US20160250351A1 - Treating Lymphomas - Google Patents

Treating Lymphomas Download PDF

Info

Publication number
US20160250351A1
US20160250351A1 US15/030,567 US201515030567A US2016250351A1 US 20160250351 A1 US20160250351 A1 US 20160250351A1 US 201515030567 A US201515030567 A US 201515030567A US 2016250351 A1 US2016250351 A1 US 2016250351A1
Authority
US
United States
Prior art keywords
abraxane
complexes
dose
albumin
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
US15/030,567
Inventor
Svetomir N. Markovic
Wendy K. Nevala
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.)
Mayo Foundation for Medical Education and Research
Original Assignee
Mayo Foundation for Medical Education and Research
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
Application filed by Mayo Foundation for Medical Education and Research filed Critical Mayo Foundation for Medical Education and Research
Priority to US15/030,567 priority Critical patent/US20160250351A1/en
Assigned to MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH reassignment MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MARKOVIC, SVETOMIR N, NEVALA, WENDY K
Publication of US20160250351A1 publication Critical patent/US20160250351A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • A61K47/48884
    • 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
    • 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
    • A61K31/555Heterocyclic compounds containing heavy metals, e.g. hemin, hematin, melarsoprol
    • 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
    • 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
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • A61K47/48284
    • A61K47/48384
    • A61K47/48561
    • 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
    • A61K47/51Medicinal 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
    • A61K47/68Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • 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
    • A61K47/68Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • A61K47/6835Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6849Medicinal 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 an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2887Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against CD20
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/54Medicinal preparations containing antigens or antibodies characterised by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/20Immunoglobulins specific features characterized by taxonomic origin
    • C07K2317/24Immunoglobulins specific features characterized by taxonomic origin containing regions, domains or residues from different species, e.g. chimeric, humanized or veneered

Definitions

  • lymphomas e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas.
  • lymphomas e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas.
  • this document relates to methods and materials involved in using complexes containing albumin-containing nanoparticies e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) to treat lymphomas.
  • albumin-containing nanoparticies e.g., ABRAXANE® nanoparticles
  • antibodies e
  • Lymphomas are blood cancers that occur when B or T lymphocytes become cancerous. They can develop in the lymph nodes, spleen, bone marrow, blood, or other organs and eventually form a tumor.
  • lymphomas e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas.
  • this document provides methods and materials for using complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) to treat lymphomas.
  • albumin-containing nanoparticles e.g., ABRAXANE® nanoparticles
  • antibodies e.g., anti-CD20 polypeptide antibodies such as Rituximab
  • ABRAXANE® is available from Celgene Corp. and is a nanopartick formulation that combines paclitaxel with human albumin.
  • Anti-CD20 polypeptide antibodies such as Rituximab are available from Genentech Inc., Roche, and Aryogen Biopharma under trade names such as RituxanTM, MabTheraTM, and ZytuxTM.
  • Rituximab is a chimeric monoclonal antibody against CD20 polypeptides presents on surface of lymphocyte cells (see, e.g., U.S. Pat. No. 5,736,137).
  • macromolecular complexes the characteristics of which (e.g., size, antibody content, or chemotherapeutic drug content) can be customized depending on need.
  • macromolecular complexes can retain antibody mediated target binding specificity, can retain or exhibit enhanced chemotherapeutic tumor cell cytotoxicity, and can exhibit no additional toxicity beyond that of ABRAXANE® nanoparticles alone.
  • contacting ABRAXANE® with an anti-CD20 polypeptide antibody e.g., Rituximab
  • a human e.g., a human lymphoma cancer patient
  • a complex that, when administered as a complex, has an increased ability to treat lymphoma as compared to a treatment regimen that includes administering ABRAXANE® and the anti-CD20 polypeptide antibody separately in a manner that does not form ABRAXANE®/anti-CD20 polypeptide antibody complexes.
  • the methods and materials provided herein can be used to increase the progression-free survival rate in lymphoma patients. Increasing progression-free survival can allow lymphoma cancer patients to live longer.
  • one aspect of this document features a method for treating a mammal having lymphoma.
  • the method comprises, or consisting essentially of administering to the mammal a composition comprising nanoparticles containing albumin and placitaxel complexed with an anti-CD20 polypeptide antibody under conditions wherein the length of progression-free survival is increased.
  • the mammal can be a human.
  • the lymphoma can be a Hodgkin lymphomas.
  • the composition can comprise Rituximab complexed with the nanoparticles.
  • the composition can comprise an alkylating agent complexed with the nanoparticles.
  • the alkylating agent can be a platinum compound.
  • the platinum compound can be carboplatin.
  • the anti-CD20 polypeptide antibody can be a humanized antibody.
  • the anti-CD20 polypeptide antibody can be a chimeric antibody.
  • the composition can be administered by injection.
  • the progression-free survival can be increased by 15 percent.
  • the progression-free survival can be increased by 25 percent.
  • the progression-free survival can be increased by 50 percent.
  • the progression-free survival can be increased by 75 percent.
  • the progression-free survival can be increased by 100 percent.
  • the composition can be administered under conditions wherein the time to progression is increased.
  • this document features a method for treating a mammal having lymphoma.
  • the method comprises, or consists essentially of, administering, to the mammal, a composition comprising albumin-containing nanoparticle/antibody complexes, wherein the average diameter of the complexes is between 0.1 and 0.9 ⁇ m, and wherein the antibody is an anti-CD20 antibody.
  • the mammal can be a human
  • the lymphoma can be a mature B-cell neoplasm.
  • the lymphoma can be a mature T neoplasm.
  • the lymphoma can be a Hodgkin lymphoma.
  • the albumin-containing nanopartictelantibody complexes can be ABRAXANE®/Rituximab complexes.
  • the composition or the albumin-containing nanopartictelantibody complexes can comprise an atkylating agent.
  • the aikyiating agent can be a platinum compound.
  • the platinum compound can be carboplatin.
  • the composition can comprise an anti-inflammatory agent.
  • the anti-CD20 polypeptide antibodies can be humanized antibodies.
  • the anti-CD20 polypeptide antibodies can be chimeric antibodies.
  • the composition can be administered by injection.
  • the administration of the composition can be effective to increase progression-free survival by 25 percent.
  • the administration of the composition can be effective to increase progression-free survival by 50 percent.
  • the administration of the composition can be effective to increase progression-free survival by 75 percent.
  • the administration of the composition can be effective to increase progression-free survival by 100 percent.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 150 days.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 165 days.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 170 days.
  • the average diameter of the complexes can be from 0,1 ⁇ m to 0.3 ⁇ m.
  • the average diameter of the complexes can be from 0.15 ⁇ m to 0.3 ⁇ m.
  • the average diameter of the complexes can be from 0.2 ⁇ m to 0.5 ⁇ m.
  • the average diameter of the complexes can be from 0.3 ⁇ m to 0.5 ⁇ m.
  • the average diameter of the complexes can be from 0.2 ⁇ m to 0.8 ⁇ m.
  • the average diameter of the complexes can be from 0.2 ⁇ m to 0.7 ⁇ m.
  • this document features a method for treating a mammal having lymphoma.
  • the method comprises, or consists essentially of, administering, to the mammal, a composition comprising albumin-containing nanoparticle/antibody complexes, wherein the average diameter of at least 5 percent of the complexes of the composition is between 0.1 and 0.9 ⁇ m, and wherein the antibodies are anti-CD20 antibodies.
  • the mammal can be a human.
  • the lymphoma can be a mature B-cell neoplasm.
  • the lymphoma can be a mature T neoplasm.
  • the lymphoma can be a Hodgkin lymphoma.
  • albumin-containing nanoparticte/antibody complexes can be ABRAXANE®/Rituximab complexes.
  • the composition or the albumin-containing nanoparticte/antibody complexes can comprise an alkylating agent.
  • the alkylating agent can be a platinum compound.
  • the platinum compound can be carboplatin.
  • the composition can comprise an anti-inflammatory agent.
  • the anti-CD20 polypeptide antibodies can be humanized antibodies.
  • the anti-CD20 polypeptide antibodies can be chimeric antibodies.
  • the composition can be administered by injection.
  • the administration of the composition can be effective to increase progression-free survival by 25 percent.
  • the administration of the composition can be effective to increase progression-free survival by 50 percent.
  • the administration of the composition can be effective to increase progression-free survival by 75 percent.
  • the administration of the composition can be effective to increase progression-free survival by 100 percent.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 150 days.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 165 days.
  • the administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 170 days.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.9 run.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.8 ⁇ m.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.7 ⁇ m.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.6 ⁇ m.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.5 ⁇ m.
  • the average diameter of at least 5 percent of the complexes of the composition can be from 0.2 ⁇ m to 0.4 ⁇ m.
  • the average diameter of at least 10 percent of the complexes of the composition can be between 0.1 and 0.9 ⁇ m.
  • the average diameter of at least 50 percent of the complexes of the composition can be between 0.1 and 0.9 ⁇ m.
  • the average diameter of at least 75 percent of the complexes of the composition can be between 0.1 and 0.9 ⁇ m.
  • the average diameter of at least 90 percent of the complexes of the composition can be between and 0.9 ⁇ m.
  • FIG. 1 is a graph plotting the particle size distribution for ABRAXANE® (ABX) dissolved in Rituxan (RIT) as determined using a Mastersizer 2000E (Malvern Instruments Ltd., Worcestershire, England). ABX (10 mg/mL) was reconstituted in 1 mL of the indicated amount of RIT, and the mixtures were incubated at room temperature for 30 minutes,
  • FIG. 2 is a graph plotting percent change at seven days in tumor size from baseline of lymphoma (Daudi cell line) tumor bearing nude mice treated with PBS, Rituxan (RIT; 12 mg/kg) only, ABRAXANE® (30 mg/kg) only, Rituxan (RIT; 12 mg/kg) followed by ABRAXANE® (30 mg/kg) one day later, or AR160 complexes (0.159 ⁇ m; complexes where ABRAXANE® (10 mg/mL) was premixed with 2 mg/mL Rituxan and incubated for 60 minutes before injection).
  • FIG. 3 is a Kaplan Meier graph plotting survival of lymphoma (Daudi cell line) tumor bearing nude mice treated with PBS, Rituxan (RIT; 12 mg/kg) only, ABRAXANE® (30 mg/kg) only, Rituxan (RIT; 12 mg/kg) followed by ABRAXANE® (30 mg/kg) one day later, or AR160 complexes (0.159 ⁇ m; complexes where ABRAXANE® (10 mg/mL) was premixed with 2 mg/mL Rituxan and incubated for 60 minutes before injection).
  • lymphomas e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas.
  • this document provides methods and materials for using complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD2( )polypeptide antibodies such as Rituximab) to treat lymphomas.
  • albumin-containing nanoparticles e.g., ABRAXANE® nanoparticles
  • antibodies e.g., anti-CD2( )polypeptide antibodies such as Rituximab
  • the methods and materials provided herein can be used to treat any type of lymphoma.
  • the methods and materials provided herein can be used to treat mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, or non-Hodgkin lymphomas.
  • the methods and materials provided herein can be used to treat lymphomas in any type of mammal including, without limitation, mice, rats, dogs, cats, horses, cows, pigs, monkeys, and humans.
  • complexes containing albumin-containing nanoparticles e.g., ABRAXANE® nanoparticles
  • antibodies e.g., anti-CD20 polypeptide antibodies such as Rituximab
  • albumin-containing nanoparticles and antibodies can be designed to have an average diameter that is greater than 1 ⁇ m.
  • appropriate concentrations of albumin-containing nanoparticles and antibodies can be used such that complexes having an average diameter that is greater than 1 ⁇ m are formed.
  • manipulations such as centrifugation can be used to form preparations of albumin-containing nanoparticle/antibody complexes where the average diameter of those complexes is greater than 1 ⁇ m.
  • the preparations of albumin-containing nanoparticle/antibody complexes provided herein can have an average diameter that is between 1 ⁇ m and 5 ⁇ m e.g., between 1.1 ⁇ m and 5 ⁇ m, between 1.5 ⁇ m and 5 ⁇ m, between 2 ⁇ m and 5 ⁇ m, between 2.5 ⁇ m and 5 ⁇ m, between 3 ⁇ m and 5 ⁇ m, between 3.5 ⁇ m and 5 ⁇ m, between 4 ⁇ m and 5 ⁇ m, between 4.5 ⁇ m and 5 ⁇ m, between 1.1 ⁇ m and 4.5 ⁇ m, between 1.1 ⁇ m and 4 ⁇ m, between 1.1 ⁇ m and 3.5 ⁇ m, between 1.1 ⁇ m and 3 ⁇ m, between 1.1 ⁇ m and 2.5 ⁇ m, between 1.1 ⁇ m and 2 ⁇ m, or between 1.1 ⁇ m and 1.5 ⁇ m).
  • Preparations of albumin-containing nanoparticle/antibody complexes provided herein having an average diameter that is between 1 ⁇ m and 5 ⁇ m can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body.
  • the preparations of albumin-containing nanoparticie/antibody complexes provided herein can have an average diameter that is between 5 ⁇ m and 50 ⁇ m (e.g., between 6 ⁇ m and 50 ⁇ m, between 7 ⁇ m and 50 ⁇ m, ween 10 ⁇ m and 50 ⁇ m, between 15 ⁇ m and 50 ⁇ m, between 20 ⁇ m and 50 ⁇ m, between 25 ⁇ m and 50 ⁇ m, between 30 ⁇ m and 50 ⁇ m, between 35 ⁇ m and 50 ⁇ m, between 5 ⁇ m and 45 ⁇ m, between 5 ⁇ m and 40 ⁇ m, between 5 ⁇ m and 35 ⁇ m, between 5 ⁇ m and 30 ⁇ m, between 5 ⁇ m and 25 ⁇ m, between 5 ⁇ m and 20 ⁇ m, between 5 ⁇ m and 15 ⁇ m, or between 10 ⁇ m and 30 ⁇ m).
  • 5 ⁇ m and 50 ⁇ m e.g., between 6 ⁇ m and 50 ⁇ m, between 7 ⁇
  • Preparations of albumin-containing nanoparticie/antibody complexes provided herein having an average diameter that is between 5 ⁇ m and 50 ⁇ m can be administered into a tumor (e.g., intratumorally) or in a region of a tumor located within a mammal's body.
  • a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 1 ⁇ m and 5 ⁇ m (e.g., between 1.1 ⁇ m and 5 ⁇ m, between 1.5 ⁇ m and 5 ⁇ m, between 2 ⁇ m and 5 ⁇ m, between 2.5 ⁇ m and 5 ⁇ m, between 3 ⁇ m and 5 ⁇ m, between 3.5 ⁇ m and 5 ⁇ m, between 4 ⁇ m and 5 ⁇ m, between 4.5 ⁇ m and 5 ⁇ m, between 1.1 ⁇ m and 4.5 ⁇ m, between 1.1 ⁇ m and 4 ⁇ m, between 1.1 ⁇ m and 3.5 ⁇ m, between 1.1 ⁇ m and 3 ⁇ m, between 1.1 ⁇ m and 2.5 ⁇ m, between 1.1 ⁇ m and 2 ⁇ m, or between 1.1 ⁇ m and 1.5
  • Preparation of albumin-containing nanoparticle/antibody complexes provided herein having greater than 60 percent (e,g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 1 ⁇ m and 5 ⁇ m can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body.
  • a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 5 ⁇ m and 50 ⁇ m (e.g., between 6 ⁇ m and 50 ⁇ m, between 7 ⁇ m and 50 ⁇ m, between 10 ⁇ m and 50 ⁇ m, between 15 ⁇ m and 50 ⁇ m, between 20 ⁇ m and 50 ⁇ m, between 25 ⁇ m and 50 ⁇ m, between 30 ⁇ m and 50 ⁇ m, between 35 ⁇ m and 50 ⁇ m, between 5 ⁇ m and 45 ⁇ m, between 5 ⁇ m and 40 ⁇ m, between 5 ⁇ m and 35 ⁇ m, between 5 ⁇ m and 30 ⁇ m, between 5 ⁇ m and 25 ⁇ m, between 5 ⁇ m and 20 ⁇ m, between 5 ⁇ m and 15 ⁇ m, or between 10 ⁇ m and 30 ⁇ m,
  • Preparation of albumin-containing nanoparticle/antibody complexes provided herein having greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 5 ⁇ m and 50 ⁇ m can be administered into a tumor (e.g., intratumorally) or in a region of a tumor located within a mammal's body.
  • complexes containing albumin-containing nanoparticles e.g., ABRAXANE® nanoparticles
  • antibodies e.g., anti-CD20 polypeptide antibodies such as Rituximab
  • albumin-containing nanoparticles and antibodies can be designed to have an average diameter that is less than 1 ⁇ m.
  • appropriate concentrations of albumin-containing nanoparticles and antibodies e.g., Rituximab
  • complexes having an average diameter that is less than 1 ⁇ m are formed.
  • the preparations of albumin-containing nanoparticle/antibody complexes provided herein can have an average diameter that is between 0.1 ⁇ m and 1 ⁇ m (e.g., between 0.1 ⁇ m and 0.95 ⁇ m, between 0.1 ⁇ m and 0.9 ⁇ m, between 0.1 ⁇ m and 0.8 ⁇ m, between 0.1 ⁇ m and 0.7 ⁇ m, between 0.1 ⁇ m and 0.6 ⁇ m, between 0.1 ⁇ m and 0.5 ⁇ m, between 0.1 ⁇ m and 0.4 ⁇ m, between 0.1 ⁇ m and 0.3 ⁇ m, between 0.1 ⁇ m and 0.2 ⁇ m, between 0.2 ⁇ m and 1 p,m, between 0.3 ⁇ m and 1 ⁇ m, between 0.4 ⁇ m and 1 ⁇ m, between 0.5 ⁇ m and 1 ⁇ m, between 0.2 ⁇ m and 0.6 ⁇ m, between 0.3 ⁇ m and 0.6 ⁇ m, between 0.2 ⁇ m and 0.5 ⁇ m, or between
  • Preparations of albumin-containing nanoparticle/antibody complexes provided herein having an average diameter that is between 0.1 ⁇ m and 0.9 ⁇ m can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body.
  • a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 0.1 ⁇ m and 0.9 ⁇ m (e.g., between 0.1 ⁇ m and 0.95 ⁇ m, between 0.1 ⁇ m and 0.9 ⁇ m, between 0.1 ⁇ m and 0.8 ⁇ m, between 0.1 ⁇ m and 0.7 ⁇ m, between 0.1 ⁇ m and 0.6 ⁇ m, between 0.1 ⁇ m and 0.5 ⁇ m, between 0.1 ⁇ m and 0.4 ⁇ m, between 0.1 ⁇ m and 0.3 ⁇ m, between 0.1 ⁇ m and 0.2 ⁇ m, between 0.2 ⁇ m and 1 ⁇ m, between 0.3 ⁇ m and 1 ⁇ m, between 0.4 ⁇ m and 1 ⁇ m, between 0.5 ⁇ m and 1 ⁇ m, between 0.2 ⁇ m and
  • Preparation of albumin-containing nanoparticte/antibody complexes provided herein having greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 0.1 ⁇ m and 0.9 ⁇ m can be administered systemically (e.g., intravenously) to treat cancers located within a mammal's body.
  • albumin-containing nanoparticles such as ABRAXANE® can be contacted with an antibody such as an anti-CD20 polypeptide antibody (e.g., Rituximab) prior to administration to a human form an albumin-containing nanoparticle/antibody complex (e.g., an ABRAXANE®/anti-CD20 polypeptide antibody complex).
  • an antibody such as an anti-CD20 polypeptide antibody (e.g., Rituximab)
  • an albumin-containing nanoparticle/antibody complex e.g., an ABRAXANE®/anti-CD20 polypeptide antibody complex
  • Any appropriate albumin-containing nanoparticie preparation and any appropriate antibody can be used as described herein.
  • ABRAXANE® nanoparticles can be used as described herein.
  • antibodies that can be used to form albumin-containing nanoparticle/antibody complexes as described herein include, without limitation, Rituximab (e.g., RituxanTM, MabTheraTM, or ZytuxTM).
  • Rituximab e.g., RituxanTM, MabTheraTM, or ZytuxTM
  • an appropriate dose of ABRAXANE® and an appropriate dose of Rituximab can be mixed together in the same container. This mixture can be incubated at an appropriate temperature (e.g., room temperature, between 15° C. and 30° C., between 15° C and 25° C., between 20° C. and 30° C., or between 20° C.
  • ABRAXANE® can be contacted with an anti-CD20 polypeptide antibody by injecting both ABRAXANE® and the anti-CD20 polypeptide antibody either individually or as a pre-mixed combination into an IV bag containing an IV bag solution.
  • the contents of the IV bag including ABRAXANE®/anti-CD20 polypeptide antibody complexes can be introduced into the patient to be treated.
  • albumin-containing nanoparticles such as ABRAXANE® can be contacted with an antibody such as an anti-CD20 polypeptide antibody (e.g., Rituximab) to form albumin-containing nanoparticle/antibody complexes (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) that are stored prior to being administered to a cancer patient (e.g., a lymphoma patient).
  • a composition containing albumin-containing nanoparticle/antibody complexes can be formed as described herein and stored for a period of time (e.g., days or weeks) prior to being administered to a cancer patient.
  • albumin-containing nanoparticles such as ABRAXANE® and an antibody such as an anti-CD20 polypeptide antibody.
  • ABRAXANE® can be obtained from Celgene Corp. or as described elsewhere (U.S. Pat. No. 6,537,579).
  • Rituximab can be obtained from Genentech Corp. or Roche Corp. or as described elsewhere (U.S. Pat. No. 5,736,137).
  • an albumin-containing nanoparticle such as ABRAXANE® and an antibody such as anti-CD20 polypeptide antibody can include one or more other agents such as an alkylating agent (e.g., a platinum compound).
  • an alkylating agent e.g., a platinum compound
  • platinum compounds that can be used as an alkylating agent include, without carboplatin (PARAPLATIN®), cisplatin (PLATINOL®), oxaliplatin (ELOXATIN®), and BBR3464.
  • agents that can be included within an albumin-containing nanoparticle/antibody complex include, without limitation, adriamycin, cyclophosphamide, vincristine, prednisone, dexamethasone, cytarabine, methotrexate, thiotepa, ifosfamide, chlorambucil, dacarbazine, bleomycin, ibrutinib, campath-B, gemcitabine, revlimid, sirolimus, temsirolimus, bexxar, brentuximab, bendamustine, and etoposide.
  • an albumin-containing nanoparticle/antibody complex provided herein can include brentuximab, cyclophosphamide, adriamycin, or vincristine as part of the complex.
  • albumin-containing nanoparticle/antibody complex provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes)
  • a composition containing albumin-containing nanoparticle/antibody complexes such as ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered via injection(e.g., subcutaneous injection, intramuscular injection, intravenous injection, or intrathecal injection).
  • the mammal Before administering a composition containing an albumin-containing nanoparticie/antibody complex provided herein (e.g.4BRAXANE®/anti-CD20 polypeptide antibody complexes) to a mammal, the mammal can be assessed to determine whether or not the mammal has lymphoma. Any appropriate method can be used to determine whether or not a mammal has lymphoma. For example, a mammal (e.g., human) can be identified as having lymphoma using standard diagnostic techniques. In some cases, a tissue biopsy (e.g., lymph node tissue sample) can be collected and analyzed to determine whether or not a mammal has lymphoma.
  • a tissue biopsy e.g., lymph node tissue sample
  • the mammal can be administered a composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes).
  • a composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered prior to or in lieu of surgical resection of a tumor.
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered following resection of a tumor.
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein can be administered to a mammal in any appropriate amount, at any appropriate frequency, and for any appropriate duration effective to achieve a desired outcome (e.g., to increase progression-free survival).
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma to reduce the progression rate of the lymphoma by 5, 10, 25, 50, 75, 100, or more percent.
  • the progression rate can be reduced such that no additional cancer progression is detected.
  • Any appropriate method can be used to determine whether or not the progression rate of lymphoma is reduced.
  • the progression rate of lymphoma can be assessed by imaging tissue at different time points and determining the amount of cancer cells present. The amounts of cancer cells determined within tissue at different times can be compared to determine the progression rate. After treatment as described herein, the progression rate can be determined again over another time interval.
  • the stage of cancer (e,g., lymphoma) after treatment can be determined and compared to the stage before treatment to determine whether or not the progression rate was reduced.
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma under conditions where progression-free survival is increased (e.g., by 5, 10, 25, 50, 75, 100, or more percent) as compared to the median progression-free survival of corresponding mammals having untreated lymphoma or the median progression-free survival of corresponding mammals having lymphoma treated with ABRAXANE® and an antibody (e.g., an anti-CD20 polypeptide antibody) without forming ABRAXANE®/antibody complexes (e.g., without forming ABRAXNE/anti-CD20 polypeptide antibody complexes).
  • progression-free survival is increased (e.g., by 5, 10, 25, 50, 75, 100, or more percent) as compared to the median progression-free survival of corresponding mammals having untreated lymphoma or the median progression-free survival of corresponding mammals having
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma to increase progression-free survival by 5, 10, 25, 50, 75, 100, or more percent as compared to the median progression-free survival of corresponding mammals having lymphoma and having received ABRAXANE® or an antibody (e.g., an anti-CD20 polypeptide antibody) alone, Progression-free survival can be measured over any length of time (e.g., one month, two months, three months, four months, five months, six months, or longer),
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein can be administered to a mammal having lymphoma under conditions where the 8-week progression-free survival rate for a population of mammals is 65% or greater (e.g., 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80% or greater) than that observed in a population of comparable mammals not receiving a composition containing albumin-containing nanoparticie/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes).
  • composition containing albumin-containing nanoparticle/antibody complexes provided herein(e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma under conditions where the median time to progression for a population of mammals is at least 150 days (e.g., at least 155, 160, 163, 165, or 170 days).
  • An effective amount of a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be any amount that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal.
  • an effective amount of ABRAXANE® can be from about 50 mg/m 2 to about 150 mg/m 2 (e.g., about 80 mg/m 2 )
  • an effective amount of an anti-CD20 polypeptide antibody such as Rituximab can be from about 5 mg/kg to about 20 mg/kg (e.g., about 10 mg/kg or 375 mg/m 2 ).
  • the amount of ABRAXANE® or anti-CD20 polypeptide antibody can be increased by, for example, two fold. After receiving this higher concentration, the mammal can be monitored for both responsiveness to the treatment and toxicity symptoms, and adjustments made accordingly.
  • the effective amount can remain constant or can be adjusted as a sliding scale or variable dose depending on the mammal's response to treatment. Various factors can influence the actual effective amount used for a particular application. For example, the frequency of administration, duration of treatment, use of multiple treatment agents, route of administration, and severity of the lymphoma may require an increase or decrease in the actual effective amount administered.
  • the frequency of administration can be any frequency that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal.
  • the frequency of administration can be from about once a month to about three times a month, or from about twice a month to about six times a month, or from about once every two months to about three times every two months.
  • the frequency of administration can remain constant or can be variable during the duration of treatment.
  • a course of treatment with a composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can include rest periods.
  • composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered over a two week period followed by a two week rest period, and such a regimen can be repeated multiple times.
  • the effective amount various factors can influence the actual frequency of administration used for a particular application. For example, the effective amount, duration of treatment, use of multiple treatment agents, route of administration, and severity of the lymphoma may require an increase or decrease in administration frequency.
  • An effective duration for administering a composition provided herein can be any duration that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal.
  • the effective duration can vary from several days to several weeks, months, or years.
  • the effective duration for the treatment of lymphoma can range in duration from several weeks to several months. :In some cases, an effective duration can be for as long as an individual mammal is alive. Multiple factors can influence the actual effective duration used for a particular treatment. For example, an effective duration can vary with the frequency of administration, effective amount, use of multiple treatment agents, route of administration, and severity of the lymphoma.
  • a composition containing albumin-containing nanoparticle/antibody complexes provided herein can be in any appropriate form.
  • a composition provided herein can be in the form of a solution or powder with or without a diluent to make an injectable suspension.
  • a composition also can contain additional ingredients including, without limitation, pharmaceutically acceptable vehicles.
  • a pharmaceutically acceptable vehicle can be, for example, saline, water, lactic acid, mannitol, or combinations thereof.
  • the mammal After administering a composition provided herein to a mammal, the mammal can be monitored to determine whether or not the lymphoma was treated. For example, a mammal can be assessed after treatment to determine whether or not the progression rate of lymphoma was reduced (e.g., stopped). As described herein, any method can be used to assess progression and survival rates.
  • a formulation of ABRAXANE®/Rituxan complexes described in Example 1 can be administered to a human lymphoma patient as described in the methods set forth in Example 3.
  • nanoparticles containing albumin e.g., nanoparticles with an albumin shell
  • an agent other than placitaxel can be used as described herein in place of or in combination with ABRAXANE®.
  • albumin-containing nanoparticles designed to carry a cancer chemotherapeutic agent can be used to form nanoparticle/anti-CD20 polypeptide antibody complexes that can be used as described herein.
  • An example of such a cancer chemotherapeutic agent includes, without vinblastine.
  • a composition can be formulated to include nanoparticies containing albumin (e,g., nanoparticles with an albumin shell) that are conjugated to an antibody, agent, or combination of antibodies and agents to form complexes for treating lymphoma.
  • albumin e.g., nanoparticles with an albumin shell
  • albumin nanoparticles can be formulated to include adriamycin, cyclophosphamide, vincristine, prednisone, dexamethasone, cytarabine, methotrexate, thiotepa, ifosfamide, chlorambucil, dacarbazine, bleomycin, ibrutinib, campath-B, gemcitabine, revlimid, sirolimus, temsirolimus, bexxar, brentuximab, bendamustine, etoposide, or combinations thereof with or without including rituximab.
  • nanoparticles containing albumin e.g., nanoparticles with an albumin shell
  • a complex described herein e.g., ABRAXANE®/rituximab complexes
  • ABRAXANE®/rituximab complexes can be formulated to include one or more anti-chronic inflammation treatment agents designed to reduce the global state of immune dysfunction and/or chronic inflammation present within a cancer patient.
  • steroidal anti-inflammatory agents e.g., prednisone
  • non-steroidal anti-inflammatory agents e.g., naproxen
  • lympho-depleting cytotoxic agents e.g., cyclophosphamide
  • immune cell and/or cytokine targeting antibodies e.g., infliximab
  • a combination thereof can be incorporated into nanoparticles containing albumin or ABRAXANE®/rituximab complexes.
  • anti-IL-4 agents e.g., anti-IL-4 antibodies
  • anti-IL-13 agents e.g., soluble IL-13 receptor
  • combinations thereof can be incorporated into nanoparticles containing albumin or ABRAXANE®/rituximab complexes.
  • cytokine profiles e.g., IL-4, IL-13, IL-4, IL-13, IL-5, IL-10, IL-2, and interferon gamma
  • cytokine profiles e.g., IL-4, IL-13, IL-4, IL-13, IL-5, IL-10, IL-2, and interferon gamma
  • ABRAXANE® was incubated with various increasing concentrations of Rituxan® (rituximab) to form ABRAXANE®/Rituxan® complexes of increasing diameter.
  • Ten milligrams of ABRAXANE® was reconstituted in 1 mL of Rituxan® at 0, 2, 4, 6, 8, and 10 mg/mL, and the mixture was incubated at room temperature for 30 minutes. After incubation, the distributions of particle sizes were determined with the Mastersizer 2000. The median particle size ranged from 0.147 ⁇ m to 8.286 ⁇ m for 0 and 10 mg/mL Rituxan®, respectively ( FIG. 1 ).
  • mice Female athymic nude mice were injected with 1 ⁇ 10 6 lymphoma cells (Daudi cell line). Tumors were allowed to grow, and treatments were administered when tumors were between 800 and 1000 mm 3 . Mice were treated with (a) a single dose of 100 ⁇ L PBS on day 0, (b) a single dose of Rituxan (12 mg/kg) on day 0, (c) a single dose of ABRAXANE® (30 mg/kg) on day 0, (d) a single dose of Rituxan (12 mg/kg) on day 0 followed by a single dose of ABRAXANE® (30 mg/kg) on day 1, or (e) a single dose of AR160 (equivalent to 30 mg/kg of ABRAXANE®) on day 0. Tumor volumes (mm 3 ) were monitored, and the percent change in tumor volume seven days after treatment was determined.
  • the AR160 complexes were produced as follows. 10 mg ABRAXANE® was reconstituted in 2 mg of Rituxan in 500 ⁇ L 0.9% saline and incubated for 1 hour at room temperature. After incubation, AR160 was brought to 1 mL with 0.9% saline. AR160 was further diluted, and 100 ⁇ L was administered to mice at a dose equivalent to 12 mg/kg Rituxan and 30 mg/kg ABRAXANE®. Average particle size for AR160 was 0.159 ⁇ m.
  • the treatment schedule for ABRAXANE®/Rituxan complexes is repeated each month (every 28 days +/ ⁇ 3 days) or until disease progression, patient refusal, or unacceptable toxicity (Table 1) with the indicated dose escalation scheme (Table 2) and dose limiting toxicities (Table 3).
  • Dose Escalation Scheme Dose Level Dose (ABX) Dose (RIT) ⁇ 2 75 mg/m 2 30 mg/m 2 ⁇ 1 100 mg/m 2 40 mg/m 2 1* 125 mg/m 2 50 mg/m 2 2 150 mg/m 2 60 mg/m 2 3 175 mg/m 2 70 mg/m 2 *Starting dose.
  • ABX Dose Level Dose
  • RIT Dose (RIT) ⁇ 2 75 mg/m 2 30 mg/m 2 ⁇ 1 100 mg/m 2 40 mg/m 2 1* 125 mg/m 2 50 mg/m 2 2 150 mg/m 2 60 mg/m 2 3 175 mg/m 2 70 mg/m 2 *Starting dose.
  • DLT Dose Limiting Toxicities
  • the maximum tolerated dose is defined as the highest dose level among those tested where at most one out of six patients develops a DLT prior to the start of their second cycle of treatment and the next highest dose level is such that two out of a maximum of six patients treated at this dose level developed a DLT prior to the start of their second cycle of treatment.
  • a minimum of two or a maximum of six patients are accrued to a given dose level.
  • dose level 1 (and if accrued to, dose levels ⁇ 1 & ⁇ 2)
  • enrollment is temporarily halted after each patient has been enrolled in order to gather acute adverse event data over the first cycle of their treatment.
  • dose levels 2 & 3 patients are accrued to these dose levels so that at any given time no more than two patients are receiving their first cycle of treatment and acute adverse event data over the first treatment cycle for all other patients treated at the current dose level is known. If, at any time in the enrollment process, two patients treated at the current dose level develop a DLT during the first cycle of treatment, enrollment is closed to that dose level.
  • Enrollment is re-opened to the next lower dose level if fewer than six patients have been treated at that dose level. If none of the first three patients treated at a given dose level develops a DLT during the first cycle of treatment, enrollment to the dose level is closed and enrollment is reopen at next higher dose level. If there are no other higher dose levels to be tested, three additional patients are enrolled at the current dose level to confirm MTD. If one of the first three patients treated at a given dose level develops a DLT during the first cycle of treatment, three additional patients are enrolled (sequentially) onto the current dose level. If at any time in the enrollment of these three additional patients, a patient develops a DLT, enrollment is closed to this dose level.
  • Enrollment is re-opened to the next lower dose level if fewer than six patients are treated at that dose level. If none of these three additional patients develops a DLT during the first cycle of treatment, enrollment to this dose level is closed and enrollment is reopened at next higher dose level. If there are no other higher dose levels to be tooted, this is considered the MTD.
  • the patient returns for evaluation and retreatment (at least every 28+/ ⁇ 3 days) according to the schedule. If a patient fails to complete the first cycle of treatment for reasons other than toxicity, an additional patient is enrolled to replace this patient.
  • the planned duration of full-dose anticoagulation is >2 weeks, treatment may be resumed during the period of full-dose anticoagulation IF all of the criteria below are met:
  • the subject must have an in-range INR (usually 2-3) on a stable dose of warfarin, or on stable dose of heparin prior to restarting treatment.
  • the subject must not have pathological conditions that carry high risk of bleeding (e.g. tumor involving major vessels or other conditions)
  • the subject must not have had hemorrhagic events while on study If thromboemboli worsen/recur upon resumption of study therapy, discontinue treatment.
  • G-CSF colony-stimulating factor
  • GM-CSF GM-CSF Routine use of colony-stimulating factors (G-CSF or GM-CSF) is not recommended. Prophylactic use of colony-stimulating factors during the study is not allowed. Therapeutic use in patients with serious neutropenic complications such as tissue infection, sepsis syndrome, fungal infection, etc., may be considered at physician discretion. Recombinant erythropoietin to maintain adequate hemoglobin levels and avoid packed red blood cell transfusions is allowed.
  • Patients do not require premedication prior to administration of ABRAXANE®/Rituxan complexes.
  • treatment with antihistamines, H2 blockers, and corticosteroids is recommended. Patients should be pre-medicated with the typical regimen for paclitaxel regimens for subsequent cycles.
  • premedication may be administered using the premedication regimen the institution typically uses for solvent-based paclitaxel.
  • ABRAXANE®/Rituxan complexes are prepared as a hazardous low risk product.
  • ABRAXANE® is supplied as a white to off-white lyophilized powder containing 100 mg of paclitaxel and approximately 900 mg Albumin Human USP (HA) as a stabilizer in a 50 mL, single-use vial. Each vial of the lyophilized product is reconstituted as set forth below. Unreconstituted ABRAXANE® is stored at controlled room temperature in its carton. Reconstituted ABRAXANE® is used immediately.
  • Rituxan is classified as an anti-CD20 monoclonal antibody.
  • the dose appropriate number of vials of Rituxan are obtained, and each vial is further diluted per the following directions to 4 mg/mL.
  • the dose appropriate number of ABRAXANE® (paclitaxel) 100 mg vials is obtained and each vial is reconstituted per the following directions to a final concentration containing 10 mg/mL nanoparticle albumin-bound (nab) paclitaxel. It is not a requirement to use filter needles in the preparation of, or in-line filters during administration. In addition, filters of pore-size less than 15 micrometers are to be avoided.
  • the vials containing the ABRAXANE® and Rituxan are allowed to sit for 60 minutes.
  • the vial(s) are gently swirled and/or inverted every 10 minutes to continue to mix the complexes.
  • a sterile 60- to 100-mL syringe (appropriate size for the volume being administered) is used to withdraw the calculated dosing volume of ABRAXANE® and Rituxan from each vial.
  • a sufficient quantity of 0.9% Sodium Chloride Injection, USP is added to make the final concentration of ABRAXANE® 5 mg/mL and Rituxan 2 mg/mL.
  • the syringe is gently swirled and/or inverted slowly for 1 minute to mix.
  • the storage and stability is for up to 4 hours at room temperature following final dilution.
  • the IV initial complex dose is infused over 60 minutes via syringe pump.
  • the infusion may be shortened to 30 minutes if the initial infusion is well tolerated.
  • Infusion is monitored closely during the infusion process for signs/symptoms of an infusion reaction.
  • the patient's line is flushed after administration with 20 mL 0.9% Sodium Chloride.
  • Each patient's response to treatment with a ABRAXANE®/Rituxan complex formulation is monitored.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Organic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Cell Biology (AREA)
  • Endocrinology (AREA)
  • Zoology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Nanotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Dermatology (AREA)
  • Medicinal Preparation (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

This document provides methods and materials related to treating lymphomas. For example, methods and materials relating to the use of a composition containing albumin-containing nanoparticle/antibody complexes (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) to treat lymphomas are provided.

Description

    CROSS REFERENCE TO RELATED APPLICATIONS
  • This application claims the benefit of U.S. Provisional Ser. No. 62/012,190 file Jun. 13, 2014. This disclosure of the prior application is considered part of (and is incorporated by reference in) the disclosure of this application.
  • BACKGROUND
  • 1. Technical Field
  • This document relates to methods and materials involved in treating lymphomas (e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas. For example, this document relates to methods and materials involved in using complexes containing albumin-containing nanoparticies e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) to treat lymphomas.
  • 2. Background Information
  • Lymphomas are blood cancers that occur when B or T lymphocytes become cancerous. They can develop in the lymph nodes, spleen, bone marrow, blood, or other organs and eventually form a tumor.
  • SUMMARY
  • This document provides methods and materials involved in treating lymphomas (e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas). For example, this document provides methods and materials for using complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) to treat lymphomas.
  • ABRAXANE® is available from Celgene Corp. and is a nanopartick formulation that combines paclitaxel with human albumin. Anti-CD20 polypeptide antibodies such as Rituximab are available from Genentech Inc., Roche, and Aryogen Biopharma under trade names such as Rituxan™, MabThera™, and Zytux™. Rituximab is a chimeric monoclonal antibody against CD20 polypeptides presents on surface of lymphocyte cells (see, e.g., U.S. Pat. No. 5,736,137).
  • As described herein, in vitro mixing of albumin-containing nanoparticles ABRAXANE® nanoparticies) and antibodies e.g., Rituximab) can result in the formation of macromolecular complexes, the characteristics of which (e.g., size, antibody content, or chemotherapeutic drug content) can be customized depending on need. In some cases, such macromolecular complexes can retain antibody mediated target binding specificity, can retain or exhibit enhanced chemotherapeutic tumor cell cytotoxicity, and can exhibit no additional toxicity beyond that of ABRAXANE® nanoparticles alone. As also described herein, contacting ABRAXANE® with an anti-CD20 polypeptide antibody (e.g., Rituximab) prior to administration to a human (e.g., a human lymphoma cancer patient) can result in a complex that, when administered as a complex, has an increased ability to treat lymphoma as compared to a treatment regimen that includes administering ABRAXANE® and the anti-CD20 polypeptide antibody separately in a manner that does not form ABRAXANE®/anti-CD20 polypeptide antibody complexes.
  • The methods and materials provided herein can be used to increase the progression-free survival rate in lymphoma patients. Increasing progression-free survival can allow lymphoma cancer patients to live longer.
  • In general, one aspect of this document features a method for treating a mammal having lymphoma. The method comprises, or consisting essentially of administering to the mammal a composition comprising nanoparticles containing albumin and placitaxel complexed with an anti-CD20 polypeptide antibody under conditions wherein the length of progression-free survival is increased. The mammal can be a human. The lymphoma can be a Hodgkin lymphomas. The composition can comprise Rituximab complexed with the nanoparticles. The composition can comprise an alkylating agent complexed with the nanoparticles. The alkylating agent can be a platinum compound. The platinum compound can be carboplatin. The anti-CD20 polypeptide antibody can be a humanized antibody. The anti-CD20 polypeptide antibody can be a chimeric antibody. The composition can be administered by injection. The progression-free survival can be increased by 15 percent. The progression-free survival can be increased by 25 percent. The progression-free survival can be increased by 50 percent. The progression-free survival can be increased by 75 percent. The progression-free survival can be increased by 100 percent. The composition can be administered under conditions wherein the time to progression is increased.
  • In another aspect, this document features a method for treating a mammal having lymphoma. The method comprises, or consists essentially of, administering, to the mammal, a composition comprising albumin-containing nanoparticle/antibody complexes, wherein the average diameter of the complexes is between 0.1 and 0.9 μm, and wherein the antibody is an anti-CD20 antibody. The mammal can be a human The lymphoma can be a mature B-cell neoplasm. The lymphoma can be a mature T neoplasm. The lymphoma can be a Hodgkin lymphoma. The albumin-containing nanopartictelantibody complexes can be ABRAXANE®/Rituximab complexes. The composition or the albumin-containing nanopartictelantibody complexes can comprise an atkylating agent. The aikyiating agent can be a platinum compound. The platinum compound can be carboplatin. The composition can comprise an anti-inflammatory agent. The anti-CD20 polypeptide antibodies can be humanized antibodies. The anti-CD20 polypeptide antibodies can be chimeric antibodies. The composition can be administered by injection. The administration of the composition can be effective to increase progression-free survival by 25 percent. The administration of the composition can be effective to increase progression-free survival by 50 percent. The administration of the composition can be effective to increase progression-free survival by 75 percent. The administration of the composition can be effective to increase progression-free survival by 100 percent. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 150 days. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 165 days. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 170 days. The average diameter of the complexes can be from 0,1 μm to 0.3 μm. The average diameter of the complexes can be from 0.15 μm to 0.3 μm. The average diameter of the complexes can be from 0.2 μm to 0.5 μm. The average diameter of the complexes can be from 0.3 μm to 0.5 μm. The average diameter of the complexes can be from 0.2 μm to 0.8 μm. The average diameter of the complexes can be from 0.2 μm to 0.7 μm.
  • In another aspect, this document features a method for treating a mammal having lymphoma. The method comprises, or consists essentially of, administering, to the mammal, a composition comprising albumin-containing nanoparticle/antibody complexes, wherein the average diameter of at least 5 percent of the complexes of the composition is between 0.1 and 0.9 μm, and wherein the antibodies are anti-CD20 antibodies. The mammal can be a human. The lymphoma can be a mature B-cell neoplasm. The lymphoma can be a mature T neoplasm. The lymphoma can be a Hodgkin lymphoma. The method of claim 43, wherein the albumin-containing nanoparticte/antibody complexes can be ABRAXANE®/Rituximab complexes. The composition or the albumin-containing nanoparticte/antibody complexes can comprise an alkylating agent. The alkylating agent can be a platinum compound. The platinum compound can be carboplatin. The composition can comprise an anti-inflammatory agent. The anti-CD20 polypeptide antibodies can be humanized antibodies. The anti-CD20 polypeptide antibodies can be chimeric antibodies. The composition can be administered by injection. The administration of the composition can be effective to increase progression-free survival by 25 percent. The administration of the composition can be effective to increase progression-free survival by 50 percent. The administration of the composition can be effective to increase progression-free survival by 75 percent. The administration of the composition can be effective to increase progression-free survival by 100 percent. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 150 days. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 165 days. The administration of the composition can be under conditions wherein the median time to progression for a population of mammals with the lymphoma is at least 170 days. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.9 run. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.8 μm. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.7 μm. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.6 μm. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.5 μm. The average diameter of at least 5 percent of the complexes of the composition can be from 0.2 μm to 0.4 μm. The average diameter of at least 10 percent of the complexes of the composition can be between 0.1 and 0.9 μm. The average diameter of at least 50 percent of the complexes of the composition can be between 0.1 and 0.9 μm. The average diameter of at least 75 percent of the complexes of the composition can be between 0.1 and 0.9 μm. The average diameter of at least 90 percent of the complexes of the composition can be between and 0.9 μm.
  • Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention pertains. Although methods and materials similar or equivalent to those described herein can be used to practice the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.
  • The details of one or more embodiments of the invention are set forth in the accompanying drawings and the description below. Other features, objects, and advantages of the invention will be apparent from the description and drawings, and from the claims.
  • DESCRIPTION OF DRAWINGS
  • FIG. 1 is a graph plotting the particle size distribution for ABRAXANE® (ABX) dissolved in Rituxan (RIT) as determined using a Mastersizer 2000E (Malvern Instruments Ltd., Worcestershire, England). ABX (10 mg/mL) was reconstituted in 1 mL of the indicated amount of RIT, and the mixtures were incubated at room temperature for 30 minutes,
  • FIG. 2 is a graph plotting percent change at seven days in tumor size from baseline of lymphoma (Daudi cell line) tumor bearing nude mice treated with PBS, Rituxan (RIT; 12 mg/kg) only, ABRAXANE® (30 mg/kg) only, Rituxan (RIT; 12 mg/kg) followed by ABRAXANE® (30 mg/kg) one day later, or AR160 complexes (0.159 μm; complexes where ABRAXANE® (10 mg/mL) was premixed with 2 mg/mL Rituxan and incubated for 60 minutes before injection).
  • FIG. 3 is a Kaplan Meier graph plotting survival of lymphoma (Daudi cell line) tumor bearing nude mice treated with PBS, Rituxan (RIT; 12 mg/kg) only, ABRAXANE® (30 mg/kg) only, Rituxan (RIT; 12 mg/kg) followed by ABRAXANE® (30 mg/kg) one day later, or AR160 complexes (0.159 βm; complexes where ABRAXANE® (10 mg/mL) was premixed with 2 mg/mL Rituxan and incubated for 60 minutes before injection).
  • DETAILED DESCRIPTION
  • This document provides methods and materials involved in treating lymphomas (e.g., mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, and non-Hodgkin lymphomas). For example, this document provides methods and materials for using complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD2( )polypeptide antibodies such as Rituximab) to treat lymphomas.
  • The methods and materials provided herein can be used to treat any type of lymphoma. For example, the methods and materials provided herein can be used to treat mature B-cell neoplasms, mature T cell neoplasms, mature natural killer cell neoplasms, immunodeficiency-associated lymphoproliferative disorders, Hodgkin lymphomas, or non-Hodgkin lymphomas. In some cases, the methods and materials provided herein can be used to treat lymphomas in any type of mammal including, without limitation, mice, rats, dogs, cats, horses, cows, pigs, monkeys, and humans.
  • In some cases, complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) can be designed to have an average diameter that is greater than 1 μm. For example, appropriate concentrations of albumin-containing nanoparticles and antibodies can be used such that complexes having an average diameter that is greater than 1 μm are formed. In some cases, manipulations such as centrifugation can be used to form preparations of albumin-containing nanoparticle/antibody complexes where the average diameter of those complexes is greater than 1 μm. In some cases, the preparations of albumin-containing nanoparticle/antibody complexes provided herein can have an average diameter that is between 1 μm and 5 μm e.g., between 1.1 μm and 5 μm, between 1.5 μm and 5 μm, between 2 μm and 5 μm, between 2.5 μm and 5 μm, between 3 μm and 5 μm, between 3.5 μm and 5 μm, between 4 μm and 5 μm, between 4.5 μm and 5 μm, between 1.1 μm and 4.5 μm, between 1.1 μm and 4 μm, between 1.1 μm and 3.5 μm, between 1.1 μm and 3 μm, between 1.1 μm and 2.5 μm, between 1.1 μm and 2 μm, or between 1.1 μm and 1.5 μm). Preparations of albumin-containing nanoparticle/antibody complexes provided herein having an average diameter that is between 1 μm and 5 μm can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body. In some cases, the preparations of albumin-containing nanoparticie/antibody complexes provided herein can have an average diameter that is between 5 μm and 50 μm (e.g., between 6 μm and 50 μm, between 7 μm and 50 μm, ween 10 μm and 50 μm, between 15 μm and 50 μm, between 20 μm and 50 μm, between 25 μm and 50 μm, between 30 μm and 50 μm, between 35 μm and 50 μm, between 5 μm and 45 μm, between 5 μm and 40 μm, between 5 μm and 35 μm, between 5 μm and 30 μm, between 5 μm and 25 μm, between 5 μm and 20 μm, between 5 μm and 15 μm, or between 10 μm and 30 μm). Preparations of albumin-containing nanoparticie/antibody complexes provided herein having an average diameter that is between 5 μm and 50 μm can be administered into a tumor (e.g., intratumorally) or in a region of a tumor located within a mammal's body.
  • In some cases, a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 1 μm and 5 μm (e.g., between 1.1 μm and 5 μm, between 1.5 μm and 5 μm, between 2 μm and 5 μm, between 2.5 μm and 5 μm, between 3 μm and 5 μm, between 3.5 μm and 5 μm, between 4 μm and 5 μm, between 4.5 μm and 5 μm, between 1.1 μm and 4.5 μm, between 1.1 μm and 4 μm, between 1.1 μm and 3.5 μm, between 1.1 μm and 3 μm, between 1.1 μm and 2.5 μm, between 1.1 μm and 2 μm, or between 1.1 μm and 1.5 μm). Preparation of albumin-containing nanoparticle/antibody complexes provided herein having greater than 60 percent (e,g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 1 μm and 5 μm can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body. In some cases, a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 5 μm and 50 μm (e.g., between 6 μm and 50 μm, between 7 μm and 50 μm, between 10 μm and 50 μm, between 15 μm and 50 μm, between 20 μm and 50 μm, between 25 μm and 50 μm, between 30 μm and 50 μm, between 35 μm and 50 μm, between 5 μm and 45 μm, between 5 μm and 40 μm, between 5 μm and 35 μm, between 5 μm and 30 μm, between 5 μm and 25 μm, between 5 μm and 20 μm, between 5 μm and 15 μm, or between 10 μm and 30 μm). Preparation of albumin-containing nanoparticle/antibody complexes provided herein having greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 5 μm and 50 μm can be administered into a tumor (e.g., intratumorally) or in a region of a tumor located within a mammal's body.
  • In some cases, complexes containing albumin-containing nanoparticles (e.g., ABRAXANE® nanoparticles) and antibodies (e.g., anti-CD20 polypeptide antibodies such as Rituximab) can be designed to have an average diameter that is less than 1 μm. For example, appropriate concentrations of albumin-containing nanoparticles and antibodies (e.g., Rituximab) can be used such that complexes having an average diameter that is less than 1 μm are formed. In some cases, the preparations of albumin-containing nanoparticle/antibody complexes provided herein can have an average diameter that is between 0.1 μm and 1 μm (e.g., between 0.1 μm and 0.95 μm, between 0.1 μm and 0.9 μm, between 0.1 μm and 0.8 μm, between 0.1 μm and 0.7 μm, between 0.1 μm and 0.6 μm, between 0.1 μm and 0.5 μm, between 0.1 μm and 0.4 μm, between 0.1 μm and 0.3 μm, between 0.1 μm and 0.2 μm, between 0.2 μm and 1 p,m, between 0.3 μm and 1 μm, between 0.4 μm and 1 μm, between 0.5 μm and 1 μm, between 0.2 μm and 0.6 μm, between 0.3 μm and 0.6 μm, between 0.2 μm and 0.5 μm, or between 0.3 μm and 0.5 μm). Preparations of albumin-containing nanoparticle/antibody complexes provided herein having an average diameter that is between 0.1 μm and 0.9 μm can be administered systemically (e.g., intravenously) to treat lymphomas located within a mammal's body.
  • In some cases, a preparation of albumin-containing nanoparticle/antibody complexes provided herein can have greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes having a diameter that is between 0.1 μm and 0.9 μm (e.g., between 0.1 μm and 0.95 μm, between 0.1 μm and 0.9 μm, between 0.1 μm and 0.8 μm, between 0.1 μm and 0.7 μm, between 0.1 μm and 0.6 μm, between 0.1 μm and 0.5 μm, between 0.1 μm and 0.4 μm, between 0.1 μm and 0.3 μm, between 0.1 μm and 0.2 μm, between 0.2 μm and 1 μm, between 0.3 μm and 1 μm, between 0.4 μm and 1 μm, between 0.5 μm and 1 μm, between 0.2 μm and 0.6 μm, between 0.3 μm and 0.6 μm, between 0.2 μm and 0.5 μm, or between 0.3 μm and 0.5 μm). Preparation of albumin-containing nanoparticte/antibody complexes provided herein having greater than 60 percent (e.g., greater than 65, 70, 75, 80, 90, 95, or 99 percent) of the complexes with a diameter that is between 0.1 μm and 0.9 μm can be administered systemically (e.g., intravenously) to treat cancers located within a mammal's body.
  • In general, albumin-containing nanoparticles such as ABRAXANE® can be contacted with an antibody such as an anti-CD20 polypeptide antibody (e.g., Rituximab) prior to administration to a human form an albumin-containing nanoparticle/antibody complex (e.g., an ABRAXANE®/anti-CD20 polypeptide antibody complex). Any appropriate albumin-containing nanoparticie preparation and any appropriate antibody can be used as described herein. For example, ABRAXANE® nanoparticles can be used as described herein. Examples of antibodies that can be used to form albumin-containing nanoparticle/antibody complexes as described herein include, without limitation, Rituximab (e.g., Rituxan™, MabThera™, or Zytux™). For example, an appropriate dose of ABRAXANE® and an appropriate dose of Rituximab can be mixed together in the same container. This mixture can be incubated at an appropriate temperature (e.g., room temperature, between 15° C. and 30° C., between 15° C and 25° C., between 20° C. and 30° C., or between 20° C. and 25° C.) for a period of time (e.g., about 30 minutes, or between about 5 minutes and about 60 minutes, between about 5 minutes and about 45 minutes, between about 15 minutes and about 60 minutes, between about 15 minutes and about 45 minutes, between about 20 minutes and about 400 minutes, or between about 25 minutes and about 35 minutes) before being administered to a cancer patient (e.g., a lymphoma patient). In some cases, ABRAXANE® can be contacted with an anti-CD20 polypeptide antibody by injecting both ABRAXANE® and the anti-CD20 polypeptide antibody either individually or as a pre-mixed combination into an IV bag containing an IV bag solution. The contents of the IV bag including ABRAXANE®/anti-CD20 polypeptide antibody complexes can be introduced into the patient to be treated.
  • In some cases, albumin-containing nanoparticles such as ABRAXANE® can be contacted with an antibody such as an anti-CD20 polypeptide antibody (e.g., Rituximab) to form albumin-containing nanoparticle/antibody complexes (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) that are stored prior to being administered to a cancer patient (e.g., a lymphoma patient). For example, a composition containing albumin-containing nanoparticle/antibody complexes can be formed as described herein and stored for a period of time (e.g., days or weeks) prior to being administered to a cancer patient.
  • Any appropriate method can be used to obtain albumin-containing nanoparticles such as ABRAXANE® and an antibody such as an anti-CD20 polypeptide antibody. For example, ABRAXANE® can be obtained from Celgene Corp. or as described elsewhere (U.S. Pat. No. 6,537,579). Rituximab can be obtained from Genentech Corp. or Roche Corp. or as described elsewhere (U.S. Pat. No. 5,736,137).
  • In some cases, the combination of an albumin-containing nanoparticle such as ABRAXANE® and an antibody such as anti-CD20 polypeptide antibody can include one or more other agents such as an alkylating agent (e.g., a platinum compound). Examples of platinum compounds that can be used as an alkylating agent include, without carboplatin (PARAPLATIN®), cisplatin (PLATINOL®), oxaliplatin (ELOXATIN®), and BBR3464. Examples of other agents that can be included within an albumin-containing nanoparticle/antibody complex provided herein include, without limitation, adriamycin, cyclophosphamide, vincristine, prednisone, dexamethasone, cytarabine, methotrexate, thiotepa, ifosfamide, chlorambucil, dacarbazine, bleomycin, ibrutinib, campath-B, gemcitabine, revlimid, sirolimus, temsirolimus, bexxar, brentuximab, bendamustine, and etoposide. For example, an albumin-containing nanoparticle/antibody complex provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complex) can include brentuximab, cyclophosphamide, adriamycin, or vincristine as part of the complex.
  • Any appropriate method can be used to administer an albumin-containing nanoparticle/antibody complex provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) to a mammal. For example, a composition containing albumin-containing nanoparticle/antibody complexes such as ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered via injection(e.g., subcutaneous injection, intramuscular injection, intravenous injection, or intrathecal injection).
  • Before administering a composition containing an albumin-containing nanoparticie/antibody complex provided herein (e.g.4BRAXANE®/anti-CD20 polypeptide antibody complexes) to a mammal, the mammal can be assessed to determine whether or not the mammal has lymphoma. Any appropriate method can be used to determine whether or not a mammal has lymphoma. For example, a mammal (e.g., human) can be identified as having lymphoma using standard diagnostic techniques. In some cases, a tissue biopsy (e.g., lymph node tissue sample) can be collected and analyzed to determine whether or not a mammal has lymphoma.
  • After identifying a mammal as having lymphoma, the mammal can be administered a composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes). For example, a composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered prior to or in lieu of surgical resection of a tumor. In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered following resection of a tumor.
  • A composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be administered to a mammal in any appropriate amount, at any appropriate frequency, and for any appropriate duration effective to achieve a desired outcome (e.g., to increase progression-free survival). In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be administered to a mammal having lymphoma to reduce the progression rate of the lymphoma by 5, 10, 25, 50, 75, 100, or more percent. For example, the progression rate can be reduced such that no additional cancer progression is detected. Any appropriate method can be used to determine whether or not the progression rate of lymphoma is reduced. For example, the progression rate of lymphoma can be assessed by imaging tissue at different time points and determining the amount of cancer cells present. The amounts of cancer cells determined within tissue at different times can be compared to determine the progression rate. After treatment as described herein, the progression rate can be determined again over another time interval. In some cases, the stage of cancer (e,g., lymphoma) after treatment can be determined and compared to the stage before treatment to determine whether or not the progression rate was reduced.
  • In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be administered to a mammal having lymphoma under conditions where progression-free survival is increased (e.g., by 5, 10, 25, 50, 75, 100, or more percent) as compared to the median progression-free survival of corresponding mammals having untreated lymphoma or the median progression-free survival of corresponding mammals having lymphoma treated with ABRAXANE® and an antibody (e.g., an anti-CD20 polypeptide antibody) without forming ABRAXANE®/antibody complexes (e.g., without forming ABRAXNE/anti-CD20 polypeptide antibody complexes). In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma to increase progression-free survival by 5, 10, 25, 50, 75, 100, or more percent as compared to the median progression-free survival of corresponding mammals having lymphoma and having received ABRAXANE® or an antibody (e.g., an anti-CD20 polypeptide antibody) alone, Progression-free survival can be measured over any length of time (e.g., one month, two months, three months, four months, five months, six months, or longer),
  • In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be administered to a mammal having lymphoma under conditions where the 8-week progression-free survival rate for a population of mammals is 65% or greater (e.g., 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80% or greater) than that observed in a population of comparable mammals not receiving a composition containing albumin-containing nanoparticie/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes). In some cases, a composition containing albumin-containing nanoparticle/antibody complexes provided herein(e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered to a mammal having lymphoma under conditions where the median time to progression for a population of mammals is at least 150 days (e.g., at least 155, 160, 163, 165, or 170 days).
  • An effective amount of a composition containing albumin-containing nanoparticle/antibody complexes provided herein e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be any amount that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal. Typically, an effective amount of ABRAXANE® can be from about 50 mg/m2 to about 150 mg/m2 (e.g., about 80 mg/m2), and an effective amount of an anti-CD20 polypeptide antibody such as Rituximab can be from about 5 mg/kg to about 20 mg/kg (e.g., about 10 mg/kg or 375 mg/m2). If a particular mammal fails to respond to a particular amount, then the amount of ABRAXANE® or anti-CD20 polypeptide antibody can be increased by, for example, two fold. After receiving this higher concentration, the mammal can be monitored for both responsiveness to the treatment and toxicity symptoms, and adjustments made accordingly. The effective amount can remain constant or can be adjusted as a sliding scale or variable dose depending on the mammal's response to treatment. Various factors can influence the actual effective amount used for a particular application. For example, the frequency of administration, duration of treatment, use of multiple treatment agents, route of administration, and severity of the lymphoma may require an increase or decrease in the actual effective amount administered.
  • The frequency of administration can be any frequency that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal. For example, the frequency of administration can be from about once a month to about three times a month, or from about twice a month to about six times a month, or from about once every two months to about three times every two months. The frequency of administration can remain constant or can be variable during the duration of treatment. A course of treatment with a composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can include rest periods. For example, a composition containing ABRAXANE®/anti-CD20 polypeptide antibody complexes can be administered over a two week period followed by a two week rest period, and such a regimen can be repeated multiple times. As with the effective amount, various factors can influence the actual frequency of administration used for a particular application. For example, the effective amount, duration of treatment, use of multiple treatment agents, route of administration, and severity of the lymphoma may require an increase or decrease in administration frequency.
  • An effective duration for administering a composition provided herein can be any duration that reduces the progression rate of lymphoma, increases the progression-free survival rate, or increases the median time to progression without producing significant toxicity to the mammal. Thus, the effective duration can vary from several days to several weeks, months, or years. In general, the effective duration for the treatment of lymphoma can range in duration from several weeks to several months. :In some cases, an effective duration can be for as long as an individual mammal is alive. Multiple factors can influence the actual effective duration used for a particular treatment. For example, an effective duration can vary with the frequency of administration, effective amount, use of multiple treatment agents, route of administration, and severity of the lymphoma.
  • A composition containing albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be in any appropriate form. For example, a composition provided herein can be in the form of a solution or powder with or without a diluent to make an injectable suspension. A composition also can contain additional ingredients including, without limitation, pharmaceutically acceptable vehicles. A pharmaceutically acceptable vehicle can be, for example, saline, water, lactic acid, mannitol, or combinations thereof.
  • After administering a composition provided herein to a mammal, the mammal can be monitored to determine whether or not the lymphoma was treated. For example, a mammal can be assessed after treatment to determine whether or not the progression rate of lymphoma was reduced (e.g., stopped). As described herein, any method can be used to assess progression and survival rates.
  • In some cases, a formulation of ABRAXANE®/Rituxan complexes described in Example 1 can be administered to a human lymphoma patient as described in the methods set forth in Example 3.
  • In some cases, nanoparticles containing albumin (e.g., nanoparticles with an albumin shell) and an agent other than placitaxel can be used as described herein in place of or in combination with ABRAXANE®. For example, albumin-containing nanoparticles designed to carry a cancer chemotherapeutic agent can be used to form nanoparticle/anti-CD20 polypeptide antibody complexes that can be used as described herein. An example of such a cancer chemotherapeutic agent includes, without vinblastine.
  • In some cases, a composition can be formulated to include nanoparticies containing albumin (e,g., nanoparticles with an albumin shell) that are conjugated to an antibody, agent, or combination of antibodies and agents to form complexes for treating lymphoma. For example, albumin nanoparticles can be formulated to include adriamycin, cyclophosphamide, vincristine, prednisone, dexamethasone, cytarabine, methotrexate, thiotepa, ifosfamide, chlorambucil, dacarbazine, bleomycin, ibrutinib, campath-B, gemcitabine, revlimid, sirolimus, temsirolimus, bexxar, brentuximab, bendamustine, etoposide, or combinations thereof with or without including rituximab.
  • In some cases, nanoparticles containing albumin (e.g., nanoparticles with an albumin shell) or a complex described herein (e.g., ABRAXANE®/rituximab complexes can be formulated to include one or more anti-chronic inflammation treatment agents designed to reduce the global state of immune dysfunction and/or chronic inflammation present within a cancer patient. For example, steroidal anti-inflammatory agents (e.g., prednisone), non-steroidal anti-inflammatory agents (e.g., naproxen), lympho-depleting cytotoxic agents (e.g., cyclophosphamide), immune cell and/or cytokine targeting antibodies (e.g., infliximab), or a combination thereof can be incorporated into nanoparticles containing albumin or ABRAXANE®/rituximab complexes. In some cases, anti-IL-4 agents (e.g., anti-IL-4 antibodies), anti-IL-13 agents (e.g., soluble IL-13 receptor), and combinations thereof can be incorporated into nanoparticles containing albumin or ABRAXANE®/rituximab complexes.
  • Any appropriate method can be used to assess whether or not the global state of immune dysfunction and/or chronic inflammation was reduced following an anti-chronic inflammation treatment. For example, cytokine profiles (e.g., IL-4, IL-13, IL-4, IL-13, IL-5, IL-10, IL-2, and interferon gamma) present in blood can be assessed before and after an anti-chronic inflammation treatment to determine whether or not the global state of immune dysfunction and/or chronic inflammation was reduced.
  • The invention will be further described in the following examples, which do not limit the scope of the invention described in the claims.
  • EXAMPLES Example 1 Making ABRAXANE®/Rituxan® Complexes
  • ABRAXANE® was incubated with various increasing concentrations of Rituxan® (rituximab) to form ABRAXANE®/Rituxan® complexes of increasing diameter. Ten milligrams of ABRAXANE® was reconstituted in 1 mL of Rituxan® at 0, 2, 4, 6, 8, and 10 mg/mL, and the mixture was incubated at room temperature for 30 minutes. After incubation, the distributions of particle sizes were determined with the Mastersizer 2000. The median particle size ranged from 0.147 μm to 8.286 μm for 0 and 10 mg/mL Rituxan®, respectively (FIG. 1). These results demonstrate that the antibody concentration in which ABRAXANE® is incubated impacts the size of the nanoparticle. Manipulating the size of the particles can change the pharmacokinetics of the drug complex as well as its bio-distribution, which in turn can improve the clinical efficacy of the drug complex.
  • Example 2 ABRAXANE®/Rituxan Complexes Inhibit Tumor Growth More Effective Than ABRAXANE® Alone, Rituxan Alone, and the Sequential Use of Rituxan and ABRAXANE®
  • Female athymic nude mice were injected with 1×106 lymphoma cells (Daudi cell line). Tumors were allowed to grow, and treatments were administered when tumors were between 800 and 1000 mm3. Mice were treated with (a) a single dose of 100 μL PBS on day 0, (b) a single dose of Rituxan (12 mg/kg) on day 0, (c) a single dose of ABRAXANE® (30 mg/kg) on day 0, (d) a single dose of Rituxan (12 mg/kg) on day 0 followed by a single dose of ABRAXANE® (30 mg/kg) on day 1, or (e) a single dose of AR160 (equivalent to 30 mg/kg of ABRAXANE®) on day 0. Tumor volumes (mm3) were monitored, and the percent change in tumor volume seven days after treatment was determined.
  • The AR160 complexes were produced as follows. 10 mg ABRAXANE® was reconstituted in 2 mg of Rituxan in 500 μL 0.9% saline and incubated for 1 hour at room temperature. After incubation, AR160 was brought to 1 mL with 0.9% saline. AR160 was further diluted, and 100 μL was administered to mice at a dose equivalent to 12 mg/kg Rituxan and 30 mg/kg ABRAXANE®. Average particle size for AR160 was 0.159 μm.
  • On day 7 post treatment, the mice treated with AR160 exhibited tumors with significantly smaller tumor size as compared to the other treatment groups (FIG. 2). Survival data also revealed an improvement for mice treated with ABRAXANE®/Rituxan complexes (FIG. 3). These results demonstrate that the albumin-containing nanoparticle/antibody complexes provided herein (e.g., ABRAXANE®/anti-CD20 polypeptide antibody complexes) can be used effectively to treat lymphomas,
  • Example 3 ABRAXANE®/Rituxan Complexes as Targeted Therapy for Lymphomas
  • The treatment schedule for ABRAXANE®/Rituxan complexes is repeated each month (every 28 days +/−3 days) or until disease progression, patient refusal, or unacceptable toxicity (Table 1) with the indicated dose escalation scheme (Table 2) and dose limiting toxicities (Table 3).
  • TABLE 1
    Agent Dose Route Days ReRx
    ABRAXANE ®/ assigned IV over 60 minutes 1, 8 Every 28
    Rituxan at time of (only 1st dose; and days*
    complexes registration subsequent doses 15
    infused over 30
    minutes)
    *One treatment cycle = 28 days +/− 3 days
  • TABLE 2
    Dose Escalation Scheme.
    Dose Level Dose (ABX) Dose (RIT)
    −2   75 mg/m 2 30 mg/m2
    −1  100 mg/m 2 40 mg/m 2
     1* 125 mg/m2 50 mg/m 2
    2 150 mg/m 2 60 mg/m 2
    3 175 mg/m2 70 mg/m2
    *Starting dose.
  • TABLE 3
    Dose Limiting Toxicities (DLT).
    Toxicity DLT Definition
    Hematologic Grade
    4 ANC, Grade 4 Hgb, or PLT <25,000
    Renal Serum creatinine ≧2 times baseline
    Other nonhematologic grade 3 as per NCI Common Terminology
    Criteria for Adverse Events (CTCAE) version
    4.0
  • Determination of Maximum Tolerated Dose (MTD)
  • The maximum tolerated dose is defined as the highest dose level among those tested where at most one out of six patients develops a DLT prior to the start of their second cycle of treatment and the next highest dose level is such that two out of a maximum of six patients treated at this dose level developed a DLT prior to the start of their second cycle of treatment.
  • Enrollment and Determination of MTD
  • A minimum of two or a maximum of six patients are accrued to a given dose level. For dose level 1 (and if accrued to, dose levels −1 & −2), enrollment is temporarily halted after each patient has been enrolled in order to gather acute adverse event data over the first cycle of their treatment. For dose levels 2 & 3, patients are accrued to these dose levels so that at any given time no more than two patients are receiving their first cycle of treatment and acute adverse event data over the first treatment cycle for all other patients treated at the current dose level is known. If, at any time in the enrollment process, two patients treated at the current dose level develop a DLT during the first cycle of treatment, enrollment is closed to that dose level. Enrollment is re-opened to the next lower dose level if fewer than six patients have been treated at that dose level. If none of the first three patients treated at a given dose level develops a DLT during the first cycle of treatment, enrollment to the dose level is closed and enrollment is reopen at next higher dose level. If there are no other higher dose levels to be tested, three additional patients are enrolled at the current dose level to confirm MTD. If one of the first three patients treated at a given dose level develops a DLT during the first cycle of treatment, three additional patients are enrolled (sequentially) onto the current dose level. If at any time in the enrollment of these three additional patients, a patient develops a DLT, enrollment is closed to this dose level. Enrollment is re-opened to the next lower dose level if fewer than six patients are treated at that dose level. If none of these three additional patients develops a DLT during the first cycle of treatment, enrollment to this dose level is closed and enrollment is reopened at next higher dose level. If there are no other higher dose levels to be tooted, this is considered the MTD.
  • For this protocol, the patient returns for evaluation and retreatment (at least every 28+/−3 days) according to the schedule. If a patient fails to complete the first cycle of treatment for reasons other than toxicity, an additional patient is enrolled to replace this patient.
  • Dosage Modification Based on Adverse Events
  • The modifications in Table 4 are followed until individual treatment tolerance is ascertained. If multiple adverse events (Table 5) are seen, dose is administered based on greatest reduction required for any single adverse event observed. Dose modifications apply to the treatment given in the preceding cycle and are based on adverse events observed since the prior dose.
  • TABLE 4
    Dose Levels Based on Adverse Events.
    ABRAXANE ®/Rituxan complexes - Both drugs are reduced
    Dose Accompanying RIT dose
    Level ABX dose (40% of ABX dose)
    2 175 mg/m2 70 mg/m2
    −1 150 mg/m 2 60 mg/m 2
    1 125 mg/m2 50 mg/m2
    −2 100 mg/m 2 40 mg/m2
    −2  75 mg/m 2 30 mg/m2
    *Dose level 1 refers to the starting dose.
  • TABLE 5
    Use Common Terminology Criteria for Adverse Events
    (CTCAE) v. 4.0* unless otherwise specified
    CTCAE Category Adverse Event Dose Reduction
    Investigations ANC <1000 Day 1: Hold until counts above these levels.
    or Day 8: Omit dose that day and retreat at same dose
    PLT <75,000 level on day 15 if counts have recovered.
    Day 15: Omit dose that day.
    NOTE: if two consecutive cycles of therapy require
    omission of a dose, subsequent treatment cycles
    should begin (day 1) at next lower dose.
    AST or Day 1: Hold until resolved to <Grade 2 then reduce
    Alkaline dose by ONE dose level.
    Phosphatase ≧ If treatment needs to be held >4 weeks, discontinue
    Grade 2 study treatment and go to event monitoring.
    Neurology Neuropathy
    disorders Grade 2 Day 1: Hold until resolved to <Grade 2 then reduce
    dose by ONE dose level.
    Day 8 OR 15- Omit dose that day. If resolved to <
    Grade 2 by next scheduled dose, then dose reduce
    by one level
    If treatment needs to be held >4 weeks, discontinue
    study treatment and go to Event Monitoring
    All other non- ≧Grade 3 Day 1: Hold until resolved to ≦Grade 2 then reduce
    hematologic dose by ONE dose level.
    adverse events Day 8: Omit dose that day. If resolved to ≦Grade 2
    by day 15, then dose reduce by one level and retreat.
    Day 15: Omit dose that day.
    NOTE: if two consecutive cycles of therapy require
    omission of a dose, subsequent treatment cycles
    should begin (day 1) at next lower dose.
    If treatment needs to be held >4 weeks, discontinue
    study treatment and go to Event Monitoring
    Gastrointestinal Bowel Discontinue all study treatment and proceed to
    Disorders perforation Event Monitoring
    Bowel
    Obstruction
    Grade
    1 Continue patient on study for partial bowel
    obstruction NOT requiring medical intervention.
    Grade 2 Hold for partial obstruction requiring medical
    intervention. If resolved to Grade 0 within 4 weeks,
    treatment may be restarted. If treatment needs to be
    held >4 weeks, discontinue all study treatment and
    go to Event Monitoring.
    Grade 3 or 4 For complete bowel obstruction, discontinue study
    treatment and proceed to Event Monitoring
    Cardiac Disorders Hypertension
    Grade 3 Hypertension should be treated as per general
    practice. If hypertension (≧150/100) persists despite
    treatment, hold treatment until blood pressure is
    below this level
    If treatment needs to be held >4 weeks due to
    uncontrolled hypertension, discontinue study
    treatment and go to Event Monitoring.
    Left ventricular
    systolic
    function-
    Grade 3 Hold until resolution to Grade ≦1. If treatment
    needs to be held >4 weeks, discontinue all study
    treatment and go to Event Monitoring.
    Grade 4 Discontinue treatment and proceed to Event
    Monitoring
    Respiratory, Bronchopulmonary
    thoracic and Hemorrhage
    mediastinal Grade 2 Discontinue all study treatment and proceed to
    disorders Event Monitoring
    Coagulation Hemorrhage
    Grade
    3 Hold until ALL of the following criteria are met:
    1. Bleeding has resolved and Hb is stable.
    2. There is no bleeding diathesis that would increase
    the risk of therapy.
    3. There is no anatomic or pathologic condition that
    could increase the risk of hemorrhage recurrence.
    If treatment needs to be held >4 weeks, discontinue
    study treatment and go to Event Monitoring
    Patients who experience a recurrence of Grade 3
    hemorrhage are to discontinue all study treatment
    and proceed to Event Monitoring.
    Grade 4 Discontinue study treatment and proceed to Event
    Monitoring
    Bleeding
    diathesis
    Grade
    3 or 4 Discontinue study treatment and proceed to Event
    Monitoring
    Vascular disorders Venous
    thrombosis
    Grade
    3 Hold treatment. If the planned duration of full-
    or dose anticoagulation is <2 weeks, treatment should
    asymptomatic be held until the full-dose anticoagulation period
    Grade
    4 is over.
    If the planned duration of full-dose
    anticoagulation is >2 weeks, treatment may be
    resumed during the period of full-dose
    anticoagulation IF all of the criteria below are
    met:
    The subject must have an in-range INR
    (usually 2-3) on a stable dose of warfarin, or on
    stable dose of heparin prior to restarting
    treatment.
    The subject must not have pathological
    conditions that carry high risk of bleeding (e.g.
    tumor involving major vessels or other
    conditions)
    The subject must not have had hemorrhagic
    events while on study
    If thromboemboli worsen/recur upon resumption
    of study therapy, discontinue treatment.
    Symptomatic
    Grade
    4 Discontinue treatment and proceed to Event
    Arterial Monitoring
    thrombosis Discontinue treatment and proceed to Event
    (Angina, Monitoring
    myocardial
    infarction,
    transient
    ischemic attack,
    cerebrovascular
    accident, or any
    other arterial
    thromboembolic
    events) ANY
    Grade
  • Ancillary Treatment/Supportive Care
  • Routine use of colony-stimulating factors (G-CSF or GM-CSF) is not recommended. Prophylactic use of colony-stimulating factors during the study is not allowed. Therapeutic use in patients with serious neutropenic complications such as tissue infection, sepsis syndrome, fungal infection, etc., may be considered at physician discretion. Recombinant erythropoietin to maintain adequate hemoglobin levels and avoid packed red blood cell transfusions is allowed.
  • Patients should receive full supportive care while on this study. This includes blood product support, antibiotic treatment and treatment of other newly diagnosed or concurrent medica conditions. All blood products and concomitant medications such as antidiarrheals, analgesics, and anti-emetics received from the first administration of study drugs until 30 days after the final dose are to be recorded in the medical record. Patients participating in phase I program clinical trials are not to be considered for enrollment in any other study involving a pharmacologic agent-(drugs, biologics, immunotherapy approaches, gene therapy) whether for symptom control or therapeutic intent.
  • Hypersensitivity Reactions
  • Patients do not require premedication prior to administration of ABRAXANE®/Rituxan complexes. In the unlikely event of a hypersensitivity reaction, treatment with antihistamines, H2 blockers, and corticosteroids is recommended. Patients should be pre-medicated with the typical regimen for paclitaxel regimens for subsequent cycles. In the unlikely event of a mild hypersensitivity reaction, premedication may be administered using the premedication regimen the institution typically uses for solvent-based paclitaxel.
  • ABRAXANE®/Rituxan Complexes
  • ABRAXANE®/Rituxan complexes are prepared as a hazardous low risk product. ABRAXANE® is supplied as a white to off-white lyophilized powder containing 100 mg of paclitaxel and approximately 900 mg Albumin Human USP (HA) as a stabilizer in a 50 mL, single-use vial. Each vial of the lyophilized product is reconstituted as set forth below. Unreconstituted ABRAXANE® is stored at controlled room temperature in its carton. Reconstituted ABRAXANE® is used immediately. Rituxan is classified as an anti-CD20 monoclonal antibody.
  • The dose appropriate number of vials of Rituxan are obtained, and each vial is further diluted per the following directions to 4 mg/mL. The dose appropriate number of ABRAXANE® (paclitaxel) 100 mg vials is obtained and each vial is reconstituted per the following directions to a final concentration containing 10 mg/mL nanoparticle albumin-bound (nab) paclitaxel. It is not a requirement to use filter needles in the preparation of, or in-line filters during administration. In addition, filters of pore-size less than 15 micrometers are to be avoided.
  • As with other cytotoxic anticancer drugs, caution is exercised in handling ABRAXANE®. The use of gloves is recommended.
  • Using a sterile 3 mL syringe, 1.6 mL (40 mg) of Rituxan 25 mg/mL is withdraw and slowly injected, over a minimum of 1 minute, onto the inside wall of each of the vials containing 100 mg of ABRAXANE®. Unused Rituxan left in the 25 mg/mL vial is discarded, as the product contains no preservatives. Injecting the Rituxan solution directly onto the lyophilized cake is avoided as this will result in foaming. Using a sterile 12 mL sterile syringe, 8.4 mL of 0.9% Sodium Chloride injection, USP, is withdraw and slowly injected, over a minimum of 1 minute, onto the inside wall of each vial containing ABRAXANE® 100 mg and Rituxan 40 mg. Once the addition of Rituxan 1.6 mL and 0.9% Sodium Chloride Injection, USP 8.4 mL is complete in each vial, each vial is gently swirled and/or inverted slowly for at least 2 minutes until complete dissolution of any cake/powder occurs. The generation of foam is avoided. The concentration of each vial is 100 mg/10 mL ABRAXANE® and 40 mg/10 mL Rituxan. The vials containing the ABRAXANE® and Rituxan are allowed to sit for 60 minutes. The vial(s) are gently swirled and/or inverted every 10 minutes to continue to mix the complexes. After 60 minutes is elapsed, a sterile 60- to 100-mL syringe (appropriate size for the volume being administered) is used to withdraw the calculated dosing volume of ABRAXANE® and Rituxan from each vial. A sufficient quantity of 0.9% Sodium Chloride Injection, USP is added to make the final concentration of ABRAXANE® 5 mg/mL and Rituxan 2 mg/mL. The syringe is gently swirled and/or inverted slowly for 1 minute to mix. The storage and stability is for up to 4 hours at room temperature following final dilution.
  • Administration
  • The IV initial complex dose is infused over 60 minutes via syringe pump. The infusion may be shortened to 30 minutes if the initial infusion is well tolerated. Infusion is monitored closely during the infusion process for signs/symptoms of an infusion reaction. The patient's line is flushed after administration with 20 mL 0.9% Sodium Chloride. An example calculation and preparation is as follows:

  • Dose level 1: ABRAXANE® 125 mg/m2/Rituxan 50 mg/m2 BSA=2 m2
      • Doses required: ABRAXANE® 250 mg/Rituxan 100 mg
      • Obtain three 100 mg vials of ABRAXANE®.
      • Obtain one 100 mg vial of Rituxan 25 mg/mL.
      • Withdraw 1.6 mL (40 mg) of Rituxan 25 mg/mL and slowly inject over 1 minute onto the inside wall of one of the 100 mg ABRAXANE® vials. Repeat this procedure for each of the remaining two ABRAXANE 100 mg vials.
      • Add 8.4 mL 0.9% Sodium Chloride Injection, LISP onto the inside wall of one of the vials containing ABRAXANE® and Rituxan. Repeat this procedure for each of the remaining two ABRAXANE® and Rituxan vials.
      • Let mixture sit for 60 minutes (swirling every 10 minutes). The final concentration of each vial should be 100 mg ABRAXANE®/10 mL and 40 mg Rituxan/10 mL.
      • Withdraw 25 mL from the ABRAXANE° and Rituxan containing vial and place in a 100 mL sterile syringe. Add 25 mL 0.9% Sodium Chloride Injection, USP for a final ABRAXANE® concentration of 5 mg/mL and Rituxan concentration of 2 mg/mL. Infuse via syringe pump over 60 minutes (first dose; 30 minutes subsequent doses).
    Response to ABRAXANE®/Rituxan Complex Treatment
  • Each patient's response to treatment with a ABRAXANE®/Rituxan complex formulation is monitored.
  • Other Embodiments
  • It is to be understood that while the invention has been described in conjunction with the detailed description thereof, the foregoing description is intended to illustrate and not limit the scope of the invention, which is defined by the scope of the appended claims. Other aspects, advantages, and modifications are within the scope of the following claims.

Claims (2)

1-72. (canceled)
73. A method for treating a mammal having lymphoma, said method comprising administering to said mammal a composition comprising nanoparticles containing albumin and placitaxel complexed with an anti-CD20 polypeptide antibody under conditions wherein the length of progression-free survival is increased.
US15/030,567 2014-06-13 2015-06-12 Treating Lymphomas Abandoned US20160250351A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US15/030,567 US20160250351A1 (en) 2014-06-13 2015-06-12 Treating Lymphomas

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201462012190P 2014-06-13 2014-06-13
US15/030,567 US20160250351A1 (en) 2014-06-13 2015-06-12 Treating Lymphomas
PCT/US2015/035505 WO2015191969A1 (en) 2014-06-13 2015-06-12 Treating lymphomas

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2015/035505 A-371-Of-International WO2015191969A1 (en) 2014-06-13 2015-06-12 Treating lymphomas

Related Child Applications (1)

Application Number Title Priority Date Filing Date
US15/452,669 Continuation US20170216453A1 (en) 2014-06-13 2017-03-07 Treating Lymphomas

Publications (1)

Publication Number Publication Date
US20160250351A1 true US20160250351A1 (en) 2016-09-01

Family

ID=54834378

Family Applications (2)

Application Number Title Priority Date Filing Date
US15/030,567 Abandoned US20160250351A1 (en) 2014-06-13 2015-06-12 Treating Lymphomas
US15/452,669 Pending US20170216453A1 (en) 2014-06-13 2017-03-07 Treating Lymphomas

Family Applications After (1)

Application Number Title Priority Date Filing Date
US15/452,669 Pending US20170216453A1 (en) 2014-06-13 2017-03-07 Treating Lymphomas

Country Status (12)

Country Link
US (2) US20160250351A1 (en)
EP (1) EP3154586B1 (en)
JP (1) JP6695286B2 (en)
KR (1) KR102450563B1 (en)
CN (2) CN113318239A (en)
AU (2) AU2015274408A1 (en)
BR (1) BR112016029123A2 (en)
CA (1) CA2950926A1 (en)
IL (1) IL249454B (en)
MX (2) MX2016016369A (en)
RU (1) RU2708332C2 (en)
WO (1) WO2015191969A1 (en)

Cited By (18)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US10279035B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
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
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
US11351254B2 (en) 2016-02-12 2022-06-07 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US11531030B2 (en) 2017-04-21 2022-12-20 Mayo Foundation For Medical Education And Research Polypeptide-antibody complexes and uses thereof
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
US11971412B2 (en) 2022-11-08 2024-04-30 Mayo Foundation For Medical Education And Research Polypeptide-antibody complexes and uses thereof

Families Citing this family (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MX2019001517A (en) * 2016-08-05 2019-07-12 Mayo Found Medical Education & Res Modified antibody-albumin nanoparticle complexes for cancer treatment.
MX2019003988A (en) * 2016-10-10 2019-08-14 Abraxis Bioscience Llc Nanoparticle formulations and methods of making and using thereof.
WO2019157039A1 (en) * 2018-02-06 2019-08-15 Mayo Foundation For Medical Education And Research Antibody-peptide complexes and uses thereof
US20230338574A1 (en) 2020-09-02 2023-10-26 Mayo Foundation For Medical Education And Research Antibody-nanoparticle complexes and methods for making and using the same

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100112077A1 (en) * 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer

Family Cites Families (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5736137A (en) 1992-11-13 1998-04-07 Idec Pharmaceuticals Corporation Therapeutic application of chimeric and radiolabeled antibodies to human B lymphocyte restricted differentiation antigen for treatment of B cell lymphoma
US6537579B1 (en) 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
CA2577133A1 (en) * 2004-08-19 2006-03-23 Genentech, Inc. Polypeptide variants with altered effector function
KR20090082386A (en) * 2006-11-07 2009-07-30 다우 아그로사이언시즈 엘엘씨 Sprayable, controlled-release, male annihilation technique (mat) formulation and insect control method
US8345535B2 (en) * 2009-07-13 2013-01-01 Lg Electronics Inc. Method and apparatus for generating ranging preamble code in wireless communication system
AR078161A1 (en) * 2009-09-11 2011-10-19 Hoffmann La Roche VERY CONCENTRATED PHARMACEUTICAL FORMULATIONS OF AN ANTIBODY ANTI CD20. USE OF THE FORMULATION. TREATMENT METHOD
CN102933231B (en) * 2010-02-10 2015-07-29 伊缪诺金公司 CD20 antibody and uses thereof
EP2625525A4 (en) * 2010-10-08 2014-04-02 Abraxis Bioscience Llc Sparc microenvironment signature, plasma sparc, and ldh as prognostic biomarkers in the treatment of cancer
US9427477B2 (en) * 2011-05-09 2016-08-30 Mayo Foundation For Medical Education And Research Cancer treatments
EP2903610B1 (en) * 2012-10-01 2021-11-03 Mayo Foundation For Medical Education And Research Cancer treatments

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100112077A1 (en) * 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Jazirehi et al. " Rituximab (anti-CD20) selectively modifies Bcl-xl and apoptosis protease activating factor-1 (Apaf-1) expression and sensitizes human non-Hodgkin's lymphoma B cell lines to paclitaxel-induced apoptosis ", Mol Cancer Ther. 2003;2:1183-1193 *

Cited By (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US10765741B2 (en) 2011-05-09 2020-09-08 Mayo Foundation For Medical Education And Research Methods for treating VEGF-expressing cancer using preformed nanoparticle complexes comprising albumin-bound paclitaxel and bevacizumab
US10441656B2 (en) 2012-10-01 2019-10-15 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin, and methods of making the same
US11648311B2 (en) 2012-10-01 2023-05-16 Mayo Foundation For Medical Education And Research Cancer treatments
US10279035B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10307482B2 (en) 2012-10-01 2019-06-04 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, cetuximab, and albumin
US10279036B2 (en) 2012-10-01 2019-05-07 Mayo Foundation For Medical Education And Research Antibody-albumin nanoparticle complexes comprising albumin, bevacizumab, and paclitaxel, and methods of making and using the same
US10376579B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and anti-VEGF antibody for treatment of cancer
US10376580B2 (en) 2012-10-01 2019-08-13 Mayo Foundation For Medical Education And Research Methods of treating cancer with antibody-albumin nanoparticle complexes comprising albumin, trastuzumab, and paclitaxel
US10493150B2 (en) 2012-10-01 2019-12-03 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US10406224B2 (en) 2012-10-01 2019-09-10 Mayo Foundation For Medical Education And Research Nanoparticle complexes of paclitaxel, trastuzumab, and albumin
US10413606B2 (en) 2012-10-01 2019-09-17 Mayo Foundation For Medical Education And Research Methods for treating cancer with nanoparticle complexes of albumin-bound paclitaxel and anti-VEGF antibodies
US10420839B2 (en) 2012-10-01 2019-09-24 Mayo Foundation For Medical Education And Research Methods for treating CD52-expressing cancers using compositions comprising nanoparticle complexes of paclitaxel, alemtuzumab, and albumin
US10507243B2 (en) 2012-10-01 2019-12-17 Mayo Foundation For Medical Education And Research Nanoparticle complexes of rituximab, albumin and pacltaxel
US10471145B2 (en) 2012-10-01 2019-11-12 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, rituximab, and albumin
US10478495B2 (en) 2012-10-01 2019-11-19 Mayo Foundation For Medical Education And Research Methods for treating cancer using nanoparticle complexes of paclitaxel, cetuximab, and albumin
US10668151B2 (en) 2012-10-01 2020-06-02 Mayo Foundation For Medical Education And Research Nanoparticle complexes of albumin, paclitaxel, and panitumumab for treatment of cancer
US11285221B2 (en) 2014-06-16 2022-03-29 Mayo Foundation For Medical Education And Research Treating myelomas
US10213513B2 (en) 2014-06-16 2019-02-26 Mayo Foundation For Medical Education And Research Treating myelomas
US10624846B2 (en) 2014-10-06 2020-04-21 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-antibody paclitaxel nanoparticle complexes
US10993912B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10300016B2 (en) 2014-10-06 2019-05-28 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US11433023B2 (en) 2014-10-06 2022-09-06 Mayo Foundation For Medical Education And Research Albumin-PD-1 paclitaxel nanoparticle complex compositions and methods of making and using the same
US10322084B2 (en) 2014-10-06 2019-06-18 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10391055B2 (en) 2014-10-06 2019-08-27 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10772833B2 (en) 2014-10-06 2020-09-15 Mayo Foundation For Medical Education And Research Albumin-CTLA-4 paclitaxel nanopartilce complex compositions and methods of making and using the same
US10596111B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of making lyophilized compositions comprising albumin-VEGF paclitaxel nanoparticle complexes
US10780049B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprising albumin-rankl or albumin-GD2 paclitaxel nanoparticle complexes
US10780050B2 (en) 2014-10-06 2020-09-22 Mayo Foundation For Medical Education And Research Lyophilized compositions comprosing albumin-EGFR paclitaxel nanoparticle complexes
US10966923B2 (en) 2014-10-06 2021-04-06 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10610484B2 (en) 2014-10-06 2020-04-07 Mayo Foundation For Medical Education And Research Methods of using albumin-CD20 paclitaxel nanoparticle complex compositions for treating cancer
US10993911B2 (en) 2014-10-06 2021-05-04 Mayo Foundation For Medical Education And Research Carrier-antibody compositions and methods of making and using the same
US10596112B2 (en) 2014-10-06 2020-03-24 Mayo Foundation For Medical Education And Research Methods of using albumin-antibody nanoparticle complex compositions for treating cancer
US11241387B2 (en) 2015-08-18 2022-02-08 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
US11660339B2 (en) 2015-10-06 2023-05-30 Mayo Foundation For Medical Education And Research Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US10561726B2 (en) 2015-10-06 2020-02-18 Vavotar Life Sciences LLC Methods of treating cancer using compositions of antibodies and carrier proteins with antibody pretreatment
US11571469B2 (en) 2016-01-07 2023-02-07 Mayo Foundation For Medical Education And Research Methods of treating cancer with interferon wherein the cancer cells are HLA negative or have reduced HLA expression
US11351254B2 (en) 2016-02-12 2022-06-07 Mayo Foundation For Medical Education And Research Hematologic cancer treatments
US11878061B2 (en) 2016-03-21 2024-01-23 Mayo Foundation For Medical Education And Research Methods for improving the therapeutic index for a chemotherapeutic drug
US11305020B2 (en) 2016-03-21 2022-04-19 Mayo Foundation For Medical Education And Research Methods for reducing toxicity of a chemotherapeutic drug
US11655301B2 (en) 2016-04-06 2023-05-23 Mayo Foundation For Medical Education And Research Carrier-binding agent compositions and methods of making and using the same
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
US11548946B2 (en) 2016-09-01 2023-01-10 Mayo Foundation For Medical Education And Research Carrier-PD-L1 binding agent compositions for treating cancers
US11160876B2 (en) 2016-09-01 2021-11-02 Mayo Foundation For Medical Education And Research Methods and compositions for targeting t-cell cancers
US11590098B2 (en) 2016-09-06 2023-02-28 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11427637B2 (en) 2016-09-06 2022-08-30 Mayo Foundation For Medical Education And Research Methods of treating PD-L1 expressing cancer
US11311631B2 (en) 2016-09-06 2022-04-26 Mayo Foundation For Medical Education And Research Paclitaxel-albumin-binding agent compositions and methods for using and making the same
US11872205B2 (en) 2016-09-06 2024-01-16 Mayo Foundation For Medical Education And Research Methods of treating triple-negative breast cancer using compositions of antibodies and carrier proteins
US11531030B2 (en) 2017-04-21 2022-12-20 Mayo Foundation For Medical Education And Research Polypeptide-antibody complexes and uses thereof
US11971412B2 (en) 2022-11-08 2024-04-30 Mayo Foundation For Medical Education And Research Polypeptide-antibody complexes and uses thereof

Also Published As

Publication number Publication date
CN106573051B (en) 2021-07-13
EP3154586A4 (en) 2018-01-10
RU2017100445A (en) 2018-07-16
MX2016016369A (en) 2017-10-12
RU2708332C2 (en) 2019-12-06
JP6695286B2 (en) 2020-05-20
CA2950926A1 (en) 2015-12-17
MX2021002900A (en) 2021-06-08
KR20170085955A (en) 2017-07-25
KR102450563B1 (en) 2022-10-07
CN106573051A (en) 2017-04-19
WO2015191969A1 (en) 2015-12-17
EP3154586A1 (en) 2017-04-19
AU2015274408A1 (en) 2017-01-12
AU2021200489A1 (en) 2021-02-25
IL249454B (en) 2021-04-29
EP3154586B1 (en) 2020-05-27
JP2017517556A (en) 2017-06-29
CN113318239A (en) 2021-08-31
RU2017100445A3 (en) 2019-01-28
IL249454A0 (en) 2017-02-28
US20170216453A1 (en) 2017-08-03
BR112016029123A2 (en) 2017-08-22

Similar Documents

Publication Publication Date Title
US20170216453A1 (en) Treating Lymphomas
US11285221B2 (en) Treating myelomas
US20220265824A1 (en) Hematologic cancer treatments
JP2013521338A (en) Methods for treating small cell lung cancer
JP2018527377A (en) Combination of topoisomerase-I inhibitor and immunotherapy in the treatment of cancer
EP1874296A2 (en) Method of treating multiple myeloma using 17-aag of 17-ag of a prodrug of either

Legal Events

Date Code Title Description
AS Assignment

Owner name: MAYO FOUNDATION FOR MEDICAL EDUCATION AND RESEARCH

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MARKOVIC, SVETOMIR N;NEVALA, WENDY K;REEL/FRAME:038634/0861

Effective date: 20160502

STCB Information on status: application discontinuation

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