CA2598239A1 - Nanoparticulate formulations of taxanes and carrier proteins for use in combination chemotherapy - Google Patents

Nanoparticulate formulations of taxanes and carrier proteins for use in combination chemotherapy Download PDF

Info

Publication number
CA2598239A1
CA2598239A1 CA002598239A CA2598239A CA2598239A1 CA 2598239 A1 CA2598239 A1 CA 2598239A1 CA 002598239 A CA002598239 A CA 002598239A CA 2598239 A CA2598239 A CA 2598239A CA 2598239 A1 CA2598239 A1 CA 2598239A1
Authority
CA
Canada
Prior art keywords
composition
taxane
albumin
cancer
abraxanetm
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.)
Granted
Application number
CA002598239A
Other languages
French (fr)
Other versions
CA2598239C (en
Inventor
Neil P. Desai
Patrick Soon-Shiong
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.)
Abraxis Bioscience LLC
Original Assignee
Individual
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=36593193&utm_source=google_patent&utm_medium=platform_link&utm_campaign=public_patent_search&patent=CA2598239(A1) "Global patent litigation dataset” by Darts-ip is licensed under a Creative Commons Attribution 4.0 International License.
Application filed by Individual filed Critical Individual
Priority to CA3054535A priority Critical patent/CA3054535A1/en
Priority to CA2902071A priority patent/CA2902071C/en
Publication of CA2598239A1 publication Critical patent/CA2598239A1/en
Application granted granted Critical
Publication of CA2598239C publication Critical patent/CA2598239C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/14Particulate form, e.g. powders, Processes for size reducing of pure drugs or the resulting products, Pure drug nanoparticles
    • A61K9/141Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers
    • A61K9/146Intimate drug-carrier mixtures characterised by the carrier, e.g. ordered mixtures, adsorbates, solid solutions, eutectica, co-dried, co-solubilised, co-kneaded, co-milled, co-ground products, co-precipitates, co-evaporates, co-extrudates, co-melts; Drug nanoparticles with adsorbed surface modifiers with organic macromolecular compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/165Amides, e.g. hydroxamic acids having aromatic rings, e.g. colchicine, atenolol, progabide
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/16Amides, e.g. hydroxamic acids
    • A61K31/17Amides, e.g. hydroxamic acids having the group >N—C(O)—N< or >N—C(S)—N<, e.g. urea, thiourea, carmustine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/28Compounds containing heavy metals
    • A61K31/282Platinum compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/436Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a six-membered ring having oxygen as a ring hetero atom, e.g. rapamycin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/517Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with carbocyclic ring systems, e.g. quinazoline, perimidine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/66Phosphorus compounds
    • A61K31/675Phosphorus compounds having nitrogen as a ring hetero atom, e.g. pyridoxal phosphate
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/69Boron compounds
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7028Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages
    • A61K31/7034Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin
    • A61K31/704Compounds having saccharide radicals attached to non-saccharide compounds by glycosidic linkages attached to a carbocyclic compound, e.g. phloridzin attached to a condensed carbocyclic ring system, e.g. sennosides, thiocolchicosides, escin, daunorubicin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7042Compounds having saccharide radicals and heterocyclic rings
    • A61K31/7052Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides
    • A61K31/706Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom
    • A61K31/7064Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines
    • A61K31/7068Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid
    • A61K31/7072Compounds having saccharide radicals and heterocyclic rings having nitrogen as a ring hetero atom, e.g. nucleosides, nucleotides containing six-membered rings with nitrogen as a ring hetero atom containing condensed or non-condensed pyrimidines having oxo groups directly attached to the pyrimidine ring, e.g. cytidine, cytidylic acid having two oxo groups directly attached to the pyrimidine ring, e.g. uridine, uridylic acid, thymidine, zidovudine
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/243Platinum; Compounds thereof
    • 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
    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • 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
    • A61K47/69Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • A61K47/6931Medicinal 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 conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle the material constituting the nanoparticle being a polymer
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K51/00Preparations containing radioactive substances for use in therapy or testing in vivo
    • A61K51/02Preparations containing radioactive substances for use in therapy or testing in vivo characterised by the carrier, i.e. characterised by the agent or material covalently linked or complexing the radioactive nucleus
    • A61K51/04Organic compounds
    • A61K51/08Peptides, e.g. proteins, carriers being peptides, polyamino acids, proteins
    • A61K51/10Antibodies or immunoglobulins; Fragments thereof, the carrier being an antibody, an immunoglobulin or a fragment thereof, e.g. a camelised human single domain antibody or the Fc fragment of an antibody
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61NELECTROTHERAPY; MAGNETOTHERAPY; RADIATION THERAPY; ULTRASOUND THERAPY
    • A61N5/00Radiation therapy
    • A61N5/10X-ray therapy; Gamma-ray therapy; Particle-irradiation therapy
    • 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
    • A61P15/00Drugs for genital or sexual disorders; Contraceptives
    • 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
    • A61P35/00Antineoplastic agents
    • 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
    • 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
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/08Vasodilators for multiple indications
    • 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
    • BPERFORMING OPERATIONS; TRANSPORTING
    • B82NANOTECHNOLOGY
    • B82YSPECIFIC USES OR APPLICATIONS OF NANOSTRUCTURES; MEASUREMENT OR ANALYSIS OF NANOSTRUCTURES; MANUFACTURE OR TREATMENT OF NANOSTRUCTURES
    • B82Y5/00Nanobiotechnology or nanomedicine, e.g. protein engineering or drug delivery
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2121/00Preparations for use in therapy
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00

Abstract

The present invention provides combination therapy methods of treating proliferative diseases (such as cancer) comprising a first therapy comprising administering to an individual an effective amount of a nanoparticle composition (such as a nonparticle composition of a taxane), and a second therapy which may include, for example, radiation, surgery, administration of chemotherapeutic agents, or combinations thereof.
Also provided are methods of administering to an individual a drug (such as a taxane) in a nanoparticle composition based on a metronomic dosing regime.

Description

COMBINATIONS AND MODES OF ADMINISTRATION OF THERAPEUTIC
AGENTS AND COMBINATION THERAPY

RELATED APPLICATIONS
[0001] This application claims priority benefit to provisional application 60/654,245, filed on February 18, 2005, which is incorporated by reference herein in its entirety.
TECHNICAL FIELD
[0002] The present invention relates to methods and compositions for the treatment of proliferative diseases comprising the administration of a combination of a taxane and at least one other and other therapeutic agents, as well as other treatment modalities useful in the treatment of proliferative diseases. In particular, the invention relates to the use of nanoparticles comprising paclitaxel and albumin (such as AbraxaneTM) in combination with other chemotherapeutic agents or radiation, which may be used for the treatment of cancer.

BACKGROUND
[0003] The failure of a significant number of tumors to respond to drug and/or radiation therapy is a serious problem in the treatment of cancer. In fact, this is one of the main reasons why many of the most prevalent forms of human cancer still resist effective chemotherapeutic intervention, despite certain advances in the field of chemotherapy.
[0004] Cancer is now primarily treated with one or a combination of three types of therapies: surgery, radiation, and chemotherapy. Surgery is a traditional approach in which all or part of a tumor is removed from the body. Surgery generally is only effective for treating the earlier stages of cancer. While surgery is sometimes effective in removing tumors located at certain sites, for example, in the breast, colon, and skin, it cannot be used in the treatment of tumors located in other areas, inaccessible to surgeons, nor in the treatment of disseminated neoplastic conditions such as leukemia. For more than 50% of cancer individuals, by the time they are diagnosed they are no longer candidates for effective surgical treatment. Surgical procedures may increase tumor metastases through blood circulation during surgery. Most of cancer individuals do not die from the cancer at the time of diagnosis or surgery, but rather die from the metastasis and the recurrence of the cancer.
[0005] Other therapies are also often ineffective. Radiation therapy is only effective for individuals who present with clinically localized disease at early and middle stages of cancer, and is not effective for the late stages of cancer with metastasis. Radiation is generally applied to a defined area of the subject's body which contains abnormal proliferative tissue, in order to maximize the dose absorbed by the abnormal tissue and minimize the dose absorbed by the nearby normal tissue. However, it is difficult (if not impossible) to selectively administer therapeutic radiation to the abnormal tissue. Thus, normal tissue proximate to the abnormal tissue is also exposed to potentially damaging doses of radiation throughout the course of treatment. There are also some treatments that require exposure of the subject's entire body to the radiation, in a procedure called "total body irradiation", or "TBI." The efficacy of radiotherapeutic techniques in destroying abnormal proliferative cells is therefore balanced by associated cytotoxic effects on nearby normal cells. Because of this, radiotherapy techniques have an inherently narrow therapeutic index which results in the inadequate treatment of inost tumors.
Even the best radiotherapeutic techniques may result in incomplete tumor reduction, tumor recurrence, increasing tumor burden, and induction of radiation resistant tumors.
[0006] Chemotherapy involves the disruption of cell replication or cell metabolism.
Chemotherapy can be effective, but there are severe side effects, e.g., voiniting, low white blood cells (WBC), loss of hair, loss of weight and other toxic effects.
Because of the extremely toxic side effects, many cancer individuals cannot successfully finish a complete chemotherapy regime. Chemotherapy-induced side effects significantly impact the quality of life of the individual and may dramatically influence individual compliance with treatment. Additionally, adverse side effects associated with chemotherapeutic agents are generally the major dose-limiting toxicity (DLT) in the administration of these drugs. For example, mucositis is one of the major dose limiting toxicity for several anticancer agents, including the antimetabolite cytotoxic agents 5-FU, methotrexate, and antitumor antibiotics, such as doxonibicin. Many of these chemotherapy-induced side effects if severe may lead to hospitalization, or require treatment with analgesics for the treatment of pain. Some cancer individuals die from the chemotherapy due to poor tolerance to the chemotherapy. The extreme side effects of anticancer drugs are caused by the poor target specificity of such drugs. The dnigs circulate through most normal organs of individuals as well as intended target tumors. The poor target specificity that causes side effects also decreases the efficacy of chemotherapy because only a fraction of the drugs is correctly targeted. The efficacy of chemotherapy is ftirther decreased by poor retention of the anti-cancer dnigs within the target tumors.
[0007] Due to the severity and breadth of neoplasm, tumor and cancer, there is a great need for effective treatments of such diseases or disorders that overcome the shortcomings of surgery, chemotherapy, and radiation treatment.

Problems of Chemotherapeutic Agents
[0008] The drug resistance problem is a reason for the added importance of combination chemotherapy, as the therapy both has to avoid the emergence of resistant cells and to kill pre-existing cells which are already dn.ig resistant.
[0009] Drug resistance is the name given to the circumstance when a disease does not respond to a treatment drug or dnigs. Drug resistance can be either intrinsic, which means the disease has never been responsive to the drug or dnigs, or it can be acquired, wllich means the disease ceases responding to a drug or dnigs that the disease had previously been responsive to. Multidrug resistance (MDR) is a specific type of drug resistance that is characterized by cross-resistance of a disease to more than one fi.inctionally and/or structurally unrelated dnigs. Multidnig resistance in the field of cancer is discussed in greater detail in "Detoxification Mechanisms and Tumor Cell Resistance to Anticancer Drugs," by Kuzmich and Tew, particularly section VII "The Multidrug-Resistant Phenotype (MDR)," Medical Research Reviews, Vol. 11, No.
2, 185-217, (Section VII is at pp. 208-213) (1991); and in "Multidnig Resistance and Chemosensitization: Therapeutic Implications for Cancer Chemotherapy," by Georges, Sharom and Ling, Advances in Pharmacology, Vol. 21, 185-220 (1990).
[0010] One form of multi-drug resistance (MDR) is mediated by a membrane bound 170-180 kD energy-dependent efflux pump designated as P-glycoprotein (P-gp).
P-glycoprotein has been shown to play a major role in the intrinsic and acquired resistance of a number of human ti.unors against hydrophobic, natural product dn.igs.
Drugs that act as substrates for and are consequently detoxified by P-gp include the vinca alkaloids (vincristine and vinblastine), anthracyclines (Adriainycin), and epipodophyllotoxins (etoposide). While P-gp associated MDR is a major deterininant in tumor cell resistance to chemotherapeutic agents, it is clear that the phenomenon of MDR is multifactorial and involves a number of different mechanisms.
[0011] A major complication of cancer chemotherapy and of antiviral chemotherapy is damage to bone marrow cells or suppression of their fimction.
Specifically, chemotherapy damages or destroys hematopoietic precursor cells, primarily found in the bone marrow and spleen, impairing the production of new blood cells (granulocytes, lymphocytes, erythrocytes, monocytes, platelets, etc.).
Treatment of cancer individuals with 5-fluorouracil, for example, reduces the number of leukocytes (lymphocytes and/or granulocytes), and can result in enhanced susceptibility of the individuals to infection. Many cancer individuals die of infection or other consequences of hematopoietic failure subsequent to chemotherapy. Chemotherapeutic agents can also result in subnoi-mal forTnation of platelets which produces a propensity toward hemorrhage.
Inhibition of erythrocyte production can result in anemia. For some cancer individuals, the risk of damage to the hematopoietic system or other important tissues frequently limits the opportunity for chemotherapy dose escalation of chemotherapy agents high enough to provide good antitumor or antiviral efficacy. Repeated or high dose cycles of chemotherapy may be responsible for severe stem cell depletion leading to serious long-term hematopoietic sequelea and marrow exhaustion.
[0012] Prevention of, or protection from, the side effects of chemotherapy would be a great benefit to cancer individuals. For life-threatening side effects, efforts have concentrated on altering the dose and schedules of the chemotherapeutic agent to reduce the side effects. Other options are becoming available, such as the use of granulocyte colony stimulating factor (G-CSF), granulocyte-macrophage-CSF (GM-CSF), epidennal growth factor (EGF), interleukin 11, erythropoietin, thrombopoietin, megakaryocyte development and growth factor, pixykines, stem cell factor, FLT-ligand, as well as interleukins 1, 3, 6, and 7, to increase the number of norinal cells in various tissues before the start of chemotherapy (See Jimenez and Yunis, Cancer Research 52:413-415;
1992).
The mechanisms of protection by these factors, while not fully understood, are most likely associated with an increase in the number of normal critical target cells before treatment with cytotoxic agents, and not with increased stuvival of cells following chemotherapy.
Chemotherapeutic Targeting For Tumor Treatment
[0013] Both the growth and metastasis of solid tuinors are angiogenesis-dependent (Folkman, J. Carzcef- Res., 46, 467-73 (1986); Folkman, J. Nat. Canccr Inst., 82, 4-6 (1989); Folkinan et al., "Tumor Aiigiogenesis," Chapter 10, pp. 206-32, in The Molecular Basis of Caracei-, Mendelsohn et al., eds. (W. B. Saunders, 1995)). It has been shown, for example, that tumors which enlarge to greater than 2 mm in diameter must obtain their own blood supply and do so by inducing the growth of new capillary blood vessels.
After these new blood vessels become embedded in the tumor, they provide nutrients and growth factors essential for tumor growth as well as a means for tumor cells to enter the circulation and metastasize to distant sites, such as liver, lung or bone (Weidner, New Eng. J. Med., 324(1), 1-8 (1991)). When used as drugs in tumor-bearing animals, natural inhibitors of angiogenesis can prevent the growth of small tumors (O'Reilly et al., O'Reilly et al., Cell, 79, 315-28 (1994)). Indeed, in some protocols, the application of such inhibitors leads to tumor regression and doi-mancy even after cessation of treatment (O'Reilly et al., Cell, 88, 277-85 (1997)). Moreover, supplying inhibitors of angiogenesis to certain tumors can potentiate their response to other therapeutic regimes (e.g., chemotherapy) (see, e.g., Teischer et al., Int. J. Cancer, 57, 920-25 (1994)).
[0014] Protein tyrosine kinases catalyze the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth and differentiation (A.
F. Wilks, Progress in Growth Factor Research, 1990, 2, 97-111; S. A.
Courtneidge, Dev.
Supp.l, 1993, 57-64; J. A. Cooper, Semin. Cell Biol., 1994, 5(6), 377-387; R.
F. Paulson, Semin. Immunol., 1995, 7(4), 267-277; A. C. Chan, Curr. Opin. Immunol., 1996, 8(3), 394-401). Protein tyrosine kinases can be broadly classified as receptor (e.g.
EGFr, c-erbB-2, c-met, tie-2, PDGFr, FGFr) or non-receptor (e.g. c-src, Ick, Zap70) kinases.
Inappropriate or uncontrolled activation of many of these kinases, i.e.
aberrant protein tyrosine kinase activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growth. For exainple, elevated epidermal growth factor receptor (EGFR) activity has been implicated in non-small cell lung, bladder and head and neck cancers, and increased c-erbB-2 activity in breast, ovarian, gastric and pancreatic cancers.
Thus, inhibition of protein tyrosine kinases should be useful as a treatinent for tumors such as those outlined above.
[0015] Growth factors are substances that induce cell proliferation, typically by binding to specific receptors on cell surfaces. Epidermal growth factor (EGF) induces proliferation of a variety of cells in vivo, and is required for the growth of most cultured cells. The EGF receptor is a 170-180 kD ineinbrane-spanning glycoprotein, which is detectable on a wide variety of cell types. The extracellular N-terminal domain of the receptor is highly glycosylated and binds EGF antibodies that selectively bind to EGFR.
Agents that competitively bind to EGFR have been used to treat certain types of cancer, since many tumors of inesodermal and ectodeirnal origin overexpress the EGF
receptor.
For exainple, the EGF receptor has been shown to be overexpressed in many gliomas, squamous cell carcinomas, breast carcinomas, melanomas, invasive bladder carcinomas and esophageal cancers. Attempts to exploit the EGFR system for anti-turnor therapy have generally involved the use of monoclonal antibodies against the EGFR. In addition, studies with primary human mammary tumors have shown a correlation between high EGFR
expression and the presence of metastases, higher rates of proliferation, and shorter individual survival.
[0016] Herlyn et al., in U.S. Patent 5,470,571, disclose the use of radiolabeled Mab 425 for treating glioinas that express EGFR. Herlyn et al. repoi-t that anti-EGFR antibodies may either stimulate or inhibit cancer cell growth and proliferation. Other monoclonal antibodies having specificity for EGFR, either alone or conjugated to a cytotoxic compound, have been reported as being effective for treating certain types of cancer.
Bendig et al, in U.S. Patent 5,558,864, disclose therapeutic anti-EGFR Mab's for competitively binding to EGFR. Heimbrook et al., in U.S. Patent 5,690,928, disclose the use of EGF fiised to a Pseudonaonas species-derived endotoxin for the treatment of bladder cancer. Brown et al., in U.S. Patent 5,859,018, disclose a method for treating diseases characterized by cellular hyperproliferation mediated by, inter alia, EGF.
Chemotherapeutic Modes of Administration
[0017] People diagnosed as having cancer are frequently treated with single or multiple chemotherapeutic agents to kill cancer cells at the primary tumor site or at distant sites to where cancer has metastasized. Chemotherapy treatment is typically given either in a single or in several large doses or over variable times of weeks to months.
However, repeated or high dose cycles of chemotherapy may be responsible for increased toxicities and severe side effects.
[0018] New studies suggest that metronomic chemotherapy, the low-dose and frequent administration of cytotoxic agents without prolonged drug-free breaks, targets activated endothelial cells in the tumor vasculature. A munber of preclinical studies have demonstrated superior anti-tumor efficacy, potent antiangiogenic effects, and reduced toxicity and side effects (e.g., myelosuppression) of metronomic regimes compared to maximum tolerated dose (MTD) counterparts (Bocci, et al., Cancer Res, 62:6938-6943, (2002); Bocci, et al., PNAS, vol, 100(22):12917-12922, (2003); and Bertolini, et al., Cancer Res, 63(15):4342-4346, (2003)). It remains unclear wliether all chemotherapeutic dnigs exert similar effects or whether sonie are better suited for such regimes than others.
Nevertheless, metronomic chemotherapy appears to be effective in overcoming some of the major shortcomings associated with cheniotherapy.

Chemotherapeutic Agents
[0019] Paclitaxel has been shown to have significant antineoplastic and anticancer effects in drug-refractory ovarian cancer and has shown excellent antitumor activity in a wide variety of tumor models, and also inhibits angiogenesis when used at very low doses (Grant et al., Int. J. Cancer, 2003). The poor aqueous solubility of paclitaxel, however, presents a problem for human administration. Indeed, the delivery of drugs that are inherently insoluble or poorly soluble in an aqueous medium can be seriously impaired if oral delivery is not effective. Accordingly, currently used paclitaxel formulations (e.g., Taxolo) require a Cremophor to solubilize the drt.ig. The presence of Cremophor in this fonnulation has been linked to severe hypersensitivity reactions in animals (Lorenz et al., Agents Actions 7:63-67 (1987)) and humans (Weiss et al., J. Clin. Oncol.
8:1263-68 (1990)) and consequently requires preinedication of individuals with corticosteroids (dexamethasone) and antihistamines. It was also reported that clinically relevant concentrations of the formulation vehicle Cremophor EL in Taxol nullify the antiangiogenic activity of paclitaxel, suggesting that this agent or other anticancer drugs formulated in Cremophor EL may need to be used at much higher doses than anticipated to achieve effective metronomic chemotherapy (Ng et al., Cancer Res., 64:821-(2004)). As such, the advantage of the lack of undesirable side effects associated with low-dose paclitaxel regimes vs. conventional MTD chemotherapy may be compromised.
See also U.S. Patent Pub. No. 2004/0143004; W000/64437.

, Abraxane TM is a Cremophor EL-free nanoparticle albumin-bound paclitaxel
[0020] Preclinical models have shown significant improvement in the safety and efficacy of AbraxaneTM compared with Taxol (Desai et al., EORTC-NCI-AACR, 2004) and in individuals with metastatic breast cancer (O'Shaughnessy et al., San Antonio Breast Cancer Symposiuin, Abstract #1122, Dec. 2003). This is possibly due to the absence of surfactants (e.g., Cremophor or TweencR' 80, used in Taxol and Taxotere , respectively) in AbraxaneTM, and/or preferential utilization of an albumin-based transport mechanism utilizing gp60/caveolae on microvascular endothelial cells (Desai et al., EORTC-NCI-AACR, 2004). In addition, both Cremophor and Tween 80 have been shown to strongly inhibit the binding of paclitaxel to albumin, possibly affecting albumin based transport (Desai et al., EORTC-NCI-AACR, 2004).
[0021] IDN5109 (Ortataxel) is a new taxane, currently in phase II, selected for its lack of cross-resistance in tLunor cell lines expressing the multidrug resistant phenotype (MDR/Pgp) and inhibition of P-glycoprotein (Pgp) (Minderman; Ca77Ce7r Chenaotl7e7-.
Pha7=macol. 2004; 53:363-9). Due to its hydrophobicity, IDN5109 is currently formulated in the surfactant Tween 80 (same vehicle as Taxotere ). Removal of surfactants from taxane fonnulations e.g., in the case of nanoparticle albumin-bound paclitaxel (AbraxaneTM) showed improvements in safety and efficacy over their surfactant containing counterparts (O'Shaughnessy et al., San Antonio Breast Cancer Symposium, Abstract #1122, Dec. 2003). Tween 80 also strongly inhibited the binding of the taxane, paclitaxel, to albumin, possibly compromising albumin based dnig transport via the gp60 receptor on microvessel endothelial cells (Desai et al., EORTC-NCI-AACR, 2004).
[0022] The antitumor activity of colchicine, which is the major alkaloid of the autumn crocus, Colchicuna autunanale, and the African climbing lily, Gloriosa superba, was first reported at the beginning of the 20th century. The elucidation of its stnicture was finally completed from X-ray studies and a number oftotal syntheses (see Shiau et al., J.
Pharm. Sci. 1978, 67(3) 394-397). Colchicine is thought to be a mitotic poison, particularly in tyhmic, intestinal, and hermatopoietic cells, which acts as a spindle poison and blocks the kinesis. Its effect on the mitotic spindle is thought to represent a special case of its effects on various organized, labile, fibrillar systems concerned with structure and movement.
[0023] Thiocolchicine dimer IDN5404 was selected for its activity in human ovarian subline resistant to cisplatin and topotecan A2780-CIS and A2780-TOP.
This effect was related to dual mechanisms of action, i.e., microtubule activity as in Vinca alkaloids and a topoisomerase I inhibitory effect different from camptothecin.
(Raspaglio, Biochemical Pharmacology 69:113-121 (2005)).
[0024] It has been found that nanoparticle compositions of a taxane (such as albumin bound paclitaxel (AbraxaneTM)) have significantly lower toxicities than other taxanes like Taxolc" and Taxotere with significantly improved outcomes in both safety and efficacy.
[0025] Combination chemotherapy, e.g., combining one or more chemotherapeutic agents or other modes of treatment, e.g., combining for example, chemotherapy with radiation or surgery and chemotherapy, have been found to be more successfiil than single agent chemotherapeutics or individual modes of treatment respectively.
[0026] , Other references include U.S. Pub. No. 2006/0013819; U.S. Pub. No.
2006/0003931; W005/117986; W005/117978; and W005/000900.
[0027] More effective treatments for proliferative diseases, especially cancer, are needed.
[0028] The disclosures of all publications, patents, patent applications and published patent applications referred to herein are hereby incorporated herein by reference in their entirety.

BRIEF SUMMARY OF THE INVENTION
[00291 The present invention provides methods for the treatment of proliferative diseases such as cancer. The invention provides combination therapy methods of treating proliferative diseases (such as cancer), comprising a) a first therapy comprising administering to an individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin) and b) a second therapy, such as chemotherapy, radiation therapy, surgery, or combinations thereof. In another aspect, there are provided methods of administering to an individual a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin) based on a metronomic dosing regime.
[0030] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of at least one other chemotherapeutic agent. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual comprising administering to the individual a) an effective amount of a coinposition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), and b) an effective amount of at least one other chemotherapeutic agent. In some embodiments, the chemotlierapeutic agent is any of (and in some embodiments selected from the group consisting of) antimetabolites (including nucleoside analogs), platinum-based agents, alkylating agents, tyrosine kinase inhibitors, anthracycline antibiotics, vinca alkloids, proteasome inhibitors, macrolides, and topoisomerase inhibitors. In some embodiments, the chemotherapeutic agent is a platinum-based agent, such as carboplatin.

[0031] In some embodiments, the composition comprising nanoparticles (also refeiTed to as "nanoparticle composition") and the chemotherapeutic agent are administered simultaneously, either in the same composition or in separate coinpositions.
In some embodiments, the nanoparticle composition and the chemotherapeutic agent are administered sequentially, i.e., the nanoparticle composition is administered either prior to or after the administration of the chemotherapeutic agent. In some einbodiments, the administration of the nanoparticle coinposition and the chemotherapeutic agent are concurrent, i.e., the administration period of the nanoparticle composition and that of the chemotherapeutic agent overlap with each other. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent are non-concurrent. For example, in some embodiments, the administration of the nanoparticle composition is terminated before the chemotherapeutic agent is administered. In some embodiments, the administration of the chemotherapeutic agent is terminated before the nanoparticle composition is administered.

[0032] In some embodiments, the first therapy taxane is nano-particle albumin bound paxlitaxel, described, for example, in U.S. Patent 6,566,405, and commercially available under the tradename AbraxaneTM. In addition, the first tllerapy taxane is also considered to be nanoparticle albumin bound docetaxel described for example in U.S.
Patent Application Publication 2005/0004002A1.

[0033] In another aspect, there is provided a method of treating a proliferative disease (such as cancer) in an individual comprising a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) a second therapy comprising radiation therapy, surgery, or combinations thereof. In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual comprising a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising paclitaxel and an albuinin (such as AbraxaneTM), and b) a second therapy comprising radiation therapy, surgery, or combinations thereof. In some embodiments, the second therapy is radiation therapy. In some embodiments, the second therapy is surgery. In some embodiments, the first therapy is caiTied out prior to the second therapy. In some embodiments, the first therapy is carried out after the second therapy.

[0034] In another aspect, the method comprises adininistering to a mammal having a proliferative disease (such as cancer) a combination therapy comprising a first therapy comprising a taxane and a second therapy selected from the grotip consisting of chemotherapeutic agent and radiation or combinations thereof. The combination therapy may be administered in any of a variety of ways such as sequentially or simultaneously, and if sequential, the taxane may be administered before or after the second therapy although it is preferred that the first therapy comprising a taxane is administered first. It will also be understood that the second therapy can include more than one chemotherapeutic agent.

[0035] The present invention also provides metronomic therapy regimes. In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein the nanoparticle composition is adininistered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of taxane at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of paclitaxel at each administration is about 0.25%
to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the dose of the taxane (such as paclitaxel, for example AbraxaneTM) per administration is less than about any of 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 18%, 20%, 22%, 24%, or 25% of the maximum tolerated dose. In some embodiments, the nanoparticle composition is administered at least about any of lx, 2x, 3x, 4x, 5x, 6x, 7x (i.e., daily) a week. In some embodiments, the intervals between each administration are less than about any of 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, and 1 day. In some embodiments, the nanoparticle composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30 and 36 months.

[0036] In some embodiments, there is provided a method of administering a composition comprising nanoparticles coinprising a taxane and a carrier protein (such as albumin), wherein the taxane is administered over a period of at least one month, wlzerein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25 mg/m2 to about 25 mg/mz. In some embodiments, there is provided a method of administering a composition comprising nanoparticles coinprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), wherein the paclitaxel is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25 mg/m2 to about 25 mg/mz. In some embodiments, the dose of the taxane (such as paclitaxel, for example AbraxaneTM) per administration is less than about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 20, 22, and 25 mg/m2. In some embodiments, the nanoparticle coinposition is administered at least about any of lx, 2x, 3x, 4x, 5x, 6x, 7x (i.e., daily) a week. In some embodiments, the intervals between each administration are less than about any of 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, and 1 day. In some embodiments, the nanoparticle composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30 and 36 months.

[0037] The methods of the invention generally comprise administration of a composition comprising nanoparticles comprising a taxane and a carrier protein. In some embodiments, the nanoparticle composition coinprises nanoparticles comprising paclitaxel and an albumin. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 nm. In some embodiments, the paclitaxel/albumin nanoparticle composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the weight ratio of the albumin to paclitaxel in the coinposition is about 18:1 or less, such as about 9:1 or less. In some embodiments, the paclitaxel is coated with albumin. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 nm and the paclitaxel/albuinin composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 nm and the paclitaxel is coated witli albumin. Other combinations of the above characteristics are also contemplated. In some embodiments, the nanoparticle composition is AbraxaneTM. Nanoparticle compositions comprising other taxanes (such as docetaxel and ortataxel) may also comprise one or more of the above characteristics.

[0038] These and other aspects and advantages of the present invention will become apparent from the subsequent detailed description and the appended claims. It is to be understood that one, some, or all of the properties of the various einbodiments described herein may be combined to form otlier embodiments of the present invention.

BRIEF DESCRIPTION OF FIGURES
[0039] Figure 1A shows the effect of ABI-007 on rat aortic ring angiogenesis.
Figure 1 B shows the effect of ABI-007 on hunian endothelial cell proliferation. Figure 1 C
shows the effect of ABI-007 on endothelial cell tube foi-mation.

[0040] Figure 2 shows the detennination of an optimal biological dose of ABI-for metronomic dosing. Shown are the levels of viable circulating endothelial progenitors (CEPs) in peripheral blood of Balb/cJ mice in response to escalating doses of ABI-007.
Untr'd, untreated control; S/A, saline/albumin vehicle control. Bars, mean SE. *
Significantly (p < 0.05 ) different from the untreated control.
[00411 Figures 3A and 3B show the effects of ABI-007 and Taxol used in metronomic or MTD regimes on MDA-MB-231 (A) and PC3 (B) tumor growth tumor-bearing SCID mice. Figures 3C and 3D show the effects of ABI-007 and Taxol used in metronomic or MTD regimes on the body weight of MDA-MB-231 (C) and PC3 (D) tumor-bearing SCID mice.
[0042] Figures 4A and 4B show changes in the levels of viable circulating endothelial progenitors (CEPs) in peripheral blood of MDA-MB-231 (Fig. 4A) and (Fig. 4B) tumor-bearing SCID mice after treatment with A, saline/albumin; B, Cremophor EL control; C, metronomic Taxol 1.3 mg/kg; D, E, and F, metronomic ABI-007 3, 6, and mg/kg, respectively; G, MTD Taxol; H, MTD ABI-007. Bars, mean SE. a Significantly (p < 0.05) different from saline/albumin vehicle control. b Significantly (p <
0.05) different from Cremophor EL vehicle control.
[0043] Figure 5A shows intratumoral microvessel density of MDA-MB-231 (a) and PC3 (o) xenografts treated with A, saline/albumin; B, Cremophor EL
control; C, metronomic Taxol 1.3 mg/kg; D, E, and F, metronomic ABI-007 3, 6, and 10 mg/kg, respectively; G, MTD Taxol; H, MTD ABI-007. Bars, mean + SE. Figure 5B and 5C
show the correlation between intratumoral microvessel density and the number of viable CEPs in peripheral blood in MDA-MB-231 (Fig. 5B) and PC3 (Fig. 5C) tumor-bearing SCID mice.
[0044] Figure 6 shows the effects of ABI-007 or Taxol used in metronomic or MTD regimes on basic fibroblast growth factor (bFGF)-induced angiogenesis in matrigel plugs injected subcutaneously into the flanlcs of Balb/cJ mice. Treatments-A, saline/albumin; B, Cremophor EL control; C, metronomic Taxol 1.3 mg/kg; D, E, and F, metronomic ABI-007 3, 6, and 10 mg/kg, respectively; G, MTD Taxol; H, MTD ABI-007.
Matrigel implanted without bFGF (-bFGF) seived as negative control. Bars, mean SE.
[0045] Figure 7A and Figure 7B show the cytotoxic activity of nab-rapamycin in combination with AbraxaneTM on vascular smooth muscle cells. Cytotoxicity was evah.iated by staining with ethidiuin homodimer-1 (Fig. 7A) or by staining wit11 calcein (Fig. 7B).

[0046] Figure 8 shows the cytotoxic activity of nab-rapainycin in combination with AbraxaneTM in a HT29 human colon carcinoma xenograft model.
[0047) Figure 9 shows the cytotoxic activity of nab-17-AAG in combination with AbraxaneTM in a H358 human lung carcinoma xenograft model.

DETAILED DESCRIPTION OF THE INVENTION

[0048] The present invention provides methods of combination therapy comprising a first therapy comprising administration of nanoparticles comprising a taxane and a carrier protein (such as albumin) in conjunction with a second therapy such as radiation, surgery, administration of at least one other chemotherapeutic agent, or combinations thereof. The invention also provides methods of metronomic therapy.
[0049] The present invention involves the discovery that AbraxaneTM, due to its superior anti-tumor activity and reduced toxicity and side effects, can be administered in coinbination with other therapeutic drugs and/or treatment modalities and can also be used in metronomic chemotherapy. Due to significantly improved safety profiles with compositions comprising drug/carrier protein nanoparticles (such as AbraxaneTM), we believe that combination chemotherapy with such nanoparticle compositions (such as AbraxaneTM) is more effective than combination chemotherapy with other drugs.
In addition the use of nanoparticle composition (such as AbraxaneTM) in combination with radiation is also believed to be more effective than combination of other agents witli radiation. Thus, the nanoparticle compositions (especially a paclitaxel/albumin nanoparticle composition, such as AbraxaneTM), when used in combination with other chemotherapeutic agents or when combined with other treatment modalities, should be very effective and overcome the deficiencies of surgery, radiation treatment, and cheinotherapy in the treatment of proliferative disease (such as cancer).
[0050] The present invention in one its embodiments is the use of a first therapy comprising a taxane, such as AbraxaneTM, in combination with a second therapy such as another chemotherapeutic agent or agents, radiation, or the like for treating proliferative diseases such as cancer. The first therapy comprising a taxane and second therapy can be administered to a mammal having the proliferative sequentially, or they can be co-adininistered, and even administered simultaneously in the same pha.nnaceutical coinposition.

[0051] Further, a metronomic dosing regime using AbraxaneTM has been found to be more effective than the traditional MTD dosing schedule of the same dnig composition.
Such inetronoinic dosing regime of AbraxaneTM has also been found to be more effective than metronomic dosing of Taxol(D .

[0052] The methods described herein are generally usefiil for treatment of diseases, particularly proliferative diseases. As used herein, "treatment" is an approach for obtaining beneficial or desired clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, any one or more of:
alleviation of one or more symptoms, diminishment of extent of disease, stabilized (i.e., not worsening) state of disease, preventing or delaying spread (e.g., metastasis) of disease, preventing or delaying occurrence or recurrence of disease, delay or slowing of disease progression, amelioration of the disease state, and remission (whether partial or total). Also encompassed by "treatment" is a reduction of pathological consequence of a proliferative disease. The methods of the invention contemplate any one or more of these aspects of treatment.
[0053] As used herein, a "proliferative disease" is defined as a tumor disease (including benign or cancerous) and/or any metastases, wherever the tumor or the metastasis are located, more especially a tumor selected from the group coinprising one or more of (and in some embodiments selected from the group consisting of) breast cancer, genitourinary cancer, lung cancer, gastrointestinal cancer, epidermoid cancer, melanoma, ovarian cancer, pancreatic cancer, neuroblastoma, colorectal cancer, head and neck cancer.
In a broader sense of the invention, a proliferative disease may fiirthermore be selected from hyperproliferative conditions such as hyperplasias, fibrosis (especially pulmonary, but also otlier types of fibrosis, such as renal fibrosis), angiogenesis, psoriasis, atherosclerosis and smooth muscle proliferation in the blood vessels, such as stenosis or restenosis following angioplasty. In some embodiments, the proliferative disease is cancer. In some embodiments, the proliferative disease is a non-cancerous disease. In some einbodiinents, the proliferative disease is a benign or malignant tumor. Where hereinbefore and subsequently a tumor, a tumor disease, a carcinoma or a cancer are mentioned, also metastasis in the original organ or tissue and/or in any other location are implied alternatively or in addition, whatever the location of the tumor and/or metastasis is.
[0054] The tenn "effective amount" used herein refers to an amount of a compound or composition sufficient to treat a specified disorder, condition or disease such as ameliorate, palliate, lessen, and/or delay one or more of its symptoms. In reference to cancers or other unwanted cell proliferation, an effective amotmt comprises an amount sufficient to cause a tumor to shrinlc and/or to decrease the growth rate of the tumor (such as to suppress tumor growth) or to prevent or delay other unwanted cell proliferation. In some embodiments, an effective amount is an amount sufficient to delay development. In some embodiments, an effective amount is an amount sufficient to prevent or delay occurrence and/or rectuTence. An effective amount can be administered in one or more administrations. In the case of cancer, the effective amount of the drug or composition may: (i) reduce the number of cancer cells; (ii) reduce tumor size; (iii) inhibit, retard, slow to some extent and preferably stop cancer cell infiltration into peripheral organs; (iv) inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; (v) inhibit tumor growth;
(vi) prevent or delay occurrence and/or recurrence of tumor; and/or (vii) relieve to some extent one or more of the symptoms associated with the cancer.
[0055] In some embodiments, there is provided a method of treating a primary tumor. In some embodiments, there is provided a method of treating metastatic cancer (that is, cancer that has metastasized from the primary tumor). In some embodiments, there is provided a method of treating cancer at advanced stage(s). In some embodiments, there is provided a method of treating breast cancer (which may be HER2 positive or negative), including, for example, advanced breast cancer, stage IV breast cancer, locally advanced breast cancer, and metastatic breast cancer. In some embodiments, there is provided a method of treating lung cancer, including, for example, non-small cell lung cancer (NSCLC, such as advanced NSCLC), small cell lung cancer (SCLC, such as advanced SCLC), and advanced solid tumor malignancy in the lung. In some embodiments, there is provided a method of treating any of ovarian cancer, head and neck cancer, gastric malignancies, melanoma (including metastatic melanoma), colorectal cancer, pancreatic cancer, and solid tumors (such as advanced solid tumors).
In some embodiments, there is provided a method of reducing cell proliferation and/or cell migration. In some embodiments, there is provided a method of treating any of the following diseases: restenosis, stenosis, fibrosis, angiogenesis, psoriasis, atherosclerosis, and proliferation of smooth muscle cells. The present invention also provides methods of delaying development of any of the proliferative diseases described herein.
[0056] The tenn "individual" is a mainmal, including humans. An individual includes, but is not limited to, human, bovine, horse, feline, canine, rodent, or primate. In some embodiments, the individual is lluman. The individual (such as human) may have advanced disease or lesser extent of disease, such as low ttunor burden. In some embodiments, the individual is at an early stage of a proliferative disease (such as cancer).

In some embodiments, the individual is at an advanced stage of a proliferative disease (such as an advanced cancer). In some embodiments, the individual is HER2 positive. In some embodiments, the individual is HER2 negative.
[0057] The methods may be practiced in an adjuvant setting. "Adjuvant setting"
refers to a clinical setting in which an individual has had a history of a proliferative disease, particularly cancer, and generally (but not necessarily) been responsive to therapy, which includes, but is not limited to, surgery (such as surgical resection), radiotherapy, and chemotherapy. However, because of their history of the proliferative disease (such as cancer), these individuals are considered at risk of development of the disease. Treatment or administration in the "adjuvant setting" refers to a subsequent mode of treatment. The degree of risk (i.e., when an individual in the adjuvant setting is considered as "high risk"
or "low risk") depends upon several factors, most usually the extent of disease when first treated. The methods provided herein may also be practiced in a neoadjuvant setting, i.e., the method may be carried out before the primary/definitive therapy. In some embodiments, the individual has previously been treated. In some embodiments, the individual has not previously been treated. In some embodiments, the treatment is a first line therapy.
[0058] It is understood that aspect and einbodiments of the invention described herein include "consisting" and/or "consisting essentially of' aspects and embodiments.
Combination therapy with chemotherapeutic agent [00591 The present invention provides methods of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual:
a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin); and b) an effective ainount of at least one other chemotherapeutic agent. In some embodiments, the taxane is any of (and in come embodiments consisting essentially of) paclitaxel, docetaxel, and ortataxel.
In some embodiments, the nanoparticle composition comprises AbraxaneTM. In some embodiments, the chemotherapeutic agent is any of (and in some einbodiments selected from the group consisting of) antimetabolite agents (inch.tding nucleoside analogs), platinum-based agents, alkylating agents, tyrosine kinase inl7ibitors, anthracycline antibiotics, vinca alkloids, proteasome inhibitors, macrolides, and topoisomerase inhibitors.
[0060] In some embodiments, the method comprises administering to the individual: a) an effective ainount of a composition comprising nanoparticles comprising paclitaxel and an albuinin; and b) an effective amount of at least one other chemotherapeutic agent. In some einbodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 run. In some embodiments, the paclitaxel/albumin nanoparticle composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the weight ratio of the albumin to paclitaxel in the composition is about 18:1 or less, such as about 9:1 or less. In some embodiments, the paclitaxel is coated with albumin. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 nm and the paclitaxellalbumin composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 iun and the paclitaxel is coated with albumin. In some embodiments, the nanoparticle composition is AbraxaneTM
[0061] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual comprising administering to the individual a) an effective amount of AbraxaneTM, and b) an effective amount of at least one other chemotherapeutic agent. Preferred drug combinations for sequential or co-administration or simultaneous administration with AbraxaneTM are those which show enhanced antiproliferative activity when compared with the single components alone, especially combinations that that lead to regression of proliferative tissues and/or cure from proliferative diseases.
[0062] The chemotherapeutic agents described herein can be the agents themselves, pharmaceutically acceptable salts thereof, and pharmaceutically acceptable esters thereof, as well as steroisomers, enantiomers, racemic mixtures, and the like. The chemotherapeutic agent or agents as described can be administered as well as a pharnnaceutical composition containing the agent(s), wherein the pharmaceutical coinposition comprises a pharmaceutically acceptable carrier vehicle, or the like.
[0063] The cheinotherapeutic agent may be present in a nanoparticle composition.
For example, in some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual: a) an effective ainount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin); and b) an effective amount of a composition comprising nanoparticles comprising at least one other chemotherapeutic agent and a carrier protein (such as albumin). In some embodiments, the method coniprises administering to the individual: a) an effective amount of a coznposition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneT"t); and b) an effective amount of a composition comprising nanoparticles comprising at least one other chemotherapeutic agent and a carrier protein (such as albumin). In some embodiments, the chemotherapeutic agent is any of (and in some embodiments selected from the group consisting of) thiocolchicine or its derivatives (such as dimeric thiocolchicine, including for example ncrb-5404, nab-5800, and nab-5801), rapamycin or its derivatives, and geldanamycin or its derivatives (such as 17-allyl amino geldanamycin (17-AAG)). In some embodiments, the chemotherapeutic agent is rapamycin. In some embodiments, the chemotherapeutic agent is 17-AAG.

[0064] An exemplary and non-limiting list of chemotherapeutic agents contemplated is provided herein. Suitable chemotherapeutic agents include, for example, vinca alkaloids, agents that disi-upt microtubule formation (such as colchicines and its derivatives), anti-angiogenic agents, therapeutic antibodies, EGFR targeting agents, tyrosine kinase targeting agent (such as tyrosine kinase inhibitors), transitional metal complexes, proteasome inhibitors, antimetabolites (such as nucleoside analogs), alkylating agents, platinum-based agents, anthracycline antibiotics, topoisomerase inhibitors, macrolides, therapeutic antibodies, retinoids ( such as all-trans retinoic acids or a derivatives thereof); geldanamycin or a derivative thereof (such as 17-AAG), and other standard chemotherapeutic agents well recognized in the art.
[0065] In some einbodiments, the chemotherapeutic agent is any of (and in some embodiments selected from the group consisting of) adriamycin, coichicine, cyclophosphamide, actinomycin, bleomycin, duanorubicin, doxorubicin, epirubicin, mitomycin, methotrexate, mitoxantrone, fluorouracil, carboplatin, carmustine (BCNU), methyl-CCNU, cisplatin, etoposide, interferons, camptothecin and derivatives thereof, phenesterine, taxanes and derivatives thereof (e.g., paclitaxel and derivatives thereof, taxotere and derivatives thereof, and the like), topetecan, vinblastine, vincristine, tamoxifen, piposulfan, nab-5404, nab-5800, nab-5801, Irinotecan, HIU, Ortataxel, gemcitabine, Herceptin , vinorelbine, Doxilg, capecitabine, AlimtaOO, Avastin0, Velcade0, TarcevaOO, Neulasta , Lapatinib, Sorafenib, derivatives thereof, chemotherapeutic agents known in the art, and the like. In some embodiments, the chemotherapeutic agent is a composition comprising nanoparticles comprising a thiocolchicine derivative and a carrier protein (such as albumin).
[0066] In some embodiments, the chemotherapeutic agent is a antineoplastic agent including, but is not limited to, carboplatin, Navelbine (vinorelbine), anthracycline (Doxil ), lapatinib (GW57016), Herceptin0, gemcitabine (Gemzar RO), capecitabine (XelodaOO ), AliintaOO, cisplatin, 5-fluorouracil, epirubicin, cyclophosphamide, Avastin RO, VelcadeOO, etc.
[0067] In some embodiments, the chemotherapeutic agent is an antagonist of other factors that are involved in tumor growth, such as EGFR, ErbB2 (also known as Herb), ErbB3, ErbB4, or TNF. Sometimes, it may be beneficial to also administer one or more cytokines to the individual. In some embodiments, the therapeutic agent is a growth inhibitory agent. Suitable dosages for the growth inhibitory agent are those presently used and may be lowered due to the combined action (synergy) of the growth inhibitory agent and the taxane.
[0068] In some embodiments, the chemotherapeutic agent is a chemotherapeutic agent other than an anti-VEGF antibody, a HER2 antibody, interferon, and an HGF(3 antagonist.
[0069] Reference to a chemotherapeutic agent herein applies to the chemotherapeutic agent or its derivatives and accordingly the invention contemplates and includes either of these embodiments (agent; agent or derivative(s)).
"Derivatives" or "analogs" of a chemotherapeutic agent or other chemical moiety include, but are not limited to, compotmds that are structurally similar to the chemotherapeutic agent or moiety or are in the same general chemical class as the chemotherapeutic agent or moiety. In some embodiments, the derivative or analog of the chemotherapeutic agent or moiety retains similar chemical and/or physical property (including, for example, fitnctionality) of the cheinotherapeutic agent or moiety.
[0070] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective ainount of a tyrosine kinase inhibitor. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), and b) an effective amount of a tyrosine kinase inhibitor. Suitable tyrosine kinase inhibitors include, for example, imatinib (Gleevec ), gefitinib (Iressa ), Tarceva, Sutent (sunitinib malate), and Lapatinib. In some embodiments, the tyrosine kinase inhibitor is lapatinib. hi some embodiments, the tyrosine kinase inhibitor is Tarceva. Tarceva is a small molecule human epidennal growth factor type 1/epidermal growth factor receptor (HERI/EGFR) inhibitor which demonstrated, in a Phase III clinical trial, an increased survival in advanced non-small cell lung cancer (NSCLC) individuals. In some einbodiments, the method is for treatment of breast cancer, including treatment of metastatic breast cancer and treatment of breast cancer in a neoadjuvant setting. In some embodiments, the method is for treatment of advanced solid tumor. In some embodiments, there is provided a method to iifllibit the proliferation of EGFR expressing tumors in a mammal comprising administering to a mammal infected with such tumors AbraxaneTM and gefitinib, wherein the gefitinib is administered by pulse-dosing.

[00711 In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of an antimetabolite agent (such as a nucleoside analog, including for example purine analogs and pyrimidine analogs). In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), and b) an effective amount of an antimetabolite agent. An "antimetabolic agent" is an agent which is structurally similar to a metabolite, but cannot be used by the body in a productive manner. Many antimetabolite agents interfere with production of nucleic acids, RNA and DNA.
For example, the antimetabolite can be a nucleoside analog, which includes, but is not limited to, azacitidine, azathioprine, capecitabine (Xeloda ), cytarabine, cladribine, cytosine arabinoside (ara-C, cytosar), doxifluridine, fluorouracil (such as 5-fluorouracil), UFT, hydoxyurea, gemcitabine, mercaptopurine, methotrexate, thioguanine (such as 6-thioguanine). Other anti-metabolites include, for example, L-asparaginase (Elspa), decarbazine (DTIC), 2-deoxy-D-glucose, and procarbazine (matulane). In some embodiments, the nucleoside analog is any of (and in some embodiments selected from the group consisting of) gemcitabine, fluorouracil, and capecitabine. In some einbodiments, the method is for treatment of metastatic breast cancer or locally advanced breast cancer.
In some embodiments, the method is for first line treatment of metastatic breast cancer. In some embodiments, the method is for treatment of breast cancer in a neoadjuvant setting.
In some embodiments, the method is for treatment of any of NSCLC, metastatic colorectal cancer, pancreatic cancer, or advanced solid tumor.

[00721 In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a can'ier protein (such as albumin), and b) an effective ainount of an alkylating agent. In sonle embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition coinprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane1 M), and b) an effective amount of an alkylating agent. Suitable alkylating agents include, but are not limited to, cyclophosphamide (Cytoxan), mechlorethamine, chlorambucil, melphalan, carmustine (BCNU), thiotepa, busulfan, alkyl sulphonates, ethylene imines, nitrogen mustard analogs, estramustine sodium phosphate, ifosfainide, nitrosoureas, lomustine, aiid streptozocin. In some embodiments, the alkylating agent is cyclophosphamide. In some embodiments, the cyclophosphainide is administered prior to the administration of the nanoparticle composition. In some embodiments, the method is for treatment of an early stage breast cancer. In some embodiments, the method is for treatment of a breast cancer in an adjuvant or a neoadjuvant setting.
[0073] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles coinprising a taxane and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), and b) an effective amount of a platinum-based agent. Suitable platinum-based agents include, but are not limited to, carboplatin, cisplatin, and oxaliplatin. In some embodiments, the platinum-based ageiit is carboplatin.
In some embodiments, the method is for treatment of: breast cancer (HER2 positive or HER2 negative, including metastatic breast cancer and advanced breast cancer);
lung cancer (including advanced NSCLC, first line NSCLC, SCLC, and advanced solid tumor malignancies in the lung); ovarian cancer; head and neck cancer; and melanoma (including metastatic melanoma).
[0074] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, coinprising administering to the individual a) an effective amount of a coinposition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of an anthracycline antibiotic. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition coinprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective amount of an anthracycline antibiotic. Suitable anthracycline antibiotic include, but are not limited to, Doxil R, actinomycin, dactinomycin, daunorubicin (datmomycin), doxorubicin (adriamycin), epirubicin, idanibicin, mitoxantrone, valrubicin.
In some embodiments, the anthracycline is any of (and in some embodiments selected from the group consisting of) Doxil , epinibicin, and doxorubicin. In some embodiments, the method is for treatinent of an early stage breast cancer. In some embodiments, the method is for treatment of a breast cancer in an adjuvant or a neoadjuvant setting.
[00751 In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of a vinca alkloid. In some einbodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising adininistering to the individual a) an effective amount of a composition comprising nanoparticles coinprising palitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective ainount of a vinca alkloid. Suitable vinca alkaloids include, for example, vinblastine, vincristine, vindesine, vinorelbine (Navelbine ), and VP-16. In some embodiments, the vinca alkaloid is vinorelbine (Navelbine). In some embodiments, the metllod is for treatment of stage IV breast cancer and lung cancer.
[00761 In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective ainount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective ainount of a macrolide.
In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles coinprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective amount of a macrolide. Suitable macrolides include, for exainple, rapamycin, carbomycin, and erythromycin. In some embodiments, the macrolide is rapamycin or a derivative thereof In some embodiments, the method is for treatment of a solid tumor.
[0077] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective ainount of a topoisomerase inhibitor. In some embodiments, the invention provides a inethod of treating a proliferative disease (such as cancer) in an individual, coinprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albinnin), and b) an effective amount of a topoisomerase inhibitor. In some embodiments, the chemotherapeutic agent is a topoisoinerase inhibitor, including, for example, inhibitor of topoisomerase I and topoisomerase II. Exemplary inhibitors of topoisomerase I include, but are not limited to, camptothecin, such as irinotecan and topotecan.
Exemplary inhibitors of topoisomerase II include, but are not limited to, amsacrine, etoposide, etoposide phosphate, and teniposide.

[0078] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of an antiangiogenic agent. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective amount of an antiangiogenic agent. In some embodiments, the method is for treatment of metastatic breast cancer, breast cancer in an adjuvant setting or a neoadjuvant setting, hing cancer (such as first line advanced NSCLC and NSCLC), ovarian cancer, and melanoma (including metastatic melanoma).

[0079] Many anti-angiogenic agents have been identified and are la-lown in the art, including those listed by Canneliet and Jain (2000). The anti-angiogenic agent can be naturally occuning or non-naturally occurring. In some embodiments, the chemotherapeutic agent is a synthetic antiangiogenic peptide. For example, it has been previously reported that the antiangiogenic activity of small synthetic pro-apoptic peptides comprise two functional domains, one targeting the CD 13 receptors (aininopeptidase N) on tumor microvessels and the other disrupting the mitochondrial membrane following intei7lalization. Nat. Med. 1999, 5(9):1032-8. A second generation dimeric peptide, CNGRC-GG-d(KLAKLAK)2, named HKP (Hunter Killer Peptide) was found to have improved antitumor activity. Accordingly, in some embodiments, the antiangiogenic peptide is HKP. In some embodiments, the antiangiogenic agent is other than an anti-VEGF antibody (such as Avastin ).

[0080] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of a proteasome iitllibitor, such as bortezomib (Velcade). In some einbodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective amount of a proteasome inhibitor such as bortezomib (Velcade).

[0081] In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), and b) an effective amount of a therapeutic antibody. In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM) and a carrier protein (such as albumin), and b) an effective amount of a therapeutic antibody. Suitable therapeutic antibodies include, but are not limited to, anti-VEGF antibody (such as Avastin0 (bevacizumab)), anti-HER2 antibody (such as Herceptin (trastuzumab)), Erbitux (cetuximab), Campath (alemtuzumab), Myelotarg (gemtuzumab), Zevalin (ibritumomab tiuextan, Rituxan (rituximab), and Bexxar (tositumomab). In some embodiments, the chemotherapeutic agent is Erbitux0 (cetuximab). In some einbodiments, the chemotherapeutic agent is a therapeutic antibody other than an antibody against VEGF or HER2. In some embodiments, the method is for treatment of HER2 positive breast cancer, including treatment of advanced breast cancer, treatment of metastatic cancer, treatment of breast cancer in an adjuvant setting, and treatment of cancer in a neoadjuvant setting. In some embodiments, the method is for treatment of any of metastatic breast cancer, breast cancer in an adjuvant setting or a neoadjuvant setting, lung cancer (such as first line advanced NSCLC and NSCLC), ovarian cancer, head and neck cancer, and melanoma (including metastatic melanoma).
For example, in some embodiments, there is provided a method for treatment of HER2 positive metastatic breast cancer in an individual, comprising administering to the individual 125 mg/m2 paclitaxel/albumin nanoparticle composition (such as Abraxane r"') weekly for three weeks with the fourth week off, concurrent with the administration of Herceptiii ~t .
[0082] In some embodiments, two or more chemotherapeutic agents are administered in addition to the taxane in the nanoparticle composition. These two or more chemotherapeutic agents may (but not necessarily) belong to different classes of chemotherapeutic agents. Examples of these combinations are provided herein.
Other combinations are also contemplated.
[0083] In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) an effective amount of an antimetabolite (such as a nucleoside analog, for example, gemcitabine), and c) an anthracycline antibiotic (such as epirubicin). In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), b) an effective amount of an antimetabolite (such as a nucleoside analog, for example, gemcitabine), and c) an effective amount of an anthracycline antibiotic (such as epirubicin). In some embodiments, the method is for treatment of breast cancer in a neoadjuvant setting. For example, in some embodiments, there is provided a method of treating locally advanced/inflammatory cancer in an individual comprising administering to the individual 220 mg/mz paclitaxel/albumin nanoparticle composition (such as AbraxaneTM) every two weelcs; 2000 mg/mZ
gemcitabine, every two weeks; and 50 mg/m2 epirubicin, every two weeks. In some embodiments, there is provided a metllod of treating breast cancer in an individual in an adjuvant setting, comprising administering to the individual 175 ing/mZ
paclitaxel/albumin nanoparticle composition (such as AbraxaneTM) every two weeks, 2000 mg/hn2 gemcitabine, every two weeks, and 50 mg/m2 epirubicin, every two weeks.
[0084] In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) an effective amount of a platinum-based agent (such as carboplatin), and c) a therapeutic antibody (such as ant-HER2 antibody (such as HerceptinOO ) and anti-VEGF antibody (such as Avastin )). In some einbodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneT"'), b) an effective ainount of a platinum-based agent (such as carboplatin), and c) a therapeutic antibody (such as ant-HER2 antibody (such as Herceptin ) and anti-VEGF
antibody (such as Avastin(b)). In some embodiments, the method is for treatment of any of advanced breast cancer, metastatic breast cancer, breast cancer in an adjuvant setting, and lung cancer (including NSCLC and advanced NSCLC). In some embodiments, there is provided a method of treating metastatic cancer in an individual, comprising administering to the individual 75 ing/m2 paclitaxel/albumin nanoparticle composition (such as AbraxaneTM) and carboplatin, AUC=2, wherein the administration is carried out weekly for three weeks with the fourth week off. In some embodiments, the method further comprises weekly administering about 2-4 mg/kg of Herception .

[00851 In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) an effective amount of a platinum-based agent (such as carboplatin), and c) a vinca alkaloid (such as Navelbine ). In some embodiments, there is provided a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), b) an effective amount of a platinum-based agent (such as carboplatin), and c) a vinca alkaloid (such as Navelbine ). In some embodiments, the method is for treatment of lung cancer.
[00861 In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) an effective amount of an allcylating agent (such as cyclophosphamide) and c) an anthracycline antibiotic (such as adriamycin). In some embodiments, the invention provides a method of treating a proliferative disease (such as cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin, b) an effective amount of an alkylating agent (such as cyclophosphamide) and c) an anthracycline antibiotic (such as adriamycin). In. some embodiments, the method is for treatment of an early stage breast cancer. In some embodiments, the method is for treatment of a breast cancer in an adjuvant or a neoadjuvant setting. For example, in some embodiments, there is provided a method of treating an early stage breast cancer in an individual, comprising administering 260 mg/m' paclitaxel/albuinin nanoparticle composition (such as AbraxaneTM), 60 mg/m2 adriamycin, and 600 mg/mz cyclophosphamide, wherein the administration is carried out once every two weeks.
[00871 Other embodiments are provided in Table I. For exarnple, in some embodiments, there is provided a method of treating advanced breast cancer in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as AbraxaneTM), b) an effective amount of carboplatin. In some embodiments, the method fiirther comprises adininistering an effective ainount of Herceptin to the individual. In some embodiments, there is provided a method of treating metastatic breast caiicer in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTm), b) an effective amount of gemcitabine. In some embodiments, there is provided a method of treating advanced non-small cell lung cancer in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), b) an effective amount of carboplatin.
[0088] In some embodiments, there is provided a composition coinprising nanoparticles comprising a taxane (such as paclitaxel, docetaxel, or ortataxel) and a carrier protein (such as albi.unin) and at least one other chemotherapeutic agent. The compositions described herein may comprise effective amounts of the taxane and the chemotlierapeutic agent for the treatment of a proliferative disease (such as cancer). In some embodiments, the chemotherapeutic agent and the taxane are present in the coinposition at a predetermined ratio, such as the weight ratios described herein. In some embodiments, the invention provides a synergistic composition of an effective atnount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel, docetaxel, or ortataxel) and an effective amount of at least one other chemotherapeutic agent.

[0089] In some embodiments, the invention provides pharmaceutical compositions comprising nanoparticles comprising a taxane and a carrier protein (such as albumin) for use in the treatment of a proliferative disease (such as cancer), wherein said use comprises simultaneous and/or sequential administration of at least one other chemotherapeutic agent.
In some embodiments, the invention provides a pharinaceutical composition comprising a cheinotherapeutic agent for use in the treatment of a proliferative disease (such as cancer), wherein said use comprises simultaneous and/or sequential administration of a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin). In some embodiments, the invention provides taxane-containing nanoparticle compositions and compositions comprising one otller chemotherapeutic agent for simultaneous, a.nd/or sequential use for treatment of a proliferative disease (such as cancer).

Modes of adnaifaistration [0090] The composition comprising nanoparticles comprising taxane (also referred to as "nanoparticle coinposition") and the chemotherapeutic agent can be administered simultaneously (i.e., simultaneous administration) and/or sequentially (i.e., sequential adininistration).
[0091] In some embodiments, the nanoparticle composition and the chemotherapeutic agent (including the specific chemotherapeutic agents described herein) are administered simultaneously. The term "simultaneous administration," as used llerein, means that the nanoparticle composition and the cheinotherapeutic agent are administered with a time separation of no more than about 15 minute(s), such as no more than about any of 10, 5, or 1 minutes. When the drugs are administered simultaneously, the dnig in the nanoparticles and the chemotherapeutic agent may be contained in the same coinposition (e.g., a composition comprising both the nanoparticles and the chemotherapeutic agent) or in separate compositions (e.g., the nanoparticles are contained in one coinposition and the cheinotherapeutic agent is contained in another composition). For example, the taxane and the chemotherapeutic agent may be present in a single coinposition containing at least two different nanoparticles, wherein some of the nanoparticles in the composition comprise the taxane and a carrier protein, and some of the other nanoparticles in the composition comprise the chemotherapeutic agent and a carrier protein. The invention contemplates and encompasses such compositions. h1 some embodiments, only the taxane is contained in nanoparticles. In some embodiments, siniultaneous administration of the dnig in the
29 nanopar-ticle composition and the chemotherapeutic agent can be combined with supplemental doses of the taxane and/or the chemotherapeutic agent.
[0092] In some embodiments, the nanoparticle composition and the chemotherapeutic agent are administered sequentially. The terin "sequential administration" as used herein means that the drug in the nanoparticle composition and the chemotherapeutic agent are administered with a time separation of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60 or more minutes.
Either the nanoparticle composition or the chemotherapeutic agent may be administered first. The nanoparticle composition and the chemotherapeutic agent are contained in separate compositions, which may be contained in the same or different packages.
[0093] In some embodiments, the administration of the nanoparticle composition and the chernotherapeutic agent are concurrent, i.e., the administration period of the nanoparticle coinposition and that of the chemotherapeutic agent overlap with each other.
In some embodiments, the administration of the iianoparticle composition and the chemotherapeutic agent are non-concLuTent. For example, in some embodiments, the administration of the nanoparticle composition is terminated before the chemotherapeutic agent is administered. In some embodiments, the administration of the chemotherapeutic agent is terminated before the nanoparticle composition is adininistered. The time period between these two non-concurrent administrations can range from about two to eight weeks, such as about four weeks.
[0094] The dosing frequency of the drn.ig-containing nanoparticle composition and the chemotherapeutic agent may be adjusted over the course of the treatment, based on the judgment of the administering physician. When administered separately, the dnig-containing nanoparticle composition and the chemotherapeutic agent can be administered at different dosing fi-equency or intervals. For example, the dnig-containing nanoparticle composition can be administered weekly, while a chemotherapeutic agent can be administered more or less frequently. In some embodiments, sustained continuous release fonnulation of the dnig-containing nanoparticle and/or chemotherapeutic agent may be used. Various formulations and devices for achieving sustained release are known in the art.
[0095] The nanoparticle composition and the chemotherapeutic agent can be administered using the same route of administration or different routes of administration.
In some embodiments (for both simultaneous and sequential administrations), the taxane in the nanoparticle composition and the chemotherapeutic agent are administered at a predetennined ratio. For example, in some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the chemotherapeutic agent is about 1 to 1. In some embodiments, the weight ratio may be between about 0.001 to about 1 and about 1000 to about 1, or between about 0.01 to about 1 and 100 to about 1. In some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the chemotherapeutic agent is less than about any of 100:1, 50:1, 30:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, and 1:1 In some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the chemotherapeutic agent is more than about any of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 30:1, 50:1, 100:1. Other ratios are contemplated.
[0096] The doses required for the taxane and/or the chemotherapeutic agent may (but not necessarily) be lower than what is normally required when each agent is administered alone. Thus, in some embodiments, a subtherapeutic amount of the dnig in the nanoparticle composition and/or the chemotherapeutic agent are adininistered.
"Subtherapeutic amount" or "subtherapeutic level" refer to an amount that is less than the therapeutic amount, that is, less than the amount normally used when the drug in the nanoparticle composition and/or the chemotherapeutic agent are administered alone. The reduction may be reflected in terms of the amount administered at a given administration and/or the amount administered over a given period of time (reduced frequency).
[0097] In some embodiments, enough chemotherapeutic agent is administered so as to allow reduction of the normal dose of the drug in the nanoparticle composition required to effect the same degree of treatment by at least about any of 5%, 10%, 20%,
30%, 50%, 60%, 70%, 80%, 90%, or more. In some embodiments, enougli drug in the nanoparticle composition is administered so as to allow reduction of the normal dose of the chemotherapeutic agent required to effect the same degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more.
[0098] In some embodiments, the dose of both the taxane in the nanoparticle coinposition and the chemotherapeutic agent are reduced as compared to the coi-responding normal dose of each when administered alone. In some embodiments, both the taxane in the nanoparticle composition and the chemotherapeutic agent are administered at a subtherapeutic, i.e., reduced, level. In some enlbodiments, the dose of the nanoparticle coinposition and/or the chemotherapeutic agent is substantially less than the established maximuin toxic dose (MTD). For example, the dose of the nanoparticle composition and/or the chemotherapeutic agent is less than about 50%, 40%, 30%, 20%, or 10% of the MTD.
31 [0099] A combination of the administration configurations described herein can be used. The combination therapy methods described herein may be perfonned alone or in conjunction with another therapy, such as surgery, radiation, chemotherapy, immunotherapy, gene therapy, and the like. Additionally, a person having a greater risk of developing the proliferative disease may receive treatments to inhibit or and/or delay the development of the disease.
j0100] As will be understood by those of ordinary skill in the art, the appropriate doses of chemotherapeutic agents will be approximately those already employed in clinical therapies wllerein the chemotherapeutic agent are administered alone or in combination with other chemotherapeutic agents. Variation in dosage will likely occur depending on the condition being treated. As described above, in some embodiments, the chemotherapeutic agents may be administered at a reduced level.
[0101] The nanoparticle colnpositions described herein can be administered to an individual (such as human) via various routes, such as parenterally, including intravenous, intra-arterial, intraperitoneal, intrapulmonary, oral, inhalation, intravesicular, illtrainuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, or transdermal. For example, the nanoparticle composition can be administered by illhalation to treat conditions of the respiratory tract. The composition can be used to treat respiratory conditions such as pulmonary fibrosis, broncheolitis obliterans, lung cancer, bronchoalveolar carcinoma, and the like. In some embodiments, the nanoparticle composition is administrated intravenously. In some embodiments, the nanoparticle composition is administered orally.
[0102] The dosing frequency of the administration of the nanoparticle composition depends on the nature of the combination therapy and the particular disease being treated.
An exeinplary dosing frequency include, but is not limited to, weelcly without brealc;
weelcly, three out of four weelcs; once every tllree weeks; once every two weeks; weekly, two out of three weeks. See also Table 1.
[0103] The dose of the taxane in the nanoparticle composition will vary witll the nature of the combination tlzerapy and the particular disease being treated.
The dose should be sufficient to effect a desirable response, such as a therapeutic or prophylactic response against a particular disease. An exemplary dose of the taxane (in some embodiments paclitaxel) in the nanoparticle composition include, but is not limited to, about ally of 50 mg/1n2, 60 nlg/1112, 75 mg/m2, 80 mg/mZ, 90 mg/rn2, 100 mg/m2, 120 mg/ma, 160 mg/mZ, 175 mg/m2 , 200 mg/m2, 210 mg/ma, 220 mg/m', 260 mg/m2, and 300 mg/m2. For exanlple, the dosage of paclitaxel in a nanoparticle composition can be in the range of
32 mg/m2 when given on a 3 week schedule, or 50-250 mg/1-1-12 when given on a weekly schedule. See also Table 1.
[0104] Other exeinplary dosing schedules for the administration of the nanoparticle composition (such as paclitaxel/albumin nanoparticle composition, for example AbraxaneTM) include, but are not limited to, 100 mg/mZ, weekly, without break;
75 mg/m2 weekly, 3 out of four weeks; 100 ing/m2, weekly, 3 out of 4 weeks; 125 mg/m2, weekly, 3 out of 4 weeks; 125 mg/m2, weekly, 2 out of 3 weeks;' 130 mg/mZ, weekly, without brealc;
175 mg/mZ, once every 2 weeks; 260 mg/m2, once every 2 weeks; 260 mg/mZ, once every 3 weeks; 180-300 mg/ma, every three weeks; 60-175 mg/m2, weekly, without break.
In addition, the taxane (alone or in combination therapy) can be administered by following a metronomic dosing regime described herein.
[0105] Exemplary dosing regimes for the combination therapy of nanoparticle composition (such as paclitaxel/albumin nanoparticle coinposition, for exainple AbraxaneTM) and other agents include, but are not limited to, 125 mg/m2 weekly, two out of three weeks, plus 825 mg/m2 Xeloda , daily; 260 mg/mZ once every two weeks, plus 60 mg/mZ adriamycin and 600 mg/m2 cyclophosphamide, once every two weeks; 220-340 mg/m2 once every three weeks, ph.is carboplatin, AUC=6, once every three weeks; 100-150 mg/mZ weelcly, plus carboplatin, AUC=6, once every three weelcs; 175 mg/m2 once every two weeks, plus 2000 mg/mZ gemcitabine and 50 mg/m2 epirubicin, once every two weeks;
and 75 mg/m2 weekly, tliree out of four weeks, plus carboplatin, AUC=2, weekly, three out of four weeks.
[0106] In some embodiments, the nanoparticle composition of the taxane and the chemotherapeutic agent is administered according to any of the dosing regimes described in Table 1.
[01071 In some embodiments, there is provided a metliod of treating breast cancer in an individual comprising administering to the individual: a) an effective amotuit of a composition comprising nanoparticles coinprising a taxane (such as paclitaxel) and an albumin, and b) an effective amoLUlt of at least one other chemotherapeutic agent as provided in Rows 1 to 35 in Table 1. In some embodiments, the administration of the nanoparticle cornposition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 1 to 35 in Table 1. In some embodiments, there is provided a method of treating metastatic breast cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other JJ

chemotherapeutic agent as provided in Rows 2, 4-8, and 10-15 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 2, 4-8, and 10-15 in Table 1.
[01081 In some embodiments, there is provided a method of treating advanced breast cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective ainount of at least one other chemotherapeutic agent as provided in Rows I and 16 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 1 and 16 in Table 1. In some embodiments, there is provided a method of treating stage IV breast cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Row 3 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be the dosing regime as indicated in Row 3 in Table 1.
[0109] In some embodiments, there is provided a method of treating breast cancer in an individual in an adjuvant setting comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 18 to 24 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regiines as indicated in Rows 18 to 24 in Table 1.
[0110] In some embodiments, there is provided a method of treating breast cancer in an individual in a neoadjuvant setting coinprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 25 to 35 in Table 1. In some embodiments, the administration of the nanoparticle coinposition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 25 to 35 in Table 1.
[0111] In some embodiments, there is provided a method of treating lung cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albunain, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 36 to 48 in Table 1. In some embodiments, the administration of the nanoparticle composition and the clleinotherapeutic agent may be any of the dosing regimes as indicated in Rows 36 to 48 in Table 1.
[01121 In some embodiments, there is provided a method of treating NSCLC
(including advanced NSCLC and first line NSCLC) in an individual comprising administering to the individual: a) an effective amoLuit of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeLrtic agent as provided in Rows 36-40 and 42-43 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 36-40 and 42-43 in Table 1. In some embodiments, there is provided a method of treating advanced solid tumor malignancy in the lung in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Row 41 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be the dosing regimes as indicated in Row 41 in Table 1. In some embodiments, there is provided a method of treating SCLC in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Row 48 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be the dosing regimes as indicated in Row 48 in Table 1.
[0113] In some embodiments, there is provided a method of treating ovarian cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 49 to 52 in Table 1. In some embodiments, the administration of the na.noparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 49 to 52 in Table 1.
[0114] In some embodiments, there is provided a method of treating head and neclc cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles coinprising a taxane (such as paclitaxel) and an albtunin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 53 to 55 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 53 to 55 in Table 1.
[0115] In some embodiments, there is provided a method of treating solid tumor (including advanced solid tumor) in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 56 to 59 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 56 to 59 in Table 1.
[0116] In some embodiments, there is provided a method of treating melanoma (including metastatic melanoma) in an individual comprising administering to the individual: a) an effective ainount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 60-63 in Table 1. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 60 to 63 in Table 1.
[01171 In some embodiments, there is provided a method of treating metastatic colorectal cancer in an individual comprising administering to the individual:
a) an effective amount of a composition coinprising nanoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Row 64 in Table 1. In some embodiments, the administration of the nanoparticle coinposition and the chemotherapeutic agent may be the dosing regime as indicated in Row 64 in Table 1.
[01181 In some einbodiments, there is provided a lnetliod of treating pancreatic cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising iianoparticles comprising a taxane (such as paclitaxel) and an albumin, and b) an effective amount of at least one other chemotherapeutic agent as provided in Rows 65 to 66 in Table 1. In some embodiments, the adininistration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 65 to 66 in Table 1.

Row Gon7bination Regime/Dosage Study therapy Protocol title No. type Row Conibination Regime/Dosage Study tlierapy Protocol title No. type A phase II study of ABX: 100 mgJm'' D1, 8, 15 weekly dose-dense q4wk x 6 nanoparticle paclitaxel ABX + Carbo: AUC = 2 D 1, 8, 15 Advanced (ABI-007) Carboplatin + q4wk x 6 HER2+ Breast carboplatinTM, with Herceptin0 Cancer HereeptinO as first or Herceptin0: 4 mg/kg on wk 1, 2 second-line tlierapy of mg/lcg all subsequent weeks advanced HER2+
breast cancer Phase II trial of weelcly AbraxaneTM
2 ABX alone ABX: 125 mg/rn' Metastatic monotherapy for 1st-(+Herceptin0) qwk x 3/4 Breast Cancer line MBC (plus Herceptin in HER2+
pts) Ll: ABX: 80 mg/m Nav: 15 mg/rn' L2: ABX: 90 mg/m'' Nav: 20 mg/m2 L3: ABX: 100 mg/mz Phase I-II study weekly ABX + Stage IV ABX + Navelbine0, 3. Navelbine0 Nav: 22.5 mghnZ with or without G-CSF, (:~G-CSF) L4: ABX: 1 10 mg/,m Breast Cancer in stage IV breast cancer Nav: 25 mg/m2 L5: ABX: 125 mg/mz Nav: 25 n1g/m' qwk all levels ABX: 125 mg/mZ qwk x 2/3 Metastatic 4. ABX ~ Xeloda0 Phase II lst-line ABX +
Xeloda0: 825 mg/mz D1-14 Breast Cancer Xeloda0 MBC trial q3wk ABX + Metastatic Phase I/II trial ABX
5. Anthrac plus Doxil for MBC
Ycline Breast Cancer plus limited PK

Randomized Phase II
Trial of Weekly nab (nanoparticle albumin AI3X: 125 mg/rn2 bound)-Paclitaxel (nab-ABX + Metastatic paclitaxel) in 6. Gemcitabine Gem: 1000 mg/m2 Breast Cancer Combination with qwlc x 2/3 Gemcitabine in Patients with HER2 Negative Metastatic Breast Cancer ABX + Metastatic Phase 1/II AbraxaneTM
7. La atinib + GW572016 p Breast Cancer ABX: 100 m/mZ x 3/4 Phase I dose escalation ABX + g qwk Metastatic study of a 2 day oral 8' Lapatinib Lapatinib: starting at 1000 mg/d lapatinib x 2 days Breast Cancer chemosensitization Row Combination Regime/Dosage Study therapy Protocol title No. type pulse given prior to weekly intravenous AbraxaneTM in patients with advanced solid tumors Z Phase II preoperative ABX: 220 mg/m q2wk x 6 trial of AbraxaneTM
ABX +FEC followed by followed by FEC
9. Breast Cancer (+Herceptin ) FEC: 4 cycles (+Herceptin for (+gercePtm as HER2+ pts) appropriate) in breast cancer ABX: 100 mg/mZ qwlc D1, 8, 15 Metastatic Phase II safety and ABX + tolerability study of Carbo: AUC = 2 qwlc D1, 8, 15 Breast Cancer AbraxaneTM, Avastin 10. Carboplatin + z and carbo latin in tri le Avastin Avastin : 10 mg/m q2wk (HER2-, ER-, p p PR ) negative metastatic breast cancer patients ABX: 130 mg/mZ qwk + Avastin vs ABX + ABX: 260 mg/mz q2w1c Metastatic Three ann phase II trial 11. Avastin in 15C line HER2-+ Avastin@ Breast Cancer negative MBC patients vs ABX: 260 mg/m2 q3wk + Avastin Single arm study of ABX + ABX: 125 mg/mZ qwlc x 3/4 Metastatic AbraxaneTM and 12. Avastin@ Avastin in l st line + Avastin Breast Cancer MBS
ABX + Avastin qwk Randomized Phase III
ABX + Metastatic trial in l st line and 2"d 13. vs line MBC with Avastin@ Breast Cancer Taxol(i] + AvastinJ qwk biological correlates analysis Phase II AbraxaneTM in ABX + Xeloda Metastatic combination with 14. XelodaO and Lapatinib + Lapatinib Breast Cancer for metastatic breast cancer Single ABX + ABX: 3000 mg/m2 Dl q3wk Metastatic arm Phase 15. study of AbraxaneTM
Gemcitabine Gem: 1250 mg/m' Dl, 8 q3wk Breast Cancer and gemcitabine for 1s1 line MBC
Phase I/II study of AbraxaneTM in 16. ABX + RAD001 Advanced coinbination with Breast Cancer RAD001 in patients with advanced breast cancer Row Conlbination Regime/Dosage Study therapy Protocol title No, type Phase I study of 17. ABX + Sutent~li? Breast Cancer AbraxaneTM in combination with SutentCR
AC + G-CSF q2wk x 4 ABX + AC + G followed b AbraxaneT"t in dose-18. CSF (+ y Breast Cancer- dense adjuvant Herceptinn) ABX: 260 mg/mZ q2wlc x 4 Adjuvant chemotherapy for early stage breastcancer (+ HerceptincD for HER2+ pts) Dose dense AC + G-CSF -ABX + AC + G followed Phase II pilot adjuvant 19. CSF (+ by ABX Breast Cancer- ~ial of AbraxaneTM in Herceptinr~) (+ Herceptin for HER2+ pts) Adjuvant breast cancer qwk AC followed by ABX: 260 ing/m'-20. ABX + AC vs Breast Cancer- Adjuvant Dose dense Adjuvant Registrational Trial AC followed by Taxol@
Rx length 16 wks AC q2wk followed by Phase II dose dense ABX + AC ABX: 260 mg/m'' +G-CSF Breast Cancer- pilot adjuvant study of 21. (+G-CSF) q2wk Adjuvant AbraxaneTM in breast Rx lengtli 16 wks cancer ABX + AC Dose dense AC followed by 22 Breast Cancer- Pilot adjuvant breast (+ Avastint) ABX (+ Avastin in HER2+ Adjuvant cancer study AC
Breast Cancer- BIG study: Dose dense 23. ABX + AC followed by ABX Adjuvant vs standard adjuvant q2wlc or q3wk chemotherapy Phase II - Pilot Study Evaluating the Safety of a Dose-Dense Regime -AC
ABX (ABI 007) AC followed by x 4=> ABI-007 x 4 24. + AC + Breast Cancer - Q 2 WEEKS +
Neulasta~ ABX q2wk x 4 Adjuvant NeulastaCC - Given as Adjuvant Chemotherapy of High-Risk Women with Early Breast Cancer ABX: 100 mg/mZ qwlc x 12 A Phase II Study of followed by Neoadjuvant 5-FU: 500 mg/m2 q3wk Chemotherapy witli Locally Sequential Weekly 25. ABX +FEC Epinlbicin: 100mg/m2 Advanced Breast Nanoparticle Albumin (+HerceptinCR)) Cancer- Bound Paclitaxel (without Herceptin(D) Neoad'uvant J (AbraxaneTni) Followed or by 5-Fluorouracil, Z Epinibicin, Epinibicin: 75 mg/m Cyclophosphamide Row Combination Regime/Dosage Study therapy Protocol title No. type (with Herceptin R for HER2+ (FEC) in Locally pts) Advanced Breast Cyclophosphamide: 500 mg/m' Cancer q3wk Arm 1: Neoadjuvant: Gem: 2000 Phase II Trial of Dose mg/m', ABX: 175 mg/m', Epi Dense Neoadjuvant 50 mg/m' ABX + Gemcitabine, 26. Gemcitabine + q2wk x 6 Breast Cancer - Epiiubicin, ABI-007 E inibicin Neoadjuvant (GEA) in Locally p Arm 2: Adjuvant: Gem: 2000 Advanced or mghn', ABX: 220 mg/mz Inflammatory Breast q2wlc x 4 Cance ABX: 260 mg/m' q2wk +
ABX + HerceptinfR) 27 Breast Cancer - Phase II Multi-center Herceptin followed by Neoadjuvant study neoadjuvant.
Navelbinet~J + Herceptin(D
TAC
ABX + vs 3 arms Randomized Carboplatin dose dense phase II
28 AC followed by ABX + carbo Breast Cancer - trial of neoadjuvant (+ Herceptin(R) vs Neoadjuvant chemotherapy in +AC patients with breast AC followed by ABX + carbo + cancer Herceptin R, ABX: 260 mg/m' q3wk x 4 Phase II neoadjuvant 29. ABX + Breast Cancer - trial of AbraxaneTnt and Capecitabine XelodaCR) 850 mg/nl' D1-14 Neoadjuvant capecitabine in locally q3wk x 4 advanced breast cancer Phase I/II trial of neoadjuvant chemotherapy (NCT) ABX + ABX qwk with weekly 30. Carboplatin carbo qwk Breast Cancer - nanoparticle paclitaxel Neoadjuvant (ABI-007, AbraxaneTM) (+ Avastin ) + Avastirn R in HER2+ pts in combination witli carboplatin and Avastin R in clinical stage I-III.
Phase II study of ABX: 100 mg/m'- qwlc x 3/4 weekly bevacizumab administered with ABX + Carbo: AUC = 5 weekly trastuzumab, Carboplatin + Breast Cancer - ABI-007, and 31. Herce tinC7 + + Herceptin~~
p Neoadjuvant carboplatin as Avastin + Avastin(_1k) preoperative therapy in 4 week cycle x 6 HER2-neu gene amplified breast cancer tumors ABX: 260 m/m' Pilot neoadjuvant trial ABX + g q3wk Breast Cancer - with conzbination of 32. Lapatinib Neoad'uvant ABI-007 AbraxaneT"' Lapatinib: 1000 mg/day J ( ) and GW572016 Row 'Combination Regime/Dosage Study therapy Protocol title No. type (Lapatinib) ABX: 200 mg/hn' Phase II neoadjuvant ABX + q3wk x 4 Breast Cancer - trial of AbraxaneTM and
33. Ca ecitabine ~ Neoad'uvant ca ecitabine in locally p Xeloda : 1000 mg/m- J p advanced breast cancer D1-14 q3wlc x 4 Phase III trial of ABX qwk + Avastin(g) followed paclitaxel vs AgX + by A qwk + C daily AbraxaneTM with or
34. Avastin + AC vs Breast Cancer - without AvastinOO in Neoadjuvant combination with (+ G-CSF) Taxol qwlc f Avastin doxorubicin and followed by A qwk + C daily cyclophosphamide plus G-CSF
Breast Cancer - Phase II neoadjuvant
35. ABX + AC ABX followed by AC Neoadjuvant trial with gene expression analyses An open label phase II
ABX: 300 mg/m' q3wlc trial of AbraxaneTM
ABX + Carbo: AUC = 6 q3wlc 15' line carboplatin and ~
36. Carboplatin + Advanced Avastin in patients Avastin Avasting: 15 mg/kg NSCLC with advanced non-4 cycles squamous non-small cell lung cancer Ll: ABX: 225 ing/m' L2: ABX: 260 mg/m' L3: ABX: 300 mg/m'' Phase II toxicity pilot ABX + Advanced study of AbraxaneTM
37. Carboplatin Cohorts 1-4: ABX q3wk NSCLC and carboplatin in Cohorts 5-7: ABX weekly advanced non-small cell lung cancer.
Cohort 8: 75 additional patients Carbo fixed at AUC = 6 q3wk Carbo: AUC = 6 + ABX
38 ABX + vs 15f line NSCLC Phase III Registration -Carboplatin Carbo: AUC = 6 + Taxol : 225 NSCLC 1st line therapy mg/in' ABX: 100 mg/m'- dl, 8, 15 Phase II Trial of weekly
39 ABX + Carbo: AUC = 6 q4wk lst line NSCLC AbraxaneTM plus Carboplatin Amendnzent: ABX: 125 mg/m' carboplatin in lst-line D1, 8, 15 NSCLC
ABX +
40. Carboplatin + Weekly NSCLC
AvastniOO

Arm 1: ABX: 100, 125,150 Lung Cancer - Phase I Trial of ABX + mg/mz D 1, 8, 15 q4wk Advanced Solid carboplatin and
41. Carboplatin Arm 2: ABX 220, 260, 300, 340 Tumor AbraxaneTM on a m/m' 3wlc Malignancy weekly and every tluee g q3wk schedule in Row Combination Regime/Dosage Study therapy Protocol title No. type Arm 3: ABX 100, 125, 150 patients with Advanced mg/rn2 D l, 8 Solid Tuinor Carbo: AUC = 6 in all arms Malignancies ABX + AbraxaneTM in
42. Gemcitabine or NSCLC combination with ABX + gemcitabine or Avastin Avastin@
Phase I trial of
43. ABX + NSCLC AbraxaneTM in Gemcitabine combination with gemcitabine ABX: 225, 260, 300 mg/mZ Phase I/II study of ABX + AbraxaneTM and Carbo: AUC = 6 carboplatin AUC 6,
44. Carboplatin + Lung Cancer Avastin q3wk plus Avastin (Standard 3+3 Phase I
+ Avastin design; Phll: 40 pts) ABX: 220, 260, 300 mg/m' Phase I/II study of
45. ABX + Alimta q3wk Lung Cancer AbraxaneTM + Alimta Pemtrexed: 500mg q3wk for 2nd-line NSCLC
Phase I/II trial of
46. ABX + Cisplatin Ltulg Cancer AbraxaneTM plus cisplatin in advanced NSCLC
Phase I/II study of ABX + AbraxaneTM,
47. Navelbine@ + Lung Cancer Navelbine , and Cisplatin Cisplatin for treatment of advanced NSCLC
Phase II trial of ABX + ABX: 300 ing/m'' q3wk AbraxaneTM and
48. SCLC carboplatin in extensive Carboplatin Carbo: AUC =6 q3wk stage small cell lung cancer A phase II trial of ABX + ABX: 100 mg/mZ qwk x 3/4 Ovarian AbraxaneTM +
49. Carboplatin Carbo: AUC = 6 Cancer Carboplatin in recurrent ovarian cancer ABX: qwlc PhaseT~tudy of ABX + Abraxane plus carbo
50. Carboplatin ABX: q3w Ovarian Cancer for treatment of Carbo: AUC = 6 both arms advanced ovarian cancer 1 st line, optimally ABX: TBD by ABI-CA034 debulked, registration ABX + vs trial. Carbo AUC 6 +
51. Ovarian Cancer ABX vs Carbo +
Carboplatin Taxo1C~ 175 mg/mZ Taxol 175 mg/m2.
Carbo: AUC = 6 in botli arms Endpoint: relapse free survival, survival Row Combination Regime/Dosage Study therapy Protocol title No. type Phase II study of bevacizumab with ABX: 100 m1m2 x 3/4 Ovarian Cancer AbraxaneTM in patients
52. ABX + g qwk with recurrent, Avastin(RD Avastin : 10mg/m' q2wk platinum resistant priniary epithelial ovarian or primary peritoneal carcinoma ABX: D 1 Ui-iresectable localized 5-FU: 750 mg/mZ CIV x 5 head and neck cancer
53. ABX + 5-FU + Head and Neck Phase II AbraxaneTM in Cisplatin cisplatin: 75 mglm2 D1 Cancer coznbination witl2 5-FU
followed by XRT/surgery and cisplatin 5-FU: 750 mg/m' CIV x 5 Unresectable localized ABX + 5-FU + cisplatin: 75 mglmz D1 Head and Neck head and neck cancer
54. Cisplatin ~ ABX DI Cancer Phase III 5-FU and cisplatin with or without followed by XRT/surgery AbraxaneTM
Phase II multicenter trial of AbraxaneTM in ABX + Head and Neck combination with
55. Cetuximab Cancer cetuximab in 1 line treatment of locally advanced or metastatic head and neck cancer Phase I Study of ABX: 100mg/m2 qwk Rapainycin in
56. A BX + Solid Tumors Combination with Ra am cin Rapamycin: 5-40 mg dose p y escalation AbraxaueTM in Advanced Solid Tumors ABX + Pl7ase I trial of
57. Satraplatin Solid Tumors AbraxaneTM and Satraplatin ABX: 180, 220, 260, 300, 340 Phase I Trial of ABX + mg/m' q3wk Advanced Solid AbraxaneTM in
58. Gemcitabine 2 Tumors combination with Gemcitabine: 1000mg1m D1 and D8 Gemcitabine ABX: 100 m/m2 wk x 3/4 Phase I dose escalation g qwk Solid study of gefitinib
59. ABX +Gefitinib Gefitinib starting at 1000 mg/d x 'r1,1111ors chemosensitization pulse 2 given prior to weekly AbraxaneTM
Phase II study of
60. ABX + Metastatic AbraxaneTM and AvastinOP Melanoma AvastinOO in metastatic melanoma AbraxaneTM and ABX + Mela AvastinL as therapy for
61. Avastin~ noma patients with malignant melanoma Row Conibination Regime/Dosage Study therapy Protocol title No. type Phase II sh.idy of
62. ABX + Metastatic AbraxaneTM and Carboplatin Melanoma carboplatin in nietastatic melanoma Phase II study of ABX + ABX: qwk AbraxaneTM in
63. Sorafenib + Sorafenib: D2-19 Metastatic combination with Carboplatin Melanoma carboplatin and Carbo: AUC = 6 Dl sorafenib in metastatic melanoma Metastatic phase II trial of Colorectal AbraxaneTM in Cancer (after combination with
64. ABX + failure of Xeloda(:R) for previously Capecitabine oxaliplatin- treated patient with based therapy advance or inetastatic and irinotecan- colorectal cancer based therapy) Phase I study of ABX + Pancreatic AbraxaneTM in
65. Gemcitabine Weekly combination with Cancer gemcitabine in pancreatic cancer ABX + Gem ABX + Pancreatic Phase III registration trial
66. Gemcitabine vs Cancer in pancreatic cancer Gem ABX + anti- AbraxaneT'K combined
67. angiogenic with anti-angiogenic agents agents, e.g. Avastin AbraxaneTM combined ABX + with proteasome
68. proteasome inhibitors, e.g.
inhibitors V elcade AbraxaneTM combined
69. ABX + EGFR with EGFR inliibitors, inhibitors e.g. Tarceva0 [0119] As used in herein (for example in Table 1), ABX refers to AbraxaneTM;
GW572016 refers to lapatinib; Xel refers to capecitabine or Xeloda RO;
bevacizumab is also Icnown as Avastin ; trastuzumab is also lcnown as Herceptin ; pemtrexed is also lulown as Alimta ; cetuximab is also known as Erbitux@; gefitinib is also known as IressaO; FEC
refers to a combination of 5-fluorouracil, Epirubiciii and Cyclophosphainide;
AC refers to a combination of Adriamycin plus Cyclophosphainide; TAC refers to a FDA approved adjuvant breast cancer regime; RAD001 refers to a derivative of rapamycin;
NSCLC refers to non-small cell 1tu1g cancer; and SCLC refers to small cell hing cancer.

[0120] As used herein (for example in Table 1), AUC refers to area under curve;
q4wk refers to a dose every 4 weeks; q3wk refers to a dose every 3 weeks; q2wk refers to a dose every 2 weeks; qwk refers to a weekly dose; qwlc x 3/4 refers to a weekly dose for 3 weeks with the 4th week off; qwk x 2/3 refers to a weekly dose for 2 weeks with the 3 rd weelc off.

Combination therapy with radiation therapy and surgery [0121] In another aspect, the present invention provides a method of treating proliferative disease (such as cancer) comprising a first therapy comprising administering a taxane (particularly nanoparticles comprising a taxane) and a carrier protein and a second therapy comprising radiation and/or surgery.
[0122] In some embodiments, the method comprises: a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising an effective amount of a taxane and a carrier protein (such as albumin) and b) a second therapy comprising radiation therapy, surgery, or combinations thereof. In some einbodiments, the taxane is coated with the carrier protein (such as albumin).
In some embodiments, the second therapy is radiation therapy. In some embodiments, the second therapy is surgery.
[0123] In some embodiments, the method comprises a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising paclitaxel and an albumin; and b) a second therapy coinprising radiation therapy, surgery, or combinations thereof In some embodiments, the second therapy is radiation therapy. In some embodiments, the second therapy is surgery. In some embodiments, the paclitaxel/albumin nanoparticles have an average diaineter of no greater than about 200 nm. In some embodiments, the paclitaxel/albumin nanoparticle coinposition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the weight ratio of the albumin to paclitaxel in the coinposition is about 18:1 or less, such as about 9:1 or less. In some embodiments, the paclitaxel is coated with albuniin. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 run and the paclitaxel/albumin composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the paclitaxel/albumin nanoparticles have an average diaineter of no greater than about 200 nm and the paclitaxel is coated with albumin. In some embodiments, the nanoparticle coinposition is AbraxaneTM

[0124] The administration of the nanoparticle composition may be prior to the radiation and/or surgery, after the radiation and/or surgery, or concurrent with the radiation and/or surgery. For example, the administration of the nanoparticle composition may precede or follow the radiation and/or surgery therapy by intervals ranging from minutes to weelcs. In some embodiments, the time period between the first and the second therapy is such that the taxane and the radiation/surgery would still be able to exert an advantageously combined effect on the cell. For example, the taxane (such as paclitaxel) in the nanoparticle composition may be administered less than about any of 1, 3, 6, 9, 12, 18, 24, 48, 60, 72, 84, 96, 108, 120 hours prior to the radiation and/or surgery. In some embodiments, the nanoparticle composition is administered less than about 9 hours prior to the radiation and/surgery. In some embodiments, the nanoparticle composition is administered less than about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 days prior to the radiation/surgery. In some embodiments, the taxane (such as paclitaxel) in the nanoparticle composition is administered less than about any of 1, 3, 6, 9, 12, 18, 24, 48, 60, 72, 84, 96, 108, or 120 hours after the radiation and/or surgery. In some embodiments, it may be desirable to extend the time period for treatment significantly, where several days to several weeks lapse between the two therapies.
[0125] Radiation contemplated herein includes, for example, -y-rays, X-rays (external beam), and the directed delivery of radioisotopes to tumor cells.
Other forms of DNA damaging factors are also contemplated such as microwaves and UV
irradiation are also contemplated. Radiation may be given in a single dose or in a series of small doses in a dose-fractionated schedule. The amount of radiation contemplated herein ranges from about I to about 100 Gy, including, for example, about 5 to about 80, about 10 to about 50 Gy, or about 10 Gy. The total dose may be applied in a fractioned regime. For example, the regime may comprise fractionated individual doses of 2 Gy. Dosage ranges for radioisotopes vary widely, and depends on the half-life of the isotope and the strength and type of radiation emitted.
[0126] When the radiation comprises use of radioactive isotopes, the isotope may be conjugated to a targeting agent, such as a therapeutic antibody, which carries the radionucleotide to the target tissue. Suitable radioactive isotopes include, but are not limited to, astatine211, 14carbon, 51chromium, 36chlorine,57iron, 58cobalt, copper 67, 152Eu, gallium 67, ;hydrogen, iodine123, iodine13', indium' 11, 51ion, 32phosphoilis, rheniuml86, 7Sselenium, 35sulphur, technicium9'~"', and/or yttrium90.

[0127] In some embodiments, enough radiation is applied to the individual so as to allow reduction of the normal dose of the taxane (such as paclitaxel) in the nanoparticle composition required to effect the same degree of treatment by at least about any of 5%, 10%, 20 fo, 30%, 50%, 60%, 70%, 80%, 90%, or more. In some embodiments, enough taxane in the nanoparticle composition is administered so as to allow reduction of the nonnal dose of the radiation required to effect the saine degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more. In some embodiments, the dose of both the taxane (such as paclitaxel) in the nanoparticle composition and the radiation are reduced as coinpared to the corresponding noimal dose of each when used alone.

[0128] In some embodiments, the combination of administration of the nanoparticle composition and the radiation therapy produce supra-additive effect. In some embodiments, the taxane (such as paclitaxel) in the nanoparticle composition is administered once at the dose of 90 mg/lcg, and the radiation is applied five times at 80 Gy daily.

[0129] Surgery described herein includes resection in which all or part of cancerous tissue is physically removed, exercised, and/or destroyed. Tumor resection refers to physical removal of at least part of a tumor. In addition to tumor resection, treatment by surgery includes laser surgery, cryosurgery, electrosurgery, and micropically controlled surgery (Mohs surgery). Removal of superficial surgery, precancers, or normal tissues are also contemplated.

[0130] The radiation therapy and/or surgery may be carried out in addition to the administration of chemotherapeutic agents. For example, the individual may first be administered with a taxane-containing nanoparticle composition and at least one otlier chemotherapeutic agent, and subsequently be subject to radiation therapy and/or surgery.
Alternatively, the individual may first be treated with radiation therapy and/or surgery, which is then followed by the administration of a nanoparticle composition and at least one other chemotherapeutic agent. Other combinations are also contemplated.

[0131] Administration of nanoparticle compositions disclosed above in conjunction with administration of chemotherapeutic agent is equally applicable to those in conjunction with radiation therapy and/or surgery.

[0132] In some embodiments, the nanoparticle composition of the taxane and/or the chemotherapeutic agent is administered in conjunction with radiation according to any of the dosing regimes described in Table 2.

[0133] In some embodiments, there is provided a method of treating NSCLC in an individual comprises a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising taxane (such as paclitaxel) and an albumin; and b) a second tlierapy comprising radiation as provided in Rows I
to 5 in Table 2. hi some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 1 to 5 in Table 2.
[0134] In some embodiments, there is provided a method of treating head and neck cancer in an individual coinprises a) a first therapy comprising administering to the individual a composition coinprising nanoparticles comprising taxane (such as paclitaxel) and an albumin; and b) a second tlierapy coinprising radiation as provided in Rows 6 to 9 in Table 2. In some embodiments, the administration of the nanoparticle coinposition and the chemotherapeutic agent may be any of the dosing regimes as indicated in Rows 6 to 9 in Table 2.
[0135] In some embodiments, there is provided a method of treating pancreatic cancer in an individual comprises a) a first therapy comprising adininistering to the individual a composition comprising nanoparticles comprising taxane (such as paclitaxel) and an albumin; and b) a second therapy comprising radiation as provided in Row 10 in Table 2. In some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be the dosing regixnes as indicated in Row 10 in Table 2.
[0136] In some embodiments, there is provided a method of treating gastric malignancies in an individual comprises a) a first therapy comprising administering to the individual a composition comprising nanoparticles comprising taxane (such as paclitaxel) and an albuinin; and b) a second therapy comprising radiation as provided in Row 11 in Table 2. ln some embodiments, the administration of the nanoparticle composition and the chemotherapeutic agent may be the dosing regimes as indicated in Rowl 1 in Table 2.

Row Combination Regime/Dosage Study therapy Protocol title No. type ABX + Phase 1/11 trial of I Radiation NSCLC AbraxaneTM combined with radiation ABX + Phase 1/II trial of 2 Carboplatin + NSCLC AbraxaneTM and Radiation carboplatin combined with radiation.
3 ABX + 1 cycle ABX/Carbo induction NSCLC Phase II chenioradiation Row Combination Regime/Dosage Study therapy Protocol title No. type Carboplatin + followed by in NSCLC
Radiation 2 or 3 times weekly pulse ABX
+ radiation AbraxaneTM
ABX + /carboplatin induction 4 Carboplatin + NSCLC followed by Radiation AbraxaneTM + radiation in stage III A&B PS2 NSCLC patients ABX + ABX qwlc + carbo + radiation Carboplatin + followed by NSCLC Phase II study Radiation ABX q3wk + carbo ABX + Head and AbraxaneTM as a 6 Radiation Neck Cancer radiosensitizer in head and neck cancer ABX + Head and PhaseI/II AbraxaneTM
7 Cetuximab + Neck Cancer in combination with Radiation cetuximab and radiation Induction: ABX 135 mg/mz Phase I/II study of qwk + carbo: AUC = 2 induction chemotherapy ABX + with AbraxaneTM and Carboplatin + followed by carboplatin followed by 8 5-FU + Concurrent chemoradiation: Head and concomitant Hydroxyurea + ABX: 100 mg/m' Neck Cancer fluorouracil, Radiation hydroxyurea, 5-FU: 600 mg/hn, AbraxaneTM and IMRT
hydroxyurea: 5000 mg BID for locally advanced head and neck cancers ABX: 20-50 mg/m'' qwk x 7 Phase I trial of dose escalation AbraxaneTM in ABX + Locally combination with 9 Carboplatin + EribituxO: 400 mg/m' day 7, Advanced carboplatin, cetuximab ErbituxC~ + 250 mg/m'' qwk x 7 Head and and IMRT in locally Radiation Carbo: AUC = 1.5 qwk x 7 Neck Cancer advanced squamous cell cancer of the head and IMRT neck A randomized phase II
trial of weekly ABX + gemcitabine, Gemcitabine + qwk Pancreatic AbraxaneTM, and Radiation Cancer external irradiation for locally advanced pancreatic cancer Phase I/II combination ABX + of AbraxaneT"t/cisplatin 11 Cisplatin + Gastric and radiation for Radiation Malignancies patients witli resected gastric/GEJ
malignancies.
[0137] In some embodiments, the invention provides pharmaceutical compositions comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin) for use in the treatment of a proliferative disease (such as cancer), wherein said use comprises a second therapy comprising radiation therapy, surgery, or combinations thereof.

Metronomic therapy [0138] The invention also provides metronomic therapy regime. There is provided a method of administering to an individual a composition comprising nanoparticles comprising a taxane (such as paclitaxel, docetaxel, or ortataxel) and a carrier protein (such as albumin) based on a metronomic dosing regime. The methods are applicable to methods of treatment, delaying development, and other clinical settings and configurations described herein. For example, in some embodiments, the methods are useful for treatment of proliferative diseases (such as cancer).
[0139] "Metronomic dosing regime" used herein refers to frequent administration of a taxane at without prolonged brealcs at a dose below the established maximum tolerated dose via a traditional schedule with brealcs (hereinafter also referred to as a "standard MTD
schedule" or a "standard MTD regime"). In metronomic dosing, the same, lower, or higher cumulative dose over a certain time period as would be administered via a standard MTD
schedule may ultimately be administered. In some cases, this is achieved by extending the time frame and/or frequency during which the dosing regime is conducted while decreasing the amount administered at each dose. Generally, the taxane administered via the metronomic dosing regime of the present invention is better tolerated by the individual.
Metronomic dosing can also be referred to as maintenance dosing or chronic dosing.
[0140] In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, there is provided a method of adininistering a composition comprising nanoparticles comprising paclitaxel and an albumin, wherein the nanoparticle composition is administered over a period of at least one month, wherein the inteival between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25% to about 25 '0 of its maximum tolerated dose following a traditional dosing regime.

[0141] In some embodiments, the dosing of the taxane (such as paclitaxel) in the nanoparticle composition per administration is less than about any of 1%, 2%, 3&, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 18%, 20%, 22%, 24%, or 25% of the MTD for the same taxane (such as paclitaxel) in the same formulation following a given traditional dosing schedule. Traditional dosing schedule refers to the dosing schedule that is generally established in a clinical setting. For example, the tradition dosing schedule for AbraxaneTM is a three-weekly schedule, i.e., administering the composition every three weeks.
[0142] In some embodiments, the dosing of the taxane (such as paclitaxel) per administration is between about 0.25% to about 25% of the corresponding MTD
value, including for example any of about 0.25% to about 20%, about 0.25% to about 15%, about 0.25% to about 10%, about 0.25% to about 20%, and about 0.25% to about 25%, of the corresponding MTD value. The MTD value for a taxane following a traditional dosing schedule is known or can be easily detennined by a person skilled in the art.
For example, the MTD value when AbraxaneTM is administered following a traditional three-week dosing schedule is about 300 mg/m2.
[0143] In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more thali about a week, and wherein the dose of the taxane at eac11 administration is about 0.25 mg/m2 to about 25 mg/m'. .h-i some embodiments, there is provided a method of adininistering a composition comprising nanoparticles comprising paclitaxel and an albumin, wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25 mg/mz to about 25 mg/m2.
[0144] In some embodiments, the dose of the taxane (such as paclitaxel) at each administration is less than about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 20, 22, 25, and 30 mg/m2. For example, the dose of the taxane (such as paclitaxel) can range from about 0.25 mg/m2 to about 30 mg/m2, about 0.25 mg/m2 to about 25 mg/m2, about 0.25 mg/tn' to about 15 mg/mz, about 0.25 mg/mz to about 10 mg/m', and about 0.25 mg/m2 to about 5 mg/mz.
[0145] Dosing frequency for the taxane (sucli as paclitaxel) in the nanoparticle composition includes, but is not liinited to, at least about any of once a week, twice a week, three times a week, four times a week, five times a week, six times a week, or daily.
Typically, the interval between each administration is less than about a week, such as less than about any of 6, 5, 4, 3, 2, or 1 day. In some embodiments, the interval between each administration is constant. For example, the administration can be carried out daily, every two days, every three days, every four days, every five days, or weekly. In some embodiments, the administration can be carried out twice daily, three times daily, or more frequent.

[0146] The metronomic dosing regimes described herein can be extended over an extended period of time, such as from about a month up to about three years.
For example, the dosing regime can be extended over a period of any of about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, and 36 months. Generally, tllere are no brealcs in the dosing schedule.
[0147] The cumulative dose of the taxane (such as paclitaxel) administered by the metronomic regime may be higher than that of the taxane administered according to a standard MTD dosing schedule over the same time period. In some embodiments, the cumulative dose of the taxane administered by the metronomic regime equals to or is lower than that of the taxane administered according to a standard MTD dosing schedule over the same time period.

[0148] It is understood that the teaching provided herein is for examples only, and that metronomic dosing regime can be routinely designed in accordance with the teachings provided herein and based upon the individual standard MTD schedule, and that the metronomic dosing regime used in these experiments merely serves as one example of possible changes in dosing interval and duration which are made to a standard MTD
schedule to arrive at an optimal metronomic dosing regime.
[0149] The metronoinic dosing regime described herein may be used alone as a treatment of a proliferative disease, or carried out in a combination therapy context, such as the combination therapies described herein. In some embodiments, the metronomic therapy dosing regime may be used in combination or conjunction with other established therapies administered via standard MTD regimes. By "combination or in conjunction with" it is meant that the inetronomic dosing regime of the present invention is conducted either at the same time as the standard MTD regime of established therapies, or between courses of induction therapy to sustain the benefit accilied to the individual by the induction therapy, the intent is to continue to iidiibit tumor growth while not unduly compromising the individual's health or the individual's ability to withstand the next course of induction therapy. For example, a metronomic dosing regime may be adopted after an initial short course of MTD chemotherapy.
[0150] The nanopai-ticle compositions administered based on the metronomic dosing regime described herein can be administered to an individual (such as human) via various routes, such as parenterally, including intravenous, intra-arterial, intrapulmonary, oral, inhalation, intravesicular, intrainuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, or transdetmal. For example, the nanoparticle composition can be administered by inhalation to treat conditions of the respiratory tract. The composition can be used to treat respiratory conditions such as pulmonary fibrosis, broncheolitis obliterans, lung cancer, bronchoalveolar carcinoma, and the like. In some embodiments, the nanoparticle composition is administered orally.
[0151] Some various exemplary embodiments are provided below.
[0152] In some embodiments, there is provided a method of administering a composition coinprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the taxane is coated with the carrier protein (such as albumin). In some embodiments, the dose of the taxane per administration is less than about any of 1%, 2%, 3%, 4%, 5%, 6%, 7%, 8%, 9%, 10%, 11%, 12%, 13%, 14%, 15%, 18%, 20%, 22%, 24%, or 25% of the maximum tolerated dose. In some embodiments, the taxane is administered at least about any of lx, 2x, 3x, 4x, 5x, 6x, 7x (i.e., daily) a week. In some embodiments, the intervals between each administration are less than about any of 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, and 1 day. In some embodiments, the taxane is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30 and 36 months.
[0153] In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising paclitaxel and an albumin, wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 iun. In some embodiments, the paclitaxel/albumin nanoparticle composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the weight ratio of the albumin to paclitaxel in the composition is about 18:1 or less, such as about 9:1 or less. In some embodiments, the paclitaxel is coated with albumin. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 zun and the paclitaxel/albumin composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the paclitaxel/albumin nanoparticles have an average diaineter of no greater than about 200 nm and the paclitaxel is coated with albumin. In some einbodiments, the nanoparticle composition is AbraxaneTM.
[0154] In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25 mg/m2 to about 25 mg/mZ. In some einbodiments, the taxane is coated with the carrier protein (such as albumin). In some embodiments, the dose of the taxane per administration is less than about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 18, 20, 22, and 25 mg/m2. In some embodiments, the taxane is adininistered at least about any of lx, 2x, 3x, 4x, 5x, 6x, 7x (i.e., daily) a weelc. In some embodiments, the intervals between each administration are less than about any of 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, and 1 day. In some embodiments, the taxane is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30 and 36 months.
[0155] In some embodiments, there is provided a method of administering a composition comprising nanoparticles comprising paclitaxel and an albumin, wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane at each administration is about 0.25 mg/mZ to about 25 mg/m2. In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 mn. In some embodiments, the paclitaxel/albumin nanoparticle composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the weiglit ratio of the albumin to paclitaxel in the composition is about 18:1 or less, such as about 9:1 or less. In some embodiments, the paclitaxel is coated with albumin. In some elnbodiments, the paclitaxel/albumin nanopai-ticles have an average diameter of no greater than about 200 run and the paclitaxel/albi.unin composition is substantially free (such as free) of surfactant (such as Cremophor). In some embodiments, the paclitaxel/albumin nanoparticles have an average diameter of no greater than about 200 nm and the paclitaxel is coated with albumin. In some embodiments, the nanoparticle composition is AbraxaneTM
[0156] Ihi some embodiments, the Abraxane"M(or other paclitaxeUalbumin nanoparticle compositions) is administered at the dose of about 3 mg/kg to about 10 mg/lcg daily. In some einbodiments, the AbraxaneTM is administered at the dose of about 6 mg/kg to about 10 mg/kg daily. In some embodiments, the AbraxaneTM is administered at the dose of about 6 mg/kg daily. In some embodiments, AbraxaneTM is administered at the dose of about 3 mg/lcg daily.
[0157] The invention also provides compositions for use in the metronomic regime(s) described herein. In some embodiments, there is provided a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), wherein said coinposition is administered to an individual via a inetronomic dosing regiine, such as the dosing regime described herein.

Other aspects of the invention [0158] In another aspects, there are provided methods of treating proliferative diseases comprising administering a composition comprising nanoparticles comprising a taxane (including pacltiaxel, docetaxel, or ortataxel) and a carrier protein (such as albumin). In some embodiments, there is provided a metliod of treating cancer comprising administering a composition coinprising nanoparticles comprising ortataxel and a carrier protein (such as albumin).
[0159] In some embodiments, there is provided methods of treating proliferative diseases comprising administering a composition comprising nanoparticles comprising a thiocolchicine or its derivative (such as dimeric thiocolchicine) and a carrier protein (such as albumin). In some embodiments, tllere is provided a method of treating cancer comprising administering a coinposition comprising nanoparticles comprising dimeric colchicines and a carrier protein (such as albumin). In some embodiments, the nanoparticle composition is any of (and in some einbodiments selected from the group consisting of) Nab-5404, Nab-5800, and Nab-5801.
[0160] In some embodiments, there is provided a method of treating cancer comprising administering a composition comprising nanoparticles comprising paclitaxel, wherein the nanoparticle composition is administered according to any of the dosing regimes described in Table 3. In some embodiments, the cancer is a Taxane refractory metastatic bI-east cancer.

Row Combination Regimen/Dosage Study therapy Protocol title No. type Phase lI study with l. ABX alone ABX: 125 mg/mz qwk x 3/4 Metastatic weekly AbraxaneTn' Breast Cancer treatment in taxane-refractory MBC patients Arm 1: ABX 130 mg/mz qwk 2. ABX alone Arm 2: ABX 260 mg/mz q2wk Metastatic 3-arm phase II trial in 1st-Breast Cancer line Her-2- MBC patients.
Arm 3: ABX 260 mg/mz q3wk Phase II Controlled, Randomized, Open Label Study to Evaluate the ABX: 260 mg/mz q3w1c Efficacy and Safety of ABX alone Metastatic Capxol (a Ci-emophor-3. vs Breast Cancer Free Nanoparticle (Capxol) z Paclitaxel) and Taxol: 175 mglm q3wk cremophor-formulated paclitaxel injection in Patient with Metastatic Breast Cancer Arm 1: ABX weekly 3-arm phase Il trial in 1st-4. ABX alone Arm 2: ABX q3w1c Metastatic line and 2nd-line MBC, Breast Cancer with biological correlates Arm 3: Taxol weelcly analysis Phase 11 trial of neoadjuvant Stage IIA, IIB, chemotherapy (NCT) with z IIIA IIIB and nanoparticle paclitaxel 5. ABX alone ABX: 300 mg/m q3wk IV breast (ABI-007, Abraxane) in women with clinical stage cancer IIA, IIB, IIIA, IIIB and IV
(with intact primary) breast cancers 1 st-line Phase 1/II study of 6. ABX alone ABX: 125 mg/m'' qwlc x 3/4 advanced Abraxane nionotherapy in NSCLC ]st-line advanced NSCLC
ABX alone ABX 260 mg/mz Phase II ABX mono in 7. q3wk 1 st-line NSCLC 1 st-line NSCLC
Arm 1: ABX q3wk Phase 11 study of õd Abraxane monotherapy in 8. ABX alone Arm 2: ABX qwk 2 line NSCLC 2"d_line NSCLC
Doses TBD
ABX: 100mg/mz qwlc Randomized phase 11 9. ABX alone vs Prostate Cancer study AbraxaneTM weelcly vs every three weeks in ABX: 260 mg/mz q3wk fi=ont line HRP
Phase II ABX in lst-line 10. ABX alone ABX qwk Prostate Cancer prostate cancer I L ABX alone cycles 150 mg/rn2 qwk x 3/4 for 2 Prostate Cancer Phase II
S~udo,adjuvant 12. ABX alone ABX: 100 mg/mz qwlc (no brealc) Prostate Cancer Pli ase ll ABX
100 mg Row Combination Regimen/Dosage Study therapy Protocol title No. type weekly no break ABX: 100 mg/mZ (previously treated) Phase 11 previously treated 13. ABX alone ABX: 150 mg/m, (untreated) Malignant Melanoma and untreated rrrnetastatic melanonia patients qwk x 3/4 ABX: 125 m/m2 Phase II study of ABX in 14. ABX alone g Carcinoma of treatment of persistent or qwk x 3/4 the cervix recurrent carcinoma of the cervix Phase 11 study of 15. ABX alone Ovarian Cancer Abraxane for treatment of advanced ovarian cancer (3rd line) Phase II single ti-eatment ABX alone non-hematologic use of ABI-007 for the 16. treatment of non-(ABI-007) malignancies hematologic malignancies.
Compassionate tise Nanoparticle compositions [0161] The nanoparticle compositions described herein comprise nanoparticles comprising (in various embodiments consisting essentially of) a taxane (such as paclitaxel) and a carrier protein (such as albumin). Nanoparticles of poorly water soluble dnigs (such as taxane) have been disclosed in, for example, U.S. Pat. Nos. 5,916,596;
6,506,405; and 6,537,579 and also in U.S. Pat. Pub. No. 200510004002A1. Although the description provided below is specific to taxane, it is understood that the same applies to other drugs, such as rapamycin, 17-AAG, and dimeric thiocolchicine.
[0162] In some embodiments, the composition comprises nanoparticles with an average or inean diameter of no greater than about 1000 nanometers (nm), such as no greater than about any of 900, 800, 700, 600, 500, 400, 300, 200, and 100 nm.
In solne embodiments, the average or mean diameters of the nanoparticles is no greater than about 200 nm. In some embodiments, the average or mean diameters of the nanoparticles is no greater than about 150 nm. In some embodiments, the average or mean dialneters of the nanoparticles is no greater than about 100 nm. In some embodiinents, the average or mean diameter of the nanoparticles is about 20 to about 400 mn. In some embodiments, the average or mean diameter of the nanoparticles is about 40 to about 200 iuii.
In some embodiments, the nanoparticles are sterile-filterable.
[0163] The nanoparticles described herein may be present in a dry forniulation (such as lyophilized composition) or suspended in a biocompatible medium.
Suitable biocompatible media include, but are not limited to, water, buffered aqueous media, saline, buffered saline, optionally buffered solutions of amino acids, optionally buffered solutions of proteins, optionally buffered solutions of sugars, optionally buffered solutions of vitamins, optionally buffered solutions of synthetic polyiners, lipid-containing emulsions, and the like.

[0164] The tenn "proteins" refers to polypeptides or polymers of amino acids of any length (including ftill length or fragments), which may be linear or branched, comprise modified amino acids, and/or be interrupted by non-amino acids. The term also encompasses an amino acid polymer that has been modified naturally or by intervention;
for example, disulfide bond fonnation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification. Also included within this tenn are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, -unnatural amino acids, etc.), as well as other modifications known in the art.
The proteins described herein may be naturally occurring, i.e., obtained or derived from a natural source (such as blood), or synthesized (such as chemically synthesized or by synthesized by recombinant DNA techniques).

[0165] Examples of suitable carrier proteins include proteins normally found in blood or plasma, which include, but are not limited to, albumin, immunoglobulin including IgA, lipoproteins, apolipoprotein B, alpha-acid glycoprotein, beta-2-inacroglobulin, thyroglobulin, transferin, fibronectin, factor VII, factor VIII, factor IX, factor X, and the like. In some embodiments, the carrier protein is non-blood protein, such as casein, a-lactalbumin, and (.3-lactoglobulin. The carrier proteins may either be natural in origin or synthetically prepared. In some embodiments, the phannaceutically acceptable carrier comprises albumin, such as human serum albumin. Human serum albumin (HSA) is a highly soluble globular protein of M, 65K and consists of 585 amino acids. HSA
is the most abundant protein in the plasma and accounts for 70-80 % of the colloid osmotic pressure of human plasma. The amino acid sequence of HSA contains a total of disulphide bridges, one free thiol (Cys 34), and a single tryptophan (Trp 214). Intravenous use of HSA solution has been indicated for the prevention and treatment of hypovolumic shock (see, e.g., Tullis, JAMA, 237, 355-360, 460-463, (1977)) and Houser et al., Szlrgerjl, Gynecology and Obstetrics, 150, 811-816 (1980)) and in conjunction with exchange transftision in the treatment of neonatal hyperbilirubinemia (see, e.g., Finlayson, Seiriinars in Tlzr=oTizbosis and Hemostasis, 6, 85-120, (1980)). Other albumins are contemplated, such as bovine serum albumin. Use of such non-human albuinins could be appropriate, for exainple, in the context of use of these compositions in non-lnunan mainmals, such as the veterinary (including domestic pets and agricultural context).
[0166] Human senim albtunin (HSA) has multiple hydrophobic binding sites (a total of eight for fatty acids, an endogenous ligand of HSA) and binds a diverse set of taxanes, especially neutral and negatively charged hydrophobic compounds (Goodinan et al., The PharrnRacological Basis of Tlzerapeutics, 9lh ed, McGraw-Hill New York (1996)).
Two high affinity binding sites have been proposed in subdomains IIA and IIIA
of HSA, wllich are highly elongated hydrophobic pockets with charged lysine and arginine residues near the surface which fiinction as attachment points for polar ligand features (see, e.g., Fehske et al., Biochern. Phar-mcol., 30, 687-92 (198a), Vorum, Dan. Med.
Bull., 46, 379-99 (1999), Kragli-Hansen, Dn. Med. Bull., 1441, 131-40 (1990), Curry et al., Ncct. Struct.
Biol., 5, 827-35 (1998), Sugio et al., Protein. Eng., 12, 439-46 (1999), He et al., Nature, 358, 209-15 (199b), and Carter et al., Adv. Pi-otein. Cheiiz., 45, 153-203 (1994)). Paclitaxel and propofol have been shown to bind HSA (see, e.g., Paal et al., Eur. J.
Bioch.ena., 268(7), 2187-91 (200a), Purcell et al., Biochim. Biophys. Acta, 1478(a), 61-8 (2000), Altmayer et al., Afzneinaittelforschung, 45, 1053-6 (1995), and Garrido et al., Rev. Esp.
Anestestiol.
Reanim., 41, 308-12 (1994)). In addition, docetaxel has been shown to bind to human plasma proteins (see, e.g., Urien et al., Invest. New Di-ztgs, 14(b), 147-51 (1996)).
[0167] The carrier protein (such as albumin) in the composition generally serves as a carrier for the taxane, i.e., the carrier protein in the composition makes the taxane more readily suspendable in an aqueous medium or helps maintain the suspension as compared to compositions not comprising a carrier protein. This can avoid the use of toxic solvents (or surfactants) for solubilizing the taxane, and thereby can reduce one or more side effects of administration of the taxane into an individual (such as a human). Thus, in some embodiments, the composition described herein is substantially free (such as free) of surfactants, such as Cremophor (including Cremophor EL (BASF)). In some embodiments, the nanoparticle composition is substantially free (such as free) of surfactants. A composition is "substantially free of Cremophor" or "substantially free of surfactant" if the amount of Cremophor or surfactant in the composition is not sufficient to cause one or more side effect(s) in an individual when the nanoparticle composition is administered to the individual.
[0168] The ainount of carrier protein in the composition described herein will vaiy depending on other components in the coinposition. In some embodiments, the composition comprises a carrier protein in an ainount that is sufficient to stabilize the taxane in an aqueous suspension, for example, in the fonn of a stable colloidal suspension (such as a stable suspension of nanoparticles). In some embodiments, the carrier protein is in an aniount that reduces the sedimentation rate of the taxane in an aqueous medium. For particle-containing coinpositions, the amount of the carrier protein also depends on the size and density of nanoparticles of the taxane.

[0169] A taxane is "stabilized" in an aqueous suspension if it remains suspended in an aqueous medium (such as without visible precipitation or sedimentation) for an extended period of time, such as for at least about any of 0.1, 0.2, 0.25, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or 72 hours. The suspension is generally, but not necessarily, suitable for administration to an individual (such as human). Stability of the suspension is generally (but not necessarily) evaluated at a storage temperature (such as room temperature (such as 20-25 C) or refrigerated conditions (such as 4 C)). For example, a suspension is stable at a storage temperature if it exhibits no flocculation or particle agglomeration visible to the nalced eye or when viewed under the optical microscope at 1000 times, at about fifteen minutes after preparation of the suspension.
Stability can also be evaluated under accelerated testing conditions, such as at a temperature that is higher than about 40 C.

[0170] In some embodiments, the carrier protein is present in an amount that is sufficient to stabilize the taxane in an aqueous suspension at a certain concentration. For example, the concentration of the taxane in the composition is about 0.1 to about 100 mg/ml, including for exainple any of abo.ut 0.1 to about 50 mg/ml, about 0.1 to about 20 mg/ml, about 1 to about 10 mg/mi, about 2 mg/ml to about 8 mg/ml, about 4 to about 6 mg/ml, about 5 mg /ml. In some embodiments, the concentration of the taxane is at least about any of 1.3 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/inl, 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mghnl, 25 mg/ml, 30 mg/ml, 40 ing/ml, and 50 ing/ml. In some embodiments, the carrier protein is present in an ainount that avoids use of surfactants (such as Creinophor), so that the composition is free or substantially fi-ee of surfactant (such as Cremophor).

[0171] In some embodiments, the composition, in liquid form, comprises from about 0.1% to about 50% (w/v) (e.g. about 0.5% (w/v), about 5% (w/v), about 10% (w/v), about 15% (w/v), about 20% (w/v), about 30% (w/v), about 40% (w/v), or about 50%
(w/v)) of can=ier protein. In some embodiments, the composition, in liquid fonn, comprises about 0.5% to about 5% (w/v) of carrier protein.

[0172] In some embodiments, the weigllt ratio of carrier protein, e.g., albumin, to the taxane in the nanoparticle composition is such that a sufficient amount of taxane binds to, or is transpoi-ted by, the cell. While the weight ratio of carrier protein to taxane will have to be optimized for different carrier protein and taxane combinations, generally the weight ratio of carrier protein, e.g., albumin, to taxane (w/w) is about 0.01:1 to about 100:1, about 0.02:1 to about 50:1, about 0.05:1 to about 20:1, about 0.1:1 to about 20:1, about 1:1 to about 18:1, about 2:1 to about 15:1, about 3:1 to about 12:1, about 4:1 to about 10:1, about 5:1 to about 9:1, or about 9:1. In some embodiments, the carrier protein to taxane weight ratio is about any of 18:1 or less, 15:1 or less, 14:1 or less, 13:1 or less, 12:1 or less, 11:1 or less, 10:1 or less, 9:1 or less, 8:1 or less, 7:1 or less, 6:1 or less, 5:1 or less, 4:1 or less, and 3:1 or less.
[0173] In some embodiments, the carrier protein allows the composition to be administered to an individual (such as human) without significant side effects. In some embodiments, the carrier protein (such as albumin) is in an amount that is effective to reduce one or more side effects of administration of the taxane to a liuman.
The term "reducing one or more side effects of administration of the taxane" refers to reduction, alleviation, elimination, or avoidance of one or more undesirable effects caused by the taxane, as well as side effects caused by delivery vehicles (such as solvents that render the taxanes suitable for injection) used to deliver the taxane. Such side effects include, for example, myelosuppression, neurotoxicity, hypersensitivity, inflainmation, venous irritation, phlebitis, pain, skin irritation, peripheral neuropathy, neutropenic fever, anaphylactic reaction, venous thrombosis, extravasation, and combinations thereof. These side effects, however, are merely exemplary and other side effects, or combination of side effects, associated with taxanes can be reduced.
[0174] In some embodiments, the composition comprises AbraxaneTM. AbraxaneTM
is a formulation of paclitaxel stabilized by human albumin USP, wliich can be dispersed in directly injectable physiological solution. When dispersed in a suitable aqueous medium such as 0.9% sodium chloride injection or 5% dextrose injection, AbraxaneTM
forms a stable colloidal suspension of paclitaxel. The mean particle size of the nanoparticles in the colloidal suspension is about 130 nanometers. Since HSA is freely soluble in water, AbraxaneTM can be reconstituted in a wide range of concentrations ranging from dilute (0.1 mg/inl paclitaxel) to concentrated (20 mg/inl paclitaxel), including for example about 2 ing/ml to about 8 mg/ml, about 5 mg/ml.

[0175] Methods of making nanoparticle compositions are known in the art. For example, nanoparticles containing taxanes (such as paclitaxel) and carrier protein (such as albumin) can be prepared under conditions of high shear forces (e.g., sonication, high pressure homogenization, or the like). These methods are disclosed in, for example, U.S.
Pat. Nos. 5,916,596; 6,506,405; and 6,537,579 and also in U.S. Pat. Pub. No.
2005/0004002A1.

[0176] Briefly, the taxane (such as docetaxel) is dissolved in an organic solvent, and the solution can be added to a human serum albumin solution. The mixture is subjected to high pressure homogenization. The organic solvent can then be removed by evaporation. The dispersion obtained can be fiirther lyophilized. Suitable organic solvent include, for example, ketones, esters, ethers, chlorinated solvents, and other solvents known in the art. For example, the organic solvent can be methylene chloride and chloroform/ethanol (for exainple with a ratio of 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, or 9:a).

Other components in the nanoparticle compositions [0177] The nanopai-ticles described herein can be present in a composition that include other agents, excipients, or stabilizers. For example, to increase stability by increasing the negative zeta potential of nanoparticles, certain negatively charged components may be added. Such negatively charged components include, but are not limited to bile salts of bile acids consisting of glycocholic acid, cholic acid, chenodeoxycholic acid, taurocholic acid, glycochenodeoxycholic acid, taurochenodeoxycholic acid, litocholic acid, ursodeoxycholic acid, dehydrocholic acid and others; phospholipids including lecithin (egg yolk) based phospholipids which include the following phosphatidylcholines: palmitoyloleoylphosphatidylcholine, palmitoyllinoleoylphosphatidylcholine, stearoyllinoleoylphosphatidylcholine stearoyloleoylphosphatidylcholine, stearoylarachidoylphosphatidylcholine, and dipalmitoylphosphatidylcholine. Other phospholipids including L-a-dimyristoylphosphatidylcholine (DMPC), dioleoylphosphatidylcholine (DOPC), distearyolphosphatidylcholine (DSPC), hydrogenated soy phosphatidylcholine (HSPC), and other related compounds. Negatively charged surfactants or emulsifiers are also suitable as additives, e.g., sodium cholesteryl sulfate and the like.

[0178] In some embodiments, the coinposition is suitable for administration to a human. In some embodiments, the composition is suitable for administration to a mammal such as, in the veterinary context, domestic pets and agricultural animals.
There are a wide variety of suitable formulations of the nanoparticle composition (see, e.g., U.S. Pat. Nos.
5,916,596 and 6,096,331). The following formulations and methods are merely exemplary and are in no way limiting. Formulations suitable for oral administration can consist of (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules, (c) suspensions in an appropriate liquid, and (d) suitable emulsions. Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
[0179] Examples of suitable carriers, excipients, and diluents include, but are not limited to, lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, water, saline solution, syrup, methylcellulose, methyl- and propylhydroxybenzoates, talc, magnesium stearate, and mineral oil. The formulations can additionally include lubricating agents, wetting agents, emulsifying and suspending agents, preserving agents, sweetening agents or flavoring agents.
[0180] Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, wliich can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation compatible with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizers, and preservatives. The formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient, for example, water, for injections, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. Injectable formulations are preferred.

[01811 In some embodiments, the composition is fornlulated to have a pH range of about 4.5 to about 9.0, including for example pH ranges of any of about 5.0 to about 8.0, about 6.5 to about 7.5, and about 6.5 to about 7Ø In some embodiments, the pH of the composition is formulated to no less than about 6, including for example no less than about any of 6.5, 7, or 8 (such as about 8). The composition can also be made to be isotonic with blood by the addition of a suitable tonicity modifier, such as glycerol.

Kits [0182] The invention also provides kits for use in the instant methods. Kits of the invention include one or more containers comprising taxane-containing nanoparticle compositions (or unit dosage forms and/or articles of manufacture) and/or a chemotherapeutic agent, and in some embodiments, ftirther comprise instnictions for use in accordance with any of the methods described herein. The kit may fiirther comprise a description of selection an individual suitable or treatment. Instnictions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instnictions (e.g., instnictions carried on a magnetic or optical storage disk) are also acceptable.
[01831 In some embodiments, the kit comprises a) a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) an effective amount of at least one other chemotlzerapeutic agent, and c) instructions for administering the nanoparticles and the chemotherapeutic agents simultaneously and/or sequentially, for treatment of a proliferative disease (such as cancer). In some embodiments, the taxane is any of paclitaxel, docetaxel, and ortataxel. In some embodiments, the kit comprises nanoparticles comprising a) a composition comprising nanoparticles comprising paclitaxel and an albumin (such as AbraxaneTM), b) an effective amount of at least one other chemotherapeutic agent, and c) instntctions for administering the nanoparticles and the chemotherapeutic agents simultaneously and/or sequentially, for the effective treatment of a proliferative disease (such as cancer).

[0184] In some embodiments, the kit comprises a) a composition comprising nanoparticles comprising a taxane and a carrier protein (such as albumin), b) a composition comprising nanoparticles comprising at least one other chemotherapeutic agent and a caiTier protein (such as albtunin), and c) instructions for administering the nanoparticle compositions simultaneously and/or sequentially, for treatment of a proliferative disease (such as cancer). In some embodiments, the kit comprises nanoparticles comprising a) a composition comprising nanoparticles coinprising paclitaxel and an albumin (such as Abraxane rM), b) a composition comprising nanoparticles comprising at least one other chemotherapeutic agent and a carrier protein (such as albumin), and c) instructions for administering the nanoparticle compositions simultaneously and/or sequentially, for the effective treatment of a proliferative disease (such as cancer).
[0185] The nanoparticles and the chemotherapeutic agents can be present in separate containers or in a single container. It is understood that the kit may comprise one distinct composition or two or more compositions wherein one coinposition coinprises nanoparticles and one composition comprises a chemotherapeutic agent.
[0186] The kits of the invention are in suitable packaging. Suitable paclcaging include, but is not limited to, vials, bottles, jars, flexible packaging (e.g., seled Mylar or plastic bags), and the like. Kits may optionally provide additional components such as buffers and inteipretative inforination.
[0187] The instructions relating to the use of the nanoparticle compositions generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The containers may be unit doses, bulk packages (e.g., multi-dose packages) or sub-unit doses. For example, kits may be provided that contain sufficient dosages of the taxane (such as taxane) as disclosed herein to provide effective treatment of an individual for an extended period, such as any of a weelc, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 7 months, 8 months, 9 months, or more.
Kits may also include multiple unit doses of the taxane and pharmaceutical compositions and instnictions for use and packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies.
[0188] Those skilled in the art will recognize that several variations are possible within the scope and spirit of this invention. The invention will now be described in greater detail by reference to the following non-limiting examples. The following examples fiirther illustrate the invention but, of course, should not be construed as in any way limiting its scope.

EXAMPLES
Example 1. Improved response and reduced toxicities for AbraxaneTM compared to Taxol in a Phase III study of AbraxaneTM given every three weeks.

[0189] Significantly reduced incidence of neutropenia and hypersensitivity, absence of requirement of steroid premedication, shorter duration of neuropathy, shorter infusion time and higher dose.
[0190] ABI-007 (AbraxaneTM), the first biologically interactive albumin-bound paclitaxel in a nanoparticle form, free of any solvent, was compared with Cremophor -based paclitaxel (Taxol"~') in individuals with metastatic breast cancer (MBC).
This phase III study was perfoi-ined to confinn the preclinical studies demonstrating superior efficacy and reduced toxicity of ABI-007 when compared with Taxol .
Individuals were randomly assigned to 3-week cycles of either ABI-007 260 mghn2 (iv) over 30 minutes without premedication (n = 229) or Taxol 175 mg/m2 IV over 3 hours with premedication (n = 225). ABI-007 demonstrated significantly higher response rates compared with Taxol (33% vs. 19%; p= 0.001) and significantly longer time to tumor progression (23.0 vs. 16.9 weeks; HR = 0.75; p = 0.006). There was a trend for longer overall survival in individuals who received ABI-007 (65.0 vs. 55.7 weeks; p=
0.374). In an unplanned analysis, ABI-007 improved survival in individuals receiving treatment as second- or greater-line therapy (56.4 vs. 46.7 weeks; HR = 0.73; p= 0.024).
The incidence of grade 4 neutropenia was significantly lower in the ABI-007 group (9% vs.
22%;
p < 0.001) despite a 49% higher paclitaxel dose. Grade 3 sensory neuropathy was more common in the ABI-007 group than in the Taxol group (10% vs. 2%; p < 0.001) but was easily managed and improved more rapidly (median, 22 days) than for Taxol' (median 73 days). No severe (grade 3 or 4) treatment-related hypersensitivity reactions occurred in any of the individuals in the ABI-007 group despite the absence of premedication and shorter adininistration time. In contrast, grade 3 hypersensitivity reactions occurred in the Taxol group despite standard premedication (chest pain: 2 individuals; allergic reaction: 3 individuals). Per protocol, corticosteroids and antihistamines were not administered routinely to individuals in the ABI-007 group; however, premedication was administered for emesis, myalgia/arthralgia, or anorexia in 18 individuals (8%) in the ABI-007 group in 2% of the treatinent cycles, whereas 224 individuals (>99%) in the Taxol group received premedication at 95% of the cycles. The only clinical chemistry value that was notably different between the 2 treatment at-ms was higher seilim glucose levels in the Taxol -treated individuals, who also had a higher incidence of hyperglycemia reported as an AE
(adverse effects) (15 [7%] vs. 3 [1%]; p = 0.003). Overall, ABI-007 demonstrated greater efficacy and a favorable safety profile compared with Taxol in this individual population.
The improved therapeutic index and elimination of the steroid premedication required for solvent-based taxanes malce this nanoparticle albumin-bound paclitaxel an important advance in the treatment of MBC.

Example 2. Weekly AbraxaneTM in Taxane-Refractory Metastatic Breast Cancer Individuals [0191] A recent Phase II clinical study showed that weekly administration of AbraxaneTM (nanoparticle albumin-bound paclitaxel) at a dose of 125 mg/in2 resulted in long-term disease control in individuals with metastatic breast cancer whose disease had progressed while being treated with Taxol'~ or Taxotere (that is, individuals who are taxane-refractory).
[0192] AbraxaneTM is believed to represent the first biologically interactive composition that exploits the receptor-mediated (gp60) pathway found to be integral to achieving high intracellular tumor concentrations of the active ingredient -paclitaxel. The Phase II study included 75 individuals with taxane-refractory metastatic breast cancer.
AbraxaneTM was administered weekly via a 30-minute infi.ision at 125 mg/mZ
without steroid/antihistainine premedication or G-CSF prophylaxis. Individuals received tl-iree weekly doses followed by one week of rest, repeated every 28 days. Unlike Taxol or Taxotere , which contain detergents that may inhibit tumor uptalce, the mechanism of action of the albumin-bound nanoparticle paclitaxel may result in improved outcomes, especially in this difficult-to-treat individual population.
[0193] Specifically, the data showed that despite this high weekly dose of 125 mg/m2 in this highly pre-treated and prior taxane-exposed individual population, only 3 of 75 individuals (4%) had to discontinue AbraxaneTM due to peripheral neuropathy.
Furthermore, of those who experienced Grade 3 peripheral neuropathy, 80% were typically able to resuine treatinent after a delay of only 1 or 2 weeks and continued to receive AbraxaneTM at a reduced dose for an average of 4 additional months. This rapid improveinent was consistent with our observation from the Phase III trial -that the peripheral neuropathy induced by paclitaxel alone (i.e., without Cremophor ) improves rapidly as compared to that induced by Taxol . These AbraxaneTM clinical trial experiences provide the first clinical opportunity to evaluate the effects of the chemotherapeutic agent itself, paclitaxel, from the effects from those of solvents. Based upon both the Phase II and III experience, the data now suggest that the peripheral neuropatlly from AbraxaneTM is not comparable to the peripheral neuropathy from Taxol'R' or Taxoterewitli respect to duration and iinpact on the individual.

[0194] With regard to the clinical experience of peripheral neuropathy following Taxol'R' or Taxotere , Abraxis Oncology recently completed a survey of 200 oncologists who were asked how long they thought the peripheral neuropathy induced by Taxol took to improve and/or resolve: 25% reported "7-12 months" and another 23% reported "never resolved"; for Taxotere , the respective percentages were 29% and 7%. These data are consistent with the statements in the Taxotere and TaxolcR'package inserts.
[0195] Analysis of the Phase II data demonstrates AbraxaneTM to be active in this poor-prognosis individual population (87% visceral (lung and liver) disease, 69% >3 metastatic sites, 88% tumor growth while on taxanes), of taxane-refractory individuals with metastatic breast cancer. Observations included a 44% disease control in Taxotere -refractory individuals and 39% disease control in Taxol -refractory individuals. Of those individuals whose disease progressed while on TaxotereCR' alone in the metastatic setting (n=27) a 19% response rate was noted after receiving weekly AbraxaneTM. Of those individuals whose disease progressed while on Taxol alone in the metastatic setting (n=23) a 13% response rate was noted after receiving weekly AbraxaneTM
[0196] AbraxaneTM was found to be well tolerated when administered weekly over 30 minutes without steroids or G-CSF prophylaxis: Grade 4 neutropenia = 3%
(withotit G-CSF); Grade 4 anemia = 1%; no severe hypersensitivity reactions (despite absence of premedication). In this heavily pretreated individual population, 75% of individuals were treated at the full high dose of 125 mg/mZ weelcly AbraxaneTM, with no dose reductions due to toxicities/adverse events. Of the individuals who developed grade 3 sensory neuropathy, 77% were able to restart AbraxaneTM at a reduced dose (75-100 ing/m2 ) and received a mean of 12.2 (range, 1-28) additional doses of AbraxaneTM. It was reinarlcable to note that of these individuals who resumed AbraxaneTM, 80% (8 of 10) were able to restart the dnig within 14 days after improvement of neuropathy to Grade 1 or 2. These results support the observations in the pivotal Phase III trial of 260 mg/in2 AbraxaneTM
administered every 3 weeks, in which rapid improvement of neuropathy (median of 22 days) was also noted.
Taken together these two clinical trials suggest when paclitaxel is given alone, the neuropathy which occurs appears to be short-lived and is easily managed.
[0197] AbraxaneTM utilizes the gp60 receptor based pathway on the microvessel endothelial cells to transport the albumin-paclitaxel complex out of the blood vessel and into the tumor interstititim, and it has been shown that Taxol'~'' was not transported by this mechanism. Furthennore, an albumin-binding protein, SPARC, was over-expressed in breast tumors and may play a role in the increased intra-tumoral accumulation of AbraxaneTM. The proposed mechanism suggested that once in the tumor interstitium, the albtunin-paclitaxel complex would bind to SPARC that was present on the tumor cell surface and be rapidly internalized into the tumor cell by a non-lysosomal mechanism.
[0198] In addition, the surfactants/solvents commonly used in current taxane foirnulations such as Cremophor , Tween 80 and TPGS, strongly inhibit the binding of paclitaxel to albumin, thereby limiting transendothelial transport. Additional data presented showed a statistically improved efficacy of AbraxaneTM over Taxotere in the MX-1 mammary breast carcinoma xenograft at equal dose.
[0199] In conclusion, 75% of individuals were treated at full high dose with no dose reductions. Data indicate rapid improvement of peripheral neuropathy when nanoparticle albumin-bound paclitaxel is administered alone, without the solvent Cremophoro'.
Additional data provide increased evidence that mechanism of action may play important role in enhancing individual outcomes.

Example 3. AbraxaneTM (ABI-007) acts synergistically with targeted antiangiogenic pro-apoptotic peptides (HKP) in MDA-MB-435 human tumor xenografts.
[0200] The antiangiogenic activity of small synthetic pro-apoptotic peptides composed of two functional domains, one targeting the CD 13 receptors (aminopeptidase N) on tumor microvessels and the other disrupting the mitochondrial membrane following internalization have previously been reported. See Nat Med. 1999 Sep;
5(9):1032-8. A
second generation dimeric peptide, CNGRC-GG-d(KLAKLAK)2, named HKP (Hunter Killer Peptide) was found to have improved antitumor activity. Since anti-angiogenic agents such as AvastinOO exhibit synergism in combination with cytotoxic agents such as 5-fluorouracil, we evaluated the combination of the antiangiogenic HKP with AbraxaneTM
(ABI-007), an albumin nanoparticle paclitaxel that is transported by the gp60 receptor in vascular endothelium (Desai, SABCS 2003), in MDA-MB-435 human breast tumor xenografts.
[0201] Methods: MDA-MB-435 human tumor xenografls were established at an average tumor volume of 100 mm3, mice were randomized into groups of 12-13 animals and treated with HKP, AbraxaneTM, or HKP and AbraxaneTM. HKP was delivered i.v.
(250 ug), once a week, for 16 weelcs. AbraxaneTM was administered i.v., daily for 5 days at mg/kg/day only for the first week of treatment. The AbraxaneTN1 dose used was substantially below its MTD (30 mg/kg/day, qd x 5) to prevent the tumor from complete regression so effect of HIU could be noted.

[0202] Results: At nineteen weeks of treatment, tumor volume was significantly decreased between conti-ol group (10,298 mm3 zL 2,570) and HKP (4,372 mm3 zL
2,470; p <
0.05 vs control) or ABI-007 (3,909 mm3 =L 506; p < 0.01 vs control). The combination of ABI-007 and HKP significantly reduced the tumor volume over either monotherapy (411 mm3 386; p < 0.01 vs. AbraxaneTM monotherapy or HKP monotherapy). The treatments were well tolerated.

[0203] Conclusion: The combination of AbraxaneTM (ABI-007), a nanoparticle albumin-bound paclitaxel, with the vascular targeting anti-angiogenic dimeric peptide HKP
(CNGRC-GG-d(KLAI,,-LAK)Z) against the MDA-MB-435 xenograft breast tumor showed a significant reduction in tumor volume coinpared to monotherapy of either agent alone. Our results suggest that the combination of AbraxaneTM with antiangiogenic agents such as HIUs or perhaps Avastin may be beneficial.

Example 4. Metronomic ABI-007 Therapy: Antiangiogenic and Antitumor Activity of a Nanoparticle Albumin-bound Paclitaxel Example 4a [0204] Methods: The antiangiogenic activity of ABI-007 was assessed by the rat aortic ring, human umbilical vein endothelial cell (HUVEC) proliferation and tube formation assays. Optimal dose of ABI-007 for metronomic therapy was determined by measuring the levels of circulating endothelial progenitors (CEPs) in peripheral blood of Balb/c non-tumor bearing mice (n=5/group; dosing: 1-30 mg/lcg, i.p, qd x 7) with flow cytometry (Shalced et al., Cancer Cell, 7:101-111 (2005)). Subsequently, the antitumor effects of metronomic (qd; i.p.) and MTD (qd x 5, 1 cycle; i.v.) ABI-007 and Taxol R were evaluated and compared in SCID mice bearing human MDA-MD-231 breast and PC3 prostate cancer xenografts.

[0205] Results: ABI-007 at 5 nM significantly (p < 0.05) inhibited rat aortic microvessel outgrowth, human endothelial cell proliferation and tube fonnation by 53%, 24%, and 75%, respectively. The optimal dose of ABI-007 for metronomic therapy was observed to be 6-10 mg/kg based on CEP measurements. Metronomic ABI-007 (6 mg/lcg) but not Taxol'c" (1.3 mg/kg) significantly (p < 0.05) suppressed tumor growth in both xenograft models. Neither ABI-007 nor Taxol adininistered metronomically induced any weight loss. Although MTD ABI-007 (30 mg/kg) iiillibited tumor growth more effectively than MTD Taxol'D', (13 mg/kg), significant weiglit loss was noted with the fonner.

Interestingly, the antitumor effect of metronomic ABI-007 approximated that of MTD
Taxol .
[0206] Conclusion: ABI-007 exhibits potent antiangiogenic and antitumor activity when used in a metronomic regime.

Exam lp e 4b [0207] Rat Aortic Ring Assay. Twelve-well tissue culture plates were coated with Matrigel (Collaborative Biomedical Products, Bedford, MA) and allowed to gel for 30 min at 37 C and 5% COa. Thoracic aortas were excised from 8- to 10-week-old male Sprague-Dawley rats, cut into 1-inm-long cross-sections, placed on Matrigel-coated wells and covered with an additional Matrigel. After the second layer of Matrigel had set, the rings were covered with EGM-II and incubated overnight at 37 C and 5% CO2. EGM-Il consists of endothelial cell basal medium (EBM-11; Cambrex, Walkersville, MD) plus endothelial cell growth factors provided as the EGM-II Bulletkit (Cambrex). The culture medium was subsequently changed to EBM-II supplemented with 2% FBS, 0.25 g/ml amphotericin B
and 10 g/ml gentamycin. Aortic rings were treated with EBM-II containing the vellicle (0.9% saline/albumin), carboxyamidotriazole (CAI; 12 g/ml), or ABI-007 (0.05-10 nM
paclitaxel) for 4 days and photographed on the fifth day. CAI, a lcnown anti-angiogenic agent, was used at a higher than clinically achievable concentration as a positive control.
Experiments were repeated four times using aortas from four different rats.
The area of angiogenic sprouting, reported in square pixels, was quantified using Adobe Photoshop 6Ø
[0208] As shown in Figure IA, ABI-007 significantly inhibited rat aortic microvessel outgrowth in a concentration-dependent manner relative to the vehicle control, reaching statistical significance (p < 0.05) at 5 nM (53% inhibition) and 10 iiM (68%
inhibition). The amount of albumin present at each concentration of ABI-007 alone did not inhibit angiogenesis.
[0209] Endothelial Cell Proliferation Assay. Human umbilical vein endotllelial cells (IIUVEC; Cainbrex) were maintained in EGM-II at 37 C and 5% CO2. HUVECs were seeded onto 12-well plates at a density of 30,000 cells/well and allowed to attach overnight. The culture medium was then aspirated, and fresh culture medium containing either the vehicle (0.9% saline/albumin), or ABI-007 (0.05-10 nM paclitaxel) was added to each well. After 48 h, cells were trypsinized and counted with a Coulter Z1 counter (Coulter Corp., Hialeah, FL). All experiments were repeated three times.

[0210] As shown in Figure IB, human endothelial cell proliferation was significa.iitly inhibited by ABI-007 at 5 nM and 10 nM by 36% and 41%, respectively.
[0211] Endothelial Cell Tube Foimation Assay. Eight-well slide chambers were coated with Matrigel and allowed to gel at 37 C and 5% CO2 for 30 min. HUVECs were then seeded at 30,000 cells/well in EGM-II containing either the vehicle (0.9%
saline/albumin) or ABI-007 (0.05-10 nM paclitaxel) and incubated at 37 C and 5% COZ for 16 h. After incubation, slides were washed in PBS, fixed in 100% methanol for 10 s, and stained with DiffQuick solution II (Dade Behring Inc., Newark, DE) for 2 min.
To analyze tube fonnation, each well was digitally photographed using a 2.5x objective. A
threshold level was set to mask the stained tubes. The corresponding area was measured as the nuinber of pixels using MetaMorph software (Universal hnaging, Downingtown, PA).
Experiments were repeated three times.

[0212] As shown in Figure 1C, ABI-007 blocked tube foimation by 75% at both 5 nM and 10 nM.

[0213] Determination of the In Vivo Optimal Biologic Dose of ABI-007 by Measuring Circulating Endothelial Cells (CECs) and Circulating Endothelial Progenitors (CEPs). Six- to eight-week-old female Balb/cJ mice were randomized into the following eight groups (n=5 each): untreated, treated with i.p. bolus injections of either the drug vehicle (0.9% saline/ albumin), or ABI-007 at 1, 3, 6, 10, 15 or 30 mg/lcg paclitaxel daily for 7 days. At the end of the treatment period, blood samples were drawn by cardiac puncture and collected in EDTA-containing vacutainer tubes (Becton Dickinson, Franklin Lalces, NJ). CECs and CEPs were enumerated using four-color flow cytometry.
Monoclonal antibodies specific for CD45 were used to exclude CD45+
hematopoietic cells.
CECs and their CEP subset were depicted using the murine endothelial markers fetal liver kinase 1/VEGF receptor 2(flk-1/VEGFR2), CD13, and CD117 (BD Pharmingen, San Diego, CA). Nuclear staining (Procount; BD Biosciences, San Jose, CA) was performed to exclude the possibility of platelets or cellular debris interfering with the accuracy of CEC
and CEP enuineration. After red cell lysis, cell suspensions were evaluated by a FACSCalibur (BD Biosciences) using analysis gates designed to exclude dead cells, platelets, and debris. At least 100,000 events/sample were obtained in order to analyze the percentage of CECs and CEPs. The absolute number of CECs and CEPs was then calculated as the percentage of the events collected in the CEC and CEP
enumeration gates multiplied by the total white cell count. Percentages of stained cells were determined and compared to the appropriate negative controls. Positive staining was defined as being greater than non-specific background staining. 7-aminoactinomycin D (7AAD) was used to enumerate viable versus apoptotic and dead cells.
[0214] Figure 2 shows that ABI-007 administered i.p. daily for 7 days at 3, 10-mg/kg significantly decreased CEP levels in non-tumor bearing Balb/cJ mice.
However, ABI-007 at 10-30 mg/kg was associated with a significant reduction of white blood cell count indicative of toxicity. Although the reduction of CEP levels by ABI-007 at 6 mg/kg did not reach statistical significance, decrease in white blood cell count was not evident.
Therefore it was concluded that the in vivo optimal biologic dose for metronomic ABI-007 was between 3-10 mg/kg. In one study, metronomic Taxol@ at 1.3, 3, 6, or 13 mg/kg given i.p. daily for 7 days did not significantly reduce viable CEP levels, whereas inetronomic Taxol at 30 mg/kg or higlier resulted in severe toxicity and eventually mortality in mice.
It was previously reported that the i.p. administration of Taxol at doses commonly used in the clinic resulted in entrapment of paclitaxel in CremophorOO EL micelles in the peritoneal cavity and consequently, insignificant plasma paclitaxel concentration (Gelderblom et al., Clin. Cccrrcet= Res. 8:1237-41 (2002)). This would explain why doses of metronomic Taxolt7 (1.3, 3, 6, and 13 mg/kg) that did not cause death failed to change viable CEP
levels. In this case, the i.p. administration of metronomic Taxol~~ at 1.3 mg/kg would not be any different from that at 13 mg/kg. Therefore the lower dose, 1.3 mg/kg, was selected to minimize the amount of CremophorD EL per paclitaxel administration for subsequent experiments.
102151 Antitumor effects of metronomic and MTD ABI-007 compared with metronomic and MTD Taxol . Human prostate cancer cell line PC3 and human breast cancer cell line MDA-MD-231 were obtained from the .American Type Culture Collection (Manassas, VA). PC3 cells (5 x 106) were injected s.c. into 6- to 8-week-old male SCID
mice, whereas MDA-MB-231 cells (2 x 106) were implanted orthotopically into the mammary fat pad of female SCID mice. When the primary tumor volume reached approximately 150-200 mm3, animals were randomized into eight groups (n=5-10/group).
Each group was treated with either 0.9% saline/albumin vehicle control, Cremophorg EL
vehicle control, metronomic Taxol (1.3 mg/kg, i.p., qd), metronomic ABI-007 (3, 6, or 10 ing/Icg paclitaxel, i.p., qd), MTD Taxol (13 mg/kg, i.p., qd x 5, 1 cycle), or MTD ABI-007 (30 mg/kg paclitaxel, i.v., qd x 5, 1 cycle). Perpendicular tuinor dianieters were measured with a caliper once a week and their volumes were calculated. At the end of the treatment period, blood samples were drawn by cardiac puncture from niice in all groups.
CECs and CEPs were enumerated as described herein.

[0216] Metronomic ABI-007 (3, 6 and 10 mg/kg) but not TaxolOO (1.3 mg/kg) administered i.p. daily for 4 weelcs significantly (p < 0.05) inhibited growth ofboth MDA-MB-231 and PC3 hmiors (Fig. 3A and Fig. 3B). Neither ABI-007 nor Taxol''21 administered metronomically induced any weight loss (Fig. 3C and Fig. 3D). Although MTD ABI-(30 mg/kg) inhibited tumor growth more effectively than MTD Taxol'~' (13 mg/kg), significant weight loss was noted with the former, indicating toxicity. In addition, two out of five mice treated with MTD ABI-007 displayed signs of paralysis in one limb 6 days after the last dose of dnig. The paralysis was transient and resolved within 24-48 hours.
Interestingly, the antitumor effect of metronomic ABI-007 at 6 mg/kg approximated that of MTD TaxolOO in the MDA-MB-231 xenograft model (Fig. 3A). Increasing the dose of metronomic ABI-007 to 10 mg/kg did not seem to confer more pronounced tumor growth inhibition. In contrast, metronomic A,BI-007 elicited greater antitumor response at 10 mg/kg than at 3 and 6 mg/kg in the PC3 xenografts (Fig. 3B).
[0217] Metronomic ABI-007 significantly decreased the levels of viable CEPs in a dose-dependent manner in MDA-MB-231 tumor-bearing mice (Fig. 4A). Viable CEP
levels also exhibited a dose-dependent reduction in response to metronomic ABI-007 in PC3 tumor-bearing mice, but reached statistical significance only at 10 mg/kg (Fig. 4B).
The levels of CEPs were not altered by metronomic Taxol in both xenograft models (Fig.
4A and 4B).
[0218] Effects of metronomic and MTD ABI-007 and metronomic and MTD
Taxol on intratumoral microvessel density were studied. Five-urn thick sections obtained froin frozen MDA-MB-231 and PC3 tumors were stained with H&E for histological examination by standard methods known to one slcilled in the art. For detection of microvessels, sections were stained with a rat anti-mouse CD31/PECAM-1 antibody (1:1000, BD Pharmingen) followed by a Texas Red-conjugated goat anti-rat secondary antibody (1:200, Jackson IminunoResearch Laboratories, Inc., West Grove, PA).
A single microvessel was defined as a discrete cluster or single cell stained positive for CD31/PECAM-1d, and the presence of a lumen was not required for scoring as a microvessel. The MVD for each tumor was expressed as the average count of the three most densely stained fields identified with a 20x objective on a Zeiss AxioVision 3.0 fluorescence microscopic irnaging system. Four to five different tumors per eac11 vehicle control or treatment group were analyzed.
[0219] In MDA-MB-231 tumors, metronomic ABI-007 at 6 and 10 mg/kg as well as MTD ABI-007 seemed to reduce microvessel density (MVD) slightly although statistical significance was not reached (Fig. 5A). In PC3 tumors, metronomic ABI-007 at 3 and 10 mg/lcg appeared to decrease MVD but without reaching statistical sigilificance (Fig. 5A).
Interestingly, a sigilificant correlation existed between MVD and the level of viable CEPs in the MDA-MB-231 (Fig. 5B; i=0.76, P-0.04) but not in the PC3 (Fig. 5C; i=-0.071, P-0.88) model.
[0220] In vivo angiogenesis evaluation were carried out. A Matrigel plug perftision assay was perfor-med with minor modifications to methods known by one skilled in the art.
Briefly, 0.5 ml Matrigel supplemented with 500 ng/ml of basic fibroblast growth factor (bFGF; R&D Systems Inc., Minneapolis, MN) was injected s.c. on day 0 into the flanks of 10-week-old female Balb/cJ mice. On day 3, animals were randomly assigned to eight groups (n = 5 each). Each group was treated with either 0.9% saline/albumin vehicle control, Cremophorg EL vehicle control, metronomic Taxol (1.3 mg/lcg, i.p., qd), metronomic ABI-007 (3, 6, or 10 mg/kg paclitaxel, i.p., qd), MTD Taxols (13 mg/kg, i.v., qd x 5), or MTD ABI-007 (30 mg/kg paclitaxel, i.v, qd x 5). As a negative control, five additional female Balb/cJ mice of similar age were injected with Matrigel alone. On day 10, all animals were injected i.v. with 0.2 ml of 25 mg/ml FITC-dextran (Sigma, St. Louis, MO). Plasma samples were subsequently collected. Matrigel plugs were removed, incubated with Dispase (Collaborative Biomedical Products, Bedford, MA) overnight at 37 C, and then homogenized. Fluorescence readings were obtained using a FL600 fluorescence plate reader (Biotech Instruments, Winooski, VT). Angiogenic response was expressed as the ratio of Matrigel plug fluorescence to plasma fluorescence.
[0221] Metronomic ABI-007 at 6 and 10 mg/kg appeared to decrease angiogenesis although the inhibition did not reach statistical significance (Fig. 6).
Angiogenesis seemed to be unaltered by metronomic ABI-007 at 3 mg/kg, MTD ABI-007, MTD aild metronoinic Taxol RO relative to the respective vehicle controls (Fig. 6). These observations were similar to the intratumoral MVD results described herein.

Example 5. Nab-5109, A Nanoparticle albumin-bound IDN5109 (nab-5109) Shows Improved Efficacy and Lower Toxicity over the Tween formulation (Tween -5109, Ortataxel) [0222] Methods: Nanoparticle nab-5109 was prepared using raccb technology and characterized by laser light scattering. Nab-5109 and Tween-5109 were tested against Pgp+ DLD-1 (lcnown to be resistant against paclitaxel and docetaxel -Vredenburg et al, JNCl93: 1234-1245, 2001) human colon carcinoma xenograft in nude mice (n=5/group) at doses of 50 mg/kg (Tween -5109, previously shown as MTD) and 75 mg/lcg (nab-5109) given q3d x 4, i.v. Control groups of PBS and human serum albumin (HSA) were also used.
[02231 Results: Nab-5109 yielded nanoparticles with mean size, ZAve=119 iun and Zeta potential =-32.7 mV. Nab-5109 was lyophilized to a dry powder that easily dispersed in saline. In vivo, there was significantly more weight loss (ANOVA, p<0.001) in the tumor bearing animals witlz Tween -5109 (50mg/kg, 8.8% wt loss) than with nab-(75mg/kg, 3.4% wt loss) indicating substantially lower toxicity of nab-5109 despite the 50% higher dose. There was significant tumor suppression by nab-5109 and Tweeri -5109 (ANOVA, p<0.0001 vs. controls) with tumor growth delays of 36 and 28 days respectively for nab-5109 (75 mg/kg) and Tween -5109 (50 mg/kg). Nab-5109 was more effective than Tweeno"-5109 (ANOVA, p=0.0001) in suppressing tumor growth. There were no differences between the PBS and HSA control group in tern of toxicity and efficacy.
[02241 Conclusion: Nanoparticle albumin-bound, nab-5109 was successfully prepared and could be given at 50% higher dose than Tween -5109 with lower toxicity despite higher dose. At this higher dose, 75 mg/kg (q3d x 4), nab-5109 showed significantly improved efficacy in the Pgp+ DLD-1 human colon xenograft compared with Tween -5109.

Exaniple 6. Nanoparticle Albumin Bound (izab) Dimeric Thiocolchicines nab-5404, nab-5800, and nab-5801: A Comparative Evaluation of Antitumor Activity vs AbraxaneTM and Irinotecan [0225] Methods: Nanoparticle colchicines were prepared using taab technology.
Cytotoxicity was evaluated in vitro using human MX-1 breast carcinoma cultures. Ifz vivo anti-tumor activity (human HT29 colon tumor xenograft) in nude mice was compared against Irinotecan and AbraxaneTM. Dose levels for the nab-colchicines and Irinotecan were 20 mg/kg, 30 mg/lcg, and 40 mg/kg, given q3d x 4, i.v. AbraxaneTM was dosed at its MTD, 30 mg/kg, given qd x 5, j02261 Results: The hydrophobic thiocoichicine dimers yielded nanoparticles with average size ZAõe (Iun) of 119, 93, and 84 for nab-5404, nab-5800, and nab-5801, respectively. The nanoparticle suspensions were sterilized throtigh 0.22 um filters and lyophilized. Ita vitro, nab-5404 was the most potent of the three analogs against MX-1 (p <
0.0005, ANOVA), (IC5o (ug/ml): 18, 36 and 77 for nab-5404, nab-5800 and nab-respectively) as well as against the HT29 xenograft in vivo (p < 0.0001, ANOVA). Tumor volume was suppressed by 93%, 79%, and 48% with nab-5404 at doses 40 mg/lcg, mg/kg, and 20 mg/kg, respectively. In contrast, tumor volume was only suppressed by 31%, 16%, and 21% with nab-5800; and 17%, 30%, and 23% with nab-5801 at 40 mg/kg, 30 mg/kg, and 20 mg/kg, respectively. Nab-5404 was more effective than Irinotecan at all dose levels (p < 0.008, ANOVA) with tumor volumes for Irinotecan suppressed by only 48%, 34%, and 29% at dose levels of 40 mg/kg, 30 mg/kg, and 20 mg/kg, respectively. In comparison to AbraxaneTM, nab-5404 was more active at equitoxic dose (ETD) based on equal weight loss (p < 0.0001, ANOVA). Tumor volume was suppressed 93% by nab-5404 (40 mg/kg, q4d x 3) and 80% by AbraxaneTM (30 mg/kg, qd x 5) at their respective ETDs.
[0227] Conclusions: Nab technology was utilized to convert 3 hydrophobic dimeric thiocolchicines (IDN5404, IDN5800, IDN5801) to nanoparticles suitable for I.V.
administration. Nab-5404 had superior antitumor activity in vitro and in vivo compared to nab-5800 and nab-5801. Nab-5404 was more potent than Irinotecan at equal dose.
At equitoxic dose, defined by weight loss, nab-5404 was more potent than AbraxaneTM. These data warrant fiirther investigation of nab-5404.

Example 7. AbraxaneTM vs Taxotere : A Preclinical Comparison of Toxicity and Efficacy [0228] Methods: Toxicity of AbraxaneTM and Taxotere was compared in a dose-ranging study in nude mice given the drugs on a q4d x 3 schedule. The dose levels were Taxotere 7, 15, 22, 33, and 50 mg/kg and ABX 15, 30, 60, 120, and 240 mg/kg.
Antitumor activity of AbraxaneTM and Taxotere was compared in nude mice with human MX-1 mammary xenografts at a dose of 15 mg/kg weekly for 3 weeks.
[0229] Results: In the dose-escalation study in mice, the Taxotere maximum tolerated dose (MTD) was 15 mg/kg and lethal dose (LDtoo) was 50 mg/kg. In contrast, the AbraxaneTM MTD was between 120 and 240 mg/kg and LDloo was 240 mg/kg. In the tumor study AbraxaneTM was more effective than equal doses of Taxotere in tumor growtll inhibition (79.8% vs 29.1%, p < 0.0001, ANOVA).
[0230] Concluslon: Nanoparticle abumin-bound paclitaxel (AbraxaneTM) was superior to Taxotere in the MX-1 tumor model when tested at equal doses.
Furthennore, the toxicity of AbraxaneTM was significantly lower than that of Taxotere which would allow dosing of AbraxaneTM at substantially higher levels. These results are similar to the ei-dzanced therapeutic index seen with AbraxaneTM compared to Taxol0 and suggest that the presence of surfactants may impair the transport, antituinor activity and increase the toxicity of taxanes. Studies in additional tumor models comparing AbraxaneTM
and Taxoterec are ongoing.

Example 8. A Nanoparticle Albumin Bound Thiocolchicine dimer (nab-5404) with Dual Mechanisms of Action on Tubulin and Topoisomerase-1: Evaluation of In vitro and In vivo Activity [0231] Methods: IDN5404 was tested for cytotoxic activity using the MCF7-S
breast carcinoma and its multidrug resistant variant, MCF7-R (pgp+). Its cytotoxicity was also assessed against the NCI-60 human tumor cell line panel. The nanoparticle albumin bound nab-5404 was administered IV using various schedules, to SC1D mice implanted s.c.
with a human A121 ovarian tumor xenograft.
[0232] Results: Against MCF7 cell lines, the parent compound, colchicine, demonstrated tumor growth inhibition with the IC50 value (50% growth inhibitory concentration) for MCF7-S cells at 3.9 0.2 nM. The resistant variant MCF7-R
demonstrated an IC50 of 66 8.6 nM, approximately a 17-fold increase due to drug resistance. IDN5404, demonstrated increased activity against both cell lines, displaying IC50 values of 1.7 0.1 and 40 3.8 nM, respectively. These results were confiimed within the NCI 60 human tumor cell line panel with IDN5404 having a mean IC50 of <10-8M and >10 fold resistance between the MCF7-S and the MCF7-R cell lines. The COMPARE algoritlun identified IDN5404 as a tubulin binder similar to vinca alkaloids, confinliing the previous results. In vivo against the A121 ovarian tumor xenograft, efficacy and toxicity of nab-5404 was dose and schedule dependent. Nanoparticle nab-5404 was well tolerated and capable of inducing complete regressions and cures: at 24 mg/kg administered IV qd x 5, 5 of 5 mice were long-term survivors (LTS) with no evidence of tumor. However, increasing the dosage to 30 mg/kg resulted in 5 of 5 toxic deaths. On a q3d x 4 schedule, 30 mg/kg resulted in 4 of 5 mice LTS and at 50 mg/kg, 5 of 5 toxic deaths. Using a q7d x 3 schedule, 40 mg/lcg resulted in 3 of 5 mice LTS and at 50 mg/lcg, 4 of 4 LTS were noted.

[0233] Conclusions: IDN5404, a new thiocolchicine dimer witli dual mechanism of action showed activity in pgp-expressing, cisplatin and topotecan resistant cell lines. Iha vivo, nanoparticle albumin bound nab-5404 was active against A121 ovarian xenografts.
Example 9. Combination Studies of AbraxaneTM and Other Agents [0234] Due to the advantageous properties of AbraxaileTM (ABX, the nanoparticle albumin-bound paclitaxel) noted above, it was used and being used in a number of studies with different modes of administration and schedules and in combination witll other oncology drugs as well as radiation treatment. These are listed below:
[0235] In metastatic breast cancer, these sttidies include:
Randomized Phase II Trial of Weekly AbraxaneTM in Combination ABX 125, Gem 1000 mg/m', To evaluate the conibination of ABX
with Gemcitabine in Individuals D1,8; q 3wlc and Gemcitabine in lst-line MBC.
with HER2 Negative Metastatic Breast Cancer A phase II study of weekly dose-dense nanoparticle paclitaxel ABX 100 mg/mZ Data will be iinportant for using (ABI-007) carboplatin, witlz , Carbo AUC ~X in combination with carbo Herce tin as first or second-line 2, both D1,8,15; Her 2 mg/kg p (4 mg/kg on wk a) q4wk x 6 and/or Herceptin". Also helpful for therapy of advanced HER2 positive other combinations.
breast cancer Weekly Vinorelbine and L1: ABX 80, Nav 15; L2:
AbraxaneTM with or without G-CSF ABX 90, Nav 20; L3: ABX Multi-center study of ABX in in stage IV breast cancer: a phase ~ 100, Nav 22.5; L4: ABX 110, combination with Navelbine(R) in I-II study Nav 25; L5: ABX 125, Nav 25 1st-line MBC.
qwk Phase II trial of weekly AbraxaneTM A relatively large phase II of weekly inonotherapy for lst-line MBC (plus ABX 125 mg/m' Q3/4wk ABX monotherapy at 125 mg/m' in Herceptin in Her2+ pts) lst-line MBC.

Phase I/II trial AbraxaneTM phts ABX + Anthracycline Doxil(i] for MBC plus limited PK

ABX weekly (130 mg/m') vs.
3-arm phase II trial in 1 st-line MBC q2wk (260 mg/m') vs. q3wlc To optimize ABX monotherapy regime for MBC
(260 mg/m') randomized ABX MBC trial to 3-arm phase II trial in 1 st-line and obtain important data: weekly ABX
2nd-line MBC, with biolo ical ABX weekly vs. ABX q3wk g vs. Taxol weekly vs. weelcly Taxol ; weekly ABX vs.
correlates analyses 3-weekly ABX; plus biomarlcer study (caveolin-1 and SPARC).
combination of ABX and Phase I/II AbraxaneTM + GW572016 TBD GW572016 (a dual EGFR inhibitor and one of the most promising new biological agents for BC).

A phase I dose escalation study of a AbraxaneTM 100 mg/m' This phase I trial is to determine the 2 day oral gefitinib weekly, 3 out of 4 weeks; safety and tolerability of a 2 day chemosensitization pulse given prior Gefitinib starting at 1000 mg/d gefitinib pulse given prior to to weekly AbraxaneTM in individuals x 2 days AbraxaneTM administration.
with advanced solid tumors weekly ABX (125 mg/mZ, 2 To evaluat the combination of ABX
Phase II 1S' line MBC trial wk on and 1 wk off) + Xeloda~ and Xeloda in lst-line MBC, using 825 mg/hn' d 1-14 q3wk 2 weeldy on and 1 weekly off ABX
regime.
Phase II pilot adjuvant trial of Dose dense AC + G CSF --> A pilot adjuvant study of a "super AbraxaneTM in breast cancer weekly ABX --> Avastin dose dense"

AbraxaneTM in dose-dense adjuvant AC q2w x 4+ G CSF --> ABX A pilot adjuvant study of dose dense chemotherapy for early stage breast q2wk x 4 ABX regime -- an alternate of a cancer standard adjuvant regime Phase II pilot adjuvant trial of AC Q2wk --> ABX q2wk + A pilot adjuvant study in preparation AbraxaneTM in breast cancer G-CSF for phase III adjuvant trial [0236] fii Breast cancer neoadjuvant setting studies include:
Neoadjuvant: Gem 2000, Phase II Trial of Dose Dense Epi 60, ABX 175 ing/m', This neoadjuvant study is based on the Neoadjuvant Gemcitabine, Epirubicin, Neul 6 mg SC, all D 1 q2 GET data from Europe which showed ABI-007 (GEA) in Locally Advanced wk x 6 Adjuvant: Gem high activity. In the current regime, or Inflammatory Breast Cancer 2000, ABX 220, Neul 6 ABX will replace T, or Taxol .
ing D l q2wk x 4 Phase II preoperative trial of ABX 220 mg/m' q2wk x AbraxaneTM followed by FEC (+ 6 followed by FEC x 4 Herceptin as appropriate) in breast (+Herceptiri for Her2+
cancer pts) Pre-clinical study of dnig-drug ABX + other agents interaction (ABX + HerceptinCO) Phase II neoadjuvant followed by (Navelbine@
+ Herceptin ) TAC vs. AC followed To evaluate AC followed by Randomized phase II trial of ABX+carbo vs. AC ABX/carbo or ABX/carbo/Herceptin"
neoadjuvant chemotherapy in followed combinations vs TAC (a FDA
individuals with breast cancer ABX+carbo+Herceptin approved adjuvant BC
regime) in neoadjuvant setting.
Phase II neoadjuvant trial of ABX: 200 mg/m2 D1;
AbraxaneTM and capecitabine in Xel: 1000 mg/m' D1-14;
locally advanced breast cancer q3wk x 4 Phase II trial of neoadjuvant chemotherapy (NCT) with nanoparticle paclitaxel (ABI-007, , , Abraxane TM) in women with clinical ABX: 300 mg/m qDwk stage IIA, IIB, IIIA, IIIB, and IV (with intact primary) breast cancers [0237] In lung caiicer the studies include:

Pliase I/II study of AbraxaneTM The first phase 11 trial of ABX
monotherapy in 1st-line advanced ABX weekly combo with carbo in NSCLC.
NSCLC

Phase II Trial of weekly AbraxaneTM ABX: 125mg/m' plus carboplatin in lst-line NSCLC D1,8,15; Carbo: AUC
6 D l; q4 wk A Phase I Trial of Carboplatin and Arm 1: ABX 100, 125, This 2-arm phase I
study will AbraxaneTM on a weekly and every 150 mg/m D1,8,15 generate important data on three week schedule in individuals q4wk; = Arm 2: ABX ABX/carbo combination for ~
with Advanced Solid Tumor 220, 260, 300 mg/m- ftirther studies of this combo Malignancies Dl q3wk. Carbo in multiple diseases.
AUC6 in both arms ABX Level(a): 225 This phase II NSCLC study Phase 11 study of ABI 007 mg/m2; Level(b): 260 will generate data for a future (AbraxaneTM) and carboplatin in mg/m2; Level(3):300 phase III registration trial in advanced non-small cell lung cancer. mg/m2;q3wk Carbo lung cancer fixed at AUC6 q3wk Phase I study of ABI 007 ABX 3wk (AbraxaneTM) and carboplatin q3wk and Alimta can be a Phase I/II stiidy of AbraxaneTM + TBD promising combination diie to Alimta for 2nd-line NSCLC the non-overlapping toxicity profiles.

Phase 1/1l trial of AbraxaneTM plus cisplatin in advanced NSCLC
Phase I/II study of AbraxaneTM, Navelbine R, and Cisplatin for treatment of advanced NSCLC

Phase II ABX mono in 1 st-line ABX 260 mg/nz-' q3wk The 1 st ABX trial 'ni NSCLC.
NSCLC

Phase II study of AbraxaneTM Cohort 1: ABX q3wk;
monotherapy in 2nd line NSCLC Cohort 2: ABX
weekly. Doses TBD
Phase I/lI trial of weekly AbraxaneTM Ist line and carboplatin in advanced NSCLC

[0238] Studies in Prostate include:
Randomized phase II ABX 100 mg/mZ
weekly vs Q3W in front line weekly vs 260 HRP mg/rn2 q3wk Phase II ABX in 1 st-line weekly ABX Phase II shidy of weekly ABX in 1 st-line prostate cailcer HRPC

Phase II neoadjuvant study TBD A multi-center neoadjuvant trial of ABX
in prostate cancer plus biomarker study.
Phase II ABX 100 mg weekly no break [0239J Studies in ovarian cancer include:
Phase II study of AbraxaneTM
for treatment of advanced TBD
ovarian cancer (3rd-line) Phase I sttidy of AbraxaneTM ABX weekly + Carbo plus carbo for treatment of AUC 6 advanced ovarian cancer A phase Il trial of AbraxaneTMJCarboplatin in recurrent ovarian cancer [0240] Studies in Chemoradiation include:
Phase I/Il trial of AbraxaneTM
combined with radiation in NSCLC

AbraxaneTM Combined With animal model Radiation H&N (Head and Neck Cancer) TBD
[0241] Other studies include:

Phase II sh.tdy of ABX in treatment of 125 mg/mZ d1,8,15 persistent or recurrent carcinoma of the q28 days cervix PhII in preciously treated (100 ABX) and untreated (150 ABX) metastatic 26-->70 melanoma Ph II single treatment use of ABI-007 for the treatment of non-hematologic malignancies AbraxaneTM Combined With antiangiogenic agents, e.g., Avastin(l.
AbraxaneTM Combined With proteasome iiihibitors e.g., Velcade0.
AbraxaneTM Combined With EGFR
inhibitors e.g., Tarceva .

A randomized phase II trial of weekly gemcitabine, AbraxaneTM, and extemal irradiation for locally advanced pancreatic cancer Example 10. Combination of nanoparticle invention drugs with other agents and modes of therapy.

[0242] Lower toxicity of nanoparticle invention dnigs described herein allow combination with other oncology drugs and other modes of treatment with more advantageous outcome. These include nanoparticle fonns of paclitaxel, docetaxel, other taxanes and analogs, geldanamycins, colchicines and analogs, combretastatins and analogs, hydrophobic pyrimidine compounds, lomaiviticins and analogs including compounds with the lomaiviticin core structures, epothilones and analogs, discodermolide and analogs and the like. The invention dntgs may be coinbined with paclitaxel, docetaxel, carboplatin, cisplatin, other platins, doxorubicin, epirubicin, cyclophosphainide, iphosphainide, geincitabine, capecitabine, vinorelbine, topotecan, irinotecan, tamoxifen, camptothecins, 5-FU, EMP, etoposide, methotraxate and the like.

Example 11. Combination of AbraxaneTM with Carboplatin and Herceptin [0243] The combination of Taxol" and carboplatin has shown significant efficacy against metastatic breast cancer. On a weekly schedule, in this combination, Taxol" can only be dosed at up to 80mg/hn2. Higher doses cannot be tolerated due to toxicity. In addition, HER-2-positive individuals derive greater benefit when Herceptin" is included in their therapeutic regime. This open-label Phase II study was conducted to determine the synergistic therapeutic effect of ABI-007 (AbraxaneTM) with these agents. The current sttidy was initiated to evaluate the safety and antitumor activity of ABI-007/carboplatin with Herceptin ' for individuals with HER-2 positive disease. ABI-007 was given in combination with carboplatin and Herceptin " adininistered intravenously weekly to individuals with HER-2 positive advanced breast cancer. A cohort of 3 individuals received ABI-007 at a dose of 75 mg/in2 IV followed by carboplatin at target AUC = 2 weekly and Hereeptin" infusion (4 mg/lcg at week 1, and 2 mg/lcg on all subsequent weeks) for 1 cycle. These individuals tolerated the drug very well so for all subsequent cycles and individuals the dose of ABI-007 was escalated to 100 mg/mZ. Six individuals were treated to date. Of the 4 individuals that were evaluated for response, al14 (100%) showed a response to the therapy. It should be noted that due to lower toxicity of AbraxaneTM, a higher total paclitaxel dose could be given compared to Taxol wit11 resulting benefits to the individuals.

Example 12. Combination of AbraxaneTM with Carboplatin [0244] The combination of Taxol and carboplatin has shown significant efficacy in lung cancer. Another study with AbraxaneTM in coinbination with carboplatin on a 3 weekly schedule in individuals with lung cancer is ongoing.

Example 13. Use of AbraxaneTM in Combination With Radiation Example 13a [0245] AbraxaneTM, combined with clinical radiotherapy, enhances therapeutic efficacy and reduces normal tissue toxicity. AbraxaneTM is used to increase the therapeutic gain of radiotherapy for tumors; to enllance tumor response to single and fractionated iiTadiation; to eillZance noi7nal tissue response to radiation and to increase therapeutic ratio of radiotherapy.

[0246] A murine ovarian carcinoma, designated OCa-I, which has been investigated extensively is used. First, optimal timing of AbraxaneTM administration relative to local tumor radiation is timed to produce maximum antitumor efficacy. Tumors are generated in the right hind leg of mice by i.m. injection of tumor cells and treatment is initiated w11en the tumors reach 8mm in size. Mice are treated with 10 Gy single dose irradiation, a single dose of AbraxaneTM. or with combination therapy of AbraxaneTM given at different times 5 days before to 1 day after irradiation. A dose of AbraxaneTM equal to about 1%a times more than the maximuin tolerated dose of paclitaxel is used, a dose of 90 mg/kg.
The endpoint of efficacy is tuinor growtll delay. The groups consist of 8 mice each. Tumors are generated and treated as described in Aim 1. The endpoint of efficacy is tumor growth delay. Tuinors are irradiated with 5, 7.5 or 10 Gy delivered either in a single dose or in fractionated doses of 1, 1.5 or 2 Gy radiation daily for five consecutive days. Since AbraxaneTM is retained in the tumor for several days and exerts its enhancing effect on each of the five daily fractions, AbraxaneTM is given once at the beginning of the radiation regime. Since the ultimate goal in clinical radiotherapy is to achieve tumor cure, the potential for AbraxaneTM to enhance tumor radiocurability is determined. The same scheme as described for the fractionated tumor growth delay study is used, except that a range of doses from 2 to 16 Gy is given daily for five consecutive days (total radiation dose to 80 Gy). Tumors are followed for regression and regrowth for up to 120 days after irradiation, when TCD50 (the dose of radiation needed to yield local tumor cure in 50 percent of animals) is determined. There are two TCD50 assays: radiation only and AbraxaneTM plus radiation, and each assay consists of 10 radiation dose groups containing mice each. To provide therapeutic gain, any radioei-Aiancing agent, including AbraxaneTM, must increase tumor radioresponse more than increase normal tissue damage by radiation. Damage to jejunal mucosa, a highly proliferative tissue affected by taxanes is assessed. The jejunal microcolony assay is used to deterinine the survival of crypt epithelial cells in the jejunum of mice exposed to radiation. Mice are exposed to whole body irradiation (WBI) with daily doses of X-rays ranging from 3 to 7 Gy for five consecutive days. The mice are treated with AbraxaneTM, at an equivalent paclitaxel dose of 80 mg/kg, administered i.v. 24 h before the first dose of WBI and killed 3.5 days after the last dose of WBI. The jejunum is prepared for histological examination, and the number of regenerating crypts in the jejunal cross-section is counted. To constnict radiation survival curves, the number of regenerating crypts is converted to the number of surviving cells.

Exam lp e 13b [0247] The objective of this study was to assess whether ABI-007 (a) as a single agent has antitumor activity against the syngeneic murine ovarian carcinoma OCa-1 and (b) enhances the radiation response of OCa-1 tumors in a combined treatment regime as described in the previous example with the following modifications.
[0248] OCa-1 tumor cells were injected i.m. into the hind leg of C3H mice.
When tumors grew to a mean diameter of 7 mm, single treatment with local radiation (10 Gy) to the tumor-bearing leg, ABI-007 90 mg/kg i.v., or both, was initiated. To determine the optimal treatment scheduling, ABI-007 was given from 5 days to 9 hours before radiation as well as 24 hours after radiation. Treatment endpoint was absolute tiunor growth delay (AGD), defined as the difference in days to grow from 7-12 mm in diameter between treated and untreated tumors. For groups treated with the combination of ABI-007 and radiation, an enhancement factor (EF) was calculated as the ratio of the difference in days to grow from 7 to 12 mm between the tumors treated with the combination and those treated with ABI-007 alone to the AGD of tumors treated with radiation only.
To assess the radiation-enhancing effect of ABI-007 for a fractionated radiation regime on the endpoint tumor cure, a TCD50 assay was performed and analyzed 140 days post treatment.
Total doses of 5 to 80 Gy in 5 daily fractions were administered either alone or combined with ABI-007 24 hours before the first radiation dose.

[0249] As a single agent, ABI-007 significantly prolonged the growth delay of the OCa-1 tumor (37 days) compared to 16 days for untreated tumors. ABI-007 as a single ageiit was more effective than a single dose of 10 Gy, which resulted in a delay of 29 days.
For combined treatment regimes, ABI-007 given at any time up to 5 days before radiation, produced a supra-additive antitumor effect. EF was 1.3, 1.4, 2.4, 2.3, 1.9, and 1.6 at intertreatment intervals of 9h, 24 h and 2, 3, 4, and 5 days, respectively.
When ABI-007 was given after radiation, the combined antitumor treatment effect was less than additive.
Combined treatment with ABI-007 and radiation also had a significant effect on tumor cure by shifting the TCD50 of 55.3 Gy for tumors treated with radiation only to 43.9 Gy for those treated with the combination (EF 1.3).

[0250] This experiment demonstrated that ABI-007 possesses single-agent antitumor activity against OCa-1 and enhances the effect of radiotherapy when given several days prior. As previously demonstrated for paclitaxel and docetaxel, the radiation enhancement is likely a result of multiple mechanisms, with a cell cycle arrest in G2/M
being dominant at short treatment inteivals and tuinor reoxygenation at longer intervals.
Example 14. Combination of AbraxaneTM and Tyrosine Kinase Inhibitors [0251] Pulse-dosing of gefitinib in combination with the use of AbraxaneTM is useful to inhibit the proliferation of EGFr expressing tumors. 120 nude mice are inoculated with BT474 tumor cells to obtain at least 90 mice bearing BT474 xenograft tumors and split into 18 experimental arms (5 mice each). Arm 1 mice receive control i.v.
injections.
All other mice receive weekly i.v. injections of AbraxaneTM at 50 mg/lcg for 3 weeks. Arm 2 receive AbraxaneTM alone. Anns 3, 4, 5, 6, 7, 8 receive weekly AbraxaneTM
preceded by 2 days of a gefitinib pulse at increasing doses. Arms 9, 10, 11, 12, 13 receive weekly AbraxaneTM preceded by one day of a gefitinib pulse at increasing doses. Arms 14, 15, 16, 17, 18 receive weekly AbraxaneTM along with everyday administration of gefitinib at increasing doses. The maximum tolerated dose of gefitinib that can be given in a 1 or 2 day pulse preceding weekly AbraxaneTM or in continuous administration with AbraxaneTM
is established. In addition, measurement of anti-tumor responses will determine whether a dose-response relationship exists and whether 2 day pulsing or 1 day pulsing is superior.
These data are used to select the optimal dose of pulse gefitinib and that of continuous daily gefitinib given with AbraxaneTM
[0252] 120 nude mice are inoculated with BT474 tumor cells to obtain 90 mice bearing tumors. These mice are split into 6 groups (15 each). Arm 1 receive control i.v.
injections. Ann 2 receive AbraxaneTM 50 mg/kg i.v. weekly for 3 weeks. Arm 3 receive oral gefitinib at 150 mg/kg/day. Arm 4 receive AbraxaneTM 50 mg/kg along with daily gefitinib at the previously established dose. Arm 5 receive AbraxaneTM 50 mg/kg preceded by a gefitinib pulse at the previously established dose and duration. Ann 6 receive only a weekly gefitinib pulse at the previously established dose. After three weeks of therapy, mice are followed until controls reach maximum allowed tumor sizes.

Example 15. Phase II Study of Weekly, Dose-dense nabTM Paclitaxel (AbraxaneTM), Carboplatin With Trastuzumab As First-line Therapy Of Advanced HER-2 Positive Breast Cancer [0253] This study aimed to evaluate (1) the safety and tolerability and (2) the objective response rate of weekly dose-dense trastuzumab/AbraxaneTM/carboplatin as first-line cytotoxic therapy for patients with advanced/inetastatic (Stage IV
adenocarcinoma) HER-2-overexpressing breast cancer. Trastuzumab is a monoclonal antibody, also luzown as Herceptin , which binds to the extracellular seginent of the erbB2 receptor.
[0254] Briefly, patients without recent cytotoxic or radiotherapy were included.
Doses of AbraxaneTM were escalated from 75 mg/m2 as 30-min i.v. infiisions on days 1, 8, 15 up to 100 mg/m' for subsequent cycles according to the standard 3 + 3 rule.
Carboplatin AUC = 2 was given as 30-60 inin i.v. infiisions on days 1, 8, 15 a.nd for an initia129 day cycle. Trastuzumab was given as i.v. 30-90 min infusion on days 1, 8, 15, 22 at a dose of 4 mg/kg at week 1 and 2 mg/kg on all subsequent weeks.
[0255] Of 8 out of 9 patients evaluable for response the response rate (confirmed plus unconfirmed) was 63% with 38% stable disease. The lnost common toxicities were neutropenia (grade 3: 44%; grade 4: 11%) and leukocytopenia (33%).
[0256] These results suggest that trastuzumab plus AbraxaneTM plus carboplatin demonstrated a high degree of antitumor activity with acceptable tolerability as a first-line therapy for MBC.

Example 16. Phase II Trial of Capecitabine Plus nccb'M Paclitaxel(AbraxaneTn in the First Line Treatment of Metastatic Breast Cancer [0257] The purpose of this phase II study was to evaluate the safety, efficacy (time to progression and overall survival), and quality of life of patients with MBC
who received capecitabine in combination with AbraxaneTM. Capecitabine is a fluoropyrimidine carbamate also known as Xeloda'R' which has been shown to have substantial efficacy alone and in combination with taxanes in the treatment of MBC.
[0258] In this open-label, single-arm study, AbraxaneTM 125 mg/ma was given by i.v. infusion on day 1 and day 8 every 3 weeks plus capecitabine 825 mg/m2 given orally twice daily on days 1 to 14 every 3 weeks. Patients were HER-2/neu negative with a life expectancy of greater than 3 months. Patients had no prior chemotherapy for metastatic disease, no prior capecitabine therapy, and no prior fluoropyrimidine therapy and paclitaxel chemotherapy given in an adjuvant setting.

[0259] 12 patients have been enrolled with safety analysis completed on the first 6 patients and the response rate evaluable after 2 cycles in the first 8 patients. There were no unique or unexpected toxicities with no grade 4 toxicities or neuropathy greater than grade 1. Response data were confir-med on only the first 2 cycles of therapy (first evaluation point) in 6 patients. Two patients have completed 6 cycles with 1 partial response and 1 stable disease. Of the first 8 patients after 2 cycles, there were 2 partial responses and 4 with stable disease.
[0260] These results show that combination of capecitabine and weekly AbraxaneTM at effective doses is feasible with no novel toxicities to date.
AbraxaneTM
related toxicity was mainly neutropenia without clinical consequences, and hand foot syndrome was the major toxicity of capecitabine.

Example 17. Pilot Study of Dose-Dense Doxorubicin Plus Cyclophosphamide Followed by nab-paclitaxel (AbraxaneTM) in Patients with Early-Stage Breast Cancer [0261] The objective of this study was to evaluate the toxicity of doxorubicin (adriamycin) plus cyclophosphamide followed by AbraxaneTM in early stage breast cancer.
[0262] Patients had operable, histologically confirmed breast adenocarcinoma of an early stage. The patients received doxorubicin (adriamycin) 60 mg/m2 plus cyclophosphamide 600 mg/m2 (AC) every 2 weeks for 4 cycles followed by AbraxaneTM
260 mg/m2 every two weeks for 4 cycles.
[0263] 30 patients received 4 cycles of AC, and 27 of 29 patients received 4 cycles of AbraxaneTM; 33% of patients received pegfilgrastim (Neulasta ) for lack of recovery of ANC (absolute neutrophil count) during AbraxaneTM. Nine patients (31%) had AbraxaneTM
dose reductions due to non-hematologic toxicity. A total of 9 patients had grade 2 and 4 patients had grade 3 peripheral neuropathy (PN); PN improved by >1. grade within a median of 28 days.
[0264] These results indicate that dose-dense therapy with doxorubicin (60 mg/mZ) plus cyclophosphamide (600 mg/m2) every 2 weeks for 4 cycles followed by dose-dense AbraxaneTM (260 mg/m2) every 2 weeks for 4 cycles was well tolerated in patients with early-stage breast cancer.

Example 18. Weekly nab-Paclitaxel (AbraxaneTM) as First Line Treatment of Metastatic Breast Cancer with Trastuzumab Add On for HER-2/neac-Positive Patients [0265] The puipose of the current study was to move weekly Abraxane to a front-line setting and add trastuzumab for HER2/faeu-positive patients.
[0266] This phase II, open-label study included 20 HER2-postivive and 50 HER2-negative patients with locally advanced or metastatic breast cancer.
AbraxaneTM was given at 125 mg/ma by 30 minute i.v. inftision on days 1, 8, and 15 followed by a week of rest.
Trastuzurnab was given concurrently with study treatinent for patients who were HER2-positive. The primary endpoint was response rate and the secondary endpoints were time to progression (TTP), overall survival (OS), and toxicity.
[0267] In the safety population, 23 patients received a median of 3 cycles of AbraxaneTM to date. The most common treatment-related adverse event was grade neutropenia (8.7%) with no grade 4 adverse events. One out of 4 evaluable patients responded to therapy.

Example 19. Phase I Trial of iaab-Paclitaxel (AbraxaneTM) and Carboplatin [02681 The aim of the current study was to determine the maximum tolerated dose of AbraxaneTM (botll weekly and every 3 weeks) with carboplatin AUC = 6 and to compare the effects of sequence of administration on pharmacolcinetics (PK).
[0269] Patients with histologically or cytologically documented malignancy that progressed after "standard therapy" were included. Arm 1 received AbraxaneTM
every 3 weeks in a dose escalation format based on cycle 1 toxicities (220, 260, 300, 340 mg/m2) every 3 weeks followed by carboplatin AUC = 6. Arm 2 received weelcly (days 1, 8, 15 followed by 1 week off) AbraxaneTM (100, 125, 150 mg/m2) followed by carboplatin AUC
= 6. For the PK portion of the study, AbraxaneTM was followed by carboplatin in cycle 1 and the order of administration reversed in cycle 2 with PK levels detennined at initial 6, 24, 48 and 72 hours.
[0270] On the every 3 weelcs schedule, neutropenia, thrombocytopenia and neuropathy were the most common grade 3/4 toxicities (3/17 each). On the weekly schedule, neutropenia 5/13 was the most common grade 3/4 toxicity. The best responses to weekly administration at the highest dose of 125 mg/m2 (n = 6) were 2 partial responses (pancreatic cancer, melanoma) and 2 stable disease (NSCLC). The best responses to the every tllree week administration at the highest dose of 340 mg/m2 (n = 5) were 1 stable disease (NSCLC) and 2 partial responses (SCLC, esophageal).
[0271] These data indicate activity of combination of AbraxaneTM and carboplatin.
The MTD for the weekly administration was 300 mg/m2, and for the once every 3 week administration was 100 mg/ma.

Example 20. Phase II Trial of Dose-Dense Gemcitabine, Epirubicin, and laccb-Paclitaxel (AbraxaneTn~ (GEA) in Locally Advanced/Inflammatory Breast Cancer [0272] In an open-label, phase II study an induction/neoadjuvant therapy regime was instituted prior to local intervention. The therapy regime was gemcitabine 2000 mg/mZ

i.v. every 2 weeks for 6 cycles, epirubicin 50 mg/m2 every 2 weeks for 6 cycles, AbraxaneTM 175 mg/ma every 2 weeks for 6 cycles, with pegfilgrastim 6 mg s.c.
on day 2 every 2 weeks. The postoperative/adjuvant therapy regime after local intervention was gemcitabine 2000 mg/m2 every 2 weeks for 4 cycles, AbraxaneTM 220 mg/ma every weeks for 4 cycles and pegfilgrastim 6 mg s.c. day every 2 weeks. Patients included females with histologically confirmed locally advanced/inflammatory adenocarcinoma of the breast.

Example 21. Cytotoxic activity of nab-rapamycin in combination with AbraxaneTM
on vascular smooth muscle cells [0273] Vascular smooth inuscle cells (VSMC) were seeded onto 96 wells plates in the presence of increasing concentrations of nab-rapamycin and 0 M, 1 M, 10 M, or 100 M of AbraxaneTM (ABI-007). To evaluate the cytotoxic effect of nab-rapamycin and AbraxaneTM, treated VSMCs were stained with ethidium homodimer-1 (Invitrogen, Carlsbad CA) and analyzed for red fluorescence. Ethidium homodimer-1 is a high-affinity, fluorescent nucleic acid stain that is only able to pass through compromised membranes of dead cells to stain nucleic acids. As shown in Fig. 7A, nab-rapamycin, by itself, exhibited dose-dependent cell killing as demonstrated by increasing fluorescence. Cell killing by nab-rapamycin was not enhanced by AbraxaneTM at 1 M or 10 M; however, it was greatly enhanced by AbraxaneTM at 100 M (ANOVA, p < 0.0001). Cells stained with ethidium homodimer-1 as shown in Fig. 7A were also exposed to calcein. Calcein AM
(Invitrogen) is a non-fluorescent molecule that is hydrolyzed into fluorescent calcein by nonspecific cytosolic esterases. Live cells exposed to calcein AM exhibit bright green fluorescence as they are able to generate the fluorescent product and retain it. As shown in Fig. 7B, nab-rapamycin exhibited dose dependent cytotoxic activity as shown by a reduced amount of fluorescent staining by calcein. This reduction in fluorescence was enhanced by coincubation with AbraxaneTM in a dose dependent manner. ANOVA statistic gave p<
0.0001 at all drug concentrations of AbraxaneTM.

Example 22. Cytotoxic activity of nab-rapamycin in combination with AbraxaneTM
against HT29 (human colon carcinoma) tumor xenograft.

[0274] Nude mice were implanted with 106 HT29 cells on their right flanks.
Treatment was initiated once the tumor were palpable and were greater than 100-200 mm3.
The mice were randomly sorted into 4 groups (n= 8 per group). Group 1 received saline 3 times weekly for 4 weeks, i.v.; Group 2 received AbraxaneTM at 10 mg/kg, daily for 5 days, i.p.; Group 3 received nab-rapamycin at 40 mg/kg, 3 times weekly for 4 weeks, i.v.; and Group 4 received both nab-rapamycin (40 mg/kg, 3 times weekly for 4 weeks, i.v.) and AbraxaneTM (10 mg/kg, daily for 5 days, i.p.). As shown in Fig. 8, the tumor suppression was greater for the AbraxaneTM plus nab-rapamycin combination therapy than for either single therapy group.

Example 23. Cytotoxic activity of nab-17-AAG in combination with AbraxaneTM
against H358 (human lung carcinoma) tumor xenograft.

[0275] Nude mice were implanted with 107 H358 cells on their right flaiilcs.
Treatment was initiated once the tumors were palpable and were greater than 100-200 mm3.
The mice were randomly sorted into 4 groups (n= 8 per group). Group 1 received saline 3 times weekly for 4 weeks, i.v.; Group 2 received AbraxaneTM at 10 mg/kg, daily for 5 days, i.p.; Group 3 received nab-17-AAG at 80 mg/kg, 3 times weekly for 4 weeks, i.v.; and Group 4 received both nab-17-AAG (80 mg/lcg, 3 times weekly for 4 weeks, i.v.) and AbraxaneTM (10 mg/kg, daily for 5 days, i.p.). As shown in Fig. 9, the tumor suppression was greater for the nab-17-AAG plus AbraxaneTM combination therapy than for either single therapy group.
[0276] Although the foregoing invention has been described in some detail by way of illustration and example for purposes of clarity of understanding, it is apparent to those skilled in the art that certain minor changes and modifications will be practiced. Therefore, the description and examples should not be construed as limiting the scope of the invention.
[0277] All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference to the saine extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein.
,[0278] PrefeiTed embodiments of this invention are described herein, including the best mode known to the inventors for carrying out the invention. Variations of those preferred embodiments may become apparent to those of ordinary skill in the art upon reading the foregoing description. The inventors expect skilled artisans to employ such variations as appropriate, and the inventors intend for the invention to be practiced otherwise than as specifically described herein. Accordingly, this invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law. Moreover, any combination of the above-described elements in all possible variations thereof is encompassed by the invention unless otherwise indicated herein or otherwise clearly contradicted by context.

Claims (49)

1. A method of treating a proliferative disease in an individual comprising administering to the individual:

a) an effective amount of a composition comprising nanoparticles comprising a taxane and an albumin, and b) an effective amount of at least one other chemotherapeutic agent, wherein said chemotherapeutic agent is selected from the group consisting of antimetabolites, platinum-based agents, alkylating agents, tyrosine kinase inhibitors, anthracycline antibiotics, vinca alkloids, proteasome inhibitors, macrolides, and topoisomerase inhibitors.
2. The method according to claim 1, wherein said chemotherapeutic agent is an antimetabolite agent.
3. The method according to claim 2, wherein said antimetabolite agent is gemcitabine, capecitabine, or fluorouracil.
4. The method according to claim 1, wherein said chemotherapeutic agent is a platinum-based agent.
5. The method according to claim 4, wherein said platinum-based agent is cisplatin or carboplatin.
6. The method according to claim 1, wherein said chemotherapeutic agent is a tyrosine kinase inhibitor.
7. The method according to claim 6, wherein said tyrosine kinase inhibitor is lapatinib.
8. The method according to claim 1, wherein the proliferative disease is cancer.
9. The method according to claim 8, wherein the cancer is breast cancer.
10. The method according to claim 8, wherein the cancer is lung cancer.
11. The method according to claim 1, wherein the composition comprising nanoparticles comprising taxane and albumin and the chemotherapeutic agent are administered simultaneously.
12. The method according to claim 1, wherein the composition comprising nanoparticles of taxane comprising albumin and the chemotherapeutic agent are administered sequentially.
13. The method according to claim 1, wherein the taxane is paclitaxel.
14. The method according to claim 13, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
15. The method according to claim 14, wherein the weight ratio of the albumin and the paclitaxel in the nanoparticle composition is less than about 9:1.
16. The method according to claim 14, wherein the nanoparticle composition is substantially free of Cremophor.
17. The method according to claim 1, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
18. The method according to claim 17, wherein the weight ratio of the albumin and the taxane in the nanoparticle composition is less than about 9:1.
19. The method according to claim 17, wherein the nanoparticle composition is substantially free of Cremophor.
20. The method according to claim 1, wherein the individual is a human.
21. A method of treating a tumor in an individual comprising:
a) a first therapy comprising administering to the individual an effective amount of a composition comprising nanoparticles coinprising a taxane and an albumin, and b) a second therapy comprising radiation therapy, surgery, or combinations thereof.
22. The method of claim 21, wherein the second therapy is radiation therapy.
23. The method of claim 22, wherein the radiation therapy occurs before the first therapy.
24. The method of claim 21, wherein the second therapy is surgery.
25. The method according to claim 21, wherein the taxane is paclitaxel.
26. The method according to claim 25, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
27. The method according to claim 26, wherein the weight ratio of the albumin and the paclitaxel in the nanoparticle composition is less than about 9:1.
28. The method according to claim 26, wherein the nanoparticle composition is substantially free of Cremophor.
29. The method according to claim 21, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
30. The method according to claim 29, wherein the weight ratio of the albumin and the taxane in the nanoparticle composition is less than about 9:1.
31. The method according to claim 29, wherein the nanoparticle composition is substantially free of Cremophor.
32. The method according to claim 21, wherein the individual is a human.
33. A method of administering a composition comprising nanoparticles comprising a taxane and an albumin to an individual, wherein the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of taxane in the composition at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime.
34. The method according to claim 33, wherein the dose of taxane is about 0.25% to about 8% of its maximum tolerated dose following a traditional dosing regime.
35. The method according to claim 33, wherein the composition is administered at least three times a week.
36. The method according to claim 33, wherein said taxane is paclitaxel.
37. The method according to claim 36, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
38. The method according to claim 37, wherein the weight ratio of the albumin and the paclitaxel in the nanoparticle composition is less than about 9:1.
39. The method according to claim 37, wherein the nanoparticle composition is substantially free of Cremophor.
40. The method according to claim 33, wherein the average diameter of the nanoparticles in the composition is no greater than about 200 nm.
41. The method according to claim 40, wherein the weight ratio of the albumin and the taxane in the nanoparticle composition is less than about 9:1.
42. The method according to claim 40, wherein the nanoparticle composition is substantially free of Cremophor.
43. The method according to claim 33, wherein the individual is a human.
44. A method of treating a proliferative disease in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane and an albumin; and b) an effective amount of a composition comprising nanoparticles coinprising at least one other chemotherapeutic agent and an albumin.
45. The method of claim 44, wherein the taxane is paclitaxel.
46. The method of claim 44, wherein the chemotherapeutic agent is rapamycin.
47. The method of claim 44, wherein the chemotherapeutic agent is 17AAG.
48. The method of claim 44, wherein the individual is human.
49. The method of claim 44, wherein the proliferative disease is cancer.
CA2598239A 2005-02-18 2006-02-21 Nanoparticulate formulations of taxanes and carrier proteins for use in combination chemotherapy Active CA2598239C (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
CA3054535A CA3054535A1 (en) 2005-02-18 2006-02-21 Combinations and modes of administration of therapeutic agents and combination therapy
CA2902071A CA2902071C (en) 2005-02-18 2006-02-21 Combination chemotherapy regimens using albumin/paclitaxel nanoparticles and nucleoside analogs

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US65424505P 2005-02-18 2005-02-18
US60/654,245 2005-02-18
PCT/US2006/006167 WO2006089290A1 (en) 2005-02-18 2006-02-21 Combinations and modes of administration of therapeutic agents and combination therapy

Related Child Applications (2)

Application Number Title Priority Date Filing Date
CA3054535A Division CA3054535A1 (en) 2005-02-18 2006-02-21 Combinations and modes of administration of therapeutic agents and combination therapy
CA2902071A Division CA2902071C (en) 2005-02-18 2006-02-21 Combination chemotherapy regimens using albumin/paclitaxel nanoparticles and nucleoside analogs

Publications (2)

Publication Number Publication Date
CA2598239A1 true CA2598239A1 (en) 2006-08-24
CA2598239C CA2598239C (en) 2019-10-29

Family

ID=36593193

Family Applications (3)

Application Number Title Priority Date Filing Date
CA2902071A Active CA2902071C (en) 2005-02-18 2006-02-21 Combination chemotherapy regimens using albumin/paclitaxel nanoparticles and nucleoside analogs
CA3054535A Abandoned CA3054535A1 (en) 2005-02-18 2006-02-21 Combinations and modes of administration of therapeutic agents and combination therapy
CA2598239A Active CA2598239C (en) 2005-02-18 2006-02-21 Nanoparticulate formulations of taxanes and carrier proteins for use in combination chemotherapy

Family Applications Before (2)

Application Number Title Priority Date Filing Date
CA2902071A Active CA2902071C (en) 2005-02-18 2006-02-21 Combination chemotherapy regimens using albumin/paclitaxel nanoparticles and nucleoside analogs
CA3054535A Abandoned CA3054535A1 (en) 2005-02-18 2006-02-21 Combinations and modes of administration of therapeutic agents and combination therapy

Country Status (26)

Country Link
US (11) US8034375B2 (en)
EP (4) EP2301531B1 (en)
JP (8) JP5769358B2 (en)
KR (8) KR101764375B1 (en)
CN (4) CN103169729A (en)
AT (1) ATE531365T1 (en)
AU (1) AU2006213999B2 (en)
BR (1) BRPI0607809A2 (en)
CA (3) CA2902071C (en)
CY (3) CY1112907T1 (en)
DK (3) DK2301531T3 (en)
ES (3) ES2678448T3 (en)
HK (3) HK1109589A1 (en)
HU (2) HUE049789T2 (en)
IL (4) IL185288A (en)
LT (2) LT2301531T (en)
LU (1) LU92484I2 (en)
MX (2) MX2007009960A (en)
NZ (2) NZ595313A (en)
PL (3) PL3248600T3 (en)
PT (3) PT1853250E (en)
RU (2) RU2452482C2 (en)
SG (1) SG166775A1 (en)
SI (3) SI3248600T1 (en)
WO (1) WO2006089290A1 (en)
ZA (1) ZA200707491B (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9101543B2 (en) 2005-02-18 2015-08-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9561288B2 (en) 2005-02-18 2017-02-07 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy

Families Citing this family (183)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US20030133955A1 (en) * 1993-02-22 2003-07-17 American Bioscience, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US20070117862A1 (en) * 1993-02-22 2007-05-24 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US8137684B2 (en) 1996-10-01 2012-03-20 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
EP1023050B1 (en) * 1997-06-27 2013-09-25 Abraxis BioScience, LLC Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
US8853260B2 (en) 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
CN102516417B (en) 2002-09-06 2014-12-10 天蓝制药公司 Cyclodextrin-based polymers for therapeutics delivery
EP2359859A1 (en) * 2002-12-09 2011-08-24 Abraxis BioScience, LLC Compositions comprising albumin nanoparticles and methods of delivery of pharmacological agents
JP4903036B2 (en) 2003-01-06 2012-03-21 アンジオケム・インコーポレーテッド Aprotinin and analogs as carriers that cross the blood brain barrier
US20070166388A1 (en) * 2005-02-18 2007-07-19 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
KR20080027253A (en) * 2005-06-16 2008-03-26 미리어드 제네틱스, 인크. Pharmaceutical compositions and use thereof
EP2471555A3 (en) 2005-07-15 2012-10-17 Angiochem Inc. Use of aprotinin polypeptides as carriers in pharmaceutical conjugates
DE102005039579B4 (en) * 2005-08-19 2022-06-30 Magforce Ag Method for introducing therapeutic substances into cells
WO2007027941A2 (en) 2005-08-31 2007-03-08 Abraxis Bioscience, Llc. Compositions and methods for preparation of poorly water soluble drugs with increased stability
ES2720603T3 (en) * 2005-08-31 2019-07-23 Abraxis Bioscience Llc Compositions comprising low water soluble pharmaceutical agents and antimicrobial agents
RU2010104916A (en) * 2006-08-16 2011-08-20 Михаил В. Благосклонный (US) METHOD FOR PREVENTION AND TREATMENT OF AGE DISEASES
US20080280987A1 (en) * 2006-08-31 2008-11-13 Desai Neil P Methods of inhibiting angiogenesis and treating angiogenesis-associated diseases
CN101568332A (en) * 2006-08-31 2009-10-28 阿布拉科斯生物科学公司 Methods of inhibiting angiogenesis and treating angiogenesis-associated diseases
US8178564B2 (en) 2006-11-06 2012-05-15 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168661B2 (en) 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8173686B2 (en) 2006-11-06 2012-05-08 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
US8168662B1 (en) 2006-11-06 2012-05-01 Poniard Pharmaceuticals, Inc. Use of picoplatin to treat colorectal cancer
PL2117520T3 (en) * 2006-12-14 2019-03-29 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
JP2010516625A (en) 2007-01-24 2010-05-20 インサート セラピューティクス, インコーポレイテッド Polymer-drug conjugates with tether groups for controlled drug delivery
AU2015271950B2 (en) * 2007-03-07 2017-09-28 Abraxis Bioscience, Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
KR102047634B1 (en) * 2007-03-07 2019-11-21 아브락시스 바이오사이언스, 엘엘씨 Nanoparticle comprising rapamycin and albumin as anticancer agent
US8642067B2 (en) 2007-04-02 2014-02-04 Allergen, Inc. Methods and compositions for intraocular administration to treat ocular conditions
NZ580869A (en) * 2007-04-10 2011-02-25 Myriad Pharmaceuticals Inc Methods for treating cancer
AU2008236994A1 (en) * 2007-04-10 2008-10-16 Myrexis, Inc. Method of treating melanoma
NZ580868A (en) * 2007-04-10 2011-07-29 Myriad Pharmaceuticals Inc Dosages and methods for the treatment of cancer
CA2720982A1 (en) * 2007-04-10 2008-10-16 Myrexis, Inc. Method of treating brain cancer
WO2008124828A1 (en) * 2007-04-10 2008-10-16 Myriad Genetics, Inc. Methods for treating vascular disruption disorders
WO2008137148A2 (en) 2007-05-03 2008-11-13 Abraxis Bioscience, Llc Methods and compositions for treating pulmonary hypertension
WO2008140751A1 (en) * 2007-05-11 2008-11-20 Champions Biotechnology, Inc. Human leiosarcoma and non small cell lung cancer lung xenograft models
US9365634B2 (en) 2007-05-29 2016-06-14 Angiochem Inc. Aprotinin-like polypeptides for delivering agents conjugated thereto to tissues
WO2008150532A1 (en) 2007-06-01 2008-12-11 Abraxis Bioscience, Llc Methods and compositions for treating recurrent cancer
US9545384B2 (en) * 2007-06-04 2017-01-17 Bend Research, Inc. Nanoparticles comprising drug, a non-ionizable cellulosic polymer and tocopheryl polyethylene glocol succinate
KR20100023862A (en) * 2007-06-22 2010-03-04 싸이도우스 엘엘씨. Solubilized formulation of docetaxel without tween 80
CA2742388C (en) * 2007-11-08 2019-02-19 Virginia Tech Intellectual Properties, Inc. Thiolated paclitaxels for reaction with gold nanoparticles as drug delivery agents
US9233078B2 (en) 2007-12-06 2016-01-12 Bend Research, Inc. Nanoparticles comprising a non-ionizable polymer and an Amine-functionalized methacrylate copolymer
WO2009078755A1 (en) * 2007-12-19 2009-06-25 Ardenia Investments, Ltd. Drug delivery system for administration of a water soluble, cationic and amphiphilic pharmaceutically active substance
JP2011516412A (en) * 2008-03-05 2011-05-26 ビカス セラピューティクス,エルエルシー Compositions and methods for the treatment of cancer and mucositis
AU2009234127B2 (en) * 2008-04-10 2015-04-30 Abraxis Bioscience, Llc Compositions of hydrophobic taxane derivatives and uses thereof
RU2518240C2 (en) * 2008-04-18 2014-06-10 Ангиочем Инк. Composition based on hydrophobic agents and method for preparing it (versions)
PT2299984T (en) 2008-05-15 2019-02-25 Celgene Corp Oral formulations of cytidine analogs and methods of use thereof
CN102695520A (en) * 2008-05-27 2012-09-26 昂科利蒂克斯生物科技公司 Modulating interstitial pressure and oncolytic viral delivery and distribution
UA104147C2 (en) * 2008-09-10 2014-01-10 Новартис Аг Pyrrolidine dicarboxylic acid derivative and use thereof in the treatment of proliferative diseases
EP2346896A4 (en) 2008-10-15 2014-06-25 Angiochem Inc Etoposide and doxorubicin conjugates for drug delivery
US8921314B2 (en) 2008-10-15 2014-12-30 Angiochem, Inc. Conjugates of GLP-1 agonists and uses thereof
CA2745524C (en) 2008-12-05 2020-06-09 Angiochem Inc. Conjugates of neurotensin or neurotensin analogs and uses thereof
AU2009324464B2 (en) * 2008-12-11 2016-05-05 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
MX2011006685A (en) 2008-12-17 2011-09-27 Angiochem Inc Membrane type-1 matrix metalloprotein inhibitors and uses thereof.
MX2011009452A (en) * 2009-03-13 2011-11-29 Abraxis Bioscience Llc Combination therapy with thiocolchicine derivatives.
WO2010114770A1 (en) * 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-agent conjugates, particles, compositions, and related methods of use
EA201171195A8 (en) * 2009-03-30 2014-08-29 Серулин Фарма Инк. CONJUGATES, PARTICLES, POLYMER-AGENT COMPOSITIONS AND METHODS OF THEIR APPLICATION
WO2010114768A1 (en) * 2009-03-30 2010-10-07 Cerulean Pharma Inc. Polymer-epothilone conjugates, particles, compositions, and related methods of use
MY175260A (en) * 2009-04-15 2020-06-17 Abraxis Bioscience Llc Prion free nanoparticle compositions and methods of making thereof
US9173891B2 (en) 2009-04-20 2015-11-03 Angiochem, Inc. Treatment of ovarian cancer using an anticancer agent conjugated to an angiopep-2 analog
EP2437738B1 (en) * 2009-06-05 2018-08-08 Joan M. Caron Methods and compositions for the treatment of cancer
US9161988B2 (en) 2009-07-02 2015-10-20 Angiochem Inc. Multimeric peptide conjugates and uses thereof
US9340697B2 (en) 2009-08-14 2016-05-17 Nano-C, Inc. Solvent-based and water-based carbon nanotube inks with removable additives
MY162514A (en) 2009-08-25 2017-06-15 Abraxis Bioscience Llc Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors
NZ598801A (en) * 2009-09-18 2014-07-25 Abraxis Bioscience Llc Use of the sparc microenvironment signature in the treatment of cancer
US20110092579A1 (en) * 2009-10-19 2011-04-21 Scidose Llc Solubilized formulation of docetaxel
US7772274B1 (en) 2009-10-19 2010-08-10 Scidose, Llc Docetaxel formulations with lipoic acid
US8912228B2 (en) 2009-10-19 2014-12-16 Scidose Llc Docetaxel formulations with lipoic acid
US8541465B2 (en) * 2009-10-19 2013-09-24 Scidose, Llc Docetaxel formulations with lipoic acid and/or dihydrolipoic acid
US20130115295A1 (en) * 2009-11-22 2013-05-09 Qiang Wang Rare Earth-Doped Up-Conversion Nanoparticles for Therapeutic and Diagnostic Applications
NZ601348A (en) 2010-01-12 2014-07-25 Nestec Sa Methods for predicting response of triple-negative breast cancer to therapy
EP2898884B1 (en) 2010-03-26 2018-05-09 Abraxis BioScience, LLC Methods of treatment of hepatocellular carcinoma
US10660965B2 (en) * 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
SI2552415T1 (en) * 2010-03-29 2017-03-31 Abraxis Bioscience, Llc Methods of treating cancer
WO2011127405A1 (en) * 2010-04-08 2011-10-13 Sanford-Burnham Medical Research Institute Methods and compositions for enhanced delivery of compounds
EP2382993A1 (en) 2010-04-19 2011-11-02 KTB Tumorforschungsgesellschaft mbH Combination of drugs with protein-binding prodrugs
MX343671B (en) 2010-06-02 2016-11-16 Abraxis Bioscience Llc * Methods of treating bladder cancer.
CN103002905A (en) * 2010-06-03 2013-03-27 阿布拉西斯生物科学有限责任公司 Use of the SPARC microenvironment signature in the treatment of cancer
AU2011261685B2 (en) * 2010-06-04 2016-02-11 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
EP3056201A1 (en) * 2010-06-07 2016-08-17 Abraxis BioScience, LLC Combination therapy methods for treating proliferative diseases
KR101223484B1 (en) * 2010-10-05 2013-01-17 한국과학기술연구원 HUMAN SERUM ALBUMIN-siRNA NANO-SIZED CARRIER SYSTEM
US8884027B2 (en) 2010-10-22 2014-11-11 University Of Rochester Melampomagnolide B derivatives as antileukemic and cytotoxic agents
JP2014505666A (en) * 2010-11-18 2014-03-06 ザ ジェネラル ホスピタル コーポレイション Antihypertensive composition and use for cancer treatment
CN103269716B (en) 2010-12-02 2015-05-27 昂科利蒂克斯生物科技公司 Lyophilized viral formulations
WO2012075379A2 (en) 2010-12-02 2012-06-07 Oncolytics Biotech Inc. Liquid viral formulations
KR102108959B1 (en) * 2011-04-28 2020-05-11 아브락시스 바이오사이언스, 엘엘씨 Intravascular delivery of nanoparticle compositions and uses thereof
DK2707030T3 (en) 2011-05-09 2020-05-18 Mayo Found Medical Education & Res CANCER TREATMENTS
IN2014CN02703A (en) * 2011-10-14 2015-07-03 Hoffmann La Roche
US9125884B2 (en) 2011-11-01 2015-09-08 Celgene Corporation Methods for treating cancers using oral formulations of cytidine analogs
WO2013085902A1 (en) 2011-12-05 2013-06-13 The University Of Texas M.D. Combination therapy methods for treating an inflammatory breast cancer
US9585960B2 (en) 2011-12-14 2017-03-07 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
GB201121924D0 (en) * 2011-12-20 2012-02-01 Fahy Gurteen Labs Ltd Detection of breast cancer
EP2630971B8 (en) 2012-02-21 2017-12-13 Vergell Medical S.A. Combinations of albumin-based drug delivery systems
AU2013243873B2 (en) 2012-04-04 2016-08-25 Halozyme, Inc. Combination therapy with an anti - hyaluronan agent and a tumor - targeted taxane
WO2014026143A1 (en) * 2012-08-09 2014-02-13 Pono Corporation Conjugated anti-microbial compounds and conjugated anti-cancer compounds and uses thereof
PT2892535T (en) * 2012-09-04 2021-12-15 Novartis Ag Method of adjuvant cancer treatment
WO2014055415A1 (en) 2012-10-01 2014-04-10 Mayo Foundation For Medical Education And Research Cancer treatments
US20140094432A1 (en) 2012-10-02 2014-04-03 Cerulean Pharma Inc. Methods and systems for polymer precipitation and generation of particles
US9149455B2 (en) * 2012-11-09 2015-10-06 Abraxis Bioscience, Llc Methods of treating melanoma
TWI598341B (en) 2012-11-12 2017-09-11 伊格尼塔公司 Bendamustine derivatives and methods of using same
US9757432B2 (en) 2012-11-14 2017-09-12 Ohio State Innovation Foundation Materials and methods useful for treating glioblastorna
MX370519B (en) 2012-11-30 2019-12-17 Novomedix Llc Substituted biaryl sulfonamides and the use thereof.
US20140163095A1 (en) * 2012-12-04 2014-06-12 Eisai R&D Management Co., Ltd. Use of eribulin in the treatment of breast cancer
CN103333179B (en) * 2012-12-21 2017-06-16 百奥泰生物科技(广州)有限公司 Class maytansine derivative and its production and use
US9511046B2 (en) 2013-01-11 2016-12-06 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US20140199405A1 (en) * 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on mutation status of k-ras
US20140199404A1 (en) * 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on level of a nucleoside transporter
US9962452B2 (en) 2013-02-04 2018-05-08 Zhuhai Beihai Biotech Co., Ltd. Soluble complexes of drug analogs and albumin
KR102148551B1 (en) * 2013-02-11 2020-08-26 아브락시스 바이오사이언스, 엘엘씨 Methods of treating melanoma
ES2804323T3 (en) 2013-03-12 2021-02-05 Abraxis Bioscience Llc Lung Cancer Treatment Procedures
WO2014143613A1 (en) 2013-03-13 2014-09-18 Abraxis Bioscience, Llc Methods of treatment of pediatric solid tumor
SI2968294T1 (en) 2013-03-13 2019-08-30 Oncoceutics, Inc. 7-benzyl-10-(2-methylbenzyl)-2,6,7,8,9,10-hexahydroimidazo(1,2-a)pyrido(4,3-d)pyrimidin-5(3h)-on for use in the treatment of cancer
US9376437B2 (en) 2013-03-13 2016-06-28 Oncoceutics, Inc 7-benzyl-4-(2-methylbenzyl)-2,4,6,7,8,9-hexahydroimidazo[1,2-a]pyrido[3,4-e]pyrimidin-5(1H)-one, salts thereof and methods of using the same in combination therapy
US9688679B2 (en) 2013-03-13 2017-06-27 Oncoceutics, Inc. 7-benzyl-4-(methylbenzyl)-2,4,6,7,8,9-hexahydroimidazo[1,2-A]pyrido[3,4-E]pyrimidin-5 (1H)-one, salts thereof and methods of using the same in combination therapy
CA2903548A1 (en) 2013-03-14 2014-09-25 Abraxis Bioscience, Llc Methods of treating bladder cancer
US20150104520A1 (en) * 2013-04-05 2015-04-16 Vuong Trieu Nanoparticle Formulations in Biomarker Detection
KR101329646B1 (en) * 2013-05-02 2013-11-14 주식회사 지니스 Targeting-enhancing anticancer nanoparticles and preparation method the same
WO2015042234A1 (en) * 2013-09-20 2015-03-26 Igdrasol Conditionally stable micelle compositions for cancer treatment including ovarian cancer
US20170065723A1 (en) * 2013-09-27 2017-03-09 Cerulean Pharma Inc. Treatment of cancer
CN105873529A (en) * 2013-10-25 2016-08-17 墨卡托医疗系统公司 Maintenance of bronchial patency by local delivery of cytotoxic, cytostatic, or anti-neoplastic agent
US10842969B2 (en) 2013-10-25 2020-11-24 Mercator Medsystems, Inc. Systems and methods of treating malacia by local delivery of hydrogel to augment tissue
JP6670751B2 (en) * 2013-11-15 2020-03-25 オンコシューティクス インコーポレイテッドOncoceutics,Inc. 7-benzyl-4- (2-methylbenzyl) -2,4,6,7,8,9-hexahydroimidazo [1,2-A] pyrido [3,4-E] pyrimidine-5 (1H)- ON, its salt and its use
FI20130341L (en) 2013-11-19 2015-05-20 Safemed Ltd Oy Transport of poorly water-soluble pharmaceuticals with alpha-fetoprotein balanced by metal ions
JP2016539149A (en) * 2013-12-06 2016-12-15 ノバルティス アーゲー Alpha-isoform selective phosphatidylinositol 3-kinase inhibitor dosing regimen
NO2699580T3 (en) 2014-01-24 2018-02-24
EP2924022A1 (en) * 2014-03-27 2015-09-30 INDENA S.p.A. Amorphous form of a thiocolchicine derivative
EA201692502A1 (en) 2014-06-16 2017-09-29 Мэйо Фаундейшн Фор Медикал Эдьюкейшн Энд Рисерч MIEL TREATMENT
WO2015195634A1 (en) 2014-06-17 2015-12-23 Celgne Corporation Methods for treating epstein-barr virus (ebv) associated cancers using oral formulations of 5-azacytidine
WO2015200837A1 (en) 2014-06-27 2015-12-30 Fl Therapeutics Llc Abiraterone derivatives and non-covalent complexes with albumin
CN104434808A (en) 2014-07-03 2015-03-25 石药集团中奇制药技术(石家庄)有限公司 Therapeutic nanoparticles and preparation method thereof
US9446148B2 (en) 2014-10-06 2016-09-20 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10201617B2 (en) 2014-10-24 2019-02-12 Zhuhai Beihai Biotech Co., Ltd. 3-substituted piperidine-2, 6-diones and non-covalent complexes with albumin
WO2016065283A1 (en) * 2014-10-24 2016-04-28 Bishop Alexander James Roy Methods and compositions for enhancing chemotherapy
RS61243B1 (en) 2015-01-30 2021-01-29 Oncoceutics Inc 7-benzyl-4-(4-(trifluoromethyl)benzyl)-1,2,6,7,8,9-hexahydroimidazo[1,2-a]pyrido[3,4-e]pyrimidin-5(4h)-one and salts thereof and their use in therapy
US10705070B1 (en) 2015-03-05 2020-07-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and poorly water soluble drug
US10527604B1 (en) 2015-03-05 2020-01-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and paclitaxel
US10669311B2 (en) 2015-04-23 2020-06-02 Sanford Burnham Prebys Medical Discovery Institute Targeted delivery system and methods of use therefor
DK3307326T3 (en) 2015-06-15 2020-10-19 Angiochem Inc METHODS FOR THE TREATMENT OF LEPTOMENINGAL CARCINOMATOSIS
HRP20211718T1 (en) 2015-06-29 2022-03-04 Abraxis Bioscience, Llc Nanoparticles comprising sirolimus and an albumin for use in treating epithelioid cell tumors
TW201707725A (en) 2015-08-18 2017-03-01 美國馬友醫藥教育研究基金會 Carrier-antibody compositions and methods of making and using the same
TW201713360A (en) 2015-10-06 2017-04-16 Mayo Foundation Methods of treating cancer using compositions of antibodies and carrier proteins
EP3389720A1 (en) 2015-12-18 2018-10-24 The General Hospital Corporation Polyacetal polymers, conjugates, particles and uses thereof
EP3399861A4 (en) 2016-01-07 2019-08-07 Mayo Foundation for Medical Education and Research Methods of treating cancer with interferon
CA3014531A1 (en) 2016-02-12 2017-08-17 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
EP3417289B1 (en) 2016-02-19 2020-12-23 Nant Holdings IP, LLC Methods of immunogenic modulation
WO2017165440A1 (en) 2016-03-21 2017-09-28 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
CA3018340A1 (en) 2016-03-21 2017-09-28 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
EP3475434A4 (en) 2016-03-24 2020-07-29 Nantcell, Inc. Sequence arrangements and sequences for neoepitope presentation
US10618969B2 (en) 2016-04-06 2020-04-14 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
JP2019517490A (en) 2016-06-02 2019-06-24 イノファーマックス インコーポレイテッド Metronomic oral gemcitabine for cancer therapy
SG11201811074RA (en) 2016-06-30 2019-01-30 Nant Holdings Ip Llc Nant cancer vaccine
WO2018026965A1 (en) 2016-08-02 2018-02-08 Isi Life Sciences, Inc. Compositions and methods for detecting cancer cells in a tissue sample
WO2018027205A1 (en) 2016-08-05 2018-02-08 Mayo Foundation For Medical Education And Research Modified antibody-albumin nanoparticle complexes for cancer treatment
CN107714652B (en) * 2016-08-12 2021-03-02 四川科伦药物研究院有限公司 Tesirolimus albumin nano composition and freeze-dried preparation, preparation method and application thereof
CA3035334C (en) 2016-08-31 2022-05-10 Fujifilm Corporation Combination of paclitaxel and 1-(2-deoxy-2-fluoro-4-thio-beta-d-arabinofuranosyl)-cytosine as anti-tumor agent
CA3035378A1 (en) 2016-09-01 2018-03-08 Mayo Foundation For Medical Education And Research Carrier-pd-l1 binding agent compositions for treating cancers
KR20190053203A (en) 2016-09-01 2019-05-17 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 Methods and compositions for targeting T-cell cancers
KR102486055B1 (en) * 2016-09-06 2023-01-09 메이오 파운데이션 포 메디칼 에쥬케이션 앤드 리써치 Paclitaxel-albumin-binder compositions and methods of use and preparation thereof
WO2018048816A1 (en) 2016-09-06 2018-03-15 Mayo Foundation For Medical Education And Research Methods of treating pd-l1 expressing cancer
EP3509635A1 (en) 2016-09-06 2019-07-17 Vavotar Life Sciences LLC Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
AU2017327392B9 (en) * 2016-09-13 2023-10-12 Tiziana Life Sciences Plc Dactinomycin compositions and methods for the treatment of myelodysplastic syndrome and acute myeloid leukemia
CA3047936A1 (en) 2016-11-21 2018-05-24 Bexion Pharmaceuticals, Inc. A combination therapy including sapc-dops for the treatment of pancreatic cancer
CA3044424C (en) 2016-11-21 2023-05-23 Nant Holdings Ip, Llc Fractal combination therapy
US20210128694A1 (en) * 2016-12-21 2021-05-06 Prometic Pharma Smt Limited Methods and compositions for preventing or minimizing epithelial-mesenchymal transition
AU2018219862B2 (en) 2017-02-07 2020-09-17 Nant Holding IP, LLC Maximizing T-cell memory and compositions and methods therefor
US20180235936A1 (en) * 2017-02-17 2018-08-23 University Of Notre Dame Du Lac Cancer treatment methods
AU2018258119B2 (en) 2017-04-24 2021-04-01 Nantcell, Inc. Targeted neoepitope vectors and methods therefor
US10753942B2 (en) 2017-05-15 2020-08-25 Indicator Systems International, Inc. Methods to detect remnant cancer cells
WO2019018813A1 (en) 2017-07-21 2019-01-24 Varian Medical Systems, Inc. Methods of use of ultra-high dose rate radiation and therapeutic agents
JP7250764B2 (en) * 2017-08-18 2023-04-03 コセラ バイオサイエンス, インコーポレイテッド Polymorphic forms of TG02
CA3081750A1 (en) 2017-11-06 2019-05-09 Rapt Therapeutics, Inc. Anticancer agents
US11773187B2 (en) 2018-01-17 2023-10-03 Nantbio, Inc. Enhanced immunogenicity for GPI-anchored antigens
EP3743091A4 (en) 2018-01-26 2021-12-15 The Regents of the University of California Methods and compositions for treatment of angiogenic disorders using anti-vegf agents
EP3773477A1 (en) 2018-04-03 2021-02-17 Vaxess Technologies, Inc. Microneedle comprising silk fibroin applied to a dissolvable base
US11823773B2 (en) 2018-04-13 2023-11-21 Nant Holdings Ip, Llc Nant cancer vaccine strategies
EP3858846A4 (en) * 2018-09-30 2023-08-09 Bai Yao Zhi Da (Beijing) Nanobio Technology Co., Ltd. Nucleic acid nanocarrier medicament, preparation method therefor, pharmaceutical composition thereof, and use thereof
EP3926047A4 (en) * 2018-10-16 2023-08-09 Bai Yao Zhi Da (Beijing) Nanobio Technology Co., Ltd. Nucleic acid nano-carrier drug, preparation method therefor, and pharmaceutical composition and application thereof
WO2020112868A1 (en) * 2018-11-30 2020-06-04 Aileron Therapeutics, Inc. Combination therapy of peptidomimetic macrocycles
RU2706347C1 (en) * 2019-07-24 2019-11-18 ФЕДЕРАЛЬНОЕ ГОСУДАРСТВЕННОЕ БЮДЖЕТНОЕ УЧРЕЖДЕНИЕ "РОССИЙСКИЙ НАУЧНЫЙ ЦЕНТР РАДИОЛОГИИ И ХИРУРГИЧЕСКИХ ТЕХНОЛОГИЙ ИМЕНИ АКАДЕМИКА А.М. ГРАНОВА" МИНИСТЕРСТВА ЗДРАВООХРАНЕНИЯ РОССИЙСКОЙ ФЕДЕРАЦИИ / ФГБУ "РНЦРХТ им. ак. А.М. Гранова" Минздрава России Method of treating operable pancreatic head adenocarcinoma
WO2021086946A1 (en) 2019-10-28 2021-05-06 Abraxis Bioscience, Llc Pharmaceutical compositions of albumin and rapamycin
IL293131A (en) 2019-11-25 2022-07-01 Univ California Long-acting vegf inhibitors for intraocular neovascularization
WO2022232420A1 (en) * 2021-04-29 2022-11-03 Insmed Incorporated Certain n-(1-cyano-2-phenylethyl)-1,4-oxazepane-2-carboxamides for treating cancer
WO2023174210A1 (en) 2022-03-14 2023-09-21 Laekna Limited Combination treatment for cancer
WO2023250117A2 (en) 2022-06-24 2023-12-28 Vaxess Technologies, Inc. Applicator for medicament patch

Family Cites Families (151)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5206018A (en) * 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
US5470571A (en) 1988-01-27 1995-11-28 The Wistar Institute Method of treating human EGF receptor-expressing gliomas using radiolabeled EGF receptor-specific MAB 425
FR2660556B1 (en) 1990-04-06 1994-09-16 Rhone Poulenc Sante MICROSPHERES, THEIR PREPARATION PROCESS AND THEIR USE.
IL98528A0 (en) 1990-06-21 1992-07-15 Merck & Co Inc Pharmaceutical compositions containing hybrid for killing bladder cancer cells
US5399363A (en) 1991-01-25 1995-03-21 Eastman Kodak Company Surface modified anticancer nanoparticles
HU219537B (en) 1991-03-06 2001-05-28 Merck Patent Gmbh. Humanized and chimaeric monoclonal antibodies comprising of them pharmaceutical composition and the antibodies coding sequence containing of expression vectors, and process for the praparation of antibodies
US6515009B1 (en) * 1991-09-27 2003-02-04 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US5811447A (en) * 1993-01-28 1998-09-22 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
CA2086874E (en) * 1992-08-03 2000-01-04 Renzo Mauro Canetta Methods for administration of taxol
US6306421B1 (en) * 1992-09-25 2001-10-23 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
FR2697752B1 (en) * 1992-11-10 1995-04-14 Rhone Poulenc Rorer Sa Antitumor compositions containing taxane derivatives.
US5981568A (en) * 1993-01-28 1999-11-09 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
US6491938B2 (en) * 1993-05-13 2002-12-10 Neorx Corporation Therapeutic inhibitor of vascular smooth muscle cells
ATE264671T1 (en) 1993-02-22 2004-05-15 American Bioscience Inc METHOD FOR THE IN VIVO ADMINISTRATION OF BIOLOGICAL SUBSTANCES AND COMPOSITIONS USABLE THEREFOR
US5362478A (en) 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
US5650156A (en) * 1993-02-22 1997-07-22 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of nutriceuticals and compositions useful therefor
US6749868B1 (en) * 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US6096331A (en) * 1993-02-22 2000-08-01 Vivorx Pharmaceuticals, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US20030068362A1 (en) 1993-02-22 2003-04-10 American Bioscience, Inc. Methods and formulations for the delivery of pharmacologically active agents
US5439686A (en) 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US5997904A (en) * 1993-02-22 1999-12-07 American Bioscience, Inc. Total nutrient admixtures as stable multicomponent liquids or dry powders and methods for the preparation thereof
US6537579B1 (en) * 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
US6528067B1 (en) 1993-02-22 2003-03-04 American Bioscience, Inc. Total nutrient admixtures as stable multicomponent liquids or dry powders and methods for the preparation thereof
US6753006B1 (en) * 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
US5665382A (en) * 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of pharmaceutically active agents for in vivo delivery
US20070117862A1 (en) * 1993-02-22 2007-05-24 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030133955A1 (en) * 1993-02-22 2003-07-17 American Bioscience, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US5665383A (en) * 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of immunostimulating agents for in vivo delivery
US20030073642A1 (en) * 1993-02-22 2003-04-17 American Bioscience, Inc. Methods and formulations for delivery of pharmacologically active agents
US5916596A (en) * 1993-02-22 1999-06-29 Vivorx Pharmaceuticals, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
ATE502664T1 (en) * 1993-07-19 2011-04-15 Angiotech Pharm Inc METHOD OF PRODUCTION OF A STENT WITH ANTI-ANGIOGENIC COMPOSITION
US5994341A (en) * 1993-07-19 1999-11-30 Angiogenesis Technologies, Inc. Anti-angiogenic Compositions and methods for the treatment of arthritis
US6441026B1 (en) * 1993-11-08 2002-08-27 Aventis Pharma S.A. Antitumor compositions containing taxane derivatives
EP0739203A4 (en) 1994-01-14 2000-12-20 Cell Therapeutics Inc Method for treating diseases mediated by cellular proliferation in response to pdgf, egf, fgf and vegf
US5626862A (en) * 1994-08-02 1997-05-06 Massachusetts Institute Of Technology Controlled local delivery of chemotherapeutic agents for treating solid tumors
US5662883A (en) 1995-01-10 1997-09-02 Nanosystems L.L.C. Microprecipitation of micro-nanoparticulate pharmaceutical agents
US5534270A (en) 1995-02-09 1996-07-09 Nanosystems Llc Method of preparing stable drug nanoparticles
US5510118A (en) 1995-02-14 1996-04-23 Nanosystems Llc Process for preparing therapeutic compositions containing nanoparticles
CA2178541C (en) * 1995-06-07 2009-11-24 Neal E. Fearnot Implantable medical device
US5609629A (en) * 1995-06-07 1997-03-11 Med Institute, Inc. Coated implantable medical device
US5744460A (en) * 1996-03-07 1998-04-28 Novartis Corporation Combination for treatment of proliferative diseases
EP0932399B1 (en) * 1996-03-12 2006-01-04 PG-TXL Company, L.P. Water soluble paclitaxel prodrugs
US6441025B2 (en) 1996-03-12 2002-08-27 Pg-Txl Company, L.P. Water soluble paclitaxel derivatives
ATE340586T1 (en) * 1996-07-30 2006-10-15 Novartis Pharma Gmbh PHARMACEUTICAL COMPOSITIONS FOR THE TREATMENT OF TRANSPLANT REJECTION AND AUTOIMMUNE OR INFLAMMATORY CONDITIONS
US5997899A (en) 1996-10-01 1999-12-07 Skyepharma Inc. Method for producing liposomes with increased percent of compound encapsulated
US8137684B2 (en) * 1996-10-01 2012-03-20 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US6495579B1 (en) * 1996-12-02 2002-12-17 Angiotech Pharmaceuticals, Inc. Method for treating multiple sclerosis
US6143276A (en) * 1997-03-21 2000-11-07 Imarx Pharmaceutical Corp. Methods for delivering bioactive agents to regions of elevated temperatures
US6884879B1 (en) * 1997-04-07 2005-04-26 Genentech, Inc. Anti-VEGF antibodies
AU743758B2 (en) 1997-04-07 2002-02-07 Genentech Inc. Anti-VEGF antibodies
EP1023050B1 (en) 1997-06-27 2013-09-25 Abraxis BioScience, LLC Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US8853260B2 (en) 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
US6258084B1 (en) 1997-09-11 2001-07-10 Vnus Medical Technologies, Inc. Method for applying energy to biological tissue including the use of tumescent tissue compression
RS49779B (en) * 1998-01-12 2008-06-05 Glaxo Group Limited, Byciclic heteroaromatic compounds as protein tyrosine kinase inhibitors
EP1071440B1 (en) 1998-04-16 2007-01-03 Eurovita A/S Novel synergistic compositions containing aromatic compounds and terpenoids present in alpinia galanga
CA2362338A1 (en) * 1998-07-30 2000-02-10 Human Rt. Pharmaceutically acceptable composition comprising an aqueous solution of paclitaxel and albumin
US6682758B1 (en) * 1998-12-22 2004-01-27 The United States Of America As Represented By The Department Of Health And Human Services Water-insoluble drug delivery system
US6740665B1 (en) 1999-02-10 2004-05-25 Ramachandran Murali Tyrosine kinase inhibitors and methods of using the same
US6537585B1 (en) * 1999-03-26 2003-03-25 Guilford Pharmaceuticals, Inc. Methods and compositions for treating solid tumors
AU777528B2 (en) 1999-04-22 2004-10-21 Abraxis Bioscience, Llc Long term administration of pharmacologically active agents
CA2371912C (en) 1999-05-21 2010-02-16 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
JP2003500368A (en) 1999-05-24 2003-01-07 ソーナス ファーマシューティカルス,インコーポレイテッド Emulsion vehicle for poorly soluble drugs
EP1267913A2 (en) 1999-11-12 2003-01-02 EntreMed, Inc. Methods for administration of therapeutic agents on an antiangiogenic schedule
US7740841B1 (en) * 2000-01-28 2010-06-22 Sunnybrook Health Science Center Therapeutic method for reducing angiogenesis
IT1318401B1 (en) * 2000-03-17 2003-08-25 Indena Spa N-DESACETYLTHIOCOLCHYCIN DERIVATIVES AND PHARMACEUTICAL COMPOSITIONS WHICH CONTAIN.
CA2406484A1 (en) 2000-04-10 2001-10-18 Moshe Flashner-Barak Method and composition for treating cancer by administration of apoptosis-inducing chemotherapeutic agents
ITMI20001107A1 (en) * 2000-05-18 2001-11-18 Acs Dobfar Spa METHOD FOR TREATMENT OF SOLIC TUMORS BY INCORPORATING PACLITAXEL MICROPARTICLES OF ALBUMIN
ES2280382T3 (en) * 2000-06-30 2007-09-16 Glaxo Group Limited COMPOUNDS OF QUINAZOLINE SALTS DITOSYLATE.
AU2001292548B2 (en) 2000-07-28 2005-06-16 Sloan-Kettering Institute For Cancer Research Methods for treating cell proliferative disorders and viral infections
CZ2003837A3 (en) 2000-09-22 2004-12-15 Bristol-Myers Squibb Company Method of reducing toxicity when employing combined chemotherapies
JP4458746B2 (en) * 2001-01-16 2010-04-28 グラクソ グループ リミテッド How to treat cancer
WO2002056755A2 (en) 2001-01-19 2002-07-25 Bionumerik Pharmaceuticals, Inc. Method for treating cancer
US6548531B2 (en) * 2001-02-09 2003-04-15 Hoffmann-La Roche Inc. Method for cancer therapy
GB0103668D0 (en) * 2001-02-15 2001-03-28 Biointeractions Ltd Methods and clinical devices for the inhibition or prevention of mammalian cell growth
US6770653B2 (en) 2001-03-06 2004-08-03 Bristol-Myers Squibb Company Method and dosage form for treating tumors by the administration of tegafur, uracil, folinic acid, paclitaxel and carboplatin
WO2002069949A2 (en) * 2001-03-06 2002-09-12 Prendergast Patrick T Combination therapy for reduction of toxycity of chemotherapeutic agents
US6752922B2 (en) * 2001-04-06 2004-06-22 Fluidigm Corporation Microfluidic chromatography
US7638161B2 (en) 2001-07-20 2009-12-29 Applied Materials, Inc. Method and apparatus for controlling dopant concentration during BPSG film deposition to reduce nitride consumption
US6872715B2 (en) 2001-08-06 2005-03-29 Kosan Biosciences, Inc. Benzoquinone ansamycins
US7056338B2 (en) * 2003-03-28 2006-06-06 Conor Medsystems, Inc. Therapeutic agent delivery device with controlled therapeutic agent release rates
US20040033271A1 (en) * 2001-12-03 2004-02-19 Seth Lederman Methods for contemporaneous administration of levamisole and 5-fluorouracil
US20040143004A1 (en) 2002-02-26 2004-07-22 Joseph Fargnoli Metronomic dosing of taxanes
ITMI20020681A1 (en) * 2002-03-29 2003-09-29 Acs Dobfar Spa PROCEDURE FOR THE PRODUCTION OF PACLITAXEL AND ALBUMINA NANOPARTICLES
ITMI20020680A1 (en) * 2002-03-29 2003-09-29 Acs Dobfar Spa IMPROVED ANTI-TUMOR COMPOSITION BASED ON PACLITAXEL AND METHOD FOR ITS OBTAINING
US20040126400A1 (en) * 2002-05-03 2004-07-01 Iversen Patrick L. Delivery of therapeutic compounds via microparticles or microbubbles
US7794743B2 (en) * 2002-06-21 2010-09-14 Advanced Cardiovascular Systems, Inc. Polycationic peptide coatings and methods of making the same
WO2004006859A2 (en) 2002-07-16 2004-01-22 Sonus Pharmaceuticals, Inc. Platinum compound
RS20050106A (en) 2002-08-19 2007-11-15 Pfizer Products Inc., Combination therapy for hyperproliferative diseases
US20040047835A1 (en) 2002-09-06 2004-03-11 Cell Therapeutics, Inc. Combinatorial drug therapy using polymer drug conjugates
US20050158375A1 (en) * 2002-11-15 2005-07-21 Toshikiro Kimura Pharmaceutical composition containing liposomes for treating cancer
US20050095267A1 (en) * 2002-12-04 2005-05-05 Todd Campbell Nanoparticle-based controlled release polymer coatings for medical implants
WO2004052401A2 (en) 2002-12-09 2004-06-24 American Bioscience, Inc. Compositions and methods of delivery of pharmacological agents
EP2359859A1 (en) 2002-12-09 2011-08-24 Abraxis BioScience, LLC Compositions comprising albumin nanoparticles and methods of delivery of pharmacological agents
CA2513399A1 (en) * 2003-01-10 2004-07-29 Threshold Pharmaceuticals, Inc. Treatment of cancer with 2-deoxyglucose
MXPA05010388A (en) 2003-03-28 2006-03-08 Kosan Biosciences Inc Devices, methods, and compositions to prevent restenosis.
US20060165744A1 (en) 2003-05-22 2006-07-27 Neopharm, Inc Combination liposomal formulations
US20050026893A1 (en) 2003-05-30 2005-02-03 Kosan Biosciences, Inc. Method for treating diseases using HSP90-inhibiting agents in combination with immunosuppressants
CN1829741A (en) 2003-05-30 2006-09-06 健泰科生物技术公司 Treatment with anti-VEGF antibodies
AR046510A1 (en) * 2003-07-25 2005-12-14 Regeneron Pharma COMPOSITION OF A VEGF ANTAGONIST AND AN ANTI-PROLIFERATIVE AGENT
WO2005027972A2 (en) 2003-09-23 2005-03-31 Novartis Ag Combination of a vegf receptor inhibitor with a chemotherapeutic agent
EP2286794B8 (en) 2003-10-15 2016-06-01 SynCore Biotechnology CO., LTD Use of cationic liposomes comprising paclitaxel
TW200522978A (en) 2003-10-29 2005-07-16 Sonus Pharma Inc Tocopherol-modified therapeutic drug compounds
US20080045559A1 (en) 2003-10-29 2008-02-21 Sonus Pharmaceuticals, Inc. Tocopherol-modified therapeutic drug compounds
US20050203174A1 (en) 2004-03-09 2005-09-15 Kosan Biosciences, Inc. Combination therapies using leptomycin B
US7129368B2 (en) 2004-03-15 2006-10-31 Sonus Pharmaceuticals, Inc. Platinum carboxylate anticancer compounds
WO2006001911A2 (en) * 2004-05-06 2006-01-05 Genentech, Inc. Crystal structure of the hepatocyte growth factor beta chain and methods of use
EP2784082B1 (en) 2004-05-14 2017-12-27 Abraxis BioScience, LLC Disease site-delivering pharmaceutical compositions comprising paclitaxel.
US8420603B2 (en) * 2004-05-14 2013-04-16 Abraxis Bioscience, Llc SPARC and methods of use thereof
CN114053429A (en) 2004-06-01 2022-02-18 健泰科生物技术公司 Antibody-drug conjugates and methods
CA2567836A1 (en) * 2004-06-03 2005-12-15 F. Hoffmann-La Roche Ag Treatment with gemcitabine and an egfr-inhibitor
BRPI0510915A (en) 2004-06-04 2007-11-13 Genentech Inc Method for treating multiple sclerosis and manufactured article
SV2006002143A (en) 2004-06-16 2006-01-26 Genentech Inc USE OF AN ANTIBODY FOR THE TREATMENT OF CANCER RESISTANT TO PLATINUM
CN101160321A (en) * 2005-02-18 2008-04-09 阿布拉西斯生物科学公司 Q3 sparc deletion mutant and uses thereof
EP2301531B1 (en) 2005-02-18 2018-06-06 Abraxis BioScience, LLC Combinations and modes of administration of therapeutic agents and combination therapy
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US20070166388A1 (en) * 2005-02-18 2007-07-19 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
CA2601067A1 (en) 2005-05-04 2006-11-09 Medigene Ag Method of administering a cationic liposomal preparation comprising paclitaxel
US20080221135A1 (en) 2005-05-13 2008-09-11 Bristol-Myers Squibb Company Combination therapy
ES2720603T3 (en) 2005-08-31 2019-07-23 Abraxis Bioscience Llc Compositions comprising low water soluble pharmaceutical agents and antimicrobial agents
WO2007027941A2 (en) * 2005-08-31 2007-03-08 Abraxis Bioscience, Llc. Compositions and methods for preparation of poorly water soluble drugs with increased stability
WO2007059116A2 (en) 2005-11-14 2007-05-24 Abraxis Bioscience, Inc. Geldanamycin derivatives and pharmaceutical compositions thereof
CA2922029C (en) 2006-03-22 2017-11-28 Medigene Ag A combination of a cationic liposomal preparation comprising an antimitotic agent and a non-liposomal preparation comprising an antimitotic agent
US9037257B2 (en) * 2006-04-07 2015-05-19 Medtronic, Inc. Resonance tuning module for implantable devices and leads
SG136814A1 (en) 2006-04-12 2007-11-29 Tencube Pte Ltd System for tracking mobile equipment and associated mobile subscribersæ identity
US20080280987A1 (en) * 2006-08-31 2008-11-13 Desai Neil P Methods of inhibiting angiogenesis and treating angiogenesis-associated diseases
US20100112077A1 (en) 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer
PL2117520T3 (en) 2006-12-14 2019-03-29 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
KR102047634B1 (en) 2007-03-07 2019-11-21 아브락시스 바이오사이언스, 엘엘씨 Nanoparticle comprising rapamycin and albumin as anticancer agent
WO2008137148A2 (en) 2007-05-03 2008-11-13 Abraxis Bioscience, Llc Methods and compositions for treating pulmonary hypertension
WO2008150532A1 (en) 2007-06-01 2008-12-11 Abraxis Bioscience, Llc Methods and compositions for treating recurrent cancer
US20090018078A1 (en) 2007-07-09 2009-01-15 Vinod Labhasetwar Apoptosis-Modulating Protein Therapy for Proliferative Disorders and Nanoparticles Containing the Same
WO2010118365A1 (en) 2009-04-10 2010-10-14 Abraxis Bioscience, Llc Nanoparticle formulations and uses therof
AU2009234127B2 (en) * 2008-04-10 2015-04-30 Abraxis Bioscience, Llc Compositions of hydrophobic taxane derivatives and uses thereof
AU2009324464B2 (en) 2008-12-11 2016-05-05 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
MX2011009452A (en) 2009-03-13 2011-11-29 Abraxis Bioscience Llc Combination therapy with thiocolchicine derivatives.
MY175260A (en) 2009-04-15 2020-06-17 Abraxis Bioscience Llc Prion free nanoparticle compositions and methods of making thereof
MY162514A (en) 2009-08-25 2017-06-15 Abraxis Bioscience Llc Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors
EP2898884B1 (en) 2010-03-26 2018-05-09 Abraxis BioScience, LLC Methods of treatment of hepatocellular carcinoma
US10660965B2 (en) 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
SI2552415T1 (en) 2010-03-29 2017-03-31 Abraxis Bioscience, Llc Methods of treating cancer
WO2011137114A1 (en) 2010-04-26 2011-11-03 Abraxis Bioscience, Llc Sparc binding antibodies and uses thereof
MX343671B (en) 2010-06-02 2016-11-16 Abraxis Bioscience Llc * Methods of treating bladder cancer.
AU2011261685B2 (en) 2010-06-04 2016-02-11 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
EP3056201A1 (en) 2010-06-07 2016-08-17 Abraxis BioScience, LLC Combination therapy methods for treating proliferative diseases
KR102108959B1 (en) 2011-04-28 2020-05-11 아브락시스 바이오사이언스, 엘엘씨 Intravascular delivery of nanoparticle compositions and uses thereof
US9585960B2 (en) 2011-12-14 2017-03-07 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
US9149455B2 (en) 2012-11-09 2015-10-06 Abraxis Bioscience, Llc Methods of treating melanoma
US9511046B2 (en) 2013-01-11 2016-12-06 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US20140199404A1 (en) 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on level of a nucleoside transporter
US20140199405A1 (en) 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on mutation status of k-ras

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US9101543B2 (en) 2005-02-18 2015-08-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9561288B2 (en) 2005-02-18 2017-02-07 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy

Also Published As

Publication number Publication date
IL185288A0 (en) 2008-02-09
JP2018090643A (en) 2018-06-14
US20130209518A1 (en) 2013-08-15
EP2301531A2 (en) 2011-03-30
HK1119071A1 (en) 2009-02-27
RU2012106620A (en) 2013-08-27
IL220547A0 (en) 2012-08-30
AU2006213999B2 (en) 2012-04-05
CN101160123B (en) 2013-07-17
US20140017315A1 (en) 2014-01-16
HK1109589A1 (en) 2008-06-13
KR20170090534A (en) 2017-08-07
PT1853250E (en) 2012-02-03
JP2015155454A (en) 2015-08-27
EP3248600A1 (en) 2017-11-29
CN103285395A (en) 2013-09-11
KR20180120798A (en) 2018-11-06
CA2902071A1 (en) 2006-08-24
US8034375B2 (en) 2011-10-11
KR101914254B1 (en) 2018-11-02
US20180169017A1 (en) 2018-06-21
US9101543B2 (en) 2015-08-11
US20120177743A1 (en) 2012-07-12
ATE531365T1 (en) 2011-11-15
LU92484I2 (en) 2015-10-27
BRPI0607809A2 (en) 2009-06-13
LT3248600T (en) 2020-07-27
HUE049789T2 (en) 2020-10-28
NZ595313A (en) 2013-03-28
JP2012193205A (en) 2012-10-11
IL185288A (en) 2017-06-29
US7780984B2 (en) 2010-08-24
US20070116774A1 (en) 2007-05-24
SI1853250T1 (en) 2012-01-31
KR20070114753A (en) 2007-12-04
US20060263434A1 (en) 2006-11-23
US20130195922A1 (en) 2013-08-01
US20090098210A1 (en) 2009-04-16
EP1853250B1 (en) 2011-11-02
JP2013064014A (en) 2013-04-11
DK1853250T3 (en) 2012-01-23
EP2301531A3 (en) 2011-07-27
PL1853250T3 (en) 2012-03-30
US20150079177A1 (en) 2015-03-19
JP2014224157A (en) 2014-12-04
NZ560879A (en) 2010-04-30
WO2006089290A1 (en) 2006-08-24
SG166775A1 (en) 2010-12-29
EP3698783A1 (en) 2020-08-26
KR20160060788A (en) 2016-05-30
ES2678448T3 (en) 2018-08-10
CA3054535A1 (en) 2006-08-24
KR101764375B1 (en) 2017-08-03
JP6092824B2 (en) 2017-03-08
JP2017025090A (en) 2017-02-02
KR20190062506A (en) 2019-06-05
RU2007134571A (en) 2009-03-27
HUE038768T2 (en) 2018-11-28
JP2020012005A (en) 2020-01-23
US20140023717A1 (en) 2014-01-23
KR20200038337A (en) 2020-04-10
US8257733B2 (en) 2012-09-04
SI2301531T1 (en) 2018-11-30
US20080063724A1 (en) 2008-03-13
ES2375828T3 (en) 2012-03-06
CY1112907T1 (en) 2016-04-13
ZA200707491B (en) 2008-12-31
SI3248600T1 (en) 2020-09-30
PT2301531T (en) 2018-07-30
DK3248600T3 (en) 2020-07-06
EP3248600B1 (en) 2020-04-08
JP5769358B2 (en) 2015-08-26
HK1250006A1 (en) 2018-11-23
KR101976003B1 (en) 2019-05-09
EP3248600B8 (en) 2020-06-24
JP6513467B2 (en) 2019-05-15
CN105288630A (en) 2016-02-03
DK2301531T3 (en) 2018-07-30
KR20140016402A (en) 2014-02-07
KR101625347B1 (en) 2016-05-27
RU2632103C2 (en) 2017-10-02
CA2902071C (en) 2019-10-08
EP1853250A1 (en) 2007-11-14
US8268348B2 (en) 2012-09-18
CA2598239C (en) 2019-10-29
KR20150043560A (en) 2015-04-22
CN101160123A (en) 2008-04-09
AU2006213999A1 (en) 2006-08-24
MX2007009960A (en) 2008-03-11
PT3248600T (en) 2020-07-16
EP2301531B1 (en) 2018-06-06
JP6685875B2 (en) 2020-04-22
IL252715A0 (en) 2017-08-31
RU2452482C2 (en) 2012-06-10
PL3248600T3 (en) 2020-09-07
ES2802541T3 (en) 2021-01-20
MX339142B (en) 2016-05-13
LT2301531T (en) 2018-09-25
JP2008530248A (en) 2008-08-07
US7758891B2 (en) 2010-07-20
CN103169729A (en) 2013-06-26
PL2301531T3 (en) 2018-11-30
IL273765A (en) 2020-05-31
CY1123110T1 (en) 2021-10-29
CY1120445T1 (en) 2019-07-10

Similar Documents

Publication Publication Date Title
CA2902071C (en) Combination chemotherapy regimens using albumin/paclitaxel nanoparticles and nucleoside analogs
US9561288B2 (en) Combinations and modes of administration of therapeutic agents and combination therapy
US20070166388A1 (en) Combinations and modes of administration of therapeutic agents and combination therapy
US20100112077A1 (en) Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer
AU2016204834B2 (en) Combinations and modes of administration of therapeutic agents and combination therapy
AU2012201568B2 (en) Combinations and modes of administration of therapeutic agents and combination therapy

Legal Events

Date Code Title Description
EEER Examination request