fusion proteins of the present invention. Biologically active fragments are those exhibiting activity similar, but not necessarily identical, to an activity of a Therapeutic protein portion and/or albumin component of the albumin fusion proteins of the present invention. The biological activity of the fragments may include an improved desired activity, or a decreased undesirable activity. [0867]
  • Additionally, this invention provides a method of screening compounds to identify those which modulate the action of an albumin fusion protein of the present invention. An example of such an assay comprises combining a mammalian fibroblast cell, an albumin fusion protein of the present invention, and the compound to be screened and [0868] 3[H] thymidine under cell culture conditions where the fibroblast cell would normally proliferate. A control assay may be performed in the absence of the compound to be screened and compared to the amount of fibroblast proliferation in the presence of the compound to determine if the compound stimulates proliferation by determining the uptake of 3[H] thymidine in each case. The amount of fibroblast cell proliferation is measured by liquid scintillation chromatography which measures the incorporation of 3[H] thymidine. Both agonist and antagonist compounds may be identified by this procedure.
  • In another method, a mammalian cell or membrane preparation expressing a receptor for the Therapeutic protien component of a fusion protine of the invention is incubated with a labeled fusion protein of the present invention in the presence of the compound. The ability of the compound to enhance or block this interaction could then be measured. Alternatively, the response of a known second messenger-system following interaction of a compound to be screened and the receptor is measured and the ability of the compound to bind to the receptor and elicit a second messenger response is measured to determine if the compound is a potential fusion protein. Such second-messenger systems include but are not limited to, cAMP guanylate cyclase, ion channels or phosphoinositide hydrolysis. [0869]
  • All of these above assays can be used as diagnostic or prognostic markers. The molecules discovered using these assays can be used to treat disease or to bring about a particular result in a patient (e.g., blood vessel growth) by activating or inhibiting the fusion protein/molecule. Moreover, the assays can discover agents which may inhibit or enhance the production of the albumin fusion proteins of the invention from suitably manipulated cells or tissues. [0870]
  • Therefore, the invention includes a method of identifying compounds which bind to an albumin fusion protein of the invention comprising the steps of: (a) incubating a candidate binding compound with an albumin fusion protein of the present invention; and (b) determining if binding has occurred. Moreover, the invention includes a method of identifying agonists/antagonists comprising the steps of: (a) incubating a candidate compound with an albumin fusion protein of the present invention, (b) assaying a biological activity, and (b) determining if a biological activity of the fusion protein has been altered. [0871]
  • Targeted Delivery [0872]
  • In another embodiment, the invention provides a method of delivering compositions to targeted cells expressing a receptor for a component of an albumin fusion protein of the invention. [0873]
  • As discussed herein, fusion proteins of the invention may be associated with heterologous polypeptides, heterologous nucleic acids, toxins, or prodrugs via hydrophobic, hydrophilic, ionic and/or covalent interactions. In one embodiment, the invention provides a method for the specific delivery of compositions of the invention to cells by administering fusion proteins of the invention (including antibodies) that are associated with heterologous polypeptides or nucleic acids. In one example, the invention provides a method for delivering a Therapeutic protein into the targeted cell. In another example, the invention provides a method for delivering a single stranded nucleic acid (e.g., antisense or ribozymes) or double stranded nucleic acid (e.g., DNA that can integrate into the cell's genome or replicate episomally and that can be transcribed) into the targeted cell. [0874]
  • In another embodiment, the invention provides a method for the specific destruction of cells (e.g., the destruction of tumor cells) by administering an albumin fusion protein of the invention (e.g., polypeptides of the invention or antibodies of the invention) in association with toxins or cytotoxic prodrugs. [0875]
  • By “toxin” is meant compounds that bind and activate endogenous cytotoxic effector systems, radioisotopes, holotoxins, modified toxins, catalytic subunits of toxins, or any molecules or enzymes not normally present in or on the surface of a cell that under defined conditions cause the cell's death. Toxins that may be used according to the methods of the invention include, but are not limited to, radioisotopes known in the art, compounds such as, for example, antibodies (or complement fixing containing portions thereof) that bind an inherent or induced endogenous cytotoxic effector system, thymidine kinase, endonuclease, RNAse, alpha toxin, ricin, abrin, Pseudomonas exotoxin A, diphtheria toxin, saporin, momordin, gelonin, pokeweed antiviral protein, alpha-sarcin and cholera toxin. By “cytotoxic prodrug” is meant a non-toxic compound that is converted by an enzyme, normally present in the cell, into a cytotoxic compound. Cytotoxic prodrugs that may be used according to the methods of the invention include, but are not limited to, glutamyl derivatives of benzoic acid mustard alkylating agent, phosphate derivatives of etoposide or mitomycin C, cytosine arabinoside, daunorubisin, and phenoxyacetamide derivatives of doxorubicin. [0876]
  • Drug Screening [0877]
  • Further contemplated is the use of the albumin fusion proteins of the present invention, or the polynucleotides encoding these fusion proteins, to screen for molecules which modify the activities of the albumin fusion protein of the present invention or proteins corresponding to the Therapeutic protein portion of the albumin fusion protein. Such a method would include contacting the fusion protein with a selected compound(s) suspected of having antagonist or agonist activity, and assaying the activity of the fusion protein following binding. [0878]
  • This invention is particularly useful for screening therapeutic compounds by using the albumin fusion proteins of the present invention, or binding fragments thereof, in any of a variety of drug screening techniques. The albumin fusion protein employed in such a test may be affixed to a solid support, expressed on a cell surface, free in solution, or located intracellularly. One method of drug screening utilizes eukaryotic or prokaryotic host cells which are stably transformed with recombinant nucleic acids expressing the albumin fusion protein. Drugs are screened against such transformed cells or supernatants obtained from culturing such cells, in competitive binding assays. One may measure, for example, the formulation of complexes between the agent being tested and an albumin fusion protein of the present invention. [0879]
  • Thus, the present invention provides methods of screening for drugs or any other agents which affect activities mediated by the albumin fusion proteins of the present invention. These methods comprise contacting such an agent with an albumin fusion protein of the present invention or a fragment thereof and assaying for the presence of a complex between the agent and the albumin fusion protein or a fragment thereof, by methods well known in the art. In such a competitive binding assay, the agents to screen are typically labeled. Following incubation, free agent is separated from that present in bound form, and the amount of free or uncomplexed label is a measure of the ability of a particular agent to bind to the albumin fusion protein of the present invention. [0880]
  • Another technique for drug screening provides high throughput screening for compounds having suitable binding affinity to an albumin fusion protein of the present invention, and is described in great detail in European Patent Application 84/03564, published on Sep. 13, 1984, which is incorporated herein by reference herein. Briefly stated, large numbers of different small peptide test compounds are synthesized on a solid substrate, such as plastic pins or some other surface. The peptide test compounds are reacted with an albumin fusion protein of the present invention and washed. Bound peptides are then detected by methods well known in the art. Purified albumin fusion protein may be coated directly onto plates for use in the aforementioned drug screening techniques. In addition, non-neutralizing antibodies may be used to capture the peptide and immobilize it on the solid support. [0881]
  • This invention also contemplates the use of competitive drug screening assays in which neutralizing antibodies capable of binding an albumin fusion protein of the present invention specifically compete with a test compound for binding to the albumin fusion protein or fragments thereof. In this manner, the antibodies are used to detect the presence of any peptide which shares one or more antigenic epitopes with an albumin fusion protein of the invention. [0882]
  • Binding Peptides and Other Molecules [0883]
  • The invention also encompasses screening methods for identifying polypeptides and nonpolypeptides that bind albumin fusion proteins of the invention, and the binding molecules identified thereby. These binding molecules are useful, for example, as agonists and antagonists of the albumin fusion proteins of the invention. Such agonists and antagonists can be used, in accordance with the invention, in the therapeutic embodiments described in detail, below. [0884]
  • This method comprises the steps of: [0885]
  • contacting an albumin fusion protein of the invention with a plurality of molecules; and [0886]
  • identifying a molecule that binds the albumin fusion protein. [0887]
  • The step of contacting the albumin fusion protein of the invention with the plurality of molecules may be effected in a number of ways. For example, one may contemplate immobilizing the albumin fusion protein on a solid support and bringing a solution of the plurality of molecules in contact with the immobilized polypeptides. Such a procedure would be akin to an affinity chromatographic process, with the affinity matrix being comprised of the immobilized albumin fusion protein of the invention. The molecules having a selective affinity for the albumin fusion protein can then be purified by affinity selection. The nature of the solid support, process for attachment of the albumin fusion protein to the solid support, solvent, and conditions of the affinity isolation or selection are largely conventional and well known to those of ordinary skill in the art. [0888]
  • Alternatively, one may also separate a plurality of polypeptides into substantially separate fractions comprising a subset of or individual polypeptides. For instance, one can separate the plurality of polypeptides by gel electrophoresis, column chromatography, or like method known to those of ordinary skill for the separation of polypeptides. The individual polypeptides can also be produced by a transformed host cell in such a way as to be expressed on or about its outer surface (e.g., a recombinant phage). Individual isolates can then be “probed” by an albumin fusion protein of the invention, optionally in the presence of an inducer should one be required for expression, to determine if any selective affinity interaction takes place between the albumin fusion protein and the individual clone. Prior to contacting the albumin fusion protein with each fraction comprising individual polypeptides, the polypeptides could first be transferred to a solid support for additional convenience. Such a solid support may simply be a piece of filter membrane, such as one made of nitrocellulose or nylon. In this manner, positive clones could be identified from a collection of transformed host cells of an expression library, which harbor a DNA construct encoding a polypeptide having a selective affinity for an albumin fusion protein of the invention. Furthermore, the amino acid sequence of the polypeptide having a selective affinity for an albumin fusion protein of the invention can be determined directly by conventional means or the coding sequence of the DNA encoding the polypeptide can frequently be determined more conveniently. The primary sequence can then be deduced from the corresponding DNA sequence. If the amino acid sequence is to be determined from the polypeptide itself, one may use microsequencing techniques. The sequencing technique may include mass spectroscopy. [0889]
  • In certain situations, it may be desirable to wash away any unbound polypeptides from a mixture of an albumin fusion protein of the invention and the plurality of polypeptides prior to attempting to determine or to detect the presence of a selective affinity interaction. Such a wash step may be particularly desirable when the albumin fusion protein of the invention or the plurality of polypeptides are bound to a solid support. [0890]
  • The plurality of molecules provided according to this method may be provided by way of diversity libraries, such as random or combinatorial peptide or nonpeptide libraries which can be screened for molecules that specifically bind an albumin fusion protein of the invention. Many libraries are known in the art that can be used, e.g., chemically synthesized libraries, recombinant (e.g., phage display libraries), and in vitro translation-based libraries. Examples of chemically synthesized libraries are described in Fodor et al., Science 251:767-773 (1991); Houghten et al., Nature 354:84-86 (1991); Lam et al., Nature 354:82-84 (1991); Medynski, Bio/Technology 12:709-710 (1994); Gallop et al., J. Medicinal Chemistry 37(9):1233-1251 (1994); Ohlmeyer et al., Proc. Natl. Acad. Sci. USA 90:10922-10926 (1993); Erb et al., Proc. Natl. Acad. Sci. USA 91:11422-11426 (1994); Houghten et al., Biotechniques 13:412 (1992); Jayawickreme et al., Proc. Natl. Acad. Sci. USA 91:1614-1618 (1994); Salmon et al., Proc. Natl. Acad. Sci. USA 90:11708-11712 (1993); PCT Publication No. WO 93/20242; and Brenner and Lerner, Proc. Natl. Acad. Sci. USA 89:5381-5383 (1992). [0891]
  • Examples of phage display libraries are described in Scott et al., Science 249:386-390 (1990); Devlin et al., Science, 249:404-406 (1990); Christian et al., 1992, J. Mol. Biol. 227:711-718 1992); Lenstra, J. Immunol. Meth. 152:149-157 (1992); Kay et al., Gene 128:59-65 (1993); and PCT Publication No. WO 94/18318 dated Aug. 18, 1994. [0892]
  • In vitro translation-based libraries include but are not limited to those described in PCT Publication No. WO 91/05058 dated Apr. 18, 1991; and Mattheakis et al., Proc. Natl. Acad. Sci. USA 91:9022-9026 (1994). [0893]
  • By way of examples of nonpeptide libraries, a benzodiazepine library (see e.g., Bunin et al., Proc. Natl. Acad. Sci. USA 91:4708-4712 (1994)) can be adapted for use. Peptoid libraries (Simon et al., Proc. Natl. Acad. Sci. USA 89:9367-9371 (1992)) can also be used. Another example of a library that can be used, in which the amide functionalities in peptides have been permethylated to generate a chemically transformed combinatorial library, is described by Ostresh et al. (Proc. Natl. Acad. Sci. USA 91:11138-11142 (1994)). [0894]
  • The variety of non-peptide libraries that are useful in the present invention is great. For example, Ecker and Crooke (Bio/Technology 13:351-360 (1995) list benzodiazepines, hydantoins, piperazinediones, biphenyls, sugar analogs, beta-mercaptoketones, arylacetic acids, acylpiperidines, benzopyrans, cubanes, xanthines, aminimides, and oxazolones as among the chemical species that form the basis of various libraries. [0895]
  • Non-peptide libraries can be classified broadly into two types: decorated monomers and oligomers. Decorated monomer libraries employ a relatively simple scaffold structure upon which a variety functional groups is added. Often the scaffold will be a molecule with a known useful pharmacological activity. For example, the scaffold might be the benzodiazepine structure. [0896]
  • Non-peptide oligomer libraries utilize a large number of monomers that are assembled together in ways that create new shapes that depend on the order of the monomers. Among the monomer units that have been used are carbamates, pyrrolinones, and morpholinos. Peptoids, peptide-like oligomers in which the side chain is attached to the alpha amino group rather than the alpha carbon, form the basis of another version of non-peptide oligomer libraries. The first non-peptide oligomer libraries utilized a single type of monomer and thus contained a repeating backbone. Recent libraries have utilized more than one monomer, giving the libraries added flexibility. [0897]
  • Screening the libraries can be accomplished by any of a variety of commonly known methods. See, e.g., the following references, which disclose screening of peptide libraries: Parmley et al., Adv. Exp. Med. Biol. 251:215-218 (1989); Scott et al, Science 249:386-390 (1990); Fowlkes et al., BioTechniques 13:422-427 (1992); Oldenburg et al., Proc. Natl. Acad. Sci. USA 89:5393-5397 (1992); Yu et al., Cell 76:933-945 (1994); Staudt et al., Science 241:577-580 (1988); Bock et al., Nature 355:564-566 (1992); Tuerk et al., Proc. Natl. Acad. Sci. USA 89:6988-6992 (1992); Ellington et al., Nature 355:850-852 (1992); U.S. Pat. No. 5,096,815, U.S. Pat. No. 5,223,409, and U.S. Pat. No. 5,198,346, all to Ladner et al.; Rebar et al., Science 263:671-673 (1993); and PCT Publication No. WO 94/18318. [0898]
  • In a specific embodiment, screening to identify a molecule that binds an albumin fusion protein of the invention can be carried out by contacting the library members with an albumin fusion protein of the invention immobilized on a solid phase and harvesting those library members that bind to the albumin fusion protein. Examples of such screening methods, termed “panning” techniques are described by way of example in Parmley et al., Gene 73:305-318 (1988); Fowlkes et al., BioTechniques 13:422-427 (1992); PCT Publication No. WO 94/18318; and in references cited herein. [0899]
  • In another embodiment, the two-hybrid system for selecting interacting proteins in yeast (Fields et al., Nature 340:245-246 (1989); Chien et al., Proc. Natl. Acad. Sci. USA 88:9578-9582 (1991) can be used to identify molecules that specifically bind to polypeptides of the invention. [0900]
  • Where the binding molecule is a polypeptide, the polypeptide can be conveniently selected from any peptide library, including random peptide libraries, combinatorial peptide libraries, or biased peptide libraries. The term “biased” is used herein to mean that the method of generating the library is manipulated so as to restrict one or more parameters that govern the diversity of the resulting collection of molecules, in this case peptides. [0901]
  • Thus, a truly random peptide library would generate a collection of peptides in which the probability of finding a particular amino acid at a given position of the peptide is the same for all 20 amino acids. A bias can be introduced into the library, however, by specifying, for example, that a lysine occur every fifth amino acid or that positions 4, 8, and 9 of a decapeptide library be fixed to include only arginine. Clearly, many types of biases can be contemplated, and the present invention is not restricted to any particular bias. Furthermore, the present invention contemplates specific types of peptide libraries, such as phage displayed peptide libraries and those that utilize a DNA construct comprising a lambda phage vector with a DNA insert. [0902]
  • As mentioned above, in the case of a binding molecule that is a polypeptide, the polypeptide may have about 6 to less than about 60 amino acid residues, preferably about 6 to about 10 amino acid residues, and most preferably, about 6 to about 22 amino acids. In another embodiment, a binding polypeptide has in the range of 15-100 amino acids, or 20-50 amino acids. [0903]
  • The selected binding polypeptide can be obtained by chemical synthesis or recombinant expression. [0904]
  • Other Activities [0905]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention, may be employed in treatment for stimulating re-vascularization of ischemic tissues due to various disease conditions such as thrombosis, arteriosclerosis, and other cardiovascular conditions. The albumin fusion proteins of the invention and/or polynucleotides encoding albumin fusion proteins of the invention may also be employed to stimulate angiogenesis and limb regeneration, as discussed above. [0906]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed for treating wounds due to injuries, burns, post-operative tissue repair, and ulcers since they are mitogenic to various cells of different origins, such as fibroblast cells and skeletal muscle cells, and therefore, facilitate the repair or replacement of damaged or diseased tissue. [0907]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed stimulate neuronal growth and to treat and prevent neuronal damage which occurs in certain neuronal disorders or neuro-degenerative conditions such as Alzheimer's disease, Parkinson's disease, and AIDS-related complex. An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may have the ability to stimulate chondrocyte growth, therefore, they may be employed to enhance bone and periodontal regeneration and aid in tissue transplants or bone grafts. [0908]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be also be employed to prevent skin aging due to sunburn by stimulating keratinocyte growth. [0909]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed for preventing hair loss, since FGF family members activate hair-forming cells and promotes melanocyte growth. Along the same lines, an albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be employed to stimulate growth and differentiation of hematopoietic cells and bone marrow cells when used in combination with other cytokines. [0910]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed to maintain organs before transplantation or for supporting cell culture of primary tissues. An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be employed for inducing tissue of mesodermal origin to differentiate in early embryos. [0911]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also increase or decrease the differentiation or proliferation of embryonic stem cells, besides, as discussed above, hematopoietic lineage. An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be used to modulate mammalian characteristics, such as body height, weight, hair color, eye color, skin, percentage of adipose tissue, pigmentation, size, and shape (e.g., cosmetic surgery). Similarly, an albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be used to modulate mammalian metabolism affecting catabolism, anabolism, processing, utilization, and storage of energy. [0912]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may be used to change a mammal's mental state or physical state by influencing biorhythms, caricadic rhythms, depression (including depressive disorders), tendency for violence, tolerance for pain, reproductive capabilities (preferably by Activin or Inhibin-like activity), hormonal or endocrine levels, appetite, libido, memory, stress, or other cognitive qualities. [0913]
  • An albumin fusion protein of the invention and/or polynucleotide encoding an albumin fusion protein of the invention may also be used as a food additive or preservative, such as to increase or decrease storage capabilities, fat content, lipid, protein, carbohydrate, vitamins, minerals, cofactors or other nutritional components. [0914]
  • The above-recited applications have uses in a wide variety of hosts. Such hosts include, but are not limited to, human, murine, rabbit, goat, guinea pig, camel, horse, mouse, rat, hamster, pig, micro-pig, chicken, goat, cow, sheep, dog, cat, non-human primate, and human. In specific embodiments, the host is a mouse, rabbit, goat, guinea pig, chicken, rat, hamster, pig, sheep, dog or cat. In preferred embodiments, the host is a mammal. In most preferred embodiments, the host is a human. [0915]
  • Having generally described the invention, the same will be more readily understood by reference to the following examples, which are provided by way of illustration and are not intended as limiting. [0916]
  • Without further description, it is believed that one of ordinary skill in the art can, using the preceding description and the following illustrative examples, make and utilize the alterations detected in the present invention and practice the claimed methods. The following working examples therefore, specifically point out preferred embodiments of the present invention, and are not to be construed as limiting in any way the remainder of the disclosure. [0917]
  • EXAMPLES Example 1 Preparation of HA-hGH Fusion Proteins
  • An HA-hGH fusion protein was prepared as follows: [0918]
  • Cloning of hGH cDNA [0919]
  • The hGH cDNA was obtained from a human pituitary gland cDNA library (catalogue number HL1097v, Clontech Laboratories, Inc) by PCR amplification. Two oligonucleotides suitable for PCR amplification of the hGH cDNA, HGH1 and HGH2, were synthesized using an Applied Biosystems 380B Oligonucleotide Synthesizer. [0920]
    (SEQ ID NO: 1)
    HGH1: 5′-CCCAAGAATTCCCTTATCCAGGC-3′
    (SEQ ID NO: 2)
    HGH2: 5′-GGGAAGCTTAGAAGCCACAGGATCCCTCCACAG-3′
  • [0921] HGH 1 and HGH2 differed from the equivalent portion of the hGH cDNA sequence (Martial et. al., 1979) by two and three nucleotides, respectively, such that after PCR amplification an EcoRI site would be introduced to the 5′ end of the cDNA and a BamH1 site would be introduced into the 3′ end of the cDNA. In addition, HGH2 contained a HindIII site immediately downstream of the hGH sequence.
  • PCR amplification using a Perkin-Elmer-Cetus Thermal Cycler 9600 and a Perkin-Elmer-Cetus PCR kit, was performed using single-stranded DNA template isolated from the phage particles of the cDNA library as follows: 10 μL phage particles were lysed by the addition of 10 μL phage lysis buffer (280 μg/mL proteinase K in TE buffer) and incubation at 55° C. for 15 min followed by 85° C. for 15 min. After a 1 min. incubation on ice, phage debris was pelleted by centrifugation at 14,000 rpm for 3 min. The PCR mixture contained 6 μL of this DNA template, 0.1 μM of each primer and 200 μM of each deoxyribonucleotide. PCR was carried out for 30 cycles, denaturing at 94° C. for 30 s, annealing at 65° C. for 30 s and extending at 72° C. for 30 s, increasing the extension time by 1 s per cycle. [0922]
  • Analysis of the reaction by gel electrophoresis showed a single product of the expected size (589 base pairs). [0923]
  • The PCR product was purified using Wizard PCR Preps DNA Purification System (Promega Corp) and then digested with EcoRI and HindIII. After further purification of the EcoRI-HindIII fragment by gel electrophoresis, the product was cloned into pUC19 (GIBCO BRL) digested with EcoRI and HindIII, to give pHGH1. DNA sequencing of the EcoRI HindIII region showed that the PCR product was identical in sequence to the hGH sequence (Martial et al., 1979), except at the 5′ and 3′ ends, where the EcoRI and BamHI sites had been introduced, respectively. [0924]
  • Expression of the hGH cDNA. [0925]
  • The polylinker sequence of the phagemid pBluescribe (+) (Stratagene) was replaced by inserting an oligonucleotide linker, formed by annealing two 75-mer oligonucleotides, between the EcoRI and HindIII sites to form pBST(+). The new polylinker included a unique NotI site. [0926]
  • The NotI HA expression cassette of pAYE309 (EP 431 880) comprising the PRBI promoter, DNA encoding the HA/MF -I hybrid leader sequence, DNA encoding HA and the ADH1 terminator, was transferred to pBST(+) to form pHA1. The HA coding sequence was removed from this plasmid by digestion with HindIII followed by religation to form pHA2. [0927]
  • Cloning of the hGH cDNA, as described in Example 1, provided the hGH coding region lacking the pro-hGH sequence and the first 8 base pairs (bp) of the mature hGH sequence. In order to construct an expression plasmid for secretion of hGH from yeast, a yeast promoter, signal peptide and the first 8 bp of the hGH sequence were attached to the 5′ end of the cloned hGH sequence as follows: The HindIII-SfaNI fragment from [0928] pHA 1 was attached to the 5′ end of the EcoRI/HindIII fragment from pHGHI via two synthetic oligonucleotides, HGH3 and HGH4 (which can anneal to one another in such a way as to generate a double stranded fragment of DNA with sticky ends that can anneal with SfaNIand EcoRI sticky ends):
    HGH3: 5′-GATAAAGATTCCCAAC-3′ (SEQ ID NO: 3)
    HGH4: 5′-AATTGTTGGGAATCTTT-3′ (SEQ ID NO: 4)
  • The HindIII fragment so formed was cloned into HindIII-digested pHA2 to make pHGH2, such that the hGH cDNA was positioned downstream of the PRBI promoter and HA/MF -1 fusion leader sequence (WO 90/01063). The NotI expression cassette contained in pHGH2, which included the ADHI terminator downstream of the hGH cDNA, was cloned into NotI-digested pSAC35 (Sleep et al., BioTechnology 8:42 (1990)) to make pHGH12. This plasmid comprised the entire 2 μm plasmid to provide replication functions and the LEU2 gene for selection of transformants. [0929]
  • pHGH12 was introduced into [0930] S. cerevisiae D88 by transformation and individual transformants were grown for 3 days at 30° C. in 10 mL YEPD (1% w/v yeast extract, 2% w/v, peptone, 2% w/v, dextrose).
  • After centrifugation of the cells, the supernatants were examined by SDS-polyacrylamide gel electrophoresis (SDS-PAGE) and were found to contain protein which was of the expected size and which was recognized by anti-hGH antiserum (Sigma, Poole, UK) on Western blots. [0931]
  • Cloning and Expression of an HA-hGH Fusion Protein. [0932]
  • In order to fuse the HA cDNA to the 5′ end of the hGH cDNA, the pHA1 HindIII-Bsu361 fragment (containing most of the HA cDNA) was joined to the pHGH1 EcoRI-HindIII fragment (containing most of the hGH cDNA) via two oligonucleotides, HGH7 and HGH8 [0933]
    HGH7: 5′-TTAGGCTTATTCCCAAC 3′ (SEQ ID NO: 5)
    HGH8: 5′-AATTGTTGGGAATAAGCC 3′ (SEQ ID NO: 6)
  • The HindIII fragment so formed was cloned into pHA2 digested with HindIII to make pHGH10, and the NotI expression cassette of this plasmid was cloned into NotI-digested pSAC35 to make pHGH16. [0934]
  • pHGH16 was used to transform [0935] S. cerevisiae D88 and supernatants of cultures were analyzed as described above. A predominant band was observed that had a molecular weight of approximately 88 kD, corresponding to the combined masses of HA and hGH. Western blotting using anti-HA and anti-hGH antisera (Sigma) confirmed the presence of the two constituent parts of the albumin fusion protein.
  • The albumin fusion protein was purified from culture supernatant by cation exchange chromatography, followed by anion exchange and gel permeation chromatography. Analysis of the N-terminus of the protein by amino acid sequencing confirmed the presence of the expected albumin sequence. [0936]
  • An in vitro growth hormone activity assay (Ealey et al, Growth Regulation 5:36 (1995)) indicated that the albumin fusion protein possessed full hGH activity. In a hypophysectomised rat weight gain model, performed essentially as described in the European Pharmacopoeia (1987, monograph 556), the fusion molecule was more potent than hGH when the same number of units of activity (based on the above in vitro assay) were administered daily. Further experiments in which the albumin fusion protein was administered once every four days showed a similar overall growth response to a daily administration of hGH. Pharmacokinetic experiments in which [0937] 125I-labeled protein was administered to rats indicated an approximately ten-fold increase in circulatory half-life for the albumin fusion protein compared to hGH.
  • A similar plasmid was constructed in which DNA encoding the [0938] S. cerevisiae invertase (SUC2) leader sequence replaced the sequence for the hybrid leader, such that the encoded leader and the junction (↓) with the HA sequence were as follows:
    (SEQ ID NO: 7)
    . . . MLLQAFLFLLAGFAAKISA ↓ DAHKS . . .
    Invertase leader
                 HA sequence . . .
  • On introduction into [0939] S. cerevisiae DBI, this plasmid directed the expression and secretion of the albumin fusion protein at a level similar to that obtained with pHGH16. Analysis of the N-terminus of the albumin fusion protein indicated precise and efficient cleavage of the leader sequence from the mature protein.
  • Cloning and Expression of an hGH-HA Fusion Protein. [0940]
  • In order to fuse the hGH cDNA to the 5′ end of the HA cDNA, the HA cDNA was first altered by site-directed mutagenesis to introduce an EcoNI site near the 5′ end of the coding region. This was done by the method of Kunkel et al. (Methods in Enzymol. 154:367 (1987)) using single-stranded DNA template prepared from pHAI and a synthetic oligonucleotide, LEU4: [0941]
    LEU4: 5′-GAGATGCACACCTGAGTGAGG-3′ (SEQ ID NO: 8)
  • Site-directed mutagenesis using this oligonucleotide changed the coding sequence of the HA cDNA from Lys4 to Leu4 (K4L). However, this change was repaired when the hGH cDNA was subsequently joined at the 5′ end by linking the pHGH2 NotI-BamHI fragment to the EcoNI-NotI fragment of the mutated pHAI, via the two oligonucleotides HGH5 and HGH6: [0942]
    HGH5:
    5′-GATCCTGTGGCTTCGATGCACACAAGA-3′ (SEQ ID NO: 9)
    HGH6:
    5′-CTCTTGTGTGCATCGAAGCCACAG-3′ (SEQ ID NO: 10)
  • The NotI fragment so formed was cloned into NotI-digested pSAC35 to make pHGH14. pHGH14 was used to transform [0943] S. cerevisiae D88 and supernatants of culture were analyzed as above. A predominant band was observed that had a molecular weight of approximately 88 kD, corresponding to the combined masses of hGH and HA. Western blotting using anti-HA and anti-hGH antisera confirmed the presence of the two constituent parts of the albumin fusion protein.
  • The albumin fusion protein was purified from culture supernatant by cation exchange chromatography, followed by anion exchange and gel permeation chromatography. Analysis of the N-terminus of the protein by amino acid sequencing confirmed the presence of the expected hGH sequence. [0944]
  • In vitro studies showed that the albumin fusion protein retained hGH activity, but was significantly less potent than an albumin fusion protein comprising full length HA (1-585) as the N-terminal portion and hGH as the C-terminal portion, as described above. [0945]
  • Construction of Plasmids for the Expression of hGH Fusions to Domains of HA. [0946]
  • Fusion polypeptides were made in which the hGH molecule was fused to the first two domains of HA ([0947] residues 1 to 387). Fusion to the N terminus of hGH was achieved by joining the pHA1 HindIII-Sap1 fragment, which contained most of the coding sequence for domains 1 and 2 of HA, to the pHGHI EcoR1-HindIII fragment, via the oligonucleotides HGH 11 and HGH 12:
    (SEQ ID NO: 11)
    HGH11: 5′-TGTGGAAGAGCCTCAGAATTTATTCCCAAC-3′
    (SEQ ID NO: 12)
    HGH12: 5′-AATTGTTGGGAATAAATTCTGAGGCTCTTCC-3′
  • The HindIII fragment so formed was cloned into HindIII-digested pHA2 to make pHGH37 and the Not1 expression cassette of this plasmid was cloned into Not1-digested pSAC35. [0948]
  • The resulting plasmid, pHGH38, contained an expression cassette that was found to direct secretion of the fusion polypeptide into the supernatant when transformed into [0949] S. cerevisiae DB 1. Western blotting using anti-HA and anti-hGH antisera confirmed the presence of the two constituent parts of the albumin fusion protein.
  • The albumin fusion protein was purified from culture supernatant by cation exchange chromatography followed by gel permeation chromatography. [0950]
  • In vivo studies with purified protein indicated that the circulatory half-life was longer than that of hGH, and similar to that of an albumin fusion protein comprising full-length HA (1-585) as the N-terminal portion and hGH as the C-terminal portion, as described above. In vitro studies showed that the albumin fusion protein retained hGH activity. [0951]
  • Using a similar strategy as detailed above, an albumin fusion protein comprising the first domain of HA (residues 1-194) as the N-terminal portion and hGH as the C-terminal portion, was cloned and expressed in [0952] S. cerevisiae DBL. Western blotting of culture supernatant using anti-HA and anti-hGH antisera confirmed the presence of the two constituent parts of the albumin fusion protein.
  • Fusion of HA to hGH Using a Flexible Linker Sequence [0953]
  • Flexible linkers, comprising repeating units of [Gly-Gly-Gly-Gly-Ser][0954] n, where n was either 2 or 3, were introduced between the HA and hGH albumin fusion protein by cloning of the oligonucleotides HGH16, HGH17, HGH18 and HGH19:
    HGH16:
    5′-TTAGGCTTAGGTGGCGGTGGATCCGGCGGTG (SEQ ID NO: 13)
    GTGGATCTTTCCCA AC-3′
    HGH17:
    5′-AATTGTTGGGAAAGATCCACCACCGCCGGAT (SEQ ID NO: 14)
    CCACCGCCACCTAAGCC-3′
    HGH18:
    5′-TTAGGCTTAGGCGGTGGTGGATCTGGTGGCG (SEQ ID NO: 15)
    GCGGATCTGGTGGCGGTGGATCCTTCCCAAC-3′
    HGH19:
    5′-AATTGTTGGGAAGGATCCACCGCCACCAGAT (SEQ ID NO: 16)
    CCGCCGCCACCAGATCCACCACCGCCTAAGCC-
    3′
  • Annealing of HGH16 with HGH17 resulted in n=2, while HGH18 annealed to HGH19 resulted in n=3. After annealing, the double-stranded oligonucleotides were cloned with the EcoRI-Bsu361 fragment isolated from pHGH1 into Bsu361-digested pHGH10 to make pHGH56 (where n=2) and pHGH57 (where n=3). The NotI expression cassettes from these plasmids were cloned into NotI-digested pSAC35 to make pHGH58 and pHGH59, respectively. [0955]
  • Cloning of the oligonucleotides to make pHGH56 and pHGH57 introduced a BamHI site in the linker sequences. It was therefore possible to construct linker sequences in which n=1 and n=4, by joining either the HindIII-BamHI fragment from pHGH56 to the BamHI-HindIII fragment from pHGH57 (making n=1), or the HindIII-BamHI fragment from pHGH57 to the BamHI-HindIII fragment from pHGH56 (making n=2). Cloning of these fragments into the HindIII site of pHA2, resulted in pHGH60 (n=1) and pHGH61 (n=4). The NotI expression cassettes from pHGH60 and pHGH61 were cloned into NotI-digested pSAC35 to make pHGH62 and pHGH63, respectively. [0956]
  • Transformation of [0957] S. cerevisiae with pHGH58, pHGH59, pHGH62 and pHGH63 resulted in transformants that secreted the fusion polypeptides into the supernatant. Western blotting using anti-HA and anti-hGH antisera confirmed the presence of the two constituent parts of the albumin fusion proteins.
  • The albumin fusion proteins were purified from culture supernatant by cation exchange chromatography, followed by anion exchange and gel permeation chromatography. Analysis of the N-termini of the proteins by amino acid sequencing confirmed the presence of the expected albumin sequence. Analysis of the purified proteins by electrospray mass spectrometry confirmed an increase in mass of 315 D (n−1), 630 D (n=2), 945 D (n=3) and 1260 D (n=4) compared to the HA-hGH fusion protein described above. The purified protein was found to be active in vitro. [0958]
  • Increased Shelf-Life of HA-hGH Fusion Proteins: Methods [0959]
  • HA-hGH and hGH were separately diluted in cell culture media containing 5% horse serum to final concentrations of 100-200 μg/ml and incubated at 4, 37 or 50° C. At time zero and at weekly intervals thereafter, aliquots of the samples were tested for their biological activity in the Nb2 cell proliferation assay, and the data normalized to the biological activity of the control (hGH solution at time zero). In other assays hGH and HA-hGH were incubated in phosphate buffer saline in at 4, 37 and 50 degree C. [0960]
  • Nb2 cell proliferation assay: The growth of these cells is dependent on hGH or other lactogenic hormones. In a [0961] typical experiment 104 cells/well are plated in 96-well plate in the presence of different concentration of hGH or HA-hGH in media such as DMEM containing 5-10% horse serum for 24-48 hrs in the incubator. After the incubation period, 1:10 volume of MTT (5 mg/ml in H2O) is added to each well and the plate is incubated for a further 6-16 hrs. The growing cells convert MTT to insoluble formazan. The formazan is solublized by acidic isopropanol, and the color produced is measured at 570 nm on microtiter plate reader. The extent of formazan formation reflects the level of cellular proliferation.
  • Increased Shelf-Life of HA-hGH Fusion Proteins: Results [0962]
  • The fusion of Therapeutic proteins to albumin confers stability in aqueous or other solution. FIG. 1 depicts the extended shelf-life of an HA fusion protein in terms of the biological activity of HA-hGH remaining after storage in cell culture media for up to 5 weeks at 37° C. A solution of 200 μg/ml HA-hGH was prepared in tissue culture media containing 5% horse serum, and the solution incubated at 37° C. starting at time zero. At the indicated times, a sample was removed and tested for its biological activity in the Nb2 cell assay, at 2 ng/ml final concentration. As shown in FIG. 1, the biological activity of HA-hGH remains essentially intact (within experimental variation) after 5 weeks of incubation at 37° C. The recombinant hGH used as control for this experiment lost its biological activity in the first week of the experiment. [0963]
  • FIG. 2 shows the stability of HA-hGH after storage in cell culture media for up to 3 weeks at 4, 37, or 50° C. At time zero, a solution of HA-hGH was prepared in tissue culture media containing 5% horse serum, and incubated at 4, 37, and 50° C. At the indicated periods a sample was removed and assayed for its biological activity in the Nb2 cell proliferation assay, at 60 ng/ml final concentration. HA-hGH retains over 90% of its initial activity at all temperatures tested for at least 3 weeks after incubation while hGH loses its biological activity within the first week. This level of activity is further retained for at least 7 weeks at 37° C. and 5 weeks at 50° C. These results indicate that HA-hGH is highly stable in aqueous solution even under temperature stress. [0964]
  • FIGS. 3A and 3B show the stable biological activity of HA-hGH compared to hGH in the Nb2 cell proliferation assay. Nb2 cells were grown in the presence of increasing concentrations of recombinant hGH or HA-hGH, added at time zero. The cells were incubated for 24 or 48 hours before measuring the extent of proliferation by the MTT method. The increased stability of HA-hGH in the assay results in essentially the same proliferative activity at 24 hours (FIG. 3A) as at 48 hours (FIG. 3B) while hGH shows a significant reduction in its proliferative activity after 48 hours of incubation (FIGS. 3A and 3B). Compared to hGH, the HA-hGH has lower biological potency after 1 day; the albumin fusion protein is about 5 fold less potent than hGH. However, after 2 days the HA-hGH shows essentially the same potency as hGH due to the short life of hGH in the assay. This increase in the stability of the hGH as an albumin fusion protein has a major unexpected impact on the biological activity of the protein. Although the potency of the albumin fusion proteins is slightly lower than the unfused counterparts in rapid bioassays, their biological stability results in much higher biological activity in the longer term in vitro assay or in vivo assays. [0965]
  • Example 2 Preparation of HA-Fusion Proteins
  • FIG. 4 shows a map of a plasmid (pPPC0005) that can be used as the base vector for cloning the cDNAs of therapeutic partners to form HA-fusions. For example, digestion of this vector with the restriction enzymes Bsu36I/Partial HindIII will allow for the insertion of a cDNA modified at the 5′ end to encode the last 5 amino acids of HA including the Bsu36I site and at the 3′ end to include a double stop codon and HindIII site. As another example, digestion of this vector with the restriction enzymes Bsu3611, SphI allows for the insertion of a cDNA modified at the 5′ end to encode the last 5 amino acids of HA including the Bsu36I site and at the 3′ end to include a double stop codon, HindIII site and the ADHI terminator sequence up to and including the SphI site. [0966]
  • This plasmid may easily be modified by one of skill in the art, for example, to modify, add or delete restriction sites so that one may more easily clone a Therapeutic protein, or fragment or variant of into the vector for the purpose of making an albumin fusion protein of the invention. [0967]
  • For example, for the purpose of making an albumin fusion protein where the Therapeutic moiety is placed N-terminal to the (mature) albumin protein, restriction sites were added at the 5′ end of the DNA encoding HA in pPPC0005 shown in FIG. 4). [0968]
  • Because it was desired to add unique XhoI and ClaI sites at the 5′ end of the DNA encoding the HA protein in pPPC0005, it was first necessary to remove those same sites from the plasmid (located 3′ of the ADH1 terminator sequence). This was accomplished by cutting pPPC0005 with XhoI and ClaI, filling in the sticky ends with T4 DNA polymerase, and religating the blunt ends to create pPPC0006 [0969]
  • Engineering the Xho and Cla I restriction sites into the Fusion leader sequence just 5′ of the DNA encoding the HA protein in pPPC0006 was accomplished using two rounds of PCR. The first pair of oligonucleotides are those of SEQ ID NO:19 and SEQ ID NO:20. SEQ ID 19 contains four point mutations relative to the DNA sequence encoding the Fusion leaadr sequence and the beginning of the HA protein. These mutations are necessary to create the XhoI site in the fusion leader sequence and the Cla I site just at the beginning of the DNA encoding the HA protein. These four mutations are underlined in the sequence shown below. In pPPC0006 the nucleotides at these four positions from 5′ to 3′ are T, G, T, and G. 5′-GCCTCGAGAAAAGAGATGCACACAAGAGTGAGGTTGCTCATCGATTTAAAG ATTTGGG-3′ (SEQ ID NO:19) 5′-AATCGATGAGCAACCTCACTCTTGTGTGCATCTCTTTTCTCGAGGCTCCTGG AATAAGC-3′ (SEQ ID NO:20). A second round of PCR is then performed with an upstream flanking primer, 5′-TACAAACTTAAGAGTCCAATTAGC-3′ (SEQ ID NO:21) and a [0970] downstream flanking primer 5′-CACTTCTCTAGAGTGGTTTCATATGTCTT-3′ (SEQ ID NO:22). The resulting PCR product is then purified and then digested with AflI and XbaI and ligated into the same sites in pPPC0006 creating pScCHSA. The resulting plasmid will have an XhoI sites engineered into the fusion leader sequence. The presence of the XhoI site creates a single amino acid change in the end of fusion leader sequence from LDKR to LEKR. The D to E change will not be present in the final albumin fusion protein expression plasmid if one ligates into the XhoI and Cla I sites a fragment comprising the Therapeutic moiety which has a 5′ SalI sticky end (which is compatible with the XhoI end) and a 3′ClaI end. Ligation of the XhoI to the SalI restores the original amino acid sequence of the Fusion leader sequence. The therapeutic protein moiety may be inserted after the Kex2 site (Kex2 claeves after the dibasic amino acid sequence KR at the end of the Fusion leader sequence) and before the ClaI site.
  • In addition, for the purpose of making an albumin fusion protein where the Therapeutic moiety is placed C-terminal to the (mature) albumin protein, four, eight-base-pair restriction sites were added at the 3′ end of the DNA encoding HA in pScCHSA. As an example, it was felt to be desirable to incorporate AscI, FseI, and PmeI restriction sites in between the Bsu36I and HindIII sites at the end of the DNA encoding the HA protein in pScCHSA. This was accomplished through the use of two complementary synthetic oligonucleotides (SEQ ID NO:19 and SEQ ID NO:20) which contain the desired restriction sites. 5′-AAGCTGCCTTAGGCTTATAATAAGGCGCGCCGGCCGGCCGTTTAAACTAAG CTTAATTCT-3′ (SEQ ID NO:23) and 5-AGAATTAAGCTTAGTTTAAACGGCCGGCCGGCGCGCCTTATTATAAGCCTAA GGCAGCTT-3′ (SEQ ID NO:24). These oligonucleotides may be annealed and digested with Bsu36I and HindIII and ligated into the same sites located at the end of the DNA encoding the HA protein in pScCHSA creating pScNHSA, using techniques known in the art. [0971]
  • Making Vectors Comprising Albumin Fusion Proteins Where the Albumin Moiety is N-Terminal to the Therapeutic Moiety. [0972]
  • The DNA encoding the Therapeutic moiety may be PCR amplified using primers that will add DNA encoding the last five amino acids of the HA (and containing the Bsu36I site) onto the 5′ end of the DNA encoding a Therapeutic protein and a STOP codon and appropriate cloning sites onto the 3′ end of the coding sequence. For instance, the forward primer used to amplify the DNA encoding a therapeutic protein might have the sequence, 5′-aagctGCCTTAGGCTTA(N)[0973] 15-3′ (SEQ ID NO:25) where the underlined sequence is a Bsu36I site, the upper case nucleotides encode the last four amino acids of the mature HA protein (ALGL), and (N)15 is identical to the first 15 nucleotides encoding the Therapetic protein of interest. Similarly, the reverse primer used to amplify the DNA encoding a therapeutic protein might have the sequence, 5′-GCGCGCGTTTAAACGGCCGGCCGGCGCGCCTTATTA(N)15-3′ (SEQ ID NO:26) where the italicized nucleotides is a PmeI site, the double underlined nucleotides are a FseI site, the singly underlined text is a PmeI site, the boxed nucleotides are the reverse complement of two tandem stop codons, and (N)15 is identical to the reverse complement of the last 15 nucleotides encoding the Therapeutic protein of interest. Once the PCR product is amplified it may be cut with Bsu36I and one of (AscI, FseI, or PmeI) and ligated into pScNHSA.
  • Making Vectors Comprising Albumin Fusion Proteins Where the Albumin Moiety is N-Terminal to the Therapeutic Moiety. [0974]
  • The DNA encoding the Therapeutic moiety may be PCR amplified using primers that will add DNA encoding the last three amino acids of the Fusion leader sequence (and containing a SalI site) onto the 5′ end of the DNA encoding a Therapeutic protein and the first few amino acids of the HA (and containing a ClaI site. For instance, the forward primer used to amplify the DNA encoding a therapeutic protein might have the sequence, 5′-aggagcgtcGACAAAAGA(N)[0975] 15-3′ (SEQ ID NO:27) where the underlined sequence is a Sal I site, the upper case nucleotides encode the last three amino acids of the Fusion leader sequence (DKR), and (N)15 is identical to the first 15 nucleotides encoding the Therapetic protein of interest. Similarly, the reverse primer used to amplify the DNA encoding a therapeutic protein might have the sequence, 5′-CTTTAAATCGATGAGCAACCTCACTCTTGTGTGCATC(N)15-3′ (SEQ ID NO:28) where the italicized nucleotides are a ClaI site, the underlined nucleotides are the reverse complement of the DNA encoding the first 9 amino acids of HA (DAHKSEVAH), and (N)15 is identical to the reverse complement of the last 15 nucleotides encoding the Therapeutic protein of interest. Once the PCR product is amplified it may be cut with SalI and ClaI and ligated into pScCHSA digested with XhoI and Cla I.
  • Expression of an Albumin Fusion Protein in Yeast. [0976]
  • The Not I fragment containing the DNA encoding either an N-terminal or C-terminal albumin fusion protein generated from pScCHSA or pScNHSA may then be cloned in to the NotI site of pSAC35. [0977]
  • Expression of an Albumin Fusion Protein from Mammalian Cell Lines [0978]
  • The HSA gene has also been cloned into a the pC4 vector which is more suitable for mammalian culture systems creating plasmid pC4:HSA. More specifically, pC4HSA was generated by PCR amplifying the mature HSA gene with a 5′ primer (SEQ ID NO:30) that anneals to the 5′ end of DNA encoding the mature form of the HSA protein (e.g, DNA in plasmid pScCHSA),incorporates BamHI (Shown in italics below) and HindIII (shown singly underlined below) cloning sites, attaches a kozak sequence (shown double underlined below) and DNA encoding the natural HSA signal peptide (MKWVSFISLLFLFSSAYSRSLDKR, SEQ ID NO:29) (shown in bold below), and a 3′ primer (SEQ ID NO:31) that anneals to the 3′ end of DNA encoding the mature form of the HSA protein and incorporates an Asp718 restriction site (shown in bold below). The DNA encoding the natural human serum albumin leader sequence in SEQ ID NO:30 also contains a modification that introduces a XhoI site that is boxed below. [0979]
    (SEQ ID NO: 30)
    5′-TCAGGGATCC AAGCTT CCGCCACCATGAAGTGGGTAACCTTTATTTC
    CCTTCTTTTTCTCTTTAGCTCGGCTTA |CTCGAG| GGGTGTGTTTCGTCG
    AGATGCACACAAGAGTGAG-3′
    (SEQ ID NO: 31)
    5″-GCAGCGGTACCGAATTCGGCGCGCCTTATAAGCCTAAGGCAGC-3′
  • This PCR product (1.85 kb) is then purified and digested with BamHI and Asp718 and cloned into the same sites in pC4 (ATCC Accession No. 209646) to produce pC4:HSA [0980]
  • Making Vectors Comprising Albumin Fusion Proteins Where the Albumin Moiety is C-Terminal to the Therapeutic Moiety Using the pC4:HSA Vector [0981]
  • Using pC4:HSA, albumin fusion proteins in which the Therapeutic protein moiety is N terminal to the albumin sequence, one can clone DNA encoding a Therapeutic protein that has its own signal sequence between the Bam HI (or HindIII) and ClaI sites. When cloning into either the BamHI or Hind III site remember to include Kozak sequence (CCGCCACCATG) prior to translational start codon of DNA encoding the Therapeutic Protein to be subcloned. If the Therapeutic does not have a signal sequence, the DNA encoding that Therapeutic protein may be cloned in between the XhoI and ClaI sites. When using the XhoI site, the following 5′ (SEQ ID NO:32) and 3′ (SEQ ID NO:33) PCR primers may be used: [0982]
    (SEQ ID NO: 32)
    5′-CCGCCGCTCGAGGGGTGTGTTTCGTCGA(N)18-3′
    (SEQ ID NO: 33)
    5′-AGTCCCATCGATGAGCAACCTCACTCTTGTGTGCATC(N)18-3′
  • In SEQ ID NO:32, the underlined sequence is an XhoI site; and the XhoI site and the DNA following the XhoI site encode for the last seven amino acids of the leader sequence of natural human serum albumin. In SEQ ID NO:33, the underlined sequence is a ClaI site; and the ClaI site and the DNA following it encode are the reverse complement of the DNA encoding the first 10 amino acids of themature HSA protein (SEQ ID NO:18). In SEQ ID NO:32 “(N)[0983] 18” is DNA identical to the first 18 nucleotides encoding the Therapeutic protein of interest.). In SEQ ID NO:33 “(N)18” is the reverse complement of DNA encoding the last 18 nucleotides encoding the Therapeutic protein of interest. Using these two primers, one may PCR amplify the Therapeutic protein of interest, purify the PCR product, digest it with XhoI and ClaI restriction enzymes and then and clone it into the with XhoI and ClaI sites in the pC4:HSA vector.
  • Making Vectors Comprising Albumin Fusion Proteins Where the Albumin Moiety is N-Terminal to the Therapeutic Moiety Using the pC4:HSA Vector [0984]
  • Using pC4:HSA, albumin fusion proteins in which the Therapeutic protein moiety is N terminal to the albumin sequence, one can clone DNA encoding a Therapeutic protein between the Bsu36I and AscI restriction sites. When cloning into the Bsu36I and AscI, the same primer design used to clone in the yeast vector system (SEQ ID NO:25 and 26) may be employed. [0985]
  • The pC4 vector is especially suitable for expression of albumin fusion proteins from CHO cells. For expression, in other mammalian cell types, e.g., NSO cells, it may be useful to subclone the HindIII-EcoRI fragment containing the DNA encoding an albumin fusion protein (from a pC4 vector in which the DNA encoding the Therapeutic protein has already been cloned in frame with the DNA encoding (the mature form of) human serum albumin) into another expression vector (such as any of the mammalian expression vectors described herein). [0986]
  • Example 3 Preparation of HA-Cytokine or HA-Growth Factor Fusion Proteins (such as EPO, GMCSF, GCSF)
  • The cDNA for the cytokine or growth factor of interest, such as EPO, can be isolated by a variety of means including from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins are known and available, for instance, in U.S. Pat. Nos. 4,703,008, 4,810,643 and 5,908,763. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. EPO (or other cytokine) cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines, a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [0987]
  • Example 4 Preparation of HA-IFN Fusion Proteins (such as IFNα)
  • The cDNA for the interferon of interest such as IFNα can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for interferons, such as IFNα are known and available, for instance, in U.S. Pat. Nos. 5,326,859 and 4,588,585, in EP 32 134, as well as in public databases such as GenBank. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used to clone the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus of the HA sequence, with or without the use of a spacer sequence. The IFNα (or other interferon) cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast (see FIG. 8). The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [0988]
  • Maximum Protein Recovery from Vials [0989]
  • The albumin fusion proteins of the invention have a high degree of stability even when they are packaged at low concentrations. In addition, in spite of the low protein concentration, good fusion-protein recovery is observed even when the aqueous solution includes no other protein added to minimize binding to the vial walls. FIG. 5 compares the recovery of vial-stored HA-IFN solutions with a stock solution. 6 or 30 μg/ml HA-IFN solutions were placed in vials and stored at 4° C. After 48 or 72 hrs a volume originally equivalent to 10 ng of sample was removed and measured in an IFN sandwich ELISA. The estimated values were compared to that of a high concentration stock solution. As shown, there is essentially no loss of the sample in these vials, indicating that addition of exogenous material such as albumin is not necessary to prevent sample loss to the wall of the vials [0990]
  • In vivo Stability and Bioavailability of HA-α-IFN Fusions [0991]
  • To determine the in vivo stability and bioavailability of a HA-α-IFN fusion molecule, the purified fusion molecule (from yeast) was administered to monkeys at the dosages and time points described in FIGS. 6 and 7. Pharmaceutical compositions formulated from HA-α-IFN fusions may account for the extended serum half-life and bioavailability exemplified in FIGS. 6 and 7. Accordingly, pharmaceutical compositions may be formulated to contain lower dosages of alpha-interferon activity compared to the native alpha-interferon molecule. [0992]
  • Pharmaceutical compositions containing HA-α-IFN fusions may be used to treat or prevent disease in patients with any disease or disease state that can be modulated by the administration of α-IFN. Such diseases include, but are not limited to, hairy cell leukemia, Kaposi's sarcoma, genital and anal warts, chronic hepatitis B, chronic non-A, non-B hepatitis, in particular hepatitis C, hepatitis D, chronic myelogenous leukemia, renal cell carcinoma, bladder carcinoma, ovarian and cervical carcinoma, skin cancers, recurrent respirator papillomatosis, non-Hodgkin's and cutaneous T-cell lymphomas, melanoma, multiple myeloma, A/DS, multiple sclerosis, gliobastoma, etc. (see Interferon Alpha, In: AHFS Drug Information, 1997. [0993]
  • Accordingly, the invention includes pharmaceutical compositions containing a HA-α-IFN fusion protein, polypeptide or peptide formulated with the proper dosage for human administration. The invention also includes methods of treating patients in need of such treatment comprising at least the step of administering a pharmaceutical composition containing at least one HA-α-IFN fusion protein, polypeptide or peptide. [0994]
  • Bifunctional HA- -IFN Fusions [0995]
  • The HA-α-IFN expression vector of FIG. 8 is modified to include an insertion for the expression of bifunctional HA-α-IFN fusion proteins. For instance, the cDNA for a second protein of interest may be inserted in frame downstream of the “rHA-IFN” sequence after the double stop codon has been removed or shifted downstream of the coding sequence. [0996]
  • In one version of a bifunctional HA-α-IFN fusion protein, an antibody or fragment against B-lymphocyte stimulator protein (GenBank Acc 4455139) or polypeptide may be fused to one end of the HA component of the fusion molecule. This bifunctional protein is useful for modulating any immune response generated by the α-IFN component of the fusion. [0997]
  • Example 5 Preparation of HA-Hormone Fusion Protein (such as Insulin, LH, FSH)
  • The cDNA for the hormone of interest such as insulin can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins are known and available, for instance, in public databases such as GenBank. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The hormone cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [0998]
  • Example 6 Preparation of HA-Soluble Receptor or HA-Binding Protein Fusion Protein such as HA-TNF Receptor
  • The cDNA for the soluble receptor or binding protein of interest such as TNF receptor can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for all of these proteins are known and available, for instance, in GenBank. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The receptor cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [0999]
  • Example 7 Preparation of HA-Growth Factors such as HA-IGF-1 Fusion Protein
  • The cDNA for the growth factor of interest such as IGF-1 can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods (see GenBank Acc. No.NP[1000] 000609). The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The growth factor cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines.
  • Example 8 Preparation of HA-Single Chain Antibody Fusion Proteins
  • Single chain antibodies are produced by several methods including but not limited to: selection from phage libraries, cloning of the variable region of a specific antibody by cloning the cDNA of the antibody and using the flanking constant regions as the primer to clone the variable region, or by synthesizing an oligonucleotide corresponding to the variable region of any specific antibody. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The cell cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. [1001]
  • In fusion molecules of the invention, the V[1002] H and VL can be linked by one of the following means or a combination thereof: a peptide linker between the C-terminus of the VH and the N-terminus of the VL;; a Kex2p protease cleavage site between the VH and VL such that the two are cleaved apart upon secretion and then self associate; and cystine residues positioned such that the VH and VL can form a disulphide bond between them to link them together (see FIG. 14). An alternative option would be to place the VH at the N-terminus of HA or an HA domain fragment and the VL at the C-terminus of the HA or HA domain fragment.
  • The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. The antibody produced in this manner can be purified from media and tested for its binding to its antigen using standard immunochemical methods. [1003]
  • Example 9 Preparation of HA-Cell Adhesion Molecule Fusion Proteins
  • The cDNA for the cell adhesion molecule of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The nucleotide sequences for the known cell adhesion molecules are known and available, for instance, in GenBank. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The cell adhesion molecule cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [1004]
  • Example 10 Preparation of Inhibitory Factors and Peptides as HA Fusion Proteins (such as HA-Antiviral, HA-Antibiotic, HA-Enzyme Inhibitor and HA-Anti-Allergic Proteins)
  • The cDNA for the peptide of interest such as an antibiotic peptide can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The peptide cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [1005]
  • Example 11 Preparation of Targeted HA Fusion Proteins
  • The cDNA for the protein of interest can be isolated from cDNA library or can be made synthetically using several overlapping oligonucleotides using standard molecular biology methods. The appropriate nucleotides can be engineered in the cDNA to form convenient restriction sites and also allow the attachment of the protein cDNA to albumin cDNA similar to the method described for hGH. Also a targeting protein or peptide cDNA such as single chain antibody or peptides, such as nuclear localization signals, that can direct proteins inside the cells can be fused to the other end of albumin. The protein of interest and the targeting peptide is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA which allows the fusion with albumin cDNA. In this manner both N- and C-terminal end of albumin are fused to other proteins. The fused cDNA is then excised from pPPC0005 and is inserted into a plasmid such as pSAC35 to allow the expression of the albumin fusion protein in yeast. All the above procedures can be performed using standard methods in molecular biology. The albumin fusion protein secreted from yeast can be collected and purified from the media and tested for its biological activity and its targeting activity using appropriate biochemical and biological tests. [1006]
  • Example 12 Preparation of HA-Enzymes Fusions
  • The cDNA for the enzyme of interest can be isolated by a variety of means including but not exclusively, from cDNA libraries, by RT-PCR and by PCR using a series of overlapping synthetic oligonucleotide primers, all using standard methods. The cDNA can be tailored at the 5′ and 3′ ends to generate restriction sites, such that oligonucleotide linkers can be used, for cloning of the cDNA into a vector containing the cDNA for HA. This can be at the N or C-terminus with or without the use of a spacer sequence. The enzyme cDNA is cloned into a vector such as pPPC0005 (FIG. 4), pScCHSA, pScNHSA, or pC4:HSA from which the complete expression cassette is then excised and inserted into the plasmid pSAC35 to allow the expression of the albumin fusion protein in yeast. The albumin fusion protein secreted from the yeast can then be collected and purified from the media and tested for its biological activity. For expression in mammalian cell lines a similar procedure is adopted except that the expression cassette used employs a mammalian promoter, leader sequence and terminator (See Example 2). This expression cassette is then excised and inserted into a plasmid suitable for the transfection of mammalian cell lines. [1007]
  • Example 13 Bacterial Expression of an Albumin Fusion Protein
  • A polynucleotide encoding an albumin fusion protein of the present invention comprising a bacterial signal sequence is amplified using PCR oligonucleotide primers corresponding to the 5′ and 3′ ends of the DNA sequence, to synthesize insertion fragments. The primers used to amplify the polynucleotide encoding insert should preferably contain restriction sites, such as BamHI and XbaI, at the 5′ end of the primers in order to clone the amplified product into the expression vector. For example, BamHI and XbaI correspond to the restriction enzyme sites on the bacterial expression vector pQE-9. (Qiagen, Inc., Chatsworth, Calif.). This plasmid vector encodes antibiotic resistance (Amp[1008] r), a bacterial origin of replication (ori), an IPTG-regulatable promoter/operator (P/O), a ribosome binding site (RBS), a 6-histidine tag (6-His), and restriction enzyme cloning sites.
  • The pQE-9 vector is digested with BamHI and XbaI and the amplified fragment is ligated into the pQE-9 vector maintaining the reading frame initiated at the bacterial RBS. The ligation mixture is then used to transform the [1009] E. coli strain M15/rep4 (Qiagen, Inc.) which contains multiple copies of the plasmid pREP4, which expresses the lacI repressor and also confers kanamycin resistance (Kanr). Transformants are identified by their ability to grow on LB plates and ampicillin/kanamycin resistant colonies are selected. Plasmid DNA is isolated and confirmed by restriction analysis.
  • Clones containing the desired constructs are grown overnight (O/N) in liquid culture in LB media supplemented with both Amp (100 ug/ml) and Kan (25 ug/ml). The O/N culture is used to inoculate a large culture at a ratio of 1:100 to 1:250. The cells are grown to an optical density 600 (O.D.[1010] 600) of between 0.4 and 0.6. IPTG (Isopropyl-B-D-thiogalacto pyranoside) is then added to a final concentration of 1 mM. IPTG induces by inactivating the lacI repressor, clearing the P/O leading to increased gene expression.
  • Cells are grown for an extra 3 to 4 hours. Cells are then harvested by centrifugation (20 mins at 6000×g). The cell pellet is solubilized in the [1011] chaotropic agent 6 Molar Guanidine HCl or preferably in 8 M urea and concentrations greater than 0.14 M 2-mercaptoethanol by stirring for 3-4 hours at 4° C. (see, e.g., Burton et al., Eur. J. Biochem. 179:379-387 (1989)). The cell debris is removed by centrifugation, and the supernatant containing the polypeptide is loaded onto a nickel-nitrilo-tri-acetic acid (“Ni-NTA”) affinity resin column (available from QIAGEN, Inc., supra). Proteins with a 6×His tag bind to the Ni-NTA resin with high affinity and can be purified in a simple one-step procedure (for details see: The QIAexpressionist (1995) QIAGEN, Inc., supra).
  • Briefly, the supernatant is loaded onto the column in 6 M guanidine-HCl, [1012] pH 8. The column is first washed with 10 volumes of 6 M guanidine-HCl, pH 8, then washed with 10 volumes of 6 M guanidine-HCl pH 6, and finally the polypeptide is eluted with 6 M guanidine-HCl, pH 5.
  • The purified protein is then renatured by dialyzing it against phosphate-buffered saline (PBS) or 50 mM Na-acetate, [1013] pH 6 buffer plus 200 mM NaCl. Alternatively, the protein can be successfully refolded while immobilized on the Ni-NTA column. Exemplary conditions are as follows: renature using a linear 6M-1M urea gradient in 500 mM NaCl, 20% glycerol, 20 mM Tris/HCl pH 7.4, containing protease inhibitors. The renaturation should be performed over a period of 1.5 hours or more. After renaturation the proteins are eluted by the addition of 250 mM immidazole. Immidazole is removed by a final dialyzing step against PBS or 50 mM sodium acetate pH 6 buffer plus 200 mM NaCl. The purified protein is stored at 4° C. or frozen at −80 C.
  • In addition to the above expression vector, the present invention further includes an expression vector, called pHE4a (ATCC Accession Number 209645, deposited on Feb. 25, 1998) which contains phage operator and promoter elements operatively linked to a polynucleotide encoding an albumin fusion protein of the present invention, called pHE4a. (ATCC Accession Number 209645, deposited on Feb. 25, 1998.) This vector contains: 1) a neomycinphosphotransferase gene as a selection marker, 2) an [1014] E. coli origin of replication, 3) a T5 phage promoter sequence, 4) two lac operator sequences, 5) a Shine-Delgarno sequence, and 6) the lactose operon repressor gene (lacIq). The origin of replication (oriC) is derived from pUC19 (LTI, Gaithersburg, Md.). The promoter and operator sequences are made synthetically.
  • DNA can be inserted into the pHE4a by restricting the vector with NdeI and XbaI, BamHI, XhoI, or Asp718, running the restricted product on a gel, and isolating the larger fragment (the stuffer fragment should be about 310 base pairs). The DNA insert is generated according to PCR protocols described herein or otherwise known in the art, using PCR primers having restriction sites for NdeI (5′ primer) and XbaI, BamHI, XhoI, or Asp718 (3′ primer). The PCR insert is gel purified and restricted with compatible enzymes. The insert and vector are ligated according to standard protocols. [1015]
  • The engineered vector may be substituted in the above protocol to express protein in a bacterial system. [1016]
  • Example 14 Expression of an Albumin Fusion Protein in Mammalian Cells
  • The albumin fusion proteins of the present invention can be expressed in a mammalian cell. A typical mammalian expression vector contains a promoter element, which mediates the initiation of transcription of mRNA, a protein coding sequence, and signals required for the termination of transcription and polyadenylation of the transcript. Additional elements include enhancers, Kozak sequences and intervening sequences flanked by donor and acceptor sites for RNA splicing. Highly efficient transcription is achieved with the early and late promoters from SV40, the long terminal repeats (LTRs) from Retroviruses, e.g., RSV, HTLVI, HIVI and the early promoter of the cytomegalovirus (CMV). However, cellular elements can also be used (e.g., the human actin promoter). [1017]
  • Suitable expression vectors for use in practicing the present invention include, for example, vectors such as, pSVL and pMSG (Pharmacia, Uppsala, Sweden), pRSVcat (ATCC 37152), pSV2dhfr (ATCC 37146), pBC12MI (ATCC 67109), pCMVSport 2.0, and pCMVSport 3.0. Mammalian host cells that could be used include, but are not limited to, human Hela, 293, H9 and Jurkat cells, mouse NIH3T3 and C127 cells, [1018] Cos 1, Cos 7 and CV1, quail QC1-3 cells, mouse L cells and Chinese hamster ovary (CHO) cells.
  • Alternatively, the albumin fusion protein can be expressed in stable cell lines containing the polynucleotide encoding the albumin fusion protein integrated into a chromosome. The co-transfection with a selectable marker such as DHFR, gpt, neomycin, or hygromycin allows the identification and isolation of the transfected cells. [1019]
  • The transfected polynucleotide encoding the fusion protein can also be amplified to express large amounts of the encoded fusion protein. The DHFR (dihydrofolate reductase) marker is useful in developing cell lines that carry several hundred or even several thousand copies of the gene of interest. (See, e.g., Alt et al., J. Biol. Chem. 253:1357-1370 (1978); Hamlin et al., Biochem. et Biophys. Acta, 1097:107-143 (1990); Page et al., Biotechnology 9:64-68 (1991)). Another useful selection marker is the enzyme glutamine synthase (GS) (Murphy et al., Biochem J. 227:277-279 (1991); Bebbington et al., Bio/Technology 10:169-175 (1992). Using these markers, the mammalian cells are grown in selective medium and the cells with the highest resistance are selected. These cell lines contain the amplified gene(s) integrated into a chromosome. Chinese hamster ovary (CHO) and NSO cells are often used for the production of proteins. [1020]
  • Derivatives of the plasmid pSV2-dhfr (ATCC Accession No. 37146), the expression vectors pC4 (ATCC Accession No. 209646) and pC6 (ATCC Accession No.209647) contain the strong promoter (LTR) of the Rous Sarcoma Virus (Cullen et al., Molecular and Cellular Biology, 438-447 (March, 1985)) plus a fragment of the CMV-enhancer (Boshart et al., Cell 41:521-530 (1985)). Multiple cloning sites, e.g., with the restriction enzyme cleavage sites BamHI, XbaI and Asp718, facilitate the cloning of the gene of interest. The vectors also contain the 3′ intron, the polyadenylation and termination signal of the rat preproinsulin gene, and the mouse DHFR gene under control of the SV40 early promoter. [1021]
  • Specifically, the plasmid pC6, for example, is digested with appropriate restriction enzymes and then dephosphorylated using calf intestinal phosphates by procedures known in the art. The vector is then isolated from a 1% agarose gel. [1022]
  • A polynucleotide encoding an albumin fusion protein of the present invention is generated using techniques known in the art and this polynucleotide is amplified using PCR technology known in the art. If a naturally occurring signal sequence is used to produce the fusion protein of the present invention, the vector does not need a second signal peptide. Alternatively, if a naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e.g., International Publication No. WO 96/34891.) [1023]
  • The amplified fragment encoding the fusion protein of the invention is isolated from a 1% agarose gel using a commercially available kit (“Geneclean,” [1024] BIO 101 Inc., La Jolla, Calif.). The fragment then is digested with appropriate restriction enzymes and again purified on a 1% agarose gel.
  • The amplified fragment encoding the albumin fusion protein of the invention is then digested with the same restriction enzyme and purified on a 1% agarose gel. The isolated fragment and the dephosphorylated vector are then ligated with T4 DNA ligase. [1025] E. coli HB101 or XL-1 Blue cells are then transformed and bacteria are identified that contain the fragment inserted into plasmid pC6 using, for instance, restriction enzyme analysis.
  • Chinese hamster ovary cells lacking an active DHFR gene is used for transfection. Five ug of the expression plasmid pC6 or pC4 is cotransfected with 0.5 μg of the plasmid pSVneo using lipofectin (Felgner et al., supra). The plasmid pSV2-neo contains a dominant selectable marker, the neo gene from Tn5 encoding an enzyme that confers resistance to a group of antibiotics including G418. The cells are seeded in alpha minus MEM supplemented with 1 mg/ml G418. After 2 days, the cells are trypsinized and seeded in hybridoma cloning plates (Greiner, Germany) in alpha minus MEM supplemented with 10, 25, or 50 ng/ml of methotrexate plus 1 mg/ml G418. After about 10-14 days single clones are trypsinized and then seeded in 6-well petri dishes or 10 ml flasks using different concentrations of methotrexate (50 nM, 100 nM, 200 nM, 400 nM, 800 nM). Clones growing at the highest concentrations of methotrexate are then transferred to new 6-well plates containing even higher concentrations of methotrexate (1 μM, 2 μM, 5 μM, 10 mM, 20 mM). The same procedure is repeated until clones are obtained which grow at a concentration of 100-200 μM. Expression of the desired fusion protein is analyzed, for instance, by SDS-PAGE and Western blot or by reversed phase HPLC analysis. [1026]
  • Example 15 Multifusion Fusions
  • The albumin fusion proteins (e.g, containing a Therapeutic protein (or fragment or variant thereof) fused to albumin (or a fragment or variant thereof)) may additionally be fused to other proteins to generate “multifusion proteins”. These multifusion proteins can be used for a variety of applications. For example, fusion of the albumin fusion proteins of the invention to His-tag, HA-tag, protein A, IgG domains, and maltose binding protein facilitates purification. (See e.g, EP A 394,827; Traunecker et al., Nature 331:84-86 (1988)). Nuclear localization signals fused to the polypeptides of the present invention can target the protein to a specific subcellular localization, while covalent heterodimer or homodimers can increase or decrease the activity of an albumin fusion protein. Furthermore, the fusion of additional protein sequences to the albumin fusion proteins of the invention may further increase the solubility and/or stability of the fusion protein. The fusion proteins described above can be made using or routinely modifting techniques known in the art and/or by modifying the following protocol, which outlines the fusion of a polypeptide to an IgG molecule. [1027]
  • Briefly, the human Fc portion of the IgG molecule can be PCR amplified, using primers that span the 5′ and 3′ ends of the sequence described below. These primers also should have convenient restriction enzyme sites that will facilitate cloning into an expression vector, preferably a mammalian or yeast expression vector. [1028]
  • For example, if pC4 (ATCC Accession No. 209646) is used, the human Fc portion can be ligated into the BamHI cloning site. Note that the 3′ BamHI site should be destroyed. Next, the vector containing the human Fc portion is re-restricted with BamHI, linearizing the vector, and a polynucleotide encoding an albumin fusion protein of the present invention (generateed and isolated using techniques known in the art), is ligated into this BamHI site. Note that the polynucleotide encoding the fusion protein of the invention is cloned without a stop codon, otherwise a Fc containing fusion protein will not be produced. [1029]
  • If the naturally occurring signal sequence is used to produce the albumin fusion protein of the present invention, pC4 does not need a second signal peptide. Alternatively, if the naturally occurring signal sequence is not used, the vector can be modified to include a heterologous signal sequence. (See, e.g., International Publication No. WO 96/34891.) [1030]
    Human IgG Fc region:
    (SEQ ID NO: 36)
    GGGATCCGGAGCCCAAATCTTCTGACAAAACTCACACATGCCCACCGTGC
    CCAGCACCTGAATTCGAGGGTGCACCGTCAGTCTTCCTCTTCCCCCCAAA
    ACCCAAGGACACCCTCATGATCTCCCGGACTCCTGAGGTCACATGCGTGG
    TGGTGGACGTAAGCCACGAAGACCCTGAGGTCAAGTTCAACTGGTACGTG
    GACGGCGTGGAGGTGCATAATGCCAAGACAAAGCCGCGGGAGGAGCAGTA
    CAACAGCACGTACCGTGTGGTCAGCGTCCTCACCGTCCTGCACCAGGACT
    GGCTGAATGGCAAGGAGTACAAGTGCAAGGTCTCCAACAAAGCCCTCCCA
    ACCCCCATCGAGAAAACCATCTCCAAAGCCAAAGGGCAGCCCCGAGAACC
    ACAGGTGTACACCCTGCCCCCATCCCGGGATGAGCTGACCAAGAACCAGG
    TCAGCCTGACCTGCCTGGTCAAAGGCTTCTATCCAAGCGACATCGCCGTG
    GAGTGGGAGAGCAATGGGCAGCCGGAGAACAACTACAAGACCACGCCTCC
    CGTGCTGGACTCCGACGGCTCCTTCTTCCTCTACAGCAAGCTCACCGTGG
    ACAAGAGCAGGTGGCAGCAGGGGAACGTCTTCTCATGCTCCGTGATGCAT
    GAGGCTCTGCACAACCACTACACGCAGAAGAGCCTCTCCCTGTCTCCGGG
    TAAATGAGTGCGACGGCCGCGACTCTAGAGGAT
  • Example 16 Production of an Antibody from an Albumin Fusion Protein
  • a) Hybridoma Technology [1031]
  • Antibodies that bind the albumin fusion proteins of the present invention and portions of the albumin fusion proteins of the present invention (e.g., the Therapeutic protein portion or albumin portion of the fusion protein) can be prepared by a variety of methods. (See, Current Protocols, [1032] Chapter 2.) As one example of such methods, a preparation of an albumin fusion protein of the invention or a portion of an albumin fusion protein of the invention is prepared and purified to render it substantially free of natural contaminants. Such a preparation is then introduced into an animal in order to produce polyclonal antisera of greater specific activity.
  • Monoclonal antibodies specific for an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention, are prepared using hybridoma technology (Kohler et al., Nature 256:495 (1975); Kohler et al., Eur. J. Immunol. 6:511 (1976); Kohler et al., Eur. J. Immunol. 6:292 (1976); Hammerling et al., in: Monoclonal Antibodies and T-Cell Hybridomas, Elsevier, N.Y., pp. 563-681 (1981)). In general, an animal (preferably a mouse) is immunized with an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. The splenocytes of such mice are extracted and fused with a suitable myeloma cell line. Any suitable myeloma cell line may be employed in accordance with the present invention; however, it is preferable to employ the parent myeloma cell line (SP20), available from the ATCC. After fusion, the resulting hybridoma cells are selectively maintained in HAT medium, and then cloned by limiting dilution as described by Wands et al. (Gastroenterology 80:225-232 (1981)). The hybridoma cells obtained through such a selection are then assayed to identify clones which secrete antibodies capable of binding an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. [1033]
  • Alternatively, additional antibodies capable of binding to an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention can be produced in a two-step procedure using anti-idiotypic antibodies. Such a method makes use of the fact that antibodies are themselves antigens, and therefore, it is possible to obtain an antibody which binds to a second antibody. In accordance with this method, protein specific antibodies are used to immunize an animal, preferably a mouse. The splenocytes of such an animal are then used to produce hybridoma cells, and the hybridoma cells are screened to identify clones which produce an antibody whose ability to bind to the an albumin fusion protein of the invention (or portion of an albumin fusion protein of the invention)-specific antibody can be blocked by the fusion protein of the invention, or a portion of an albumin fusion protein of the invention. Such antibodies comprise anti-idiotypic antibodies to the fusion protein of the invention (or portion of an albumin fusion protein of the invention)-specific antibody and are used to immunize an animal to induce formation of further fusion protein of the invention (or portion of an albumin fusion protein of the invention)-specific antibodies. [1034]
  • For in vivo use of antibodies in humans, an antibody is “humanized”. Such antibodies can be produced using genetic constructs derived from hybridoma cells producing the monoclonal antibodies described above. Methods for producing chimeric and humanized antibodies are known in the art and are discussed herein. (See, for review, Morrison, Science 229:1202 (1985); Oi et al., BioTechniques 4:214 (1986); Cabilly et al., U.S. Pat. No. 4,816,567; Taniguchi et al., EP 171496; Morrison et al., EP 173494; Neuberger et al., WO 8601533; Robinson et al., International Publication No. WO 8702671; Boulianne et al., Nature 312:643 (1984); Neuberger et al., Nature 314:268 (1985)). [1035]
  • b) Isolation Of Antibody Fragments Directed Against an Albumin Fusion Protein of the Invention, or a Portion of an Albumin Fusion Protein of the Invention From a Library of scFvs [1036]
  • Naturally occurring V-genes isolated from human PBLs are constructed into a library of antibody fragments which contain reactivities against an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention, to which the donor may or may not have been exposed (see e.g., U.S. Pat. No. 5,885,793 incorporated herein by reference in its entirety). [1037]
  • Rescue of the Library. A library of scFvs is constructed from the RNA of human PBLs as described in International Publication No. WO 92/01047. To rescue phage displaying antibody fragments, approximately 109 [1038] E. coli harboring the phagemid are used to inoculate 50 ml of 2×TY containing 1% glucose and 100 μg/ml of ampicillin (2×TY-AMP-GLU) and grown to an O.D. of 0.8 with shaking. Five ml of this culture is used to inoculate 50 ml of 2×TY-AMP-GLU, 2×108 TU of delta gene 3 helper (M13 delta gene III, see International Publication No. WO 92/01047) are added and the culture incubated at 37° C. for 45 minutes without shaking and then at 37° C. for 45 minutes with shaking. The culture is centrifuged at 4000 r.p.m. for 10 min. and the pellet resuspended in 2 liters of 2×TY containing 100 μg/ml ampicillin and 50 ug/ml kanamycin and grown overnight. Phage are prepared as described in International Publication No. WO 92/01047.
  • M13 delta gene III is prepared as follows: M13 delta gene III helper phage does not encode gene III protein, hence the phage(mid) displaying antibody fragments have a greater avidity of binding to antigen. Infectious M13 delta gene III particles are made by growing the helper phage in cells harboring a pUC19 derivative supplying the wild type gene III protein during phage morphogenesis. The culture is incubated for 1 hour at 37° C. without shaking and then for a further hour at 37° C. with shaking. Cells are spun down (IEC-Centra 8,400 r.p.m. for 10 min), resuspended in 300 [1039] ml 2×TY broth containing 100 μg ampicillin/ml and 25 μg kanamycin/ml (2×TY-AMP-KAN) and grown overnight, shaking at 37° C. Phage particles are purified and concentrated from the culture medium by two PEG-precipitations (Sambrook et al., 1990), resuspended in 2 ml PBS and passed through a 0.45 μm filter (Minisart NML; Sartorius) to give a final concentration of approximately 1013 transducing units/ml (ampicillin-resistant clones).
  • Panning of the Library. Immunotubes (Nunc) are coated overnight in PBS with 4 ml of either 100 μg/ml or 10 μg/ml of an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention. Tubes are blocked with 2% Marvel-PBS for 2 hours at 37° C. and then washed 3 times in PBS. Approximately 10[1040] 13 TU of phage is applied to the tube and incubated for 30 minutes at room temperature tumbling on an over and under turntable and then left to stand for another 1.5 hours. Tubes are washed 10 times with PBS 0.1% Tween-20 and 10 times with PBS. Phage are eluted by adding 1 ml of 100 mM triethylamine and rotating 15 minutes on an under and over turntable after which the solution is immediately neutralized with 0.5 ml of 1.0M Tris-HCl, pH 7.4. Phage are then used to infect 10 ml of mid-log E. coli TG1 by incubating eluted phage with bacteria for 30 minutes at 37° C. The E. coli are then plated on TYE plates containing 1% glucose and 100 μg/ml ampicillin. The resulting bacterial library is then rescued with delta gene 3 helper phage as described above to prepare phage for a subsequent round of selection. This process is then repeated for a total of 4 rounds of affinity purification with tube-washing increased to 20 times with PBS, 0.1% Tween-20 and 20 times with PBS for rounds 3 and 4.
  • Characterization of Binders. Eluted phage from the 3rd and 4th rounds of selection are used to infect [1041] E. coli HB 2151 and soluble scFv is produced (Marks, et al., 1991) from single colonies for assay. ELISAs are performed with microtitre plates coated with either 10 pg/ml of an albumin fusion protein of the invention, or a portion of an albumin fusion protein of the invention, in 50 mM bicarbonate pH 9.6. Clones positive in ELISA are further characterized by PCR fingerprinting (see, e.g., International Publication No. WO 92/01047) and then by sequencing. These ELISA positive clones may also be further characterized by techniques known in the art, such as, for example, epitope mapping, binding affinity, receptor signal transduction, ability to block or competitively inhibit antibody/antigen binding, and competitive agonistic or antagonistic activity.
  • Example 17 Method of Treatment Using Gene Therapy-Ex Vivo
  • One method of gene therapy transplants fibroblasts, which are capable of expressing an albumin fusion protein of the present invention, onto a patient. Generally, fibroblasts are obtained from a subject by skin biopsy. The resulting tissue is placed in tissue-culture medium and separated into small pieces. Small chunks of the tissue are placed on a wet surface of a tissue culture flask, approximately ten pieces are placed in each flask. The flask is turned upside down, closed tight and left at room temperature over night. After 24 hours at room temperature, the flask is inverted and the chunks of tissue remain fixed to the bottom of the flask and fresh media (e.g., Ham's F12 media, with 10% FBS, penicillin and streptomycin) is added. The flasks are then incubated at 37 degree C. for approximately one week. [1042]
  • At this time, fresh media is added and subsequently changed every several days. After an additional two weeks in culture, a monolayer of fibroblasts emerge. The monolayer is trypsinized and scaled into larger flasks. pMV-7 (Kirschmeier, P. T. et al., DNA, 7:219-25 (1988)), flanked by the long terminal repeats of the Moloney murine sarcoma virus, is digested with EcoRI and HindIII and subsequently treated with calf intestinal phosphatase. The linear vector is fractionated on agarose gel and purified, using glass beads. [1043]
  • Polynucleotides encoding an albumin fusion protein of the invention can be generated using techniques known in the art amplified using PCR primers which correspond to the 5′ and 3′ end sequences and optionally having appropriate restriction sites and initiation/stop codons, if necessary. Preferably, the 5′ primer contains an EcoRI site and the 3′ primer includes a HindIII site. Equal quantities of the Moloney murine sarcoma virus linear backbone and the amplified EcoRI and HindIII fragment are added together, in the presence of T4 DNA ligase. The resulting mixture is maintained under conditions appropriate for ligation of the two fragments. The ligation mixture is then used to transform bacteria HB101, which are then plated onto agar containing kanamycin for the purpose of confirming that the vector has the gene of interest properly inserted. [1044]
  • The amphotropic pA317 or GP+am12 packaging cells are grown in tissue culture to confluent density in Dulbecco's Modified Eagles Medium (DMEM) with 10% calf serum (CS), penicillin and streptomycin. The MSV vector containing the gene is then added to the media and the packaging cells transduced with the vector. The packaging cells now produce infectious viral particles containing the gene (the packaging cells are now referred to as producer cells). [1045]
  • Fresh media is added to the transduced producer cells, and subsequently, the media is harvested from a 10 cm plate of confluent producer cells. The spent media, containing the infectious viral particles, is filtered through a millipore filter to remove detached producer cells and this media is then used to infect fibroblast cells. Media is removed from a sub-confluent plate of fibroblasts and quickly replaced with the media from the producer cells. This media is removed and replaced with fresh media. If the titer of virus is high, then virtually all fibroblasts will be infected and no selection is required. If the titer is very low, then it is necessary to use a retroviral vector that has a selectable marker, such as neo or his. Once the fibroblasts have been efficiently infected, the fibroblasts are analyzed to determine whether the albumin fusion protein is produced. [1046]
  • The engineered fibroblasts are then transplanted onto the host, either alone or after having been grown to confluence on [1047] cytodex 3 microcarrier beads.
  • Example 18 Method of Treatment Using Gene Therapy—In Vivo
  • Another aspect of the present invention is using in vivo gene therapy methods to treat disorders, diseases and conditions. The gene therapy method relates to the introduction of naked nucleic acid (DNA, RNA, and antisense DNA or RNA) sequences encoding an albumin fusion protein of the invention into an animal. Polynucleotides encoding albumin fusion proteins of the present invention may be operatively linked to (i.e., associated with) a promoter or any other genetic elements necessary for the expression of the polypeptide by the target tissue. Such gene therapy and delivery techniques and methods are known in the art, see, for example, WO90/11092, WO98/11779; U.S. Pat. No. 5,693,622, 5705151, 5580859; Tabata et al., Cardiovasc. Res. 35(3):470-479 (1997); Chao et al., Pharmacol. Res. 35(6):517-522 (1997); Wolff, Neuromuscul. Disord. 7(5):314-318 (1997); Schwartz et al., Gene Ther. 3(5):405-411 (1996); Tsurumi et al., Circulation 94(12):3281-3290 (1996) (incorporated herein by reference). [1048]
  • The polynucleotide constructs may be delivered by any method that delivers injectable materials to the cells of an animal, such as, injection into the interstitial space of tissues (heart, muscle, skin, lung, liver, intestine and the like). The polynucleotide constructs can be delivered in a pharmaceutically acceptable liquid or aqueous carrier. [1049]
  • The term “naked” polynucleotide, DNA or RNA, refers to sequences that are free from any delivery vehicle that acts to assist, promote, or facilitate entry into the cell, including viral sequences, viral particles, liposome formulations, lipofectin or precipitating agents and the like. However, polynucleotides encoding albumin fusion proteins of the present invention may also be delivered in liposome formulations (such as those taught in Felgner P. L. et al. (1995) Ann. NY Acad. Sci. 772:126-139 and Abdallah B. et al. (1995) Biol. Cell 85(1):1-7) which can be prepared by methods well known to those skilled in the art. [1050]
  • The polynucleotide vector constructs used in the gene therapy method are preferably constructs that will not integrate into the host genome nor will they contain sequences that allow for replication. Any strong promoter known to those skilled in the art can be used for driving the expression of DNA. Unlike other gene therapy techniques, one major advantage of introducing naked nucleic acid sequences into target cells is the transitory nature of the polynucleotide synthesis in the cells. Studies have shown that non-replicating DNA sequences can be introduced into cells to provide production of the desired polypeptide for periods of up to six months. [1051]
  • The polynucleotide construct can be delivered to the interstitial space of tissues within an animal, including muscle, skin, brain, lung, liver, spleen, bone marrow, thymus, heart, lymph, blood, bone, cartilage, pancreas, kidney, gall bladder, stomach, intestine, testis, ovary, uterus, rectum, nervous system, eye, gland, and connective tissue. Interstitial space of the tissues comprises the intercellular fluid, mucopolysaccharide matrix among the reticular fibers of organ tissues, elastic fibers in the walls of vessels or chambers, collagen fibers of fibrous tissues, or that same matrix within connective tissue ensheathing muscle cells or in the lacunae of bone. It is similarly the space occupied by the plasma of the circulation and the lymph fluid of the lymphatic channels. Delivery to the interstitial space of muscle tissue is preferred for the reasons discussed below. They may be conveniently delivered by injection into the tissues comprising these cells. They are preferably delivered to and expressed in persistent, non-dividing cells which are differentiated, although delivery and expression may be achieved in non-differentiated or less completely differentiated cells, such as, for example, stem cells of blood or skin fibroblasts. In vivo muscle cells are particularly competent in their ability to take up and express polynucleotides. [1052]
  • For the naked polynucleotide injection, an effective dosage amount of DNA or RNA will be in the range of from about 0.05 g/kg body weight to about 50 mg/kg body weight. Preferably the dosage will be from about 0.005 mg/kg to about 20 mg/kg and more preferably from about 0.05 mg/kg to about 5 mg/kg. Of course, as the artisan of ordinary skill will appreciate, this dosage will vary according to the tissue site of injection. The appropriate and effective dosage of nucleic acid sequence can readily be determined by those of ordinary skill in the art and may depend on the condition being treated and the route of administration. The preferred route of administration is by the parenteral route of injection into the interstitial space of tissues. However, other parenteral routes may also be used, such as, inhalation of an aerosol formulation particularly for delivery to lungs or bronchial tissues, throat or mucous membranes of the nose. In addition, naked polynucleotide constructs can be delivered to arteries during angioplasty by the catheter used in the procedure. [1053]
  • The dose response effects of injected polynucleotide in muscle in vivo is determined as follows. Suitable template DNA for production of mRNA coding for polypeptide of the present invention is prepared in accordance with a standard recombinant DNA methodology. The template DNA, which may be either circular or linear, is either used as naked DNA or complexed with liposomes. The quadriceps muscles of mice are then injected with various amounts of the template DNA. [1054]
  • Five to six week old female and male Balb/C mice are anesthetized by intraperitoneal injection with 0.3 ml of 2.5% Avertin. A 1.5 cm incision is made on the anterior thigh, and the quadriceps muscle is directly visualized. The template DNA is injected in 0.1 ml of carrier in a 1 cc syringe through a 27 gauge needle over one minute, approximately 0.5 cm from the distal insertion site of the muscle into the knee and about 0.2 cm deep. A suture is placed over the injection site for future localization, and the skin is closed with stainless steel clips. [1055]
  • After an appropriate incubation time (e.g., 7 days) muscle extracts are prepared by excising the entire quadriceps. Every fifth 15 um cross-section of the individual quadriceps muscles is histochemically stained for protein expression. A time course for fusion protein expression may be done in a similar fashion except that quadriceps from different mice are harvested at different times. Persistence of DNA in muscle following injection may be determined by Southern blot analysis after preparing total cellular DNA and HIRT supernatants from injected and control mice. The results of the above experimentation in mice can be used to extrapolate proper dosages and other treatment parameters in humans and other animals using naked DNA. [1056]
  • Example 19 Transgenic Animals
  • The albumin fusion proteins of the invention can also be expressed in transgenic animals. Animals of any species, including, but not limited to, mice, rats, rabbits, hamsters, guinea pigs, pigs, micro-pigs, goats, sheep, cows and non-human primates, e.g., baboons, monkeys, and chimpanzees may be used to generate transgenic animals. In a specific embodiment, techniques described herein or otherwise known in the art, are used to express fusion proteins of the invention in humans, as part of a gene therapy protocol. [1057]
  • Any technique known in the art may be used to introduce the polynucleotides encoding the albumin fusion proteins of the invention into animals to produce the founder lines of transgenic animals. Such techniques include, but are not limited to, pronuclear microinjection (Paterson et al., Appl. Microbiol. Biotechnol. 40:691-698 (1994); Carver et al., Biotechnology (NY) 11:1263-1270 (1993); Wright et al., Biotechnology (NY) 9:830-834 (1991); and Hoppe et al., U.S. Pat. No. 4,873,191 (1989)); retrovirus mediated gene transfer into germ lines (Van der Putten et al., Proc. Natl. Acad. Sci., USA 82:6148-6152 (1985)), blastocysts or embryos; gene targeting in embryonic stem cells (Thompson et al., Cell 56:313-321 (1989)); electroporation of cells or embryos (Lo, 1983, Mol Cell. Biol. 3:1803-1814 (1983)); introduction of the polynucleotides of the invention using a gene gun (see, e.g., Ulmer et al., Science 259:1745 (1993); introducing nucleic acid constructs into embryonic pleuripotent stem cells and transferring the stem cells back into the blastocyst; and sperm-mediated gene transfer (Lavitrano et al., Cell 57:717-723 (1989); etc. For a review of such techniques, see Gordon, “Transgenic Animals,” Intl. Rev. Cytol. 115:171-229 (1989), which is incorporated by reference herein in its entirety. [1058]
  • Any technique known in the art may be used to produce transgenic clones containing polynucleotides encoding albumin fusion proteins of the invention, for example, nuclear transfer into enucleated oocytes of nuclei from cultured embryonic, fetal, or adult cells induced to quiescence (Campell et al., Nature 380:64-66 (1996); Wilmut et al., Nature 385:810-813 (1997)). [1059]
  • The present invention provides for transgenic animals that carry the polynucleotides encoding the albumin fusion proteins of the invention in all their cells, as well as animals which carry these polynucleotides in some, but not all their cells, i.e., mosaic animals or chimeric. The transgene may be integrated as a single transgene or as multiple copies such as in concatamers, e.g., head-to-head tandems or head-to-tail tandems. The transgene may also be selectively introduced into and activated in a particular cell type by following, for example, the teaching of Lasko et al. (Lasko et al., Proc. Natl. Acad. Sci. USA 89:6232-6236 (1992)). The regulatory sequences required for such a cell-type specific activation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. When it is desired that the polynucleotide encoding the fusion protein of the invention be integrated into the chromosomal site of the endogenous gene corresponding to the Therapeutic protein portion or ablumin portion of the fusion protein of the invention, gene targeting is preferred. Briefly, when such a technique is to be utilized, vectors containing some nucleotide sequences homologous to the endogenous gene are designed for the purpose of integrating, via homologous recombination with chromosomal sequences, into and disrupting the function of the nucleotide sequence of the endogenous gene. The transgene may also be selectively introduced into a particular cell type, thus inactivating the endogenous gene in only that cell type, by following, for example, the teaching of Gu et al. (Gu et al., Science 265:103-106 (1994)). The regulatory sequences required for such a cell-type specific inactivation will depend upon the particular cell type of interest, and will be apparent to those of skill in the art. [1060]
  • Once transgenic animals have been generated, the expression of the recombinant gene may be assayed utilizing standard techniques. Initial screening may be accomplished by Southern blot analysis or PCR techniques to analyze animal tissues to verify that integration of the polynucleotide encoding the fsuion protien of the invention has taken place. The level of mRNA expression of the polynucleotide encoding the fusion protein of the invention in the tissues of the transgenic animals may also be assessed using techniques which include, but are not limited to, Northern blot analysis of tissue samples obtained from the animal, in situ hybridization analysis, and reverse transcriptase-PCR (rt-PCR). Samples of fusion protein-expressing tissue may also be evaluated immunocytochemically or immunohistochemically using antibodies specific for the fusion protein. [1061]
  • Once the founder animals are produced, they may be bred, inbred, outbred, or crossbred to produce colonies of the particular animal. Examples of such breeding strategies include, but are not limited to: outbreeding of founder animals with more than one integration site in order to establish separate lines; inbreeding of separate lines in order to produce compound transgenics that express the transgene at higher levels because of the effects of additive expression of each transgene; crossing of heterozygous transgenic animals to produce animals homozygous for a given integration site in order to both augment expression and eliminate the need for screening of animals by DNA analysis; crossing of separate homozygous lines to produce compound heterozygous or homozygous lines; and breeding to place the transgene (i.e., polynucleotide encoding an albumin fusion protein of the invention) on a distinct background that is appropriate for an experimental model of interest. [1062]
  • Transgenic animals of the invention have uses which include, but are not limited to, animal model systems useful in elaborating the biological function of fusion proteins of the invention and the Therapeutic protein and/or albumin component of the fusion protein of the invention, studying conditions and/or disorders associated with aberrant expression, and in screening for compounds effective in ameliorating such conditions and/or disorders. [1063]
  • Example 20 Assays Detecting Stimulation or Inhibition of B Cell Proliferation and Differentiation
  • Generation of functional humoral immune responses requires both soluble and cognate signaling between B-lineage cells and their microenvironment. Signals may impart a positive stimulus that allows a B-lineage cell to continue its programmed development, or a negative stimulus that instructs the cell to arrest its current developmental pathway. To date, numerous stimulatory and inhibitory signals have been found to influence B cell responsiveness including IL-2, IL-4, IL-5, IL-6, IL-7, IL10, IL-13, IL-14 and IL-15. Interestingly, these signals are by themselves weak effectors but can, in combination with various co-stimulatory proteins, induce activation, proliferation, differentiation, homing, tolerance and death among B cell populations. One of the best studied classes of B-cell co-stimulatory proteins is the TNF-superfamily. Within this family CD40, CD27, and CD30 along with their respective ligands CD154, CD70, and CD153 have been found to regulate a variety of immune responses. Assays which allow for the detection and/or observation of the proliferation and differentiation of these B-cell populations and their precursors are valuable tools in determining the effects various proteins may have on these B-cell populations in terms of proliferation and differentiation. Listed below are two assays designed to allow for the detection of the differentiation, proliferation, or inhibition of B-cell populations and their precursors. [1064]
  • In Vitro Assay—Albumin fusion proteins of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin) can be assessed for its ability to induce activation, proliferation, differentiation or inhibition and/or death in B-cell populations and their precursors. The activity of an albumin fusion protein of the invention on purified human tonsillar B cells, measured qualitatively over the dose range from 0.1 to 10,000 ng/mL, is assessed in a standard B-lymphocyte co-stimulation assay in which purified tonsillar B cells are cultured in the presence of either formalin-fixed [1065] Staphylococcus aureus Cowan I (SAC) or immobilized anti-human IgM antibody as the priming agent. Second signals such as IL-2 and IL-15 synergize with SAC and IgM crosslinking to elicit B cell proliferation as measured by tritiated-thymidine incorporation. Novel synergizing agents can be readily identified using this assay. The assay involves isolating human tonsillar B cells by magnetic bead (MACS) depletion of CD3-positive cells. The resulting cell population is greater than 95% B cells as assessed by expression of CD45R(B220).
  • Various dilutions of each sample are placed into individual wells of a 96-well plate to which are added 10[1066] 5 B-cells suspended in culture medium (RPMI 1640 containing 10% FBS, 5×10−5M 2ME, 100U/ml penicillin, 10 ug/ml streptomycin, and 10−5 dilution of SAC) in a total volume of 150 ul. Proliferation or inhibition is quantitated by a 20 h pulse (1 uCi/well) with 3H-thymidine (6.7 Ci/mM) beginning 72 h post factor addition. The positive and negative controls are IL2 and medium respectively.
  • In vivo Assay—BALB/c mice are injected (i.p.) twice per day with buffer only, or 2 mg/Kg of an albumin fusion protein of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin). Mice receive this treatment for 4 consecutive days, at which time they are sacrificed and various tissues and serum collected for analyses. Comparison of H&E sections from normal spleens and spleens treated with the albumin fusion protein of the invention identify the results of the activity of the fusion protein on spleen cells, such as the diffusion of peri-arterial lymphatic sheaths, and/or significant increases in the nucleated cellularity of the red pulp regions, which may indicate the activation of the differentiation and proliferation of B-cell populations. Immunohistochemical studies using a B cell marker, anti-CD45R(B220), are used to determine whether any physiological changes to splenic cells, such as splenic disorganization, are due to increased B-cell representation within loosely defined B-cell zones that infiltrate established T-cell regions. [1067]
  • Flow cytometric analyses of the spleens from mice treated with the albumin fusion protein is used to indicate whether the albumin fusion protein specifically increases the proportion of ThB+, CD45R(B220)dull B cells over that which is observed in control mice. [1068]
  • Likewise, a predicted consequence of increased mature B-cell representation in vivo is a relative increase in serum Ig titers. Accordingly, serum IgM and IgA levels are compared between buffer and fusion protein treated mice. [1069]
  • The studies described in this example tested activity of fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1070]
  • Example 21 T Cell Proliferation Assay
  • A CD3-induced proliferation assay is performed on PBMCs and is measured by the uptake of [1071] 3H-thymidine. The assay is performed as follows. Ninety-six well plates are coated with 100 μL/well of mAb to CD3 (HIT3a, Pharmingen) or isotype-matched control mAb (B33.1) overnight at 4 degrees C. (1 μg/ml in 0.05M bicarbonate buffer, pH 9.5), then washed three times with PBS. PBMC are isolated by F/H gradient centrifugation from human peripheral blood and added to quadruplicate wells (5×104/well) of mAb coated plates in RPMI containing 10% FCS and P/S in the presence of varying concentrations of an albumin fusion protein of the invention (including fusion proteins containing fragments or variants of Therapeutic proteins and/or albumin or fragments or variants of albumin) (total volume 200 ul). Relevant protein buffer and medium alone are controls. After 48 hr. culture at 37 degrees C., plates are spun for 2 min. at 1000 rpm and 100 μl of supernatant is removed and stored −20 degrees C. for measurement of IL-2 (or other cytokines) if effect on proliferation is observed. Wells are supplemented with 100 ul of medium containing 0.5 uCi of 3H-thymidine and cultured at 37 degrees C. for 18-24 hr. Wells are harvested and incorporation of 3H-thymidine used as a measure of proliferation. Anti-CD3 alone is the positive control for proliferation. IL-2 (100 U/ml) is also used as a control which enhances proliferation. Control antibody which does not induce proliferation of T cells is used as the negative control for the effects of fusion proteins of the invention.
  • The studies described in this example tested activity of fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins or polynucleotides of the invention (e.g., gene therapy). [1072]
  • Example 22 Effect of Fusion Proteins of the Invention on the Expression of MHC Class II, Costimulatory and Adhesion Molecules and Cell Differentiation of Monocytes and Monocyte-Derived Human Dendritic Cells
  • Dendritic cells are generated by the expansion of proliferating precursors found in the peripheral blood: adherent PBMC or elutriated monocytic fractions are cultured for 7-10 days with GM-CSF (50 ng/ml) and IL-4 (20 ng/ml). These dendritic cells have the characteristic phenotype of immature cells (expression of CD1, CD80, CD86, CD40 and MHC class II antigens). Treatment with activating factors, such as TNF-α, causes a rapid change in surface phenotype (increased expression of MHC class I and II, costimulatory and adhesion molecules, downregulation of FCγRII, upregulation of CD83). These changes correlate with increased antigen-presenting capacity and with functional maturation of the dendritic cells. [1073]
  • FACS analysis of surface antigens is performed as follows. Cells are treated 1-3 days with increasing concentrations of an albumin fusion protein of the invention or LPS (positive control), washed with PBS containing 1% BSA and 0.02 mM sodium azide, and then incubated with 1:20 dilution of appropriate FITC- or PE-labeled monoclonal antibodies for 30 minutes at 4 degrees C. After an additional wash, the labeled cells are analyzed by flow cytometry on a FACScan (Becton Dickinson). [1074]
  • Effect on the production of cytokines. Cytokines generated by dendritic cells, in particular IL-12, are important in the initiation of T-cell dependent immune responses. IL-12 strongly influences the development of Th1 helper T-cell immune response, and induces cytotoxic T and NK cell function. An ELISA is used to measure the IL-12 release as follows. Dendritic cells (10[1075] 6/ml) are treated with increasing concentrations of an albumin fusion protein of the invention for 24 hours. LPS (100 ng/ml) is added to the cell culture as positive control. Supernatants from the cell cultures are then collected and analyzed for IL-12 content using commercial ELISA kit (e.g., R & D Systems (Minneapolis, Minn.)). The standard protocols provided with the kits are used.
  • Effect on the expression of MHC Class II, costimulatory and adhesion molecules. Three major families of cell surface antigens can be identified on monocytes: adhesion molecules, molecules involved in antigen presentation, and Fc receptor. Modulation of the expression of MHC class II antigens and other costimulatory molecules, such as B7 and ICAM-1, may result in changes in the antigen presenting capacity of monocytes and ability to induce T cell activation. Increased expression of Fc receptors may correlate with improved monocyte cytotoxic activity, cytokine release and phagocytosis. [1076]
  • FACS analysis is used to examine the surface antigens as follows. Monocytes are treated 1-5 days with increasing concentrations of an albumin fusion protein of the invention or LPS (positive control), washed with PBS containing 1% BSA and 0.02 mM sodium azide, and then incubated with 1:20 dilution of appropriate FITC- or PE-labeled monoclonal antibodies for 30 minutes at 4 degrees C. After an additional wash, the labeled cells are analyzed by flow cytometry on a FACScan (Becton Dickinson). [1077]
  • Monocyte activation and/or increased survival. Assays for molecules that activate (or alternatively, inactivate) monocytes and/or increase monocyte survival (or alternatively, decrease monocyte survival) are known in the art and may routinely be applied to determine whether a molecule of the invention functions as an inhibitor or activator of monocytes. Albumin fusion proteins of the invention can be screened using the three assays described below. For each of these assays, Peripheral blood mononuclear cells (PBMC) are purified from single donor leukopacks (American Red Cross, Baltimore, Md.) by centrifugation through a Histopaque gradient (Sigma). Monocytes are isolated from PBMC by counterflow centrifugal elutriation. [1078]
  • Monocyte Survival Assay. Human peripheral blood monocytes progressively lose viability when cultured in absence of serum or other stimuli. Their death results from internally regulated processes (apoptosis). Addition to the culture of activating factors, such as TNF-alpha dramatically improves cell survival and prevents DNA fragmentation. Propidium iodide (PI) staining is used to measure apoptosis as follows. Monocytes are cultured for 48 hours in polypropylene tubes in serum-free medium (positive control), in the presence of 100 ng/ml TNF-alpha (negative control), and in the presence of varying concentrations of the fusion protein to be tested. Cells are suspended at a concentration of 2×10[1079] 6/ml in PBS containing PI at a final concentration of 5 μg/ml, and then incubated at room temperature for 5 minutes before FACScan analysis. PI uptake has been demonstrated to correlate with DNA fragmentation in this experimental paradigm.
  • Effect on cytokine release. An important function of monocytes/macrophages is their regulatory activity on other cellular populations of the immune system through the release of cytokines after stimulation. An ELISA to measure cytokine release is performed as follows. Human monocytes are incubated at a density of 5×10[1080] 5 cells/ml with increasing concentrations of an albumin fusion protein of the invention and under the same conditions, but in the absence of the fusion protein. For IL-12 production, the cells are primed overnight with IFN (100 U/ml) in the presence of the fusion protein. LPS (10 ng/ml) is then added. Conditioned media are collected after 24 h and kept frozen until use. Measurement of TNF-alpha, IL-10, MCP-1 and IL-8 is then performed using a commercially available ELISA kit (e.g., R & D Systems (Minneapolis, Minn.)) and applying the standard protocols provided with the kit.
  • Oxidative burst. Purified monocytes are plated in 96-w plate at 2-1×10[1081] 5 cell/well. Increasing concentrations of an albumin fusion protein of the invention are added to the wells in a total volume of 0.2 ml culture medium (RPMI 1640+10% FCS, glutamine and antibiotics). After 3 days incubation, the plates are centrifuged and the medium is removed from the wells. To the macrophage monolayers, 0.2 ml per well of phenol red solution (140 mM NaCl, 10 mM potassium phosphate buffer pH 7.0, 5.5 mM dextrose, 0.56 mM phenol red and 19 U/ml of HRPO) is added, together with the stimulant (200 nM PMA). The plates are incubated at 37° C. for 2 hours and the reaction is stopped by adding 20 μl 1N NaOH per well. The absorbance is read at 610 nm. To calculate the amount of H2O2 produced by the macrophages, a standard curve of a H2O2 solution of known molarity is performed for each experiment.
  • The studies described in this example tested activity of fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins or polynucleotides of the invention (e.g., gene therapy). [1082]
  • Example 23 Biological Effects of Fusion Proteins of the Invention
  • Astrocyte and Neuronal Assays [1083]
  • Albumin fusion proteins of the invention can be tested for activity in promoting the survival, neurite outgrowth, or phenotypic differentiation of cortical neuronal cells and for inducing the proliferation of glial fibrillary acidic protein immunopositive cells, astrocytes. The selection of cortical cells for the bioassay is based on the prevalent expression of FGF-1 and FGF-2 in cortical structures and on the previously reported enhancement of cortical neuronal survival resulting from FGF-2 treatment. A thymidine incorporation assay, for example, can be used to elucidate an albumin fusion protein of the invention's activity on these cells. [1084]
  • Moreover, previous reports describing the biological effects of FGF-2 (basic FGF) on cortical or hippocampal neurons in vitro have demonstrated increases in both neuron survival and neurite outgrowth (Walicke et al., “Fibroblast growth factor promotes survival of dissociated hippocampal neurons and enhances neurite extension.” [1085] Proc. Natl. Acad. Sci. USA 83:3012-3016. (1986), assay herein incorporated by reference in its entirety). However, reports from experiments done on PC-12 cells suggest that these two responses are not necessarily synonymous and may depend on not only which FGF is being tested but also on which receptor(s) are expressed on the target cells. Using the primary cortical neuronal culture paradigm, the ability of an albumin fusion protein of the invention to induce neurite outgrowth can be compared to the response achieved with FGF-2 using, for example, a thymidine incorporation assay.
  • Fibroblast and Endothelial Cell Assays [1086]
  • Human lung fibroblasts are obtained from Clonetics (San Diego, Calif.) and maintained in growth media from Clonetics. Dermal microvascular endothelial cells are obtained from Cell Applications (San Diego, Calif.). For proliferation assays, the human lung fibroblasts and dermal microvascular endothelial cells can be cultured at 5,000 cells/well in a 96-well plate for one day in growth medium. The cells are then incubated for one day in 0.1% BSA basal medium. After replacing the medium with fresh 0.1% BSA medium, the cells are incubated with the test fusion protein of the invention proteins for 3 days. Alamar Blue (Alamar Biosciences, Sacramento, Calif.) is added to each well to a final concentration of 10%. The cells are incubated for 4 hr. Cell viability is measured by reading in a CytoFluor fluorescence reader. For the PGE[1087] 2 assays, the human lung fibroblasts are cultured at 5,000 cells/well in a 96-well plate for one day. After a medium change to 0.1% BSA basal medium, the cells are incubated with FGF-2 or fusion protein of the invention with or without IL-1α for 24 hours. The supernatants are collected and assayed for PGE2 by EIA kit (Cayman, Ann Arbor, Mich.). For the IL-6 assays, the human lung fibroblasts are cultured at 5,000 cells/well in a 96-well plate for one day. After a medium change to 0.1% BSA basal medium, the cells are incubated with FGF-2 or with or without an albumin fusion protein of the invention and/or IL-1α for 24 hours. The supernatants are collected and assayed for IL-6 by ELISA kit (Endogen, Cambridge, Mass.).
  • Human lung fibroblasts are cultured with FGF-2 or an albumin fusion protein of the invention for 3 days in basal medium before the addition of Alamar Blue to assess effects on growth of the fibroblasts. FGF-2 should show a stimulation at 10-2500 ng/ml which can be used to compare stimulation with the fusion protein of the invention. [1088]
  • Cell Proliferation Based on [3H]Thymidine Incorporation [1089]
  • The following [3H]Thymidine incorporation assay can be used to measure the effect of a Therapeutic proteins, e.g., growth factor proteins, on the proliferation of cells such as fibroblast cells, epithelial cells or immature muscle cells. [1090]
  • Sub-confluent cultures are arrested in G1 phase by an 18 h incubation in serum-free medium. Therapeutic proteins are then added for 24 h and during the last 4 h, the cultures are labeled with [3H]thymidine, at a final concentration of 0.33 μM (25 Ci/mmol, Amersham, Arlington Heights, Ill.). The incorporated [3H]thymidine is precipitated with ice-cold 10% trichloroacetic acid for 24 h. Subsequently, the cells are rinsed sequentially with ice-cold 10% trichloroacetic acid and then with ice-cold water. Following lysis in 0.5 M NaOH, the lysates and PBS rinses (500 ml) are pooled, and the amount of radioactivity is measured. [1091]
  • Parkinson Models. [1092]
  • The loss of motor function in Parkinson's disease is attributed to a deficiency of striatal dopamine resulting from the degeneration of the nigrostriatal dopaminergic projection neurons. An animal model for Parkinson's that has been extensively characterized involves the systemic administration of 1-methyl-4 [1093] phenyl 1,2,3,6-tetrahydropyridine (MPTP). In the CNS, MPTP is taken-up by astrocytes and catabolized by monoamine oxidase B to 1-methyl-4-phenyl pyridine (MPP+) and released. Subsequently, MPP+ is actively accumulated in dopaminergic neurons by the high-affinity reuptake transporter for dopamine. MPP+ is then concentrated in mitochondria by the electrochemical gradient and selectively inhibits nicotidamide adenine disphosphate: ubiquinone oxidoreductionase (complex I), thereby interfering with electron transport and eventually generating oxygen radicals.
  • It has been demonstrated in tissue culture paradigms that FGF-2 (basic FGF) has trophic activity towards nigral dopaminergic neurons (Ferrari et al., Dev. Biol. 1989). Recently, Dr. Unsicker's group has demonstrated that administering FGF-2 in gel foam implants in the striatum results in the near complete protection of nigral dopaminergic neurons from the toxicity associated with MPTP exposure (Otto and Unsicker, J. Neuroscience, 1990). [1094]
  • Based on the data with FGF-2, an albumin fusion protein of the invention can be evaluated to determine whether it has an action similar to that of FGF-2 in enhancing dopaminergic neuronal survival in vitro and it can also be tested in vivo for protection of dopaminergic neurons in the striatum from the damage associated with MPTP treatment. The potential effect of an albumin fusion protein of the invention is first examined in vitro in a dopaminergic neuronal cell culture paradigm. The cultures are prepared by dissecting the midbrain floor plate from gestation day 14 Wistar rat embryos. The tissue is dissociated with trypsin and seeded at a density of 200,000 cells/cm[1095] 2 on polyorthinine-laminin coated glass coverslips. The cells are maintained in Dulbecco's Modified Eagle's medium and F12 medium containing hormonal supplements (N1). The cultures are fixed with paraformaldehyde after 8 days in vitro and are processed for tyrosine hydroxylase, a specific marker for dopaminergic neurons, immunohistochemical staining. Dissociated cell cultures are prepared from embryonic rats. The culture medium is changed every third day and the factors are also added at that time.
  • Since the dopaminergic neurons are isolated from animals at gestation day 14, a developmental time which is past the stage when the dopaminergic precursor cells are proliferating, an increase in the number of tyrosine hydroxylase immunopositive neurons would represent an increase in the number of dopaminergic neurons surviving in vitro. Therefore, if a therapeutic protein of the invention acts to prolong the survival of dopaminergic neurons, it would suggest that the fusion protein may be involved in Parkinson's Disease. [1096]
  • The studies described in this example tested activity of albumin fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1097]
  • Example 24 The Effect of Albumin Fusion Proteins of the Invention on the Growth of Vascular Endothelial Cells
  • On [1098] day 1, human umbilical vein endothelial cells (HUVEC) are seeded at 2-5×104 cells/35 mm dish density in M199 medium containing 4% fetal bovine serum (FBS), 16 units/ml heparin, and 50 units/ml endothelial cell growth supplements (ECGS, Biotechnique, Inc.). On day 2, the medium is replaced with M199 containing 10% FBS, 8 units/ml heparin. An albumin fusion protein of the invention, and positive controls, such as VEGF and basic FGF (bFGF) are added, at varying concentrations. On days 4 and 6, the medium is replaced. On day 8, cell number is determined with a Coulter Counter.
  • An increase in the number of HUVEC cells indicates that the fusion protein may proliferate vascular endothelial cells, while a decrease in the number of HUVEC cells indicates that the fusion protein inhibits vascular endothelial cells. [1099]
  • The studies described in this example tested activity of an albumin fusion protein of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of a fusion protiem and polynucleotides of the invention. [1100]
  • Example 25 Rat Corneal Wound Healing Model
  • This animal model shows the effect of an albumin fusion protein of the invention on neovascularization. The experimental protocol includes: [1101]
  • Making a 1-1.5 mm long incision from the center of cornea into the stromal layer. [1102]
  • Inserting a spatula below the lip of the incision facing the outer corner of the eye. [1103]
  • Making a pocket (its base is 1-1.5 mm form the edge of the eye). [1104]
  • Positioning a pellet, containing 50 ng-5 ug of an albumin fusion protein of the invention, within the pocket. [1105]
  • Treatment with an an albumin fusion protein of the invention can also be applied topically to the corneal wounds in a dosage range of 20 mg-500 mg (daily treatment for five days). [1106]
  • The studies described in this example test the activity of an albumin fusion protein of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1107]
  • Example 26 Diabetic Mouse and Glucocorticoid-Impaired Wound Healing Models
  • Diabetic db+/db+ Mouse Model. [1108]
  • To demonstrate that an albumin fusion protein of the invention accelerates the healing process, the genetically diabetic mouse model of wound healing is used. The full thickness wound healing model in the db+/db+ mouse is a well characterized, clinically relevant and reproducible model of impaired wound healing. Healing of the diabetic wound is dependent on formation of granulation tissue and re-epithelialization rather than contraction (Gartner, M. H. et al., [1109] J. Surg. Res. 52:389 (1992); Greenhalgh, D. G. et al., Am. J. Pathol. 136:1235 (1990)).
  • The diabetic animals have many of the characteristic features observed in Type II diabetes mellitus. Homozygous (db+/db+) mice are obese in comparison to their normal heterozygous (db+/+m) littermates. Mutant diabetic (db+/db+) mice have a single autosomal recessive mutation on chromosome 4 (db+) (Coleman et al. [1110] Proc. Natl. Acad. Sci. USA 77:283-293 (1982)). Animals show polyphagia, polydipsia and polyuria. Mutant diabetic mice (db+/db+) have elevated blood glucose, increased or normal insulin levels, and suppressed cell-mediated immunity (Mandel et al, J. Immunol. 120:1375 (1978); Debray-Sachs, M. et al., Clin. Exp. Immunol. 51(1):1-7 (1983); Leiter et al., Am. J. of Pathol. 114:46-55 (1985)). Peripheral neuropathy, myocardial complications, and microvascular lesions, basement membrane thickening and glomerular filtration abnormalities have been described in these animals (Norido, F. et al., Exp. Neurol. 83(2):221-232 (1984); Robertson et al., Diabetes 29(1):60-67 (1980); Giacomelli et al., Lab Invest. 40(4):460-473 (1979); Coleman, D. L., Diabetes 31 (Suppl):1-6 (1982)). These homozygous diabetic mice develop hyperglycemia that is resistant to insulin analogous to human type II diabetes (Mandel et al., J. Immunol. 120:1375-1377 (1978)).
  • The characteristics observed in these animals suggests that healing in this model may be similar to the healing observed in human diabetes (Greenhalgh, et al., [1111] Am. J. of Pathol. 136:1235-1246 (1990)).
  • Genetically diabetic female C57BL/KsJ (db+/db+) mice and their non-diabetic (db+/+m) heterozygous littermates are used in this study (Jackson Laboratories). The animals are purchased at 6 weeks of age and are 8 weeks old at the beginning of the study. Animals are individually housed and received food and water ad libitum. All manipulations are performed using aseptic techniques. The experiments are conducted according to the rules and guidelines of Human Genome Sciences, Inc. Institutional Animal Care and Use Committee and the Guidelines for the Care and Use of Laboratory Animals. [1112]
  • Wounding protocol is performed according to previously reported methods (Tsuboi, R. and Rifkin, D. B., [1113] J. Exp. Med. 172:245-251 (1990)). Briefly, on the day of wounding, animals are anesthetized with an intraperitoneal injection of Avertin (0.01 mg/mL), 2,2,2-tribromoethanol and 2-methyl-2-butanol dissolved in deionized water. The dorsal region of the animal is shaved and the skin washed with 70% ethanol solution and iodine. The surgical area is dried with sterile gauze prior to wounding. An 8 mm full-thickness wound is then created using a Keyes tissue punch. Immediately following wounding, the surrounding skin is gently stretched to eliminate wound expansion. The wounds are left open for the duration of the experiment. Application of the treatment is given topically for 5 consecutive days commencing on the day of wounding. Prior to treatment, wounds are gently cleansed with sterile saline and gauze sponges.
  • Wounds are visually examined and photographed at a fixed distance at the day of surgery and at two day intervals thereafter. Wound closure is determined by daily measurement on days 1-5 and on [1114] day 8. Wounds are measured horizontally and vertically using a calibrated Jameson caliper. Wounds are considered healed if granulation tissue is no longer visible and the wound is covered by a continuous epithelium.
  • An albumin fusion protein of the invention is administered using at a range different doses, from 4 mg to 500 mg per wound per day for 8 days in vehicle. Vehicle control groups received 50 mL of vehicle solution. [1115]
  • Animals are euthanized on [1116] day 8 with an intraperitoneal injection of sodium pentobarbital (300 mg/kg). The wounds and surrounding skin are then harvested for histology and immunohistochemistry. Tissue specimens are placed in 10% neutral buffered formalin in tissue cassettes between biopsy sponges for further processing.
  • Three groups of 10 animals each (5 diabetic and 5 non-diabetic controls) are evaluated: 1) Vehicle placebo control, 2) untreated group, and 3) treated group. [1117]
  • Wound closure is analyzed by measuring the area in the vertical and horizontal axis and obtaining the total square area of the wound. Contraction is then estimated by establishing the differences between the initial wound area (day 0) and that of post treatment (day 8). The wound area on [1118] day 1 is 64 mm2, the corresponding size of the dermal punch. Calculations are made using the following formula:
  • [Open area on day 8]−[Open area on day 1]/[Open area on day 1]
  • Specimens are fixed in 10% buffered formalin and paraffin embedded blocks are sectioned perpendicular to the wound surface (5 mm) and cut using a Reichert-Jung microtome. Routine hematoxylin-eosin (H&E) staining is performed on cross-sections of bisected wounds. Histologic examination of the wounds are used to assess whether the healing process and the morphologic appearance of the repaired skin is altered by treatment with an albumin fusion protein of-the invention., This assessment included verification of the presence of cell accumulation, inflammatory cells, capillaries, fibroblasts, re-epithelialization and epidermal maturity (Greenhalgh, D. G. et al., [1119] Am. J. Pathol. 136:1235 (1990)). A calibrated lens micrometer is used by a blinded observer.
  • Tissue sections are also stained immunohistochemically with a polyclonal rabbit anti-human keratin antibody using ABC Elite detection system. Human skin is used as a positive tissue control while non-immune IgG is used as a negative control. Keratinocyte growth is determined by evaluating the extent of reepithelialization of the wound using a calibrated lens micrometer. [1120]
  • Proliferating cell nuclear antigen/cyclin (PCNA) in skin specimens is demonstrated by using anti-PCNA antibody (1:50) with an ABC Elite detection system. Human colon cancer served as a positive tissue control and human brain tissue is used as a negative tissue control. Each specimen included a section with omission of the primary antibody and substitution with non-immune mouse IgG. Ranking of these sections is based on the extent of proliferation on a scale of 0-8, the lower side of the scale reflecting slight proliferation to the higher side reflecting intense proliferation. [1121]
  • Experimental data are analyzed using an unpaired t test. A p value of <0.05 is considered significant. [1122]
  • Steroid Impaired Rat Model [1123]
  • The inhibition of wound healing by steroids has been well documented in various in vitro and in vivo systems (Wahl, Glucocorticoids and Wound healing. In: Anti-Inflammatory Steroid Action: Basic and Clinical Aspects. 280-302 (1989); Wahl et al., [1124] J. Immunol. 115: 476-481 (1975); Werb et al., J. Exp. Med. 147:1684-1694 (1978)). Glucocorticoids retard wound healing by inhibiting angiogenesis, decreasing vascular permeability (Ebert et al., An. Intern. Med. 37:701-705 (1952)), fibroblast proliferation, and collagen synthesis (Beck et al., Growth Factors. 5: 295-304 (1991); Haynes et al., J. Clin. Invest. 61: 703-797 (1978)) and producing a transient reduction of circulating monocytes (Haynes et al., J. Clin. Invest. 61: 703-797 (1978); Wahl, “Glucocorticoids and wound healing”, In: Antiinflammatory Steroid Action: Basic and Clinical Aspects, Academic Press, New York, pp. 280-302 (1989)). The systemic administration of steroids to impaired wound healing is a well establish phenomenon in rats (Beck et al., Growth Factors. 5: 295-304 (1991); Haynes et al., J. Clin. Invest. 61: 703-797 (1978); Wahl, “Glucocorticoids and wound healing”, In: Antiinflammatory Steroid Action: Basic and Clinical Aspects, Academic Press, New York, pp. 280-302 (1989); Pierce et al., Proc. Natl. Acad. Sci. USA 86: 2229-2233 (1989)).
  • To demonstrate that an albumin fusion protein of the invention can accelerate the healing process, the effects of multiple topical applications of the fusion protein on full thickness excisional skin wounds in rats in which healing has been impaired by the systemic administration of methylprednisolone is assessed. [1125]
  • Young adult male Sprague Dawley rats weighing 250-300 g (Charles River Laboratories) are used in this example. The animals are purchased at 8 weeks of age and are 9 weeks old at the beginning of the study. The healing response of rats is impaired by the systemic administration of methylprednisolone (17 mg/kg/rat intramuscularly) at the time of wounding. Animals are individually housed and received food and water ad libitum. All manipulations are performed using aseptic techniques. This study is conducted according to the rules and guidelines of Human Genome Sciences, Inc. Institutional Animal Care and Use Committee and the Guidelines for the Care and Use of Laboratory Animals. [1126]
  • The wounding protocol is followed according to that described above. On the day of wounding, animals are anesthetized with an intramuscular injection of ketamine (50 mg/kg) and xylazine (5 mg/kg). The dorsal region of the animal is shaved and the skin washed with 70% ethanol and iodine solutions. The surgical area is dried with sterile gauze prior to wounding. An 8 mm full-thickness wound is created using a Keyes tissue punch. The wounds are left open for the duration of the experiment. Applications of the testing materials are given topically once a day for 7 consecutive days commencing on the day of wounding and subsequent to methylprednisolone administration. Prior to treatment, wounds are gently cleansed with sterile saline and gauze sponges. [1127]
  • Wounds are visually examined and photographed at a fixed distance at the day of wounding and at the end of treatment. Wound closure is determined by daily measurement on days 1-5 and on [1128] day 8. Wounds are measured horizontally and vertically using a calibrated Jameson caliper. Wounds are considered healed if granulation tissue is no longer visible and the wound is covered by a continuous epithelium.
  • The fusion protein of the invention is administered using at a range different doses, from 4 mg to 500 mg per wound per day for 8 days in vehicle. Vehicle control groups received 50 mL of vehicle solution. [1129]
  • Animals are euthanized on [1130] day 8 with an intraperitoneal injection of sodium pentobarbital (300 mg/kg). The wounds and surrounding skin are then harvested for histology. Tissue specimens are placed in 10% neutral buffered formalin in tissue cassettes between biopsy sponges for further processing.
  • Three groups of 10 animals each (5 with methylprednisolone and 5 without glucocorticoid) are evaluated: 1) Untreated group 2) Vehicle placebo control 3) treated groups. [1131]
  • Wound closure is analyzed by measuring the area in the vertical and horizontal axis and obtaining the total area of the wound. Closure is then estimated by establishing the differences between the initial wound area (day 0) and that of post treatment (day 8). The wound area on [1132] day 1 is 64 mm2, the corresponding size of the dermal punch. Calculations are made using the following formula:
  • [Open area on day 8]−[Open area on day 1]/[Open area on day 1]
  • Specimens are fixed in 10% buffered formalin and paraffin embedded blocks are sectioned perpendicular to the wound surface (5 mm) and cut using an Olympus microtome. Routine hematoxylin-eosin (H&E) staining is performed on cross-sections of bisected wounds. Histologic examination of the wounds allows assessment of whether the healing process and the morphologic appearance of the repaired skin is improved by treatment with an albumin fusion protein of the invention. A calibrated lens micrometer is used by a blinded observer to determine the distance of the wound gap. [1133]
  • Experimental data are analyzed using an unpaired t test. A p value of <0.05 is considered significant. [1134]
  • The studies described in this example tested activity of an albumin fusion protein of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1135]
  • Example 27 Lymphedema Animal Model
  • The purpose of this experimental approach is to create an appropriate and consistent lymphedema model for testing the therapeutic effects of an albumin fusion protein of the invention in lymphangiogenesis and re-establishment of the lymphatic circulatory system in the rat hind limb. Effectiveness is measured by swelling volume of the affected limb, quantification of the amount of lymphatic vasculature, total blood plasma protein, and histopathology. Acute lymphedema is observed for 7-10 days. Perhaps more importantly, the chronic progress of the edema is followed for up to 3-4 weeks. [1136]
  • Prior to beginning surgery, blood sample is drawn for protein concentration analysis. Male rats weighing approximately 350 g are dosed with Pentobarbital. Subsequently, the right legs are shaved from knee to hip. The shaved area is swabbed with gauze soaked in 70% EtOH. Blood is drawn for serum total protein testing. Circumference and volumetric measurements are made prior to injecting dye into paws after marking 2 measurement levels (0.5 cm above heel, at mid-pt of dorsal paw). The intradermal dorsum of both right and left paws are injected with 0.05 ml of 1% Evan's Blue. Circumference and volumetric measurements are then made following injection of dye into paws. [1137]
  • Using the knee joint as a landmark, a mid-leg inguinal incision is made circumferentially allowing the femoral vessels to be located. Forceps and hemostats are used to dissect and separate the skin flaps. After locating the femoral vessels, the lymphatic vessel that runs along side and underneath the vessel(s) is located. The main lymphatic vessels in this area are then electrically coagulated or suture ligated. [1138]
  • Using a microscope, muscles in back of the leg (near the semitendinosis and adductors) are bluntly dissected. The popliteal lymph node is then located. The 2 proximal and 2 distal lymphatic vessels and distal blood supply of the popliteal node are then ligated by suturing. The popliteal lymph node, and any accompanying adipose tissue, is then removed by cutting connective tissues. [1139]
  • Care is taken to control any mild bleeding resulting from this procedure. After lymphatics are occluded, the skin flaps are sealed by using liquid skin (Vetbond) (A J Buck). The separated skin edges are sealed to the underlying muscle tissue while leaving a gap of ˜0.5 cm around the leg. Skin also may be anchored by suturing to underlying muscle when necessary. [1140]
  • To avoid infection, animals are housed individually with mesh (no bedding). Recovering animals are checked daily through the optimal edematous peak, which typically occurred by day 5-7. The plateau edematous peak are then observed. To evaluate the intensity of the lymphedema, the circumference and volumes of 2 designated places on each paw before operation and daily for 7 days are measured. The effect of plasma proteins on lymphedema is determined and whether protein analysis is a useful testing perimeter is also investigated. The weights of both control and edematous limbs are evaluated at 2 places. Analysis is performed in a blind manner. [1141]
  • Circumference Measurements: Under brief gas anesthetic to prevent limb movement, a cloth tape is used to measure limb circumference. Measurements are done at the ankle bone and dorsal paw by 2 different people and those 2 readings are averaged. Readings are taken from both control and edematous limbs. [1142]
  • Volumetric Measurements: On the day of surgery, animals are anesthetized with Pentobarbital and are tested prior to surgery. For daily volumetrics animals are under brief halothane anesthetic (rapid immobilization and quick recovery), and both legs are shaved and equally marked using waterproof marker on legs. Legs are first dipped in water, then dipped into instrument to each marked level then measured by Buxco edema software (Chen/Victor). Data is recorded by one person, while the other is dipping the limb to marked area. [1143]
  • Blood-plasma protein measurements: Blood is drawn, spun, and serum separated prior to surgery and then at conclusion for total protein and Ca2[1144] + comparison.
  • Limb Weight Comparison: After drawing blood, the animal is prepared for tissue collection. The limbs are amputated using a quillitine, then both experimental and control legs are cut at the ligature and weighed. A second weighing is done as the tibio-cacaneal joint is disarticulated and the foot is weighed. [1145]
  • Histological Preparations: The transverse muscle located behind the knee (popliteal) area is dissected and arranged in a metal mold, filled with freezeGel, dipped into cold methylbutane, placed into labeled sample bags at −80EC until sectioning. Upon sectioning, the muscle is observed under fluorescent microscopy for lymphatics. [1146]
  • The studies described in this example tested activity of fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion protein and polynucleotides of the invention (e.g., gene therapy). [1147]
  • Example 28 Suppression of TNF Alpha-Induced Adhesion Molecule Expression by an Albumin Fusion Protein of the Invention
  • The recruitment of lymphocytes to areas of inflammation and angiogenesis involves specific receptor-ligand interactions between cell surface adhesion molecules (CAMs) on lymphocytes and the vascular endothelium. The adhesion process, in both normal and pathological settings, follows a multi-step cascade that involves intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and endothelial leukocyte adhesion molecule-1 (E-selectin) expression on endothelial cells (EC). The expression of these molecules and others on the vascular endothelium determines the efficiency with which leukocytes may adhere to the local vasculature and extravasate into the local tissue during the development of an inflammatory response. The local concentration of cytokines and growth factor participate in the modulation of the expression of these CAMs. [1148]
  • Tumor necrosis factor alpha (TNF-a), a potent proinflammatory cytokine, is a stimulator of all three CAMs on endothelial cells and may be involved in a wide variety of inflammatory responses, often resulting in a pathological outcome. [1149]
  • The potential of an albumin fusion protein of the invention to mediate a suppression of TNF-a induced CAM expression can be examined. A modified ELISA assay which uses ECs as a solid phase absorbent is employed to measure the amount of CAM expression on TNF-a treated ECs when co-stimulated with a member of the FGF family of proteins. [1150]
  • To perform the experiment, human umbilical vein endothelial cell (HUVEC) cultures are obtained from pooled cord harvests and maintained in growth medium (EGM-2; Clonetics, San Diego, Calif.) supplemented with 10% FCS and 1% penicillin/streptomycin in a 37 degree C. humidified incubator containing 5% CO[1151] 2. HUVECs are seeded in 96-well plates at concentrations of 1×104 cells/well in EGM medium at 37 degree C. for 18-24 hrs or until confluent. The monolayers are subsequently washed 3 times with a serum-free solution of RPMI-1640 supplemented with 100 U/ml penicillin and 100 mg/ml streptomycin, and treated with a given cytokine and/or growth factor(s) for 24 h at 37 degree C. Following incubation, the cells are then evaluated for CAM expression.
  • Human Umbilical Vein Endothelial cells (HUVECs) are grown in a standard 96 well plate to confluence. Growth medium is removed from the cells and replaced with 90 ul of 199 Medium (10% FBS). Samples for testing and positive or negative controls are added to the plate in triplicate (in 10 ul volumes). Plates are incubated at 37 degree C. for either 5 h (selectin and integrin expression) or 24 h (integrin expression only). Plates are aspirated to remove medium and 100 μl of 0.1% paraformaldehyde-PBS (with Ca++ and Mg++) is added to each well. Plates are held at 4° C. for 30 min. [1152]
  • Fixative is then removed from the wells and wells are washed 1×with PBS (+Ca,Mg)+0.5% BSA and drained. Do not allow the wells to dry. Add 10 μl of diluted primary antibody to the test and control wells. Anti-ICAM-1-Biotin, Anti-VCAM-1-Biotin and Anti-E-selectin-Biotin are used at a concentration of 10 μg/ml (1:10 dilution of 0.1 mg/ml stock antibody). Cells are incubated at 37° C. for 30 min. in a humidified environment. Wells are washed ×3 with PBS (+Ca,Mg)+0.5% BSA. [1153]
  • Then add 20 μl of diluted ExtrAvidin-Alkaline Phosphotase (1:5,000 dilution) to each well and incubated at 37° C. for 30 min. Wells are washed ×3 with PBS (+Ca,Mg)+0.5% BSA. 1 tablet of p-Nitrophenol Phosphate pNPP is dissolved in 5 ml of glycine buffer (pH 10.4). 100 ul of pNPP substrate in glycine buffer is added to each test well. Standard wells in triplicate are prepared from the working dilution of the ExtrAvidin-Alkaline Phosphotase in glycine buffer: 1:5,000 (10[1154] 0)>10−0.5>10−1>101.5. 5 μl of each dilution is added to triplicate wells and the resulting AP content in each well is 5.50 ng, 1.74 ng, 0.55 ng, 0.18 ng. 100 μl of pNNP reagent must then be added to each of the standard wells. The plate must be incubated at 37° C. for 4 h. A volume of 50 ill of 3M NaOH is added to all wells. The results are quantified on a plate reader at 405 nm. The background subtraction option is used on blank wells filled with glycine buffer only. The template is set up to indicate the concentration of AP-conjugate in each standard well [5.50 ng; 1.74 ng; 0.55 ng; 0.18 ng]. Results are indicated as amount of bound AP-conjugate in each sample.
  • The studies described in this example tested activity of fusion proteins of the invention. However, one skilled in the art could easily modify the exemplified studies to test the activity of fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1155]
  • Example 29 Construction of GAS Reporter Construct
  • One signal transduction pathway involved in the differentiation and proliferation of cells is called the Jaks-STATs pathway. Activated proteins in the Jaks-STATs pathway bind to gamma activation site “GAS” elements or interferon-sensitive responsive element (“ISRE”), located in the promoter of many genes. The binding of a protein to these elements alter the expression of the associated gene. [1156]
  • GAS and ISRE elements are recognized by a class of transcription factors called Signal Transducers and Activators of Transcription, or “STATs.” There are six members of the STATs family. Stat1 and Stat3 are present in many cell types, as is Stat2 (as response to IFN-alpha is widespread). Stat4 is more restricted and is not in many cell types though it has been found in T helper class I, cells after treatment with IL-12. Stat5 was originally called mammary growth factor, but has been found at higher concentrations in other cells including myeloid cells. It can be activated in tissue culture cells by many cytokines. [1157]
  • The STATs are activated to translocate from the cytoplasm to the nucleus upon tyrosine phosphorylation by a set of kinases known as the Janus Kinase (“Jaks”) family. Jaks represent a distinct family of soluble tyrosine kinases and include Tyk2, Jak1, Jak2, and Jak3. These kinases display significant sequence similarity and are generally catalytically inactive in resting cells. [1158]
  • The Jaks are activated by a wide range of receptors summarized in the Table below. (Adapted from review by Schidler and Darnell, Ann. Rev. Biochem. 64:621-51 (1995)). A cytokine receptor family, capable of activating Jaks, is divided into two groups: (a) [1159] Class 1 includes receptors for IL-2, IL-3, IL-4, IL-6, IL-7, IL-9, IL-11, IL-12, IL-15, Epo, PRL, GH, G-CSF, GM-CSF, LIF, CNTF, and thrombopoietin; and (b) Class 2 includes IFN-a, IFN-g, and IL-10. The Class 1 receptors share a conserved cysteine motif (a set of four conserved cysteines and one tryptophan) and a WSXWS motif (a membrane proximal region encoding Trp-Ser-Xaa-Trp-Ser (SEQ ID NO: 37)).
  • Thus, on binding of a ligand to a receptor, Jaks are activated, which in turn activate STATs, which then translocate and bind to GAS elements. This entire process is encompassed in the Jaks-STATs signal transduction pathway. Therefore, activation of the Jaks-STATs pathway, reflected by the binding of the GAS or the ISRE element, can be used to indicate proteins involved in the proliferation and differentiation of cells. For example, growth factors and cytokines are known to activate the Jaks-STATs pathway (See Table below). Thus, by using GAS elements linked to reporter molecules, activators of the Jaks-STATs pathway can be identified. [1160]
    GAS
    JAKs (elements)
    Ligand tyk2 Jak1 Jak2 Jak3 STATS or ISRE
    IFN family
    IFN-a/B + + 1, 2, 3 ISRE
    IFN-g + + 1 GAS(IRF1 >
    Lys6 > IFP)
    Il-10 + ? ? 1, 3
    gp130 family
    IL-6(Pleiotropic) + + + ? 1, 3 GAS(IRF1 >
    Lys6 > IFP)
    Il-11(Pleiotropic) ? + ? ? 1, 3
    OnM(Pleiotropic) ? + + ? 1, 3
    LIF(Pleiotropic) ? + + ? 1, 3
    CNTF(Pleiotropic) −/+ + + ? 1, 3
    G-CSF(Pleiotropic) ? + ? ? 1, 3
    IL-12(Pleiotropic) + + + 1, 3
    g-C family
    IL-2(lymphocytes) + + 1, 3, 5 GAS
    IL-4(lymph/ + + 6 GAS
    myeloid) (IRF1 =
    IFP >> Ly6)
    (IgH)
    IL-7(lymphocytes) + + 5 GAS
    IL-9(lymphocytes) + + 5 GAS
    IL-13(lymphocyte) + ? ? 6 GAS
    IL-15 ? + ? + 5 GAS
    gp140 family
    IL-3(myeloid) + 5 GAS(IRF1 >
    IFP >> Ly6)
    IL-5(myeloid) + 5 GAS
    GM-CSF(myeloid) + 5 GAS
    Growth hormone
    family
    GH ? + 5
    PRL ? +/− + 1, 3, 5
    EPO ? + 5 GAS
    (B-CAS >
    IRF1 =
    IFP >> Ly6)
    Receptor Tyrosine
    Kinases
    EGF ? + + 1, 3 GAS(IRF1)
    PDGF ? + + 1, 3
    CSF-1 ? + + 1, 3 GAS
    (not IRF1)
  • To construct a synthetic GAS containing promoter element, which is used in the Biological Assays described in Examples 32-33, a PCR based strategy is employed to generate a GAS-SV40 promoter sequence. The 5′ primer contains four tandem copies of the GAS binding site found in the IRF1 promoter and previously demonstrated to bind STATs upon induction with a range of cytokines (Rothman et al., Immunity 1:457-468 (1994).), although other GAS or ISRE elements can be used instead. The 5′ primer also contains 18 bp of sequence complementary to the SV40 early promoter sequence and is flanked with an XhoI site. The sequence of the 5′ primer is: [1161]
    (SEQ ID NO: 38)
    5′:GCGCCTCGAGATTTCCCCGAAATCTAGATTTCCCCGAAATGATTTCC
    CCGAAATGATTTCCCCGAAATATCTGCCATCTCAATTAG:3′
  • The downstream primer is complementary to the SV40 promoter and is flanked with a Hind III site: 5′:GCGGCAAGCTTTTTGCAAAGCCTAGGC:3′ (SEQ ID NO: 39) PCR amplification is performed using the SV40 promoter template present in the B-gal:promoter plasmid obtained from Clontech. The resulting PCR fragment is digested with XhoI/Hind III and subcloned into BLSK2-. (Stratagene.) Sequencing with forward and reverse primers confirms that the insert contains the following sequence: [1162]
    (SEQ ID NO: 40)
    5′-CTCGAGATTTCCCCGAAATCTAGATTTCCCCGAAATGATTTCCCCGA
    AATGATTTCCCCGAAATATCTGCCATCTCAATTAGTCAGCAACCATAGTC
    CCGCCCCTAACTCCGCCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCA
    TTCTCCGCCCCATGGCTGACTAATTTTTTTTATTTATGCAGAGGCCGAGG
    CCGCCTCGGCCTCTGAGCTATTCCAGAAGTAGTGAGGAGGCTTTTTTGGA
    GGCCTAGGCTTTTGCAAAAAGCTT:3′
  • With this GAS promoter element linked to the SV40 promoter, a GAS:SEAP2 reporter construct is next engineered. Here, the reporter molecule is a secreted alkaline phosphatase, or “SEAP.” Clearly, however, any reporter molecule can be instead of SEAP, in this or in any of the other Examples. Well known reporter molecules that can be used instead of SEAP include chloramphenicol acetyltransferase (CAT), luciferase, alkaline phosphatase, B-galactosidase, green fluorescent protein (GFP), or any protein detectable by an antibody. [1163]
  • The above sequence confirmed synthetic GAS-SV40 promoter element is subcloned into the pSEAP-Promoter vector obtained from Clontech using HindIII and XhoI, effectively replacing the SV40 promoter with the amplified GAS:SV40 promoter element, to create the GAS-SEAP vector. However, this vector does not contain a neomycin resistance gene, and therefore, is not preferred for mammalian expression systems. [1164]
  • Thus, in order to generate mammalian stable cell lines expressing the GAS-SEAP reporter, the GAS-SEAP cassette is removed from the GAS-SEAP vector using SalI and NotI, and inserted into a backbone vector containing the neomycin resistance gene, such as pGFP-1 (Clontech), using these restriction sites in the multiple cloning site, to create the GAS-SEAP/Neo vector. Once this vector is transfected into mammalian cells, this vector can then be used as a reporter molecule for GAS binding as described in Examples 32-33. [1165]
  • Other constructs can be made using the above description and replacing GAS with a different promoter sequence. For example, construction of reporter molecules containing EGR and NF-KB promoter sequences are described in Examples 34 and 35. However, many other promoters can be substituted using the protocols described in these Examples. For instance, SRE, IL-2, NFAT, or Osteocalcin promoters can be substituted, alone or in combination (e.g., GAS/NF-KB/EGR, GAS/NF-KB, Il-2/NFAT, or NF-KB/GAS). Similarly, other cell lines can be used to test reporter construct activity, such as HELA (epithelial), HUVEC (endothelial), Reh (B-cell), Saos-2 (osteoblast), HUVAC (aortic), or Cardiomyocyte. [1166]
  • Example 30 Assay for SEAP Activity
  • As a reporter molecule for the assays described in examples disclosed herein, SEAP activity is assayed using the Tropix Phospho-light Kit (Cat. BP-400) according to the following general procedure. The Tropix Phospho-light Kit supplies the Dilution, Assay, and Reaction Buffers used below. [1167]
  • Prime a dispenser with the 2.5×Dilution Buffer and dispense 15 ul of 2.5×dilution buffer into Optiplates containing 35 ul of a solution containing an albumin fusion protein of the invention. Seal the plates with a plastic sealer and incubate at 65 degree C. for 30 min. Separate the Optiplates to avoid uneven heating. [1168]
  • Cool the samples to room temperature for 15 minutes. Empty the dispenser and prime with the Assay Buffer. Add 50 ml Assay Buffer and incubate at [1169] room temperature 5 min. Empty the dispenser and prime with the Reaction Buffer (see the Table below). Add 50 ul Reaction Buffer and incubate at room temperature for 20 minutes. Since the intensity of the chemiluminescent signal is time dependent, and it takes about 10 minutes to read 5 plates on a luminometer, thus one should treat 5 plates at each time and start the second set 10 minutes later.
  • Read the relative light unit in the luminometer. Set H12 as blank, and print the results. An increase in chemiluminescence indicates reporter activity. [1170]
    Reaction Buffer Formulation:
    # of Rxn buffer diluent CSPD
    plates (ml) (ml)
    10 60 3
    11 65 3.25
    12 70 3.5
    13 75 3.75
    14 80 4
    15 85 4.25
    16 90 4.5
    17 95 4.75
    18 100 5
    19 105 5.25
    20 110 5.5
    21 115 5.75
    22 120 6
    23 125 6.25
    24 130 6.5
    25 135 6.75
    26 140 7
    27 145 7.25
    28 150 7.5
    29 155 7.75
    30 160 8
    31 165 8.25
    32 170 8.5
    33 175 8.75
    34 180 9
    35 185 9.25
    36 190 9.5
    37 195 9.75
    38 200 10
    39 205 10.25
    40 210 10.5
    41 215 10.75
    42 220 11
    43 225 11.25
    44 230 11.5
    45 235 11.75
    46 240 12
    47 245 12.25
    48 250 12.5
    49 255 12.75
    50 260 13
  • Example 31 Assay Identifying Neuronal Activity.
  • When cells undergo differentiation and proliferation, a group of genes are activated through many different signal transduction pathways. One of these genes, EGR1 (early growth response gene 1), is induced in various tissues and cell types upon activation. The promoter of EGR1 is responsible for such induction. Using the EGR1 promoter linked to reporter molecules, the ability of fusion proteins of the invention to activate cells can be assessed. [1171]
  • Particularly, the following protocol is used to assess neuronal activity in PC12 cell lines. PC12 cells (rat phenochromocytoma cells) are known to proliferate and/or differentiate by activation with a number of mitogens, such as TPA (tetradecanoyl phorbol acetate), NGF (nerve growth factor), and EGF (epidermal growth factor). The EGR1 gene expression is activated during this treatment. Thus, by stably transfecting PC12 cells with a construct containing an EGR promoter linked to SEAP reporter, activation of PC12 cells by an albumin fusion protein of the present invention can be assessed. [1172]
  • The EGR/SEAP reporter construct can be assembled by the following protocol. The EGR-1 promoter sequence (−633 to +1)(Sakamoto K et al., Oncogene 6:867-871 (1991)) can be PCR amplified from human genomic DNA using the following primers: [1173]
    (SEQ ID NO: 41)
    5′ GCGCTCGAGGGATGACAGCGATAGAACCCCGG-3′
    (SEQ ID NO: 42)
    5′ GCGAAGCTTCGCGACTCCCCGGATCCGCCTC-3′
  • Using the GAS:SEAP/Neo vector produced in Example 29, EGR1 amplified product can then be inserted into this vector. Linearize the GAS:SEAP/Neo vector using restriction enzymes XhoI/HindIII, removing the GAS/SV40 stuffer. Restrict the EGR1 amplified product with these same enzymes. Ligate the vector and the EGR1 promoter. [1174]
  • To prepare 96 well-plates for cell culture, two mls of a coating solution (1:30 dilution of collagen type I (Upstate Biotech Inc. Cat#08-115) in 30% ethanol (filter sterilized)) is added per one 10 cm plate or 50 ml per well of the 96-well plate, and allowed to air dry for 2 hr. [1175]
  • PC 12 cells are routinely grown in RPMI-1640 medium (Bio Whittaker) containing 10% horse serum (JRH BIOSCIENCES, Cat. # 12449-78P), 5% heat-inactivated fetal bovine serum (FBS) supplemented with 100 units/ml penicillin and 100 ug/ml streptomycin on a precoated 10 cm tissue culture dish. One to four split is done every three to four days. Cells are removed from the plates by scraping and resuspended with pipetting up and down for more than 15 times. [1176]
  • Transfect the EGR/SEAP/Neo construct into PC12 using techniques known in the art. EGR-SEAP/PC12 stable cells are obtained by growing the cells in 300 ug/ml G418. The G418-free medium is used for routine growth but every one to two months, the cells should be re-grown in 300 ug/ml G418 for couple of passages. [1177]
  • To assay for neuronal activity, a 10 cm plate with cells around 70 to 80% confluent is screened by removing the old medium. Wash the cells once with PBS (Phosphate buffered saline). Then starve the cells in low serum medium (RPMI-1640 containing 1% horse serum and 0.5% FBS with antibiotics) overnight. [1178]
  • The next morning, remove the medium and wash the cells with PBS. Scrape off the cells from the plate, suspend the cells well in 2 ml low serum medium. Count the cell number and add more low serum medium to reach final cell density as 5×10[1179] 5 cells/ml.
  • Add 200 ul of the cell suspension to each well of 96-well plate (equivalent to 1×10[1180] 5 cells/well). Add a series of different concentrations of an albumin fusion protein of the inventon, 37 degree C. for 48 to 72 hr. As a positive control, a growth factor known to activate PC12 cells through EGR can be used, such as 50 ng/ul of Neuronal Growth Factor (NGF). Over fifty-fold induction of SEAP is typically seen in the positive control wells. SEAP assay may be routinely performed using techniques known in the art and/or as described in Example 30.
  • Example 32 Assay for T-Cell Activity
  • The following protocol is used to assess T-cell activity by identifying factors, and determining whether an albumin fusion protein of the invention proliferates and/or differentiates T-cells. T-cell activity is assessed using the GAS/SEAP/Neo construct produced in Example 29. Thus, factors that increase SEAP activity indicate the ability to activate the Jaks-STATS signal transduction pathway. The T-cell used in this assay is Jurkat T-cells (ATCC Accession No. TIB-152), although Molt-3 cells (ATCC Accession No. CRL-1552) and Molt-4 cells (ATCC Accession No. CRL-1582) cells can also be used. [1181]
  • Jurkat T-cells are lymphoblastic CD4[1182] + Th1 helper cells. In order to generate stable cell lines, approximately 2 million Jurkat cells are transfected with the GAS-SEAP/neo vector using DMRIE-C (Life Technologies)(transfection procedure described below). The transfected cells are seeded to a density of approximately 20,000 cells per well and transfectants resistant to 1 mg/ml genticin selected. Resistant colonies are expanded and then tested for their response to increasing concentrations of interferon gamma. The dose response of a selected clone is demonstrated.
  • Specifically, the following protocol will yield sufficient cells for 75 wells containing 200 ul of cells. Thus, it is either scaled up, or performed in multiple to generate sufficient cells for multiple 96 well plates. Jurkat cells are maintained in RPMI+10% serum with 1%Pen-Strep. Combine 2.5 mls of OPTI-MEM (Life Technologies) with 10 ug of plasmid DNA in a T25 flask. Add 2.5 ml OPTI-MEM containing 50 ul of DMRIE-C and incubate at room temperature for 15-45 mins. [1183]
  • During the incubation period, count cell concentration, spin down the required number of cells (10[1184] 7 per transfection), and resuspend in OPTI-MEM to a final concentration of 107 cells/ml. Then add 1 ml of 1×107 cells in OPTI-MEM to T25 flask and incubate at 37 degree C. for 6 hrs. After the incubation, add 10 ml of RPMI+15% serum.
  • The Jurkat:GAS-SEAP stable reporter lines are maintained in RPMI+10% serum, 1 mg/ml Genticin, and 1% Pen-Strep. These cells are treated with varying concentrations of one or more fusion proteins of the present invention. [1185]
  • On the day of treatment with the fusion protein, the cells should be washed and resuspended in fresh RPMI+10% serum to a density of 500,000 cells per ml. The exact number of cells required will depend on the number of fusion proteins and the number of different concentrations of fusion proteins being screened. For one 96 well plate, approximately 10 million cells (for 10 plates, 100 million cells) are required. [1186]
  • The well dishes containing Jurkat cells treated with the fusion protein are placed in an incubator for 48 hrs (note: this time is variable between 48-72 hrs). 35 ul samples from each well are then transferred to an opaque 96 well plate using a 12 channel pipette. The opaque plates should be covered (using sellophene covers) and stored at −20 degree C. until SEAP assays are performed according to Example 30. The plates containing the remaining treated cells are placed at 4 degree C. and serve as a source of material for repeating the assay on a specific well if desired. [1187]
  • As a positive control, 100 Unit/ml interferon gamma can be used which is known to activate Jurkat T cells. Over 30 fold induction is typically observed in the positive control wells. [1188]
  • The above protocol may be used in the generation of both transient, as well as, stable transfected cells, which would be apparent to those of skill in the art. [1189]
  • Example 33 Assay for T-Cell Activity
  • NF-KB (Nuclear Factor KB) is a transcription factor activated by a wide variety of agents including the inflammatory cytokines IL-1 and TNF, CD30 and CD40, lymphotoxin-alpha and lymphotoxin-beta, by exposure to LPS or thrombin, and by expression of certain viral gene products. As a transcription factor, NF-KB regulates the expression of genes involved in immune cell activation, control of apoptosis (NF-KB appears to shield cells from apoptosis), B and T-cell development, anti-viral and antimicrobial responses, and multiple stress responses. [1190]
  • In non-stimulated conditions, NF-KB is retained in the cytoplasm with I-KB (Inhibitor KB). However, upon stimulation, I-KB is phosphorylated and degraded, causing NF-KB to shuttle to the nucleus, thereby activating transcription of target genes. Target genes activated by NF-KB include IL-2, 1L-6, GM-CSF, ICAM-1 and [1191] class 1 MHC.
  • Due to its central role and ability to respond to a range of stimuli, reporter constructs utilizing the NF-KB promoter element are used to screen the fusion protein. Activators or inhibitors of NF-KB would be useful in treating, preventing, and/or diagnosing diseases. For example, inhibitors of NF-KB could be used to treat those diseases related to the acute or chronic activation of NF-KB, such as rheumatoid arthritis. [1192]
  • To construct a vector containing the NF-KB promoter element, a PCR based strategy is employed. The upstream primer contains four tandem copies of the NF-KB binding site (GGGGACTTTCCC) (SEQ BD NO: 43), 18 bp of sequence complementary to the 5′ end of the SV40 early promoter sequence, and is flanked with an XhoI site: [1193]
    (SEQ ID NO: 44)
    5′:GCGGCCTCGAGGGGACTTTCCCGGGGACTTTCCGGGGACTTTCCGGG
    ACTTTCCATCCTGCCATCTCAATTAG:3′
  • The downstream primer is complementary to the 3′ end of the SV40 promoter and is flanked with a Hind III site: [1194]
    5′:GCGGCAAGCTTTTTGCAAAGCCTAGGC:3′ (SEQ ID NO: 39)
  • PCR amplification is performed using the SV40 promoter template present in the pB-gal:promoter plasmid obtained from Clontech. The resulting PCR fragment is digested with XhoI and Hind III and subcloned into BLSK2-. (Stratagene) Sequencing with the T7 and T3 primers confirms the insert contains the following sequence: [1195]
    (SEQ ID NO: 45)
    5′:CTCGAGGGGACTTTCCCGGGGACTTTCCGGGGACTTTCCGGGACTTT
    CCATCTGCCATCTCAATTAGTCAGCAACCATAGTCCCGCCCCTAACTCCG
    CCCATCCCGCCCCTAACTCCGCCCAGTTCCGCCCATTCTCCGCCCCATGG
    CTGACTAATTTTTTTTATTTATGCAGAGGCCGAGGCCGCCTCGGCCTCTG
    AGCTATTCCAGAAGTAGTGAGGAGGCTTTTTTGGAGGCCTAGGCTTTTGC
    AAAAAGCTT:3′
  • Next, replace the SV40 minimal promoter element present in the pSEAP2-promoter plasmid (Clontech) with this NF-KB/SV40 fragment using XhoI and HindIII. However, this vector does not contain a neomycin resistance gene, and therefore, is not preferred for mammalian expression systems. [1196]
  • In order to generate stable mammalian cell lines, the NF-KB/SV40/SEAP cassette is removed from the above NF-KB/SEAP vector using restriction enzymes SalI and NotI, and inserted into a vector containing neomycin resistance. Particularly, the NF-KB/SV40/SEAP cassette was inserted into pGFP-1 (Clontech), replacing the GFP gene, after restricting pGFP-1 with SalI and NotI. [1197]
  • Once NF-KB/SV40/SEAP/Neo vector is created, stable Jurkat T-cells are created and maintained according to the protocol described in Example 32. Similarly, the method for assaying fusion proteins with these stable Jurkat T-cells is also described in Example 32. As a positive control, exogenous TNF alpha (0.1, 1, 10 ng) is added to wells H9, H1O, and H11, with a 5-10 fold activation typically observed. [1198]
  • Example 33 Assay Identifying Myeloid Activity
  • The following protocol is used to assess myeloid activity of an albumin fusion protein of the present invention by determining whether the fusion protein proliferates and/or differentiates myeloid cells. Myeloid cell activity is assessed using the GAS/SEAP/Neo construct produced in Example 29. Thus, factors that increase SEAP activity indicate the ability to activate the Jaks-STATS signal transduction pathway. The myeloid cell used in this assay is U937, a pre-monocyte cell line, although TF-1, HL60, or KG1 can be used. [1199]
  • To transiently transfect U937 cells with the GAS/SEAP/Neo construct produced in Example 29, a DEAE-Dextran method (Kharbanda et. al., 1994, Cell Growth & Differentiation, 5:259-265) is used. First, harvest 2×10[1200] 7 U937 cells and wash with PBS. The U937 cells are usually grown in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum (FBS) supplemented with 100 units/ml penicillin and 100 mg/ml streptomycin.
  • Next, suspend the cells in 1 ml of 20 mM Tris-HCl (pH 7.4) buffer containing 0.5 mg/ml DEAE-Dextran, 8 ug GAS-SEAP2 plasmid DNA, 140 mM NaCl, 5 mM KCl, 375 uM Na[1201] 2HPO4.7H2O, 1 mM MgCl2, and 675 uM CaCl2. Incubate at 37 degrees C. for 45 min.
  • Wash the cells with RPMI 1640 medium containing 10% FBS and then resuspend in 10 ml complete medium and incubate at 37 degree C. for 36 hr. [1202]
  • The GAS-SEAP/U937 stable cells are obtained by growing the cells in 400 ug/ml G418. The G418-free medium is used for routine growth but every one to two months, the cells should be re-grown in 400 ug/ml G418 for couple of passages. [1203]
  • These cells are tested by harvesting 1×10[1204] 8 cells (this is enough for ten 96-well plates assay) and wash with PBS. Suspend the cells in 200 ml above described growth medium, with a final density of 5×105 cells/ml. Plate 200 ul cells per well in the 96-well plate (or 1×105 cells/well).
  • Add different concentrations of the fusion protein. Incubate at 37 degee C for 48 to 72 hr. As a positive control, 100 Unit/ml interferon gamma can be used which is known to activate U937 cells. Over 30 fold induction is typically observed in the positive control wells. SEAP assay the supernatant according to methods known in the art and/or the protocol described in Example 30. [1205]
  • Example 34 Assay Identifying Changes in Small Molecule Concentration and Membrane Permeability
  • Binding of a ligand to a receptor is known to alter intracellular levels of small molecules, such as calcium, potassium, sodium, and pH, as well as alter membrane potential. These alterations can be measured in an assay to identify fusion proteins which bind to receptors of a particular cell. Although the following protocol describes an assay for calcium, this protocol can easily be modified to detect changes in potassium, sodium, pH, membrane potential, or any other small molecule which is detectable by a fluorescent probe. [1206]
  • The following assay uses Fluorometric Imaging Plate Reader (“FLIPR”) to measure changes in fluorescent molecules (Molecular Probes) that bind small molecules. Clearly, any fluorescent molecule detecting a small molecule can be used instead of the calcium fluorescent molecule, fluo-4 (Molecular Probes, Inc.; catalog no. F-14202), used here. [1207]
  • For adherent cells, seed the cells at 10,000-20,000 cells/well in a Co-star black 96-well plate with clear bottom. The plate is incubated in a CO[1208] 2 incubator for 20 hours. The adherent cells are washed two times in Biotek washer with 200 ul of HBSS (Hank's Balanced Salt Solution) leaving 100 ul of buffer after the final wash.
  • A stock solution of 1 mg/ml fluo-4 is made in 10% pluronic acid DMSO. To load the cells with fluo-4, 50 ul of 12 ug/ml fluo-4 is added to each well. The plate is incubated at 37 degrees C. in a CO[1209] 2 incubator for 60 min. The plate is washed four times in the Biotek washer with HBSS leaving 100 ul of buffer.
  • For non-adherent cells, the cells are spun down from culture media. Cells are re-suspended to 2-5×10[1210] 6 cells/ml with HBSS in a 50-ml conical tube. 4 ul of 1 mg/ml fluo-4 solution in 10% pluronic acid DMSO is added to each ml of cell suspension. The tube is then placed in a 37 degrees C. water bath for 30-60 min. The cells are washed twice with HBSS, resuspended to 1×106 cells/ml, and dispensed into a microplate, 100 ul/well. The plate is centrifuged at 1000 rpm for 5 min. The plate is then washed once in Denley Cell Wash with 200 ul, followed by an aspiration step to 100 ul final volume.
  • For a non-cell based assay, each well contains a fluorescent molecule, such as fluo-4. The fusion protein of the invention is added to the well, and a change in fluorescence is detected. [1211]
  • To measure the fluorescence of intracellular calcium, the FLIPR is set for the following parameters: (1) System gain is 300-800 mW; (2) Exposure time is 0.4 second; (3) Camera F/stop is F/2; (4) Excitation is 488 nm; (5) Emission is 530 nm; and (6) Sample addition is 50 ul. Increased emission at 530 nm indicates an extracellular signaling event caused by an albumin fusion protein of the present invention or a molecule induced by an albumin fusion protein of the present invention, which has resulted in an increase in the intracellular Ca[1212] ++ concentration.
  • Example 35 Assay Identifying Tyrosine Kinase Activity
  • The Protein Tyrosine Kinases (PTK) represent a diverse group of transmembrane and cytoplasmic kinases. Within the Receptor Protein Tyrosine Kinase (RPTK) group are receptors for a range of mitogenic and metabolic growth factors including the PDGF, FGF, EGF, NGF, HGF and Insulin receptor subfamilies. In addition there are a large family of RPTKs for which the corresponding ligand is unknown. Ligands for RPTKs include mainly secreted small proteins, but also membrane-bound and extracellular matrix proteins. [1213]
  • Activation of RPTK by ligands involves ligand-mediated receptor dimerization, resulting in transphosphorylation of the receptor subunits and activation of the cytoplasmic tyrosine kinases. The cytoplasmic tyrosine kinases include receptor associated tyrosine kinases of the src-family (e.g., src, yes, lck, lyn, fyn) and non-receptor linked and cytosolic protein tyrosine kinases, such as the Jak family, members of which mediate signal transduction triggered by the cytokine superfamily of receptors (e.g., the Interleukins, Interferons, GM-CSF, and Leptin). [1214]
  • Because of the wide range of known factors capable of stimulating tyrosine kinase activity, identifying whether an albumin fusion protein of the present invention or a molecule induced by a fusion proetin of the present invention is capable of activating tyrosine kinase signal transduction pathways is of interest. Therefore, the following protocol is designed to identify such molecules capable of activating the tyrosine kinase signal transduction pathways. [1215]
  • Seed target cells (e.g., primary keratinocytes) at a density of approximately 25,000 cells per well in a 96 well Loprodyne Silent Screen Plates purchased from Nalge Nunc (Naperville, Ill.). The plates are sterilized with two 30 minute rinses with 100% ethanol, rinsed with water and dried overnight. Some plates are coated for 2 hr with 100 ml of cell culture grade type I collagen (50 mg/ml), gelatin (2%) or polylysine (50 mg/ml), all of which can be purchased from Sigma Chemicals (St. Louis, Mo.) or 10% Matrigel purchased from Becton Dickinson (Bedford, Mass.), or calf serum, rinsed with PBS and stored at 4 degree C. Cell growth on these plates is assayed by seeding 5,000 cells/well in growth medium and indirect quantitation of cell number through use of alamarBlue as described by the manufacturer Alamar Biosciences, Inc. (Sacramento, Calif.) after 48 hr. Falcon plate covers #3071 from Becton Dickinson (Bedford, Mass.) are used to cover the Loprodyne Silent Screen Plates. Falcon Microtest III cell culture plates can also be used in some proliferation experiments. [1216]
  • To prepare extracts, A431 cells are seeded onto the nylon membranes of Loprodyne plates (20,000/200 ml/well) and cultured overnight in complete medium. Cells are quiesced by incubation in serum-free basal medium for 24 hr. After 5-20 minutes treatment with EGF (60 ng/ml) or a different concentrations of an albumin fusion protein of the invention, the medium was removed and 100 ml of extraction buffer ((20 mM HEPES pH 7.5, 0.15 M NaCl, 1% Triton X-100, 0.1% SDS, 2 mM Na3VO4, 2 mM Na4P207 and a cocktail of protease inhibitors (# 1836170) obtained from Boeheringer Mannheim (Indianapolis, Ind.)) is added to each well and the plate is shaken on a rotating shaker for 5 minutes at 4° C. The plate is then placed in a vacuum transfer manifold and the extract filtered through the 0.45 mm membrane bottoms of each well using house vacuum. Extracts are collected in a 96-well catch/assay plate in the bottom of the vacuum manifold and immediately placed on ice. To obtain extracts clarified by centrifugation, the content of each well, after detergent solubilization for 5 minutes, is removed and centrifuged for 15 minutes at 4 degree C. at 16,000×g. [1217]
  • Test the filtered extracts for levels of tyrosine kinase activity. Although many methods of detecting tyrosine kinase activity are known, one method is described here. [1218]
  • Generally, the tyrosine kinase activity of an albumin fusion protein of the invention is evaluated by determining its ability to phosphorylate a tyrosine residue on a specific substrate (a biotinylated peptide). Biotinylated peptides that can be used for this purpose include PSK1 (corresponding to amino acids 6-20 of the cell division kinase cdc2-p34) and PSK2 (corresponding to amino acids 1-17 of gastrin). Both peptides are substrates for a range of tyrosine kinases and are available from Boehringer Mannheim. [1219]
  • The tyrosine kinase reaction is set up by adding the following components in order. First, add 10 ul of 5 uM Biotinylated Peptide, then 10 ul ATP/Mg[1220] 2+ (5 mM ATP/50 mM MgCl2), then 10 ul of 5×Assay Buffer (40 mM imidazole hydrochloride, pH 7.3, 40 mM beta-glycerophosphate, 1 mM EGTA, 100 mM MgCl2, 5 mM MnCl2, 0.5 mg/ml BSA), then 5 ul of Sodium Vanadate(1 mM), and then 5 ul of water. Mix the components gently and preincubate the reaction mix at 30 degree C. for 2 min. Initial the reaction by adding 10 ul of the control enzyme or the filtered supernatant.
  • The tyrosine kinase assay reaction is then terminated by adding 10 ul of 120 mm EDTA and place the reactions on ice. [1221]
  • Tyrosine kinase activity is determined by transferring 50 ul aliquot of reaction mixture to a microtiter plate (MTP) module and incubating at 37 degree C. for 20 min. This allows the streptavidin coated 96 well plate to associate with the biotinylated peptide. Wash the MTP module with 300 ul/well of PBS four times. Next add 75 ul of anti-phospolyrosine antibody conjugated to horse radish peroxidase (anti-P-Tyr-POD(0.5 u/ml)) to each well and incubate at 37 degree C. for one hour. Wash the well as above. [1222]
  • Next add 100 ul of peroxidase substrate solution (Boehringer Mannheim) and incubate at room temperature for at least 5 mins (up to 30 min). Measure the absorbance of the sample at 405 m by using ELISA reader. The level of bound peroxidase activity is quantitated using an ELISA reader and reflects the level of tyrosine kinase activity. [1223]
  • Example 36 Assay Identifying Phosphorylation Activity
  • As a potential alternative and/or complement to the assay of protein tyrosine kinase activity described in Example 35, an assay which detects activation (phosphorylation) of major intracellular signal transduction intermediates can also be used. For example, as described below one particular assay can detect tyrosine phosphorylation of the Erk-1 and Erk-2 kinases. However, phosphorylation of other molecules, such as Raf, JNK, p38 MAP, Map kinase kinase (MEK), MEK kinase, Src, Muscle specific kinase (MuSK), IRAK, Tec, and Janus, as well as any other phosphoserine, phosphotyrosine, or phosphothreonine molecule, can be detected by substituting these molecules for Erk-1 or Erk-2 in the following assay. [1224]
  • Specifically, assay plates are made by coating the wells of a 96-well ELISA plate with 0.1 ml of protein G (lug/ml) for 2 hr at room temp, (RT). The plates are then rinsed with PBS and blocked with 3% BSA/PBS for 1 hr at RT. The protein G plates are then treated with 2 commercial monoclonal antibodies (100 ng/well) against Erk-1 and Erk-2 (1 hr at RT) (Santa Cruz Biotechnology). (To detect other molecules, this step can easily be modified by substituting a monoclonal antibody detecting any of the above described molecules.) After 3-5 rinses with PBS, the plates are stored at 4 degree C. until use. [1225]
  • A431 cells are seeded at 20,000/well in a 96-well Loprodyne filterplate and cultured overnight in growth medium. The cells are then starved for 48 hr in basal medium (DMEM) and then treated with EGF (6 ng/well) or varying concentrations of the fusion protein of the invention for 5-20 minutes. The cells are then solubilized and extracts filtered directly into the assay plate. [1226]
  • After incubation with the extract for 1 hr at RT, the wells are again rinsed. As a positive control, a commercial preparation of MAP kinase (10 ng/well) is used in place of A431 extract. Plates are then treated with a commercial polyclonal (rabbit) antibody (lug/ml) which specifically recognizes the phosphorylated epitope of the Erk-1 and Erk-2 kinases (1 hr at RT). This antibody is biotinylated by standard procedures. The bound polyclonal antibody is then quantitated by successive incubations with Europium-streptavidin and Europium fluorescence enhancing reagent in the Wallac DELFIA instrument (time-resolved fluorescence). An increased fluorescent signal over background indicates a phosphorylation by the fusion protein of the present invention or a molecule induced by an albumin fusion protein of the present invention. [1227]
  • Example 37 Assay for the Stimulation of Bone Marrow CD34+ Cell Proliferation
  • This assay is based on the ability of human CD34+ to proliferate in the presence of hematopoietic growth factors and evaluates the ability of fusion proteins of the inventon to stimulate proliferation of CD34+ cells. [1228]
  • It has been previously shown that most mature precursors will respond to only a single signal. More immature precursors require at least two signals to respond. Therefore, to test the effect of fusion proteins of the invention on hematopoietic activity of a wide range of progenitor cells, the assay contains a given fusion protein of the invention in the presence or absence of hematopoietic growth factors. Isolated cells are cultured for 5 days in the presence of Stem Cell Factor (SCF) in combination with tested sample. SCF alone has a very limited effect on the proliferation of bone marrow (BM) cells, acting in such conditions only as a “survival” factor. However, combined with any factor exhibiting stimulatory effect on these cells (e.g., IL-3), SCF will cause a synergistic effect. Therefore, if the tested fusion protein has a stimulatory effect on hematopoietic progenitors, such activity can be easily detected. Since normal BM cells have a low level of cycling cells, it is likely that any inhibitory effect of a given fusion protein might not be detected. Accordingly, assays for an inhibitory effect on progenitors is preferably tested in cells that are first subjected to in vitro stimulation with SCF+IL+3, and then contacted with the compound that is being evaluated for inhibition of such induced proliferation. [1229]
  • Briefly, CD34+ cells are isolated using methods known in the art. The cells are thawed and resuspended in medium ([1230] QBSF 60 serum-free medium with 1% L-glutamine (500 ml) Quality Biological, Inc., Gaithersburg, Md. Cat# 160-204-101). After several gentle centrifugation steps at 200×g, cells are allowed to rest for one hour. The cell count is adjusted to 2.5×105 cells/ml. During this time, 100 μl of sterile water is added to the peripheral wells of a 96-well plate. The cytokines that can be tested with an albumin fusion protein of the invention in this assay is rhSCF (R&D Systems, Minneapolis, Minn., Cat# 255-SC) at 50 ng/ml alone and in combination with rhSCF and rhIL-3 (R&D Systems, Minneapolis, Minn., Cat# 203-ML) at 30 ng/ml. After one hour, 10 μl of prepared cytokines, varying concentrations of an albumin fusion protein of the invention, and 20 μl of diluted cells are added to the media which is already present in the wells to allow for a final total volume of 100 μl. The plates are then placed in a 37° C./5% CO2 incubator for five days.
  • Eighteen hours before the assay is harvested, 0.5 μCi/well of [3H] Thymidine is added in a 10 μl volume to each well to determine the proliferation rate. The experiment is terminated by harvesting the cells from each 96-well plate to a filtermat using the Tomtec Harvester 96. After harvesting, the filtermats are dried, trimmed and placed into OmniFilter assemblies consisting of one OmniFilter plate and one OmniFilter Tray. 60 μl Microscint is added to each well and the plate sealed with TopSeal-A press-on sealing film A bar code 15 sticker is affixed to the first plate for counting. The sealed plates are then loaded and the level of radioactivity determined via the Packard Top Count and the printed data collected for analysis. The level of radioactivity reflects the amount of cell proliferation. [1231]
  • The studies described in this example test the activity of a given fusion protein to stimulate bone marrow CD34+ cell proliferation. One skilled in the art could easily modify the exemplified studies to test the activity of fusion porteins and polynucleotides of the invention (e.g., gene therapy) as well as agonists and antagonists thereof. The ability of an albumin fusion protein of the invention to stimulate the proliferation of bone marrow CD34+ cells indicates that the albumin fusion protein and/or polynucleotides corresponding to the fusion protein are useful for the diagnosis and treatment of disorders affecting the immune system and hematopoiesis. Representative uses are described in the “Immune Activity” and “Infectious Disease” sections above, and elsewhere herein. [1232]
  • Example 38 Assay for Extracellular Matrix Enhanced Cell Response (EMECR)
  • The objective of the Extracellular Matrix Enhanced Cell Response (EMECR) assay is to evaluate the ability of fusion proteins of the invention to act on hematopoietic stem cells in the context of the extracellular matrix (ECM) induced signal. [1233]
  • Cells respond to the regulatory factors in the context of signal(s) received from the surrounding microenvironment. For example, fibroblasts, and endothelial and epithelial stem cells fail to replicate in the absence of signals from the ECM. Hematopoietic stem cells can undergo self-renewal in the bone marrow, but not in in vitro suspension culture. The ability of stem cells to undergo self-renewal in vitro is dependent upon their interaction with the stromal cells and the ECM protein fibronectin (fn). Adhesion of cells to fn is mediated by the α[1234] 51 and α41 integrin receptors, which are expressed by human and mouse hematopoietic stem cells. The factor(s) which integrate with the ECM environment and are responsible for stimulating stem cell self-renewal havea not yet been identified. Discovery of such factors should be of great interest in gene therapy and bone marrow transplant applications
  • Briefly, polystyrene, non tissue culture treated, 96-well plates are coated with fn fragment at a coating concentration of 0.2 μg/cm[1235] 2. Mouse bone marrow cells are plated (1,000 cells/well) in 0.2 ml of serum-free medium. Cells cultured in the presence of IL-3 (5 ng/ml)+SCF (50 ng/ml) would serve as the positive control, conditions under which little self-renewal but pronounced differentiation of the stem cells is to be expected. Albumin fusion proteins of the invention are tested with appropriate negative controls in the presence and absence of SCF (5.0 ng/ml), where volume of the administed composition containing the albumin fusion protein of the invention represents 10% of the total assay volume. The plated cells are then allowed to grow by incubating in a low oxygen environment (5% CO2, 7% O2, and 88% N2) tissue culture incubator for 7 days. The number of proliferating cells within the wells is then quantitated by measuring thymidine incorporation into cellular DNA. Verification of the positive hits in the assay will require phenotypic characterization of the cells, which can be accomplished by scaling up of the culture system and using appropriate antibody reagents against cell surface antigens and FACScan.
  • One skilled in the art could easily modify the exemplified studies to test the activity of albumin fusion proteins and polynucleotides of the invention (e.g., gene therapy). [1236]
  • If a particular fusion protein of the present invention is found to be a stimulator of hematopoietic progenitors, the fusion protein and polynucleotides corresponding to the fusion protein may be useful for example, in the diagnosis and treatment of disorders affecting the immune system and hematopoiesis. Representative uses are described in the “Immune Activity” and “Infectious Disease” sections above, and elsewhere herein. The fusion protein may also be useful in the expansion of stem cells and committed progenitors of various blood lineages, and in the differentiation and/or proliferation of various cell types. [1237]
  • Additionally, the albumin fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention, may also be employed to inhibit the proliferation and differentiation of hematopoietic cells and therefore may be employed to protect bone marrow stem cells from chemotherapeutic agents during chemotherapy. This antiproliferative effect may allow administration of higher doses of chemotherapeutic agents and, therefore, more effective chemotherapeutic treatment. [1238]
  • Moreover, fusion proteins of the invention and polynucleotides encoding albumin fusion proteins of the invention may also be useful for the treatment and diagnosis of hematopoietic related disorders such as, anemia, pancytopenia, leukopenia, thrombocytopenia or leukemia, since stromal cells are important in the production of cells of hematopoietic lineages. The uses include bone marrow cell ex-vivo culture, bone marrow transplantation, bone marrow reconstitution, radiotherapy or chemotherapy of neoplasia. [1239]
  • Example 39 Human Dermal Fibroblast and Aortic Smooth Muscle Cell Proliferation
  • An albumin fusion protein of the invention is added to cultures of normal human dermal fibroblasts (NHDF) and human aortic smooth muscle cells (AoSMC) and two co-assays are performed with each sample. The first assay examines the effect of the fusion protein on the proliferation of normal human dermal fibroblasts (NHDF) or aortic smooth muscle cells (AoSMC). Aberrant growth of fibroblasts or smooth muscle cells is a part of several pathological processes, including fibrosis, and restenosis. The second assay examines IL6 production by both NHDF and SMC. IL6 production is an indication of functional activation. Activated cells will have increased production of a number of cytokines and other factors, which can result in a proinflammatory or immunomodulatory outcome. Assays are run with and without co-TNFa stimulation, in order to check for costimulatory or inhibitory activity. [1240]
  • Briefly, on [1241] day 1, 96-well black plates are set up with 1000 cells/well (NHDF) or 2000 cells/well (AoSMC) in 100 μl culture media. NHDF culture media contains: Clonetics FB basal media, 1 mg/ml hFGF, 5 mg/ml insulin, 50 mg/ml gentamycin, 2%FBS, while AoSMC culture media contains Clonetics SM basal media, 0.5 μg/ml hEGF, 5 mg/ml insulin, 1 μg/ml hFGF, 50 mg/ml gentamycin, 50 μg/ml Amphotericin B, 5%FBS. After incubation at 37° C. for at least 4-5 hours culture media is aspirated and replaced with growth arrest media. Growth arrest media for NHDF contains fibroblast basal media, 50 mg/ml gentamycin, 2% FBS, while growth arrest media for AoSMC contains SM basal media, 50 mg/ml gentamycin, 50 μg/ml Amphotericin B, 0.4% FBS. Incubate at 37° C. until day 2.
  • On [1242] day 2, serial dilutions and templates of an albumin fusion protein of the invention are designed such that they always include media controls and known-protein controls. For both stimulation and inhibition experiments, proteins are diluted in growth arrest media. For inhibition experiments, TNFa is added to a final concentration of 2 ng/ml (NHDF) or 5 ng/ml (AoSMC). Add ⅓ vol media containing controls or an albumin fusion protein of the invention and incubate at 37 degrees C./5% CO2 until day 5.
  • [1243] Transfer 60 μl from each well to another labeled 96-well plate, cover with a plate-sealer, and store at 4 degrees C. until Day 6 (for IL6 ELISA). To the remaining 100 μl in the cell culture plate, aseptically add Alamar Blue in an amount equal to 10% of the culture volume (10 μl). Return plates to incubator for 3 to 4 hours. Then measure fluorescence with excitation at 530 nm and emission at 590 nm using the CytoFluor. This yields the growth stimulation/inhibition data.
  • On [1244] day 5, the IL6 ELISA is performed by coating a 96 well plate with 50-100 ul/well of Anti-Human IL6 Monoclonal antibody diluted in PBS, pH 7.4, incubate ON at room temperature.
  • On [1245] day 6, empty the plates into the sink and blot on paper towels. Prepare Assay Buffer containing PBS with 4% BSA. Block the plates with 200 μl/well of Pierce Super Block blocking buffer in PBS for 1-2 hr and then wash plates with wash buffer (PBS, 0.05% Tween-20). Blot plates on paper towels. Then add 50 μL/well of diluted Anti-Human IL-6 Monoclonal, Biotin-labeled antibody at 0.50 mg/ml. Make dilutions of IL-6 stock in media (30, 10, 3, 1, 0.3, 0 ng/ml). Add duplicate samples to top row of plate. Cover the plates and incubate for 2 hours at RT on shaker.
  • Plates are washed with wash buffer and blotted on paper towels. Dilute EU-labeled Streptavidin 1:1000 in Assay buffer, and add 100 μl/well. Cover the plate and incubate 1 h at RT. Plates are again washed with wash buffer and blotted on paper towels. [1246]
  • Add 100 μl/well of Enhancement Solution. Shake for 5 minutes. Read the plate on the Wallac DELFIA Fluorometer. Readings from triplicate samples in each assay were tabulated and averaged. [1247]
  • A positive result in this assay suggests AoSMC cell proliferation and that the albumin fusion protein may be involved in dermal fibroblast proliferation and/or smooth muscle cell proliferation. A positive result also suggests many potential uses of the fusion protein and polynucleotides encoding the albumin fusion protein. For example, inflammation and immune responses, wound healing, and angiogenesis, as detailed throughout this specification. Particularly, fusion proteins may be used in wound healing and dermal regeneration, as well as the promotion of vasculogenesis, both of the blood vessels and lymphatics. The growth of vessels can be used in the treatment of, for example, cardiovascular diseases. Additionally, fusion proteins showing antagonistic activity in this assay may be useful in treating diseases, disorders, and/or conditions which involve angiogenesis by acting as an anti-vascular agent (e.g., anti-angiogenesis). These diseases, disorders, and/or conditions are known in the art and/or are described herein, such as, for example, malignancies, solid tumors, benign tumors, for example hemangiomas, acoustic neuromas, neurofibromas, trachomas, and pyogenic granulomas; artheroscleric plaques; ocular angiogenic diseases, for example, diabetic retinopathy, retinopathy of prematurity, macular degeneration, corneal graft rejection, neovascular glaucoma, retrolental fibroplasia, rubeosis, retinoblastoma, uvietis and Pterygia (abnormal blood vessel growth) of the eye; rheumatoid arthritis; psoriasis; delayed wound healing; endometriosis; vasculogenesis; granulations; hypertrophic scars (keloids); nonunion fractures; scleroderma; trachoma; vascular adhesions; myocardial angiogenesis; coronary collaterals; cerebral collaterals; arterioyenous malformations; ischemic limb angiogenesis; Osler-Webber Syndrome; plaque neovascularization; telangiectasia; hemophiliac joints; angiofibroma; fibromuscular dysplasia; wound granulation; Crohn's disease; and atherosclerosis. Moreover, albumin fusion proteins that act as antagonists in this assay may be useful in treating anti-hyperproliferative diseases and/or anti-inflammatory known in the art and/or described herein. [1248]
  • Example 40 Cellular Adhesion Molecule (CAM) Expression on Endothelial Cells
  • The recruitment of lymphocytes to areas of inflammation and angiogenesis involves specific receptor-ligand interactions between cell surface adhesion molecules (CAMs) on lymphocytes and the vascular endothelium. The adhesion process, in both normal and pathological settings, follows a multi-step cascade that involves intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1), and endothelial leukocyte adhesion molecule-1 (E-selectin) expression on endothelial cells (EC). The expression of these molecules and others on the vascular endothelium determines the efficiency with which leukocytes may adhere to the local vasculature and extravasate into the local tissue during the development of an inflammatory response. The local concentration of cytokines and growth factor participate in the modulation of the expression of these CAMs. [1249]
  • Briefly, endothelial cells (e.g., Human Umbilical Vein Endothelial cells (HUVECs)) are grown in a standard 96 well plate to confluence, growth medium is removed from the cells and replaced with 100 μl of 199 Medium (10% fetal bovine serum (FBS)). Samples for testing (containing an albumin fusion protein of the invention) and positive or negative controls are added to the plate in triplicate (in 10 μl volumes). Plates are then incubated at 37° C. for either 5 h (selectin and integrin expression) or 24 h (integrin expression only). Plates are aspirated to remove medium and 100 μl of 0.1% paraformaldehyde-PBS (with Ca++ and Mg++) is added to each well. Plates are held at 4° C. for 30 min. Fixative is removed from the wells and wells are washed 1×with PBS (+Ca,Mg)+0.5% BSA and drained. 10 μl of diluted primary antibody is added to the test and control wells. Anti-ICAM-1-Biotin, Anti-VCAM-1-Biotin and Anti-E-selectin-Biotin are used at a concentration of 10 μg/ml (1:10 dilution of 0.1 mg/ml stock antibody). Cells are incubated at 37° C. for 30 min. in a humidified environment. Wells are washed three times with PBS (+Ca,Mg)+0.5% BSA. 20 μl of diluted ExtrAvidin-Alkaline Phosphatase (1:5,000 dilution, referred to herein as the working dilution) are added to each well and incubated at 37° C. for 30 min. Wells are washed three times with PBS (+Ca,Mg)+0.5% BSA. Dissolve 1 tablet of p-Nitrophenol Phosphate pNPP per 5 ml of glycine buffer (pH 10.4). 100 μl of pNPP substrate in glycine buffer is added to each test well. Standard wells in triplicate are prepared from the working dilution of the ExtrAvidin-Alkaline Phosphotase in glycine buffer: 1:5,000 (10[1250] 0)>10−0.5>10−1>10−1.5. 5 μl of each dilution is added to triplicate wells and the resulting AP content in each well is 5.50 ng, 1.74 ng, 0.55 ng, 0.18 ng. 100 μl of pNNP reagent is then added to each of the standard wells. The plate is incubated at 37° C. for 4 h. A volume of 50 μl of 3M NaOH is added to all wells. The plate is read on a plate reader at 405 nm using the background subtraction option on blank wells filled with glycine buffer only. Additionally, the template is set up to indicate the concentration of AP-conjugate in each standard well [5.50 ng; 1.74 ng; 0.55 ng; 0.18 ng]. Results are indicated as amount of bound AP-conjugate in each sample.
  • Example 41 Alamar Blue Endothelial Cells Proliferation Assay
  • This assay may be used to quantitatively determine protein mediated inhibition of bFGF-induced proliferation of Bovine Lymphatic Endothelial Cells (LECs), Bovine Aortic Endothelial Cells (BAECs) or Human Microvascular Uterine Myometrial Cells (UTMECs). This assay incorporates a fluorometric growth indicator based on detection of metabolic activity. A standard Alamar Blue Proliferation Assay is prepared in EGM-2MV with 10 ng/ml of bFGF added as a source of endothelial cell stimulation. This assay may be used with a variety of endothelial cells with slight changes in growth medium and cell concentration. Dilutions of protein batches to be tested are diluted as appropriate. Serum-free medium (GIBCO SFM) without bFGF is used as a non-stimulated control and Angiostatin or TSP-1 are included as a known inhibitory controls. [1251]
  • Briefly, LEC, BAECs or UTMECs are seeded in growth media at a density of 5000 to 2000 cells/well in a 96 well plate and placed at 37 degreesC overnight. After the overnight incubation of the cells, the growth media is removed and replaced with GIBCO EC-SFM. The cells are treated with the appropriate dilutions of an albumin fusion protein of the invention or control protein sample(s) (prepared in SFM) in triplicate wells with additional bFGF to a concentration of 10 ng/ml. Once the cells have been treated with the samples, the plate(s) is/are placed back in the 37° C. incubator for three days. After three [1252] days 10 ml of stock alamar blue (Biosource Cat# DAL1100) is added to each well and the plate(s) is/are placed back in the 37° C. incubator for four hours. The plate(s) are then read at 530 nm excitation and 590 nm emission using the CytoFluor fluorescence reader. Direct output is recorded in relative fluorescence units.
  • Alamar blue is an oxidation-reduction indicator that both fluoresces and changes color in response to chemical reduction of growth medium resulting from cell growth. As cells grow in culture, innate metabolic activity results in a chemical reduction of the immediate surrounding environment. Reduction related to growth causes the indicator to change from oxidized (non-fluorescent blue) form to reduced (fluorescent red) form (i.e., stimulated proliferation will produce a stronger signal and inhibited proliferation will produce a weaker signal and the total signal is proportional to the total number of cells as well as their metabolic activity). The background level of activity is observed with the starvation medium alone. This is compared to the output observed from the positive control samples (bFGF in growth medium) and protein dilutions. [1253]
  • Example 42 Detection of Inhibition of a Mixed Lymphocyte Reaction
  • This assay can be used to detect and evaluate inhibition of a Mixed Lymphocyte Reaction (MLR) by fusion proteins of the invention. Inhibition of a MLR may be due to a direct effect on cell proliferation and viability, modulation of costimulatory molecules on interacting cells, modulation of adhesiveness between lymphocytes and accessory cells, or modulation of cytokine production by accessory cells. Multiple cells may be targeted by the albumin fusion proteins that inhibit MLR since the peripheral blood mononuclear fraction used in this assay includes T, B and natural killer lymphocytes, as well as monocytes and dendritic cells. [1254]
  • Albumin fusion proteins of the invention found to inhibit the MLR may find application in diseases associated with lymphocyte and monocyte activation or proliferation. These include, but are not limited to, diseases such as asthma, arthritis, diabetes, inflammatory skin conditions, psoriasis, eczema, systemic lupus erythematosus, multiple sclerosis, glomerulonephritis, inflammatory bowel disease, crohn's disease, ulcerative colitis, arteriosclerosis, cirrhosis, graft vs. host disease, host vs. graft disease, hepatitis, leukemia and lymphoma. [1255]
  • Briefly, PBMCs from human donors are purified by density gradient centrifugation using Lymphocyte Separation Medium (LSM®, density 1.0770 g/ml, Organon Teknika Corporation, West Chester, Pa.). PBMCs from two donors are adjusted to 2×10[1256] 6 cells/ml in RPMI-1640 (Life Technologies, Grand Island, N.Y.) supplemented with 10% FCS and 2 mM glutamine. PBMCs from a third donor is adjusted to 2×105 cells/ml. Fifty microliters of PBMCs from each donor is added to wells of a 96-well round bottom microtiter plate. Dilutions of the fusion protein test material (50 μl) is added in triplicate to microtiter wells. Test samples (of the protein of interest) are added for final dilution of 1:4; rhuIL-2 (R&D Systems, Minneapolis, Minn., catalog number 202-IL) is added to a final concentration of 1 μg/ml; anti-CD4 mAb (R&D Systems, clone 34930.11, catalog number MAB379) is added to a final concentration of 10 μg/ml. Cells are cultured for 7-8 days at 37° C. in 5% CO2, and 1 μC of [3H] thymidine is added to wells for the last 16 hrs of culture. Cells are harvested and thymidine incorporation determined using a Packard TopCount. Data is expressed as the mean and standard deviation of triplicate determinations.
  • Samples of the fusion protein of interest are screened in separate experiments and compared to the negative control treatment, anti-CD4 mAb, which inhibits proliferation of lymphocytes and the positive control treatment, IL-2 (either as recombinant material or supernatant), which enhances proliferation of lymphocytes. [1257]
  • Example 43 Assays for Protease Activity
  • The following assay may be used to assess protease activity of an albumin fusion protein of the invention. [1258]
  • Gelatin and casein zymography are performed essentially as described (Heusen et al., [1259] Anal Biochem., 102:196-202 (1980); Wilson et al., Journal of Urology, 149:653-658 (1993)). Samples are run on 10% polyacryamide/0.1% SDS gels containing 1% gelain orcasein, soaked in 2.5% triton at room temperature for 1 hour, and in 0.1M glycine, pH 8.3 at 37° C. 5 to 16 hours. After staining in amido black areas of proteolysis apear as clear areas agains the blue-black background. Trypsin (Sigma T8642) is used as a positive control.
  • Protease activity is also determined by monitoring the cleavage of n-a-benzoyl-L-arginine ethyl ester (BAEE) (Sigma B-4500. Reactions are set up in (25 mMNaPO[1260] 4, 1 mM EDTA, and 1 mM BAEE), pH 7.5. Samples are added and the change in adsorbance at 260 nm is monitored on the Beckman DU-6 spectrophotometer in the time-drive mode. Trypsin is used as a positive control.
  • Additional assays based upon the release of acid-soluble peptides from casein or hemoglobin measured as adsorbance at 280 nm or calorimetrically using the Folin method are performed as described in Bergmeyer, et al., [1261] Methods of Enzymatic Analysis, 5 (1984). Other assays involve the solubilization of chromogenic substrates (Ward, Applied Science, 251-317 (1983)).
  • Example 44 Identifying Serine Protease Substrate Specificity
  • Methods known in the art or described herein may be used to determine the substrate specificity of the albumin fusion proteins of the present invention having serine protease activity. A preferred method of determining substrate specificity is by the use of positional scanning synthetic combinatorial libraries as described in [1262] GB 2 324 529 (incorporated herein in its entirety).
  • Example 45 Ligand Binding Assays
  • The following assay may be used to assess ligand binding activity of an albumin fusion protein of the invention. [1263]
  • Ligand binding assays provide a direct method for ascertaining receptor pharmacology and are adaptable to a high throughput format. The purified ligand for an albumin fusion protein of the invention is radiolabeled to high specific activity (50-2000 Ci/mmol) for binding studies. A determination is then made that the process of radiolabeling does not diminish the activity of the ligand towards the fusion protein. Assay conditions for buffers, ions, pH and other modulators such as nucleotides are optimized to establish a workable signal to noise ratio for both membrane and whole cell polypeptide sources. For these assays, specific polypeptide binding is defined as total associated radioactivity minus the radioactivity measured in the presence of an excess of unlabeled competing ligand. Where possible, more than one competing ligand is used to define residual nonspecific binding. [1264]
  • Example 46 Functional Assay in Xenopus Oocytes
  • Capped RNA transcripts from linearized plasmid templates encoding an albumin fusion protein of the invention is synthesized in vitro with RNA polymerases in accordance with standard procedures. In vitro transcripts are suspended in water at a final concentration of 0.2 mg/ml. Ovarian lobes are removed from adult female toads, Stage V defolliculated oocytes are obtained, and RNA transcripts (10 ng/oocytc) are injected in a 50 nl bolus using a microinjection apparatus. Two electrode voltage clamps are used to measure the currents from individual Xenopus oocytes in response fusion protein and polypeptide agonist exposure. Recordings are made in Ca2+ free Barth's medium at room temperature. The Xenopus system can be used to screen known ligands and tissue/cell extracts for activating ligands. [1265]
  • Example 47 Microphysiometric Assays
  • Activation of a wide variety of secondary messenger systems results in extrusion of small amounts of acid from a cell. The acid formed is largely as a result of the increased metabolic activity required to fuel the intracellular signaling process. The pH changes in the media surrounding the cell are very small but are detectable by the CYTOSENSOR microphysiometer (Molecular Devices Ltd., Menlo Park, Calif.). The CYTOSENSOR is thus capable of detecting the ability of an albumin fusion protein of the invention to activate secondary messengers that are coupled to an energy utilizing intracellular signaling pathway. [1266]
  • Example 48 Extract/Cell Supernatant Screening
  • A large number of mammalian receptors exist for which there remains, as yet, no cognate activating ligand (agonist). Thus, active ligands for these receptors may not be included within the ligands banks as identified to date. Accordingly, the albumin fusion proteins of the invention can also be functionally screened (using calcium, cAMP, microphysiometer, oocyte electrophysiology, etc., functional screens) against tissue extracts to identify natural ligands for the Therapeutic protein portion and/or albumin protein portion of an albumin fusion protein of the invention. Extracts that produce positive functional responses can be sequentially subfractionated until an activating ligand is isolated and identified. [1267]
  • Example 49 ATP-Binding Assay
  • The following assay may be used to assess ATP-binding activity of fusion proteins of the invention. [1268]
  • ATP-binding activity of an albumin fusion protein of the invention may be detected using the ATP-binding assay described in U.S. Pat. No. 5,858,719, which is herein incorporated by reference in its entirety. Briefly, ATP-binding to an albumin fusion protein of the invention is measured via photoaffinity labeling with 8-azido-ATP in a competition assay. Reaction mixtures containing 1 mg/ml of ABC transport protein are incubated with varying concentrations of ATP, or the non-hydrolyzable ATP analog adenyl-5′-imidodiphosphate for 10 minutes at 4° C. A mixture of 8-azido-ATP (Sigma Chem. Corp., St. Louis, Mo.) plus 8-azido-ATP ([1269] 32P-ATP) (5 mCi/μmol, ICN, Irvine Calif.) is added to a final concentration of 100 μM and 0.5 ml aliquots are placed in the wells of a porcelain spot plate on ice. The plate is irradiated using a short wave 254 nm UV lamp at a distance of 2.5 cm from the plate for two one-minute intervals with a one-minute cooling interval in between. The reaction is stopped by addition of dithiothreitol to a final concentration of 2 mM. The incubations are subjected to SDS-PAGE electrophoresis, dried, and autoradiographed. Protein bands corresponding to the albumin fusion proteins of the invention are excised, and the radioactivity quantified. A decrease in radioactivity with increasing ATP or adenly-5′-imidodiphosphate provides a measure of ATP affinity to the fusion protein.
  • Example 50 Phosphorylation Assay
  • In order to assay for phosphorylation activity of an albumin fusion protein of the invention, a phosphorylation assay as described in U.S. Pat. No. 5,958,405 (which is herein incorporated by reference) is utilized. Briefly, phosphorylation activity may be measured by phosphorylation of a protein substrate using gamma-labeled [1270] 32P-ATP and quantitation of the incorporated radioactivity using a gamma radioisotope counter. The fusion portein of the invention is incubated with the protein substrate, 32P-ATP, and a kinase buffer. The incorporated into the substrate is then separated from free 32P-ATP by electrophoresis, and the incorporated 32P is counted and compared to a negative control. Radioactivity counts above the negative control are indicative of phosphorylation activity of the fusion protein.
  • Example 51 Detection of Phosphorylation Activity (Activation) of an Albumin Fusion Protein of the Invention in the Presence of Polypeptide Ligands
  • Methods known in the art or described herein may be used to determine the phosphorylation activity of an albumin fusion protein of the invention. A preferred method of determining phosphorylation activity is by the use of the tyrosine phosphorylation assay as described in U.S. Pat. No. 5,817,471 (incorporated herein by reference). [1271]
  • Example 52 Identification of Signal Transduction Proteins that Interact with an Albumin Fusion Protein of the Present Invention
  • Albumin fusion proteins of the invention may serve as research tools for the identification, characterization and purification of signal transduction pathway proteins or receptor proteins. Briefly, a labeled fusion protein of the invention is useful as a reagent for the purification of molecules with which it interacts. In one embodiment of affinity purification, an albumin fusion protein of the invention is covalently coupled to a chromatography column. Cell-free extract derived from putative target cells, such as carcinoma tissues, is passed over the column, and molecules with appropriate affinity bind to the albumin fusion protein. The protein complex is recovered from the column, dissociated, and the recovered molecule subjected to N-terminal protein sequencing. This amino acid sequence is then used to identify the captured molecule or to design degenerate oligonucleotide probes for cloning the relevant gene from an appropriate cDNA library. [1272]
  • Example 53 IL-6 Bioassay
  • A variety of assays are known in the art for testing the proliferative effects of an albumin fusion protein of the invention. For example, one such asssay is the IL-6 Bioassay as described by Marz et al. ([1273] Proc. Natl. Acad. Sci., U.S.A., 95:3251-56 (1998), which is herein incorporated by reference). After 68 hrs. at 37° C., the number of viable cells is measured by adding the tetrazolium salt thiazolyl blue (MTT) and incubating for a further 4 hrs. at 37° C. B9 cells are lysed by SDS and optical density is measured at 570 nm. Controls containing IL-6 (positive) and no cytokine (negative) are Briefly, IL-6 dependent B9 murine cells are washed three times in IL-6 free medium and plated at a concentration of 5,000 cells per well in 50 μl, and 50 μl of fusion protein of the invention is added utilized. Enhanced proliferation in the test sample(s) (containing an albumin fusion protein of the invention) relative to the negative control is indicative of proliferative effects mediated by the fusion protein.
  • Example 54 Support of Chicken Embryo Neuron Survival
  • To test whether sympathetic neuronal cell viability is supported by an albumin fusion protein of the invention, the chicken embryo neuronal survival assay of Senaldi et al may be utilized ([1274] Proc. Natl. Acad. Sci., U.S.A., 96:11458-63 (1998), which is herein incorporated by reference). Briefly, motor and sympathetic neurons are isolated from chicken embryos, resuspended in L15 medium (with 10% FCS, glucose, sodium selenite, progesterone, conalbumin, putrescine, and insulin; Life Technologies, Rockville, Md.) and Dulbecco's modified Eagles medium [with 10% FCS, glutamine, penicillin, and 25 mM Hepes buffer (pH 7.2); Life Technologies, Rockville, Md.], respectively, and incubated at 37° C. in 5% CO2 in the presence of different concentrations of the purified fusion protein of the invention, as well as a negative control lacking any cytokine. After 3 days, neuron survival is determined by evaluation of cellular morphology, and through the use of the colorimetric assay of Mosmann (Mosmann, T., J. Immunol. Methods, 65:55-63 (1983)). Enhanced neuronal cell viability as compared to the controls lacking cytokine is indicative of the ability of the albumin fusion protein to enhance the survival of neuronal cells.
  • Example 55 Assay for Phosphatase Activity
  • The following assay may be used to assess serine/threonine phosphatase (PTPase) activity of an albumin fusion protein of the invention. [1275]
  • In order to assay for serine/threonine phosphatase (PTPase) activity, assays can be utilized which are widely known to those skilled in the art. For example, the serine/threonine phosphatase (PSPase) activity of an albumin fusion protein of the invention may be measured using a PSPase assay kit from New England Biolabs, Inc. Myelin basic protein (MyBP), a substrate for PSPase, is phosphorylated on serine and threonine residues with cAMP-dependent Protein Kinase in the presence of [[1276] 32P]ATP. Protein serine/threonine phosphatase activity is then determined by measuring the release of inorganic phosphate from 32P-labeled MyBP.
  • Example 56 Interaction of Serine/Threonine Phosphatases with Other Proteins
  • Fusion protein of the invention having serine/threonine phosphatase activity (e.g., as determined in Example 55) are useful, for example, as research tools for the identification, characterization and purification of additional interacting proteins or receptor proteins, or other signal transduction pathway proteins. Briefly, a labeled fusion protein of the invention is useful as a reagent for the purification of molecules with which it interacts. In one embodiment of affinity purification, an albumin fusion protein of the invention is covalently coupled to a chromatography column. Cell-free extract derived from putative target cells, such as neural or liver cells, is passed over the column, and molecules with appropriate affinity bind to the fusion protein. The fusion protein-complex is recovered from the column, dissociated, and the recovered molecule subjected to N-terminal protein sequencing. This amino acid sequence is then used to identify the captured molecule or to design degenerate oligonucleotide probes for cloning the relevant gene from an appropriate cDNA library. [1277]
  • Example 57 Assaying for Heparanase Activity
  • There a numerous assays known in the art that may be employed to assay for heparanase activity of an albumin fusion protein of the invention. In one example, heparanase activity of an albumin fusion protein of the invention, is assayed as described by Vlodavsky et al., (Vlodavsky et al., Nat. Med., 5:793-802 (1999)). Briefly, cell lysates, conditioned media, intact cells (1×10[1278] 6 cells per 35-mm dish), cell culture supernatant, or purified fusion protein are incubated for 18 hrs at 37° C., pH 6.2-6.6, with 35S-labeled ECM or soluble ECM derived peak I proteoglycans. The incubation medium is centrifuged and the supernatant is analyzed by gel filtration on a Sepharose CL-6B column (0.9×30 cm). Fractions are eluted with PBS and their radioactivity is measured. Degradation fragments of heparan sulfate side chains are eluted from Sepharose 6B at 0.5<Kav<0.8 (peak II). Each experiment is done at least three times. Degradation fragments corresponding to “peak II,” as described by Vlodavsky et al., is indicative of the activity of an albumin fusion protein of the invention in cleaving heparan sulfate.
  • Example 58 Immobilization of Biomolecules
  • This example provides a method for the stabilization of an albumin fusion protein of the invention in non-host cell lipid bilayer constucts (see, e.g., Bieri et al., Nature Biotech 17:1105-1108 (1999), hereby incorporated by reference in its entirety herein) which can be adapted for the study of fusion proteins of the invention in the various functional assays described above. Briefly, carbohydrate-specific chemistry for biotinylation is used to confine a biotin tag to an albumin fusion protein of the invention, thus allowing uniform orientation upon immobilization. A 50 uM solution of an albumin fusion protein of the invention in washed membranes is incubated with 20 mM NaIO4 and 1.5 mg/ml (4 mM) BACH or 2 mg/ml (7.5 mM) biotin-hydrazide for 1 hr at room temperature (reaction volume, 150 ul). Then the sample is dialyzed (Pierce Slidealizer Cassett, 10 kDa cutoff; Pierce Chemical Co., Rockford Ill.) at 4C first for 5 h, exchanging the buffer after each hour, and finally for 12 h against 500 ml buffer R (0.15 M NaCl, 1 mM MgCl2, 10 mM sodium phosphate, pH 7). Just before addition into a cuvette, the sample is diluted 1:5 in buffer ROG50 (Buffer R supplemented with 50 mM octylglucoside). [1279]
  • Example 59 Assays for Metalloproteinase Activity
  • Metalloproteinases are peptide hydrolases which use metal ions, such as Zn[1280] 2+, as the catalytic mechanism. Metalloproteinase activity of an albumin fusion protein of the present invention can be assayed according to methods known in the art. The following exemplary methods are provided:
  • Proteolysis of Alpha-2-Macroglobulin [1281]
  • To confirm protease activity, a purified fusion protein of the invention is mixed with the substrate alpha-2-macroglobulin (0.2 unit/ml; Boehringer Mannheim, Germany) in 1×assay buffer (50 mM HEPES, pH 7.5, 0.2 M NaCl, 10 mM CaCl[1282] 2, 25 μM ZnCl2 and 0.05% Brij-35) and incubated at 37° C. for 1-5 days. Trypsin is used as positive control. Negative controls contain only alpha-2-macroglobulin in assay buffer. The samples are collected and boiled in SDS-PAGE sample buffer containing 5% 2-mercaptoethanol for 5-min, then loaded onto 8% SDS-polyacrylamide gel. After electrophoresis the proteins are visualized by silver staining. Proteolysis is evident by the appearance of lower molecular weight bands as compared to the negative control.
  • Inhibition of Alpha-2-Macroglobulin Proteolysis by Inhibitors of Metalloproteinases [1283]
  • Known metalloproteinase inhibitors (metal chelators (EDTA, EGTA, AND HgCl[1284] 2), peptide metalloproteinase inhibitors (TIMP-1 and TIMP-2), and commercial small molecule MMP inhibitors) may also be used to characterize the proteolytic activity of an albumin fusion protein of the invention. Three synthetic MMP inhibitors that may be used are: MMP inhibitor I, [IC50=1.0 μM against MMP-1 and MMP-8; IC50=30 μM against MMP-9; IC50=150 μM against MMP-3]; MMP-3 (stromelysin-1) inhibitor I [IC50=5 μM against MMP-3], and MMP-3 inhibitor II [Kl=130 nM against MMP-3]; inhibitors available through Calbiochem, catalog # 444250, 444218, and 444225, respectively). Briefly, different concentrations of the small molecule MMP inhibitors are mixed with a purified fusion protein of the invention (50 μg/ml) in 22.9 μl of 1×HEPES buffer (50 mM HEPES, pH 7.5, 0.2 M NaCl, 10 mM CaCl2, 25 μM ZnCl2 and 0.05%Brij-35) and incubated at room temperature (24° C.) for 2-hr, then 7.1 μl of substrate alpha-2-macroglobulin (0.2 unit/ml) is added and incubated at 37° C. for 20-hr. The reactions are stopped by adding 4×sample buffer and boiled immediately for 5 minutes. After SDS-PAGE, the protein bands are visualized by silver stain.
  • Synthetic Fluorogenic Peptide Substrates Cleavage Assay [1285]
  • The substrate specificity for fusion proteins of the invention with demonstrated metalloproteinase activity may be determined using techniques knonw in the art, such as using synthetic fluorogenic peptide substrates (purchased from BACHEM Bioscience Inc). Test substrates include, M-1985, M-2225, M-2105, M-2110, and M-2255. The first four are MMP substrates and the last one is a substrate of tumor necrosis factor-α (TNF-α) converting enzyme (TACE). These substrastes are preferably prepared in 1:1 dimethyl sulfoxide (DMSO) and water. The stock solutions are 50-500 μM. Fluorescent assays are performed by using a Perkin Elmer LS 50B luminescence spectrometer equipped with a constant temperature water bath. The excitation λ is 328 nm and the emission k is 393 nm. Briefly, the assay is carried out by incubating 176 [1286] μl 1×HEPES buffer (0.2 M NaCl, 10 mM CaCl2, 0.05% Brij-35 and 50 mM HEPES, pH 7.5) with 4 μl of substrate solution (50 μM) at 25° C. for 15 minutes, and then adding 20 μl of a purified fusion protein of the invention into the assay cuvett. The final concentration of substrate is 1 μM. Initial hydrolysis rates are monitored for 30-min.
  • Example 60 Identification and Cloning of VH and VL Domains
  • One method to identfy and clone VH and VL domains from cell lines expressing a particular antibody is to perform PCR with VH and VL specific primers on cDNA made from the antibody expressing cell lines. Briefly, RNA is isolated from the cell lines and used as a template for RT-PCR designed to amplify the VH and VL domains of the antibodies expressed by the EBV cell lines. Cells may be lysed in the TRIzol® reagent (Life Technologies, Rockville. MD) and extracted with one fifth volume of chloroform. After addition of chloroform, the solution is allowed to incubate at room temperature for 10 minutes, and the centrifuged at 14,000 rpm for 15 minutes at 4° C. in a tabletop centrifuge. The supernatant is collected and RNA is precipitated using an equal volume of isopropanol. Precipitated RNA is pelleted by centrifuging at 14,000 rpm for 15 minutes at 4° C. in a tabletop centrifuge. Following centrifugation, the supernatant is discarded and washed with 75% ethanol. Follwing washing, the RNA is centrifuged again at 800 rpm for 5 minutes at 4° C. The supernatant is discarded and the pellet allowed to air dry. RNA is the dissolved in DEPC water and heated to 60° C. for 10 minutes. Quantities of RNA can determined using optical density measurements. [1287]
  • cDNA may be synthesized, according to methods well-known in the art, from 1.5-2.5 micrograms of RNA using reverse transciptase and random hexamer primers. cDNA is then used as a template for PCR amplification of VH and VL domains. Primers used to amplify VH and VL genes are shown in Table 3. Typically a PCR reaction makes use of a single 5′ primer and a single 3′ primer. Sometimes, when the amount of available RNA template is limiting, or for greater efficiency, groups of 5′ and/or 3′ primers may be used. For example, sometimes all five VH-5′ primers and all JH3′ primers are used in a single PCR reaction. The PCR reaction is carried out in a 50 microliter volume containing 1×PCR buffer, 2 mM of each dNTP, 0.7 units of High Fidelity Taq polymerse, 5′ primer mix, 3′ primer mix and 7.5 microliters of cDNA. The 5′ and 3′ primer mix of both VH and VL can be made by pooling together 22 pmole and 28 pmole, respectively, of each of the individual primers. PCR conditions are: 96° C. for 5 minutes; followed by 25 cycles of 94° C. for 1 minute, 50° C. for 1 minute, and 72° C. for 1 minute; followed by an extension cycle of 72° C. for 10 minutes. After the reaction is completed, sample tubes are stored 4° C. [1288]
    TABLE 3
    Primer Sequences Used to Amplify VH and VL
    domains.
    Primer name SEQ ID NO Primer Sequence (5′-3′)
    VH Primers
    Hu VH1-5′ 36 CAGGTGCAGCTGGTGCAGTCTGG
    Hu VH2-5′ 37 CAGGTCAACTTAAGGGAGTCTGG
    Hu VH3-5′ 38 GAGGTGCAGCTGGTGGAGTCTGG
    Hu VH4-5′ 39 CAGGTGCAGCTGCAGGAGTCGGG
    Hu VH5-5′ 40 GAGGTGCAGCTGTTGCAGTCTGC
    Hu VH6-5′ 41 CAGGTACAGCTGCAGCAGTCAGG
    Hu JH1,2-5′ 42 TGAGGAGACGGTGACCAGGGTGCC
    Hu JH3-5′ 43 TGAAGAGACGGTGACCATTGTCCC
    Hu JH4,5-5′ 44 TGAGGAGACGGTGACCAGGGTTCC
    Hu JH6-5′ 45 TGAGGAGACGGTGACCGTGGTCCC
    VL Primers
    Hu Vkappa1-5′ 46 GACATCCAGATGACCCAGTCTCC
    Hu Vkappa2a-5′ 47 GATGTTGTGATGACTCAGTCTCC
    Hu Vkappa2b-5′ 48 GATATTGTGATGACTCAGTCTCC
    Hu Vkappa3-5′ 49 GAAATTGTGTTGACGCAGTCTCC
    Hu Vkappa4-5′ 50 GACATCGTGATGACCCAGTCTCC
    Hu Vkappa5-5′ 51 GAAACGACACTCACGCAGTCTCC
    Hu Vkappa6-5′ 52 GAAATTGTGCTGACTCAGTCTCC
    Hu Vlambda1-5′ 53 CAGTCTGTGTTGACGCAGCCGCC
    Hu Vlambda2-5′ 54 CAGTCTGCCCTGACTCAGCCTGC
    Hu Vlambda3-5′ 55 TCCTATGTGCTGACTCAGCCACC
    Hu Vlambda3b-5′ 56 TCTTCTGAGCTGACTCAGGACCC
    Hu Vlambda4-5′ 57 CACGTTATACTGACTCAACCGCC
    Hu Vlambda5-5′ 58 CAGGCTGTGCTCACTCAGCCGTC
    Hu Vlambda6-5′ 59 AATTTTATGCTGACTCAGCCCCA
    Hu Jkappa1-3′ 60 ACGTTTGATTTCCACCTTGGTCCC
    Hu Jkappa2-3′ 61 ACGTTTGATCTCCAGCTTGGTCCC
    Hu Jkappa3-3′ 62 ACGTTTGATATCCACTTTGGTCCC
    Hu Jkappa4-3′ 63 ACGTTTGATCTCCACCTTGGTCCC
    Hu Jkappa5-3′ 64 ACGTTTAATCTCCAGTCGTGTCCC
    Hu Jlambda2-3′ 65 CAGTCTGTGTTGACGCAGCCGCC
    Hu Jlambda2-3′ 66 CAGTCTGCCCTGACTCAGCCTGC
    Hu Jlambda3-−3′ 67 TCCTATGTGCTGACTCAGCCACC
    Hu Jlambda3b-3′ 68 TCTTCTGAGCTGACTCAGGACCC
    Hu Jlambda4-3′ 69 CACGTTATACTGACTCAACCGCC
    Hu Jlambda5-3′ 70 CAGGCTGTGCTCACTCAGCCGTC
    Hu Jlambda6-3′ 71 AATTTTATGCTGACTCAGCCCCA
  • PCR samples are then electrophoresed on a 1.3% agarose gel. DNA bands of the expected sizes (˜506 base pairs for VH domains, and 344 base pairs for VL domains) can be cut out of the gel and purified using methods well known in the art. Purified PCR products can be ligated into a PCR cloning vector (TA vector from Invitrogen Inc., Carlsbad, Calif.). Individual cloned PCR products can be isolated after transfection of [1289] E. coli and blue/white color selection. Cloned PCR products may then be sequenced using methods commonly known in the art.
  • The PCR bands containing the VH domain and the VL domains can also be used to create full-length Ig expression vectors. VH and VL domains can be cloned into vectors containing the nucleotide sequences of a heavy (e.g., human IgG1 or human IgG4) or light chain (human kappa or human lambda) constant regions such that a complete heavy or light chain molecule could be expressed from these vectors when transfected into an appropriate host cell. Further, when cloned heavy and light chains are both expressed in one cell line (from either one or two vectors), they can assemble into a complete functional antibody molecule that is secreted into the cell culture medium. Methods using polynucleotides encoding VH and VL antibody domain to generate expression vectors that encode complete antibody molecules are well known within the art. [1290]
  • It will be clear that the invention may be practiced otherwise than as particularly described in the foregoing description and examples. Numerous modifications and variations of the present invention are possible in light of the above teachings and, therefore, are within the scope of the appended claims. [1291]
  • The entire disclosure of each document cited (including patents, patent applications, patent publications, journal articles, abstracts, laboratory manuals, books, or other disclosures) as well as information available through Identifiers specific to databases such as GenBank, GeneSeq, or the CAS Registry, referred to in this application are herein incorporated by reference in their entirety. The specification and sequence listing of each of the following U.S. applications are herein incorporated by reference in their entirety: [1292]
    ApplNumber File Date
    60/047,599 23-May-1997
    60/043,576 11-Apr-1997
    60/043,578 11-Apr-1997
    60/043,568 11-Apr-1997
    60/040,162 07-Mar-1997
    60/047,601 23-May-1997
    60/047,632 23-May-1997
    60/047,595 23-May-1997
    60/056,845 22-Aug-1997
    60/056,664 22-Aug-1997
    60/056,892 22-Aug-1997
    60/056,632 22-Aug-1997
    60/043,580 11-Apr-1997
    60/043,314 11-Apr-1997
    60/056,631 22-Aug-1997
    60/043,674 11-Apr-1997
    60/043,670 11-Apr-1997
    60/043,569 11-Apr-1997
    60/043,671 11-Apr-1997
    60/047,596 23-May-1997
    60/056,876 22-Aug-1997
    60/047,612 23-May-1997
    60/047,588 23-May-1997
    60/056,864 22-Aug-1997
    60/043,311 11-Apr-1997
    60/040,333 07-Mar-1997
    60/043,312 11-Apr-1997
    60/056,909 22-Aug-1997
    60/056,910 22-Aug-1997
    60/056,636 22-Aug-1997
    60/047,586 23-May-1997
    60/043,315 11-Apr-1997
    60/043,669 11-Apr-1997
    60/043,672 11-Apr-1997
    60/047,585 23-May-1997
    60/047,598 23-May-1997
    60/056,874 22-Aug-1997
    60/047,582 23-May-1997
    60/056,881 22-Aug-1997
    60/043,313 11-Apr-1997
    60/047,613 23-May-1997
    60/038,621 07-Mar-1997
    60/040,161 07-Mar-1997
    60/056,911 22-Aug-1997
    60/047,492 23-May-1997
    60/047,584 23-May-1997
    60/047,500 23-May-1997
    60/047,587 23-May-1997
    60/056,880 22-Aug-1997
    60/056,894 22-Aug-1997
    60/056,879 22-Aug-1997
    60/047,590 23-May-1997
    60/047,581 23-May-1997
    60/047,592 23-May-1997
    60/056,637 22-Aug-1997
    60/047,503 23-May-1997
    60/056,903 22-Aug-1997
    60/056,888 22-Aug-1997
    60/040,626 07-Mar-1997
    60/056,882 22-Aug-1997
    60/056,875 22-Aug-1997
    60/047,501 23-May-1997
    60/056,878 22-Aug-1997
    60/056,662 22-Aug-1997
    60/056,872 22-Aug-1997
    60/047,589 23-May-1997
    60/056,884 22-Aug-1997
    60/056,862 22-Aug-1997
    60/040,334 07-Mar-1997
    60/047,583 23-May-1997
    60/047,618 23-May-1997
    60/047,594 23-May-1997
    60/047,617 23-May-1997
    60/047,633 23-May-1997
    60/047,593 23-May-1997
    60/056,630 22-Aug-1997
    60/047,502 23-May-1997
    60/040,336 07-Mar-1997
    60/056,887 22-Aug-1997
    60/056,893 22-Aug-1997
    60/056,889 22-Aug-1997
    60/047,614 23-May-1997
    60/047,615 23-May-1997
    60/040,163 07-Mar-1997
    60/047,597 23-May-1997
    60/056,877 22-Aug-1997
    60/047,600 23-May-1997
    60/056,908 22-Aug-1997
    60/056,886 22-Aug-1997
    60/040,710 14-Mar-1997
    60/050,934 30-May-1997
    60/048,100 30-May-1997
    60/048,189 30-May-1997
    60/048,357 30-May-1997
    60/040,762 14-Mar-1997
    60/041,277 21-Mar-1997
    60/048,135 30-May-1997
    60/048,350 30-May-1997
    60/048,094 30-May-1997
    60/048,188 30-May-1997
    60/048,099 30-May-1997
    60/050,937 30-May-1997
    60/042,344 21-Mar-1997
    60/048,352 30-May-1997
    60/048,187 30-May-1997
    60/048,186 30-May-1997
    60/048,095 30-May-1997
    60/048,131 30-May-1997
    60/048,069 30-May-1997
    60/041,276 21-Mar-1997
    60/048,096 30-May-1997
    60/048,154 30-May-1997
    60/048,160 30-May-1997
    60/041,281 21-Mar-1997
    60/048,355 30-May-1997
    60/048,351 30-May-1997
    60/048,068 30-May-1997
    60/042,825 08-Apr-1997
    60/048,184 30-May-1997
    60/042,727 08-Apr-1997
    60/048,070 30-May-1997
    60/042,728 08-Apr-1997
    60/042,726 08-Apr-1997
    60/042,754 08-Apr-1997
    60/048,190 30-May-1997
    60/044,039 30-May-1997
    60/048,093 30-May-1997
    60/057,645 05-Sep-1997
    60/049,375 06-Jun-1997
    60/048,885 06-Jun-1997
    60/057,668 05-Sep-1997
    60/057,642 05-Sep-1997
    60/057,635 05-Sep-1997
    60/048,881 06-Jun-1997
    60/048,880 06-Jun-1997
    60/049,020 06-Jun-1997
    60/057,667 05-Sep-1997
    60/057,666 05-Sep-1997
    60/048,876 06-Jun-1997
    60/057,627 05-Sep-1997
    60/048,896 06-Jun-1997
    60/057,764 05-Sep-1997
    60/048,895 06-Jun-1997
    60/048,884 06-Jun-1997
    60/057,769 05-Sep-1997
    60/057,643 05-Sep-1997
    60/048,894 06-Jun-1997
    60/048,971 06-Jun-1997
    60/057,763 05-Sep-1997
    60/057,584 05-Sep-1997
    60/048,964 06-Jun-1997
    60/048,893 06-Jun-1997
    60/057,661 05-Sep-1997
    60/048,899 06-Jun-1997
    60/057,647 05-Sep-1997
    60/048,882 06-Jun-1997
    60/057,650 05-Sep-1997
    60/048,892 06-Jun-1997
    60/048,901 06-Jun-1997
    60/048,900 06-Jun-1997
    60/057,646 05-Sep-1997
    60/057,651 05-Sep-1997
    60/057,662 05-Sep-1997
    60/048,915 06-Jun-1997
    60/057,654 05-Sep-1997
    60/048,916 06-Jun-1997
    60/048,972 06-Jun-1997
    60/057,775 05-Sep-1997
    60/057,644 05-Sep-1997
    60/049,019 06-Jun-1997
    60/057,765 05-Sep-1997
    60/048,970 06-Jun-1997
    60/057,762 05-Sep-1997
    60/057,774 05-Sep-1997
    60/057,649 05-Sep-1997
    60/057,648 05-Sep-1997
    60/049,374 06-Jun-1997
    60/048,875 06-Jun-1997
    60/049,373 06-Jun-1997
    60/057,771 05-Sep-1997
    60/048,917 06-Jun-1997
    60/048,949 06-Jun-1997
    60/057,770 05-Sep-1997
    60/048,897 06-Jun-1997
    60/057,761 05-Sep-1997
    60/057,776 05-Sep-1997
    60/057,760 05-Sep-1997
    60/048,974 06-Jun-1997
    60/048,883 06-Jun-1997
    60/048,898 06-Jun-1997
    60/057,778 05-Sep-1997
    60/057,629 05-Sep-1997
    60/057,777 05-Sep-1997
    60/048,962 06-Jun-1997
    60/048,963 06-Jun-1997
    60/048,878 06-Jun-1997
    60/048,877 06-Jun-1997
    60/057,628 05-Sep-1997
    60/057,634 05-Sep-1997
    60/058,669 12-Sep-1997
    60/058,668 12-Sep-1997
    60/058,750 12-Sep-1997
    60/049,566 13-Jun-1997
    60/052,989 13-Jun-1997
    60/049,608 13-Jun-1997
    60/049,609 13-Jun-1997
    60/049,607 13-Jun-1997
    60/058,665 12-Sep-1997
    60/058,971 12-Sep-1997
    60/058,972 12-Sep-1997
    60/058,975 12-Sep-1997
    60/049,611 13-Jun-1997
    60/050,901 13-Jun-1997
    60/056,250 29-Aug-1997
    60/056,296 29-Aug-1997
    60/056,293 29-Aug-1997
    60/048,356 30-May-1997
    60/048,101 30-May-1997
    60/050,935 30-May-1997
    60/049,550 13-Jun-1997
    60/049,610 13-Jun-1997
    60/060,841 02-Oct-1997
    60/060,834 02-Oct-1997
    60/061,060 02-Oct-1997
    60/049,606 13-Jun-1997
    60/049,548 13-Jun-1997
    60/049,549 13-Jun-1997
    60/060,865 02-Oct-1997
    60/060,844 02-Oct-1997
    60/049,547 13-Jun-1997
    60/061,059 02-Oct-1997
    60/051,480 01-Jul-1997
    60/051,381 01-Jul-1997
    60/058,663 12-Sep-1997
    60/058,598 12-Sep-1997
    60/051,925 08-Jul-1997
    60/052,793 08-Jul-1997
    60/058,664 12-Sep-1997
    60/058,660 12-Sep-1997
    60/058,661 12-Sep-1997
    60/051,926 08-Jul-1997
    60/051,929 08-Jul-1997
    60/058,785 12-Sep-1997
    60/055,722 18-Aug-1997
    60/051,932 08-Jul-1997
    60/052,732 08-Jul-1997
    60/052,803 08-Jul-1997
    60/055,949 18-Aug-1997
    60/055,948 18-Aug-1997
    60/055,723 18-Aug-1997
    60/051,931 08-Jul-1997
    60/051,930 08-Jul-1997
    60/051,920 08-Jul-1997
    60/051,918 08-Jul-1997
    60/051,916 08-Jul-1997
    60/055,953 18-Aug-1997
    60/055,950 18-Aug-1997
    60/055,947 18-Aug-1997
    60/055,964 18-Aug-1997
    60/056,360 18-Aug-1997
    60/055,954 18-Aug-1997
    60/055,984 18-Aug-1997
    60/055,684 18-Aug-1997
    60/052,795 08-Jul-1997
    60/052,733 08-Jul-1997
    60/051,928 08-Jul-1997
    60/051,919 08-Jul-1997
    60/055,952 18-Aug-1997
    60/055,985 18-Aug-1997
    60/056,359 18-Aug-1997
    60/055,725 18-Aug-1997
    60/052,661 16-Jul-1997
    60/052,870 16-Jul-1997
    60/052,871 16-Jul-1997
    60/052,872 16-Jul-1997
    60/052,875 16-Jul-1997
    60/052,873 16-Jul-1997
    60/052,874 16-Jul-1997
    60/055,724 18-Aug-1997
    60/055,726 18-Aug-1997
    60/056,361 18-Aug-1997
    60/055,946 18-Aug-1997
    60/053,441 22-Jul-1997
    60/053,440 22-Jul-1997
    60/055,683 18-Aug-1997
    60/055,989 18-Aug-1997
    60/053,442 22-Jul-1997
    60/054,209 30-Jul-1997
    60/055,968 18-Aug-1997
    60/055,972 18-Aug-1997
    60/055,969 18-Aug-1997
    60/054,212 30-Jul-1997
    60/054,234 30-Jul-1997
    60/055,986 18-Aug-1997
    60/055,386 05-Aug-1997
    60/055,970 18-Aug-1997
    60/054,807 05-Aug-1997
    60/056,561 19-Aug-1997
    60/054,214 30-Jul-1997
    60/054,218 30-Jul-1997
    60/054,236 30-Jul-1997
    60/054,215 30-Jul-1997
    60/056,543 19-Aug-1997
    60/056,729 19-Aug-1997
    60/056,534 19-Aug-1997
    60/054,217 30-Jul-1997
    60/054,213 30-Jul-1997
    60/056,730 19-Aug-1997
    60/056,727 19-Aug-1997
    60/054,211 30-Jul-1997
    60/056,554 19-Aug-1997
    60/055,309 05-Aug-1997
    60/056,563 19-Aug-1997
    60/054,798 05-Aug-1997
    60/056,557 19-Aug-1997
    60/055,312 05-Aug-1997
    60/056,371 19-Aug-1997
    60/056,732 19-Aug-1997
    60/055,310 05-Aug-1997
    60/056,556 19-Aug-1997
    60/056,535 19-Aug-1997
    60/056,369 19-Aug-1997
    60/056,555 19-Aug-1997
    60/056,370 19-Aug-1997
    60/054,804 05-Aug-1997
    60/056,731 19-Aug-1997
    60/056,366 19-Aug-1997
    60/054,809 05-Aug-1997
    60/054,806 05-Aug-1997
    60/054,803 05-Aug-1997
    60/056,364 19-Aug-1997
    60/055,311 05-Aug-1997
    60/056,367 19-Aug-1997
    60/056,365 19-Aug-1997
    60/054,808 05-Aug-1997
    60/056,368 19-Aug-1997
    60/056,726 19-Aug-1997
    60/056,728 19-Aug-1997
    60/056,628 19-Aug-1997
    60/056,629 19-Aug-1997
    60/056,271 29-Aug-1997
    60/056,247 29-Aug-1997
    60/056,073 29-Aug-1997
    60/056,270 29-Aug-1997
    60/057,626 05-Sep-1997
    60/057,663 05-Sep-1997
    60/057,669 05-Sep-1997
    60/058,974 12-Sep-1997
    60/058,973 12-Sep-1997
    60/058,667 12-Sep-1997
    60/058,666 12-Sep-1997
    60/060,837 02-Oct-1997
    60/060,862 02-Oct-1997
    60/060,839 02-Oct-1997
    60/060,866 02-Oct-1997
    60/060,843 02-Oct-1997
    60/060,836 02-Oct-1997
    60/060,874 02-Oct-1997
    60/060,838 02-Oct-1997
    60/060,884 02-Oct-1997
    60/060,833 02-Oct-1997
    60/060,880 02-Oct-1997
    60/061,529 09-Oct-1997
    60/071,498 09-Oct-1997
    60/061,463 09-Oct-1997
    60/061,536 09-Oct-1997
    60/061,527 09-Oct-1997
    60/061,532 09-Oct-1997
    60/063,100 24-Oct-1997
    60/062,784 24-Oct-1997
    60/063,148 24-Oct-1997
    60/063,387 24-Oct-1997
    60/063,386 24-Oct-1997
    60/063,089 24-Oct-1997
    60/063,091 24-Oct-1997
    60/063,090 24-Oct-1997
    60/063,092 24-Oct-1997
    60/063,111 24-Oct-1997
    60/063,109 24-Oct-1997
    60/063,110 24-Oct-1997
    60/063,101 24-Oct-1997
    60/063,098 24-Oct-1997
    60/063,097 24-Oct-1997
    60/074,037 09-Feb-1998
    60/074,157 09-Feb-1998
    60/074,118 09-Feb-1998
    60/076,053 26-Feb-1998
    60/076,057 26-Feb-1998
    60/076,054 26-Feb-1998
    60/076,052 26-Feb-1998
    60/076,051 26-Feb-1998
    60/113,006 18-Dec-1998
    60/112,809 17-Dec-1998
    60/116,330 19-Jan-1999
    60/119,468 10-Feb-1999
    60/125,055 18-Mar-1999
    60/128,693 09-Apr-1999
    60/130,991 26-Apr-1999
    60/137,725 07-Jun-1999
    60/145,220 23-Jul-1999
    60/149,182 17-Aug-1999
    60/152,317 03-Sep-1999
    60/152,315 03-Sep-1999
    60/155,709 24-Sep-1999
    60/163,085 02-Nov-1999
    60/172,411 17-Dec-1999
    60/134,068 13-May-1999
    US00/12788 11-May-2000
    60/167,061 23-Nov-1999
    60/124,146 12-Mar-1999
    US00/06043 09-Mar-2000
    60/124,093 12-Mar-1999
    US00/06012 09-Mar-2000
    60/166,989 23-Nov-1999
    US00/06058 09-Mar-2000
    60/124,145 12-Mar-1999
    60/168,654 03-Dec-1999
    60/124,099 12-Mar-1999
    US00/06044 09-Mar-2000
    60/168,661 03-Dec-1999
    60/124,096 12-Mar-1999
    60/168,622 03-Dec-1999
    US00/06059 09-Mar-2000
    US00/06042 09-Mar-2000
    60/124,143 12-Mar-1999
    60/168,663 03-Dec-1999
    60/124,095 12-Mar-1999
    US00/06014 09-Mar-2000
    60/138,598 11-Jun-1999
    60/168,665 03-Dec-1999
    US00/06013 09-Mar-2000
    60/138,626 11-Jun-1999
    60/168,662 03-Dec-1999
    60/125,360 19-Mar-1999
    US00/06049 09-Mar-2000
    60/168,667 03-Dec-1999
    60/138,574 11-Jun-1999
    60/124,144 12-Mar-1999
    60/124,142 12-Mar-1999
    60/138,597 11-Jun-1999
    60/168,666 03-Dec-1999
    US00/06057 09-Mar-2000
    60/125,359 19-Mar-1999
    60/168,664 03-Dec-1999
    US00/06824 16-Mar-2000
    60/126,051 23-Mar-1999
    60/169,906 10-Dec-1999
    US00/06765 16-Mar-2000
    60/125,362 19-Mar-1999
    US00/06792 16-Mar-2000
    60/169,980 10-Dec-1999
    US00/06830 16-Mar-2000
    60/125,361 19-Mar-1999
    60/169,910 10-Dec-1999
    60/125,812 23-Mar-1999
    60/169,936 10-Dec-1999
    US00/06782 16-Mar-2000
    60/126,054 23-Mar-1999
    US00/06822 16-Mar-2000
    60/169,916 10-Dec-1999
    US00/06791 16-Mar-2000
    60/169,946 10-Dec-1999
    60/125,815 23-Mar-1999
    60/125,358 19-Mar-1999
    US00/06828 16-Mar-2000
    60/169,616 08-Dec-1999
    60/125,364 19-Mar-1999
    60/169,623 08-Dec-1999
    US00/06823 16-Mar-2000
    60/169,617 08-Dec-1999
    US00/06781 16-Mar-2000
    60/125,363 19-Mar-1999
    60/172,410 17-Dec-1999
    60/126,502 26-Mar-1999
    US00/07505 22-Mar-2000
    60/126,503 26-Mar-1999
    60/172,409 17-Dec-1999
    US00/07440 22-Mar-2000
    60/126,505 26-Mar-1999
    60/172,412 17-Dec-1999
    US00/07506 22-Mar-2000
    60/126,594 26-Mar-1999
    60/172,408 17-Dec-1999
    US00/07507 22-Mar-2000
    60/172,413 17-Dec-1999
    US00/07535 22-Mar-2000
    60/126,511 26-Mar-1999
    60/126,595 26-Mar-1999
    60/171,549 22-Dec-1999
    US00/07525 22-Mar-2000
    60/171,504 22-Dec-1999
    60/126,598 26-Mar-1999
    US00/07534 22-Mar-2000
    US00/07483 22-Mar-2000
    60/171,552 22-Dec-1999
    60/126,596 26-Mar-1999
    60/126,600 26-Mar-1999
    US00/07526 22-Mar-2000
    60/171,550 22-Dec-1999
    60/126,501 26-Mar-1999
    60/171,551 22-Dec-1999
    US00/07527 22-Mar-2000
    60/126,504 26-Mar-1999
    US00/07661 23-Mar-2000
    60/174,847 07-Jan-2000
    60/174,853 07-Jan-2000
    US00/07579 23-Mar-2000
    60/126,509 26-Mar-1999
    60/174,852 07-Jan-2000
    60/242,710 25-Oct-2000
    60/126,506 26-Mar1999
    US00/07723 23-Mar-2000
    60/174,850 07-Jan-2000
    US00/07724 23-Mar-2000
    60/126,510 26-Mar-1999
    60/138,573 11-Jun-1999
    US00/14929 01-Jun-2000
    60/174,851 07-Jan-2000
    60/126,508 26-Mar-1999
    US00/07722 23-Mar-2000
    60/174,871 07-Jan-2000
    US00/07578 23-Mar-2000
    60/174,872 07-Jan-2000
    60/126,507 26-Mar-1999
    60/174,877 07-Jan-2000
    US00/07726 23-Mar-2000
    60/126,597 26-Mar-1999
    60/126,601 26-Mar-1999
    US00/07677 23-Mar-2000
    60/176,064 14-Jan-2000
    60/154,373 17-Sep-1999
    60/176,063 14-Jan-2000
    US00/07725 23-Mar-2000
    60/126,602 26-Mar-1999
    60/176,052 14-Jan-2000
    US00/09070 06-Apr-2000
    60/128,695 09-Apr-1999
    US00/08982 06-Apr-2000
    60/128,696 09-Apr-1999
    60/176,069 14-Jan-2000
    60/176,068 14-Jan-2000
    US00/08983 06-Apr-2000
    60/128,703 09-Apr-1999
    60/176,929 20-Jan-2000
    US00/09067 06-Apr-2000
    60/128,697 09-Apr-1999
    US00/09066 06-Apr-2000
    60/176,926 20-Jan-2000
    60/128,698 09-Apr-1999
    US00/09068 06-Apr-2000
    60/128,699 09-Apr-1999
    60/177,050 20-Jan-2000
    60/177,166 20-Jan-2000
    US00/08981 06-Apr-2000
    60/128,701 09-Apr-1999
    60/128,700 09-Apr-1999
    60/176,930 20-Jan-2000
    US00/08980 06-Apr-2000
    60/128,694 09-Apr-1999
    US00/09071 06-Apr-2000
    60/176,931 20-Jan-2000
    US00/09069 06-Apr-2000
    60/177,049 20-Jan-2000
    60/128,702 09-Apr-1999
    60/138,629 11-Jun-1999
    US00/15136 01-Jun-2000
    60/138,628 11-Jun-1999
    US00/14926 01-Jun-2000
    60/138,631 11-Jun-1999
    US00/14963 01-Jun-2000
    60/138,632 11-Jun-1999
    US00/15135 01-Jun-2000
    US00/14934 01-Jun-2000
    60/138,599 11-Jun-1999
    60/138,572 11-Jun-1999
    US00/14933 01-Jun-2000
    US00/15137 01-Jun-2000
    60/138,625 11-Jun-1999
    60/138,633 11-Jun-1999
    US00/14928 01-Jun-2000
    60/,138,630 11-Jun-1999
    US00/14973 01-Jun-2000
    US00/14964 01-Jun-2000
    60/138,627 11-Jun-1999
    US00/26376 26-Sep-2000
    60/155,808 27-Sep-1999
    US00/26371 26-Sep-2000
    60/155,804 27-Sep-1999
    60/155,807 27-Sep-1999
    US00/26324 26-Sep-2000
    US00/26323 26-Sep-2000
    60/155,805 27-Sep-1999
    US00/26337 26-Sep-2000
    60/155,806 27-Sep-1999
    60/201,194 02-May-2000
    60/212,142 16-Jun-2000
    US00/29363 25-Oct-2000
    60/215,139 30-Jun-2000
    60/162,239 29-Oct-1999
    US00/29360 25-Oct-2000
    60/162,211 29-Oct-1999
    60/215,138 30-Jun-2000
    60/162,240 29-Oct-1999
    US00/29362 25-Oct-2000
    60/215,131 30-Jun-2000
    US00/29365 25-Oct-2000
    60/162,237 29-Oct-1999
    60/219,666 21-Jul-2000
    60/215,134 30-Jun-2000
    US00/29364 25-Oct-2000
    60/162,238 29-Oct-1999
    60/163,580 05-Nov-1999
    60/215,130 30-Jun-2000
    US00/30040 01-Nov-2000
    US00/30037 01-Nov-2000
    60/163,577 05-Nov-1999
    60/215,137 30-Jun-2000
    60/215,133 30-Jun-2000
    60/163,581 05-Nov-1999
    US00/30045 01-Nov-2000
    60/163,576 05-Nov-1999
    US00/30036 01-Nov-2000
    60/221,366 27-Jul-2000
    60/221,367 27-Jul-2000
    60/195,296 07-Apr-2000
    US00/30039 01-Nov-2000
    60/164,344 09-Nov-1999
    60/221,142 27-Jul-2000
    US00/30654 08-Nov-2000
    60/164,835 12-Nov-1999
    60/164,744 12-Nov-1999
    US00/30628 08-Nov-2000
    60/215,140 30-Jun-20O0
    US00/30653 08-Nov-2000
    60/164,735 12-Nov-1999
    60/221,193 27-Jul-2000
    60/222,904 03-Aug-2000
    60/164,825 12-Nov-1999
    US00/30629 08-Nov-2000
    60/224,007 04-Aug-2000
    US00/30679 08-Nov-2000
    60/164,834 12-Nov-1999
    60/164,750 12-Nov-1999
    60/215,128 30-Jun-2000
    US00/30674 08-Nov-2000
    60/166,415 19-Nov-1999
    US00/31162 15-Nov-2000
    60/215,136 30-Jun-2000
    60/166,414 19-Nov-1999
    60/219,665 21-Jul-2000
    US00/31282 15-Nov-2000
    60/215,132 30-Jun-2000
    60/164,731 12-Nov-1999
    US00/30657 08-Nov-2000
    60/256,968 21-Dec-2000
    60/226,280 18-Aug-2000
    US01/01396 17-Jan-2001
    60/226,380 18-Aug-2000
    US01/01387 17-Jan-2001
    60/259,803 05-Jan-2001
    US01/01567 17-Jan-2001
    60/228,084 28-Aug-2000
    US01/01431 17-Jan-2001
    60/231,968 12-Sep-2000
    60/234,211 20-Sep-2000
    US01/00544 09-Jan-2001
    60/226,282 18-Aug-2000
    US01/01435 17-Jan-2001
    60/232,104 12-Sep-2000
    US01/01386 17-Jan-2001
    60/234,210 20-Sep-2000
    US01/01565 17-Jan-2001
    60/259,805 05-Jan-2001
    US01/01394 17-Jan-2001
    60/226,278 18-Aug-2000
    60/259,678 05-Jan-2001
    60/226,279 18-Aug-2000
    US01/01434 17-Jan-2001
    60/226,281 18-Aug-2000
    US01/01397 17-Jan-2001
    60/231,969 12-Sep-2000
    US01/01385 17-Jan-2001
    60/228,086 28-Aug-2000
    60/259,516 04-Jan-2001
    US01/01384 17-Jan-2001
    60/228,083 28-Aug-2000
    60/259,804 05-Jan-2001
    US01/01383 17-Jan-2001
    60/064,988 07-Nov-1997
    60/064,912 07-Nov-1997
    60/064,900 07-Nov-1997
    60/064,983 07-Nov-1997
    60/064,911 07-Nov-1997
    60/064,985 07-Nov-1997
    60/064,908 07-Nov-1997
    60/064,984 07-Nov-1997
    60/064,987 07-Nov-1997
    60/066,100 17-Nov-1997
    60/066,095 17-Nov-1997
    60/066,089 17-Nov-1997
    60/066,090 17-Nov-1997
    60/066,094 17-Nov-1997
    60/068,007 18-Dec-1997
    60/068,006 18-Dec-1997
    60/068,008 18-Dec-1997
    60/068,057 18-Dec-1997
    60/070,923 18-Dec-1997
    60/068,053 18-Dec-1997
    60/068,054 18-Dec-1997
    60/068,064 18-Dec-1997
    60/068,369 19-Dec-1997
    60/068,367 19-Dec-1997
    60/068,169 19-Dec-1997
    60/068,365 19-Dec-1997
    60/070,704 07-Jan-1998
    60/070,657 07-Jan-1998
    60/070,658 07-Jan-1998
    60/070,692 07-Jan-1998
    60/073,160 30-Jan-1998
    60/073,159 30-Jan-1998
    60/073,165 30-Jan-1998
    60/073,164 30-Jan-1998
    60/073,167 30-Jan-1998
    60/073,162 30-Jan-1998
    60/073,161 30-Jan-1998
    60/073,170 30-Jan-1998
    60/074,341 09-Feb-1998
    60/074,141 09-Feb-1998
    60/077,714 12-Mar-1998
    60/077,687 12-Mar-1998
    60/077,696 12-Mar-1998
    60/077,686 12-Mar-1998
    60/078,574 19-Mar-1998
    60/078,566 19-Mar-1998
    60/078,576 19-Mar-1998
    60/078,577 19-Mar-1998
    60/078,581 19-Mar-1998
    60/078,573 19-Mar-1998
    60/078,578 19-Mar-1998
    60/078,563 19-Mar-1998
    60/080,313 01-Apr-1998
    60/080,312 01-Apr-1998
    60/080,314 01-Apr-1998
    60/085,105 12-May-1998
    60/085,180 12-May-1998
    60/085,093 12-May-1998
    60/085,094 12-May-1998
    60/085,928 18-May-1998
    60/085,920 18-May-1998
    60/085,906 18-May-1998
    60/085,927 18-May-1998
    60/085,924 18-May-1998
    60/085,925 18-May-1998
    60/085,921 18-May-1998
    60/085,922 18-May-1998
    60/085,923 18-May-1998
    60/090,112 22-Jun-1998
    60/089,507 16-Jun-1998
    60/089,508 16-Jun-1998
    60/089,510 16-Jun-1998
    60/089,509 16-Jun-1998
    60/090,113 22-Jun-1998
    60/092,956 15-Jul-1998
    60/092,921 15-Jul-1998
    60/092,922 15-Jul-1998
    60/094,657 30-Jul-1998
    60/095,486 05-Aug-1998
    60/096,319 12-Aug-1998
    60/095,455 06-Aug-1998
    60/095,454 06-Aug-1998
    60/097,917 25-Aug-1998
    60/098,634 31-Aug-1998
    60/101,546 23-Sep-1998
    60/102,895 02-Oct-1998
    60/108,207 12-Nov-1998
    60/236,326 29-Sep-2000
    US01/01432 17-Jan-2001
    US98/04482 06-Mar-1998
    09/148,545 04-Sep-1998
    09/149,476 08-Sep-1998
    US98/04493 06-Mar-1998
    60/190,068 17-Mar-2000
    US98/04858 12-Mar-1998
    09/152,060 11-Sep-1998
    60/265,583 02-Feb-2001
    09/154,707 17-Sep-1998
    US98/05311 19-Mar-1998
    US98/06801 07-Apr-1998
    09/166,780 06-Oct-1998
    09/189,144 10-Nov-1998
    US98/10868 28-May-1998
    60/184,836 24-Feb-2000
    60/193,170 29-Mar-2000
    US98/11422 04-Jun-1998
    09/205,258 04-Dec-1998
    US98/12125 11-Jun-1998
    09/209,462 11-Dec-1998
    09/213,365 17-Dec-1998
    US98/13608 30-Jun-1998
    60/239,899 13-Oct-2000
    US98/13684 07-Jul-1998
    60/180,909 08-Feb-2000
    US98/14613 15-Jul-1998
    09/229,982 14-Jan-1999
    US98/15949 29-Jul-1998
    09/236,557 26-Jan-1999
    09/666,984 21-Sep-2000
    2,298,852 29-Jul-1998
    60/238,291 06-Oct-2000
    US98/16235 04-Aug-1998
    09/244,112 04-Feb-1999
    09/251,329 17-Feb-1999
    US98/17044 18-Aug-1998
    09/716,128 17-Nov-2000
    09/729,835 06-Dec-2000
    US98/17709 27-Aug-1998
    09/257,179 25-Feb-1999
    US98/18360 03-Sep-1998
    60/262,066 18-Jan-2001
    09/722,329 28-Nov-2000
    09/262,109 04-Mar-1999
    US98/20775 01-Oct-1998
    09/281,976 31-Mar-1999
    09/288,143 08-Apr-1999
    60/244,591 01-Nov-2000
    US98/21142 08-Oct-1998
    US98/22376 23-Oct-1998
    09/296,622 23-Apr-1999
    60/239,893 13-Oct-2000
    09/305,736 05-May-1999
    US98/23435 04-Nov-1998
    US98/27059 17-Dec-1998
    09/334,595 17-Jun-1999
    09/348,457 07-Jul-1999
    09/739,907 20-Dec-2000
    US99/00108 06-Jan-1999
    60/232,150 12-Sep-2000
    09/363,044 29-Jul-1999
    US99/01621 27-Jan-1999
    US99/02293 04-Feb-1999
    09/369,247 05-Aug-1999
    09/716,129 17-Nov-2000
    US99/03939 24-Feb-1999
    US99/05721 11-Mar-1999
    09/393,022 09-Sep-1999
    09/397,945 17-Sep-1999
    60/231,846 11-Sep-2000
    US99/09847 06-May-1999
    09/437,658 10-Nov-1999
    US99/13418 15-Jun-1999
    60/263,681 24-Jan-2001
    60/263,230 23-Jan-2001
    09/461,325 14-Dec-1999
    US99/15849 14-Jul-1999
    60/234,925 25-Sep-2001
    US01/00911 12-Jan-2001
    09/482,273 13-Jan-2000
    09/489,847 24-Jan-2000
    US99/17130 29-Jul-1999
    US99/19330 24-Aug-1999
    09/511,554 23-Feb-2000
    09/531,119 20-Mar-2000
    US99/22012 22-Sep-1999
    US99/26409 09-Nov-1999
    09/565,391 05-May-2000
    US99/29950 16-Dec-1999
    09/591,316 09-Jun-2000
    09/618,150 17-Jul-2000
    US00/00903 18-Jan-2000
    09/628,508 28-Jul-2000
    US00/03062 08-Feb-2000
    US00/06783 16-Mar-2000
    09/661,453 13-Sep-2000
    09/684,524 10-Oct-2000
    US00/08979 06-Apr-2000
    09/726,643 01-Dec-2000
    US00/15187 02-Jun-2000
    09/756,168 09-Jan-2001
    US00/19735 20-Jul-2000
    US00/22325 16-Aug-2000
    US00/24008 31-Aug-2000
    US00/26013 22-Sep-2000
    US00/28664 17-Oct-2000
    60/277340 21-Mar-01
    09/781,417 13-Feb-01
    09/789,561 22-Feb-01
    09/800,729 08-Mar-01
    09/756,168 09-Jan-01
  • Furthermore, the contents and sequence listings of application Ser. No. 09/091,873 filed Jun. 25, 1998; No. 60/229,358 filed on Apr. 12, 2000; No. 60/199,384 filed on Apr. 25, 2000 and No. 60/256,931 filed on Dec. 21, 2000 are hereby incorporation by reference in their entirety. [1293]
  • 0
    SEQUENCE LISTING
    The patent application contains a lengthy “Sequence Listing” section. A copy of the “Sequence Listing” is available in electronic form from the USPTO
    web site (http://seqdata.uspto.gov/sequence.html?DocID=20040010134). An electronic copy of the “Sequence Listing” will also be available from the
    USPTO upon request and payment of the fee set forth in 37 CFR 1.19(b)(3).

Claims (29)

What is claimed:
1. An albumin fusion protein comprising a member selected from the group consisting of:
(a) a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:18;
(b) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:18, wherein said fragment or variant has albumin activity;
(c) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:18, wherein said fragment or variant has albumin activity, and further wherein said albumin activity is the ability to prolong the shelf life of the Therapeutic protein:X compared to the shelf-life of the Therapeutic protein:X in an unfused state;
(d) a Therapeutic protein:X and a fragment or a variant of the amino acid sequence of SEQ ID NO:18, wherein said fragment or variant has albumin activity, and further wherein the fragment or variant comprises the amino acid sequence of amino acids 1-387 of SEQ ID NO:18;
(e) a fragment or variant of a Therapeutic protein:X and albumin comprising the amino acid sequence of SEQ ID NO:18, wherein said fragment or variant has a biological activity of the Therapeutic protein:X;
(f) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the N-terminus of albumin, or the N-terminus of the fragment or variant of albumin;
(g) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the C-terminus of albumin, or the C-terminus of the fragment or variant of albumin;
(h) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), wherein the Therapeutic protein:X, or fragment or variant thereof, is fused to the N-terminus and C-terminus of albumin, or the N-terminus and the C-terminus of the fragment or variant of albumin;
(i) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (e), which comprises a first Therapeutic protein:X, or fragment or variant thereof, and a second Therapeutic protein:X, or fragment or variant thereof, wherein said first Therapeutic protein:X, or fragment or variant thereof, is different from said second Therapeutic protein:X, or fragment or variant thereof,
(j) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (i), wherein the Therapeutic protein:X, or fragment or variant thereof, is separated from the albumin or the fragment or variant of albumin by a linker; and
(k) a Therapeutic protein:X, or fragment or variant thereof, and albumin, or fragment or variant thereof, of (a) to (j), wherein the albumin fusion protein has the following formula:
R1-L-R2; R2-L-R1; or R1-L-R2-L-R1,
and further wherein R1 is Therapeutic protein:X, or fragment or variant thereof, L is a peptide linker, and R2 is albumin comprising the amino acid sequence of SEQ II) NO: 18 or a fragment or variant of albumin.
2. The albumin fusion protein of claim 1, wherein the shelf-life of the albumin fusion protein is greater than the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
3. The albumin fusion protein of claim 1, wherein the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin, or fragment or variant thereof, is greater than the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
4. The albumin fusion protein of claim 1, wherein the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin, or fragment or variant thereof, is greater than the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
5. An albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin, or fragment or variant thereof, comprising the amino acid sequence of SEQ ID NO:18 or fragment or variant thereof.
6. An albumin fusion protein comprising a Therapeutic protein:X, or fragment or variant thereof, inserted into an albumin, or fragment or variant thereof, comprising an amino acid sequence selected from the group consisting of:
(a) amino acids 54 to 61 of SEQ ID NO: 18;
(b) amino acids 76 to 89 of SEQ ID NO: 18;
(c) amino acids 92 to 100 of SEQ ID NO: 18;
(d) amino acids 170 to 176 of SEQ ID NO: 18;
(e) amino acids 247 to 252 of SEQ ID NO: 18;
(f) amino acids 266 to 277 of SEQ ID NO: 18;
(g) amino acids 280 to 288 of SEQ ID NO:18;
(h) amino acids 362 to 368 of SEQ ID NO: 18;
(i) amino acids 439 to 447 of SEQ ID NO: 18;
(j) amino acids 462 to 475 of SEQ ID NO: 18;
(k) amino acids 478 to 486 of SEQ ID NO: 18; and
(l) amino acids 560 to 566 of SEQ ID NO:18.
7. The albumin fusion protein of claim 5, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, as compared to the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
8. The albumin fusion protein of claim 6, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, as compared to the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
9. The albumin fusion protein of claim 5, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin as compared to the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
10. The albumin fusion protein of claim 6, wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin as compared to the in vitro biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
11. The albumin fusion protein of claim 5 wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin compared to the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
12. The albumin fusion protein of claim 6 wherein said albumin fusion protein comprises a portion of albumin sufficient to prolong the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, fused to albumin compared to the in vivo biological activity of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
13. The albumin fusion protein of any one of claims 1-12, which is non-glycosylated.
14. The albumin fusion protein of any one of claims 1-12, which is expressed in yeast.
15. The albumin fusion protein of claim 14, wherein the yeast is glycosylation deficient.
16. The albumin fusion protein of claim 14 wherein the yeast is glycosylation and protease deficient.
17. The albumin fusion protein of any one of claims 1-12, which is expressed by a mammalian cell.
18. The albumin fusion protein of any one of claims 1-12, wherein the albumin fusion protein is expressed by a mammalian cell in culture.
19. The albumin fusion protein of any one of claims 1-12, wherein the albumin fusion protein further comprises a secretion leader sequence.
20. A composition comprising the albumin fusion protein of any one of claims 1-12 and a pharmaceutically acceptable carrier.
21. A kit comprising the composition of claim 20.
22. A method of treating a disease or disorder in a patient, comprising the step of administering the albumin fusion protein of any one of claims 1-12.
23. The method of claim 22, wherein the disease or disorder comprises indication:Y.
24. A method of treating a patient with a disease or disorder that is modulated by Therapeutic protein:X, or fragment or variant thereof, comprising the step of administering an effective amount of the albumin fusion protein of any one of claims 1-12.
25. The method of claim 24, wherein the disease or disorder is indication:Y.
26. A method of extending the shelf life of Therapeutic protein:X, or fragment or variant thereof, comprising the step of fusing the Therapeutic protein:X, or fragment or variant thereof, to albumin, or fragment or variant thereof, sufficient to extend the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, compared to the shelf-life of the Therapeutic protein:X, or fragment or variant thereof, in an unfused state.
27. A nucleic acid molecule comprising a polynucleotide sequence encoding the albumin fusion protein of any one of claims 1-12.
28. A vector comprising the nucleic acid molecule of claim 27.
29. A host cell comprising the nucleic acid molecule of claim 28.
US09/833,245 1999-03-12 2001-04-12 Albumin fusion proteins Abandoned US20040010134A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US09/833,245 US20040010134A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US10/472,964 US20070032414A1 (en) 1999-03-12 2002-03-26 Human secreted proteins
US10/472,965 US20070026454A1 (en) 1999-03-12 2002-03-26 Human secreted proteins
US10/868,184 US20070048818A1 (en) 1999-03-12 2004-06-16 Human secreted proteins
US11/264,096 US20060084794A1 (en) 2001-04-12 2005-11-02 Albumin fusion proteins
US11/545,766 US20070099833A1 (en) 2001-04-12 2006-10-11 Albumin fusion proteins
US12/570,397 US20100286048A1 (en) 2001-04-12 2009-09-30 Albumin Fusion Proteins

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US22935800P 2000-04-12 2000-04-12
US19938400P 2000-04-25 2000-04-25
US25693100P 2000-12-21 2000-12-21
US09/833,245 US20040010134A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US95008201A Continuation-In-Part 1999-03-12 2001-09-12
US10/868,184 Continuation-In-Part US20070048818A1 (en) 1999-03-12 2004-06-16 Human secreted proteins
US11/264,096 Division US20060084794A1 (en) 2001-04-12 2005-11-02 Albumin fusion proteins

Publications (1)

Publication Number Publication Date
US20040010134A1 true US20040010134A1 (en) 2004-01-15

Family

ID=27394014

Family Applications (23)

Application Number Title Priority Date Filing Date
US09/833,118 Expired - Lifetime US6905688B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/833,041 Expired - Lifetime US6994857B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/832,929 Expired - Lifetime US6926898B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/833,117 Abandoned US20030171267A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/833,245 Abandoned US20040010134A1 (en) 1999-03-12 2001-04-12 Albumin fusion proteins
US09/832,501 Abandoned US20030199043A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US11/078,663 Expired - Lifetime US7507414B2 (en) 2000-04-12 2005-03-14 Albumin fusion proteins
US11/078,914 Expired - Lifetime US7482013B2 (en) 2000-04-12 2005-03-14 Albumin fusion proteins
US11/927,617 Abandoned US20080269128A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,607 Abandoned US20080269126A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,593 Abandoned US20080261877A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,602 Active 2024-07-29 US10080785B2 (en) 2000-04-12 2007-10-29 Albumin fusion proteins
US11/927,583 Abandoned US20120141415A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,600 Expired - Fee Related US9821039B2 (en) 2000-04-12 2007-10-29 Albumin fusion proteins
US11/927,555 Abandoned US20090285816A9 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,610 Abandoned US20080269127A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/929,677 Expired - Fee Related US7785599B2 (en) 2000-04-12 2007-10-30 Albumin fusion proteins
US12/365,878 Abandoned US20110280830A9 (en) 2000-04-12 2009-02-04 Albumin Fusion Proteins
US13/212,879 Abandoned US20120252732A1 (en) 2000-04-12 2011-08-18 Albumin fusion proteins
US13/464,248 Expired - Fee Related US8946156B2 (en) 2000-04-12 2012-05-04 Albumin Fusion Proteins
US13/855,454 Expired - Fee Related US9849162B2 (en) 2000-04-12 2013-04-02 Treatment with factor VII-albumin fusion protein
US13/855,434 Expired - Lifetime US9775888B2 (en) 2000-04-12 2013-04-02 Treatment with factor ix-albumin fusion protein
US15/850,371 Abandoned US20180200346A1 (en) 2000-04-12 2017-12-21 Albumin fusion proteins

Family Applications Before (4)

Application Number Title Priority Date Filing Date
US09/833,118 Expired - Lifetime US6905688B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/833,041 Expired - Lifetime US6994857B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/832,929 Expired - Lifetime US6926898B2 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US09/833,117 Abandoned US20030171267A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins

Family Applications After (18)

Application Number Title Priority Date Filing Date
US09/832,501 Abandoned US20030199043A1 (en) 2000-04-12 2001-04-12 Albumin fusion proteins
US11/078,663 Expired - Lifetime US7507414B2 (en) 2000-04-12 2005-03-14 Albumin fusion proteins
US11/078,914 Expired - Lifetime US7482013B2 (en) 2000-04-12 2005-03-14 Albumin fusion proteins
US11/927,617 Abandoned US20080269128A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,607 Abandoned US20080269126A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,593 Abandoned US20080261877A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,602 Active 2024-07-29 US10080785B2 (en) 2000-04-12 2007-10-29 Albumin fusion proteins
US11/927,583 Abandoned US20120141415A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,600 Expired - Fee Related US9821039B2 (en) 2000-04-12 2007-10-29 Albumin fusion proteins
US11/927,555 Abandoned US20090285816A9 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/927,610 Abandoned US20080269127A1 (en) 2000-04-12 2007-10-29 Albumin Fusion Proteins
US11/929,677 Expired - Fee Related US7785599B2 (en) 2000-04-12 2007-10-30 Albumin fusion proteins
US12/365,878 Abandoned US20110280830A9 (en) 2000-04-12 2009-02-04 Albumin Fusion Proteins
US13/212,879 Abandoned US20120252732A1 (en) 2000-04-12 2011-08-18 Albumin fusion proteins
US13/464,248 Expired - Fee Related US8946156B2 (en) 2000-04-12 2012-05-04 Albumin Fusion Proteins
US13/855,454 Expired - Fee Related US9849162B2 (en) 2000-04-12 2013-04-02 Treatment with factor VII-albumin fusion protein
US13/855,434 Expired - Lifetime US9775888B2 (en) 2000-04-12 2013-04-02 Treatment with factor ix-albumin fusion protein
US15/850,371 Abandoned US20180200346A1 (en) 2000-04-12 2017-12-21 Albumin fusion proteins

Country Status (10)

Country Link
US (23) US6905688B2 (en)
EP (21) EP2236152B1 (en)
JP (9) JP2004506407A (en)
AU (7) AU2001274809A1 (en)
BE (1) BE2016C059I2 (en)
CA (8) CA2405563A1 (en)
DK (2) DK2236152T3 (en)
ES (2) ES2484966T3 (en)
FR (1) FR16C0043I2 (en)
WO (7) WO2001079442A2 (en)

Cited By (55)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20030027264A1 (en) * 1997-09-18 2003-02-06 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030125247A1 (en) * 2000-04-12 2003-07-03 Rosen Craig A. Albumin fusion proteins
US20030143654A1 (en) * 2000-05-12 2003-07-31 Matthias Grell F-box containing protein
US20030211513A1 (en) * 2000-06-08 2003-11-13 Henry Yue Intracellular signaling proteins
US20040082761A1 (en) * 2001-12-18 2004-04-29 Duggan Brendan M. Cell adhesion proteins
US20050054570A1 (en) * 2001-12-21 2005-03-10 Rosen Craig A. Albumin fusion proteins
US20050079546A1 (en) * 2003-05-01 2005-04-14 Dasa Lipovsek Serum albumin scaffold-based proteins and uses thereof
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
US20050123955A1 (en) * 2000-05-23 2005-06-09 Turner C. A.Jr. Novel human thrombospondin-like proteins and polynucleotides encoding the same
US20050186664A1 (en) * 2001-12-21 2005-08-25 Rosen Craig A. Albumin fusion proteins
US20050196407A1 (en) * 2000-12-05 2005-09-08 Young John A.T. Receptor for B. anthracis toxin
WO2005082400A1 (en) * 2004-02-27 2005-09-09 Leangene Ab Therapeutic proteins for treating medical conditions associated with obesity and/or insulin resistance
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050239167A1 (en) * 1992-01-31 2005-10-27 Aventis Behring L.L.C. Fusion polypeptides of human serum albumin and a therapeutically active polypeptide
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20050281772A1 (en) * 2004-06-17 2005-12-22 Bromley Philip J Compositions for mucosal delivery of agents
EP1617875A1 (en) * 2004-04-27 2006-01-25 Seoul National University Industry Foundation Novel use of aim3 acting as a tumor suppressor
WO2006039504A2 (en) * 2004-10-01 2006-04-13 Cell Ionix, Inc. Method and formulation for stem cells’ stimulation
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20060166329A1 (en) * 2001-10-05 2006-07-27 Human Genome Sciences Inc. Albumin fusion proteins
US20070053903A1 (en) * 2005-05-12 2007-03-08 Zeren Gao Methods of using pHHLA2 to co-stimulate T-cells
US20070141061A1 (en) * 2003-09-05 2007-06-21 Hudson Lindsey J Protein involved in carcinoma
US7329729B1 (en) * 2000-06-21 2008-02-12 Amgen Inc. Secreted epithelial colon stromal-1 molecules and uses thereof
US20080226662A1 (en) * 2000-04-28 2008-09-18 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US7507413B2 (en) 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US7550432B2 (en) 1995-12-30 2009-06-23 Novozymes Biopharma Uk Limited Recombinant fusion proteins to growth hormone and serum albumin
US20090176317A1 (en) * 2006-04-20 2009-07-09 Mayo Foundation For Medical Education And Research Soluble B7-H1
US20090215084A1 (en) * 2006-01-05 2009-08-27 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
US20090240666A1 (en) * 2008-03-19 2009-09-24 Sony Ericsson Mobile Communications Japan, Inc. Mobile terminal device and computer program
US20100015642A1 (en) * 2006-01-05 2010-01-21 Kwon Eugene D B7-h1 and survivin in cancer
US20110020325A1 (en) * 2008-02-29 2011-01-27 Kwon Eugene D Methods for reducing granulomatous inflammation
US20110195068A1 (en) * 2008-08-25 2011-08-11 Solomon Langermann Pd-1 antagonists and methods of use thereof
US20110200620A1 (en) * 2004-10-06 2011-08-18 Lieping Chen B7-h1 and methods of diagnosis, prognosis, and treatment of cancer
EP2359842A1 (en) 2004-07-14 2011-08-24 University of Utah Research Foundation Netrin-related compositions and uses
US8039589B1 (en) 2002-10-04 2011-10-18 Mayo Foundation For Medical Education And Research B7-DC variants
US8153595B2 (en) 2007-07-13 2012-04-10 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
US20120322075A1 (en) * 2009-10-26 2012-12-20 Externautics S.P.A. Lung Tumor Markers and Methods of Use Thereof
US20120329986A1 (en) * 2009-11-17 2012-12-27 Universite De Montreal Heteropeptides useful for reducing nonspecific adsorption
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
US8709416B2 (en) 2008-08-25 2014-04-29 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
US20150051378A1 (en) * 2002-04-18 2015-02-19 The General Hospital Corporation Drg11-responsive (dragon) gene family
US9212228B2 (en) 2005-11-24 2015-12-15 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US9512232B2 (en) 2012-05-09 2016-12-06 Ganymed Pharmaceuticals Ag Antibodies against Claudin 18.2 useful in cancer diagnosis
US9775785B2 (en) 2004-05-18 2017-10-03 Ganymed Pharmaceuticals Ag Antibody to genetic products differentially expressed in tumors and the use thereof
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US10414824B2 (en) 2002-11-22 2019-09-17 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US10656156B2 (en) 2012-07-05 2020-05-19 Mepur Ravindranath Diagnostic and therapeutic potential of HLA-E monospecific monoclonal IgG antibodies directed against tumor cell surface and soluble HLA-E
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation
US10800847B2 (en) 2012-01-11 2020-10-13 Dr. Mepur Ravindranath Anti-HLA class-IB antibodies mimic immunoreactivity and immunomodulatory functions of intravenous immunoglobulin (IVIG) useful as therapeutic IVIG mimetics and methods of their use
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1

Families Citing this family (583)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6057287A (en) 1994-01-11 2000-05-02 Dyax Corp. Kallikrein-binding "Kunitz domain" proteins and analogues thereof
WO2001034769A2 (en) * 1999-11-05 2001-05-17 Human Genome Sciences, Inc. 24 human secreted proteins
WO2002068638A1 (en) * 2001-02-23 2002-09-06 Human Genome Sciences, Inc. 83 human secreted proteins
WO2002018435A1 (en) * 2000-08-28 2002-03-07 Human Genome Sciences, Inc. 18 human secreted proteins
WO2002016388A1 (en) * 2000-08-18 2002-02-28 Human Genome Sciences, Inc. 21 human secreted proteins
US20020137890A1 (en) 1997-03-31 2002-09-26 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
EP0892047A3 (en) * 1997-07-09 2000-03-08 Hoechst Marion Roussel Deutschland GmbH Human and murine semaphorin L
US20050181482A1 (en) * 2004-02-12 2005-08-18 Meade Harry M. Method for the production of an erythropoietin analog-human IgG fusion proteins in transgenic mammal milk
EP1088084B1 (en) * 1998-06-15 2006-09-13 GTC Biotherapeutics, Inc. Erythropoietin analog-human serum albumin fusion protein
US20040199099A1 (en) * 1998-07-10 2004-10-07 Matson James R Hemofiltration systems, methods and devices used to treat inflammatory mediator related disease
US20030190740A1 (en) 1998-10-13 2003-10-09 The University Of Georgia Research Foundation, Inc Stabilized bioactive peptides and methods of identification, synthesis, and use
ATE298796T1 (en) 1998-10-13 2005-07-15 Univ Georgia Res Found STABILIZED BIOACTIVE PEPTIDES AND METHODS FOR THEIR IDENTIFICATION, SYNTHESIS AND USE
AU771534B2 (en) * 1999-05-05 2004-03-25 Phylogica Limited Isolating biological modulators from biodiverse gene fragment libraries
US6946129B1 (en) 1999-06-08 2005-09-20 Seattle Genetics, Inc. Recombinant anti-CD40 antibody and uses thereof
EP1224285A4 (en) * 1999-10-29 2004-12-08 Human Genome Sciences Inc 27 human secreted proteins
AU1449001A (en) * 1999-11-05 2001-06-06 Human Genome Sciences, Inc. 28 human secreted proteins
CA2389724A1 (en) * 1999-11-12 2001-05-17 Human Genome Sciences, Inc. 28 human secreted proteins
ES2270893T3 (en) 1999-12-24 2007-04-16 Genentech, Inc. PROCEDURE AND COMPOSITIONS TO EXTEND THE AVERAGE LIFE OF BIOACTIVE COMPOUNDS.
US7534417B2 (en) 2000-02-24 2009-05-19 Agensys, Inc. 103P2D6: tissue specific protein highly expressed in various cancers
US7291122B2 (en) * 2000-03-24 2007-11-06 Immunocept, L.L.C. Hemofiltration methods for treatment of diseases in a mammal
US6787040B2 (en) * 2000-05-16 2004-09-07 Immunocept, L.L.C. Method and system for colloid exchange therapy
JP2004500863A (en) 2000-06-06 2004-01-15 ブリストル−マイヤーズ スクイブ カンパニー B7-related nucleic acids and polypeptides useful for immunomodulation
EP1311669A4 (en) * 2000-08-18 2004-07-14 Human Genome Sciences Inc 11 human secreted proteins
AU2001232810A1 (en) * 2000-08-18 2002-03-04 Human Genome Sciences, Inc. 23 human secreted proteins
AU2001229563A1 (en) * 2000-09-20 2002-04-02 Human Genome Sciences, Inc. 21 human secreted proteins
AU2001227920A1 (en) * 2000-09-29 2002-04-15 Human Genome Sciences, Inc. 24 human secreted proteins
AU2001296235A1 (en) * 2000-10-12 2002-04-22 Hyseq, Inc. Novel nucleic acids and polypeptides
US6531297B2 (en) * 2000-10-20 2003-03-11 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
US6949371B2 (en) 2000-10-20 2005-09-27 Applera Corporation Isolated human drug-metabolizing proteins, nucleic acid molecules encoding human drug-metabolizing proteins, and uses thereof
FR2815964A1 (en) * 2000-10-30 2002-05-03 Inst Nat Sante Rech Med RENIN AND / OR PRORENIN RECEPTOR PROTEIN, NUCLEIC ACID ENCODING THE RECEPTOR AND THEIR APPLICATIONS
SK288088B6 (en) 2000-12-07 2013-06-03 Eli Lilly And Company GLP-1 fusion proteins
US20030148920A1 (en) * 2000-12-27 2003-08-07 Steven Rosen Sulfatases and methods of use thereof
WO2002053737A1 (en) * 2000-12-28 2002-07-11 Asahi Kasei Kabushiki Kaisha Nf-kb activating gene
CA2441006A1 (en) * 2001-03-08 2002-09-19 Hyseq, Inc. Methods and materials relating to fibulin-like polypeptides and polynucleotides
WO2002083921A2 (en) 2001-04-10 2002-10-24 Agensys, Inc. Nuleic acids and corresponding proteins useful in the detection and treatment of various cancers
US20030125521A1 (en) 2001-06-20 2003-07-03 Genentech, Inc. Compositions and methods for the diagnosis and treatment of disorders involving angiogenesis
AU2002328853A1 (en) * 2001-07-06 2003-01-21 Geneprot, Inc. Carcinoma-related peptides
US8129504B2 (en) 2001-08-30 2012-03-06 Biorexis Technology, Inc. Oral delivery of modified transferrin fusion proteins
US7176278B2 (en) 2001-08-30 2007-02-13 Biorexis Technology, Inc. Modified transferrin fusion proteins
DE10251673A1 (en) 2001-11-09 2003-07-10 Hoffmann La Roche Alström syndrome gene, gene variants, encoded protein and method for diagnosing Alström syndrome
KR100406760B1 (en) * 2001-11-16 2003-11-21 신코엠 주식회사 Semiconductor memory device
WO2003051921A1 (en) 2001-12-17 2003-06-26 Applied Research Systems Ars Holding N.V. Chemokine mutants acting as chemokine antagonists
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
WO2005077042A2 (en) * 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
US20080194481A1 (en) * 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
US20060253913A1 (en) * 2001-12-21 2006-11-09 Yue-Jin Huang Production of hSA-linked butyrylcholinesterases in transgenic mammals
US7081446B2 (en) * 2002-01-31 2006-07-25 The Trustees Of Columbia University In The City Of New York Long-acting follicle stimulating hormone analogues and uses thereof
US20060241027A1 (en) * 2002-02-07 2006-10-26 Hans-Peter Hauser Hiv inhibiting proteins
US20050222023A1 (en) * 2002-02-07 2005-10-06 Hans-Peter Hauser Albumin-fused kunitz domain peptides
AU2003244515A1 (en) * 2002-02-07 2003-09-02 Novozymes Delta Limited Albumin-fused anti-angiogenesis peptides
DE10205520A1 (en) * 2002-02-08 2003-08-14 Aventis Behring Gmbh Inhibitory, monoclonal antibody against the clotting factor VII activating protease
AU2003216345A1 (en) * 2002-02-21 2003-09-09 Wyeth Follistatin domain containing proteins
CA2477614A1 (en) * 2002-03-05 2003-09-18 Merck & Co., Inc. Biomarker for efficacy of appetite suppressant drugs
US20030191056A1 (en) * 2002-04-04 2003-10-09 Kenneth Walker Use of transthyretin peptide/protein fusions to increase the serum half-life of pharmacologically active peptides/proteins
US20070015230A1 (en) * 2002-04-15 2007-01-18 Hammond David J Identification and characterization of analytes from whole blood
EP1494695A4 (en) * 2002-04-15 2006-01-25 American Nat Red Cross Plasma protein-binding ligands
US7141381B2 (en) 2002-04-25 2006-11-28 Bristol-Myers Squibb Company Human leucine-rich repeat-containing proteins specifically expressed in the nervous system
GB0210464D0 (en) 2002-05-08 2002-06-12 Svanborg Catharina Therapeutic treatment
US7153829B2 (en) 2002-06-07 2006-12-26 Dyax Corp. Kallikrein-inhibitor therapies
DK2311432T3 (en) 2002-06-07 2015-02-02 Dyax Corp Modified Kunitz domain polypeptides and their use in reducing ischemia or the onset of a systemic inflammatory response associated with a surgical procedure
ES2381265T3 (en) * 2002-06-07 2012-05-24 Zymogenetics, Inc. Use of IL-21 and monoclonal antibody to treat solid cancers
US7425618B2 (en) 2002-06-14 2008-09-16 Medimmune, Inc. Stabilized anti-respiratory syncytial virus (RSV) antibody formulations
US7132100B2 (en) 2002-06-14 2006-11-07 Medimmune, Inc. Stabilized liquid anti-RSV antibody formulations
US8029803B2 (en) 2002-06-20 2011-10-04 Paladin Labs, Inc. Chimeric antigens for eliciting an immune response
CN101172091B (en) * 2007-09-25 2011-04-27 北京美福源生物医药科技有限公司 Technique for preparing amalgamation protein skin-protection product containing albuminar and skin cell growth factor, and uses of the same
CN1241946C (en) 2002-07-01 2006-02-15 美国福源集团 Human serum albumins recombined merge protein having hyperplasia stimulation function to multiple cells
AU2003245089A1 (en) * 2002-07-03 2004-01-23 Nexgen Biotechnologies, Inc. Fusion polypeptide comprising epidermal growth factor and human serum albumin
EP2332956A1 (en) * 2002-07-08 2011-06-15 Genentech, Inc. Antibody binding to PRO71238
JPWO2004009626A1 (en) * 2002-07-22 2005-11-17 アステラス製薬株式会社 Rheumatoid arthritis-related novel gene
GB0217033D0 (en) 2002-07-23 2002-08-28 Delta Biotechnology Ltd Gene and polypeptide sequences
CA2496002A1 (en) * 2002-08-07 2004-02-19 Delta Biotechnology Ltd. Albumin-fused ciliary neurotrophic factor
KR20050083627A (en) * 2002-08-13 2005-08-26 아비오스 테크놀러지스 인코포레이티드 Selective plasma exchange therapy
EP1542710B1 (en) 2002-08-28 2012-09-26 Dyax Corp. Methods for preserving organs and tissues
US20050271660A1 (en) * 2002-09-06 2005-12-08 Alexion Pharmaceuticals, Inc. Nebulization of monoclonal antibodies for treating pulmonary diseases
US9415102B2 (en) 2002-09-06 2016-08-16 Alexion Pharmaceuticals, Inc. High concentration formulations of anti-C5 antibodies
AU2003270330B2 (en) * 2002-09-06 2009-07-30 Alexion Pharmaceuticals, Inc. Method of treatment of asthma using antibodies to complement component C5
NZ539218A (en) * 2002-09-18 2008-03-28 Univ Montreal Ct Hospitalier Chum Synthetic GHRH analogues of 29 amino acids or more
EP1572739B1 (en) * 2002-12-20 2008-07-16 Geneos OY Asthma susceptibility locus
WO2004058817A1 (en) * 2002-12-26 2004-07-15 Takeda Pharmaceutical Company Limited Novel proteins and use thereof
CA2513213C (en) * 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
CN101444621B (en) * 2003-02-11 2013-11-20 夏尔人类遗传性治疗公司 Diagnosis and treatment of multiple sulfatase deficiency and other using a formylglycine generating enzyme (FGE)
US7381410B2 (en) * 2003-03-12 2008-06-03 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
US7862816B2 (en) * 2003-03-12 2011-01-04 Vasgene Therapeutics, Inc. Polypeptide compounds for inhibiting angiogenesis and tumor growth
WO2004082640A2 (en) * 2003-03-19 2004-09-30 New Century Pharmaceuticals, Inc. Human serum albumin conjugates with therapeutic compounds
EP1613661B1 (en) * 2003-04-04 2011-06-29 Université de Lausanne Peptabody for cancer treatment
KR20110094361A (en) 2003-04-11 2011-08-23 메디뮨 엘엘씨 Recombinant il-9 antibodies and uses thereof
WO2004094589A2 (en) * 2003-04-18 2004-11-04 Incyte Corporation Secreted proteins
US6987270B2 (en) 2003-05-07 2006-01-17 General Electric Company Method to account for event losses due to positron range in positron emission tomography and assay of positron-emitting isotopes
WO2005001093A1 (en) * 2003-06-30 2005-01-06 Nihon University Protein capable of deposition onto extracellular matrix
JP2008500005A (en) 2003-07-15 2008-01-10 バロス リサーチ インスティテュート Compositions and methods for immunotherapy of cancer and infectious diseases
US8007805B2 (en) * 2003-08-08 2011-08-30 Paladin Labs, Inc. Chimeric antigens for breaking host tolerance to foreign antigens
SI1682178T1 (en) 2003-11-04 2010-11-30 Novartis Vaccines & Diagnostic Methods of therapy for cancers expressing the cd40 antigen
WO2005054286A2 (en) * 2003-12-03 2005-06-16 Delta Biotechnology Limited Interleukin-11 fusion proteins
CN100379762C (en) * 2003-12-08 2008-04-09 中国人民解放军军事医学科学院生物工程研究所 Interfusion protein between human serum albumin and interleukin, and encoding genes
US9050378B2 (en) 2003-12-10 2015-06-09 Board Of Regents, The University Of Texas System N2S2 chelate-targeting ligand conjugates
US7371381B2 (en) 2003-12-12 2008-05-13 Amgen Inc. Anti-galanin antibodies and uses thereof
GB0329681D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Gene expression technique
GB0329722D0 (en) 2003-12-23 2004-01-28 Delta Biotechnology Ltd Modified plasmid and use thereof
WO2005061704A1 (en) * 2003-12-24 2005-07-07 Takeda Pharmaceutical Company Limited Preventive/remedy for cancer
EP1721008B1 (en) * 2004-03-03 2010-02-24 EVOTEC Neurosciences GmbH Diagnostic and therapeutic use of mal2 gene and protein for neurodegenerative diseases
JP2007528393A (en) * 2004-03-11 2007-10-11 カイセラ バイオファーマシューティカルズ, インコーポレイテッド Compositions and methods for preventing and treating skin conditions and hair conditions
US20050201959A1 (en) * 2004-03-11 2005-09-15 Vvii Newco 2003, Inc. Methods and compositions for altering skin coloration
CN102746401A (en) 2004-03-12 2012-10-24 瓦斯基因治疗公司 Polypeptide compound for inhibiting angiogenesis and tumor growth
US9592277B2 (en) * 2004-04-14 2017-03-14 Avirid, Inc. Compositions with modified nucleases targeted to viral nucleic acids and methods of use for prevention and treatment of viral diseases
EP1745078B1 (en) * 2004-04-23 2009-06-17 ConjuChem Biotechnologies Inc. Method for the purification of albumin conjugates
JP4948399B2 (en) * 2004-06-03 2012-06-06 ヴァーミリオン インコーポレイテッド Biomarkers for peripheral arterial disease
EP1784511A4 (en) * 2004-08-13 2009-03-11 Millennium Pharm Inc Genes, compositions, kits, and methods for identification, assessment, prevention, and therapy of prostate cancer
EP1793850A4 (en) 2004-09-21 2010-06-30 Medimmune Inc Antibodies against and methods for producing vaccines for respiratory syncytial virus
WO2006034455A2 (en) 2004-09-23 2006-03-30 Vasgene Therapeutics, Inc. Polipeptide compounds for inhibiting angiogenesis and tumor growth
US7235530B2 (en) * 2004-09-27 2007-06-26 Dyax Corporation Kallikrein inhibitors and anti-thrombolytic agents and uses thereof
WO2006042197A2 (en) * 2004-10-11 2006-04-20 The Board Of Trustees Of The Leland Standford Junior University Use of del-1 in hair, bone and cartilage regeneration
AU2005298245B2 (en) * 2004-10-25 2011-03-10 Immune System Key Ltd A thymus-specific protein
KR100583350B1 (en) * 2004-11-03 2006-06-05 (주)넥스젠 -1 Method for Producing Epidermal Growth Factor Using Fusion Proteins Comprising Fas-1 Domain
ES2312037T3 (en) * 2004-11-09 2009-02-16 Ares Trading S.A. METHOD FOR PURUIFYING FSH.
DK2330200T3 (en) 2004-12-23 2017-07-24 Albumedix As GENE EXPRESSION TECHNIQUE
EP2446897A1 (en) 2005-01-06 2012-05-02 Novo Nordisk A/S Anti-KIR combination treatments and methods
US20060178301A1 (en) * 2005-02-04 2006-08-10 Mathias Jurs Albumin-fused ciliary neurotrophic factor
PT1853718E (en) 2005-02-15 2015-11-12 Univ Duke Anti-cd19 antibodies and uses in oncology
US20110230407A1 (en) * 2005-03-14 2011-09-22 Alexander Yuzhakov Hepatocyte growth factor pathway activators in demyelinating diseases and central nervous system trauma
CA2601086A1 (en) * 2005-03-23 2006-10-12 Wyeth Detection of an immune response to gdf-8 modulating agents
US20060240487A1 (en) * 2005-03-23 2006-10-26 Nowak John A Detection of GDF-8 modulating agents
BRPI0609079A2 (en) 2005-04-18 2010-11-16 Novo Nordisk As peptide, pharmaceutical composition, method for the treatment of cancer, use of a peptide nucleic acid construct, vector, host, and, antibody
US7833979B2 (en) * 2005-04-22 2010-11-16 Amgen Inc. Toxin peptide therapeutic agents
WO2006114115A1 (en) 2005-04-26 2006-11-02 Trion Pharma Gmbh Combination of antibodies and glucocorticoids for treating cancer
US9345745B2 (en) 2005-04-29 2016-05-24 Bo Wang Methods for treating inflammatory disorders and traumatic brain injury using stabilized non-hematopoietic EPO short peptides
US9585932B2 (en) 2005-04-29 2017-03-07 Peter C. Dowling Use of EPO-derived peptide fragments for the treatment of neurodegenerative disorders
WO2007052154A2 (en) 2005-04-29 2007-05-10 University Of Medicine And Dentistry Of New Jersey Erythropoietin-derived short peptide and its mimics as immuno/inflammatory modulators
EP1885755A4 (en) 2005-05-05 2009-07-29 Univ Duke Anti-cd19 antibody therapy for autoimmune disease
CN101355965A (en) 2005-06-08 2009-01-28 达纳-法伯癌症研究院 Methods and compositions for the treatment of persistent infections and cancers by inhibiting the programmed cell death 1 (PD-1) phthway
WO2007014167A2 (en) * 2005-07-22 2007-02-01 Five Prime Therapeutics, Inc. Compositions for and methods of treating epithelial diseases with growth factors
US8323666B2 (en) * 2005-08-01 2012-12-04 Allergan, Inc. Botulinum toxin compositions
CA2618681C (en) 2005-08-10 2015-10-27 Macrogenics, Inc. Identification and engineering of antibodies with variant fc regions and methods of using same
US8008453B2 (en) 2005-08-12 2011-08-30 Amgen Inc. Modified Fc molecules
PL1917276T3 (en) 2005-08-26 2018-08-31 Ares Trading S.A. Process for the preparation of glycosylated interferon beta
US7713715B2 (en) * 2005-09-06 2010-05-11 University Of Tennessee Research Foundation Method for diagnosing infections
US7846445B2 (en) * 2005-09-27 2010-12-07 Amunix Operating, Inc. Methods for production of unstructured recombinant polymers and uses thereof
US20090099031A1 (en) * 2005-09-27 2009-04-16 Stemmer Willem P Genetic package and uses thereof
EP1929073A4 (en) * 2005-09-27 2010-03-10 Amunix Inc Proteinaceous pharmaceuticals and uses thereof
US7855279B2 (en) * 2005-09-27 2010-12-21 Amunix Operating, Inc. Unstructured recombinant polymers and uses thereof
JP2009509564A (en) * 2005-10-03 2009-03-12 アストラゼネカ・アクチエボラーグ Fusion proteins with regulated plasma half-life
US8168592B2 (en) * 2005-10-21 2012-05-01 Amgen Inc. CGRP peptide antagonists and conjugates
US8604175B2 (en) * 2005-12-09 2013-12-10 Ares Trading S.A. Method for purifying FSH or a FSH mutant
JP2009518441A (en) * 2005-12-09 2009-05-07 シアトル ジェネティクス,インコーポレーテッド How to use a CD40 binder
US8841255B2 (en) 2005-12-20 2014-09-23 Duke University Therapeutic agents comprising fusions of vasoactive intestinal peptide and elastic peptides
US20130172274A1 (en) 2005-12-20 2013-07-04 Duke University Methods and compositions for delivering active agents with enhanced pharmacological properties
EP1976876A4 (en) * 2005-12-22 2010-01-13 Conjuchem Biotechnologies Inc Process for the production of preformed conjugates of albumin and a therapeutic agent
CA2658227A1 (en) 2006-01-17 2007-07-26 Health Research, Inc. Heteroduplex tracking assay
US20070248646A1 (en) * 2006-02-16 2007-10-25 Ali Hafezi-Moghadam Use of azurocidin inhibitors in prevention and treatment of ocular vascular leakage
WO2007103469A2 (en) 2006-03-06 2007-09-13 Aeres Biomedical Ltd. Humanized anti-cd22 antibodies and their use in treatment of oncology, transplantation and autoimmune disease
TWI392684B (en) 2006-04-05 2013-04-11 Abbott Biotech Ltd Antibody purification
DK2007885T3 (en) 2006-04-11 2010-11-08 Csl Behring Gmbh Method for enhancing the in vivo recovery of therapeutic polypeptides
AU2007258609B2 (en) * 2006-06-07 2013-01-24 Human Genome Sciences, Inc. Albumin fusion proteins
JP5800458B2 (en) * 2006-06-14 2015-10-28 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー Fusion protein cleavable by proteolysis with blood clotting factor
AU2013202566C1 (en) * 2006-06-14 2018-07-12 Csl Behring Gmbh Proteolytically cleavable fusion protein comprising a blood coagulation factor
EP1867660A1 (en) 2006-06-14 2007-12-19 CSL Behring GmbH Proteolytically cleavable fusion protein comprising a blood coagulation factor
US7939632B2 (en) * 2006-06-14 2011-05-10 Csl Behring Gmbh Proteolytically cleavable fusion proteins with high molar specific activity
US9139611B2 (en) * 2006-07-13 2015-09-22 Novozymes Biopharma Dk A/S Process for preparing particles of proteinaceous material
DK2046826T3 (en) 2006-07-24 2011-10-24 Biorexis Pharmaceutical Corp Exendin-fusion proteins
CA2659809A1 (en) * 2006-08-04 2008-02-14 Pharmathene Inc. Long half-life recombinant butyrylcholinesterase
CN103230598A (en) * 2006-09-06 2013-08-07 费斯生物制药公司 Fusion peptide therapeutic compositions
CN104193815A (en) 2006-09-08 2014-12-10 Ambrx公司 Modified human plasma polypeptide or Fc scaffolds and their uses
US10925977B2 (en) 2006-10-05 2021-02-23 Ceil>Point, LLC Efficient synthesis of chelators for nuclear imaging and radiotherapy: compositions and applications
US7834164B2 (en) * 2006-10-25 2010-11-16 Amgen Inc. DNA encoding OSK1 toxin peptide analogs and vectors and cells for combinant expression
CA2666426A1 (en) 2006-10-26 2008-05-02 Novo Nordisk A/S Il-21 variants
CA2669424A1 (en) 2006-11-07 2008-05-29 Merck & Co., Inc. Antagonists of pcsk9
ES2535358T3 (en) 2006-12-12 2015-05-08 Biorexis Pharmaceutical Corporation Transferrin fusion protein libraries
WO2008074840A2 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against a metalloproteinase from the adam family and polypeptides comprising the same for the treatment of adam-related diseases and disorders
CA2673331A1 (en) 2006-12-19 2008-06-26 Ablynx N.V. Amino acid sequences directed against gpcrs and polypeptides comprising the same for the treatment of gpcr-related diseases and disorders
EP3231440A1 (en) 2006-12-22 2017-10-18 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
EP1935430A1 (en) * 2006-12-22 2008-06-25 CSL Behring GmbH Modified coagulation factors with prolonged in vivo half-life
EP2423222B2 (en) 2007-01-30 2020-07-29 Epivax, Inc. Regulatory t cell epitopes, compositions and uses thereof
WO2008098720A1 (en) * 2007-02-12 2008-08-21 Csl Behring Gmbh Therapeutic application of kazal-type serine protease inhibitors
CA2680832A1 (en) 2007-03-27 2008-10-02 Merck & Co., Inc. Method for detecting autoprocessed, secreted pcsk9
KR20100017514A (en) 2007-05-07 2010-02-16 메디뮨 엘엘씨 Anti-icos antibodies and their use in treatment of oncology, transplantation and autoimmune disease
PL2068927T3 (en) 2007-05-14 2016-06-30 Medimmune Llc Methods of reducing eosinophil levels
EP2738257A1 (en) 2007-05-22 2014-06-04 Amgen Inc. Compositions and methods for producing bioactive fusion proteins
EP1997830A1 (en) 2007-06-01 2008-12-03 AIMM Therapeutics B.V. RSV specific binding molecules and means for producing them
WO2009012502A1 (en) * 2007-07-19 2009-01-22 The Arizona Board Of Regents, A Body Corporate Of The State Of Arizona Acting For And On Behalf Of Arizona State University Self- anchoring mems intrafascicular neural electrode
KR20100058541A (en) * 2007-08-15 2010-06-03 아뮤닉스 인코포레이티드 Compositions and methods for modifying properties of biologically active polypeptides
EP3354662B1 (en) 2007-08-29 2020-08-19 Sanofi Humanized anti-cxcr5 antibodies, derivatives thereof and their uses
JP2009055838A (en) * 2007-08-31 2009-03-19 Nipro Corp Fusion protein, gene related to the fusion protein, vector, transformant and anti-inflammatory medicinal composition
US9139634B2 (en) * 2007-09-21 2015-09-22 The Regents Of The University Of California Interferon-antibody fusion proteins demonstrating potent apoptotic and anti-tumor activities
EP2050764A1 (en) 2007-10-15 2009-04-22 sanofi-aventis Novel polyvalent bispecific antibody format and uses thereof
WO2009053358A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying fc-fusion proteins
AU2008314689A1 (en) * 2007-10-22 2009-04-30 Merck Serono S.A. Method for purifying an Fc-containing protein
WO2009070642A1 (en) 2007-11-28 2009-06-04 Medimmune, Llc Protein formulation
US8092804B2 (en) 2007-12-21 2012-01-10 Medimmune Limited Binding members for interleukin-4 receptor alpha (IL-4Rα)-173
EP3211010A1 (en) 2007-12-21 2017-08-30 Medimmune Limited Binding members for interleukin-4 receptor alpha (il-4r) - 173
WO2009097318A1 (en) * 2008-01-28 2009-08-06 Bristol-Myers Squibb Company A fluorescence polarization binding assay for characterizing glucokinase ligands
AR070316A1 (en) 2008-02-07 2010-03-31 Merck & Co Inc PCSK9 ANTAGONISTS (SUBTILISINE-KEXINA TYPE 9 PROPROTEIN)
AR070315A1 (en) 2008-02-07 2010-03-31 Merck & Co Inc ANTIBODIES 1B20 ANTAGONISTS OF PCSK9
EP2262539B1 (en) 2008-04-01 2015-07-15 Novo Nordisk A/S Insulin albumin conjugates
CA2720763A1 (en) 2008-04-07 2009-10-15 Ablynx Nv Amino acid sequences directed against the notch pathways and uses thereof
PT2439212T (en) 2008-05-02 2017-03-29 Novartis Ag Improved fibronectin-based binding molecules and uses thereof
DK2291523T3 (en) 2008-06-24 2015-03-23 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
AU2013202564B2 (en) * 2008-06-24 2015-09-17 Csl Behring Gmbh Factor VIII, von Willebrand factor or complexes thereof with prolonged in vivo half-life
JP2011526303A (en) 2008-06-27 2011-10-06 デューク ユニバーシティ A therapeutic agent containing an elastin-like peptide
EP2313093A4 (en) 2008-07-10 2012-03-28 Angion Biomedica Corp Methods and compositions of small molecule modulators of hepatocyte growth factor (scatter factor) activity
CA2731617A1 (en) 2008-07-22 2010-01-28 Maria Joao Saraiva Amino acid sequences directed against multitarget scavenger receptors and polypeptides
ES2541142T3 (en) 2008-08-05 2015-07-16 Novartis Ag Compositions and methods for antibodies directed against complement C5 protein
BRPI0823049A2 (en) 2008-09-07 2015-06-16 Glyconex Inc Anti-extended type 1 glycosphingolipid antibodies, derivatives thereof and use.
BRPI0921845A2 (en) 2008-11-12 2019-09-17 Medimmune Llc stable sterile aqueous formulation, pharmaceutical unit dosage form, pre-filled syringe, and methods for treating a disease or disorder, treating or preventing rejection, depleting unique expressing t cells in a human patient, and disrupting central germinal architecture in a secondary lymphoid organ of a primate
US8927694B2 (en) 2008-11-18 2015-01-06 Merrimack Pharmaceuticals, Inc. Human serum albumin linkers and conjugates thereof
DK2370593T3 (en) 2008-11-28 2016-07-04 Univ Emory A method for determining the effect of PD-1 Antagonists
TWI605828B (en) 2008-12-09 2017-11-21 建南德克公司 Anti-pd-l1 antibodies and their use to enhance t-cell function
WO2010068526A1 (en) 2008-12-12 2010-06-17 Merck Sharp & Dohme Corp. Pcsk9 immunoassay
CN102307594A (en) 2009-01-06 2012-01-04 戴埃克斯有限公司 Treatment of mucositis with kallikrein inhibitors
NZ594055A (en) 2009-01-16 2013-03-28 Teva Pharma New stable formulations of recombinant human albumin-human granulocyte colony stimulating factor fusion protein
US8852608B2 (en) 2009-02-02 2014-10-07 Medimmune, Llc Antibodies against and methods for producing vaccines for respiratory syncytial virus
CN108530543B (en) 2009-02-03 2023-06-23 阿穆尼克斯制药公司 Extended recombinant polypeptides and compositions comprising the same
US8703717B2 (en) 2009-02-03 2014-04-22 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
US8680050B2 (en) * 2009-02-03 2014-03-25 Amunix Operating Inc. Growth hormone polypeptides fused to extended recombinant polypeptides and methods of making and using same
WO2010092135A2 (en) 2009-02-11 2010-08-19 Novozymes Biopharma Uk Ltd. Albumin variants and conjugates
JP2012521197A (en) 2009-03-20 2012-09-13 アムジエン・インコーポレーテツド Carrier immunoglobulin and use thereof
WO2010128143A1 (en) 2009-05-07 2010-11-11 Novozymes Biopharma Dk A/S Method of controlling o-linked glycosylation of antibodies
US11512326B2 (en) * 2009-05-26 2022-11-29 University Of Florida Research Foundation, Incorporated Small angiotensin peptide expression system in mammalian cells
EP2258398A1 (en) * 2009-05-26 2010-12-08 Araclón Biotech, S. L. Albumin-amyloid peptide conjugates and uses thereof
AU2010258898B8 (en) 2009-06-08 2015-02-05 Amunix Operating Inc. Glucose-regulating polypeptides and methods of making and using same
US9849188B2 (en) 2009-06-08 2017-12-26 Amunix Operating Inc. Growth hormone polypeptides and methods of making and using same
CN101628346B (en) * 2009-07-25 2012-05-16 大连理工大学 Method for slowing down outlet speed of carbon fiber composite material hand-made hole and buffer device thereof
EP3381937A3 (en) 2009-08-13 2018-10-31 The Johns Hopkins University Methods of modulating immune function
JP6250282B2 (en) 2009-08-20 2017-12-20 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー Albumin fusion clotting factor for non-intravenous administration in the treatment and prophylactic treatment of bleeding disorders
JP2013502458A (en) 2009-08-24 2013-01-24 アムニクス オペレーティング インコーポレイテッド Coagulation factor VII composition and methods of making and using the same
US10420862B2 (en) 2009-08-24 2019-09-24 Aresenal AAA, LLC. In-situ forming foams for treatment of aneurysms
US20110202016A1 (en) * 2009-08-24 2011-08-18 Arsenal Medical, Inc. Systems and methods relating to polymer foams
US9044580B2 (en) 2009-08-24 2015-06-02 Arsenal Medical, Inc. In-situ forming foams with outer layer
US9173817B2 (en) 2009-08-24 2015-11-03 Arsenal Medical, Inc. In situ forming hemostatic foam implants
US8451450B2 (en) * 2009-09-14 2013-05-28 Bio-Rad Laboratories, Inc. Near real time optical phase conjugation
UY32917A (en) 2009-10-02 2011-04-29 Boehringer Ingelheim Int DLL-4 BINDING MOLECULES
US20110172398A1 (en) 2009-10-02 2011-07-14 Boehringer Ingelheim International Gmbh Bispecific binding molecules for anti-angiogenesis therapy
US8568726B2 (en) 2009-10-06 2013-10-29 Medimmune Limited RSV specific binding molecule
ES2672121T3 (en) 2009-10-07 2018-06-12 Macrogenics, Inc. Polypeptides containing Fc region that have an enhanced effector function due to alterations in the degree of fucosylation, and methods for their use
EP2493507A4 (en) 2009-10-30 2013-11-20 Merck Sharp & Dohme Ax213 and ax132 pcsk9 antagonists and variants
WO2011051327A2 (en) 2009-10-30 2011-05-05 Novartis Ag Small antibody-like single chain proteins
EP2493505A4 (en) 2009-10-30 2013-06-12 Merck Sharp & Dohme Ax1 and ax189 pcsk9 antagonists and variants
WO2011051489A2 (en) 2009-10-30 2011-05-05 Novozymes Biopharma Dk A/S Albumin variants
WO2011051466A1 (en) 2009-11-02 2011-05-05 Novartis Ag Anti-idiotypic fibronectin-based binding molecules and uses thereof
EP2506868B1 (en) 2009-12-06 2017-11-15 Bioverativ Therapeutics Inc. Factor viii-fc chimeric and hybrid polypeptides, and methods of use thereof
AU2010328305B2 (en) 2009-12-08 2015-05-14 Teva Pharmaceutical Industries Ltd. BChE albumin fusions for the treatment of cocaine abuse
EP2513145B1 (en) 2009-12-14 2018-01-24 Ablynx N.V. Single variable domain antibodies against ox40l, constructs and therapeutic use
JP2013526840A (en) 2009-12-21 2013-06-27 ファーマシーネ,インコーポレイテッド Recombinant butyrylcholinesterase and its truncated form
AU2011204349C1 (en) 2010-01-06 2017-02-16 Takeda Pharmaceutical Company Limited Plasma kallikrein binding proteins
AR079944A1 (en) 2010-01-20 2012-02-29 Boehringer Ingelheim Int NEUTRALIZING ANTIBODY OF THE ACTIVITY OF AN ANTICOAGULANT
US9695226B2 (en) 2010-01-22 2017-07-04 Novo Nordisk Healthcare Ag Growth hormones with prolonged in-vivo efficacy
JP5980689B2 (en) 2010-01-22 2016-08-31 ノヴォ・ノルディスク・ヘルス・ケア・アーゲー Stable growth hormone compound
WO2011092233A1 (en) 2010-01-29 2011-08-04 Novartis Ag Yeast mating to produce high-affinity combinations of fibronectin-based binders
US9120855B2 (en) 2010-02-10 2015-09-01 Novartis Ag Biologic compounds directed against death receptor 5
EP2547328B1 (en) * 2010-02-11 2017-06-07 Ecole Polytechnique Federale de Lausanne (EPFL) Ccr7 ligand delivery and co-delivery in immunotherapy
WO2011107507A1 (en) 2010-03-03 2011-09-09 Boehringer Ingelheim International Gmbh Biparatopic abeta binding polypeptides
EP2371857A1 (en) 2010-04-01 2011-10-05 CSL Behring GmbH Factor XII inhibitors for treating interstitial lung disease
CN106977608A (en) 2010-04-09 2017-07-25 阿尔布麦狄克斯公司 Albumin derivant and variant
MA34287B1 (en) 2010-05-06 2013-06-01 Novartis Ag COMPOSITIONS AND METHODS OF USING LOW DENSITY THERAPEUTIC MULTIVALENT ANTIBODIES OF LIPOPROTEIN 6-RELATED PROTEIN (LRP6)
ES2659406T3 (en) 2010-05-06 2018-03-15 Novartis Ag Compositions and methods of use for therapeutic antibodies against protein 6 related to low density lipoproteins (LRP6)
WO2011144749A1 (en) 2010-05-20 2011-11-24 Ablynx Nv Biological materials related to her3
KR20140017480A (en) 2010-07-09 2014-02-11 바이오겐 이데크 헤모필리아 인코포레이티드 Factor ix polypeptides and methods of use thereof
EP3508573A1 (en) 2010-07-09 2019-07-10 Bioverativ Therapeutics Inc. Systems for factor viii processing and methods thereof
WO2012019061A2 (en) 2010-08-05 2012-02-09 Stem Centrx, Inc. Novel effectors and methods of use
CA2807552A1 (en) 2010-08-06 2012-02-09 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US9850296B2 (en) 2010-08-10 2017-12-26 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
US9517257B2 (en) 2010-08-10 2016-12-13 Ecole Polytechnique Federale De Lausanne (Epfl) Erythrocyte-binding therapeutics
CN103282380B (en) 2010-08-10 2018-02-16 洛桑聚合联合学院 Erythrocyte binding therapeutic agent
NZ607337A (en) 2010-08-20 2015-06-26 Novartis Ag Antibodies for epidermal growth factor receptor 3 (her3)
EP2608807A1 (en) 2010-08-27 2013-07-03 Stem Centrx, Inc. Notum protein modulators and methods of use
US20120225081A1 (en) 2010-09-03 2012-09-06 Boehringer Ingelheim International Gmbh Vegf-binding molecules
BR112013005116A2 (en) 2010-09-03 2019-09-24 Stem Centrx Inc modulators and methods of use
SG188591A1 (en) 2010-09-22 2013-04-30 Amgen Inc Carrier immunoglobulins and uses thereof
BR122021020041B1 (en) 2010-09-28 2023-03-07 Amylin Pharmaceuticals, Llc CHIMERIC POLYPEPTIDE, ITS USE AND COMPOSITION INCLUDING IT
EP2857499A1 (en) 2010-10-01 2015-04-08 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
US8892184B2 (en) 2010-10-18 2014-11-18 Siemens Medical Solutions Usa, Inc. Systems and methods for reducing interference in a dual modality imaging system
US8853490B2 (en) * 2010-10-26 2014-10-07 Pioneer Hi Bred International Inc Antifungal proteins and methods of use
US20130225496A1 (en) 2010-11-01 2013-08-29 Novozymes Biopharma Dk A/S Albumin Variants
CN108341868B (en) 2010-11-05 2022-06-07 酵活有限公司 Stable heterodimeric antibody design with mutations in the Fc domain
UA115027C2 (en) 2010-11-08 2017-09-11 Новартіс Аг Cxcr2 binding polypeptides
US9095606B1 (en) 2010-11-13 2015-08-04 Sirbal Ltd. Molecular and herbal combinations for treating psoriasis
US9066974B1 (en) 2010-11-13 2015-06-30 Sirbal Ltd. Molecular and herbal combinations for treating psoriasis
EP2643353A1 (en) 2010-11-24 2013-10-02 Novartis AG Multispecific molecules
AR084208A1 (en) 2010-12-08 2013-05-02 Stem Centrx Inc MODULATORS OF THE EFRINE A BINDING (EFNA) AND METHODS FOR USE
CN103370620B (en) 2010-12-28 2016-01-20 大日本住友制药株式会社 The diagnosis medicine of Alzheimer disease and diagnostic method
US20120171195A1 (en) 2011-01-03 2012-07-05 Ravindranath Mepur H Anti-hla-e antibodies, therapeutic immunomodulatory antibodies to human hla-e heavy chain, useful as ivig mimetics and methods of their use
KR102320178B1 (en) 2011-01-06 2021-11-02 다케다 파머수티컬 컴패니 리미티드 Plasma kallikrein binding proteins
SA112330278B1 (en) 2011-02-18 2015-10-09 ستيم سينتركس، انك. Novel modulators and methods of use
WO2012117336A2 (en) 2011-02-28 2012-09-07 Istituto Di Ricovero E Cura A Carattere Scientifico Materno-Infantile Burlo Garofolo - Ospedale Di Alta Specializzazione E Di Rilievo Nazionale Per La Salute Della Donna E Del Bambino Apoptosis-inducing molecules and uses therefor
US9499605B2 (en) 2011-03-03 2016-11-22 Zymeworks Inc. Multivalent heteromultimer scaffold design and constructs
PL2683397T3 (en) 2011-03-09 2018-01-31 Csl Behring Gmbh Factor xii inhibitors for the administration with medical procedures comprising contact with artificial surfaces
EP2497489A1 (en) 2011-03-09 2012-09-12 CSL Behring GmbH Factor XII inhibitors for the treatment of silent brain ischemia and ischemia of other organs
US9340590B2 (en) 2011-03-16 2016-05-17 Amgen Inc. Potent and selective inhibitors of NaV1.3 and NaV1.7
TWI700093B (en) 2011-03-16 2020-08-01 法商賽諾菲公司 Uses of a dual v region antibody-like protein
WO2012135500A1 (en) 2011-03-29 2012-10-04 The General Hospital Corporation Engineered thioredoxin-like fold proteins
WO2012130834A1 (en) 2011-03-30 2012-10-04 Boehringer Ingelheim International Gmbh Anticoagulant antidotes
EP2691101A2 (en) 2011-03-31 2014-02-05 Moderna Therapeutics, Inc. Delivery and formulation of engineered nucleic acids
US9527925B2 (en) 2011-04-01 2016-12-27 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to VEGF and ANG2
US20130078247A1 (en) 2011-04-01 2013-03-28 Boehringer Ingelheim International Gmbh Bispecific binding molecules binding to dii4 and ang2
EP2515112B1 (en) * 2011-04-22 2015-08-12 Sysmex Corporation Method for electrochemically detecting analyte
WO2012149197A2 (en) 2011-04-27 2012-11-01 Abbott Laboratories Methods for controlling the galactosylation profile of recombinantly-expressed proteins
UA117218C2 (en) 2011-05-05 2018-07-10 Мерк Патент Гмбх Amino acid sequences directed against il-17a, il-17f and/or il17-a/f and polypeptides comprising the same
JP6430828B2 (en) 2011-05-05 2018-11-28 アルブミディクス リミティド Albumin variant
US9244074B2 (en) 2011-06-07 2016-01-26 University Of Hawaii Biomarker of asbestos exposure and mesothelioma
WO2012170742A2 (en) 2011-06-07 2012-12-13 University Of Hawaii Treatment and prevention of cancer with hmgb1 antagonists
AU2012267484B2 (en) 2011-06-10 2017-03-23 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof
WO2012172495A1 (en) 2011-06-14 2012-12-20 Novartis Ag Compositions and methods for antibodies targeting tem8
US20160145589A1 (en) 2011-06-24 2016-05-26 Green Cross Corporation Composition and formulation comprising recombinant human iduronate-2-sulfatase and preparation method thereof
CN110066340A (en) * 2011-06-28 2019-07-30 英伊布里克斯有限合伙公司 Serine protease inhibitor fused polypeptide and its application method
WO2013004607A1 (en) 2011-07-01 2013-01-10 Bayer Intellectual Property Gmbh Relaxin fusion polypeptides and uses thereof
CA2838964C (en) 2011-07-05 2021-07-13 Novozymes Biopharma Dk A/S Albumin formulation and use
KR20140039257A (en) 2011-07-08 2014-04-01 바이엘 인텔렉쳐 프로퍼티 게엠베하 Fusion proteins releasing relaxin and uses thereof
WO2013009539A1 (en) 2011-07-08 2013-01-17 Amylin Pharmaceuticals, Inc. Engineered polypeptides having enhanced duration of action and reduced immunogenicity
CA2842323A1 (en) * 2011-07-18 2013-01-24 Arts Biologics A/S Long acting biologically active luteinizing hormone (lh) compound
JP6253578B2 (en) 2011-07-22 2017-12-27 ツェー・エス・エル・ベーリング・ゲー・エム・ベー・ハー Inhibitory anti-factor XII / XIIA monoclonal antibodies and their use
US20130058947A1 (en) 2011-09-02 2013-03-07 Stem Centrx, Inc Novel Modulators and Methods of Use
US9464124B2 (en) 2011-09-12 2016-10-11 Moderna Therapeutics, Inc. Engineered nucleic acids and methods of use thereof
RU2648950C2 (en) 2011-10-03 2018-04-02 Модерна Терапьютикс, Инк. Modified nucleosides, nucleotides and nucleic acids and their application
US8993831B2 (en) * 2011-11-01 2015-03-31 Arsenal Medical, Inc. Foam and delivery system for treatment of postpartum hemorrhage
AU2012332590B2 (en) 2011-11-01 2016-10-20 Bionomics, Inc. Anti-GPR49 antibodies
AU2012332593B2 (en) 2011-11-01 2016-11-17 Bionomics, Inc. Anti-GPR49 antibodies
EP2773373B1 (en) 2011-11-01 2018-08-22 Bionomics, Inc. Methods of blocking cancer stem cell growth
US9221906B2 (en) 2011-11-01 2015-12-29 Bionomics Inc. Methods of inhibiting solid tumor growth by administering GPR49 antibodies
US9574010B2 (en) 2011-11-04 2017-02-21 Zymeworks Inc. Stable heterodimeric antibody design with mutations in the Fc domain
EP3252075A1 (en) 2011-11-04 2017-12-06 Novartis AG Low density lipoprotein-related protein 6 (lrp6) - half life extender constructs
US20140315817A1 (en) 2011-11-18 2014-10-23 Eleven Biotherapeutics, Inc. Variant serum albumin with improved half-life and other properties
CN104159924B (en) 2011-12-05 2018-03-16 诺华股份有限公司 The antibody of EGF-R ELISA 3 (HER3)
AU2012349736A1 (en) 2011-12-05 2014-06-26 Novartis Ag Antibodies for epidermal growth factor receptor 3 (HER3) directed to domain II of HER3
PL2791160T3 (en) 2011-12-16 2022-06-20 Modernatx, Inc. Modified mrna compositions
CA2859493A1 (en) 2011-12-21 2013-06-27 Novartis Ag Compositions and methods for antibodies targeting factor p
CA2858984C (en) 2011-12-22 2021-01-05 Csl Behring Gmbh Use of c1-inhibitor for the treatment of secondary edema of the central nervous system
US20130177574A1 (en) 2012-01-11 2013-07-11 Paul I. Terasaki Foundation Laboratory ANTI-HLA CLASS-Ib ANTIBODIES MIMIC IMMUNOREACTIVITY AND IMMUNOMODULATORY FUNCTIONS OF INTRAVENOUS IMMUNOGLOBULIN (IVIg) USEFUL AS THERAPEUTIC IVIg MIMETICS AND METHODS OF THEIR USE
CN111499761A (en) 2012-01-12 2020-08-07 比奥贝拉蒂治疗公司 Chimeric factor VIII polypeptides and uses thereof
EP2623110A1 (en) 2012-01-31 2013-08-07 CSL Behring GmbH Factor XII inhibitors for the treatment of neurological inflammatory disorders
ES2781773T3 (en) 2012-02-15 2020-09-07 Ecole Polytechnique Fed Lausanne Epfl Erythrocyte binding therapeutics
WO2013120939A1 (en) * 2012-02-15 2013-08-22 Csl Behring Gmbh Von willebrand factor variants having improved factor viii binding affinity
SG10201606783RA (en) 2012-02-15 2016-10-28 Amunix Operating Inc Factor viii compositions and methods of making and using same
WO2013123457A1 (en) 2012-02-15 2013-08-22 Biogen Idec Ma Inc. Recombinant factor viii proteins
EP3093294A1 (en) 2012-02-24 2016-11-16 Stemcentrx, Inc. Dll3 modulators and methods of use
KR102008136B1 (en) 2012-02-27 2019-08-08 베링거 인겔하임 인터내셔날 게엠베하 Cx3cr1-binding polypeptides
BR112014018679A2 (en) 2012-03-16 2017-07-04 Novozymes Biopharma Dk As albumin variants
GB201204868D0 (en) * 2012-03-20 2012-05-02 San Raffaele Centro Fond Peptides
US9592289B2 (en) 2012-03-26 2017-03-14 Sanofi Stable IgG4 based binding agent formulations
JP6254146B2 (en) 2012-03-27 2017-12-27 エヌジーエム バイオファーマシューティカルス,インコーポレーテッド Compositions and methods for treating metabolic disorders
US9878056B2 (en) 2012-04-02 2018-01-30 Modernatx, Inc. Modified polynucleotides for the production of cosmetic proteins and peptides
US9572897B2 (en) 2012-04-02 2017-02-21 Modernatx, Inc. Modified polynucleotides for the production of cytoplasmic and cytoskeletal proteins
US9283287B2 (en) 2012-04-02 2016-03-15 Moderna Therapeutics, Inc. Modified polynucleotides for the production of nuclear proteins
JP2015513913A (en) 2012-04-02 2015-05-18 モデルナ セラピューティクス インコーポレイテッドModerna Therapeutics,Inc. Modified polynucleotide
US9150645B2 (en) 2012-04-20 2015-10-06 Abbvie, Inc. Cell culture methods to reduce acidic species
WO2013158273A1 (en) 2012-04-20 2013-10-24 Abbvie Inc. Methods to modulate c-terminal lysine variant distribution
US9067990B2 (en) 2013-03-14 2015-06-30 Abbvie, Inc. Protein purification using displacement chromatography
WO2013163631A2 (en) * 2012-04-27 2013-10-31 Cytomx Therapeutics, Inc. Activatable antibodies that bind epidermal growth factor receptor and methods of use thereof
WO2013166290A1 (en) 2012-05-04 2013-11-07 Abbvie Biotherapeutics Inc. P21 biomarker assay
US9328174B2 (en) 2012-05-09 2016-05-03 Novartis Ag Chemokine receptor binding polypeptides
ES2843054T3 (en) 2012-05-10 2021-07-15 Zymeworks Inc Heteromultimeric constructions of immunoglobulin heavy chains with mutations in the Fc domain
WO2013176754A1 (en) 2012-05-24 2013-11-28 Abbvie Inc. Novel purification of antibodies using hydrophobic interaction chromatography
WO2013177386A1 (en) 2012-05-24 2013-11-28 Abbvie Biotherapeutics Inc. Biomarkers for predicting response to tweak receptor (tweakr) agonist therapy
BR112014029553B1 (en) 2012-05-30 2019-11-26 Bayer Cropscience Ag composition comprising a biological control agent and a fungicide, its uses, pathogen resistant seed, method for reducing total damage to plants and plant parts, and parts kit
CN108669088A (en) 2012-05-30 2018-10-19 拜耳作物科学股份公司 Include the composition of biocontrol agent and the fungicide selected from Cell wall synthesis inhibitor
US9497972B2 (en) 2012-05-30 2016-11-22 Bayer Cropscience Ag Compositions comprising a biological control agent and an insecticide
MX355327B (en) 2012-05-30 2018-04-16 Bayer Cropscience Ag Compositions comprising a biological control agent and a fungicide from the group consisting of inhibitors of the respiratory chain at complex i or ii.
MX360058B (en) 2012-05-30 2018-10-19 Bayer Cropscience Ag Compositions comprising a biological control agent and an insecticide.
PT2854548T (en) 2012-05-30 2018-12-06 Bayer Cropscience Ag Composition comprising a biological control agent and a fungicide selected from metalaxyl and metalaxyl-m
BR112014029120A2 (en) 2012-05-30 2017-06-27 Bayer Cropscience Ag composition comprising a biological control agent and a fungicide
NZ701724A (en) 2012-05-30 2016-11-25 Bayer Cropscience Ag Compositions comprising a biological control agent and an insecticide
WO2013178658A1 (en) 2012-05-30 2013-12-05 Bayer Cropscience Ag Compositions comprising a biological control agent and an insecticide
MX356529B (en) 2012-05-30 2018-06-01 Bayer Cropscience Ag Composition comprising a biological control agent and a fungicide selected from inhibitors of the mitosis and cell division or compounds having a multi-site action.
AR092326A1 (en) 2012-05-30 2015-04-15 Bayer Cropscience Ag COMPOSITION THAT INCLUDES A BIOLOGICAL CONTROL AGENT AND AN INSECTICIDE
PL2854547T3 (en) 2012-05-30 2019-02-28 Bayer Cropscience Ag Composition comprising a biological control agent and trifloxystrobin
CN104519897A (en) 2012-06-08 2015-04-15 比奥根艾迪克Ma公司 Procoagulant compounds
CN104427995A (en) 2012-06-08 2015-03-18 比奥根艾迪克Ma公司 Chimeric clotting factors
US9150841B2 (en) 2012-06-29 2015-10-06 Shire Human Genetic Therapies, Inc. Cells for producing recombinant iduronate-2-sulfatase
KR101380740B1 (en) 2012-06-29 2014-04-11 쉐어 휴먼 제네텍 세러피스, 인코포레이티드 Purification of iduronate-2-sulfatase
EP3674410A1 (en) 2012-07-11 2020-07-01 Bioverativ Therapeutics Inc. Factor viii complex with xten and von willebrand factor protein, and uses thereof
AU2013289881B2 (en) 2012-07-13 2018-01-18 Zymeworks Bc Inc. Multivalent heteromultimer scaffold design and constructs
WO2014036520A1 (en) 2012-08-30 2014-03-06 Merrimack Pharmaceuticals, Inc. Combination therapies comprising anti-erbb3 agents
KR20150043523A (en) 2012-09-02 2015-04-22 애브비 인코포레이티드 Methods to control protein heterogeneity
US9512214B2 (en) 2012-09-02 2016-12-06 Abbvie, Inc. Methods to control protein heterogeneity
US10391152B2 (en) 2012-10-18 2019-08-27 Bioverativ Therapeutics Inc. Methods of using a fixed dose of a clotting factor
RU2670063C2 (en) 2012-11-08 2018-10-17 Альбумедикс А/С Albumin variants
KR20240033145A (en) 2012-11-13 2024-03-12 비온테크 에스이 Agents for treatment of claudin expressing cancer diseases
DK2922554T3 (en) 2012-11-26 2022-05-23 Modernatx Inc Terminalt modificeret rna
WO2014084859A1 (en) 2012-11-30 2014-06-05 Novartis Ag Molecules and methods for modulating tmem16a activities
AP2015008365A0 (en) 2012-12-05 2015-04-30 Novartis Ag Compositions and methods for antibodies targeting epo
EP3907237A1 (en) 2012-12-20 2021-11-10 Amgen Inc. Apj receptor agonists and uses thereof
MX2015009141A (en) 2013-01-15 2016-03-16 Teva Pharma Formulations of albu-bche, preparation and uses thereof.
KR101503907B1 (en) * 2013-01-17 2015-03-20 서울대학교산학협력단 Recombinant protein vaccine for preventing Actinobacillus pleuropneumoniae and Mycoplasma hyopneumoniae infection diseases
US9161966B2 (en) 2013-01-30 2015-10-20 Ngm Biopharmaceuticals, Inc. GDF15 mutein polypeptides
KR101993714B1 (en) 2013-01-30 2019-06-28 엔지엠 바이오파마슈티컬스, 아이엔씨. Compositions and methods of use in treating metabolic disorders
MX2015010259A (en) 2013-02-11 2015-10-29 Bayer Cropscience Lp Compositions comprising a streptomyces-based biological control agent and another biological control agent.
EP3889173B1 (en) 2013-02-15 2023-07-05 Bioverativ Therapeutics Inc. Optimized factor viii gene
DK2956002T3 (en) 2013-02-16 2017-12-11 Albumedix As Pharmacokinetic animal model
ES2609070T3 (en) 2013-02-28 2017-04-18 Csl Behring Gmbh Therapeutic agent for amniotic fluid embolism
US9255262B2 (en) * 2013-03-06 2016-02-09 Vision Global Holdings Ltd. Albumin-binding arginine deminase and the use thereof
USRE48805E1 (en) 2013-03-06 2021-11-02 Vision Global Holdings Ltd. Method for cancer targeting treatment and detection of arginine using albumin-binding arginine deiminase fusion protein
AU2014224599B2 (en) 2013-03-08 2018-11-08 Csl Behring Gmbh Treatment and prevention of remote ischemia-reperfusion injury
CA2940513C (en) 2013-03-11 2023-08-15 University Of Florida Research Foundation, Inc. Delivery of card protein as therapy for ocular inflammation
AU2013381687A1 (en) 2013-03-12 2015-09-24 Abbvie Inc. Human antibodies that bind human TNF-alpha and methods of preparing the same
EP2970408B1 (en) 2013-03-12 2018-01-10 Amgen Inc. Potent and selective inhibitors of nav1.7
US20160152686A1 (en) 2013-03-13 2016-06-02 Bristol-Myers Squibb Company Fibronectin based scaffold domains linked to serum albumin or moiety binding thereto
US20160024181A1 (en) * 2013-03-13 2016-01-28 Moderna Therapeutics, Inc. Long-lived polynucleotide molecules
US8921526B2 (en) 2013-03-14 2014-12-30 Abbvie, Inc. Mutated anti-TNFα antibodies and methods of their use
WO2014159239A2 (en) 2013-03-14 2014-10-02 Novartis Ag Antibodies against notch 3
WO2014151878A2 (en) 2013-03-14 2014-09-25 Abbvie Inc. Methods for modulating protein glycosylation profiles of recombinant protein therapeutics using monosaccharides and oligosacharides
US9017687B1 (en) 2013-10-18 2015-04-28 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same using displacement chromatography
SG11201505926VA (en) 2013-03-15 2015-09-29 Biogen Ma Inc Factor ix polypeptide formulations
US8980864B2 (en) 2013-03-15 2015-03-17 Moderna Therapeutics, Inc. Compositions and methods of altering cholesterol levels
JP6505079B2 (en) 2013-03-29 2019-04-24 アレクシオン ファーマシューティカルズ, インコーポレイテッド Compositions and methods for increasing the serum half-life of a therapeutic that targets complement C5
WO2014166836A1 (en) * 2013-04-05 2014-10-16 Novo Nordisk A/S Growth hormone compound formulation
EP2796145B1 (en) 2013-04-22 2017-11-01 CSL Ltd. A covalent complex of von willebrand factor and faktor viii linked by a disulphide bridge
US9562101B2 (en) 2013-06-21 2017-02-07 Novartis Ag Lectin-like oxidized LDL receptor 1 antibodies and methods of use
AR096601A1 (en) 2013-06-21 2016-01-20 Novartis Ag ANTIBODIES OF LEXINED OXIDATED LDL RECEIVER 1 AND METHODS OF USE
KR20160026905A (en) 2013-06-28 2016-03-09 체에스엘 베링 게엠베하 Combination therapy using a factor xii inhibitor and a c1-inhibitor
CN104342420B (en) * 2013-07-30 2017-09-15 惠觅宙 A kind of recombinant long-acting people hyaluronidase, its encoding gene, production method and application
US10548953B2 (en) 2013-08-14 2020-02-04 Bioverativ Therapeutics Inc. Factor VIII-XTEN fusions and uses thereof
WO2015023894A1 (en) 2013-08-14 2015-02-19 Biogen Idec Ma Inc. Recombinant factor viii proteins
CA2920835A1 (en) 2013-08-20 2015-02-26 Anutra Medical, Inc. Syringe fill system and method
EP3038634A4 (en) 2013-08-28 2017-10-11 AbbVie Stemcentrx LLC Novel sez6 modulators and methods of use
EP3892294A1 (en) 2013-08-28 2021-10-13 AbbVie Stemcentrx LLC Site-specific antibody conjugation methods and compositions
ES2900425T3 (en) 2013-09-25 2022-03-16 Bioverativ Therapeutics Inc Column viral inactivation methods
CN103468662A (en) * 2013-09-29 2013-12-25 惠觅宙 Recombined human hyaluronidase, production and purification method and preparations thereof, use method and application
EP3052106A4 (en) 2013-09-30 2017-07-19 ModernaTX, Inc. Polynucleotides encoding immune modulating polypeptides
BR112016007255A2 (en) 2013-10-03 2017-09-12 Moderna Therapeutics Inc polynucleotides encoding low density lipoprotein receptor
US9598667B2 (en) 2013-10-04 2017-03-21 Abbvie Inc. Use of metal ions for modulation of protein glycosylation profiles of recombinant proteins
WO2015057403A2 (en) * 2013-10-17 2015-04-23 Tarix Pharmaceuticals Ltd. Compositions and methods for treatment of inflammatory bowel disease
US9181337B2 (en) 2013-10-18 2015-11-10 Abbvie, Inc. Modulated lysine variant species compositions and methods for producing and using the same
US9085618B2 (en) 2013-10-18 2015-07-21 Abbvie, Inc. Low acidic species compositions and methods for producing and using the same
US8946395B1 (en) 2013-10-18 2015-02-03 Abbvie Inc. Purification of proteins using hydrophobic interaction chromatography
US9540440B2 (en) 2013-10-30 2017-01-10 Cytomx Therapeutics, Inc. Activatable antibodies that bind epidermal growth factor receptor and methods of use thereof
WO2015066557A1 (en) 2013-10-31 2015-05-07 Resolve Therapeutics, Llc Therapeutic nuclease molecules with altered glycosylation and methods
KR101977449B1 (en) * 2013-11-01 2019-05-10 유니버시티에트 이 오슬로 Albumin variants and uses thereof
EP3065769A4 (en) 2013-11-08 2017-05-31 Biogen MA Inc. Procoagulant fusion compound
WO2015073884A2 (en) 2013-11-15 2015-05-21 Abbvie, Inc. Glycoengineered binding protein compositions
WO2015089283A1 (en) 2013-12-11 2015-06-18 Cytomx Therapeutics, Inc. Antibodies that bind activatable antibodies and methods of use thereof
US8986691B1 (en) 2014-07-15 2015-03-24 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
US8980273B1 (en) 2014-07-15 2015-03-17 Kymab Limited Method of treating atopic dermatitis or asthma using antibody to IL4RA
ES2902835T3 (en) 2014-01-09 2022-03-30 Hadasit Med Res Service Improved cell compositions and methods for cancer therapy
SG11201605242YA (en) 2014-01-10 2016-07-28 Biogen Ma Inc Factor viii chimeric proteins and uses thereof
US10946079B2 (en) 2014-02-21 2021-03-16 Ecole Polytechnique Federale De Lausanne Glycotargeting therapeutics
US10821157B2 (en) 2014-02-21 2020-11-03 Anokion Sa Glycotargeting therapeutics
US10953101B2 (en) 2014-02-21 2021-03-23 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
US10046056B2 (en) 2014-02-21 2018-08-14 École Polytechnique Fédérale De Lausanne (Epfl) Glycotargeting therapeutics
ES2911714T3 (en) 2014-03-11 2022-05-20 Univ Florida AAV-expressed protein M013 as an anti-inflammatory therapeutic for use in a method of treating inflammatory eye disease
US20170107294A1 (en) * 2014-03-21 2017-04-20 Nordlandssykehuset Hf Anti-cd14 antibodies and uses thereof
WO2015160618A1 (en) 2014-04-16 2015-10-22 Bayer Cropscience Lp Compositions comprising ningnanmycin and a biological control agent
WO2015164615A1 (en) 2014-04-24 2015-10-29 University Of Oslo Anti-gluten antibodies and uses thereof
BR112016025470A2 (en) 2014-05-02 2017-08-15 Cerenis Therapeutics Holding S A therapeutic hdl?
CN105198999A (en) * 2014-05-27 2015-12-30 上海生物制品研究所有限责任公司 Fusion protein and its preparation method and use
USD763433S1 (en) 2014-06-06 2016-08-09 Anutra Medical, Inc. Delivery system cassette
USD774182S1 (en) 2014-06-06 2016-12-13 Anutra Medical, Inc. Anesthetic delivery device
USD750768S1 (en) 2014-06-06 2016-03-01 Anutra Medical, Inc. Fluid administration syringe
EP4257152A3 (en) 2014-06-10 2023-12-06 Amgen Inc. Apelin polypeptides
CN106456778A (en) 2014-06-18 2017-02-22 德国杰特贝林生物制品有限公司 Therapy using a factor XII inhibitor in a neurotraumatic disorder
US20170204149A1 (en) 2014-06-23 2017-07-20 Novartis Ag Hsa-gdf-15 fusion polypeptide and use thereof
CN106459222A (en) 2014-06-24 2017-02-22 诺和诺德股份有限公司 MIC-1 fusion proteins and uses thereof
EP3160478A4 (en) 2014-06-30 2018-05-16 Bioverativ Therapeutics Inc. Optimized factor ix gene
EP3174894B1 (en) 2014-07-30 2021-06-23 NGM Biopharmaceuticals, Inc. Compositions and methods of use for treating metabolic disorders
CU24453B1 (en) 2014-08-07 2019-11-04 Novartis Ag ANTI-PROTEIN ANTIBODIES SIMILAR TO ANGIOPOYETIN 4
TW201613977A (en) 2014-08-07 2016-04-16 Novartis Ag Angiopoetin-like 4 (ANGPTL4) antibodies and methods of use
EP3182988A4 (en) * 2014-08-22 2018-05-23 National Cheng Kung University Disintegrin variants and pharmaceutical uses thereof
CA2961382C (en) 2014-09-17 2024-02-13 Bayer Cropscience Lp Compositions comprising recombinant bacillus cells and another biological control agent
US9616114B1 (en) 2014-09-18 2017-04-11 David Gordon Bermudes Modified bacteria having improved pharmacokinetics and tumor colonization enhancing antitumor activity
JP2018500272A (en) 2014-09-26 2018-01-11 バイエル ファーマ アクチエンゲゼルシャフト Stabilized adrenomedullin derivatives and uses thereof
MA41044A (en) 2014-10-08 2017-08-15 Novartis Ag COMPOSITIONS AND METHODS OF USE FOR INCREASED IMMUNE RESPONSE AND CANCER TREATMENT
EP3207130B1 (en) 2014-10-14 2019-08-07 Halozyme, Inc. Compositions of adenosine deaminase-2 (ada2), variants thereof and methods of using same
MA40835A (en) 2014-10-23 2017-08-29 Biogen Ma Inc ANTI-GPIIB / IIIA ANTIBODIES AND THEIR USES
DK3212226T3 (en) 2014-10-31 2020-06-15 Ngm Biopharmaceuticals Inc COMPOSITIONS AND METHODS OF USE FOR TREATING METABOLIC DISORDERS
MA40861A (en) 2014-10-31 2017-09-05 Biogen Ma Inc ANTI-GLYCOPROTEIN IIB / IIIA ANTIBODIES
US20160130324A1 (en) * 2014-10-31 2016-05-12 Shire Human Genetic Therapies, Inc. C1 Inhibitor Fusion Proteins and Uses Thereof
GB201420139D0 (en) 2014-11-12 2014-12-24 Ucl Business Plc Factor IX gene therapy
UY36449A (en) 2014-12-19 2016-07-29 Novartis Ag COMPOSITIONS AND METHODS FOR ANTIBODIES DIRECTED TO BMP6
EP3236939B1 (en) * 2014-12-23 2020-04-01 Merz Pharma GmbH & Co. KGaA Botulinum toxin prefilled container
CA2981312C (en) * 2015-03-30 2023-09-26 Regeneron Pharmaceuticals, Inc. Heavy chain constant regions with reduced binding to fc gamma receptors
JP2016204292A (en) * 2015-04-21 2016-12-08 国立大学法人 熊本大学 Bmp7 mutant-albumin cointegrate, and renal disease therapeutic agent containing the same
CN107531771A (en) * 2015-04-29 2018-01-02 意大利梅迪奥兰制药公司 Soluble chimeric Interleukin 10 receptor and its therapeutical uses
WO2016195723A1 (en) * 2015-06-03 2016-12-08 Beller Pharmaceuticals LLC Methods of treatment for conditions mediated by substance p
WO2016193872A2 (en) 2015-06-05 2016-12-08 Novartis Ag Antibodies targeting bone morphogenetic protein 9 (bmp9) and methods therefor
WO2016201202A1 (en) * 2015-06-11 2016-12-15 Attwill Medical Solutions Inc. Medical devices, systems, and methods utilizing antithrombin-heparin compositions
JOP20200312A1 (en) 2015-06-26 2017-06-16 Novartis Ag Factor xi antibodies and methods of use
JP2018522030A (en) * 2015-07-30 2018-08-09 エンドール テクノロジーズ, エス.エレ.Endor Technologies, S.L. Colony stimulating factor for the treatment of pancreatic or colon cancer
AU2016301303B2 (en) 2015-08-03 2021-10-07 Bioverativ Therapeutics Inc. Factor IX fusion proteins and methods of making and using same
CN108350072B (en) 2015-08-03 2022-05-24 诺华股份有限公司 Methods of treating FGF 21-associated disorders
KR20180057609A (en) * 2015-08-06 2018-05-30 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Use thereof in GLP-1 and compositions for the treatment of metabolic diseases
WO2017029407A1 (en) 2015-08-20 2017-02-23 Albumedix A/S Albumin variants and conjugates
WO2017043569A1 (en) 2015-09-08 2017-03-16 Jcrファーマ株式会社 Novel human serum albumin mutant
JP6516925B2 (en) 2015-09-09 2019-05-22 ノバルティス アーゲー Thymic stromal lymphopoietic factor (TSLP) binding antibodies and methods of using the antibodies
MY186352A (en) 2015-09-09 2021-07-15 Novartis Ag Thymic stromal lymphopoietin (tslp)-binding antibodies and methods of using the antibodies
AU2016318621A1 (en) * 2015-09-11 2018-05-10 The Board Of Trustees Of The Leland Stanford Junior University Biologically relevant orthogonal cytokine/receptor pairs
EP3383425B1 (en) 2015-12-04 2020-07-15 Boehringer Ingelheim International GmbH Biparatopic polypeptides antagonizing wnt signaling in tumor cells
CN116077648A (en) 2015-12-11 2023-05-09 武田药品工业株式会社 Plasma kallikrein inhibitors and their use for the treatment of hereditary angioedema attacks
UY37030A (en) 2015-12-18 2017-07-31 Novartis Ag ANTIBODIES DIRECTED TO CD32B AND METHODS OF USE OF THE SAME
WO2017109706A1 (en) 2015-12-22 2017-06-29 Novartis Ag Methods of treating or ameliorating metabolic disorders using growth differentiation factor 15 (gdf-15)
CA3009001A1 (en) 2016-01-11 2017-07-20 Universitat Zurich Immune-stimulating humanized monoclonal antibodies against human interleukin-2, and fusion proteins thereof
EP3405492B1 (en) 2016-01-21 2020-10-21 Novartis AG Multispecific molecules targeting cll-1
CA3012695A1 (en) 2016-02-01 2017-08-10 Bioverativ Therapeutics Inc. Optimized factor viii genes
WO2017159540A1 (en) 2016-03-14 2017-09-21 Jcrファーマ株式会社 Serum albumin-20 k growth hormone fusion protein
WO2017161414A1 (en) 2016-03-22 2017-09-28 Bionomics Limited Administration of an anti-lgr5 monoclonal antibody
WO2017172260A1 (en) 2016-03-31 2017-10-05 Ngm Bioparmaceuticals, Inc. Binding proteins and methods of use thereof
US11174321B2 (en) 2016-04-06 2021-11-16 Csl Limited Method of treating atherosclerosis
EP3448887A1 (en) 2016-04-27 2019-03-06 Novartis AG Antibodies against growth differentiation factor 15 and uses thereof
HUE053452T2 (en) 2016-05-18 2021-07-28 Boehringer Ingelheim Int Anti pd-1 and anti-lag3 antibodies for cancer treatment
JP7231411B2 (en) 2016-06-15 2023-03-01 ノバルティス アーゲー Methods of treating diseases using inhibitors of bone morphogenetic protein 6 (BMP6)
WO2018007601A1 (en) * 2016-07-08 2018-01-11 Csl Behring Recombinant Facility Ag Subcutaneous administration of long-acting factor ix in humans
WO2018014260A1 (en) 2016-07-20 2018-01-25 Nanjing Legend Biotech Co., Ltd. Multispecific antigen binding proteins and methods of use thereof
CN109562149A (en) 2016-08-05 2019-04-02 德国杰特贝林生物制品有限公司 The pharmaceutical preparation of c1 esterase inhibitor
US20190183991A1 (en) 2016-08-23 2019-06-20 Csl Behring Gmbh Method of preventing acute attacks of hereditary angioedema associated with c1 esterase inhibitor deficiency
WO2018068201A1 (en) 2016-10-11 2018-04-19 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against ctla-4
WO2018082758A1 (en) 2016-11-04 2018-05-11 Aarhus Universitet Identification and treatment of tumors characterized by an overexpression of the neonatal fc receptor
JP2019535834A (en) * 2016-11-18 2019-12-12 ユニベルシタ デ バルセロナ CD6 and imipenem combination therapy for the treatment of infectious diseases and related inflammatory processes
WO2018096396A1 (en) 2016-11-22 2018-05-31 University Of Oslo Albumin variants and uses thereof
JP2020500874A (en) 2016-12-02 2020-01-16 バイオベラティブ セラピューティクス インコーポレイテッド Method of treating hemophilic arthropathy using chimeric clotting factor
CA3044838A1 (en) 2016-12-02 2018-06-07 Bioverativ Therapeutics Inc. Methods of inducing immune tolerance to clotting factors
US11129906B1 (en) 2016-12-07 2021-09-28 David Gordon Bermudes Chimeric protein toxins for expression by therapeutic bacteria
US11180535B1 (en) 2016-12-07 2021-11-23 David Gordon Bermudes Saccharide binding, tumor penetration, and cytotoxic antitumor chimeric peptides from therapeutic bacteria
EP3562840A1 (en) 2016-12-16 2019-11-06 Biogen MA Inc. Stabilized proteolytically activated growth differentiation factor 11
EP3360898A1 (en) 2017-02-14 2018-08-15 Boehringer Ingelheim International GmbH Bispecific anti-tnf-related apoptosis-inducing ligand receptor 2 and anti-cadherin 17 binding molecules for the treatment of cancer
EP3559047A1 (en) 2016-12-23 2019-10-30 Novartis AG Factor xi antibodies and methods of use
JP7456605B2 (en) 2016-12-23 2024-03-27 プレジデント アンド フェローズ オブ ハーバード カレッジ PCSK9 gene editing
CA3048157A1 (en) 2016-12-23 2018-06-28 Novartis Ag Methods of treatment with anti-factor xi/xia antibodies
US20190382462A1 (en) 2017-01-13 2019-12-19 Pietro P. Sanna Methods and compositions for treating hpa hyperactivity
EP3576762A1 (en) 2017-01-31 2019-12-11 Bioverativ Therapeutics Inc. Factor ix fusion proteins and methods of making and using same
TWI786087B (en) 2017-02-08 2022-12-11 瑞士商諾華公司 Fgf21 mimetic antibodies and uses thereof
WO2018193033A1 (en) 2017-04-20 2018-10-25 Novo Nordisk A/S Methods of purification of albumin fusion proteins
TWI710377B (en) 2017-05-23 2020-11-21 丹麥商諾佛 儂迪克股份有限公司 Mic-1 compounds and uses thereof
MA48760A (en) 2017-05-31 2020-04-08 Boehringer Ingelheim Int POLYPEPTIDES HAVING AN ANTAGONIST EFFECT ON WNT SIGNALING IN TUMOR CELLS
WO2018229715A1 (en) 2017-06-16 2018-12-20 Novartis Ag Compositions comprising anti-cd32b antibodies and methods of use thereof
US11253579B2 (en) 2017-06-16 2022-02-22 The University Of Chicago Compositions and methods for inducing immune tolerance
CA3068360A1 (en) 2017-06-29 2019-01-03 CSL Behring Lengnau AG 21-day dosing regimen for fusion proteins comprising factor ix and human albumin for prophylactic treatment of hemophilia and methods thereof
US20210346513A1 (en) 2017-08-04 2021-11-11 Amgen Inc. Method of conjugation of cys-mabs
CA3072334A1 (en) 2017-08-09 2019-02-14 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof
US11485781B2 (en) 2017-08-17 2022-11-01 Massachusetts Institute Of Technology Multiple specificity binders of CXC chemokines
US20200276283A1 (en) 2017-09-15 2020-09-03 Cedars-Sinai Medical Center Methods for improving organ function in organ transplant patients
EP3697812A4 (en) 2017-10-18 2021-09-22 CSL Limited Human serum albumin variants and uses thereof
US20210040205A1 (en) 2017-10-25 2021-02-11 Novartis Ag Antibodies targeting cd32b and methods of use thereof
JP7317827B2 (en) 2017-12-15 2023-07-31 シーエスエル、リミテッド Use of FXIIa inhibitors in the treatment of renal fibrosis and/or chronic kidney disease
WO2019129054A1 (en) 2017-12-27 2019-07-04 信达生物制药(苏州)有限公司 Triabody, preparation method and use thereof
WO2019129221A1 (en) 2017-12-28 2019-07-04 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against tigit
US11246908B2 (en) * 2018-01-10 2022-02-15 The Johns Hopkins University Compositions comprising albumin-FMS-like tyrosine kinase 3 ligand fusion proteins and uses thereof
WO2019137541A1 (en) 2018-01-15 2019-07-18 Nanjing Legend Biotech Co., Ltd. Single-domain antibodies and variants thereof against pd-1
EP3746136A1 (en) 2018-02-01 2020-12-09 Bioverativ Therapeutics Inc. Use of lentiviral vectors expressing factor viii
PL3765055T3 (en) * 2018-03-13 2023-11-27 Sepsia Therapeutics, S.L. Bacterial-binding peptides for treatment of infectious diseases and related inflammatory processes
CA3093477A1 (en) 2018-03-13 2019-09-19 Zymeworks Inc. Anti-her2 biparatopic antibody-drug conjugates and methods of use
AU2019244091B2 (en) 2018-03-28 2023-12-07 Bristol-Myers Squibb Company Interleukin-2/Interleukin-2 receptor alpha fusion proteins and methods of use
KR102119197B1 (en) * 2018-04-23 2020-06-05 주식회사 엘베이스 Composition for autophagy inhibiting in cell, And pharmaceutical composition for preventing or treating tumor disease, or inhibiting anti-cancer agents resistance containing the same
DK3794024T3 (en) 2018-05-14 2023-08-14 Werewolf Therapeutics Inc ACTIVABLE INTERLEUKIN-2 POLYPEPTIDES AND METHODS OF USING THEREOF
BR112020023167A2 (en) 2018-05-14 2021-02-09 Werewolf Therapeutics, Inc. activatable cytokine polypeptides and methods of using these
US20190375822A1 (en) 2018-05-18 2019-12-12 Bioverativ Therapeutics Inc. Methods of treating hemophilia a
EP3569618A1 (en) 2018-05-19 2019-11-20 Boehringer Ingelheim International GmbH Antagonizing cd73 antibody
AR126019A1 (en) 2018-05-30 2023-09-06 Novartis Ag ANTIBODIES AGAINST ENTPD2, COMBINATION THERAPIES AND METHODS OF USE OF ANTIBODIES AND COMBINATION THERAPIES
TW202016136A (en) 2018-06-01 2020-05-01 瑞士商諾華公司 Binding molecules against bcma and uses thereof
BR112020026512A2 (en) 2018-07-03 2021-04-06 Bristol-Myers Squibb Company FGF-21 FORMULATIONS
WO2020033863A1 (en) 2018-08-09 2020-02-13 Bioverativ Therapeutics Inc. Nucleic acid molecules and uses thereof for non-viral gene therapy
US10842885B2 (en) 2018-08-20 2020-11-24 Ucl Business Ltd Factor IX encoding nucleotides
MA53495A (en) 2018-08-31 2021-12-08 Regeneron Pharma DOSING STRATEGY TO MITIGATE CYTOKINE RELEASE SYNDROME FOR CD3/C20 BISPECIFIC ANTIBODIES
US20220056115A1 (en) * 2018-09-17 2022-02-24 Kyoto University Administration of an anti-c5 agent for treatment of hepatic injury or failure
JP2022502088A (en) 2018-09-27 2022-01-11 エクシリオ デベロップメント, インコーポレイテッド Masked cytokine polypeptide
UY38407A (en) 2018-10-15 2020-05-29 Novartis Ag TREM2 STABILIZING ANTIBODIES
KR20210111245A (en) * 2018-10-29 2021-09-10 스핀 테라퓨틱스, 엘엘씨 Compositions and methods for alpha-1-antitrypsin disorders
GB201818477D0 (en) 2018-11-13 2018-12-26 Emstopa Ltd Tissue plasminogen activator antibodies and method of use thereof
WO2020109978A1 (en) 2018-11-26 2020-06-04 Novartis Ag Lpl-gpihbp1 fusion polypeptides
KR20210100661A (en) 2018-12-06 2021-08-17 바이오버라티브 테라퓨틱스 인크. Use of Lentiviral Vectors Expressing Factor IX
KR20220044612A (en) 2019-03-22 2022-04-08 리플렉시온 파마슈티컬스, 인크. D-peptidic compounds for vegf
US20230051872A1 (en) 2019-03-22 2023-02-16 Reflexion Pharmaceuticals, Inc. Multivalent D-Peptidic Compounds for Target Proteins
EP3947442A2 (en) 2019-03-28 2022-02-09 Danisco US Inc. Engineered antibodies
JP2022526827A (en) 2019-04-11 2022-05-26 アンジオン バイオメディカ コーポレーション Solid form of (E) -3- [2- (2-thienyl) vinyl] -1H-pyrazole
EP3969035A4 (en) 2019-05-14 2023-06-21 Werewolf Therapeutics, Inc. Separation moieties and methods and use thereof
US20230071196A1 (en) 2019-05-21 2023-03-09 Novartis Ag Variant cd58 domains and uses thereof
CR20210576A (en) 2019-05-21 2021-12-15 Novartis Ag Cd19 binding molecules and uses thereof
JP2022532928A (en) 2019-05-24 2022-07-20 サノフイ Methods for treating systemic scleroderma
US20220387608A1 (en) 2019-06-18 2022-12-08 Bayer Aktiengesellschaft Adrenomedullin-analogues for long-term stabilization and their use
CA3152990A1 (en) 2019-09-06 2021-03-11 Novartis Ag Therapeutic fusion proteins
JP2022548881A (en) 2019-09-18 2022-11-22 ノバルティス アーゲー ENTPD2 Antibodies, Combination Therapy and Methods of Using Antibodies and Combination Therapy
TW202124446A (en) 2019-09-18 2021-07-01 瑞士商諾華公司 Combination therapies with entpd2 antibodies
WO2021067389A1 (en) 2019-09-30 2021-04-08 Bioverativ Therapeutics Inc. Lentiviral vector formulations
JP2021066730A (en) 2019-10-17 2021-04-30 Jcrファーマ株式会社 Method for producing fusion protein of serum albumin and growth hormone
CN114728043A (en) 2019-10-30 2022-07-08 Jcr制药股份有限公司 Aqueous pharmaceutical composition containing fusion protein of serum albumin and growth hormone
WO2021111143A1 (en) 2019-12-04 2021-06-10 Albumedix Limited Methods and compositions produced thereby
WO2021119534A2 (en) 2019-12-13 2021-06-17 Synthekine, Inc. Il-2 orthologs and methods of use
US20210181200A1 (en) * 2019-12-17 2021-06-17 Women's College Hospital Ovarian cancer biomarker and methods of using same
WO2021202473A2 (en) 2020-03-30 2021-10-07 Danisco Us Inc Engineered antibodies
MX2022014538A (en) 2020-05-19 2022-12-15 Boehringer Ingelheim Int Binding molecules for the treatment of cancer.
EP4232474A1 (en) 2020-10-21 2023-08-30 Boehringer Ingelheim International GmbH Bispecific anti-vegf and anti-trkb binding molecules for the treatment of eye diseases
IL302569A (en) 2020-11-06 2023-07-01 Novartis Ag Cd19 binding molecules and uses thereof
WO2022166720A1 (en) * 2021-02-05 2022-08-11 华南理工大学 Serum albumin-based fusion protein, and nano-assembly, preparation method therefor and application thereof
EP4294834A2 (en) 2021-02-19 2023-12-27 The United States of America, as represented by The Secretary, Department of Health and Human Services Single domain antibodies that neutralize sars-cov-2
WO2022263507A1 (en) 2021-06-17 2022-12-22 Boehringer Ingelheim International Gmbh Novel tri-specific binding molecules
CN114133458B (en) * 2021-12-08 2023-11-14 福州大学 Method for fusing polypeptide in human serum albumin
US20230357381A1 (en) 2022-04-26 2023-11-09 Novartis Ag Multispecific antibodies targeting il-13 and il-18
WO2024015953A1 (en) 2022-07-15 2024-01-18 Danisco Us Inc. Methods for producing monoclonal antibodies
WO2024013315A1 (en) 2022-07-15 2024-01-18 Boehringer Ingelheim International Gmbh Binding molecules for the treatment of cancer

Citations (88)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4264731A (en) * 1977-05-27 1981-04-28 The Regents Of The University Of California DNA Joining method
US4440859A (en) * 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4447538A (en) * 1978-04-19 1984-05-08 Regents Of The University Of California Microorganism containing gene for human chorionic somatomammotropin
US4450103A (en) * 1982-03-01 1984-05-22 Cetus Corporation Process for recovering human IFN-β from a transformed microorganism
US4462940A (en) * 1982-09-23 1984-07-31 Cetus Corporation Process for the recovery of human β-interferon-like polypeptides
US4492684A (en) * 1983-06-08 1985-01-08 Connaught Laboratories Limited Slow release injectable insulin composition
US4499188A (en) * 1982-05-05 1985-02-12 Cetus Corporation Bacterial production of heterologous polypeptides under the control of a repressible promoter-operator
US4652525A (en) * 1978-04-19 1987-03-24 The Regents Of The University Of California Recombinant bacterial plasmids containing the coding sequences of insulin genes
US4835260A (en) * 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
US4840934A (en) * 1983-01-25 1989-06-20 Eleanor Roosevelt Institute For Cancer Research, Inc. Therapeutic method using T cell growth factor
US4908434A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Process for preparing purified interleukin-2
US4908433A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Uses of interleukin-2
US4914026A (en) * 1983-04-07 1990-04-03 Chiron Corporation Alpha factor leader sequence directed secretion of insulin
US4916212A (en) * 1984-05-30 1990-04-10 Novo Industri A/S DNA-sequence encoding biosynthetic insulin precursors and process for preparing the insulin precursors and human insulin
US4925919A (en) * 1984-04-25 1990-05-15 Roland Mertelsmann Purified interleukin 2
US4929442A (en) * 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US4999339A (en) * 1988-03-28 1991-03-12 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5002764A (en) * 1986-08-12 1991-03-26 Schering Corporation Treatment of actinic keratoses with alpha2 interferon
US5010003A (en) * 1983-04-25 1991-04-23 Genentech, Inc. Use of yeast homologous signals to secrete heterologous proteins
US5015575A (en) * 1983-04-07 1991-05-14 Chiron Corporation Hybrid DNA synthesis of insulin
US5028422A (en) * 1986-05-27 1991-07-02 Schering Corporation Treatment of basal cell carcinoma intralesionally with recombinant human alpha interferon
US5096885A (en) * 1988-04-15 1992-03-17 Genentech, Inc. Human growth hormone formulation
US5096707A (en) * 1988-04-15 1992-03-17 The United States Of America As Represented By The Department Of Health And Human Services Flavone-8-acetic acid and interleukin-2 in a method of treating certain cancers
US5102872A (en) * 1985-09-20 1992-04-07 Cetus Corporation Controlled-release formulations of interleukin-2
US5106954A (en) * 1989-07-26 1992-04-21 Behringwerke Aktiengesellschaft Erythropoietin (epo) peptides
US5126129A (en) * 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
US5128126A (en) * 1989-04-11 1992-07-07 Boehringer Ingelheim International Gmbh Use of pharmaceutical compositions containing at least one cytokine for the systemic treatment of preneoplastic lesions
US5208018A (en) * 1990-03-19 1993-05-04 Brigham And Women's Hospital Treatment of cachexia with interleukin 2
US5219565A (en) * 1990-04-17 1993-06-15 Roussel Uclaf Treatment of primary cancers of the pleura
US5304473A (en) * 1991-06-11 1994-04-19 Eli Lilly And Company A-C-B proinsulin, method of manufacturing and using same, and intermediates in insulin production
US5322930A (en) * 1990-08-07 1994-06-21 Scios Nova Inc. Expression of recombinant polypeptides with improved purification
US5395922A (en) * 1989-03-03 1995-03-07 Novo Nordisk A/S Yeast processing system
US5409815A (en) * 1988-02-16 1995-04-25 The Green Cross Corporation DNA's encoding signal peptides
US5503993A (en) * 1987-12-02 1996-04-02 The Green Cross Corporation Method of preparing foreign protein in yeast, recombinant DNA, transformant
US5508031A (en) * 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
US5512549A (en) * 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US5521086A (en) * 1993-09-16 1996-05-28 Cephalon, Inc. Secretion sequence for the production of a heterologous protein in yeast
US5602232A (en) * 1993-02-25 1997-02-11 Schering Corporation Method for producing metal-interferon-α crystals
US5618676A (en) * 1981-02-25 1997-04-08 Genentech, Inc. Expression of polypeptides in yeast
US5618698A (en) * 1983-12-13 1997-04-08 Kirin-Amgen, Inc. Production of erythropoietin
US5625041A (en) * 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
US5629286A (en) * 1994-03-31 1997-05-13 Brewitt; Barbara Homeopathic dilutions of growth factors
US5637504A (en) * 1987-04-09 1997-06-10 Delta Biotechnology Limited Stable yeast 2 μm vector
US5639642A (en) * 1994-06-16 1997-06-17 Novo Nordisk A/S Synthetic leader peptide sequences
US5641663A (en) * 1985-11-06 1997-06-24 Cangene Corporation Expression system for the secretion of bioactive human granulocyte macrophage colony stimulating factor (GM-CSF) and other heterologous proteins from steptomyces
US5726038A (en) * 1993-07-08 1998-03-10 Novo Nordisk A/S DNA construct encoding the YAP3 signal peptide
US5728553A (en) * 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
US5728707A (en) * 1995-07-21 1998-03-17 Constantia Gruppe Treatment and prevention of primary and metastatic neoplasms with salts of aminoimidazole carboxamide
US5739007A (en) * 1986-08-29 1998-04-14 Delta Biotechnology Limited Hybrid GAL10/pgk yeast promoter
US5741815A (en) * 1995-06-02 1998-04-21 Lai; Ching-San Methods for in vivo reduction of nitric oxide levels and compositions useful therefor
US5763394A (en) * 1988-04-15 1998-06-09 Genentech, Inc. Human growth hormone aqueous formulation
US5766620A (en) * 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5767097A (en) * 1996-01-23 1998-06-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
US5908830A (en) * 1996-10-31 1999-06-01 Merck & Co., Inc. Combination therapy for the treatment of diabetes and obesity
US5912229A (en) * 1996-03-01 1999-06-15 Novo Nordisk Als Use of a pharmaceutical composition comprising an appetite-suppressing peptide
US6017545A (en) * 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
US6031004A (en) * 1997-12-08 2000-02-29 Bristol-Myers Squibb Company Salts of metformin and method
US6045788A (en) * 1996-02-28 2000-04-04 Cornell Research Foundation, Inc. Method of stimulation of immune response with low doses of IL-2
US6048724A (en) * 1991-11-05 2000-04-11 Transkaryotic Therapies Inc. Method of producing clonal cell strains which express exogenous DNA encoding glucagon-like peptide 1
US6054489A (en) * 1996-10-15 2000-04-25 Loyola University Of Chicago Method for the enhancement of lymphocyte activity against tumors
US6063373A (en) * 1989-09-19 2000-05-16 Maxim Pharmaceuticals, Inc. Enhanced activation of NK cells using an NK cell activator and a hydrogen peroxide scavenger or inhibitor
US6069135A (en) * 1989-09-21 2000-05-30 Hyal Pharmaceutical Corporation Use of hyaluronic acid or its derivatives to enhance delivery of therapeutic agents
US6071923A (en) * 1994-09-16 2000-06-06 Bar-Ilan University Retinoyloxy aryl-substituted alkylene butyrates useful for the treatment of cancer and other proliferative diseases
US6080877A (en) * 1996-05-22 2000-06-27 Neuromedica, Inc. Taxanes
US6172046B1 (en) * 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
US6171828B1 (en) * 1996-03-04 2001-01-09 Suntory Limited Method for culturing microorganisms having a methanol metabolic pathway
US6191102B1 (en) * 1996-11-05 2001-02-20 Eli Lilly And Company Use of GLP-1 analogs and derivatives administered peripherally in regulation of obesity
US6193997B1 (en) * 1998-09-27 2001-02-27 Generex Pharmaceuticals Inc. Proteinic drug delivery system using membrane mimetics
US6201072B1 (en) * 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6214863B1 (en) * 1992-11-10 2001-04-10 Aventis Pharma S.A. Antitumor compositions containing taxane derivatives
US6214547B1 (en) * 1997-01-24 2001-04-10 Novo Nordisk A/S Synthetic leader peptide sequences
US6217893B1 (en) * 1997-04-18 2001-04-17 Pharma Biotech Sustained-release compositions and method for preparing same
US6221958B1 (en) * 1993-01-06 2001-04-24 Societe De Conseils De Recherches Et D'applications Scientifiques, Sas Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
US6221378B1 (en) * 1998-02-10 2001-04-24 Generex Pharmaceuticals Incorporated Mixed micellar delivery system and method of preparation
US20010002394A1 (en) * 1992-03-19 2001-05-31 Suad Efendic Use of a peptide
US6242479B1 (en) * 1998-12-17 2001-06-05 Loma Linda University Medical Center Use of γ-tocopherol and its oxidative metabolite LLU-α in the treatment of disease
US6340742B1 (en) * 1999-07-02 2002-01-22 Roche Diagnostics Gmbh Erythropoietin conjugates
US6346543B1 (en) * 1998-08-17 2002-02-12 Aventis Pharma S.A. Use of a taxoid to treat abnormal cell proliferation in the brain
US6348327B1 (en) * 1991-12-06 2002-02-19 Genentech, Inc. Non-endocrine animal host cells capable of expressing variant proinsulin and processing the same to form active, mature insulin and methods of culturing such cells
US6348192B1 (en) * 1999-05-11 2002-02-19 Bayer Corporation Interleukin-2 mutein expressed from mammalian cells
US20020037841A1 (en) * 2000-05-15 2002-03-28 Apollon Papadimitriou Erythropoietin composition
US20020048571A1 (en) * 1999-07-19 2002-04-25 Jeno Gyuris Chimeric polypeptides of serum albumin and uses related thereto
US6387365B1 (en) * 1995-05-19 2002-05-14 Schering Corporation Combination therapy for chronic hepatitis C infection
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US6569832B1 (en) * 1999-11-12 2003-05-27 Novo Nordisk A/S Inhibition of beta cell degeneration
US20040063635A1 (en) * 2002-07-01 2004-04-01 Zailin Yu Recombinant human albumin fusion proteins with long-lasting biological effects
US20040121426A1 (en) * 2002-12-18 2004-06-24 Palo Alto Research Center Incorporated Process for preparing albumin protein conjugated oligonucleotide probes

Family Cites Families (429)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3773919A (en) 1969-10-23 1973-11-20 Du Pont Polylactide-drug mixtures
US4179337A (en) 1973-07-20 1979-12-18 Davis Frank F Non-immunogenic polypeptides
DE2449885C3 (en) * 1974-10-21 1980-04-30 Biotest-Serum-Institut Gmbh, 6000 Frankfurt Process for the production of chemically modified, long-life hemoglobin preparations as well as the modified hemoglobin preparation produced by this process
US4002531A (en) 1976-01-22 1977-01-11 Pierce Chemical Company Modifying enzymes with polyethylene glycol and product produced thereby
US4363877B1 (en) 1977-09-23 1998-05-26 Univ California Recombinant dna transfer vectors
US4283489A (en) 1977-09-23 1981-08-11 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4407948A (en) 1977-09-23 1983-10-04 The Regents Of The University Of California Purification of nucleotide sequences suitable for expression in bacteria
US4366246A (en) 1977-11-08 1982-12-28 Genentech, Inc. Method for microbial polypeptide expression
US4263428A (en) 1978-03-24 1981-04-21 The Regents Of The University Of California Bis-anthracycline nucleic acid function inhibitors and improved method for administering the same
US5514567A (en) * 1979-01-30 1996-05-07 Juridical Foundation, Japanese Foundation For Cancer Research DNA and recombinant plasmid
US5326859A (en) 1979-10-30 1994-07-05 Juridical Foundation, Japanese Foundation For Cancer Research DNA and recombinant plasmid
US4342832A (en) 1979-07-05 1982-08-03 Genentech, Inc. Method of constructing a replicable cloning vehicle having quasi-synthetic genes
AU538665B2 (en) 1979-10-30 1984-08-23 Juridical Foundation, Japanese Foundation For Cancer Research Human interferon dna
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
FI86558C (en) 1980-01-08 1992-09-10 Biogen Inc DNA sequences, recombinant DNA molecules and methods for producing human-type interferon and selection of the DNA sequence
US4530901A (en) 1980-01-08 1985-07-23 Biogen N.V. Recombinant DNA molecules and their use in producing human interferon-like polypeptides
YU45104B (en) 1980-04-03 1992-03-10 Biogen Nv Process for producing recombinant dnk molecule, capable of inducting polypeptide expression in an one-cell host
DE3023787A1 (en) 1980-06-25 1982-01-21 Studiengesellschaft Kohle mbH, 4330 Mülheim METHOD FOR INCREASING THE INCORPORATION AND EXPRESSION OF GENETIC MATERIAL IN THE CORE OF INTACT CELLS BY LIPOSOME
US6610830B1 (en) 1980-07-01 2003-08-26 Hoffman-La Roche Inc. Microbial production of mature human leukocyte interferons
FR2490675B1 (en) 1980-09-25 1985-11-15 Genentech Inc MICROBIAL PRODUCTION OF HUMAN FIBROPLASTER INTERFERON
EP0052322B1 (en) 1980-11-10 1985-03-27 Gersonde, Klaus, Prof. Dr. Method of preparing lipid vesicles by ultrasonic treatment, the use of this method and apparatus for its application
US4456748A (en) 1981-02-23 1984-06-26 Genentech, Inc. Hybrid human leukocyte interferons
EP0070906B1 (en) 1981-02-04 1986-10-15 Juridical Foundation Japanese Foundation For Cancer Research Human interferon-beta gene
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
JPS57149228A (en) 1981-03-11 1982-09-14 Ajinomoto Co Inc Novel erythropoietin and its preparation
US4873191A (en) 1981-06-12 1989-10-10 Ohio University Genetic transformation of zygotes
US4792602A (en) 1981-06-19 1988-12-20 Cornell Research Foundation, Inc. Adaptors, and synthesis and cloning of proinsulin genes
US4474893A (en) 1981-07-01 1984-10-02 The University of Texas System Cancer Center Recombinant monoclonal antibodies
US4714681A (en) 1981-07-01 1987-12-22 The Board Of Reagents, The University Of Texas System Cancer Center Quadroma cells and trioma cells and methods for the production of same
US4485045A (en) 1981-07-06 1984-11-27 Research Corporation Synthetic phosphatidyl cholines useful in forming liposomes
IL66614A (en) 1981-08-28 1985-09-29 Genentech Inc Method of constructing a dna sequence encoding a polypeptide,microbial production of human serum albumin,and pharmaceutical compositions comprising it
AU561343B2 (en) 1981-10-19 1987-05-07 Genentech Inc. Human immune interferon by recombinant dna
EP0079739A3 (en) 1981-11-12 1984-08-08 The Upjohn Company Albumin-based nucleotides, their replication and use, and plasmids for use therein
EP0091527A3 (en) 1981-12-14 1984-07-25 The President And Fellows Of Harvard College Dna sequences, recombinant dna molecules and processes for producing human serum albumin-like polypeptides
JPS58118008A (en) 1982-01-06 1983-07-13 Nec Corp Data processor
IT1167610B (en) 1982-01-19 1987-05-13 Cetus Corp MULTICLASS HYBRID INTERFRERON, PHARMACEUTICAL COMPOSITION CONTAINING IT AND PRODUCTION PROCESS
DE3374837D1 (en) 1982-02-17 1988-01-21 Ciba Geigy Ag Lipids in the aqueous phase
US4775622A (en) 1982-03-08 1988-10-04 Genentech, Inc. Expression, processing and secretion of heterologous protein by yeast
US4670393A (en) * 1982-03-22 1987-06-02 Genentech, Inc. DNA vectors encoding a novel human growth hormone-variant protein
US4778879A (en) 1982-04-20 1988-10-18 Sloan-Kettering Institute For Cancer Research Highly purified human interleukin 2 and method
US6936694B1 (en) 1982-05-06 2005-08-30 Intermune, Inc. Manufacture and expression of large structural genes
DE3218121A1 (en) 1982-05-14 1983-11-17 Leskovar, Peter, Dr.-Ing., 8000 München Pharmaceutical compositions for tumour treatment
EP0102324A3 (en) 1982-07-29 1984-11-07 Ciba-Geigy Ag Lipids and surfactants in an aqueous medium
US4716111A (en) 1982-08-11 1987-12-29 Trustees Of Boston University Process for producing human antibodies
US4992271A (en) 1982-09-23 1991-02-12 Cetus Corporation Formulation for lipophilic IL-2 proteins
IE56026B1 (en) 1982-10-19 1991-03-27 Cetus Corp Cysteine-depleted muteins of biologically active proteins
US4588585A (en) * 1982-10-19 1986-05-13 Cetus Corporation Human recombinant cysteine depleted interferon-β muteins
US4966843A (en) * 1982-11-01 1990-10-30 Cetus Corporation Expression of interferon genes in Chinese hamster ovary cells
US4741900A (en) 1982-11-16 1988-05-03 Cytogen Corporation Antibody-metal ion complexes
EP0116201B1 (en) 1983-01-12 1992-04-22 Chiron Corporation Secretory expression in eukaryotes
NZ207394A (en) 1983-03-08 1987-03-06 Commw Serum Lab Commission Detecting or determining sequence of amino acids
GB8308235D0 (en) 1983-03-25 1983-05-05 Celltech Ltd Polypeptides
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
US4518584A (en) 1983-04-15 1985-05-21 Cetus Corporation Human recombinant interleukin-2 muteins
WO1984004330A1 (en) 1983-04-22 1984-11-08 Amgen Secretion of exogenous polypeptides from yeast
NZ207926A (en) 1983-04-25 1988-04-29 Genentech Inc Use of yeast #a#-factor to assist in expression of proteins heterologus to yeast
US4544545A (en) 1983-06-20 1985-10-01 Trustees University Of Massachusetts Liposomes containing modified cholesterol for organ targeting
US4576813A (en) 1983-07-05 1986-03-18 Monsanto Company Heat recovery from concentrated sulfuric acid
HUT35524A (en) 1983-08-02 1985-07-29 Hoechst Ag Process for preparing pharmaceutical compositions containing regulatory /regulative/ peptides providing for the retarded release of the active substance
JPS6087792A (en) 1983-09-23 1985-05-17 ジェネックス・コーポレイション Variant control region
EP0350973B1 (en) 1983-09-26 1997-11-05 Udo Dr. Ehrenfeld Means and product for the diagnosis and therapy of tumours and for the treatment of weaknesses of the cellular and humoral immune system
US4518564A (en) * 1983-10-03 1985-05-21 Jeneric Industries, Inc. Gallium and silver free, palladium based dental alloys for porcelain-fused-to-metal restorations
DE3474511D1 (en) 1983-11-01 1988-11-17 Terumo Corp Pharmaceutical composition containing urokinase
GB8334102D0 (en) 1983-12-21 1984-02-01 Searle & Co Interferons with cysteine pattern
US4703008A (en) 1983-12-13 1987-10-27 Kiren-Amgen, Inc. DNA sequences encoding erythropoietin
US4855238A (en) 1983-12-16 1989-08-08 Genentech, Inc. Recombinant gamma interferons having enhanced stability and methods therefor
GB8334261D0 (en) 1983-12-22 1984-02-01 Bass Plc Fermentation processes
JPS60136596A (en) 1983-12-26 1985-07-20 Suntory Ltd Peptide and diuretic comprising it as active ingredient
CA1213537A (en) 1984-05-01 1986-11-04 Canadian Patents And Development Limited - Societe Canadienne Des Brevets Et D'exploitation Limitee Polypeptide expression method
FR2564106B1 (en) * 1984-05-09 1988-04-22 Transgene Sa FACTOR IX EXPRESSION VECTORS, CELLS TRANSFORMED BY THESE VECTORS, AND PROCESS FOR THE PREPARATION OF FACTOR IX.
EP0172619A1 (en) 1984-06-20 1986-02-26 Takeda Chemical Industries, Ltd. Novel transformant and use thereof
US4736866A (en) 1984-06-22 1988-04-12 President And Fellows Of Harvard College Transgenic non-human mammals
US5908763A (en) 1984-07-06 1999-06-01 Novartis Corporation DNA encoding GM-CSF and a method of producing GM-CSF protein
JPS6147500A (en) 1984-08-15 1986-03-07 Res Dev Corp Of Japan Chimera monoclonal antibody and its preparation
EP0173494A3 (en) 1984-08-27 1987-11-25 The Board Of Trustees Of The Leland Stanford Junior University Chimeric receptors by dna splicing and expression
US5807715A (en) 1984-08-27 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods and transformed mammalian lymphocyte cells for producing functional antigen-binding protein including chimeric immunoglobulin
US4716217A (en) 1984-08-31 1987-12-29 University Patents, Inc. Hybrid lymphoblastoid-leukocyte human interferons
US4734491A (en) 1984-08-31 1988-03-29 University Patents, Inc. DNA sequences encoding hybrid lymphoblastoid-leukocyte human interferons
GB8422238D0 (en) 1984-09-03 1984-10-10 Neuberger M S Chimeric proteins
US4959314A (en) * 1984-11-09 1990-09-25 Cetus Corporation Cysteine-depleted muteins of biologically active proteins
US4897355A (en) 1985-01-07 1990-01-30 Syntex (U.S.A.) Inc. N[ω,(ω-1)-dialkyloxy]- and N-[ω,(ω-1)-dialkenyloxy]-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US5049386A (en) 1985-01-07 1991-09-17 Syntex (U.S.A.) Inc. N-ω,(ω-1)-dialkyloxy)- and N-(ω,(ω-1)-dialkenyloxy)Alk-1-YL-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4946787A (en) 1985-01-07 1990-08-07 Syntex (U.S.A.) Inc. N-(ω,(ω-1)-dialkyloxy)- and N-(ω,(ω-1)-dialkenyloxy)-alk-1-yl-N,N,N-tetrasubstituted ammonium lipids and uses therefor
US4970300A (en) * 1985-02-01 1990-11-13 New York University Modified factor VIII
GB8504099D0 (en) * 1985-02-18 1985-03-20 Wellcome Found Physiologically active substances
FR2579224B1 (en) * 1985-03-25 1987-05-22 Genetica PROCESS FOR THE MICROBIOLOGICAL PREPARATION OF HUMAN SERUM-ALBUMIN
US4751180A (en) * 1985-03-28 1988-06-14 Chiron Corporation Expression using fused genes providing for protein product
CA1260858A (en) 1985-03-28 1989-09-26 Lawrence S. Cousens Expression using fused genes providing for protein product
GB2183662B (en) 1985-04-01 1989-01-25 Celltech Ltd Transformed myeloma cell-line and a process for the expression of a gene coding for a eukaryotic polypeptide employing same
GR860984B (en) 1985-04-17 1986-08-18 Zymogenetics Inc Expression of factor vii and ix activities in mammalian cells
GB8510219D0 (en) 1985-04-22 1985-05-30 Bass Plc Isolation of fermentation products
AU5890286A (en) 1985-06-17 1986-12-24 Genex Corp. Cloned human serum albumin gene
US4980286A (en) 1985-07-05 1990-12-25 Whitehead Institute For Biomedical Research In vivo introduction and expression of foreign genetic material in epithelial cells
US4810643A (en) 1985-08-23 1989-03-07 Kirin- Amgen Inc. Production of pluripotent granulocyte colony-stimulating factor
US4676980A (en) 1985-09-23 1987-06-30 The United States Of America As Represented By The Secretary Of The Department Of Health And Human Services Target specific cross-linked heteroantibodies
JPS6296086A (en) 1985-10-21 1987-05-02 Agency Of Ind Science & Technol Composite plasmid
US5139941A (en) 1985-10-31 1992-08-18 University Of Florida Research Foundation, Inc. AAV transduction vectors
EP0247091B1 (en) 1985-11-01 1993-09-29 Xoma Corporation Modular assembly of antibody genes, antibodies prepared thereby and use
US5576195A (en) 1985-11-01 1996-11-19 Xoma Corporation Vectors with pectate lyase signal sequence
CA1295566C (en) 1987-07-21 1992-02-11 Robert T. Garvin Characterization and structure of genes for protease a and protease b from streptomyces griseus
GB8601597D0 (en) 1986-01-23 1986-02-26 Wilson R H Nucleotide sequences
FR2594846B1 (en) * 1986-02-21 1989-10-20 Genetica PROCESS FOR THE PREPARATION OF MATURE HUMAN ALBUMIN SERUM
EP0237019A3 (en) 1986-03-14 1988-03-09 Toray Industries, Inc. Interferon conjugate and production thereof using recombinant gene
IT1203758B (en) 1986-03-27 1989-02-23 Univ Roma CLONING AND EXPRESSION VECTORS OF HETEROLOGICAL GENES IN YEASTS AND YEASES TRANSFORMED WITH SUCH CARRIERS
US5225539A (en) 1986-03-27 1993-07-06 Medical Research Council Recombinant altered antibodies and methods of making altered antibodies
GB8607679D0 (en) 1986-03-27 1986-04-30 Winter G P Recombinant dna product
DK179286D0 (en) 1986-04-18 1986-04-18 Nordisk Gentofte INSULIN PREPARATION
US4765980A (en) * 1986-04-28 1988-08-23 International Minerals & Chemical Corp. Stabilized porcine growth hormone
US4859609A (en) 1986-04-30 1989-08-22 Genentech, Inc. Novel receptors for efficient determination of ligands and their antagonists or agonists
IT1204400B (en) * 1986-06-20 1989-03-01 Sclavo Spa PHARMACEUTICAL COMPOSITIONS CONTAINING A CALCITONIN
GB8615701D0 (en) 1986-06-27 1986-08-06 Delta Biotechnology Ltd Stable gene integration vector
IT1196484B (en) 1986-07-11 1988-11-16 Sclavo Spa YEAST EXPRESSION AND SECRETION VECTOR, USEFUL FOR THE PREPARATION OF HETEROLOGICAL PROTEINS
GR871067B (en) 1986-07-18 1987-11-19 Chugai Pharmaceutical Co Ltd Process for producing stable pharmaceutical preparation containing granulocyte colony stimulating factor
US4857467A (en) 1986-07-23 1989-08-15 Phillips Petroleum Company Carbon and energy source markers for transformation of strains of the genes Pichia
US4801575A (en) * 1986-07-30 1989-01-31 The Regents Of The University Of California Chimeric peptides for neuropeptide delivery through the blood-brain barrier
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
AU624789B2 (en) 1987-02-19 1992-06-25 Amgen, Inc. Purified platelet-derived growth factor and methods for purification thereof
EP0682110A1 (en) 1987-04-22 1995-11-15 Chiron Corporation Recombinant DNA encoding PDGF A-chain polypeptides
US5258498A (en) 1987-05-21 1993-11-02 Creative Biomolecules, Inc. Polypeptide linkers for production of biosynthetic proteins
US5489425A (en) 1987-06-24 1996-02-06 The Dow Chemical Company Functionalized polyamine chelants
US4994560A (en) 1987-06-24 1991-02-19 The Dow Chemical Company Functionalized polyamine chelants and radioactive rhodium complexes thereof for conjugation to antibodies
GB8717430D0 (en) 1987-07-23 1987-08-26 Celltech Ltd Recombinant dna product
JPH02476A (en) 1987-07-28 1990-01-05 Gist Brocades Nv Kluyveromyces as host cell
JP2627899B2 (en) 1987-08-19 1997-07-09 株式会社 ビタミン研究所 Production method of gene-encapsulated liposome
SE459586B (en) 1987-09-14 1989-07-17 Mta Szegedi Biolog Koezponti THE STRUCTURES CODING FOR AUTHENTIC HUMAN SERUM ALBUMIN AND PROCEDURES FOR ITS PREPARATION
NZ226414A (en) 1987-10-02 1992-07-28 Genentech Inc Cd4 peptide adhesion variants and their preparation and use
US5336603A (en) * 1987-10-02 1994-08-09 Genentech, Inc. CD4 adheson variants
GB8725529D0 (en) 1987-10-30 1987-12-02 Delta Biotechnology Ltd Polypeptides
JPH0811074B2 (en) 1987-10-30 1996-02-07 財団法人化学及血清療法研究所 Recombinant plasmid incorporating a gene encoding prealbumin and method for producing prealbumin using the same
PT89484B (en) 1988-01-22 1994-03-31 Gen Hospital Corp GENE CLONED CODES OF FUSAO PROTEINS IG-CD4 AND ITS UTILIZATION
JPH01215289A (en) 1988-02-22 1989-08-29 Toa Nenryo Kogyo Kk Production of normal human serum albumin a through gene recombination
US5066489A (en) 1988-03-28 1991-11-19 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5061488A (en) 1988-04-15 1991-10-29 The United States Of America As Represented Department Of Health & Human Services Flavone-8-acetic acid and interleukin-2 for cancer therapy
GB8809129D0 (en) 1988-04-18 1988-05-18 Celltech Ltd Recombinant dna methods vectors and host cells
IL89992A0 (en) 1988-04-25 1989-12-15 Phillips Petroleum Co Expression of human serum albumin in methylotrophic yeasts
NZ229700A (en) 1988-06-24 1993-01-27 Dow Chemical Co Tetraazacyclododecane derivatives containing a linker/spacer moiety capable of forming antibody conjugates; complexes with radionuclides and conjugates of such compounds and complexes with antibodies or antibody fragments
CN1033030C (en) 1988-06-24 1996-10-16 唐化学原料公司 Macrocyclic bifunctional chelants, complexes thereof and their antibody conjugates
US5756065A (en) 1988-06-24 1998-05-26 The Dow Chemical Company Macrocyclic tetraazacyclododecane conjugates and their use as diagnostic and therapeutic agents
US5274119A (en) 1988-07-01 1993-12-28 The Dow Chemical Company Vicinal diols
EP0387319B1 (en) * 1988-07-23 1996-03-06 Delta Biotechnology Limited Secretory leader sequences
US4925648A (en) 1988-07-29 1990-05-15 Immunomedics, Inc. Detection and treatment of infectious and inflammatory lesions
FR2649991B2 (en) 1988-08-05 1994-03-04 Rhone Poulenc Sante USE OF STABLE DERIVATIVES OF PLASMID PKD1 FOR THE EXPRESSION AND SECRETION OF HETEROLOGOUS PROTEINS IN YEASTS OF THE GENUS KLUYVEROMYCES
FR2635115B1 (en) 1988-08-05 1992-04-03 Rhone Poulenc Sante PROCESS FOR THE PREPARATION OF HUMAN ALBUMIN SERUM FROM YEAST
US5601819A (en) 1988-08-11 1997-02-11 The General Hospital Corporation Bispecific antibodies for selective immune regulation and for selective immune cell binding
EP0768377A1 (en) 1988-09-02 1997-04-16 Protein Engineering Corporation Generation and selection of recombinant varied binding proteins
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5260202A (en) 1988-09-07 1993-11-09 Delta Biotechnology Limited Fermentation method
JPH02227079A (en) 1988-10-06 1990-09-10 Tonen Corp Human serum albumin fragment
US5349052A (en) 1988-10-20 1994-09-20 Royal Free Hospital School Of Medicine Process for fractionating polyethylene glycol (PEG)-protein adducts and an adduct for PEG and granulocyte-macrophage colony stimulating factor
US5298243A (en) 1988-10-20 1994-03-29 Denki Kagaku Kogyo Kabushiki Kaisha Colony stimulating factor-gelatin conjugate
US5759802A (en) 1988-10-26 1998-06-02 Tonen Corporation Production of human serum alubumin A
JPH02117384A (en) 1988-10-26 1990-05-01 Tonen Corp Production of human serum albumin a by yeast host
US5696239A (en) 1988-10-31 1997-12-09 The Dow Chemical Company Conjugates possessing ortho ligating functionality and complexes thereof
US5342604A (en) 1988-10-31 1994-08-30 The Dow Chemical Company Complexes possessing ortho ligating functionality
US5256410A (en) 1988-12-01 1993-10-26 Schering Corporation Treatment of squamous cell carcinoma intralesionally with recombinant human alpha interferon
US4975271A (en) 1988-12-19 1990-12-04 Vipont Pharmaceutical, Inc. Muscosal delivery systems for treatment of periodontal disease
JP2989002B2 (en) 1988-12-22 1999-12-13 キリン―アムジエン・インコーポレーテツド Chemically modified granulocyte colony stimulating factor
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5198346A (en) 1989-01-06 1993-03-30 Protein Engineering Corp. Generation and selection of novel DNA-binding proteins and polypeptides
US5096815A (en) 1989-01-06 1992-03-17 Protein Engineering Corporation Generation and selection of novel dna-binding proteins and polypeptides
US5116964A (en) * 1989-02-23 1992-05-26 Genentech, Inc. Hybrid immunoglobulins
US5705363A (en) * 1989-03-02 1998-01-06 The Women's Research Institute Recombinant production of human interferon τ polypeptides and nucleic acids
US5703055A (en) 1989-03-21 1997-12-30 Wisconsin Alumni Research Foundation Generation of antibodies through lipid mediated DNA delivery
DK0465529T3 (en) 1989-03-21 1998-10-05 Vical Inc Expression of exogenous polynucleotide sequences in a vertebrate
US5693622A (en) 1989-03-21 1997-12-02 Vical Incorporated Expression of exogenous polynucleotide sequences cardiac muscle of a mammal
US5231020A (en) 1989-03-30 1993-07-27 Dna Plant Technology Corporation Genetic engineering of novel plant phenotypes
US5328470A (en) 1989-03-31 1994-07-12 The Regents Of The University Of Michigan Treatment of diseases by site-specific instillation of cells or site-specific transformation of cells and kits therefor
EP0395918A3 (en) 1989-04-13 1991-10-23 Vascular Laboratory, Inc. Plasminogen activator complex of pure pro-urokinase covalently bound by a disulfide bridge to human serum albumin
US5324844A (en) 1989-04-19 1994-06-28 Enzon, Inc. Active carbonates of polyalkylene oxides for modification of polypeptides
EP0394827A1 (en) 1989-04-26 1990-10-31 F. Hoffmann-La Roche Ag Chimaeric CD4-immunoglobulin polypeptides
GB8909916D0 (en) * 1989-04-29 1989-06-14 Delta Biotechnology Ltd Polypeptides
US5766883A (en) * 1989-04-29 1998-06-16 Delta Biotechnology Limited Polypeptides
ATE92107T1 (en) 1989-04-29 1993-08-15 Delta Biotechnology Ltd N-TERMINAL FRAGMENTS OF HUMAN SERUM ALBUMIN-CONTAINING FUSION PROTEINS.
US5332671A (en) 1989-05-12 1994-07-26 Genetech, Inc. Production of vascular endothelial cell growth factor and DNA encoding same
DE69024261T2 (en) 1989-05-24 1996-07-18 Merck & Co Inc Purification and characterization of a growth factor derived from a glioma
US5808003A (en) 1989-05-26 1998-09-15 Perimmune Holdings, Inc. Polyaminocarboxylate chelators
US5330971A (en) * 1989-06-09 1994-07-19 Gropep Pty. Ltd. Growth hormone fusion proteins, methods of production, and methods of treatment
JPH0327320A (en) 1989-06-26 1991-02-05 Ajinomoto Co Inc Human b cell differentiation factor pharmaceutical composition
DK0479909T3 (en) 1989-06-29 1997-04-07 Medarex Inc Bispecific reagents for AIDS treatment
US5413923A (en) 1989-07-25 1995-05-09 Cell Genesys, Inc. Homologous recombination for universal donor cells and chimeric mammalian hosts
FR2650598B1 (en) 1989-08-03 1994-06-03 Rhone Poulenc Sante DERIVATIVES OF ALBUMIN WITH THERAPEUTIC FUNCTION
CU22222A1 (en) 1989-08-03 1995-01-31 Cigb PROCEDURE FOR THE EXPRESSION OF HETEROLOGICAL PROTEINS PRODUCED IN A FUSION FORM IN ESCHERICHIA COLI, ITS USE, EXPRESSION VECTORS AND RECOMBINANT STRAINS
GB8927480D0 (en) 1989-12-05 1990-02-07 Delta Biotechnology Ltd Mutant fungal strain detection and new promoter
US5073627A (en) * 1989-08-22 1991-12-17 Immunex Corporation Fusion proteins comprising GM-CSF and IL-3
US5436146A (en) 1989-09-07 1995-07-25 The Trustees Of Princeton University Helper-free stocks of recombinant adeno-associated virus vectors
IE66494B1 (en) 1989-09-26 1996-01-10 Immunex Corp Granulocyte-colony stimulating factor receptors
ES2118066T3 (en) 1989-10-05 1998-09-16 Optein Inc SYNTHESIS AND ISOLATION, EXEMPTED FROM CELLS, FROM NEW GENES AND POLYPEPTIDES.
FR2653020B1 (en) 1989-10-17 1993-03-26 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR THE PREPARATION OF PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF LEUKEMIA.
GB8924021D0 (en) 1989-10-25 1989-12-13 Celltech Ltd Recombinant dna method and vectors for the use therein
AU625013B2 (en) 1989-11-03 1992-06-25 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5676954A (en) 1989-11-03 1997-10-14 Vanderbilt University Method of in vivo delivery of functioning foreign genes
US5173408A (en) 1989-11-13 1992-12-22 Lange Louis George Iii Mammalian pancreatic cholesterol esterase
US5580560A (en) * 1989-11-13 1996-12-03 Novo Nordisk A/S Modified factor VII/VIIa
GB8928874D0 (en) 1989-12-21 1990-02-28 Celltech Ltd Humanised antibodies
JPH03201987A (en) * 1989-12-29 1991-09-03 Tonen Corp Human serum albumin fragment
US5116944A (en) * 1989-12-29 1992-05-26 Neorx Corporation Conjugates having improved characteristics for in vivo administration
US5780225A (en) 1990-01-12 1998-07-14 Stratagene Method for generating libaries of antibody genes comprising amplification of diverse antibody DNAs and methods for using these libraries for the production of diverse antigen combining molecules
AU7247191A (en) 1990-01-11 1991-08-05 Molecular Affinities Corporation Production of antibodies using gene libraries
JP3068180B2 (en) 1990-01-12 2000-07-24 アブジェニックス インコーポレイテッド Generation of heterologous antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5545618A (en) 1990-01-24 1996-08-13 Buckley; Douglas I. GLP-1 analogs useful for diabetes treatment
DK0439442T3 (en) * 1990-01-25 1996-07-08 Univ Washington Factor X-LACI hybrid protein
JPH04211375A (en) 1990-02-05 1992-08-03 Ajinomoto Co Inc Synthetic gene and production of human serum albumin using the synthetic gene
US5747334A (en) 1990-02-15 1998-05-05 The University Of North Carolina At Chapel Hill Random peptide library
US5264618A (en) 1990-04-19 1993-11-23 Vical, Inc. Cationic lipids for intracellular delivery of biologically active molecules
US5427908A (en) 1990-05-01 1995-06-27 Affymax Technologies N.V. Recombinant library screening methods
AU7906691A (en) 1990-05-23 1991-12-10 United States of America, as represented by the Secretary, U.S. Department of Commerce, The Adeno-associated virus (aav)-based eucaryotic vectors
US5766897A (en) * 1990-06-21 1998-06-16 Incyte Pharmaceuticals, Inc. Cysteine-pegylated proteins
DK0538300T3 (en) 1990-07-10 1994-10-10 Boehringer Ingelheim Int O-Glycosylated IFN-alpha
GB9015198D0 (en) 1990-07-10 1990-08-29 Brien Caroline J O Binding substance
US5225341A (en) 1990-07-19 1993-07-06 The Regents Of The University Of California Biologically safe plant transformation system using a ds transposon
US5071872A (en) 1990-08-14 1991-12-10 The Ohio State University Research Foundation Method for improving interleukin-2 activity using aci-reductone compounds
US5625126A (en) 1990-08-29 1997-04-29 Genpharm International, Inc. Transgenic non-human animals for producing heterologous antibodies
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
KR100272077B1 (en) 1990-08-29 2000-11-15 젠팜인터내셔날,인코포레이티드 Transgenic non-human animals capable of producing heterologous antibodies
US5633425A (en) 1990-08-29 1997-05-27 Genpharm International, Inc. Transgenic non-human animals capable of producing heterologous antibodies
US5814318A (en) 1990-08-29 1998-09-29 Genpharm International Inc. Transgenic non-human animals for producing heterologous antibodies
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5391183A (en) * 1990-09-21 1995-02-21 Datascope Investment Corp Device and method sealing puncture wounds
IT1242149B (en) 1990-09-27 1994-02-16 Consiglio Nazionale Ricerche CODING NUCLEOTID SEQUENCE FOR A HUMAN PROTEIN WITH REGULATORY PROPERTIES OF ANGIOGENESIS
US5698426A (en) 1990-09-28 1997-12-16 Ixsys, Incorporated Surface expression libraries of heteromeric receptors
ES2129029T5 (en) 1990-10-05 2005-10-16 Celldex Therapeutics, Inc. DIRECT IMMUNOSTIMULATION WITH BISPECIFIC REAGENTS.
JPH07108232B2 (en) 1990-10-09 1995-11-22 エム・ディ・リサーチ株式会社 Method for producing peptide or protein
EP0557300B1 (en) 1990-10-29 1997-11-19 Chiron Corporation Bispecific antibodies, method of production, and uses thereof
FR2668368B1 (en) 1990-10-30 1995-03-10 Roussel Uclaf USE OF A POLYPEPTIDE HAVING THE ACTIVITY OF HUMAN INTERLEUKIN 2 FOR PREPARING A PHARMACEUTICAL COMPOSITION FOR THE TREATMENT OF MALIGNANT EPITHELIAL TUMORS.
US5173414A (en) 1990-10-30 1992-12-22 Applied Immune Sciences, Inc. Production of recombinant adeno-associated virus vectors
JPH0638771A (en) 1990-10-31 1994-02-15 Tonen Corp Expression of human protein disulfide isomerase gene and production of polypeptide by co-expression with the gene
US5353535A (en) 1990-11-05 1994-10-11 Plumly George W Floor type advertising apparatus
AU656353B2 (en) 1990-11-29 1995-02-02 Institut National De La Recherche Agronomique (Inra) New variants derived from interferons of type I, production process thereof and their applications
CA2095633C (en) 1990-12-03 2003-02-04 Lisa J. Garrard Enrichment method for variant proteins with altered binding properties
US5272080A (en) * 1991-02-19 1993-12-21 Pharmavene, Inc. Production of butyrylcholinesterase
US5833982A (en) 1991-02-28 1998-11-10 Zymogenetics, Inc. Modified factor VII
US5272070A (en) * 1991-03-08 1993-12-21 Board Of Regents, The University Of Texas System Method for the preparation of cell lines producing Man3 GlcNac 2 asparagine-linked gylcans and cell lines produced thereby
CA2062659A1 (en) 1991-03-12 1992-09-13 Yasutaka Igari Composition for sustained-release of erythropoietin
US5817471A (en) 1991-03-14 1998-10-06 The United States Of America As Represented By The Department Of Health And Human Services Trk tyrosine kinase receptor is the physiological receptor for nerve growth factor
DE69229454T2 (en) 1991-03-28 2000-01-05 Merck & Co Inc Subunit-C of the vascular endothelial cell growth factor
US5374617A (en) 1992-05-13 1994-12-20 Oklahoma Medical Research Foundation Treatment of bleeding with modified tissue factor in combination with FVIIa
WO1992018619A1 (en) 1991-04-10 1992-10-29 The Scripps Research Institute Heterodimeric receptor libraries using phagemids
EP0509841A3 (en) 1991-04-18 1993-08-18 Tonen Corporation Co-expression system of protein disulfide isomerase gene and useful polypeptide gene and process for producing the polypeptide using its system
CA2058820C (en) 1991-04-25 2003-07-15 Kotikanyad Sreekrishna Expression cassettes and vectors for the secretion of human serum albumin in pichia pastoris cells
EP0582595A1 (en) 1991-04-26 1994-02-16 Surface Active Limited Novel antibodies, and methods for their use
US5330901A (en) 1991-04-26 1994-07-19 Research Corporation Technologies, Inc. Expression of human serum albumin in Pichia pastoris
FR2676070B1 (en) 1991-04-30 1994-09-30 Rhone Poulenc Rorer Sa YEAST PROMOTER AND ITS USE.
US5646012A (en) * 1991-04-30 1997-07-08 Rhone-Poulenc Rorer S.A. Yeast promoter and use thereof
DE69233482T2 (en) 1991-05-17 2006-01-12 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
WO1992022324A1 (en) 1991-06-14 1992-12-23 Xoma Corporation Microbially-produced antibody fragments and their conjugates
FR2677996B1 (en) 1991-06-21 1993-08-27 Rhone Poulenc Rorer Sa CLONING AND / OR EXPRESSION VECTORS PREPARATION AND USE.
US5851795A (en) 1991-06-27 1998-12-22 Bristol-Myers Squibb Company Soluble CTLA4 molecules and uses thereof
US5844095A (en) * 1991-06-27 1998-12-01 Bristol-Myers Squibb Company CTLA4 Ig fusion proteins
US5223408A (en) * 1991-07-11 1993-06-29 Genentech, Inc. Method for making variant secreted proteins with altered properties
JPH05292972A (en) 1991-07-29 1993-11-09 Tonen Corp Improved yeast expression system
US5723719A (en) 1991-08-08 1998-03-03 Health Research Inc. Transgenic mouse as model for diseases involving dopaminergic dysfunction
DE4126968A1 (en) 1991-08-14 1993-02-18 Detlev Prof Dr Med Ganten Transgenic rats that contain at least one human gene in their genome that is involved in blood pressure regulation
ES2136092T3 (en) 1991-09-23 1999-11-16 Medical Res Council PROCEDURES FOR THE PRODUCTION OF HUMANIZED ANTIBODIES.
US6270989B1 (en) 1991-11-05 2001-08-07 Transkaryotic Therapies, Inc. Protein production and delivery
US5641670A (en) 1991-11-05 1997-06-24 Transkaryotic Therapies, Inc. Protein production and protein delivery
AU665599B2 (en) * 1991-11-08 1996-01-11 Hemosol Inc. Hemoglobins as drug delivery agents
US5786883A (en) * 1991-11-12 1998-07-28 Pilkington Barnes Hind, Inc. Annular mask contact lenses
ES2313867T3 (en) 1991-12-02 2009-03-16 Medical Research Council ANTI-AUTO ANTIBODY PRODUCTION OF ANTIBODY SEGMENT REPERTORIES EXPRESSED ON THE PAYMENT SURFACE.
US5540923A (en) * 1991-12-06 1996-07-30 Landsforeningen Til Kraeftens Bekaemplse Interferon proteins
US5428139A (en) 1991-12-10 1995-06-27 The Dow Chemical Company Bicyclopolyazamacrocyclophosphonic acid complexes for use as radiopharmaceuticals
ATE243746T1 (en) 1992-01-07 2003-07-15 Elan Pharm Inc TRANSGENIC ANIMAL MODELS FOR ALZHEIMER'S DISEASE
GB9200417D0 (en) * 1992-01-09 1992-02-26 Bagshawe Kenneth D Cytotoxic drug therapy
FR2686620B1 (en) * 1992-01-27 1995-06-23 Rhone Poulenc Rorer Sa HUMAN SERUM-ALBUMIN, PREPARATION AND USE.
FR2686899B1 (en) * 1992-01-31 1995-09-01 Rhone Poulenc Rorer Sa NOVEL BIOLOGICALLY ACTIVE POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686901A1 (en) 1992-01-31 1993-08-06 Rhone Poulenc Rorer Sa NOVEL ANTITHROMBOTIC POLYPEPTIDES, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
FR2686900B1 (en) * 1992-01-31 1995-07-21 Rhone Poulenc Rorer Sa NOVEL POLYPEPTIDES HAVING GRANULOCYTE COLONY STIMULATION ACTIVITY, THEIR PREPARATION AND PHARMACEUTICAL COMPOSITIONS CONTAINING THEM.
WO1993017715A1 (en) 1992-03-05 1993-09-16 Board Of Regents, The University Of Texas System Diagnostic and/or therapeutic agents, targeted to neovascular endothelial cells
US5230886A (en) 1992-03-18 1993-07-27 Trustees Of Boston University Tumor cell suppression
US5733743A (en) 1992-03-24 1998-03-31 Cambridge Antibody Technology Limited Methods for producing members of specific binding pairs
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
US5460954A (en) 1992-04-01 1995-10-24 Cheil Foods & Chemicals, Inc. Production of human proinsulin using a novel vector system
US5229109A (en) 1992-04-14 1993-07-20 Board Of Regents, The University Of Texas System Low toxicity interleukin-2 analogues for use in immunotherapy
US5775745A (en) 1992-06-09 1998-07-07 Hoppe Holding Ag Latch and lockset system
US5686268A (en) 1992-06-19 1997-11-11 Pfizer Inc. Fused proteins
US5505931A (en) 1993-03-04 1996-04-09 The Dow Chemical Company Acid cleavable compounds, their preparation and use as bifunctional acid-labile crosslinking agents
JP3269504B2 (en) 1992-07-08 2002-03-25 三菱ウェルファーマ株式会社 Method for producing human serum albumin
FR2694294B1 (en) * 1992-07-30 1994-09-09 Rhone Poulenc Rorer Sa Yeast promoter and its user.
AU686567C (en) 1992-07-31 2002-08-08 Genentech Inc. Human growth hormone aqueous formulation
US5602307A (en) 1992-08-12 1997-02-11 Baylor College Of Medicine Non-human animal having predefined allele of a cellular adhesion gene
DE4226971C2 (en) * 1992-08-14 1997-01-16 Widmar Prof Dr Tanner Modified fungal cells and processes for the production of recombinant products
US5639641A (en) 1992-09-09 1997-06-17 Immunogen Inc. Resurfacing of rodent antibodies
US5731490A (en) 1992-09-29 1998-03-24 The Ontario Cancer Institute Mutant mouse lacking the expression of interferon regulatory factor 1 (IRF-1)
WO1994008599A1 (en) * 1992-10-14 1994-04-28 The Regents Of The University Of Colorado Ion-pairing of drugs for improved efficacy and delivery
TW402639B (en) 1992-12-03 2000-08-21 Transkaryotic Therapies Inc Protein production and protein delivery
US5478745A (en) 1992-12-04 1995-12-26 University Of Pittsburgh Recombinant viral vector system
US5441050A (en) 1992-12-18 1995-08-15 Neoprobe Corporation Radiation responsive surgical instrument
US5489743A (en) 1993-01-19 1996-02-06 Amgen Inc. Transgenic animal models for thrombocytopenia
US5593972A (en) 1993-01-26 1997-01-14 The Wistar Institute Genetic immunization
FR2701953B1 (en) 1993-02-22 1995-05-24 Centre Nat Rech Scient Multi-VIP fusion protein and method for preparing recombinant VIP.
US5780021A (en) 1993-03-05 1998-07-14 Georgetown University Method for treating type 1 diabetes using α-interferon and/or β-i
US5869445A (en) * 1993-03-17 1999-02-09 University Of Washington Methods for eliciting or enhancing reactivity to HER-2/neu protein
ATE210458T1 (en) * 1993-05-21 2001-12-15 Zymogenetics Inc MODIFIED FACTOR VII TO INHIBIT VASCULAR RESTENOSIS AND THROMBOCYTE DEPOSITION
ATE301201T1 (en) 1993-06-07 2005-08-15 Vical Inc PLASMIDS USABLE FOR GENE THERAPY
US5621039A (en) * 1993-06-08 1997-04-15 Hallahan; Terrence W. Factor IX- polymeric conjugates
GB9317618D0 (en) 1993-08-24 1993-10-06 Royal Free Hosp School Med Polymer modifications
US5643575A (en) 1993-10-27 1997-07-01 Enzon, Inc. Non-antigenic branched polymer conjugates
US5459031A (en) 1993-11-05 1995-10-17 Amgen Inc. Methods for controlling sialic acid derivatives in recombinant glycoproteins
WO1995015982A2 (en) 1993-12-08 1995-06-15 Genzyme Corporation Process for generating specific antibodies
AU1736495A (en) 1994-01-31 1995-08-15 Trustees Of Boston University Polyclonal antibody libraries
US5834252A (en) 1995-04-18 1998-11-10 Glaxo Group Limited End-complementary polymerase reaction
US5605793A (en) 1994-02-17 1997-02-25 Affymax Technologies N.V. Methods for in vitro recombination
US5837458A (en) 1994-02-17 1998-11-17 Maxygen, Inc. Methods and compositions for cellular and metabolic engineering
US5932780A (en) 1994-02-28 1999-08-03 Yissum Research Development Company Of Hebrew University Of Jerusalem Transgenic non-human animal assay system for anti-cholinesterase substances
GB9404270D0 (en) 1994-03-05 1994-04-20 Delta Biotechnology Ltd Yeast strains and modified albumins
US6608182B1 (en) 1994-03-08 2003-08-19 Human Genome Sciences, Inc. Human vascular endothelial growth factor 2
US5646113A (en) 1994-04-07 1997-07-08 Genentech, Inc. Treatment of partial growth hormone insensitivity syndrome
FR2719593B1 (en) 1994-05-06 1996-05-31 Rhone Poulenc Rorer Sa New biologically active polypeptides, their preparation and pharmaceutical composition containing them.
GB9411356D0 (en) 1994-06-07 1994-07-27 Delta Biotechnology Ltd Yeast strains
US5516637A (en) 1994-06-10 1996-05-14 Dade International Inc. Method involving display of protein binding pairs on the surface of bacterial pili and bacteriophage
US5985660A (en) 1994-06-15 1999-11-16 Systemix, Inc. Method of identifying biological response modifiers involved in dendritic and/or lymphoid progenitor cell proliferation and/or differentiation
US5623054A (en) 1994-06-23 1997-04-22 The General Hospital Corporation Crucifer AFT proteins and uses thereof
JPH0851982A (en) 1994-08-11 1996-02-27 Asahi Glass Co Ltd Modified gene coding human serum albumen
US5574008A (en) 1994-08-30 1996-11-12 Eli Lilly And Company Biologically active fragments of glucagon-like insulinotropic peptide
FR2726471B1 (en) * 1994-11-07 1997-01-31 Pf Medicament PROCESS FOR IMPROVING THE IMMUNOGENICITY OF AN IMMUNOGENIC COMPOUND OR A HAPTENA AND APPLICATION TO THE PREPARATION OF VACCINES
WO1996014328A1 (en) 1994-11-07 1996-05-17 Human Genome Sciences, Inc. Tumor necrosis factor-gamma
FR2726576B1 (en) 1994-11-07 1997-01-31 Pf Medicament PRODUCTION OF HYDROPHOBIC PEPTIDE-LIKE PEPTIDES, RECOMBINANT PEPTIDE, CORRESPONDING DNA SEQUENCE
AT403167B (en) * 1994-11-14 1997-11-25 Immuno Ag SELECTION AND EXPRESSION OF FOREIGN PROTEINS BY MEANS OF A SELECTION-AMPLIFICATION SYSTEM
US5695750A (en) 1994-11-25 1997-12-09 The United States Of America As Represented By The Secretary Of The Army Compositions for use to deactivate organophosphates
DE69534265T2 (en) * 1994-12-12 2006-05-04 Beth Israel Deaconess Medical Center, Inc., Boston CHIMERIC CYTOKINS AND ITS USE
US5652224A (en) 1995-02-24 1997-07-29 The Trustees Of The University Of Pennsylvania Methods and compositions for gene therapy for the treatment of defects in lipoprotein metabolism
US5928939A (en) 1995-03-01 1999-07-27 Ludwig Institute For Cancer Research Vascular endothelial growth factor-b and dna coding therefor
US5837281A (en) * 1995-03-17 1998-11-17 Takeda Chemical Industries, Ltd. Stabilized interface for iontophoresis
JP2758154B2 (en) 1995-04-06 1998-05-28 エフ・ホフマン−ラ ロシユ アーゲー Liquid preparations containing interferon
AU2363695A (en) 1995-04-27 1996-11-18 Human Genome Sciences, Inc. Human tumor necrosis factor receptors
ATE390933T1 (en) 1995-04-27 2008-04-15 Amgen Fremont Inc HUMAN ANTIBODIES AGAINST IL-8 DERIVED FROM IMMUNIZED XENOMICES
AU2466895A (en) 1995-04-28 1996-11-18 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5480640A (en) 1995-05-02 1996-01-02 Schering Corporation Alpha interferon for treating prostate cancer
KR19990008335A (en) 1995-05-05 1999-01-25 로버트 에리치. 벤슨 Human Chemokine Beta-8, Chemokine Beta-1, and Macrophage Inflammatory Protein-4
US5728915A (en) 1995-05-08 1998-03-17 Children's Hospital, Inc. Transgenic mice which express simian SV 40 T-antigen under control of the retinoblastoma gene promoter
US5705151A (en) 1995-05-18 1998-01-06 National Jewish Center For Immunology & Respiratory Medicine Gene therapy for T cell regulation
US6001625A (en) 1995-05-19 1999-12-14 The United States Of America As Represented By The Secretary Of The Army Site-directed mutagenesis of esterases
US5804162A (en) 1995-06-07 1998-09-08 Alliance Pharmaceutical Corp. Gas emulsions stabilized with fluorinated ethers having low Ostwald coefficients
US5811097A (en) 1995-07-25 1998-09-22 The Regents Of The University Of California Blockade of T lymphocyte down-regulation associated with CTLA-4 signaling
AU7473096A (en) 1995-11-02 1997-05-22 Schering Corporation Continuous low-dose cytokine infusion therapy
US6048964A (en) * 1995-12-12 2000-04-11 Stryker Corporation Compositions and therapeutic methods using morphogenic proteins and stimulatory factors
GB9526733D0 (en) * 1995-12-30 1996-02-28 Delta Biotechnology Ltd Fusion proteins
US6110707A (en) * 1996-01-19 2000-08-29 Board Of Regents, The University Of Texas System Recombinant expression of proteins from secretory cell lines
US6087129A (en) * 1996-01-19 2000-07-11 Betagene, Inc. Recombinant expression of proteins from secretory cell lines
US6150337A (en) 1996-01-23 2000-11-21 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by Ribavirin in activated T-lymphocytes
JP2978435B2 (en) 1996-01-24 1999-11-15 チッソ株式会社 Method for producing acryloxypropyl silane
DE19639601A1 (en) 1996-02-28 1997-09-04 Bayer Ag Parapox viruses that contain foreign DNA, their production and their use in vaccines
WO1997033904A1 (en) 1996-03-12 1997-09-18 Human Genome Sciences, Inc. Death domain containing receptors
AU5711196A (en) 1996-03-14 1997-10-01 Human Genome Sciences, Inc. Apoptosis inducing molecule i
AU728146B2 (en) 1996-03-14 2001-01-04 Immune Response Corporation, The Targeted delivery of genes encoding interferon
CA2248136A1 (en) * 1996-03-21 1997-09-25 Human Genome Sciences, Inc. Human endometrial specific steroid-binding factor i, ii and iii
EP0904278A4 (en) 1996-03-22 1999-09-15 Human Genome Sciences Inc Apoptosis inducing molecule ii
US6204022B1 (en) * 1996-04-12 2001-03-20 Pepgen Corporation And University Of Florida Low-toxicity human interferon-alpha analogs
US6497878B1 (en) * 1996-04-23 2002-12-24 Chugai Seiyaku Kabushiki Kaisha Treatment of cerebral disorders by inhibition of IL-8 binding to receptor
US6300065B1 (en) * 1996-05-31 2001-10-09 Board Of Trustees Of The University Of Illinois Yeast cell surface display of proteins and uses thereof
US6277618B1 (en) * 1996-06-06 2001-08-21 Roche Diagnostics Gmbh Recombinant blood-coagulation proteases
US6828426B1 (en) 1996-07-15 2004-12-07 Chugai Seiyaku Kabushiki Kaisha VEGF-like factor
ATE397660T1 (en) 1996-08-16 2008-06-15 Schering Corp MAMMAL CELL SURFACE ANTIGEN AND RELATED REAGENTS
EP2025682A3 (en) 1996-08-16 2009-06-17 Human Genome Sciences, Inc. Human endokine alpha
EP2107109B1 (en) 1996-08-23 2012-06-20 Vegenics Pty Ltd Recombinant vascular endothelial cell growth factor D (VEGF-D)
EP0961832A4 (en) * 1996-09-18 2003-02-26 Human Genome Sciences Inc Human tumor necrosis factor receptor-like genes
US6225290B1 (en) 1996-09-19 2001-05-01 The Regents Of The University Of California Systemic gene therapy by intestinal cell transformation
US5994112A (en) 1996-10-09 1999-11-30 Incyte Pharmaceuticals, Inc. Human protein tyrosine kinase
US5916771A (en) 1996-10-11 1999-06-29 Abgenix, Inc. Production of a multimeric protein by cell fusion method
AU4988697A (en) 1996-10-24 1998-05-15 Vion Pharmaceuticals, Inc. Monophosphate prodrugs of beta-l-fd4c and beta-l-fddc as potent antiviral agents
KR20000052796A (en) 1996-10-25 2000-08-25 벤슨 로버트 에이치. Neutrokine alpha
JPH10134761A (en) 1996-10-30 1998-05-22 Ebara Corp Ion implantation device and method
GB9623205D0 (en) * 1996-11-07 1997-01-08 Eurand Int Novel pharmaceutical compositions
KR100643058B1 (en) 1996-12-03 2006-11-13 아브게닉스, 인크. Transgenic mammals having human ig loci including plural vh and vk regions and antibodies produced therefrom
US5833994A (en) 1997-01-08 1998-11-10 Paracelsian, Inc. Use of the AH receptor and AH receptor ligands to treat or prevent cytopathicity of viral infection
ATE321855T1 (en) 1997-01-14 2006-04-15 Human Genome Sciences Inc TUMOR NECROSIS FACTOR RECEPTOR 5
ES2284199T5 (en) 1997-01-28 2011-11-14 Human Genome Sciences, Inc. RECEIVER 4 CONTAINING DEATH DOMAIN (DR4: DEATH RECEIVER 4), MEMBER OF THE TNF RECEPTORS SUPERFAMILY AND TRAIL UNION (APO-2L).
JP2001508783A (en) 1997-01-29 2001-07-03 ポリマスク・ファーマシューティカルズ・パブリック・リミテッド・カンパニー PEGylation method
GB2324529A (en) 1997-02-21 1998-10-28 Merck & Co Inc A combinatorial library based on a tetrapeptide substituted with aminomethylcoumarin for characterizing proteases
WO1999019339A1 (en) * 1997-10-09 1999-04-22 Human Genome Sciences, Inc. 53 human secreted proteins
US7026447B2 (en) * 1997-10-09 2006-04-11 Human Genome Sciences, Inc. 53 human secreted proteins
DK0970126T3 (en) 1997-04-14 2001-08-13 Micromet Ag Hitherto unknown method for the preparation of anti-human antigen receptors and uses thereof
US6235883B1 (en) 1997-05-05 2001-05-22 Abgenix, Inc. Human monoclonal antibodies to epidermal growth factor receptor
WO1998050347A1 (en) 1997-05-05 1998-11-12 The Regents Of The University Of California Naphthols useful in antiviral methods
EP1003767A4 (en) 1997-05-30 2003-01-02 Human Genome Sciences Inc Human tumor necrosis factor receptor tr10
US6071743A (en) 1997-06-02 2000-06-06 Subsidiary No. 3, Inc. Compositions and methods for inhibiting human immunodeficiency virus infection by down-regulating human cellular genes
AU7830398A (en) 1997-06-11 1998-12-30 Human Genome Sciences, Inc. Human tumor necrosis factor receptor tr9
GB9713412D0 (en) 1997-06-26 1997-08-27 Delta Biotechnology Ltd Improved protein expression strains
US5858719A (en) 1997-07-17 1999-01-12 Incyte Pharmaceuticals, Inc. Polynucleotides encoding human ATP binding-cassette transport protein and methods of use
US6472373B1 (en) 1997-09-21 2002-10-29 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in antiviral treatment naive patients having chronic hepatitis C infection
DK1136075T3 (en) 1997-09-21 2003-04-28 Schering Corp Combination therapy for eradication of detectable HCV RNA in patients with chronic hepatitis C infection
EP1042343B1 (en) 1997-11-03 2008-01-23 Human Genome Sciences, Inc. Vegi, an inhibitor of angiogenesis and tumor growth
DE69824039T2 (en) * 1997-12-08 2005-08-18 Lexigen Pharmaceuticals Corp., Lexington HETERODIMARY FUSION PROTEINS FOR THE USE OF TARGETED IMMUNOTHERAPY AND GENERAL IMMUNE REGION
DE19813802A1 (en) 1998-03-27 1999-11-11 Retro Tech Gmbh Anti-viral effects of propolis through inhibition of viral nucleic acid polymerases
GB9806631D0 (en) 1998-03-28 1998-05-27 Safeglass Europ Limited Safetyglass
US6251868B1 (en) 1998-04-30 2001-06-26 Teijin Limited Method for treating a human immunodeficiency virus infection
PT956861E (en) 1998-05-15 2002-08-30 Schering Corp COMBINED THERAPY UNDERSTANDING RIBAVIRIN AND ALPHA INTERFERENCE IN PATIENTS WITHOUT PREVIOUS ANTIVIRAL TREATMENT HAVING CHRONIC INFECTION BY HEPATITIS C VIRUS
CN1119352C (en) 1998-05-15 2003-08-27 中国科学院上海生物化学研究所 Express and purification of human serum albumin in pichia
US5970300A (en) * 1998-06-01 1999-10-19 Xerox Corporation Apparatus for applying scents to paper in a printer/copier
EP1088084B1 (en) * 1998-06-15 2006-09-13 GTC Biotherapeutics, Inc. Erythropoietin analog-human serum albumin fusion protein
CN1105727C (en) 1998-06-17 2003-04-16 上海海济医药生物工程有限公司 Process for preparing recombined human serum albumin
BR9911457A (en) 1998-06-24 2001-12-11 Univ Emory Use of 3'-azide-2 ', 3'-dideoxyuridine in combination with additional anti-HIV drugs for the manufacture of a drug for the treatment of HIV
GB9817084D0 (en) 1998-08-06 1998-10-07 Wood Christopher B A method for promoting extra-heptic production of proteins for the correction of hypoalbuminaemia,anaemia,thrombocytopenia and/or coagulation disorders
GB9902000D0 (en) 1999-01-30 1999-03-17 Delta Biotechnology Ltd Process
US7109292B2 (en) * 1999-03-08 2006-09-19 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
KR20020018197A (en) 1999-05-19 2002-03-07 추후보정 EXPRESSION AND EXPORT OF INTERFERON-ALPHA PROTEINS AS Fc FUSION PROTEINS
US7144574B2 (en) * 1999-08-27 2006-12-05 Maxygen Aps Interferon β variants and conjugates
CA2388432A1 (en) 1999-10-21 2001-04-26 Monsanto Company Post-translational modification of recombinant proteins produced in plants
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
JP2004506407A (en) 2000-04-12 2004-03-04 ヒューマン ゲノム サイエンシズ インコーポレイテッド Albumin fusion protein
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US7101561B2 (en) * 2000-12-01 2006-09-05 Innogenetics N.V. Purified hepatitis C virus envelope proteins for diagnostic and therapeutic use
SK288088B6 (en) 2000-12-07 2013-06-03 Eli Lilly And Company GLP-1 fusion proteins
US7070973B2 (en) 2000-12-26 2006-07-04 Board Of Regents Of The University Of Nebraska Butyrylcholinesterase variants and methods of use
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20050054051A1 (en) 2001-04-12 2005-03-10 Human Genome Sciences, Inc. Albumin fusion proteins
US20030143191A1 (en) * 2001-05-25 2003-07-31 Adam Bell Chemokine beta-1 fusion proteins
WO2003013573A1 (en) 2001-08-10 2003-02-20 Epix Medical, Inc. Polypeptide conjugates with extended circulating half-lives
WO2003030821A2 (en) 2001-10-05 2003-04-17 Human Genome Sciences, Inc. Albumin fusion proteins
WO2003031464A2 (en) * 2001-10-10 2003-04-17 Neose Technologies, Inc. Remodeling and glycoconjugation of peptides
WO2005077042A2 (en) 2004-02-09 2005-08-25 Human Genome Sciences, Inc. Albumin fusion proteins
US20080194481A1 (en) 2001-12-21 2008-08-14 Human Genome Sciences, Inc. Albumin Fusion Proteins
US20080167238A1 (en) 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
KR101271635B1 (en) * 2001-12-21 2013-06-12 휴먼 게놈 사이언시즈, 인코포레이티드 Albumin fusion proteins
AU2002364586A1 (en) 2001-12-21 2003-07-30 Delta Biotechnology Limited Albumin fusion proteins
AU2002353374A1 (en) 2001-12-21 2003-07-09 Nexia Biotechnologies, Inc. Production of butyrylcholinesterases in transgenic mammals
WO2003059934A2 (en) 2001-12-21 2003-07-24 Human Genome Sciences, Inc. Albumin fusion proteins
MXPA04012496A (en) * 2002-06-21 2005-09-12 Novo Nordisk Healthcare Ag Pegylated factor vii glycoforms.
CA2513213C (en) 2003-01-22 2013-07-30 Human Genome Sciences, Inc. Albumin fusion proteins
EP2266630B1 (en) * 2003-02-27 2013-12-25 Baxter International Inc. Device for calibration in a method for the validatable inactivation of pathogens in a biological fluid by irradiation
US20060051859A1 (en) * 2004-09-09 2006-03-09 Yan Fu Long acting human interferon analogs
US7436410B2 (en) * 2005-04-01 2008-10-14 Seiko Epson Corporation System and method for programming a controller
CA2618476A1 (en) 2005-08-12 2007-02-22 Human Genome Sciences, Inc. Albumin fusion proteins
US7438904B1 (en) 2005-10-04 2008-10-21 University Of Kentucky Research Foundation High-activity mutants of butyrylcholinesterase for cocaine hydrolysis and method of generating the same
CA2562249A1 (en) 2005-10-20 2007-04-20 University Of Ottawa Heart Institute Anf analogue
EP1816201A1 (en) * 2006-02-06 2007-08-08 CSL Behring GmbH Modified coagulation factor VIIa with extended half-life
AU2007258609B2 (en) 2006-06-07 2013-01-24 Human Genome Sciences, Inc. Albumin fusion proteins
AU2012267484B2 (en) * 2011-06-10 2017-03-23 Bioverativ Therapeutics Inc. Pro-coagulant compounds and methods of use thereof

Patent Citations (99)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5905143A (en) * 1919-09-12 1999-05-18 Delta Biotechnology Limited Purification of proteins
US4264731A (en) * 1977-05-27 1981-04-28 The Regents Of The University Of California DNA Joining method
US4440859A (en) * 1977-05-27 1984-04-03 The Regents Of The University Of California Method for producing recombinant bacterial plasmids containing the coding sequences of higher organisms
US4447538A (en) * 1978-04-19 1984-05-08 Regents Of The University Of California Microorganism containing gene for human chorionic somatomammotropin
US4652525A (en) * 1978-04-19 1987-03-24 The Regents Of The University Of California Recombinant bacterial plasmids containing the coding sequences of insulin genes
US5856123A (en) * 1981-02-25 1999-01-05 Washington Research Foundation Expression of polypeptides in yeast
US5618676A (en) * 1981-02-25 1997-04-08 Genentech, Inc. Expression of polypeptides in yeast
US4450103A (en) * 1982-03-01 1984-05-22 Cetus Corporation Process for recovering human IFN-β from a transformed microorganism
US4499188A (en) * 1982-05-05 1985-02-12 Cetus Corporation Bacterial production of heterologous polypeptides under the control of a repressible promoter-operator
US4462940A (en) * 1982-09-23 1984-07-31 Cetus Corporation Process for the recovery of human β-interferon-like polypeptides
US4840934A (en) * 1983-01-25 1989-06-20 Eleanor Roosevelt Institute For Cancer Research, Inc. Therapeutic method using T cell growth factor
US5015575A (en) * 1983-04-07 1991-05-14 Chiron Corporation Hybrid DNA synthesis of insulin
US4914026A (en) * 1983-04-07 1990-04-03 Chiron Corporation Alpha factor leader sequence directed secretion of insulin
US5010003A (en) * 1983-04-25 1991-04-23 Genentech, Inc. Use of yeast homologous signals to secrete heterologous proteins
US4492684A (en) * 1983-06-08 1985-01-08 Connaught Laboratories Limited Slow release injectable insulin composition
US5618698A (en) * 1983-12-13 1997-04-08 Kirin-Amgen, Inc. Production of erythropoietin
US4925919A (en) * 1984-04-25 1990-05-15 Roland Mertelsmann Purified interleukin 2
US4908433A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Uses of interleukin-2
US4908434A (en) * 1984-04-25 1990-03-13 Sloan-Kettering Institute For Cancer Research Process for preparing purified interleukin-2
US4916212A (en) * 1984-05-30 1990-04-10 Novo Industri A/S DNA-sequence encoding biosynthetic insulin precursors and process for preparing the insulin precursors and human insulin
US5102872A (en) * 1985-09-20 1992-04-07 Cetus Corporation Controlled-release formulations of interleukin-2
US5641663A (en) * 1985-11-06 1997-06-24 Cangene Corporation Expression system for the secretion of bioactive human granulocyte macrophage colony stimulating factor (GM-CSF) and other heterologous proteins from steptomyces
US5028422A (en) * 1986-05-27 1991-07-02 Schering Corporation Treatment of basal cell carcinoma intralesionally with recombinant human alpha interferon
US5002764A (en) * 1986-08-12 1991-03-26 Schering Corporation Treatment of actinic keratoses with alpha2 interferon
US5739007A (en) * 1986-08-29 1998-04-14 Delta Biotechnology Limited Hybrid GAL10/pgk yeast promoter
US4929442A (en) * 1986-09-26 1990-05-29 Exovir, Inc. Compositions suitable for human topical application including a growth factor and/or related materials
US5508031A (en) * 1986-11-21 1996-04-16 Cetus Oncology Corporation Method for treating biological damage using a free-radial scavenger and interleukin-2
US4835260A (en) * 1987-03-20 1989-05-30 Genetics Institute, Inc. Erythropoietin composition
US5637504A (en) * 1987-04-09 1997-06-10 Delta Biotechnology Limited Stable yeast 2 μm vector
US5503993A (en) * 1987-12-02 1996-04-02 The Green Cross Corporation Method of preparing foreign protein in yeast, recombinant DNA, transformant
US5409815A (en) * 1988-02-16 1995-04-25 The Green Cross Corporation DNA's encoding signal peptides
US4999339A (en) * 1988-03-28 1991-03-12 Cetus Corporation Combination therapy of IL-2 and DTIC for the treatment of melanoma
US5096885A (en) * 1988-04-15 1992-03-17 Genentech, Inc. Human growth hormone formulation
US5763394A (en) * 1988-04-15 1998-06-09 Genentech, Inc. Human growth hormone aqueous formulation
US5096707A (en) * 1988-04-15 1992-03-17 The United States Of America As Represented By The Department Of Health And Human Services Flavone-8-acetic acid and interleukin-2 in a method of treating certain cancers
US5126129A (en) * 1988-05-23 1992-06-30 The Government Of The United States Of America As Represented By The Secretary Of The Department Of Health & Human Services Cancer therapy using interleukin-2 and flavone compounds
US5395922A (en) * 1989-03-03 1995-03-07 Novo Nordisk A/S Yeast processing system
US5128126A (en) * 1989-04-11 1992-07-07 Boehringer Ingelheim International Gmbh Use of pharmaceutical compositions containing at least one cytokine for the systemic treatment of preneoplastic lesions
US5106954A (en) * 1989-07-26 1992-04-21 Behringwerke Aktiengesellschaft Erythropoietin (epo) peptides
US6063373A (en) * 1989-09-19 2000-05-16 Maxim Pharmaceuticals, Inc. Enhanced activation of NK cells using an NK cell activator and a hydrogen peroxide scavenger or inhibitor
US6069135A (en) * 1989-09-21 2000-05-30 Hyal Pharmaceutical Corporation Use of hyaluronic acid or its derivatives to enhance delivery of therapeutic agents
US5208018A (en) * 1990-03-19 1993-05-04 Brigham And Women's Hospital Treatment of cachexia with interleukin 2
US5219565A (en) * 1990-04-17 1993-06-15 Roussel Uclaf Treatment of primary cancers of the pleura
US5322930A (en) * 1990-08-07 1994-06-21 Scios Nova Inc. Expression of recombinant polypeptides with improved purification
US5625041A (en) * 1990-09-12 1997-04-29 Delta Biotechnology Limited Purification of proteins
US5304473A (en) * 1991-06-11 1994-04-19 Eli Lilly And Company A-C-B proinsulin, method of manufacturing and using same, and intermediates in insulin production
US6048724A (en) * 1991-11-05 2000-04-11 Transkaryotic Therapies Inc. Method of producing clonal cell strains which express exogenous DNA encoding glucagon-like peptide 1
US6348327B1 (en) * 1991-12-06 2002-02-19 Genentech, Inc. Non-endocrine animal host cells capable of expressing variant proinsulin and processing the same to form active, mature insulin and methods of culturing such cells
US20010002394A1 (en) * 1992-03-19 2001-05-31 Suad Efendic Use of a peptide
US6034221A (en) * 1992-09-23 2000-03-07 Delta Biotechnology Limited High purity albumin
US5728553A (en) * 1992-09-23 1998-03-17 Delta Biotechnology Limited High purity albumin and method of producing
US6239167B1 (en) * 1992-11-10 2001-05-29 Aventis Pharma S.A. Antitumor compositions containing taxane derivatives
US6214863B1 (en) * 1992-11-10 2001-04-10 Aventis Pharma S.A. Antitumor compositions containing taxane derivatives
US6221958B1 (en) * 1993-01-06 2001-04-24 Societe De Conseils De Recherches Et D'applications Scientifiques, Sas Ionic molecular conjugates of biodegradable polyesters and bioactive polypeptides
US5602232A (en) * 1993-02-25 1997-02-11 Schering Corporation Method for producing metal-interferon-α crystals
US5726038A (en) * 1993-07-08 1998-03-10 Novo Nordisk A/S DNA construct encoding the YAP3 signal peptide
US5521086A (en) * 1993-09-16 1996-05-28 Cephalon, Inc. Secretion sequence for the production of a heterologous protein in yeast
US5629286A (en) * 1994-03-31 1997-05-13 Brewitt; Barbara Homeopathic dilutions of growth factors
US5639642A (en) * 1994-06-16 1997-06-17 Novo Nordisk A/S Synthetic leader peptide sequences
US6071923A (en) * 1994-09-16 2000-06-06 Bar-Ilan University Retinoyloxy aryl-substituted alkylene butyrates useful for the treatment of cancer and other proliferative diseases
US5512549A (en) * 1994-10-18 1996-04-30 Eli Lilly And Company Glucagon-like insulinotropic peptide analogs, compositions, and methods of use
US6387365B1 (en) * 1995-05-19 2002-05-14 Schering Corporation Combination therapy for chronic hepatitis C infection
US5741815A (en) * 1995-06-02 1998-04-21 Lai; Ching-San Methods for in vivo reduction of nitric oxide levels and compositions useful therefor
US5861406A (en) * 1995-07-21 1999-01-19 Constantia Gruppe Treatment and prevention of neoplasms with salts of aminoimidazole carboxamide and 5-amino or substituted amino 1,2,3-triazoles
US5728707A (en) * 1995-07-21 1998-03-17 Constantia Gruppe Treatment and prevention of primary and metastatic neoplasms with salts of aminoimidazole carboxamide
US5766620A (en) * 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5863555A (en) * 1995-10-23 1999-01-26 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US6063772A (en) * 1996-01-23 2000-05-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
US5767097A (en) * 1996-01-23 1998-06-16 Icn Pharmaceuticals, Inc. Specific modulation of Th1/Th2 cytokine expression by ribavirin in activated T-lymphocytes
US6045788A (en) * 1996-02-28 2000-04-04 Cornell Research Foundation, Inc. Method of stimulation of immune response with low doses of IL-2
US5912229A (en) * 1996-03-01 1999-06-15 Novo Nordisk Als Use of a pharmaceutical composition comprising an appetite-suppressing peptide
US6171828B1 (en) * 1996-03-04 2001-01-09 Suntory Limited Method for culturing microorganisms having a methanol metabolic pathway
US6080877A (en) * 1996-05-22 2000-06-27 Neuromedica, Inc. Taxanes
US6054489A (en) * 1996-10-15 2000-04-25 Loyola University Of Chicago Method for the enhancement of lymphocyte activity against tumors
US5908830A (en) * 1996-10-31 1999-06-01 Merck & Co., Inc. Combination therapy for the treatment of diabetes and obesity
US6191102B1 (en) * 1996-11-05 2001-02-20 Eli Lilly And Company Use of GLP-1 analogs and derivatives administered peripherally in regulation of obesity
US6583111B1 (en) * 1996-11-05 2003-06-24 Eli Lilly And Company Use of GLP-1 analogs and derivative adminstered peripherally in regulation of obesity
US6214547B1 (en) * 1997-01-24 2001-04-10 Novo Nordisk A/S Synthetic leader peptide sequences
US6030961A (en) * 1997-03-11 2000-02-29 Bar-Ilan Research & Development Co., Ltd. Oxyalkylene phosphate compounds and uses thereof
US6217893B1 (en) * 1997-04-18 2001-04-17 Pharma Biotech Sustained-release compositions and method for preparing same
US6172046B1 (en) * 1997-09-21 2001-01-09 Schering Corporation Combination therapy for eradicating detectable HCV-RNA in patients having chronic Hepatitis C infection
US6201072B1 (en) * 1997-10-03 2001-03-13 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co- glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6031004A (en) * 1997-12-08 2000-02-29 Bristol-Myers Squibb Company Salts of metformin and method
US6231882B1 (en) * 1998-02-10 2001-05-15 Generex Pharmaceuticals Inc. Mixed micellar delivery system and method of preparation
US6221378B1 (en) * 1998-02-10 2001-04-24 Generex Pharmaceuticals Incorporated Mixed micellar delivery system and method of preparation
US6017545A (en) * 1998-02-10 2000-01-25 Modi; Pankaj Mixed micellar delivery system and method of preparation
US6346543B1 (en) * 1998-08-17 2002-02-12 Aventis Pharma S.A. Use of a taxoid to treat abnormal cell proliferation in the brain
US6193997B1 (en) * 1998-09-27 2001-02-27 Generex Pharmaceuticals Inc. Proteinic drug delivery system using membrane mimetics
US6242479B1 (en) * 1998-12-17 2001-06-05 Loma Linda University Medical Center Use of γ-tocopherol and its oxidative metabolite LLU-α in the treatment of disease
US6348192B1 (en) * 1999-05-11 2002-02-19 Bayer Corporation Interleukin-2 mutein expressed from mammalian cells
US6340742B1 (en) * 1999-07-02 2002-01-22 Roche Diagnostics Gmbh Erythropoietin conjugates
US20020048571A1 (en) * 1999-07-19 2002-04-25 Jeno Gyuris Chimeric polypeptides of serum albumin and uses related thereto
US6514500B1 (en) * 1999-10-15 2003-02-04 Conjuchem, Inc. Long lasting synthetic glucagon like peptide {GLP-!}
US20030108567A1 (en) * 1999-10-15 2003-06-12 BRIDON Dominique P. Long lasting synthetic glucagon-like peptide {GLP-1}
US20030108568A1 (en) * 1999-10-15 2003-06-12 BRIDON Dominique P. Long lasting synthetic glucagon-like peptide {GLP-1}
US6569832B1 (en) * 1999-11-12 2003-05-27 Novo Nordisk A/S Inhibition of beta cell degeneration
US20020037841A1 (en) * 2000-05-15 2002-03-28 Apollon Papadimitriou Erythropoietin composition
US20040063635A1 (en) * 2002-07-01 2004-04-01 Zailin Yu Recombinant human albumin fusion proteins with long-lasting biological effects
US20040121426A1 (en) * 2002-12-18 2004-06-24 Palo Alto Research Center Incorporated Process for preparing albumin protein conjugated oligonucleotide probes

Cited By (128)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060105429A1 (en) * 1992-01-31 2006-05-18 Aventis Behring L.L.C. Fusion polypeptides of human serum albumin and a therapeutically active polypeptide
US20050239167A1 (en) * 1992-01-31 2005-10-27 Aventis Behring L.L.C. Fusion polypeptides of human serum albumin and a therapeutically active polypeptide
US8642542B2 (en) 1995-12-30 2014-02-04 Novozymes Biopharma Dk A/S Recombinant fusion proteins to growth hormone and serum albumin
US7550432B2 (en) 1995-12-30 2009-06-23 Novozymes Biopharma Uk Limited Recombinant fusion proteins to growth hormone and serum albumin
US20100261650A1 (en) * 1995-12-30 2010-10-14 Human Genome Sciences, Inc. Recombinant fusion proteins to growth hormone and serum albumin
US20030077732A1 (en) * 1997-09-18 2003-04-24 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030027264A1 (en) * 1997-09-18 2003-02-06 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US20030166113A1 (en) * 1997-09-18 2003-09-04 Genentech, Inc. Secreted and transmembrane polypeptides and nucleic acids encoding the same
US8460927B2 (en) 1999-11-30 2013-06-11 Mayo Foundation For Medical Education And Research B7-H1 antibodies and method of use
US6946134B1 (en) 2000-04-12 2005-09-20 Human Genome Sciences, Inc. Albumin fusion proteins
US9821039B2 (en) 2000-04-12 2017-11-21 Albumedix A/S Albumin fusion proteins
US20030125247A1 (en) * 2000-04-12 2003-07-03 Rosen Craig A. Albumin fusion proteins
US6905688B2 (en) 2000-04-12 2005-06-14 Human Genome Sciences, Inc. Albumin fusion proteins
US20080269125A1 (en) * 2000-04-12 2008-10-30 Human Genome Sciences, Inc. Albumin Fusion Proteins
US9775888B2 (en) 2000-04-12 2017-10-03 Albumedix A/S Treatment with factor ix-albumin fusion protein
US20080267962A1 (en) * 2000-04-12 2008-10-30 Human Genome Sciences, Inc. Albumin Fusion Proteins
US7482013B2 (en) 2000-04-12 2009-01-27 Human Genome Sciences, Inc. Albumin fusion proteins
US20030171267A1 (en) * 2000-04-12 2003-09-11 Rosen Craig A. Albumin fusion proteins
US9849162B2 (en) 2000-04-12 2017-12-26 Albumedix A/S Treatment with factor VII-albumin fusion protein
US20050266533A1 (en) * 2000-04-12 2005-12-01 Human Genome Sciences, Inc. Albumin fusion proteins
US10080785B2 (en) 2000-04-12 2018-09-25 Novozymes Biopharma Dk A/S Albumin fusion proteins
US20090285816A9 (en) * 2000-04-12 2009-11-19 Human Genome Sciences, Inc. Albumin Fusion Proteins
US6994857B2 (en) 2000-04-12 2006-02-07 Human Genome Sciences, Inc. Albumin fusion proteins
US9370565B2 (en) 2000-04-28 2016-06-21 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US8053414B2 (en) 2000-04-28 2011-11-08 The Johns Hopkins University Methods of using B7-DC molecules to induce or enhance an immune response
US8053558B2 (en) 2000-04-28 2011-11-08 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US20080241175A1 (en) * 2000-04-28 2008-10-02 The Johns Hopkins University Dendritic Cell Co-Stimulatory Molecules
US20080226662A1 (en) * 2000-04-28 2008-09-18 The Johns Hopkins University Dendritic cell co-stimulatory molecules
US20030143654A1 (en) * 2000-05-12 2003-07-31 Matthias Grell F-box containing protein
US20050123955A1 (en) * 2000-05-23 2005-06-09 Turner C. A.Jr. Novel human thrombospondin-like proteins and polynucleotides encoding the same
US20030211513A1 (en) * 2000-06-08 2003-11-13 Henry Yue Intracellular signaling proteins
US7329729B1 (en) * 2000-06-21 2008-02-12 Amgen Inc. Secreted epithelial colon stromal-1 molecules and uses thereof
US20110151579A1 (en) * 2000-06-21 2011-06-23 Amgen, Inc. Secreted Epithelial Stromal-1 Molecules and Uses Thereof
US20050196407A1 (en) * 2000-12-05 2005-09-08 Young John A.T. Receptor for B. anthracis toxin
US7435418B2 (en) * 2000-12-05 2008-10-14 Wisconsin Alumni Research Foundation Receptor for B. anthracis toxin
US20100249026A1 (en) * 2001-04-12 2010-09-30 Human Genome Sciences, Inc. Albumin fusion proteins
US7507413B2 (en) 2001-04-12 2009-03-24 Human Genome Sciences, Inc. Albumin fusion proteins
US20050100991A1 (en) * 2001-04-12 2005-05-12 Human Genome Sciences, Inc. Albumin fusion proteins
US20050244931A1 (en) * 2001-04-12 2005-11-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20060084794A1 (en) * 2001-04-12 2006-04-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20060166329A1 (en) * 2001-10-05 2006-07-27 Human Genome Sciences Inc. Albumin fusion proteins
US20040082761A1 (en) * 2001-12-18 2004-04-29 Duggan Brendan M. Cell adhesion proteins
US7238660B2 (en) 2001-12-21 2007-07-03 Human Genome Sciences, Inc. Albumin fusion proteins
US20080167239A1 (en) * 2001-12-21 2008-07-10 Human Genome Sciences, Inc. Albumin Fusion Proteins
US7189690B2 (en) 2001-12-21 2007-03-13 Human Genome Sciences, Inc. Albumin fusion proteins
US20060276396A1 (en) * 2001-12-21 2006-12-07 Human Genome Sciences, Inc. Albumin fusion proteins
US20070244047A1 (en) * 2001-12-21 2007-10-18 Human Genome Sciences, Inc. Albumin fusion proteins
US20080213886A1 (en) * 2001-12-21 2008-09-04 Human Genome Sciences, Inc. Albumin fusion proteins
US20090099073A1 (en) * 2001-12-21 2009-04-16 Human Genome Sciences, Inc. Albumin Fusion Proteins
US9296809B2 (en) 2001-12-21 2016-03-29 Human Genome Sciences, Inc. Albumin fusion proteins
US8252739B2 (en) 2001-12-21 2012-08-28 Human Genome Sciences, Inc. Albumin fusion proteins
US9221896B2 (en) 2001-12-21 2015-12-29 Human Genome Sciences, Inc. Albumin fusion proteins
US8211439B2 (en) 2001-12-21 2012-07-03 Human Genome Sciences, Inc. Albumin fusion proteins comprising insulin polypeptides
US8513189B2 (en) 2001-12-21 2013-08-20 Human Genome Sciences, Inc. Albumin fusion proteins
US20080153751A1 (en) * 2001-12-21 2008-06-26 Human Genome Sciences, Inc. Albumin Fusion Proteins
US7799759B2 (en) 2001-12-21 2010-09-21 Human Genome Sciences, Inc. Albumin fusion proteins
US8071539B2 (en) 2001-12-21 2011-12-06 Human Genome Sciences, Inc. Albumin fusion proteins
US7847079B2 (en) 2001-12-21 2010-12-07 Human Genome Sciences, Inc. Albumin fusion proteins
US20070259815A1 (en) * 2001-12-21 2007-11-08 Human Genome Sciences, Inc. Albumin fusion proteins
US20050054570A1 (en) * 2001-12-21 2005-03-10 Rosen Craig A. Albumin fusion proteins
US8012464B2 (en) 2001-12-21 2011-09-06 Human Genome Sciences, Inc. G-CSF-albumin fusion proteins
US20080146503A1 (en) * 2001-12-21 2008-06-19 Human Genome Sciences, Inc. Albumin Fusion Proteins
US20050186664A1 (en) * 2001-12-21 2005-08-25 Rosen Craig A. Albumin fusion proteins
US7977306B2 (en) 2001-12-21 2011-07-12 Human Genome Sciences, Inc. Albumin fusion proteins
US8993517B2 (en) 2001-12-21 2015-03-31 Human Genome Sciences, Inc. Albumin fusion proteins
US20150051378A1 (en) * 2002-04-18 2015-02-19 The General Hospital Corporation Drg11-responsive (dragon) gene family
US9193783B2 (en) 2002-04-18 2015-11-24 The General Hospital Corporation DRG11-responsive (dragon) polypeptides
US9475868B2 (en) 2002-04-18 2016-10-25 The General Hospital Corporation Antibodies that specifically bind to DRG11-responsive (DRAGON) proteins
US9644027B2 (en) 2002-04-18 2017-05-09 The General Hospital Corporation Antibodies that specifically bind to DRG11-responsive (DRAGON) proteins
US9868782B2 (en) 2002-04-18 2018-01-16 The General Hospital Corporation DRG11-responsive (DRAGON) gene family
US9416177B2 (en) 2002-04-18 2016-08-16 The General Hospital Corporation Antibodies that specifically bind to DRG11-responsive (dragon) proteins
US9637539B2 (en) * 2002-04-18 2017-05-02 The General Hospital Corporation Antibodies that specifically bind to DRG11-responsive (dragon) proteins
US8039589B1 (en) 2002-10-04 2011-10-18 Mayo Foundation For Medical Education And Research B7-DC variants
US8273864B2 (en) 2002-10-04 2012-09-25 Mayo Foundation For Medical Education And Research Nucleic acid molecules encoding B7-DC variants
US10414824B2 (en) 2002-11-22 2019-09-17 Ganymed Pharmaceuticals Ag Genetic products differentially expressed in tumors and the use thereof
US20050079546A1 (en) * 2003-05-01 2005-04-14 Dasa Lipovsek Serum albumin scaffold-based proteins and uses thereof
US20070141061A1 (en) * 2003-09-05 2007-06-21 Hudson Lindsey J Protein involved in carcinoma
WO2005082400A1 (en) * 2004-02-27 2005-09-09 Leangene Ab Therapeutic proteins for treating medical conditions associated with obesity and/or insulin resistance
EP1617875A1 (en) * 2004-04-27 2006-01-25 Seoul National University Industry Foundation Novel use of aim3 acting as a tumor suppressor
EP1617875A4 (en) * 2004-04-27 2007-10-10 Seoul Nat Univ Ind Foundation Novel use of aim3 acting as a tumor suppressor
US9775785B2 (en) 2004-05-18 2017-10-03 Ganymed Pharmaceuticals Ag Antibody to genetic products differentially expressed in tumors and the use thereof
US20110117184A1 (en) * 2004-06-17 2011-05-19 Philip James Bromley Compositions for mucosal delivery of agents
US7906140B2 (en) 2004-06-17 2011-03-15 Virun, Inc. Compositions for mucosal delivery of agents
US8414914B2 (en) 2004-06-17 2013-04-09 Virun, Inc. Compositions for mucosal delivery of agents
US8252323B2 (en) 2004-06-17 2012-08-28 Virun, Inc. Compositions for mucosal delivery of agents
US20050281772A1 (en) * 2004-06-17 2005-12-22 Bromley Philip J Compositions for mucosal delivery of agents
EP2359842A1 (en) 2004-07-14 2011-08-24 University of Utah Research Foundation Netrin-related compositions and uses
WO2006039504A2 (en) * 2004-10-01 2006-04-13 Cell Ionix, Inc. Method and formulation for stem cells’ stimulation
WO2006039504A3 (en) * 2004-10-01 2007-01-25 Cell Ionix Inc Method and formulation for stem cells’ stimulation
US11242387B2 (en) 2004-10-06 2022-02-08 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US9803015B2 (en) 2004-10-06 2017-10-31 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US20110200620A1 (en) * 2004-10-06 2011-08-18 Lieping Chen B7-h1 and methods of diagnosis, prognosis, and treatment of cancer
US8747833B2 (en) 2004-10-06 2014-06-10 Mayo Foundation For Medical Education And Research B7-H1 and methods of diagnosis, prognosis, and treatment of cancer
US11939378B2 (en) 2004-10-06 2024-03-26 Mayo Foundation For Medical Education And Research Costimulatory B7-H1 in renal cell carcinoma patients: indicator of tumor aggressiveness and potential therapeutic target
US20070053903A1 (en) * 2005-05-12 2007-03-08 Zeren Gao Methods of using pHHLA2 to co-stimulate T-cells
US10738108B2 (en) 2005-11-24 2020-08-11 Astellas Pharma Inc. Monoclonal antibodies against claudin-18 for treatment of cancer
US9499609B2 (en) 2005-11-24 2016-11-22 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US9212228B2 (en) 2005-11-24 2015-12-15 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US10017564B2 (en) 2005-11-24 2018-07-10 Ganymed Pharmaceuticals Gmbh Monoclonal antibodies against claudin-18 for treatment of cancer
US10174104B2 (en) 2005-11-24 2019-01-08 Ganymed Pharmaceuticals Gmbh Monoclonal antibodies against claudin-18 for treatment of cancer
US9751934B2 (en) 2005-11-24 2017-09-05 Ganymed Pharmaceuticals Ag Monoclonal antibodies against claudin-18 for treatment of cancer
US11739139B2 (en) 2005-11-24 2023-08-29 Astellas Pharma Inc. Monoclonal antibodies against Claudin-18 for treatment of cancer
US20100015642A1 (en) * 2006-01-05 2010-01-21 Kwon Eugene D B7-h1 and survivin in cancer
US20090215084A1 (en) * 2006-01-05 2009-08-27 Mayo Foundation For Medical Education And Research B7-h1 and b7-h4 in cancer
US8765915B2 (en) 2006-02-06 2014-07-01 Csl Behring Gmbh Modified coagulation factor VIIa with extended half-life
US20090176317A1 (en) * 2006-04-20 2009-07-09 Mayo Foundation For Medical Education And Research Soluble B7-H1
US8445447B2 (en) 2007-07-13 2013-05-21 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
US8153595B2 (en) 2007-07-13 2012-04-10 The Johns Hopkins University B7-DC variants immunogenic compositions and methods of use thereof
US20110020325A1 (en) * 2008-02-29 2011-01-27 Kwon Eugene D Methods for reducing granulomatous inflammation
US20090240666A1 (en) * 2008-03-19 2009-09-24 Sony Ericsson Mobile Communications Japan, Inc. Mobile terminal device and computer program
US8709416B2 (en) 2008-08-25 2014-04-29 Amplimmune, Inc. Compositions of PD-1 antagonists and methods of use
US20110195068A1 (en) * 2008-08-25 2011-08-11 Solomon Langermann Pd-1 antagonists and methods of use thereof
US20120322075A1 (en) * 2009-10-26 2012-12-20 Externautics S.P.A. Lung Tumor Markers and Methods of Use Thereof
US8680233B2 (en) * 2009-11-17 2014-03-25 Valorisation-Recherche, Limited Partnership Heteropeptides useful for reducing nonspecific adsorption
US20120329986A1 (en) * 2009-11-17 2012-12-27 Universite De Montreal Heteropeptides useful for reducing nonspecific adsorption
WO2012140627A1 (en) 2011-04-15 2012-10-18 Compugen Ltd. Polypeptides and polynucleotides, and uses thereof for treatment of immune related disorders and cancer
US10800847B2 (en) 2012-01-11 2020-10-13 Dr. Mepur Ravindranath Anti-HLA class-IB antibodies mimic immunoreactivity and immunomodulatory functions of intravenous immunoglobulin (IVIG) useful as therapeutic IVIG mimetics and methods of their use
US10053512B2 (en) 2012-05-09 2018-08-21 Ganymed Pharmaceuticals Ag Antibodies against claudin 18.2 useful in cancer diagnosis
US9512232B2 (en) 2012-05-09 2016-12-06 Ganymed Pharmaceuticals Ag Antibodies against Claudin 18.2 useful in cancer diagnosis
US10656156B2 (en) 2012-07-05 2020-05-19 Mepur Ravindranath Diagnostic and therapeutic potential of HLA-E monospecific monoclonal IgG antibodies directed against tumor cell surface and soluble HLA-E
US10167336B2 (en) 2013-03-14 2019-01-01 Mayo Foundation For Medical Education And Research Methods and materials for treating cancer
US11136393B2 (en) 2013-10-01 2021-10-05 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of Bim
US10259875B2 (en) 2013-10-01 2019-04-16 Mayo Foundation For Medical Education And Research Methods for treating cancer in patients with elevated levels of BIM
US10302653B2 (en) 2014-05-22 2019-05-28 Mayo Foundation For Medical Education And Research Distinguishing antagonistic and agonistic anti B7-H1 antibodies
US11504376B2 (en) 2014-07-23 2022-11-22 Mayo Foundation For Medical Education And Research Targeting DNA-PKCS and B7-H1 to treat cancer
US10517875B2 (en) 2014-07-23 2019-12-31 Mayo Foundation for Medical Engineering and Research Targeting DNA-PKcs and B7-H1 to treat cancer
US10875923B2 (en) 2015-10-30 2020-12-29 Mayo Foundation For Medical Education And Research Antibodies to B7-H1
US10767164B2 (en) 2017-03-30 2020-09-08 The Research Foundation For The State University Of New York Microenvironments for self-assembly of islet organoids from stem cells differentiation

Also Published As

Publication number Publication date
EP1274719A2 (en) 2003-01-15
EP1278544A4 (en) 2004-08-18
EP1832599A2 (en) 2007-09-12
US7507414B2 (en) 2009-03-24
EP2298355A2 (en) 2011-03-23
WO2001079480A1 (en) 2001-10-25
EP2275557A1 (en) 2011-01-19
EP1276856A4 (en) 2004-06-09
EP1983055A1 (en) 2008-10-22
AU2001259066A1 (en) 2001-10-30
WO2001079442A3 (en) 2002-06-06
US20030171267A1 (en) 2003-09-11
JP2003531590A (en) 2003-10-28
JP2004506407A (en) 2004-03-04
CA2405912A1 (en) 2001-10-18
EP1274720A1 (en) 2003-01-15
JP2003530838A (en) 2003-10-21
EP1274720A4 (en) 2004-08-18
US9821039B2 (en) 2017-11-21
US20130266553A1 (en) 2013-10-10
BE2016C059I2 (en) 2020-08-20
US7785599B2 (en) 2010-08-31
US20070287173A9 (en) 2007-12-13
EP2236152A1 (en) 2010-10-06
ES2529300T3 (en) 2015-02-18
US6994857B2 (en) 2006-02-07
US20080269126A1 (en) 2008-10-30
JP2003530852A (en) 2003-10-21
EP1276756A1 (en) 2003-01-22
EP2213743A1 (en) 2010-08-04
US20080267962A1 (en) 2008-10-30
AU2001264563A1 (en) 2001-10-30
WO2001077137A1 (en) 2001-10-18
WO2001079271A1 (en) 2001-10-25
US20120141449A1 (en) 2012-06-07
EP2067488A1 (en) 2009-06-10
EP2295456A1 (en) 2011-03-16
US20080269127A1 (en) 2008-10-30
DK2236152T3 (en) 2014-07-07
EP2267026A1 (en) 2010-12-29
US20120141415A1 (en) 2012-06-07
US20080269125A1 (en) 2008-10-30
WO2001079442A2 (en) 2001-10-25
US20040171123A1 (en) 2004-09-02
CA2405525A1 (en) 2001-10-25
US20080131399A1 (en) 2008-06-05
AU2001266557A1 (en) 2001-10-23
EP1803730A1 (en) 2007-07-04
US20140004095A1 (en) 2014-01-02
US6905688B2 (en) 2005-06-14
JP2003530846A (en) 2003-10-21
US20030199043A1 (en) 2003-10-23
WO2001079258A9 (en) 2002-02-28
EP1274719A4 (en) 2004-05-19
EP2311872A1 (en) 2011-04-20
US20100189686A1 (en) 2010-07-29
AU2001261024A1 (en) 2001-10-30
US8946156B2 (en) 2015-02-03
CA2405709A1 (en) 2001-10-25
EP2216409A1 (en) 2010-08-11
US6926898B2 (en) 2005-08-09
WO2001079444A3 (en) 2002-05-23
EP1278767A1 (en) 2003-01-29
EP2298355A3 (en) 2011-06-29
US10080785B2 (en) 2018-09-25
FR16C0043I2 (en) 2020-04-10
US20080261877A1 (en) 2008-10-23
JP2003530839A (en) 2003-10-21
US20090285816A9 (en) 2009-11-19
JP2014057589A (en) 2014-04-03
CA2405701A1 (en) 2001-10-25
AU2001262942A1 (en) 2001-10-30
CA2405563A1 (en) 2001-10-25
US20140010798A1 (en) 2014-01-09
US20030219875A1 (en) 2003-11-27
EP1278544A2 (en) 2003-01-29
WO2001079480A9 (en) 2003-09-18
US20120252732A1 (en) 2012-10-04
ES2484966T3 (en) 2014-08-12
US20180200346A1 (en) 2018-07-19
EP2216409B1 (en) 2014-12-03
AU2001274809A1 (en) 2001-10-30
WO2001077137A9 (en) 2002-05-02
US9849162B2 (en) 2017-12-26
WO2001079258A1 (en) 2001-10-25
CA2405550A1 (en) 2001-10-25
US7482013B2 (en) 2009-01-27
EP2357008A1 (en) 2011-08-17
JP2011217750A (en) 2011-11-04
US20080269128A1 (en) 2008-10-30
US20050266532A1 (en) 2005-12-01
EP1276856A1 (en) 2003-01-22
EP2236152B1 (en) 2014-06-04
EP1278767A4 (en) 2003-11-12
DK2216409T3 (en) 2015-01-05
CA2405557A1 (en) 2001-10-25
EP1276849A2 (en) 2003-01-22
WO2001079271A9 (en) 2002-02-28
WO2001079444A2 (en) 2001-10-25
US20050266533A1 (en) 2005-12-01
CA2747325A1 (en) 2001-10-25
WO2001079443A2 (en) 2001-10-25
EP2206720A1 (en) 2010-07-14
CA2405557C (en) 2013-09-24
WO2001079443A3 (en) 2002-02-21
AU2001259063A1 (en) 2001-10-30
EP1276756A4 (en) 2004-06-09
FR16C0043I1 (en) 2016-12-09
EP1276849A4 (en) 2004-06-09
JP2003530847A (en) 2003-10-21
US20030125247A1 (en) 2003-07-03
US9775888B2 (en) 2017-10-03
US20110280830A9 (en) 2011-11-17
EP1832599A3 (en) 2007-11-21

Similar Documents

Publication Publication Date Title
US6946134B1 (en) Albumin fusion proteins
US6994857B2 (en) Albumin fusion proteins
US7507413B2 (en) Albumin fusion proteins
US20060084794A1 (en) Albumin fusion proteins
US20060166329A1 (en) Albumin fusion proteins
EP2474318A1 (en) Albumin fusion proteins
AU2010227035A1 (en) Albumin fusion proteins
US20090075880A1 (en) Albumin Fusion Proteins

Legal Events

Date Code Title Description
AS Assignment

Owner name: HUMAN GENOME SCIENCES, INC., MARYLAND

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:ROSEN, CRAIG A.;HASELTINE, WILLIAM A.;REEL/FRAME:012587/0368

Effective date: 20010808

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION