US20140017316A1 - Methods and compositions for treating recurrent cancer - Google Patents

Methods and compositions for treating recurrent cancer Download PDF

Info

Publication number
US20140017316A1
US20140017316A1 US13/781,482 US201313781482A US2014017316A1 US 20140017316 A1 US20140017316 A1 US 20140017316A1 US 201313781482 A US201313781482 A US 201313781482A US 2014017316 A1 US2014017316 A1 US 2014017316A1
Authority
US
United States
Prior art keywords
cancer
composition
individual
taxane
recurrent
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
US13/781,482
Inventor
Neil P. Desai
Patrick Soon-Shiong
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Abraxis Bioscience LLC
Original Assignee
Abraxis Bioscience LLC
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 Abraxis Bioscience LLC filed Critical Abraxis Bioscience LLC
Priority to US13/781,482 priority Critical patent/US20140017316A1/en
Publication of US20140017316A1 publication Critical patent/US20140017316A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/42Proteins; Polypeptides; Degradation products thereof; Derivatives thereof, e.g. albumin, gelatin or zein
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • 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
    • A61K47/48284
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being a protein, peptide or polyamino acid
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/643Albumins, e.g. HSA, BSA, ovalbumin or a Keyhole Limpet Hemocyanin [KHL]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/69Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
    • A61K47/6921Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere
    • A61K47/6927Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores
    • A61K47/6929Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit the form being a particulate, a powder, an adsorbate, a bead or a sphere the form being a solid microparticle having no hollow or gas-filled cores the form being a nanoparticle, e.g. an immuno-nanoparticle
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/48Preparations in capsules, e.g. of gelatin, of chocolate
    • A61K9/50Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
    • A61K9/51Nanocapsules; Nanoparticles
    • A61K9/5107Excipients; Inactive ingredients
    • A61K9/513Organic macromolecular compounds; Dendrimers
    • A61K9/5169Proteins, e.g. albumin, gelatin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/14Vasoprotectives; Antihaemorrhoidals; Drugs for varicose therapy; Capillary stabilisers
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/70Nanostructure
    • Y10S977/773Nanoparticle, i.e. structure having three dimensions of 100 nm or less
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10STECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10S977/00Nanotechnology
    • Y10S977/902Specified use of nanostructure
    • Y10S977/904Specified use of nanostructure for medical, immunological, body treatment, or diagnosis
    • Y10S977/906Drug delivery

Definitions

  • the present invention relates to methods and compositions for the treatment of recurrent cancer, particularly recurrent ovarian cancer, comprising the administration of compositions comprising nanoparticles comprising taxane and a carrier protein (such as albumin).
  • a carrier protein such as albumin
  • Ovarian cancer forms in tissues of an ovary (one of a pair of female reproductive glands in which the ova, or eggs, are formed). Most ovarian cancers are either ovarian epithelial carcinomas (cancer that begins in the cells on the surface of the ovary) or malignant germ cell tumors (cancer that begins in egg cells). According to the National Cancer Institute, ovarian cancer is the seventh most common cancer, with an estimated 20,180 new cases in 2006, but is the fourth most deadly, with an estimated 15,310 deaths in 2006.
  • a possible genetic contribution to ovarian cancer risk is indicated by the increased incidence of this cancer among women with a family history, and by the observation of rare families in which multiple family members are affected with ovarian cancer, in a pattern compatible with autosomal dominant inheritance of cancer susceptibility.
  • Formal studies of families have subsequently proven the existence of autosomal dominant predispositions to ovarian cancer and have led to the identification of several highly penetrant genes as the cause of inherited cancer risk in many cancer-prone families. Mutations in these genes are rare in the general population and are estimated to account for no more than 5% to 10% of ovarian cancer cases overall.
  • first-line chemotherapy fails to produce a remission in more than 70% of patients with ovarian cancer. Furthermore, approximately 40-50% of the women who do achieve a remission after first-line chemotherapy will experience a recurrence of cancer within 3 years. Patients with recurrent ovarian, peritoneal, or fallopian tube cancer generally have a poor outcome with current therapies. There is a need for effective treatment method for patients with recurrent ovarian cancers. Preferably, the treatments overcome the shortcomings of current drug and transplant treatments, such as hypersensitivity reactions due to the solvent/surfactant in which drugs are dissolved.
  • anti-proliferative agents are dissolved in a solvent/surfactant which produces hypersensitivity reactions.
  • Great efforts have been invested on the development of water soluble prodrugs and derivatives of anti-proliferative agents with higher hydrophilic groups to enhance water solubility and thus obviate the need for potentially toxic solvents/surfactants.
  • Another approach to address the problem associated with the poor water solubility of anti-proliferative agents is the development of various formulations such as nanoparticles, oil-in-water emulsions, and liposomes.
  • Abraxane® is a nanoparticle composition of paclitaxel and albumin. Nanoparticle compositions of substantially poorly water soluble drugs and uses thereof have been disclosed, for example, in U.S. Pat. Nos.
  • the present invention provides methods for the treatment of a recurrent cancer (such as a recurrent gynecological cancer) by administering a composition comprising nanoparticles comprising a taxane and a carrier protein (hereinafter referred to as “taxane nanoparticle composition”).
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • the recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum sensitive. In some embodiments, the recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum resistant. In some embodiments, the recurrent cancer is a recurrent lung cancer.
  • the recurrent cancer is a recurrent gynecological cancer.
  • the recurrent gynecological cancer is a recurrent ovarian cancer (such as a recurrent epithelial ovarian cancer).
  • the recurrent gynecological cancer is a recurrent peritoneal cancer.
  • the recurrent gynecological cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas).
  • Other recurrent gynecological cancers such as recurrent malignant mixed mullerian tumor and serous endo can also be treated.
  • the individual is a woman who is about 40 to about 85 years old, including for example about 60 to about 70 years old.
  • the individual has an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 (such as any of 0, 1, or 2) prior to the administration of the taxane nanoparticle composition.
  • ECOG Eastern Cooperative Oncology Group
  • the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 3, 6, or 9 months since the completion of prior chemotherapy.
  • the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy.
  • the prior chemotherapy has a different mechanism of action than that of the taxane.
  • the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition.
  • the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition.
  • the individual has not previously been treated with a taxane-based therapy.
  • the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 3, 6, or 9 months prior to the initiation of the methods described herein. In some embodiments, the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months prior to the initiation of the methods described herein. In some embodiments, the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the methods described herein (such as within 12, 9, 6, 5, 4, 3, 2, or 1 month prior to the initiation of the methods described herein). In some embodiments, the individual does not show a symptom of hypersensitivity throughout the treatment period with methods described herein. In some embodiments, the individual does not show a symptom of hypersensitivity upon completion of the treatment with methods described herein.
  • a prior cancer therapy such as chemotherapy
  • the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 12, 18, 24, 36
  • the individual when the method is directed to treatment of a recurrent ovarian, peritoneal, or fallopian tube cancer, the individual may be confirmed of having an ovarian, peritoneal, or fallopian tube cancer histologically or cytologically. In some embodiments, the individual is determined to have an ovarian, peritoneal, or fallopian tube cancer based on RECIST (Response Evaluation Criteria in Solid Tumors).
  • RECIST Response Evaluation Criteria in Solid Tumors
  • the individual has an elevated blood level of Cancer Antigen 125 (CA-125, for example a CA-125 level of more than about 40, 50, 60, 70, 80, or 90 units/ml, or about 2 ⁇ , 3 ⁇ , 4 ⁇ , or more of that of the upper limit of a normal CA-125 level).
  • CA-125 Cancer Antigen 125
  • the individual has an altered level of a marker that is indicative of an ovarian, peritoneal, or fallopian tube cancer.
  • the individual satisfies at least two of the criteria described above.
  • the individual has a measurable disease by RECIST and an elevated blood level of CA-125.
  • the individual is confirmed of having an ovarian cancer histologically or cytologically and has only been treated with platinum-based agent(s) prior to administration of the nanoparticle compositions described above.
  • the individual satisfies at least any of two, three, four, five, or more criteria described above. In some embodiments, the individual satisfies all criteria described above.
  • the recurrent cancer (such as recurrent ovarian cancer) is platinum-sensitive.
  • the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 3, 6, or 9 months since the completion of the platinum-based chemotherapy.
  • the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of the platinum-based chemotherapy.
  • Platinum-based chemotherapy includes, but is not limited to, treatment with carboplatin, cisplatin, and oxaliplatin.
  • the platinum-based chemotherapy is treatment with carboplatin.
  • the methods described herein comprise administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • the taxane nanoparticle composition is administered in conjunction with another chemotherapeutic agent (such as a platinum-based agent).
  • the taxane nanoparticle composition and the other chemotherapeutic agent can be administered sequentially, simultaneously, or concurrently.
  • the other chemotherapeutic agent (such as platinum-based agent) is administered in the same composition as the nanoparticles comprising taxane and carrier protein.
  • the other chemotherapeutic agent (such as platinum-based agent) can also be formulated into a nanoparticle composition as described herein.
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent.
  • Suitable platinum-based agents include, but are not limited to, carboplatin, cisplatin, and oxaliplatin.
  • the platinum-based agent is carboplatin.
  • the taxane nanoparticle composition and the platinum-based agent are administered simultaneously.
  • the taxane nanoparticle composition and the platinum-based agent are administered sequentially.
  • the taxane nanoparticle composition and the platinum-based agent is administered concurrently.
  • a method of treating a recurrent ovarian cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a method of treating a primary peritoneal cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent, wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent, wherein the paclitaxel nanoparticle composition and the platinum-based agent are administered concurrently.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering (for example intravenously or intraperitoneally) to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), wherein the amount of the paclitaxel in the composition is at least about 40 mg/m 2 (including for example about any of 50 mg/m 2 , 60 mg/m 2 , 70 mg/m 2 , 80 mg/m 2 , 90 mg/m 2 , 100 mg/m 2 , 150 mg/m 2 , or 200 mg/m 2 ); b) an effective amount of a platinum-based agent (such as the platinum-based agent at the amount of AUC3, AUC4, or AUC6), wherein the taxane nanoparticle composition and the platinum
  • the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and a platinum-based agent (such as carboplatin) AUC6 every four weeks in the same treatment cycle.
  • the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle.
  • the method comprises intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle.
  • the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • a method of treating recurrent ovarian cancer comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle.
  • a platinum-based agent such as carboplatin
  • AUC5 or AUC6
  • a method of treating primary peritoneal cancer comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle.
  • a platinum-based agent such as carboplatin
  • AUC5 or AUC6
  • the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • the taxane nanoparticle composition is not administered in conjunction with a platinum-based agent, that is, the taxane nanoparticle composition is either administered in a monotherapy dosing regime or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent.
  • a platinum-based agent is not administered to the individual during the time period in which the individual is receiving one or more doses of the taxane nanoparticle composition.
  • the individual is not treated with a platinum-based agent concurrently with the administration of the taxane nanoparticle composition.
  • the taxane nanoparticle composition can be administered alone, that is, the taxane nanoparticle is administered in a monotherapy dosing regime.
  • the amount of the taxane nanoparticle composition administered alone is sufficient to result in a complete response in the individual.
  • the amount of the taxane nanoparticle composition administered alone is sufficient to result in a partial response in the individual.
  • the amount of the taxane nanoparticle composition administered alone is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the taxane nanoparticle composition.
  • the amount of taxane nanoparticle composition administered alone is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the taxane nanoparticle composition.
  • the taxane nanoparticle compositions described herein comprise nanoparticles comprising taxane (such as paclitaxel) and a carrier protein.
  • the taxane is paclitaxel, docetaxel, ortataxel, or IDN5390.
  • the carrier protein is albumin, such as human serum albumin.
  • the weight ratio of the carrier protein to the taxane in the taxane nanoparticle composition is less than about 18:1 (including for example less than about any of 15:1, 12:1, 10:1, such as 9:1).
  • the nanoparticles comprise the taxane coated with a coating comprising the carrier protein (such as albumin).
  • the coating consists essentially of or consists of the carrier protein.
  • at least a portion of the carrier protein in the nanoparticle portion of the taxane nanoparticle composition is crosslinked (for example crosslinked by disulfide bonds).
  • the nanoparticles of the composition comprise at least 5% (including for example at least any of 10%, 15%, 20%, or 25%) of carrier protein that is cross-linked.
  • the average or mean diameter of the nanoparticles in the composition is no greater than about 200 nm. In some embodiments, the average or mean diameter of the particles is between about 20 nm to about 400 nm (such as about 40 nm to about 200 nm). In some embodiments, the nanoparticles are sterile-filterable. In some embodiments, the taxane in the nanoparticles is amorphous. In some embodiments, the nanoparticles are substantially free of polymeric core materials. In some embodiments, the nanoparticles comprise a core of taxane that is substantially free of polymeric materials (such as polymertic matrix). In some embodiments, the nanoparticles in the composition have a solid core.
  • the nanoparticles in the composition have a core that is not aqueous (i.e., other than aqueous core). In some embodiments, the nanoparticles of the composition are substantially free of lipids. In some embodiments, the nanoparticles of the composition are free of lipids.
  • the composition comprises more than about 50% (for example more than about any of 60%, 70%, 80%, 90%, or 95%) of the taxane in nanoparticle form.
  • the weight percentage of the taxane in the nanoparticle portion of the taxane nanoparticle composition is at least about any of 50%, 60%, 70%, 80%, 90%, or 95% of the total weight of the nanoparticle portion of the composition.
  • the composition is administered at least about any of once every three weeks, once every two weeks, once a week, twice a week, three times a week, four times a week, five times a week, six times a week, or daily.
  • Other exemplary dosing frequencies include, but are not limited to, weekly, two out of three weeks; weekly, three out of four weeks; and weekly, four out of five weeks.
  • the composition is administered (with or without breaks in administration cycles) for at least about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more month(s).
  • the composition is administered via any of intravenous, intraperitoneal, oral or inhalational routes.
  • the nanoparticle composition is administered with premedication. In some embodiments, the nanoparticle composition is administered without premedication.
  • the dose of the taxane in the nanoparticle composition will depend on the type of cancer to be treated, the severity and course of the cancer, the individual's clinical history, and the discretion of the attending physician.
  • Suitable dosages of the taxane in the taxane nanoparticle compositions include, but are not limited to, about any of 50 mg/m 2 , 60 mg/m 2 , 75 mg/m 2 , 80 mg/m 2 , 90 mg/m 2 , 100 mg/m 2 , 120 mg/m 2 , 125 mg/m 2 , 150 mg/m 2 , 160 mg/m 2 , 180 mg/m 2 , 200 mg/m 2 , 210 mg/m 2 , 220 mg/m 2 , 230 mg/m 2 , 240 mg/m 2 , 260 mg/m 2 , and 300 mg/m 2 .
  • Exemplary dosing schedules for the administration of the taxane nanoparticle composition include, but are not limited to, 260 mg/m 2 , every three weeks; 60-150 mg/m 2 , weekly, without break, and 60-150 mg/m 2 , weekly, three out of four weeks.
  • the taxane can be administered by following a metronomic dosing regime described herein.
  • the method comprises administering Abraxane® at 260 mg/m 2 by 30 minutes IV infusion every three weeks.
  • compositions, kits and unit dosage forms that are suitable for methods described herein.
  • a composition for use in the treatment of a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • compositions for use in decreasing one or more symptoms resulting from a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a composition for use in the treatment of a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • a platinum-based agent such as platinum-based agent
  • the composition comprises nanoparticles comprising a taxane (such as paclitaxel)
  • the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • compositions for use in decreasing one or more symptoms resulting from a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • FIG. 1 a shows overall survival among patients plotted in terms of months (x-axis) to proportion of survival (y-axis).
  • FIG. 1 b shows progression free survival among patients plotted in terms of months (x-axis) to proportion not progressed (y-axis).
  • FIG. 2 shows a comparison of plasma level of paclitaxel when Nab-paclitaxel is administered intraperitoneally or intravenously (log-log).
  • FIG. 3 shows a comparison of plasma level of paclitaxel when Nab-paclitaxel is administered intraperitoneally or intravenously (log-linear).
  • the present invention provides methods for the treatment of a recurrent cancer (such as recurrent gynecological cancer) comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a recurrent cancer such as recurrent gynecological cancer
  • a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • compositions comprising nanoparticles comprising a taxane and a carrier protein
  • a composition comprising nanoparticles comprising paclitaxel and albumin
  • a composition comprising nanoparticles albumin bound paclitaxel (“Nab-paclitaxel”)
  • platinum-based agent carboplatin
  • the overall response rate for the treatment was 64% and the clinical benefit rate was 77%.
  • a taxane nanoparticle composition is particularly suitable (either as a single agent or in combination with a platinum-based agent) for treating platinum-sensitive recurrent cancer, particularly recurrent gynecological cancer.
  • the present invention in one aspect provides a method of treating a platinum-sensitive recurrent cancer comprising administering to the individual an effective amount of a composition (such as Nab-paclitaxel or Abraxane®) comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a composition such as Nab-paclitaxel or Abraxane®
  • a carrier protein such as albumin
  • compositions includes and is applicable to compositions of the invention.
  • compositions includes and is applicable to compositions of the invention.
  • invention also provides pharmaceutical compositions comprising the components described herein.
  • Taxanes include, but are not limited to, compounds that are structurally similar to or are in the same general chemical class such as paclitaxel (i.e., taxol), docetaxel (i.e., taxotere), or ortataxel, and pharmaceutically acceptable salts, derivatives, or analogs of paclitaxel, docetaxel, and ortataxel. Taxanes are antimicrobial agents that inhibit cell replication by promoting the assembly and stabilization of microtubule from tubulin dimers
  • an individual as used herein refers to human.
  • beneficial or desired clinical results include, but are not limited to, one or more of the following: decreasing one or more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), delay or slowing the progression of the disease, ameliorating the disease state, decreasing the dose of one or more other medications required to treat the disease, increasing the quality of life, and/or prolonging survival (including overall survival and progression free survival.
  • the composition reduces the severity of one or more symptoms associated with cancer by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% compared to the corresponding symptom in the same subject prior to treatment or compared to the corresponding symptom in other subjects not receiving the composition.
  • treatment is a reduction of pathological consequence of cancer. The methods of the invention contemplate any one or more of these aspects of treatment.
  • “delaying” the development of cancer means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease.
  • a method that “delays” development of cancer is a method that reduces probability of disease development in a given time frame and/or reduces the extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects. Cancer development can be detectable using standard methods, such as routine physical exams or x-ray. Development may also refer to disease progression that may be initially undetectable and includes occurrence and onset.
  • “Adjuvant setting” refers to a clinical setting in which an individual has had a history of ovarian cancer, and generally (but not necessarily) been responsive to therapy, which includes, but is not limited to, surgery (e.g., surgical resection), radiotherapy, and chemotherapy. However, because of their history of the cancer, these individuals are considered at risk of development of the disease. Treatment or administration in the “adjuvant setting” refers to a subsequent mode of treatment. The degree of risk (i.e., when an individual in the adjuvant setting is considered as “high risk” or “low risk”) depends upon several factors, most usually the extent of disease when first treated.
  • “Neoadjuvant setting” refers to a clinical setting in which the method is be carried out before the primary/definitive therapy.
  • an “at risk” individual is an individual who is at risk of developing cancer.
  • An individual “at risk” may or may not have detectable disease, and may or may not have displayed detectable disease prior to the treatment methods described herein.
  • At risk denotes that an individual has one or more so-called risk factors, which are measurable parameters that correlate with development of cancer, which are described herein. An individual having one or more of these risk factors has a higher probability of developing cancer than an individual without these risk factor(s).
  • pharmaceutically active compound is meant a chemical compound that induces a desired effect, e.g., treating, stabilizing, preventing, and/or delaying cancer.
  • combination therapy is meant a first therapy that includes nanoparticles comprising nanoparticles comprising a taxane (e.g. paclitaxel) and a carrier protein in conjunction with a second therapy (e.g., surgery or a therapeutic agent) useful for treating, stabilizing, preventing, and/or delaying cancer.
  • Administration in “conjunction with” another compound includes administration in the same or different composition(s), either sequentially, simultaneously, or continuously.
  • the combination therapy optionally includes one or more pharmaceutically acceptable carriers or excipients, non-pharmaceutically active compounds, and/or inert substances.
  • the term “effective amount” refers to an amount of a drug effective to treat cancer in the patient.
  • the effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer.
  • the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic.
  • the effective amount may extend progression free survival (e.g.
  • an “effective amount” may be in one or more doses, i.e., a single dose or multiple doses may be required to achieve the desired treatment endpoint.
  • An effective amount may be considered in the context of administering one or more therapeutic agents, and a nanoparticle composition (e.g., a composition including a taxane (e.g., paclitaxel) and a carrier protein) may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable or beneficial result may be or is achieved.
  • a nanoparticle composition e.g., a composition including a taxane (e.g., paclitaxel) and a carrier protein
  • the amount of the composition is an amount sufficient to decrease the size of a tumor, decrease the number of cancer cells, or decrease the growth rate of a tumor by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% compared to the corresponding tumor size, number of cancer cells, or tumor growth rate in the same subject prior to treatment or compared to the corresponding activity in other subjects not receiving the treatment.
  • Standard methods can be used to measure the magnitude of this effect, such as in vitro assays with purified enzyme, cell-based assays, animal models, or human testing.
  • proteins refers to polypeptides or polymers of amino acids of any length (including full length or fragments), which may be linear or branched, comprise modified amino acids, and/or be interrupted by non-amino acids.
  • the term also encompasses an amino acid polymer that has been modified naturally or by intervention, including, for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification. Also included within this term are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art.
  • the proteins described herein may be naturally-occurring, i.e., obtained or derived from a natural source (e.g., blood) or synthesized (e.g., chemically synthesized or by synthesized by recombinant DNA techniques). Exemplary carrier proteins are described herein.
  • antimicrobial agent refers to an agent that is capable of inhibiting (e.g., delaying, reducing, slowing, and/or preventing) the growth of one or more microorganisms.
  • Significant microbial growth can be measured or indicated by a number of ways known in the art, such as one or more of the following: (i) microbial growth in a composition that is enough to cause one or more adverse effects to an individual when the composition is administered to the individual; (ii) more than about 10-fold increase in microbial growth over a certain period of time (for example over a 24 hour period) upon extrinsic contamination (e.g., exposure to 10-10 3 colony forming units at a temperature in the range of 20 to 25° C.).
  • Other indicia of significant microbial growth are described in U.S. Patent Application Publication No. US20070117744 (U.S. Ser. No. 11/514,030, filed Aug. 30, 2006), which is hereby incorporated by reference in its entirety.
  • “Sugar” as used herein includes, but is not limited to, monosaccharides, disaccharides, polysaccharides, and derivatives or modifications thereof. Suitable sugars for compositions described herein include, for example, mannitol, sucrose, fructose, lactose, maltose, and trehalose.
  • pharmaceutically acceptable or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained.
  • Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • “Survival” refers to the patient remaining alive, and includes overall survival as well as progression free survival.
  • “Overall survival” refers to the patient remaining alive for a defined period of time, such as 1 year, 5 years, etc. from the time of diagnosis or treatment.
  • progression free survival refers to the patient remaining alive, without the cancer progressing or getting worse.
  • prolonging survival is meant increasing overall or progression free survival in a treated patient relative to an untreated patient (e.g. relative to a patient not treated with a taxane nanoparticle composition).
  • reference to “not” a value or parameter generally means and describes “other than” a value or parameter. For example, if a taxane is not administered, it means an agent other than a taxane is administered.
  • references to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.’
  • the present invention provides methods for the treatment of a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) by administering a composition comprising nanoparticles comprising a taxane and a carrier protein.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a composition comprising nanoparticles comprising a taxane and a carrier protein.
  • the recurrent cancer is platinum sensitive.
  • the recurrent cancer is platinum resistant.
  • the recurrent cancer is any of recurrent ovarian cancer, recurrent peritoneal cancer, recurrent fallopian tube cancer, recurrent malignant mixed mullerian tumor, and serous endo.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein.
  • a method of treating a platinum-sensitive recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein.
  • the recurrent cancer is a recurrent ovarian cancer (such as a recurrent epithelial ovarian cancer).
  • the recurrent cancer is a recurrent peritoneal cancer. In some embodiments, the recurrent cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas).
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 3, 6, 9 months since the completion of prior chemotherapy.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin)
  • the prior chemotherapy has a different mechanism of action than that of the taxane-based therapy.
  • the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 3, 6, 9 months since the completion of the platinum-based chemotherapy.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 38 months since the completion of the platinum-based chemotherapy.
  • the method further comprises administering to the individual an effective amount of a platinum-based agent (such as carboplatin).
  • the composition comprising nanoparticles comprising a taxane and a carrier protein is not administered in conjunction with a platinum agent, that is, the composition comprising nanoparticles comprising a taxane and a carrier protein is either administered alone or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent.
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 3, 6, 9 months prior to the initiation of the treatment method with the taxane nanoparticle composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 12, 18, 24, 36, or 48 months prior to the initiation of the treatment method with the taxane nanoparticle composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin)
  • the prior chemotherapy has a different mechanism of action than that of the taxane-based therapy.
  • the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 3, 6, 9 months prior to the initiation of the treatment method with the taxane nanoparticle composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 38 months prior to the initiation of the treatment method with the taxane nanoparticle composition.
  • the method further comprises administering to the individual an effective amount of a platinum-based agent.
  • the composition comprising nanoparticles comprising a taxane and a carrier protein is not administered in conjunction with a platinum agent, that is, the composition comprising nanoparticles comprising a taxane and a carrier protein is either administered alone or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent.
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the treatment method with the taxane nanoparticle composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show a symptom of hypersensitivity throughout the treatment period.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show symptoms of hypersensitivity upon completion of the treatment.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • the composition comprising nanoparticles comprising a taxane and a carrier protein is administered alone.
  • a method of treating a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • administering comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is effective to result in a complete response in the individual.
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is effective to result in a partial response in the individual.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the composition.
  • a recurrent cancer such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising paclitaxel and albumin. In some embodiments, the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein average or mean diameter of the nanoparticles in the composition is less than about 200 nm. In some embodiments, the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the nanoparticles comprise taxane coated with a coating comprising the carrier protein.
  • a taxane such as paclitaxel
  • albumin carrier protein
  • the method comprises administering to the individual an effective amount of a Nab-paclitaxel (such as Abraxane®).
  • a Nab-paclitaxel such as Abraxane®
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer comprising administering to the individual about 50 mg/m 2 to about 300 mg/m 2 (including for example about 260 mg/m 2 ) Nab-paclitaxel or Abraxane®.
  • the Nab-paclitaxel or Abraxane® is administered every three weeks.
  • the Nab-paclitaxel or Abraxane® is administered intravenously (such as by i.v. infusion over a period of about 30 minutes or less).
  • the Nab-paclitaxel or Abraxane® is administered intraperitoneally.
  • the methods of the present invention are useful for any one or more of the following (and thus in various embodiments can achieve and/or include any one or more of the following): 1) decreasing one or more symptoms resulting from recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 2) increasing overall response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 3) increasing partial response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 4) increasing complete response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 5) delaying disease progression of an individual with a recurrent cancer (such as recurrent gy
  • a method of decreasing one or more symptoms resulting from a recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a recurrent cancer such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer
  • a method of decreasing one more symptoms resulting from a recurrent cancer comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • a method of increasing response rate of recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a method of increasing response rate of recurrent cancer comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • a method of delaying disease progression of an individual with recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a method of delaying disease progression of an individual with recurrent cancer comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • a method of prolonging survival of an individual having recurrent cancer comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • a method of prolonging survival of an individual having recurrent cancer comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein one or more symptoms resulting from the recurrent cancer is decreased.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a partial response to treatment upon completion of less than about any of one, two, three, four, five, six, seven, or eight treatment cycles.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a complete response to treatment upon completion of less than about any of one, two, three, four, five, six, seven, or eight treatment cycles.
  • the treatment cycle is four weeks. In some embodiments, the treatment cycle is three weeks.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual is disease free for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual does not show a symptom resulting from the recurrent cancer for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a reduced CA-125 level (such as a level below about any of 70%, 60%, 50%, 40%, 30%, 20%, 10% of the level prior to the treatment) upon completion of the treatment.
  • the individual has a reduced CA-125 level for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment.
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering (for example intravenously or intraperitoneally) to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the taxane per administration is at least about 50 mg/m 2 (such as at least about any of 70 mg/m 2 , 100 m g/m 2 , 150 mg/m 2 , 200 mg/m 2 , or 260 mg/m 2 ), and wherein one or more symptoms resulting from the recurrent cancer is decreased.
  • a taxane such as paclitaxel
  • carrier protein such as albumin
  • a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual comprising administering (for example intravenously or intraperitoneally) to the individual an effective amount of Abraxane®, wherein the amount of paclitaxel per administration is at least about 50 mg/m 2 (such as at least about any of 70 mg/m 2 , 100 mg/m 2 , 150 mg/m 2 , 200 mg/m 2 , or 260 mg/m 2 ), and wherein one or more symptoms resulting from the recurrent cancer is decreased.
  • the composition is administered once every three weeks.
  • the composition is administered weekly.
  • the composition is administered three out of four weeks.
  • the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the treatment. In some embodiments, the individual does not show a symptom of hypersensitivity throughout the treatment period. In some embodiments, the individual does not show a symptom of hypersensitivity upon completion of the treatment.
  • a symptom of hypersensitivity such as neuropathy
  • the taxane nanoparticle composition is administered in conjunction with another chemotherapeutic agent (such as a platinum-based agent).
  • another chemotherapeutic agent such as a platinum-based agent
  • the taxane nanoparticle composition and the other chemotherapeutic agent are administered simultaneously.
  • the term “simultaneous administration,” as used herein, means that the taxane nanoparticle composition and the other chemotherapeutic agent are administered with a time separation of no more than about 15 minute(s), such as no more than about any of 10, 5, or 1 minutes.
  • the taxane in the nanoparticles and the other chemotherapeutic agent may be contained in the same composition (e.g., a composition comprising both the nanoparticles and the other chemotherapeutic agent) or in separate compositions (e.g., the nanoparticles are contained in one composition and the other chemotherapeutic agent is contained in another composition).
  • the taxane and the other chemotherapeutic agent may be present in a single composition containing at least two different nanoparticles, wherein some of the nanoparticles in the composition comprise the taxane and a carrier protein, and some of the other nanoparticles in the composition comprise the other chemotherapeutic agent and a carrier protein.
  • only the taxane is contained in nanoparticles.
  • simultaneous administration of the drug in the nanoparticle composition and the other chemotherapeutic agent can be combined with supplemental doses of the taxane and/or the other chemotherapeutic agent.
  • the nanoparticle composition and the other chemotherapeutic agent are administered sequentially.
  • the term “sequential administration” as used herein means that the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered with a time separation, of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60 or more minutes. Either the nanoparticle composition or the other chemotherapeutic agent may be administered first.
  • the nanoparticle composition and the other chemotherapeutic agent are contained in separate compositions, which may be contained in the same or different packages.
  • the administration of the nanoparticle composition and the other chemotherapeutic agent are concurrent, i.e., the administration period of the nanoparticle composition and that of the other chemotherapeutic agent overlap with each other.
  • the nanoparticle composition and the other chemotherapeutic agent are administered in the same treatment cycle(s).
  • the dosing frequency of the drug-containing nanoparticle composition and the other chemotherapeutic agent may be adjusted over the course of the treatment, based on the judgment of the administering physician.
  • the drug-containing nanoparticle composition and the other chemotherapeutic agent can be administered at different dosing frequency or intervals.
  • the drug-containing nanoparticle composition can be administered weekly, while a chemotherapeutic agent can be administered more or less frequently.
  • sustained continuous release formulation of the drug-containing nanoparticle and/or chemotherapeutic agent may be used.
  • Various formulations and devices for achieving sustained release are known in the art.
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent.
  • Suitable platinum-based agents include, but are not limited to, carboplatin, cisplatin, and oxaliplatin.
  • the platinum-based agent is carboplatin.
  • the taxane nanoparticle composition and the platinum-based agent are administered simultaneously.
  • the taxane nanoparticle composition and the platinum-based agent are administered sequentially.
  • the taxane nanoparticle composition and the platinum-based agent is administered concurrently.
  • a method of treating a recurrent ovarian cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a method of treating a primary peritoneal cancer in an individual comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent, wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent, wherein the paclitaxel nanoparticle composition and the platinum-based agent are administered concurrently.
  • a recurrent cancer such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer
  • a method of treating a recurrent cancer comprising administering (for example intravenously or intraperitoneally) to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), wherein the amount of the paclitaxel in the composition is at least about 40 mg/m 2 (including for example about any of 50 mg/m 2 , 60 mg/m 2 , 70 mg/m 2 , 80 mg/m 2 , 90 mg/m 2 , 100 mg/m 2 , 150 mg/m 2 , or 200 mg/m 2 ); b) an effective amount of a platinum-based agent (such as the platinum-based agent at the amount of AUC3, AUC4, or AUC6), wherein the taxane nanoparticle composition and the platinum
  • the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and a platinum-based agent (such as carboplatin) AUC6 every four weeks in the same treatment cycle.
  • the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle.
  • the method comprises intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle.
  • the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • a method of treating recurrent ovarian cancer comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle.
  • a platinum-based agent such as carboplatin
  • AUC5 or AUC6
  • a method of treating primary peritoneal cancer comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m 2 , three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle.
  • a platinum-based agent such as carboplatin
  • AUC5 or AUC6
  • the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • the present invention provides a method of treating a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer) in an individual.
  • a “recurrent” cancer is one which has regrown, either at the initial site or at a distant site, after a response to initial therapy.
  • the length of time between the completion of initial therapy and the development of recurrent disease is longer than about 3 month, including for example longer than about any of 4, 5, 6, 7, 8, 9, 10, or 11 months.
  • the length of time between the completion of initial therapy and the development of recurrent disease is longer than about 12 month, including for example, longer than about any of 14, 16, 18, 20, 22, 24, 36, or 48 months.
  • the recurrent cancer is a recurrent gynecological cancer.
  • “Gynecologic cancer” used herein refers to a cancer originating in the female reproductive organs. It includes cancers of the cervix, fallopian tubes, ovaries, uterus, vagina and vulva.
  • the recurrent cancer is a recurrent ovarian cancer.
  • the cancer is ovarian epithelial cancer.
  • Exemplary ovarian epithelial cancer histological classifications include: serous cystomas (e.g., serous benign cystadenomas, serous cystadenomas with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or serous cystadenocarcinomas), mucinous cystomas (e.g., mucinous benign cystadenomas, mucinous cystadenomas with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or mucinous cystadenocarcinomas), endometrioid tumors (e.g., endometrioid benign cysts, endometrioid tumors with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or endometrioid adenocarcinomas), clear cell (mesonephroid) tumors (e.g., begin clear cell tumors, clear cell
  • the recurrent cancer is a recurrent ovarian germ cell tumor.
  • Exemplary histologic subtypes include dysgerminomas or other germ cell tumors (e.g., endodermal sinus tumors such as hepatoid or intestinal tumors, embryonal carcinomas, olyembryomas, choriocarcinomas, teratomas, or mixed form tumors).
  • Exemplary teratomas are immature teratomas, mature teratomas, solid teratomas, and cystic teratomas (e.g., dermoid cysts such as mature cystic teratomas, and dermoid cysts with malignant transformation).
  • teratomas are monodermal and highly specialized, such as struma ovarii, carcinoid, struma ovarii and carcinoid, or others (e.g., malignant neuroectodermal and ependymomas).
  • the recurrent cancer is a recurrent peritoneal cancer.
  • the recurrent cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas).
  • Other recurrent cancers such as recurrent malignant mixed mullerian tumor and serous endo can also be treated.
  • Recurrence of ovarian, peritoneal, or fallopian tube cancer can be determined, for example, based on bimanual pelvic examinations, serial measurements of CA-125 or other tumor markers, one or more imaging studies reassessments, and second-look laparotomy. Imaging studies such as CT, PET, or a CT/PET combination can also be used. In some embodiments, recurrence of ovarian, peritoneal, or fallopian tube cancer can be determined based on Response Evaluation Criteria in Solid Tumors (RECIST).
  • RECIST Response Evaluation Criteria in Solid Tumors
  • “Individual” used herein refers to human.
  • the individual is a woman who is about 40 to about 85 years old, including for example about 60 to about 70 years old.
  • the individual has an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 (such as any of 0, 1, or 2) prior to the administration of the taxane nanoparticle composition.
  • ECOG Eastern Cooperative Oncology Group
  • the individual has received a prior chemotherapy and has a treatment free interval for more than about any of 3, 6, or 9 months since the completion of prior chemotherapy.
  • the individual has received a prior chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy.
  • the prior chemotherapy has a different mechanism of action than that of the taxane.
  • the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition.
  • the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition.
  • the individual has not previously been treated with a taxane-based therapy.
  • the individual when the method is directed to treatment of a recurrent ovarian, peritoneal, or fallopian tube cancer, the individual is confirmed of having an ovarian, peritoneal, or fallopian tube cancer histologically or cytologically. In some embodiments, the individual is determined to have an ovarian, peritoneal, or fallopian tube cancer based on RECIST. In some embodiments, the individual has an elevated blood level of Cancer Antigen 125 (CA-125, for example a CA-125 level of more than about 40, 50, 60, 70, 80, or 90 units/ml, or about 2 ⁇ , 3 ⁇ , 4 ⁇ , or more of that of the upper limit of a normal CA-125 level). In some embodiments, the individual has an altered expression level of another marker that is indicative of a recurrent ovarian, peritoneal, and fallopian tube cancer.
  • CA-125 Cancer Antigen 125
  • the individual has an altered expression level of another marker that is indicative of a recurrent ovarian, peri
  • the individual being treated may have one or more risk factors associated with a higher probability of developing ovarian, peritoneal, and fallopian tube cancer.
  • risk factors include, but are not limited to, age, sex, race, diet, history of previous disease, presence of precursor disease, genetic (i.e., hereditary) considerations, and environmental exposure.
  • the individual may be a human who is genetically or otherwise predisposed to developing ovarian, peritoneal, and fallopian tube cancer, particularly recurrent ovarian, peritoneal, and fallopian tube cancer.
  • Individuals at risk for ovarian, peritoneal, and fallopian tube cancer include, for example, those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers.
  • the individual may be a human who has a gene, genetic mutation, or polymorphism associated with ovarian cancer (e.g., BRCA1 or BRCA2) or has one or more extra copies of a gene associated with ovarian cancer (e.g., one or more extra copies of the HER2 gene).
  • the individual is HER2 positive.
  • the individual is HER2 negative.
  • the ovarian cancer is associated with basal cell nevus (Gorlin) syndrome, multiple endocrine neoplasia type 1 (MEN1), or hereditary nonpolyposis colon cancer (HNPCC).
  • the individual satisfies at least two of the criteria described above.
  • the individual has a measurable disease by RECIST and an elevated blood level of CA-125.
  • the individual is confirmed of having an ovarian cancer histologically or cytologically and has only been treated with platinum-based agent(s) prior to administration of the nanoparticle compositions described above.
  • the individual satisfies at least any of two, three, four, five, or more criteria described above. In some embodiments, the individual satisfies all criteria described above.
  • the recurrent cancer is platinum sensitive.
  • the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 3, 6, or 9 months since the completion of the platinum-based chemotherapy.
  • the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of the platinum-based chemotherapy.
  • the recurrent cancer is a platinum-sensitive ovarian cancer.
  • the recurrent cancer is a platinum-sensitive peritoneal cancer.
  • the recurrent cancer is a platinum-sensitive fallopian tube cancer.
  • the recurrent cancer (such as a recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum resistant.
  • platinum-resistant cancer is meant that the individual with cancer has progressed while receiving platinum-based chemotherapy (i.e. the patient is “platinum refractory”).
  • platinum-based chemotherapy is intended to mean therapy with one or more platinum-based chemotherapeutic agents, optionally in combination with one or more other chemotherapeutic agents.
  • a “platinum-based chemotherapeutic agent” comprises an organic compound which contains platinum as an integral part of the molecule. These agents are believed to inhibit cell growth by forming reactive platinum complexes which form intrastrand and interstrand cross-linking of DNA molecules and inhibit DNA synthesis. Examples of platinum-based chemotherapeutic agents include carboplatin, cisplatin, and oxaliplatinum.
  • the platinum-based chemotherapy comprises treatment with only platinum-based agents.
  • the platinum-based chemotherapy comprises treatment with carboplatin.
  • the amount of the inventive composition administered to an individual may vary with the particular composition, the method of administration, and the particular type of recurrent cancer being treated.
  • the amount should be sufficient to produce a desirable beneficial effect.
  • the amount of the composition is effective to result in an objective response (such as a partial response or a complete response).
  • the amount of the taxane nanoparticle composition is sufficient to result in a complete response in the individual.
  • the amount of the taxane nanoparticle composition is sufficient to result in a partial response in the individual.
  • the amount of the taxane nanoparticle composition administered is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the taxane nanoparticle composition.
  • Responses of an individual to the treatment of the methods described herein can be determined, for example, based on RECIST or CA-125 level. For example, when CA-125 is used, a complete response can be defined as a return to a normal range value of at least 28 days from the pretreatment value. A particle response can be defined as a sustained over 50% reduction from the pretreatment value.
  • the amount of the composition is sufficient to prolong progress-free survival of the individual (for example as measured by RECIST or CA-125 changes). In some embodiments, the amount of the composition is sufficient to prolong overall survival of the individual. In some embodiments, the amount of the composition (for example when administered along) is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the taxane nanoparticle composition.
  • the amount of the taxane (e.g., paclitaxel) in the composition is below the level that induces a toxicological effect (i.e., an effect above a clinically acceptable level of toxicity) or is at a level where a potential side effect can be controlled or tolerated when the composition is administered to the individual.
  • the amount of the composition is close to a maximum tolerated dose (MTD) of the composition following the same dosing regime.
  • the amount of the composition is more than about any of 80%, 90%, 95%, or 98% of the MTD.
  • the amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 0.5 to about 5 mg, about 5 to about 10 mg, about 10 to about 15 mg, about 15 to about 20 mg, about 20 to about 25 mg, about 20 to about 50 mg, about 25 to about 50 mg, about 50 to about 75 mg, about 50 to about 100 mg, about 75 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg.
  • a taxane e.g., paclitaxel
  • the amount of a taxane (e.g., paclitaxel) or derivative thereof in the effective amount of the composition (e.g., a unit dosage form) is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg.
  • the concentration of the taxane (e.g., paclitaxel) in the composition is dilute (about 0.1 mg/ml) or concentrated (about 100 mg/ml), including for example any of about 0.1 to about 50 mg/ml, about 0.1 to about 20 mg/ml, about 1 to about 10 mg/ml, about 2 mg/ml to about 8 mg/ml, about 4 to about 6 mg/ml, about 5 mg/ml.
  • the concentration of the taxane is at least about any of 0.5 mg/ml, 1.3 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, or 50 mg/ml.
  • the taxane e.g., paclitaxel
  • Exemplary effective amounts of a taxane (e.g., paclitaxel) in the nanoparticle composition include, but are not limited to, about any of 25 mg/m 2 , 30 mg/m 2 , 50 mg/m 2 , 60 mg/m 2 , 75 mg/m 2 , 80 mg/m 2 , 90 mg/m 2 , 100 mg/m 2 , 120 mg/m 2 , 125 mg/m 2 , 150 mg/m 2 , 160 mg/m 2 , 175 mg/m 2 , 180 mg/m 2 , 200 mg/m 2 , 210 mg/m 2 , 220 mg/m 2 , 250 mg/m 2 , 260 mg/m 2 , 300 mg/m 2 , 350 mg/m 2 , 400 mg/m 2 , 500 mg/m 2 , 540 mg/m 2 , 750 mg/m 2 , 1000 mg/m 2 , or 1080 mg/m 2 of a taxane (e.g., paclitaxel).
  • the composition includes less than about any of 350 mg/m 2 , 300 mg/m 2 , 250 mg/m 2 , 200 mg/m 2 , 150 mg/m 2 , 120 mg/m 2 , 100 mg/m 2 , 90 mg/m 2 , 50 mg/m 2 , or 30 mg/m 2 of a taxane (e.g., paclitaxel).
  • a taxane e.g., paclitaxel
  • the amount of the taxane (e.g., paclitaxel) per administration is less than about any of 25 mg/m 2 , 22 mg/m 2 , 20 mg/m 2 , 18 mg/m 2 , 15 mg/m 2 , 14 mg/m 2 , 13 mg/m 2 , 12 mg/m 2 , 11 mg/m 2 , 10 mg/m 2 , 9 mg/m 2 , 8 mg/m 2 , 7 mg/m 2 , 6 mg/m 2 , 5 mg/m 2 , 4 mg/m 2 , 3 mg/m 2 , 2 mg/m 2 , or 1 mg/m 2 .
  • the taxane e.g., paclitaxel
  • the effective amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 1 to about 5 mg/m 2 , about 5 to about 10 mg/m 2 , about 10 to about 25 mg/m 2 , about 25 to about 50 mg/m 2 , about 50 to about 75 mg/m 2 , about 75 to about 100 mg/m 2 , about 100 to about 125 mg/m 2 , about 125 to about 150 mg/m 2 , about 150 to about 175 mg/m 2 , about 175 to about 200 mg/m 2 , about 200 to about 225 mg/m 2 , about 225 to about 250 mg/m 2 , about 250 to about 300 mg/m 2 , about 300 to about 350 mg/m 2 , or about 350 to about 400 mg/m 2 .
  • a taxane e.g., paclitaxel
  • the effective amount of a taxane (e.g., paclitaxel) in the composition is about 5 to about 300 mg/m 2 , such as about 100 to about 150 mg/m 2 , about 120 mg/m 2 , about 130 mg/m 2 , or about 140 mg/m 2 .
  • the effective amount of a taxane (e.g., paclitaxel) in the composition includes at least about any of 1 mg/kg, 2.5 mg/kg, 3.5 mg/kg, 5 mg/kg, 6.5 mg/kg, 7.5 mg/kg, 10 mg/kg, 15 mg/kg, or 20 mg/kg.
  • a taxane e.g., paclitaxel
  • the effective amount of a taxane (e.g., paclitaxel) in the composition includes less than about any of 350 mg/kg, 300 mg/kg, 250 mg/kg, 200 mg/kg, 150 mg/kg, 100 mg/kg, 50 mg/kg, 25 mg/kg, 20 mg/kg, 10 mg/kg, 7.5 mg/kg, 6.5 mg/kg, 5 mg/kg, 3.5 mg/kg, 2.5 mg/kg, or 1 mg/kg of a taxane (e.g., paclitaxel).
  • Exemplary dosing frequencies include, but are not limited to, weekly without break; weekly, three out of four weeks; once every three weeks; once every two weeks; weekly, two out of three weeks.
  • the composition is administered about once every 2 weeks, once every 3 weeks, once every 4 weeks, once every 6 weeks, or once every 8 weeks.
  • the composition is administered at least about any of 1 ⁇ , 2 ⁇ , 3 ⁇ , 4 ⁇ , 5 ⁇ , 6 ⁇ , or 7 ⁇ (i.e., daily) a week.
  • the intervals between each administration are less than about any of 6 months, 3 months, 1 month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some embodiments, the intervals between each administration are more than about any of 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, or 12 months. In some embodiments, there is no break in the dosing schedule. In some embodiments, the interval between each administration is no more than about a week.
  • the administration of the composition can be extended over an extended period of time, such as from about a month up to about seven years.
  • the composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, 36, 48, 60, 72, or 84 months.
  • the taxane (e.g., paclitaxel) or derivative thereof is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 0.25 mg/m 2 to about 75 mg/m 2 , such as about 0.25 mg/m 2 to about 25 mg/m 2 or about 25 mg/m 2 to about 50 mg/m 2 .
  • the dosage of a taxane (e.g., paclitaxel) in a nanoparticle composition can be in the range of 5-400 mg/m 2 when given on a 3 week schedule, or 5-250 mg/m 2 when given on a weekly schedule.
  • the amount of a taxane (e.g., paclitaxel) is about 60 to about 300 mg/m 2 (e.g., about 260 mg/m 2 ).
  • exemplary dosing schedules for the administration of the nanoparticle composition include, but are not limited to, 100 mg/m 2 , weekly, without break; 75 mg/m 2 weekly, 3 out of four weeks; 100 mg/m 2 , weekly, 3 out of 4 weeks; 125 mg/m 2 , weekly, 3 out of 4 weeks; 125 mg/m 2 , weekly, 2 out of 3 weeks; 130 mg/m 2 , weekly, without break; 175 mg/m 2 , once every 2 weeks; 260 mg/m 2 , once every 2 weeks; 260 mg/m 2 , once every 3 weeks; 180-300 mg/m 2 , every three weeks; 60-175 mg/m 2 , weekly, without break; 20-150 mg/m 2 twice a week; and 150-250 mg/m 2 twice a week.
  • the dosing frequency of the composition may be adjusted over the course of the treatment based on the judgment of the administering physician.
  • the individual is treated for at least about any of one, two, three, four, five, six, seven, eight, nine, or ten treatment cycles.
  • compositions described herein allow infusion of the composition to an individual over an infusion time that is shorter than about 24 hours.
  • the composition is administered over an infusion period of less than about any of 24 hours, 12 hours, 8 hours, 5 hours, 3 hours, 2 hours, 1 hour, 30 minutes, 20 minutes, or 10 minutes.
  • the composition is administered over an infusion period of about 30 minutes.
  • the invention provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin).
  • a taxane e.g., paclitaxel
  • a carrier protein e.g., albumin such as human serum albumin
  • the invention also provides a method of treating cancer in an individual by intravenous, intra-arterial, intramuscular, subcutaneous, inhalation, oral, intraperitoneal, nasally, or intra-tracheal administering to the individual (e.g., a human) an effective amount of a composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin).
  • the route of administration is intraperitoneal.
  • the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous.
  • about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 to about 500 mg, of the taxane (e.g., paclitaxel) or derivative thereof is administered per dose.
  • the taxane (e.g., paclitaxel) or derivative thereof is the only pharmaceutically active agent for the treatment of cancer that is contained in the composition.
  • the method comprises intraperitoneally administering composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin) at the dose of any of about 60 mg/m 2 , 80 mg/m 2 , 100 mg/m 2 , 125 mg/m 2 , and 150 mg/m 2 .
  • a taxane e.g., paclitaxel
  • carrier protein e.g., albumin such as human serum albumin
  • compositions described herein can be administered to an individual (such as human) via various routes, including, for example, intravenous, intra-arterial, intraperitoneal, intrapulmonary, oral, inhalation, intravesicular, intramuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, transmucosal, and transdermal.
  • sustained continuous release formulation of the composition may be used.
  • nanoparticles (such as albumin nanoparticles) of the inventive compounds can be administered by any acceptable route including, but not limited to, orally, intramuscularly, transdermally, intravenously, through an inhaler or other air borne delivery systems and the like.
  • paclitaxel-containing nanoparticles composition may be administered with a second therapeutic compound and/or a second therapy.
  • the dosing frequency of the paclitaxel-containing nanoparticles composition and the second compound may be adjusted over the course of the treatment based on the judgment of the administering physician.
  • the first and second therapies are administered simultaneously, sequentially, or concurrently.
  • the taxane (e.g., paclitaxel)-containing nanoparticles composition and the second compound can be administered at different dosing frequency or intervals.
  • the taxane (e.g., paclitaxel)-containing nanoparticle composition can be administered weekly, while a second compound can be administered more or less frequently.
  • sustained continuous release formulation of the taxane (e.g., paclitaxel)-containing nanoparticle and/or second compound may be used.
  • Various formulations and devices for achieving sustained release are known in the art. A combination of the administration configurations described herein can be used.
  • the nanoparticle composition and the other chemotherapeutic agent can be administered using the same route of administration or different routes of administration.
  • the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered at a predetermined ratio.
  • the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is about 1 to 1.
  • the weight ratio may be between about 0.001 to about 1 and about 1000 to about 1, or between about 0.01 to about 1 and 100 to about 1.
  • the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is less than about any of 100:1, 50:1, 30:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, and 1:1 In some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is more than about any of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 30:1, 50:1, 100:1. Other ratios are contemplated.
  • the doses required for the taxane and/or the other chemotherapeutic agent may (but not necessarily) be lower than what is normally required when each agent is administered alone.
  • a subtherapeutic amount of the drug in the nanoparticle composition and/or the other chemotherapeutic agent are administered.
  • “Subtherapeutic amount” or “subtherapeutic level” refer to an amount that is less than the therapeutic amount, that is, less than the amount normally used when the drug in the nanoparticle composition and/or the other chemotherapeutic agent are administered alone. The reduction may be reflected in terms of the amount administered at a given administration and/or the amount administered over a given period of time (reduced frequency).
  • enough chemotherapeutic agent is administered so as to allow reduction of the normal dose of the drug in the nanoparticle composition required to effect the same degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more.
  • enough drug in the nanoparticle composition is administered so as to allow reduction of the normal dose of the other chemotherapeutic agent required to effect the same degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more.
  • the dose of both the taxane in the nanoparticle composition and the other chemotherapeutic agent are reduced as compared to the corresponding normal dose of each when administered alone.
  • both the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered at a subtherapeutic, i.e., reduced, level.
  • the dose of the nanoparticle composition and/or the other chemotherapeutic agent is substantially less than the established maximum toxic dose (MTD).
  • the dose of the nanoparticle composition and/or the other chemotherapeutic agent is less than about 50%, 40%, 30%, 20%, or 10% of the MTD.
  • the present invention also provides metronomic therapy regimes for any of the methods of treatment and methods of administration described herein.
  • Exemplary metronomic therapy regimes and embodiments for the use of metronomic therapy regimes are discussed below and disclosed in U.S. Ser. No. 11/359,286, filed Feb. 21, 2006, published as U.S. Pub. No. 2006/0263434 (such as those described in paragraphs [0138] to [0157]), which is hereby incorporated by reference in its entirety.
  • the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the nanoparticle composition is administered over a period of at least two months, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 1% to about 20% of its maximum tolerated dose following a traditional dosing regime.
  • the dose of a taxane (e.g., paclitaxel) per administration is less than about any of 25%, 24%, 23%, 22%, 20%, 18%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of the maximum tolerated dose.
  • any nanoparticle composition is administered at least about any of 1 ⁇ , 2 ⁇ , 3 ⁇ , 4 ⁇ , 5 ⁇ , 6 ⁇ , or 7 ⁇ (i.e., daily) a week.
  • the intervals between each administration are less than about any of 6 months, 3 months, 1 month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some embodiments, the intervals between each administration are more than about any of 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, or 12 months. In some embodiments, the composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, 36, 48, 60, 72, or 84 months.
  • compositions comprising nanoparticles that comprise a taxane and a carrier protein for use in methods of treatment, methods of administration, and dosage regimes described herein.
  • the carrier protein is albumin.
  • the carrier protein is human serum albumin.
  • suitable carrier proteins include proteins normally found in blood or plasma, which include, but are not limited to, albumin, immunoglobulin including IgA, lipoproteins, apolipoprotein B, ⁇ -acid glycoprotein, ⁇ -2-macroglobulin, thyroglobulin, transferin, fibronectin, factor VII, factor VIII, factor IX, factor X, and the like.
  • the carrier protein is a non-blood protein, such as casein, ⁇ -lactalbumin, or ⁇ -lactoglobulin.
  • the carrier proteins may either be natural in origin or synthetically prepared.
  • the pharmaceutical acceptable carrier comprises albumin, such as human serum albumin (HSA).
  • HSA is a highly soluble globular protein of M r 65K and consists of 585 amino acids. HSA is the most abundant protein in the plasma and accounts for 70-80% of the colloid osmotic pressure of human plasma.
  • the amino acid sequence of HSA contains a total of 17 disulphide bridges, one free thiol (Cys 34), and a single tryptophan (Trp 214).
  • Other albumins are contemplated, such as bovine serum albumin. Use of such non-human albumins could be appropriate, for example, in the context of use of these compositions in non-human mammals, such as the veterinary animals (including domestic pets and agricultural animals).
  • HSA Human serum albumin
  • hydrophobic binding sites a total of eight for fatty acids, an endogenous ligand of HSA
  • binds a diverse set of drugs, especially neutral and negatively charged hydrophobic compounds Goodman et al., The Pharmacological Basis of Therapeutics, 9 th ed, McGraw-Hill New York (1996).
  • Two high affinity binding sites have been proposed in subdomains IIA and IIIA of HSA, which are highly elongated hydrophobic pockets with charged lysine and arginine residues near the surface which function as attachment points for polar ligand features (see, e.g., Fehske et al., Biochem. Pharmcol., 30, 687-92 (1981), Vorum, Dan. Med.
  • the carrier protein (e.g., albumin) in the composition generally serves as a carrier for the taxane (e.g., paclitaxel) or derivative thereof, i.e., the carrier protein in the composition makes the taxane (e.g., paclitaxel) or derivative thereof more readily suspendable in an aqueous medium or helps maintain the suspension as compared to compositions not comprising a carrier protein. This can avoid the use of toxic solvents for solubilizing of the taxane (e.g., paclitaxel), and thereby can reduce one or more side effects of administration of the taxane (e.g., paclitaxel) into an individual (e.g., human).
  • the composition is substantially free (e.g.
  • the nanoparticles in the composition have a solid core. In some embodiments, the nanoparticles in the composition have a core that is not aqueous (i.e., other than aqueous core). In some embodiments, the nanoparticles of the composition lack a polymeric matrix. In some embodiments, the nanoparticles of the composition are filter sterilizable. In some embodiments, the nanoparticles in the composition comprise at least one cross-linked carrier protein. In some embodiments, the nanoparticles in the composition comprise at least ten-percent of carrier protein that is cross-linked.
  • the taxane (e.g., paclitaxel) is “stabilized” in an aqueous suspension if it remains suspended in an aqueous medium (e.g., without visible precipitation or sedimentation) for an extended period of time, such as for at least about any of 0.1, 0.2, 0.25, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or 72 hours.
  • the suspension is generally, but not necessarily, suitable for administration to an individual (e.g., human). Stability of the suspension is generally (but not necessarily) evaluated at storage temperature, such as room temperature (e.g., 20-25° C.) or refrigerated conditions (e.g., 4° C.).
  • a suspension is stable at a storage temperature if it exhibits no flocculation or particle agglomeration visible to the naked eye or when viewed under the optical microscope at 1000 times, at about fifteen minutes after preparation of the suspension. Stability can also be evaluated under accelerated testing conditions, such as at a temperature that is higher than about 40° C.
  • the composition comprises nanoparticles comprising (in various embodiments consisting essentially of) a taxane (e.g., paclitaxel) and a carrier protein.
  • a taxane e.g., paclitaxel
  • the particles or nanoparticles are also referred to as droplets or nanodroplets.
  • taxane is coated with the carrier protein.
  • Particles (such as nanoparticles) of poorly water soluble pharmaceutical agents have been disclosed in, for example, U.S. Pat. Nos. 5,916,596; 6,506,405; and 6,537,579 and also in U.S. Pat. App. Pub. No. 2005/0004002A1.
  • the amount of carrier protein in the composition described herein will vary depending on the taxane (e.g., paclitaxel) or derivative thereof, and other components in the composition.
  • the composition comprises a carrier protein in an amount that is sufficient to stabilize the taxane (e.g., paclitaxel) in an aqueous suspension, for example, in the form of a stable colloidal suspension (e.g., a stable suspension of nanoparticles).
  • the carrier protein is in an amount that reduces the sedimentation rate of the taxane (e.g., paclitaxel) in an aqueous medium.
  • the amount of the carrier protein also depends on the size and density of particles of the taxane (e.g., paclitaxel).
  • the taxanes in the nanoparticles are coated with a carrier protein, such as albumin (e.g., human serum albumin).
  • the taxane is paclitaxel.
  • the composition comprises more than about any of 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the taxane or derivative thereof in nanoparticle form.
  • the taxane or derivative thereof constitutes more than about any of 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the nanoparticles by weight.
  • the nanoparticles are substantially free of polymeric core materials.
  • the taxane in the nanoparticles is amorphous.
  • the taxane used for making the nanoparticle compositions is in anhydrous form.
  • the carrier protein (such as albumin) to a taxane weight ratio in the taxane nanoparticle composition is about any of 18:1 or less, 15:1 or less, 14:1 or less, 13:1 or less, 12:1 or less, 11:1 or less, 10:1 or less, 9:1 or less, 8:1 or less, 7.5:1 or less, 7:1 or less, 6:1 or less, 5:1 or less, 4:1 or less, or 3:1 or less.
  • the albumin to taxane weight ratio is between about any of 18:1 to 1:18, 10:1 to 1:10, 9:1 to 1:1, 8:1 to 1:1, 7.5:1 to 1.1, 7.1:1 to 1:1, 6:1 to 1:1, 5:1 to 1:1, 4:1 to 1:1, or 3:1 to 1:1.
  • the composition comprises a stable aqueous suspension of particles (e.g., nanoparticles) comprising a taxane and carrier protein (such as albumin, e.g., particles of paclitaxel coated with albumin).
  • the composition comprises nanoparticles of any shape (e.g., a spherical or non-spherical shape) with an average or mean diameter of no greater than about 1000 nanometers (nm), such as no greater than about any of 900 nm, 800 nm, 700 nm, 600 nm, 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm, or 50 nm. In some embodiments, the average or mean diameter of the particles is no greater than about 200 nm. In some embodiments, the composition is sterile filterable. In some embodiments, the composition is sterile filtered.
  • any shape e.g., a spherical or non-spherical shape
  • an average or mean diameter of no greater than about 1000 nanometers (nm) such as no greater than about any of 900 nm, 800 nm, 700 nm, 600 n
  • the average or mean diameter of the particles is no greater than about 100 nm. In some embodiments, the average or mean diameter of the particles is between about 20 to about 400 run. In some embodiments, the average or mean diameter of the particles is between about 40 to about 200 nm. In some embodiments, the particles are sterile-filterable. In some embodiments, the composition comprises nanoparticles with a diameter of about any of 1000 nm, 900 nm, 800 nm, 700 nm, 600 nm, 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm, or 50 nm.
  • the composition comprises nanoparticles with a diameter of between about any of 50 nm and 150 nm, 50 nm and 75 nm, 50 nm and 100 nm, 75 nm and 100 nm, 100 nm and 125 nm, 125 nm and 150 nm, 100 nm and 150 nm, 150 nm and 175 nm, or 175 nm and 200 nm.
  • the diameter of about any of 50% or more, 65% or more, 75% or more, 80% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 99.9% or more of the nanoparticles can fall within the range specified. Average or mean diameters of nanoparticles can be determined by methods known in the art, for example laser light scattering.
  • the nanoparticles described herein may be present in a dry formulation (e.g., lyophilized composition) or suspended in a biocompatible medium.
  • Suitable biocompatible media include, but are not limited to, water, buffered aqueous media, saline, buffered saline, optionally buffered solutions of amino acids, optionally buffered solutions of proteins, optionally buffered solutions of sugars, optionally buffered solutions of vitamins, optionally buffered solutions of synthetic polymers, lipid-containing emulsions, and the like.
  • the nanoparticles do not comprise a blood-insoluble gas or do not comprise gas-filled microbubbles.
  • the carrier protein is present in an effective amount to reduce one or more side effects associated with administration of taxane to a human compared to compositions without carrier protein.
  • side effects include, but are not limited to, myelosuppression, neurotoxicity, hypersensitivity, inflammation, venous irritation, phlebitis, pain, skin irritation, neutropenic fever, anaphylactic reaction, hematologic toxicity, and cerebral or neurologic toxicity, and combinations thereof.
  • a method of reducing hypersensitivity reactions associated with administration of the taxane including, for example, severe skin rashes, hives, flushing, dyspnea, tachycardia, pulmonary hypertension (e.g., lymphoma); chest pain; black, tarry stools; general feeling of illness, shortness of breath; swollen glands; weight loss; yellow skin and eyes, abdominal pain; unexplained anxiousness; bloody or cloudy urine; bone pain; chills; confusion; convulsions (seizures); cough; decreased urge to urinate; fast, slow, or irregular heartbeat; fever; frequent urge to urinate; increased thirst; loss of appetite; lower back or side pain; mood changes; muscle pain or cramps; nausea or vomiting; numbness or tingling around lips, hands, or feet; painful or difficult urination; rash; sore throat; sores or white spots on lips or in mouth; swelling of hands
  • the taxane e.g., paclitaxel
  • side effects are merely exemplary and other side effects, or combination of side effects, associated with the taxane (e.g., paclitaxel) can be reduced.
  • the side effects may be immediate or delayed (such as not occurring for a few days, weeks, months, or years after treatment begins).
  • the compositions of the invention also includes an antimicrobial agent (e.g., an agent in addition to the taxane (e.g., paclitaxel)) in an amount sufficient to significantly inhibit (e.g., delay, reduce, slow, and/or prevent) microbial growth in the composition for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • an antimicrobial agent e.g., an agent in addition to the taxane (e.g., paclitaxel)
  • an antimicrobial agent e.g., an agent in addition to the taxane (e.g., paclitaxel)
  • an antimicrobial agent e.g., an agent in addition to the taxane (e.g., paclitaxel)
  • an antimicrobial agent e.g., an agent in addition to the taxane (e.g., paclitaxel)
  • significantly inhibit e.g., delay, reduce, slow, and/or prevent
  • the antimicrobial agent is a chelating agent, such as EDTA, edetate, citrate, pentetate, tromethamine, sorbate, ascorbate, derivatives thereof; or mixtures thereof.
  • the antimicrobial agent is a polydentate chelating agent.
  • the antimicrobial agent is a non-chelating agent, such as any of sulfites, benzoic acid, benzyl alcohol, chlorobutanol, and paraben.
  • an antimicrobial other than the taxane discussed above is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • the compositions of the invention include a sugar for use in the methods of treatment described herein.
  • the compositions of the invention include both a sugar and an antimicrobial agent for use in the methods of treatment described herein.
  • Exemplary sugars and variations for the use of sugars are disclosed in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006 (such as those described in paragraphs [0084] to [0090]).
  • the sugar serves as a reconstitution enhancer which causes a lyophilized composition to dissolve or suspend in water and/or aqueous solution more quickly than the lyophilized composition would dissolve without the sugar.
  • the composition is a liquid (e.g., aqueous) composition obtained by reconstituting or resuspending a dry composition.
  • concentration of sugar in the composition is greater than about 50 mg/ml.
  • the sugar is in an amount that is effective to increase the stability of the taxane (e.g., paclitaxel) or derivative thereof in the composition as compared to a composition without the sugar.
  • the sugar is in an amount that is effective to improve filterability of the composition as compared to a composition without the sugar.
  • the sugar-containing compositions described herein may further comprise one or more antimicrobial agents, such as the antimicrobial agents described herein or in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006.
  • antimicrobial agents such as the antimicrobial agents described herein or in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006.
  • other reconstitution enhancers such as those described in U.S. Pat. App. Publication No. 2005/0152979, which is hereby incorporated by reference in its entirety
  • a sugar is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • compositions of the invention also include a stabilizing agent for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • the compositions of the invention include an antimicrobial agent and/or a sugar and/or a stabilizing agent for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • Exemplary stabilizing agents and variations for the use of stabilizing agents are disclosed in U.S. Ser. No. 11/513,756, filed Aug. 30, 2006 (such as those described in paragraphs [0038] to [0083] and [0107] to [0114]).
  • the present invention in another variation provides for compositions and methods of preparation of a taxane (e.g., paclitaxel) which retain the desirable therapeutic effects and remain physically and/or chemically stable upon exposure to certain conditions such as prolonged storage, elevated temperature, or dilution for parenteral administration.
  • the stabilizing agent includes, for example, chelating agents (e.g., citrate, malic acid, edetate, or pentetate), sodium pyrophosphate, and sodium gluconate.
  • the invention provides pharmaceutical formulations of a taxane (e.g., paclitaxel) comprising citrate, sodium pyrophosphate, EDTA, sodium gluconate, citrate and sodium chloride, and/.
  • the invention provides a composition of a taxane, wherein the taxane (e.g., paclitaxel) used for preparing the formulation is in an anhydrous form prior to being incorporated into the composition.
  • a stabilizing agent is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • compositions described herein may be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the nanoparticle composition(s) described with a pharmaceutical acceptable carrier, excipients, stabilizing agents or other agent, which are known in the art, for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • a pharmaceutical acceptable carrier such as a pharmaceutical formulation
  • excipients such as a pharmaceutical acceptable carrier
  • stabilizing agents or other agent which are known in the art, for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • certain negatively charged components may be added.
  • Such negatively charged components include, but are not limited to bile salts, bile acids, glycocholic acid, cholic acid, chenodeoxycholic acid, taurocholic acid, glycochenodeoxycholic acid, taurochenodeoxycholic acid, litocholic acid, ursodeoxycholic acid, dehydrocholic acid, and others; phospholipids including lecithin (egg yolk) based phospholipids which include the following phosphatidylcholines: palmitoyloleoylphosphatidylcholine, palmitoyllinoleoylphosphatidylcholine, stearoyllinoleoylphosphatidylcholine, stearoyloleoylphosphatidylcholine, stearoylarachidoylphosphatidylcholine, and dipalmitoylphosphatidylcholine.
  • phospholipids including lecithin (egg yolk) based phospholipids which include
  • phospholipids including L- ⁇ -dimyristoylphosphatidylcholine (DMPC), dioleoylphosphatidylcholine (DOPC), distearoylphosphatidylcholine (DSPC), hydrogenated soy phosphatidylcholine (HSPC), and other related compounds.
  • Negatively charged surfactants or emulsifiers are also suitable as additives, e.g., sodium cholesteryl sulfate and the like.
  • the composition is suitable for administration to a human.
  • suitable formulations of the inventive composition see, e.g., U.S. Pat. Nos. 5,916,596 and 6,096,331, which are hereby incorporated by reference in their entireties).
  • the following formulations and methods are merely exemplary and are in no way limiting.
  • Formulations suitable for oral administration can comprise (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules, (c) suspensions in an appropriate liquid, (d) suitable emulsions, and (e) powders.
  • liquid solutions such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice
  • capsules, sachets or tablets each containing a predetermined amount of the active ingredient, as solids or granules
  • suspensions in an appropriate liquid such as water, saline, or orange juice
  • Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients.
  • Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • a flavor usually sucrose and acacia or tragacanth
  • pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation compatible with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizing agents, and preservatives.
  • the formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient methods of treatment, methods of administration, and dosage regimes described herein (i.e., water) for injection, immediately prior to use.
  • sterile liquid excipient methods of treatment, methods of administration, and dosage regimes described herein i.e., water
  • Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. Injectable formulations are preferred.
  • the invention also includes formulations of nanoparticle compositions comprising the taxane (e.g., paclitaxel) or derivative thereof and a carrier suitable for administration by inhalation for use in the methods of the invention.
  • Formulations suitable for aerosol administration comprise the inventive composition include aqueous and non-aqueous, isotonic sterile solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes, as well as aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizing agents, and preservatives, alone or in combination with other suitable components, which can be made into aerosol formulations to be administered via inhalation.
  • aqueous and non-aqueous, isotonic sterile solutions which can contain anti-oxidants, buffers, bacteriostats, and solutes
  • aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thick
  • aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also can be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • the composition is formulated to have a pH in the range of about 4.5 to about 9.0, including for example pH ranges of any of about 5.0 to about 8.0, about 6.5 to about 7.5, and about 6.5 to about 7.0.
  • the pH of the composition is formulated to no less than about 6, including for example no less than about any of 6.5, 7, or 8 (e.g., about 8).
  • the composition can also be made to be isotonic with blood by the addition of a suitable tonicity modifier, such as glycerol.
  • the nanoparticles of this invention can be enclosed in a hard or soft capsule, can be compressed into tablets, or can be incorporated with beverages or food or otherwise incorporated into the diet.
  • Capsules can be formulated by mixing the nanoparticles with an inert pharmaceutical diluent and inserting the mixture into a hard gelatin capsule of the appropriate size. If soft capsules are desired, a slurry of the nanoparticles with an acceptable vegetable oil, light petroleum or other inert oil can be encapsulated by machine into a gelatin capsule.
  • unit dosage forms comprising the compositions and formulations described herein. These unit dosage forms can be stored in a suitable packaging in single or multiple unit dosages and may also be further sterilized and sealed.
  • the pharmaceutical composition e.g., a dosage or unit dosage form of a pharmaceutical composition
  • the pharmaceutical composition also includes one or more other compounds (or pharmaceutically acceptable salts thereof) that are useful for treating cancer.
  • the amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 5 to about 50 mg, about 20 to about 50 mg, about 50 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg.
  • a taxane e.g., paclitaxel
  • the amount of a taxane (e.g., paclitaxel) or derivative thereof in the composition is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg, of the taxane (e.g., paclitaxel) or derivative thereof.
  • the carrier is suitable for parental administration (e.g., intravenous administration).
  • the taxane (e.g., paclitaxel) or derivative thereof is the only pharmaceutically active agent for the treatment of cancer that is contained in the composition.
  • the invention features a dosage form (e.g., a unit dosage form) for the treatment of cancer comprising (i) nanoparticles that comprise a carrier protein and a taxane (e.g., paclitaxel), wherein the amount of a taxane (e.g., paclitaxel) or derivative thereof in the unit dosage from is in the range of about 5 mg to about 500 mg, and (ii) a pharmaceutically acceptable carrier.
  • the amount of the taxane (e.g., paclitaxel) or derivative thereof in the unit dosage form includes about 30 mg to about 300 mg.
  • compositions, formulations, and unit dosages described herein in suitable packaging for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • suitable packaging for compositions described herein are known in the art, and include, for example, vials (such as sealed vials), vessels (such as sealed vessels), ampules, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. These articles of manufacture may further be sterilized and/or sealed.
  • kits comprising the compositions, formulations, unit dosages, and articles of manufacture described herein for use in the methods of treatment, methods of administration, and dosage regimes described herein.
  • Kits of the invention include one or more containers comprising a taxane (e.g., paclitaxel)-containing nanoparticle compositions (formulations or unit dosage forms and/or articles of manufacture), and in some embodiments, further comprise instructions for use in accordance with any of the methods of treatment described herein.
  • a taxane e.g., paclitaxel
  • nanoparticle compositions formulations or unit dosage forms and/or articles of manufacture
  • the kit comprises i) a composition comprising nanoparticles comprising a taxane (e.g., paclitaxel) and a carrier protein (such as albumin) and ii) instructions for administering the nanoparticles and the other chemotherapeutic agents simultaneously and/or sequentially, for treatment of cancer.
  • a taxane e.g., paclitaxel
  • a carrier protein such as albumin
  • the amount of a taxane (e.g., paclitaxel) in the kit is included in any of the following ranges: about 5 mg to about 20 mg, about 20 to about 50 mg, about 50 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg.
  • a taxane e.g., paclitaxel
  • the amount of a taxane (e.g., paclitaxel) in the kit is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg.
  • the kit includes one or more other compounds (i.e., one or more compounds other than a taxane) that are useful for cancer.
  • kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable.
  • the instructions relating to the use of the nanoparticle compositions generally include information as to dosage, dosing schedule, and route of administration for the intended treatment.
  • the kit may further comprise a description of selecting an individual suitable or treatment.
  • kits comprising compositions (or unit dosages forms and/or articles of manufacture) described herein and may further comprise instruction(s) on methods of using the composition, such as uses further described herein.
  • the kit of the invention comprises the packaging described above.
  • the kit of the invention comprises the packaging described above and a second packaging comprising a buffer. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for performing any methods described herein.
  • the kit may contain instructions for administering the first and second therapies simultaneously and/or sequentially for the effective treatment of cancer.
  • the first and second therapies can be present in separate containers or in a single container. It is understood that the kit may comprise one distinct composition or two or more compositions wherein one composition comprises a first therapy and one composition comprises a second therapy.
  • Kits may also be provided that contain sufficient dosages of the taxane (e.g., paclitaxel) as disclosed herein to provide effective treatment for an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months or more. Kits may also include multiple unit doses of the taxane (e.g., paclitaxel) compositions, pharmaceutical compositions, and formulations described herein and instructions for use and packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies.
  • the taxane e.g., paclitaxel
  • the kit comprises a dry (e.g., lyophilized) composition that can be reconstituted, resuspended, or rehydrated to form generally a stable aqueous suspension of nanoparticles comprising a taxane (e.g., paclitaxel) and albumin (e.g., a taxane (e.g., paclitaxel) coated with albumin).
  • a taxane e.g., paclitaxel
  • albumin e.g., a taxane (e.g., paclitaxel) coated with albumin
  • kits of the invention are in suitable packaging.
  • suitable packaging include, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Kits may optionally provide additional components such as buffers and interpretative information.
  • compositions containing carrier proteins and poorly water soluble pharmaceutical agents are known in the art.
  • nanoparticles containing poorly water soluble pharmaceutical agents and carrier proteins e.g., albumin
  • high shear forces e.g., sonication, high pressure homogenization, or the like.
  • the taxane e.g., paclitaxel
  • Suitable organic solvents include, for example, ketones, esters, ethers, chlorinated solvents, and other solvents known in the art.
  • the organic solvent can be methylene chloride, chloroform/ethanol, or chloroform/t-butanol (for example with a ratio of about any of 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, or 9:1 or with a ratio of about any of 3:7, 5:7, 4:6, 5:5, 6:5, 8:5, 9:5, 9.5:5, 5:3, 7:3, 6:4, or 9.5:0.5).
  • the solution is added to a carrier protein (e.g., human serum albumin).
  • a carrier protein e.g., human serum albumin.
  • the mixture is subjected to high pressure homogenization (e.g., using an Avestin, APV Gaulin, MicrofluidizerTM such as a MicrofluidizerTM Processor M-110EH from Microfluidics, Stansted, or Ultra Turrax homogenizer).
  • the emulsion may be cycled through the high pressure homogenizer for between about 2 to about 100 cycles, such as about 5 to about 50 cycles or about 8 to about 20 cycles (e.g., about any of 8, 10, 12, 14, 16, 18 or 20 cycles).
  • the organic solvent can then be removed by evaporation utilizing suitable equipment known for this purpose, including, but not limited to, rotary evaporators, falling film evaporators, wiped film evaporators, spray driers, and the like that can be operated in batch mode or in continuous operation.
  • the solvent may be removed at reduced pressure (such as at about any of 25 mm Hg, 30 mm Hg, 40 mm Hg, 50 mm Hg, 100 mm Hg, 200 mm Hg, or 300 mm Hg).
  • the amount of time used to remove the solvent under reduced pressure may be adjusted based on the volume of the formulation.
  • the solvent can be removed at about 1 to about 300 mm Hg (e.g., about any of 5-100 mm Hg, 10-50 mm Hg, 20-40 mm Hg, or 25 mm Hg) for about 5 to about 60 minutes (e.g., about any of 7, 8, 9, 10, 11, 12, 13, 14, 15 16, 18, 20, 25, or 30 minutes).
  • human albumin solution may be added to the dispersion to adjust the human serum albumin to the taxane (e.g., paclitaxel) ratio or to adjust the concentration of the taxane (e.g., paclitaxel) in the dispersion.
  • human serum albumin solution e.g., 25% w/v
  • human serum albumin solution can be added to adjust the human serum albumin to a taxane (e.g., paclitaxel) ratio to about any of 18:1, 15, :1 14:1, 13:1, 12:1, 11:1, 10:1, 9:1, 8:1, 7.5:1, 7:1, 6:1, 5:1, 4:1, or 3:1.
  • human serum albumin solution e.g., 25% w/v
  • another solution is added to adjust the concentration of a taxane (e.g., paclitaxel) in the dispersion to about any of 0.5 mg/ml, 1.3 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, or 50 mg/ml.
  • a taxane e.g., paclitaxel
  • the dispersion may be serially filtered through multiple filters, such as a combination of 1.2 ⁇ m and 0.8/0.2 ⁇ m filters; the combination of 1.2 ⁇ m, 0.8 ⁇ m, 0.45 ⁇ m, and 0.22 ⁇ m filters; or the combination of any other filters known in the art.
  • the dispersion obtained can be further lyophilized.
  • the nanoparticle compositions may be made using a batch process or a continuous process (e.g., the production of a composition on a large scale).
  • a second therapy e.g., one or more compounds useful for treating cancer
  • an antimicrobial agent e.g., sugar, and/or stabilizing agent
  • This additional agent can either be admixed with the taxane (e.g., paclitaxel) and/or the carrier protein during preparation of the taxane (e.g., paclitaxel)/carrier protein composition, or added after the taxane (e.g., paclitaxel)/carrier protein composition is prepared.
  • the agent is admixed with the taxane (e.g., paclitaxel)/carrier protein composition prior to lyophilization.
  • the agent is added to the lyophilized pharmaceutical agent/carrier protein composition.
  • the pH in the composition are generally (but not necessarily) adjusted to a desired pH.
  • Exemplary pH values of the compositions include, for example, in the range of about 5 to about 8.5.
  • the pH of the composition is adjusted to no less than about 6, including for example no less than any of about 6.5, 7, or 8 (e.g., about 8).
  • This example provides formulations of paclitaxel/albumin.
  • the compositions were prepared essentially as described in U.S. Pat. Nos. 5,439,686 and 5,916,596. Briefly, paclitaxel was dissolved in an organic solvent (such as methylene chloride or a chloroform/ethanol mixture), and the solution was added to a human serum albumin solution. The mixture was homogenized for 5 minutes at low RPM to form a crude emulsion, and then transferred into a high pressure homogenizer. The emulsification was performed at 9000-40,000 psi while recycling the emulsion for at least 5 cycles.
  • an organic solvent such as methylene chloride or a chloroform/ethanol mixture
  • the resulting system was transferred into a rotary evaporator, and the organic solvent was rapidly removed at 40° C., at reduced pressure (30 mm Hg) for 20-30 minutes. The dispersion was then further lyophilized for 48 hours. The resulting cake was reconstituted to the original dispersion by addition of sterile water or saline, which may optionally contain additional antimicrobial agent(s).
  • the primary objective of this study was to determine the objective response rate in platinum-sensitive patients with recurrent ovarian, peritoneal, or fallopian tube cancer.
  • the study also evaluated the progression free survival, overall survival, quality of life during treatment, and the safety and toxicity of the treatment in this patient population.
  • PS ECOG Performance Status
  • Premedication was administered at the discretion of the treating physician.
  • Patients received 260 mg/m 2 of Nab-paclitaxel (Abraxane®) administered IV during a 30-minute period on Day 1 of each 21-day cycle. Successive cycles were initiated every 3 weeks and were continued until there was evidence of disease progression, unacceptable toxicity, or until 6 cycles.
  • up to 3 cycles of treatment were to be administered at the discretion of the treating physician before response was assessed.
  • Patients who achieved a CR could have received an additional 2 cycles at the discretion of the treating physician therefore, CR patients were eligible to receive a maximum of 8 cycles.
  • Baseline Complete medical history and physical exam, assessment of ECOG performance status and peripheral neuropathy, CBC, CMP, CA-125, pregnancy test (if appropriate), clinical and radiological assessment of the sites of disease, urinalysis, ECG, and completion FACT-O quality of life questionnaire was determined.
  • CA-125 Using CA-125, a complete response was defined as a return to a normal range value for at least 28 days from the pretreatment value. See Rustin et al. Clin. Cancer Res. 10(11):3919-26 (2004). A partial response was defined as a sustained over 50% reduction from the pretreatment value. Stable disease was defined as a sustained greater than 50% increase in CA-125 over 28 days in the absence of any new clinically measurable disease, after an adequate trial of therapy. Progressive disease was defined as a sustained over 50% increase in CA-125 or development of new clinically measurable disease after an adequate trial of therapy. An increase was measured from the nadir (the lowest CA-125 value since enrollment).
  • the Quality of Life (FACT-O) questionnaire was completed at baseline and before each cycle. Overall quality of life measured decreased from baseline at the initiation of therapy. As measured by the questionnaire, the major contributors to the initial decline in quality of life were the functional, social, and physical well being assessments. All measure improved at the completion of therapy and returned to based line.
  • Nab-paclitaxel was very active as a single agent in patients with platinum sensitive recurrent ovarian, peritoneal, or fallopian tube cancer.
  • the ORR was 64% and the clinical benefit rate was 77%. Toxicities were tolerable and manageable.
  • nab-paclitaxel a 130-nm albumin-bound particle form of paclitaxel
  • carboplatin a 130-nm albumin-bound particle form of paclitaxel
  • Eligible patients had to have either measurable disease (based on RECIST criteria) or pretreatment CA-125 levels >2 times the upper limit of normal in the absence of measurable disease. Patients also had to have good performance status, adequate hepatic/renal function, peripheral neuropathy ⁇ grade 1, and life expectancy >6 months. Patients may have received prior chemotherapy for ovarian cancer, including taxane-containing regimens, provided the treatment was completed at least 6 months before enrollment. Nab-Paclitaxel 100 mg/m 2 was administered IV over 30 minutes on days 1, 8, and 15 every 28 days. Carboplatin AUC 6 was administered IV over 1-2 hours on day 1 every 28 days. Treatment continued for 6 cycles (or longer in the absence of unacceptable toxicity). Planned sample size was 43 patients (39 evaluable). Efficacy was determined by changes in tumor size for patients with measurable disease or changes in CA-125 levels for those with non-measurable disease.
  • nab-paclitaxel 100 mg/m2 plus carboplatin AUC 6 will have antitumor activity in patients with recurrent platinum-sensitive ovarian or primary peritoneal cancer and that treatment appears to be well tolerated.
  • the aim of this study was to evaluate the effectiveness of Abraxane® plus carboplatin in the treatment of patients with recurrent platinum-sensitive ovarian or primary peritoneal cancer.
  • the purpose of this study was to compare the pharmacokinetics of intravenous and intraperitoneal administration of Nab-paclitaxel.
  • intravenous administration 20 rats were dosed with Abraxane® at 50 mg/kg. Dose volume of 10 ml/kg was delivered intravenously via the tail vein. Blood was drawn at the following intervals for LC/MS/MS analysis of paclitaxel: 0.0, 0.083, 0.25, 0.5, 1, 2, 4, 8, 24, 48, 72 hr.
  • 3 rats were dosed with Abraxane® at 50 mg/kg. Dose volume of 10 ml/kg was delivered intraperitoneally. Blood was drawn at the following intervals for LC/MS/MS analysis of paclitaxel: 0.0, 0.083, 0.25, 0.5, 1, 2, 4, 8, 24, 48, 72 hr.
  • the plasma level of paclitaxel was measured by LC/MS/MS analysis ( FIG. 2 and FIG. 3 ). As indicated in Table 7, intraperitoneal delivery leaded to a slow absorption of paclitaxel into the circulation with t max occurring at ⁇ 3 hrs. The terminal half-lives of paclitaxel when administered intraperitoneally or intravenously were about the same. Systemic exposure after IP administration was ⁇ 50% of IV (over 72 hrs), indicating substantial local intraperitoneal exposure of drug, especially in the early phase (0-4 hrs) after administration. There may be sustained exposure beyond 72 hrs; however, this study was terminated at 72 hrs.
  • Intraperitoneal therapy for recurrent diseases is an area that requires further study.
  • the purpose of this experiment is to determine the safety, efficacy, and maximum tolerable dose of Nab-paclitaxel when administered intraperitoneally.
  • a Phase I dose-escalation trial is conducted with 6 patients per cohort to provide sufficient pharmacokinetic data points. Every 4 weeks dosages are escalated so that the cumulative toxicity of weekly treatment may be evaluated. When the MTD is reached, an additional 10 patients are treated to obtain further information about the tolerability at this dose continuously (or 3 ⁇ 4 weeks) as well as preliminary information regarding efficacy.
  • Intraperitoneal dosing of Abraxane® begins at 60 mg/m 2 . Doses is escalated as follows: Dose level 1: 60 mg/m 2 , Dose level 2: 80 mg/m 2 , Dose level 3: 100 mg/m 2 , Dose level 4: 125 mg/m 2 , and Dose level 5: 150 mg/m 2 . Ten patients are added at the DLT, and the study is expected to accrue approximately 30-40 patients.
  • Patients are included in the study if they meet the following criteria: having recurrent Mullerian cancer (ovarian, peritoneal, fallopian tube, malignant mixed mullerian tumor, serous endo), having disease ⁇ 1 cm by CT/MRI or SLO, displaying adequate heme/hepatic/renal function, and PS 0-1. Patients are excluded based on meeting the following criteria: having a bowel obstruction (or impending), existing intra-abdominal infection, having significant loculations/adhesions or other contraindications to IP port placement, existing neuropathy >grade 1, having extraperitoneal disease, or displaying an inability to tolerate IV paclitaxel and docetaxel, suggestive of taxane allergy.

Abstract

The present invention provides methods of treating recurrent cancer (such as recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition (such as Nab-paclitaxel or Abraxane®) comprising nanoparticles comprising a taxane and a carrier protein.

Description

    CROSS-REFERENCE TO RELATED APPLICATIONS
  • This application claims the priority benefit of the U.S. Provisional Patent Application No. 60/932,750 filed on Jun. 1, 2007, the content of which is incorporated herein by reference in its entirety.
  • TECHNICAL FIELD
  • The present invention relates to methods and compositions for the treatment of recurrent cancer, particularly recurrent ovarian cancer, comprising the administration of compositions comprising nanoparticles comprising taxane and a carrier protein (such as albumin).
  • BACKGROUND
  • Ovarian cancer forms in tissues of an ovary (one of a pair of female reproductive glands in which the ova, or eggs, are formed). Most ovarian cancers are either ovarian epithelial carcinomas (cancer that begins in the cells on the surface of the ovary) or malignant germ cell tumors (cancer that begins in egg cells). According to the National Cancer Institute, ovarian cancer is the seventh most common cancer, with an estimated 20,180 new cases in 2006, but is the fourth most deadly, with an estimated 15,310 deaths in 2006.
  • A possible genetic contribution to ovarian cancer risk is indicated by the increased incidence of this cancer among women with a family history, and by the observation of rare families in which multiple family members are affected with ovarian cancer, in a pattern compatible with autosomal dominant inheritance of cancer susceptibility. Formal studies of families (linkage analysis) have subsequently proven the existence of autosomal dominant predispositions to ovarian cancer and have led to the identification of several highly penetrant genes as the cause of inherited cancer risk in many cancer-prone families. Mutations in these genes are rare in the general population and are estimated to account for no more than 5% to 10% of ovarian cancer cases overall.
  • Although reproductive, demographic, and lifestyle factors affect risk of ovarian cancer, the single greatest ovarian cancer risk factor is a family history of the disease. A large meta-analysis of 15 published studies estimated an odds ratio (OR) of 3.1 for the risk of ovarian cancer associated with at least one first-degree relative with ovarian cancer.
  • Despite recent improvements, initial or first-line chemotherapy fails to produce a remission in more than 70% of patients with ovarian cancer. Furthermore, approximately 40-50% of the women who do achieve a remission after first-line chemotherapy will experience a recurrence of cancer within 3 years. Patients with recurrent ovarian, peritoneal, or fallopian tube cancer generally have a poor outcome with current therapies. There is a need for effective treatment method for patients with recurrent ovarian cancers. Preferably, the treatments overcome the shortcomings of current drug and transplant treatments, such as hypersensitivity reactions due to the solvent/surfactant in which drugs are dissolved.
  • Many anti-proliferative agents are dissolved in a solvent/surfactant which produces hypersensitivity reactions. Great efforts have been invested on the development of water soluble prodrugs and derivatives of anti-proliferative agents with higher hydrophilic groups to enhance water solubility and thus obviate the need for potentially toxic solvents/surfactants. Another approach to address the problem associated with the poor water solubility of anti-proliferative agents is the development of various formulations such as nanoparticles, oil-in-water emulsions, and liposomes. For example, Abraxane® is a nanoparticle composition of paclitaxel and albumin. Nanoparticle compositions of substantially poorly water soluble drugs and uses thereof have been disclosed, for example, in U.S. Pat. Nos. 5,916,596; 6,096,331; 6,749,868; and 6,537,579; U.S. Patent Appln. Pub. No. US20030199425; and PCT Application Pub. Nos. WO98/14174, WO99/00113, WO07/027,941 and WO07/027,819. Administration of Abraxane® to a patient with recurrent ovarian cancer is described in Mida et al., Gynecologic Oncology, 100:437-438 (2006).
  • The disclosures of all publications, patents, patent applications and published patent applications referred to herein are hereby incorporated herein by reference in their entirety.
  • BRIEF SUMMARY OF THE INVENTION
  • The present invention provides methods for the treatment of a recurrent cancer (such as a recurrent gynecological cancer) by administering a composition comprising nanoparticles comprising a taxane and a carrier protein (hereinafter referred to as “taxane nanoparticle composition”). In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, the recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum sensitive. In some embodiments, the recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum resistant. In some embodiments, the recurrent cancer is a recurrent lung cancer.
  • In some embodiments, the recurrent cancer is a recurrent gynecological cancer. In some embodiments, the recurrent gynecological cancer is a recurrent ovarian cancer (such as a recurrent epithelial ovarian cancer). In some embodiments, the recurrent gynecological cancer is a recurrent peritoneal cancer. In some embodiments, the recurrent gynecological cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas). Other recurrent gynecological cancers such as recurrent malignant mixed mullerian tumor and serous endo can also be treated.
  • In some embodiments, the individual is a woman who is about 40 to about 85 years old, including for example about 60 to about 70 years old. In some embodiments, the individual has an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 (such as any of 0, 1, or 2) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 3, 6, or 9 months since the completion of prior chemotherapy. In some embodiments, the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy. In some embodiments, the prior chemotherapy has a different mechanism of action than that of the taxane. In some embodiments, the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • In some embodiments, the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 3, 6, or 9 months prior to the initiation of the methods described herein. In some embodiments, the individual has received a prior cancer therapy (such as chemotherapy) and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months prior to the initiation of the methods described herein. In some embodiments, the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the methods described herein (such as within 12, 9, 6, 5, 4, 3, 2, or 1 month prior to the initiation of the methods described herein). In some embodiments, the individual does not show a symptom of hypersensitivity throughout the treatment period with methods described herein. In some embodiments, the individual does not show a symptom of hypersensitivity upon completion of the treatment with methods described herein.
  • In some embodiments when the method is directed to treatment of a recurrent ovarian, peritoneal, or fallopian tube cancer, the individual may be confirmed of having an ovarian, peritoneal, or fallopian tube cancer histologically or cytologically. In some embodiments, the individual is determined to have an ovarian, peritoneal, or fallopian tube cancer based on RECIST (Response Evaluation Criteria in Solid Tumors). In some embodiments, the individual has an elevated blood level of Cancer Antigen 125 (CA-125, for example a CA-125 level of more than about 40, 50, 60, 70, 80, or 90 units/ml, or about 2×, 3×, 4×, or more of that of the upper limit of a normal CA-125 level). In some embodiments, the individual has an altered level of a marker that is indicative of an ovarian, peritoneal, or fallopian tube cancer.
  • In some embodiments, the individual satisfies at least two of the criteria described above. For example, in some embodiments, the individual has a measurable disease by RECIST and an elevated blood level of CA-125. In some embodiments, the individual is confirmed of having an ovarian cancer histologically or cytologically and has only been treated with platinum-based agent(s) prior to administration of the nanoparticle compositions described above. In some embodiments, the individual satisfies at least any of two, three, four, five, or more criteria described above. In some embodiments, the individual satisfies all criteria described above.
  • In some embodiments, the recurrent cancer (such as recurrent ovarian cancer) is platinum-sensitive. For example, in some embodiments, the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 3, 6, or 9 months since the completion of the platinum-based chemotherapy. In some embodiments, the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of the platinum-based chemotherapy. Platinum-based chemotherapy includes, but is not limited to, treatment with carboplatin, cisplatin, and oxaliplatin. In some embodiments, the platinum-based chemotherapy is treatment with carboplatin.
  • The methods described herein comprise administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, the taxane nanoparticle composition is administered in conjunction with another chemotherapeutic agent (such as a platinum-based agent). For example, the taxane nanoparticle composition and the other chemotherapeutic agent (such as a platinum-based agent) can be administered sequentially, simultaneously, or concurrently. In some embodiments, the other chemotherapeutic agent (such as platinum-based agent) is administered in the same composition as the nanoparticles comprising taxane and carrier protein. The other chemotherapeutic agent (such as platinum-based agent) can also be formulated into a nanoparticle composition as described herein.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent. Suitable platinum-based agents include, but are not limited to, carboplatin, cisplatin, and oxaliplatin. In some embodiments, the platinum-based agent is carboplatin. In some embodiments, the taxane nanoparticle composition and the platinum-based agent are administered simultaneously. In some embodiments, the taxane nanoparticle composition and the platinum-based agent are administered sequentially. In some embodiments, the taxane nanoparticle composition and the platinum-based agent is administered concurrently.
  • In some embodiments, there is provided a method of treating a recurrent ovarian cancer in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent. In some embodiments, there is provided a method of treating a primary peritoneal cancer in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent, wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent, wherein the paclitaxel nanoparticle composition and the platinum-based agent are administered concurrently.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering (for example intravenously or intraperitoneally) to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), wherein the amount of the paclitaxel in the composition is at least about 40 mg/m2 (including for example about any of 50 mg/m2, 60 mg/m2, 70 mg/m2, 80 mg/m2, 90 mg/m2, 100 mg/m2, 150 mg/m2, or 200 mg/m2); b) an effective amount of a platinum-based agent (such as the platinum-based agent at the amount of AUC3, AUC4, or AUC6), wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently. In some embodiments, the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and a platinum-based agent (such as carboplatin) AUC6 every four weeks in the same treatment cycle. In some embodiments, the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle. In some embodiments, the method comprises intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle. In some embodiments, the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • In some embodiments, there is provided a method of treating recurrent ovarian cancer, comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle. In some embodiments, there is provided a method of treating primary peritoneal cancer, comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle. In some embodiments, the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • In some embodiments, the taxane nanoparticle composition is not administered in conjunction with a platinum-based agent, that is, the taxane nanoparticle composition is either administered in a monotherapy dosing regime or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent. In some embodiments, a platinum-based agent is not administered to the individual during the time period in which the individual is receiving one or more doses of the taxane nanoparticle composition. In some embodiments, the individual is not treated with a platinum-based agent concurrently with the administration of the taxane nanoparticle composition.
  • In some embodiments, the taxane nanoparticle composition can be administered alone, that is, the taxane nanoparticle is administered in a monotherapy dosing regime. For example, in some embodiments, the amount of the taxane nanoparticle composition administered alone is sufficient to result in a complete response in the individual. In some embodiments, the amount of the taxane nanoparticle composition administered alone is sufficient to result in a partial response in the individual. In some embodiments, the amount of the taxane nanoparticle composition administered alone is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the taxane nanoparticle composition. In some embodiments, the amount of taxane nanoparticle composition administered alone is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the taxane nanoparticle composition.
  • The taxane nanoparticle compositions described herein comprise nanoparticles comprising taxane (such as paclitaxel) and a carrier protein. In some embodiments, the taxane is paclitaxel, docetaxel, ortataxel, or IDN5390. In some embodiments, the carrier protein is albumin, such as human serum albumin. In some embodiments, the weight ratio of the carrier protein to the taxane in the taxane nanoparticle composition is less than about 18:1 (including for example less than about any of 15:1, 12:1, 10:1, such as 9:1).
  • In some embodiments, the nanoparticles comprise the taxane coated with a coating comprising the carrier protein (such as albumin). In some embodiments, the coating consists essentially of or consists of the carrier protein. In some embodiments, at least a portion of the carrier protein in the nanoparticle portion of the taxane nanoparticle composition is crosslinked (for example crosslinked by disulfide bonds). In some embodiments, the nanoparticles of the composition comprise at least 5% (including for example at least any of 10%, 15%, 20%, or 25%) of carrier protein that is cross-linked.
  • In some embodiments, the average or mean diameter of the nanoparticles in the composition is no greater than about 200 nm. In some embodiments, the average or mean diameter of the particles is between about 20 nm to about 400 nm (such as about 40 nm to about 200 nm). In some embodiments, the nanoparticles are sterile-filterable. In some embodiments, the taxane in the nanoparticles is amorphous. In some embodiments, the nanoparticles are substantially free of polymeric core materials. In some embodiments, the nanoparticles comprise a core of taxane that is substantially free of polymeric materials (such as polymertic matrix). In some embodiments, the nanoparticles in the composition have a solid core. In some embodiments, the nanoparticles in the composition have a core that is not aqueous (i.e., other than aqueous core). In some embodiments, the nanoparticles of the composition are substantially free of lipids. In some embodiments, the nanoparticles of the composition are free of lipids.
  • In some embodiments, the composition comprises more than about 50% (for example more than about any of 60%, 70%, 80%, 90%, or 95%) of the taxane in nanoparticle form. In some embodiments, the weight percentage of the taxane in the nanoparticle portion of the taxane nanoparticle composition is at least about any of 50%, 60%, 70%, 80%, 90%, or 95% of the total weight of the nanoparticle portion of the composition.
  • In some embodiments, the composition is administered at least about any of once every three weeks, once every two weeks, once a week, twice a week, three times a week, four times a week, five times a week, six times a week, or daily. Other exemplary dosing frequencies include, but are not limited to, weekly, two out of three weeks; weekly, three out of four weeks; and weekly, four out of five weeks. In some embodiments, the composition is administered (with or without breaks in administration cycles) for at least about any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, or more month(s).
  • In some embodiments, the composition is administered via any of intravenous, intraperitoneal, oral or inhalational routes. In some embodiments, the nanoparticle composition is administered with premedication. In some embodiments, the nanoparticle composition is administered without premedication.
  • The dose of the taxane in the nanoparticle composition will depend on the type of cancer to be treated, the severity and course of the cancer, the individual's clinical history, and the discretion of the attending physician. Suitable dosages of the taxane in the taxane nanoparticle compositions include, but are not limited to, about any of 50 mg/m2, 60 mg/m2, 75 mg/m2, 80 mg/m2, 90 mg/m2, 100 mg/m2, 120 mg/m2, 125 mg/m2, 150 mg/m2, 160 mg/m2, 180 mg/m2, 200 mg/m2, 210 mg/m2, 220 mg/m2, 230 mg/m2, 240 mg/m2, 260 mg/m2, and 300 mg/m2. Exemplary dosing schedules for the administration of the taxane nanoparticle composition (such as paclitaxel/albumin nanoparticle composition, for example Abraxane®) include, but are not limited to, 260 mg/m2, every three weeks; 60-150 mg/m2, weekly, without break, and 60-150 mg/m2, weekly, three out of four weeks. In addition, the taxane can be administered by following a metronomic dosing regime described herein. In some embodiments, the method comprises administering Abraxane® at 260 mg/m2 by 30 minutes IV infusion every three weeks.
  • Also provided herein are compositions, kits and unit dosage forms that are suitable for methods described herein. For example, in some embodiments, there is provided a composition for use in the treatment of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), wherein the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a composition for use in decreasing one or more symptoms resulting from a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), wherein the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a composition for use in the treatment of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer) in conjunction with a platinum-based agent, wherein the composition comprises nanoparticles comprising a taxane (such as paclitaxel), wherein the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a composition for use in decreasing one or more symptoms resulting from a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer) in conjunction with a platinum-based agent, wherein the composition comprises nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • These and other aspects and advantages of the present invention will become apparent from the subsequent detailed description and the appended claims. It is to be understood that one, some, or all of the properties of the various embodiments described herein may be combined to form other embodiments of the present invention.
  • BRIEF DESCRIPTION OF FIGURES
  • FIG. 1 a shows overall survival among patients plotted in terms of months (x-axis) to proportion of survival (y-axis).
  • FIG. 1 b shows progression free survival among patients plotted in terms of months (x-axis) to proportion not progressed (y-axis).
  • FIG. 2 shows a comparison of plasma level of paclitaxel when Nab-paclitaxel is administered intraperitoneally or intravenously (log-log).
  • FIG. 3 shows a comparison of plasma level of paclitaxel when Nab-paclitaxel is administered intraperitoneally or intravenously (log-linear).
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention provides methods for the treatment of a recurrent cancer (such as recurrent gynecological cancer) comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). Specifically, we have shown that a composition comprising nanoparticles comprising a taxane and a carrier protein (hereinafter designated as a “taxane nanoparticle composition”), particularly, a composition comprising nanoparticles comprising paclitaxel and albumin, more particularly, a composition comprising nanoparticles albumin bound paclitaxel (“Nab-paclitaxel”), was very active as a single agent or in combination with a platinum-based agent (carboplatin) in patients with platinum sensitive recurrent gynecological cancer, including recurrent ovarian, peritoneal, or fallopian tube cancer. The overall response rate for the treatment was 64% and the clinical benefit rate was 77%. Complete response was attained in about 70% patients treated with Abraxane® and carboplatin. This demonstrates that a taxane nanoparticle composition is particularly suitable (either as a single agent or in combination with a platinum-based agent) for treating platinum-sensitive recurrent cancer, particularly recurrent gynecological cancer.
  • Accordingly, the present invention in one aspect provides a method of treating a platinum-sensitive recurrent cancer comprising administering to the individual an effective amount of a composition (such as Nab-paclitaxel or Abraxane®) comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In another aspect, there is provided a method of treating a recurrent ovarian cancer (such as platinum-sensitive recurrent ovarian cancer) comprising administering to the individual an effective amount of a composition (such as Nab-paclitaxel or Abraxane®) comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin).
  • DEFINITIONS
  • As used herein, “the composition” or “compositions” includes and is applicable to compositions of the invention. The invention also provides pharmaceutical compositions comprising the components described herein.
  • Reference to “taxane” herein applies to a taxane or its derivatives and accordingly the invention contemplates and includes all these variations. Reference to “taxane” is to simplify the description and is exemplary. Taxanes include, but are not limited to, compounds that are structurally similar to or are in the same general chemical class such as paclitaxel (i.e., taxol), docetaxel (i.e., taxotere), or ortataxel, and pharmaceutically acceptable salts, derivatives, or analogs of paclitaxel, docetaxel, and ortataxel. Taxanes are antimicrobial agents that inhibit cell replication by promoting the assembly and stabilization of microtubule from tubulin dimers
  • Unless clearly indicated otherwise, “an individual” as used herein refers to human.
  • As used herein, “treatment” or “treating” is an approach for obtaining beneficial or desired results including clinical results. For purposes of this invention, beneficial or desired clinical results include, but are not limited to, one or more of the following: decreasing one or more symptoms resulting from the disease, diminishing the extent of the disease, stabilizing the disease (e.g., preventing or delaying the worsening of the disease), delay or slowing the progression of the disease, ameliorating the disease state, decreasing the dose of one or more other medications required to treat the disease, increasing the quality of life, and/or prolonging survival (including overall survival and progression free survival. In some embodiments, the composition reduces the severity of one or more symptoms associated with cancer by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95%, or 100% compared to the corresponding symptom in the same subject prior to treatment or compared to the corresponding symptom in other subjects not receiving the composition. Also encompassed by “treatment” is a reduction of pathological consequence of cancer. The methods of the invention contemplate any one or more of these aspects of treatment.
  • As used herein, “delaying” the development of cancer means to defer, hinder, slow, retard, stabilize, and/or postpone development of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individual being treated. As is evident to one skilled in the art, a sufficient or significant delay can, in effect, encompass prevention, in that the individual does not develop the disease. A method that “delays” development of cancer is a method that reduces probability of disease development in a given time frame and/or reduces the extent of the disease in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a statistically significant number of subjects. Cancer development can be detectable using standard methods, such as routine physical exams or x-ray. Development may also refer to disease progression that may be initially undetectable and includes occurrence and onset.
  • “Adjuvant setting” refers to a clinical setting in which an individual has had a history of ovarian cancer, and generally (but not necessarily) been responsive to therapy, which includes, but is not limited to, surgery (e.g., surgical resection), radiotherapy, and chemotherapy. However, because of their history of the cancer, these individuals are considered at risk of development of the disease. Treatment or administration in the “adjuvant setting” refers to a subsequent mode of treatment. The degree of risk (i.e., when an individual in the adjuvant setting is considered as “high risk” or “low risk”) depends upon several factors, most usually the extent of disease when first treated.
  • “Neoadjuvant setting” refers to a clinical setting in which the method is be carried out before the primary/definitive therapy.
  • As used herein, an “at risk” individual is an individual who is at risk of developing cancer. An individual “at risk” may or may not have detectable disease, and may or may not have displayed detectable disease prior to the treatment methods described herein. “At risk” denotes that an individual has one or more so-called risk factors, which are measurable parameters that correlate with development of cancer, which are described herein. An individual having one or more of these risk factors has a higher probability of developing cancer than an individual without these risk factor(s).
  • As used herein, by “pharmaceutically active compound” is meant a chemical compound that induces a desired effect, e.g., treating, stabilizing, preventing, and/or delaying cancer.
  • As used herein, by “combination therapy” is meant a first therapy that includes nanoparticles comprising nanoparticles comprising a taxane (e.g. paclitaxel) and a carrier protein in conjunction with a second therapy (e.g., surgery or a therapeutic agent) useful for treating, stabilizing, preventing, and/or delaying cancer. Administration in “conjunction with” another compound includes administration in the same or different composition(s), either sequentially, simultaneously, or continuously. In some embodiments, the combination therapy optionally includes one or more pharmaceutically acceptable carriers or excipients, non-pharmaceutically active compounds, and/or inert substances.
  • The term “effective amount” refers to an amount of a drug effective to treat cancer in the patient. The effective amount of the drug may reduce the number of cancer cells; reduce the tumor size; inhibit (i.e., slow to some extent and preferably stop) cancer cell infiltration into peripheral organs; inhibit (i.e., slow to some extent and preferably stop) tumor metastasis; inhibit, to some extent, tumor growth; and/or relieve to some extent one or more of the symptoms associated with the cancer. To the extent the drug may prevent growth and/or kill existing cancer cells, it may be cytostatic and/or cytotoxic. The effective amount may extend progression free survival (e.g. as measured by Response Evaluation Criteria for Solid Tumors, RECIST, or CA-125 changes), result in an objective response (including a partial response or a complete response), increase overall survival time, and/or improve one or more symptoms of cancer (e.g. as assessed by FOSI).
  • As is understood in the art, an “effective amount” may be in one or more doses, i.e., a single dose or multiple doses may be required to achieve the desired treatment endpoint. An effective amount may be considered in the context of administering one or more therapeutic agents, and a nanoparticle composition (e.g., a composition including a taxane (e.g., paclitaxel) and a carrier protein) may be considered to be given in an effective amount if, in conjunction with one or more other agents, a desirable or beneficial result may be or is achieved.
  • In some embodiments, the amount of the composition is an amount sufficient to decrease the size of a tumor, decrease the number of cancer cells, or decrease the growth rate of a tumor by at least about any of 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 95% or 100% compared to the corresponding tumor size, number of cancer cells, or tumor growth rate in the same subject prior to treatment or compared to the corresponding activity in other subjects not receiving the treatment. Standard methods can be used to measure the magnitude of this effect, such as in vitro assays with purified enzyme, cell-based assays, animal models, or human testing.
  • The term “proteins” refers to polypeptides or polymers of amino acids of any length (including full length or fragments), which may be linear or branched, comprise modified amino acids, and/or be interrupted by non-amino acids. The term also encompasses an amino acid polymer that has been modified naturally or by intervention, including, for example, disulfide bond formation, glycosylation, lipidation, acetylation, phosphorylation, or any other manipulation or modification. Also included within this term are, for example, polypeptides containing one or more analogs of an amino acid (including, for example, unnatural amino acids, etc.), as well as other modifications known in the art. The proteins described herein may be naturally-occurring, i.e., obtained or derived from a natural source (e.g., blood) or synthesized (e.g., chemically synthesized or by synthesized by recombinant DNA techniques). Exemplary carrier proteins are described herein.
  • The term “antimicrobial agent” used herein refers to an agent that is capable of inhibiting (e.g., delaying, reducing, slowing, and/or preventing) the growth of one or more microorganisms. Significant microbial growth can be measured or indicated by a number of ways known in the art, such as one or more of the following: (i) microbial growth in a composition that is enough to cause one or more adverse effects to an individual when the composition is administered to the individual; (ii) more than about 10-fold increase in microbial growth over a certain period of time (for example over a 24 hour period) upon extrinsic contamination (e.g., exposure to 10-103 colony forming units at a temperature in the range of 20 to 25° C.). Other indicia of significant microbial growth are described in U.S. Patent Application Publication No. US20070117744 (U.S. Ser. No. 11/514,030, filed Aug. 30, 2006), which is hereby incorporated by reference in its entirety.
  • “Sugar” as used herein includes, but is not limited to, monosaccharides, disaccharides, polysaccharides, and derivatives or modifications thereof. Suitable sugars for compositions described herein include, for example, mannitol, sucrose, fructose, lactose, maltose, and trehalose.
  • As used herein, by “pharmaceutically acceptable” or “pharmacologically compatible” is meant a material that is not biologically or otherwise undesirable, e.g., the material may be incorporated into a pharmaceutical composition administered to a patient without causing any significant undesirable biological effects or interacting in a deleterious manner with any of the other components of the composition in which it is contained. Pharmaceutically acceptable carriers or excipients have preferably met the required standards of toxicological and manufacturing testing and/or are included on the Inactive Ingredient Guide prepared by the U.S. Food and Drug administration.
  • “Survival” refers to the patient remaining alive, and includes overall survival as well as progression free survival.
  • “Overall survival” refers to the patient remaining alive for a defined period of time, such as 1 year, 5 years, etc. from the time of diagnosis or treatment.
  • “Progression free survival” refers to the patient remaining alive, without the cancer progressing or getting worse.
  • By “prolonging survival” is meant increasing overall or progression free survival in a treated patient relative to an untreated patient (e.g. relative to a patient not treated with a taxane nanoparticle composition).
  • As used herein, reference to “not” a value or parameter generally means and describes “other than” a value or parameter. For example, if a taxane is not administered, it means an agent other than a taxane is administered.
  • Reference to “about” a value or parameter herein includes (and describes) variations that are directed to that value or parameter per se. For example, description referring to “about X” includes description of “X”.’
  • As used herein and in the appended claims, the singular forms “a,” “or,” and “the” include plural referents unless the context clearly dictates otherwise. It is understood that aspects and variations of the invention described herein include “consisting” and/or “consisting essentially of” aspects and variations.
  • Methods of the Present Invention
  • The present invention provides methods for the treatment of a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) by administering a composition comprising nanoparticles comprising a taxane and a carrier protein. In some embodiments, there is provided a method of treating a recurrent cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein. In some embodiments, the recurrent cancer is platinum sensitive. In some embodiments, the recurrent cancer is platinum resistant. In some embodiments, the recurrent cancer is any of recurrent ovarian cancer, recurrent peritoneal cancer, recurrent fallopian tube cancer, recurrent malignant mixed mullerian tumor, and serous endo.
  • In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein. In some embodiments, there is provided a method of treating a platinum-sensitive recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein. In some embodiments, the recurrent cancer is a recurrent ovarian cancer (such as a recurrent epithelial ovarian cancer). In some embodiments, the recurrent cancer is a recurrent peritoneal cancer. In some embodiments, the recurrent cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas).
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 3, 6, 9 months since the completion of prior chemotherapy. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy. In some embodiments, the prior chemotherapy has a different mechanism of action than that of the taxane-based therapy. In some embodiments, the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 3, 6, 9 months since the completion of the platinum-based chemotherapy. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 38 months since the completion of the platinum-based chemotherapy. In some embodiments, the method further comprises administering to the individual an effective amount of a platinum-based agent (such as carboplatin). In some embodiments, the composition comprising nanoparticles comprising a taxane and a carrier protein is not administered in conjunction with a platinum agent, that is, the composition comprising nanoparticles comprising a taxane and a carrier protein is either administered alone or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 3, 6, 9 months prior to the initiation of the treatment method with the taxane nanoparticle composition. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received a prior chemotherapy and has a treatment free interval for more about any of 12, 18, 24, 36, or 48 months prior to the initiation of the treatment method with the taxane nanoparticle composition. In some embodiments, the prior chemotherapy has a different mechanism of action than that of the taxane-based therapy. In some embodiments, the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 3, 6, 9 months prior to the initiation of the treatment method with the taxane nanoparticle composition. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual has received prior platinum-based chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 38 months prior to the initiation of the treatment method with the taxane nanoparticle composition. In some embodiments, the method further comprises administering to the individual an effective amount of a platinum-based agent. In some embodiments, the composition comprising nanoparticles comprising a taxane and a carrier protein is not administered in conjunction with a platinum agent, that is, the composition comprising nanoparticles comprising a taxane and a carrier protein is either administered alone or administered in conjunction with a chemotherapeutic agent other than a platinum-based agent.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the treatment method with the taxane nanoparticle composition. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show a symptom of hypersensitivity throughout the treatment period. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the individual does not show symptoms of hypersensitivity upon completion of the treatment.
  • In some embodiments, the composition comprising nanoparticles comprising a taxane and a carrier protein is administered alone. For example, in some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is effective to result in a complete response in the individual. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is effective to result in a partial response in the individual.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the composition. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the composition administered to the individual alone is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the composition.
  • In some embodiments, the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising paclitaxel and albumin. In some embodiments, the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein average or mean diameter of the nanoparticles in the composition is less than about 200 nm. In some embodiments, the method comprises administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the nanoparticles comprise taxane coated with a coating comprising the carrier protein. In some embodiments, the method comprises administering to the individual an effective amount of a Nab-paclitaxel (such as Abraxane®). For example, in some embodiments, there is provided a method of treating a platinum-sensitive recurrent ovarian, peritoneal, or fallopian tube cancer in the individual, comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer (such as a platinum-resistant ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual about 50 mg/m2 to about 300 mg/m2 (including for example about 260 mg/m2) Nab-paclitaxel or Abraxane®. In some embodiments, the Nab-paclitaxel or Abraxane® is administered every three weeks. In some embodiments, the Nab-paclitaxel or Abraxane® is administered intravenously (such as by i.v. infusion over a period of about 30 minutes or less). In some embodiments, the Nab-paclitaxel or Abraxane® is administered intraperitoneally.
  • The methods of the present invention are useful for any one or more of the following (and thus in various embodiments can achieve and/or include any one or more of the following): 1) decreasing one or more symptoms resulting from recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 2) increasing overall response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 3) increasing partial response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 4) increasing complete response rate of a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 5) delaying disease progression of an individual with a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 6) increasing the quality of life in an individual with recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); 7) prolonging overall survival of an individual having recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer); and 8) prolonging progression-free survival of an individual having recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer).
  • Accordingly, in some embodiments, there is provided a method of decreasing one or more symptoms resulting from a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a method of decreasing one more symptoms resulting from a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • In some embodiments, there is provided a method of increasing response rate of recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a method of increasing response rate of recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • In some embodiments, there is provided a method of delaying disease progression of an individual with recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a method of delaying disease progression of an individual with recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • In some embodiments, there is provided a method of prolonging survival of an individual having recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin). In some embodiments, there is provided a method of prolonging survival of an individual having recurrent cancer (such as recurrent gynecological cancer for example ovarian, peritoneal, and fallopian tube cancer), comprising administering to the individual an effective amount of Nab-paclitaxel or Abraxane®.
  • In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein one or more symptoms resulting from the recurrent cancer is decreased. In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a partial response to treatment upon completion of less than about any of one, two, three, four, five, six, seven, or eight treatment cycles. In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a complete response to treatment upon completion of less than about any of one, two, three, four, five, six, seven, or eight treatment cycles. In some embodiments, the treatment cycle is four weeks. In some embodiments, the treatment cycle is three weeks.
  • In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual is disease free for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment. In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual does not show a symptom resulting from the recurrent cancer for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment. In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, wherein the individual has a reduced CA-125 level (such as a level below about any of 70%, 60%, 50%, 40%, 30%, 20%, 10% of the level prior to the treatment) upon completion of the treatment. In some embodiments, the individual has a reduced CA-125 level for at least about any of 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, or 24 months upon completion of the treatment.
  • In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering (for example intravenously or intraperitoneally) to the individual an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), wherein the amount of the taxane per administration is at least about 50 mg/m2 (such as at least about any of 70 mg/m2, 100 m g/m2, 150 mg/m2, 200 mg/m2, or 260 mg/m2), and wherein one or more symptoms resulting from the recurrent cancer is decreased. In some embodiments, there is provided a method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering (for example intravenously or intraperitoneally) to the individual an effective amount of Abraxane®, wherein the amount of paclitaxel per administration is at least about 50 mg/m2 (such as at least about any of 70 mg/m2, 100 mg/m2, 150 mg/m2, 200 mg/m2, or 260 mg/m2), and wherein one or more symptoms resulting from the recurrent cancer is decreased. In some embodiments, the composition is administered once every three weeks. In some embodiments, the composition is administered weekly. In some embodiments, the composition is administered three out of four weeks. In some embodiments, the individual does not show a symptom of hypersensitivity (such as neuropathy) prior to the initiation of the treatment. In some embodiments, the individual does not show a symptom of hypersensitivity throughout the treatment period. In some embodiments, the individual does not show a symptom of hypersensitivity upon completion of the treatment.
  • In some embodiments, the taxane nanoparticle composition is administered in conjunction with another chemotherapeutic agent (such as a platinum-based agent). For example, in some embodiments, the taxane nanoparticle composition and the other chemotherapeutic agent (such as a platinum-based agent) are administered simultaneously. The term “simultaneous administration,” as used herein, means that the taxane nanoparticle composition and the other chemotherapeutic agent are administered with a time separation of no more than about 15 minute(s), such as no more than about any of 10, 5, or 1 minutes. When the drugs are administered simultaneously, the taxane in the nanoparticles and the other chemotherapeutic agent may be contained in the same composition (e.g., a composition comprising both the nanoparticles and the other chemotherapeutic agent) or in separate compositions (e.g., the nanoparticles are contained in one composition and the other chemotherapeutic agent is contained in another composition). For example, the taxane and the other chemotherapeutic agent may be present in a single composition containing at least two different nanoparticles, wherein some of the nanoparticles in the composition comprise the taxane and a carrier protein, and some of the other nanoparticles in the composition comprise the other chemotherapeutic agent and a carrier protein. In some embodiments, only the taxane is contained in nanoparticles. In some embodiments, simultaneous administration of the drug in the nanoparticle composition and the other chemotherapeutic agent can be combined with supplemental doses of the taxane and/or the other chemotherapeutic agent.
  • In some embodiments, the nanoparticle composition and the other chemotherapeutic agent (such as the platinum-based agent) are administered sequentially. The term “sequential administration” as used herein means that the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered with a time separation, of more than about 15 minutes, such as more than about any of 20, 30, 40, 50, 60 or more minutes. Either the nanoparticle composition or the other chemotherapeutic agent may be administered first. The nanoparticle composition and the other chemotherapeutic agent are contained in separate compositions, which may be contained in the same or different packages.
  • In some embodiments, the administration of the nanoparticle composition and the other chemotherapeutic agent are concurrent, i.e., the administration period of the nanoparticle composition and that of the other chemotherapeutic agent overlap with each other. In some embodiments, the nanoparticle composition and the other chemotherapeutic agent are administered in the same treatment cycle(s).
  • The dosing frequency of the drug-containing nanoparticle composition and the other chemotherapeutic agent may be adjusted over the course of the treatment, based on the judgment of the administering physician. When administered separately, the drug-containing nanoparticle composition and the other chemotherapeutic agent can be administered at different dosing frequency or intervals. For example, the drug-containing nanoparticle composition can be administered weekly, while a chemotherapeutic agent can be administered more or less frequently. In some embodiments, sustained continuous release formulation of the drug-containing nanoparticle and/or chemotherapeutic agent may be used. Various formulations and devices for achieving sustained release are known in the art.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual a) an effective amount of a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent. Suitable platinum-based agents include, but are not limited to, carboplatin, cisplatin, and oxaliplatin. In some embodiments, the platinum-based agent is carboplatin. In some embodiments, the taxane nanoparticle composition and the platinum-based agent are administered simultaneously. In some embodiments, the taxane nanoparticle composition and the platinum-based agent are administered sequentially. In some embodiments, the taxane nanoparticle composition and the platinum-based agent is administered concurrently.
  • In some embodiments, there is provided a method of treating a recurrent ovarian cancer in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent. In some embodiments, there is provided a method of treating a primary peritoneal cancer in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane (such as paclitaxel) and a carrier protein (such as albumin), and b) an effective amount of a platinum-based agent, wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently. In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), and b) an effective amount of a platinum-based agent, wherein the paclitaxel nanoparticle composition and the platinum-based agent are administered concurrently.
  • In some embodiments, there is provided a method of treating a recurrent cancer (such as a recurrent gynecological cancer, for example recurrent ovarian, peritoneal, or fallopian tube cancer) in an individual, comprising administering (for example intravenously or intraperitoneally) to the individual: a) an effective amount of a composition comprising nanoparticles comprising a paclitaxel and an albumin (such as Abraxane®), wherein the amount of the paclitaxel in the composition is at least about 40 mg/m2 (including for example about any of 50 mg/m2, 60 mg/m2, 70 mg/m2, 80 mg/m2, 90 mg/m2, 100 mg/m2, 150 mg/m2, or 200 mg/m2); b) an effective amount of a platinum-based agent (such as the platinum-based agent at the amount of AUC3, AUC4, or AUC6), wherein the taxane nanoparticle composition and the platinum-based agent are administered concurrently. In some embodiments, the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and a platinum-based agent (such as carboplatin) AUC6 every four weeks in the same treatment cycle. In some embodiments, the method comprises administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle. In some embodiments, the method comprises intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 every four weeks in the same treatment cycle. In some embodiments, the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • In some embodiments, there is provided a method of treating recurrent ovarian cancer, comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle. In some embodiments, there is provided a method of treating primary peritoneal cancer, comprising intravenously administering a composition comprising nanoparticles comprising paclitaxel and an albumin (such as Abraxane®) at about 100 mg/m2, three out of four weeks, and intravenously administering a platinum-based agent (such as carboplatin) AUC5 (or AUC6) every four weeks in the same treatment cycle. In some embodiments, the individual is treated with at least any of about one, two, three, four, five, six, seven, eight, or more such treatment cycles.
  • Individual Having Recurrent Cancer
  • The present invention provides a method of treating a recurrent cancer (such as recurrent gynecological cancer for example recurrent ovarian, peritoneal, and fallopian tube cancer) in an individual. A “recurrent” cancer is one which has regrown, either at the initial site or at a distant site, after a response to initial therapy. In some embodiments, the length of time between the completion of initial therapy and the development of recurrent disease is longer than about 3 month, including for example longer than about any of 4, 5, 6, 7, 8, 9, 10, or 11 months. In some embodiments, the length of time between the completion of initial therapy and the development of recurrent disease is longer than about 12 month, including for example, longer than about any of 14, 16, 18, 20, 22, 24, 36, or 48 months.
  • In some embodiments, the recurrent cancer is a recurrent gynecological cancer. “Gynecologic cancer” used herein refers to a cancer originating in the female reproductive organs. It includes cancers of the cervix, fallopian tubes, ovaries, uterus, vagina and vulva. In some embodiments, the recurrent cancer is a recurrent ovarian cancer. In some embodiments, the cancer is ovarian epithelial cancer. Exemplary ovarian epithelial cancer histological classifications include: serous cystomas (e.g., serous benign cystadenomas, serous cystadenomas with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or serous cystadenocarcinomas), mucinous cystomas (e.g., mucinous benign cystadenomas, mucinous cystadenomas with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or mucinous cystadenocarcinomas), endometrioid tumors (e.g., endometrioid benign cysts, endometrioid tumors with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or endometrioid adenocarcinomas), clear cell (mesonephroid) tumors (e.g., begin clear cell tumors, clear cell tumors with proliferating activity of the epithelial cells and nuclear abnormalities but with no infiltrative destructive growth, or clear cell cystadenocarcinomas), unclassified tumors that cannot be allotted to one of the above groups, or other malignant tumors.
  • In some embodiments, the recurrent cancer is a recurrent ovarian germ cell tumor. Exemplary histologic subtypes include dysgerminomas or other germ cell tumors (e.g., endodermal sinus tumors such as hepatoid or intestinal tumors, embryonal carcinomas, olyembryomas, choriocarcinomas, teratomas, or mixed form tumors). Exemplary teratomas are immature teratomas, mature teratomas, solid teratomas, and cystic teratomas (e.g., dermoid cysts such as mature cystic teratomas, and dermoid cysts with malignant transformation). Some teratomas are monodermal and highly specialized, such as struma ovarii, carcinoid, struma ovarii and carcinoid, or others (e.g., malignant neuroectodermal and ependymomas).
  • In some embodiments, the recurrent cancer is a recurrent peritoneal cancer. In some embodiments, the recurrent cancer is a recurrent fallopian tube cancer (including for example papillary serous adenocarcinomas, sarcomas, and transitional cell carcinomas). Other recurrent cancers such as recurrent malignant mixed mullerian tumor and serous endo can also be treated.
  • Recurrence of ovarian, peritoneal, or fallopian tube cancer can be determined, for example, based on bimanual pelvic examinations, serial measurements of CA-125 or other tumor markers, one or more imaging studies reassessments, and second-look laparotomy. Imaging studies such as CT, PET, or a CT/PET combination can also be used. In some embodiments, recurrence of ovarian, peritoneal, or fallopian tube cancer can be determined based on Response Evaluation Criteria in Solid Tumors (RECIST).
  • “Individual” used herein refers to human. In some embodiments, the individual is a woman who is about 40 to about 85 years old, including for example about 60 to about 70 years old. In some embodiments, the individual has an Eastern Cooperative Oncology Group (ECOG) performance status of 0-2 (such as any of 0, 1, or 2) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has received a prior chemotherapy and has a treatment free interval for more than about any of 3, 6, or 9 months since the completion of prior chemotherapy. In some embodiments, the individual has received a prior chemotherapy and has a treatment free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of prior chemotherapy. In some embodiments, the prior chemotherapy has a different mechanism of action than that of the taxane. In some embodiments, the individual has only been treated with platinum-based agent(s) prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has only been treated with one dosing regime prior to the administration of the taxane nanoparticle composition. In some embodiments, the individual has not previously been treated with a taxane-based therapy.
  • In some embodiments, when the method is directed to treatment of a recurrent ovarian, peritoneal, or fallopian tube cancer, the individual is confirmed of having an ovarian, peritoneal, or fallopian tube cancer histologically or cytologically. In some embodiments, the individual is determined to have an ovarian, peritoneal, or fallopian tube cancer based on RECIST. In some embodiments, the individual has an elevated blood level of Cancer Antigen 125 (CA-125, for example a CA-125 level of more than about 40, 50, 60, 70, 80, or 90 units/ml, or about 2×, 3×, 4×, or more of that of the upper limit of a normal CA-125 level). In some embodiments, the individual has an altered expression level of another marker that is indicative of a recurrent ovarian, peritoneal, and fallopian tube cancer.
  • The individual being treated may have one or more risk factors associated with a higher probability of developing ovarian, peritoneal, and fallopian tube cancer. These risk factors include, but are not limited to, age, sex, race, diet, history of previous disease, presence of precursor disease, genetic (i.e., hereditary) considerations, and environmental exposure. In some embodiments, the individual may be a human who is genetically or otherwise predisposed to developing ovarian, peritoneal, and fallopian tube cancer, particularly recurrent ovarian, peritoneal, and fallopian tube cancer.
  • Individuals at risk for ovarian, peritoneal, and fallopian tube cancer include, for example, those having relatives who have experienced this disease, and those whose risk is determined by analysis of genetic or biochemical markers. For example, the individual may be a human who has a gene, genetic mutation, or polymorphism associated with ovarian cancer (e.g., BRCA1 or BRCA2) or has one or more extra copies of a gene associated with ovarian cancer (e.g., one or more extra copies of the HER2 gene). In some embodiments, the individual is HER2 positive. In some embodiments, the individual is HER2 negative. In some embodiments, the ovarian cancer is associated with basal cell nevus (Gorlin) syndrome, multiple endocrine neoplasia type 1 (MEN1), or hereditary nonpolyposis colon cancer (HNPCC).
  • In some embodiments, the individual satisfies at least two of the criteria described above. For example, in some embodiments, the individual has a measurable disease by RECIST and an elevated blood level of CA-125. In some embodiments, the individual is confirmed of having an ovarian cancer histologically or cytologically and has only been treated with platinum-based agent(s) prior to administration of the nanoparticle compositions described above. In some embodiments, the individual satisfies at least any of two, three, four, five, or more criteria described above. In some embodiments, the individual satisfies all criteria described above.
  • In some embodiments, the recurrent cancer is platinum sensitive. For example, in some embodiments, the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 3, 6, or 9 months since the completion of the platinum-based chemotherapy. In some embodiments, the individual has received prior platinum-based chemotherapy and has a treatment-free interval for more than about any of 12, 18, 24, 36, or 48 months since the completion of the platinum-based chemotherapy. In some embodiments, the recurrent cancer is a platinum-sensitive ovarian cancer. In some embodiments, the recurrent cancer is a platinum-sensitive peritoneal cancer. In some embodiments, the recurrent cancer is a platinum-sensitive fallopian tube cancer.
  • In some embodiments, the recurrent cancer (such as a recurrent ovarian, peritoneal, or fallopian tube cancer) is platinum resistant. By “platinum-resistant” cancer is meant that the individual with cancer has progressed while receiving platinum-based chemotherapy (i.e. the patient is “platinum refractory”).
  • By “platinum-based chemotherapy” is intended to mean therapy with one or more platinum-based chemotherapeutic agents, optionally in combination with one or more other chemotherapeutic agents. A “platinum-based chemotherapeutic agent” comprises an organic compound which contains platinum as an integral part of the molecule. These agents are believed to inhibit cell growth by forming reactive platinum complexes which form intrastrand and interstrand cross-linking of DNA molecules and inhibit DNA synthesis. Examples of platinum-based chemotherapeutic agents include carboplatin, cisplatin, and oxaliplatinum. In some embodiments, the platinum-based chemotherapy comprises treatment with only platinum-based agents. In some embodiments, the platinum-based chemotherapy comprises treatment with carboplatin.
  • Dosing and Method of Administration
  • The amount of the inventive composition administered to an individual (such as a human) may vary with the particular composition, the method of administration, and the particular type of recurrent cancer being treated. The amount should be sufficient to produce a desirable beneficial effect. For example, in some embodiments, the amount of the composition is effective to result in an objective response (such as a partial response or a complete response). In some embodiments, the amount of the taxane nanoparticle composition is sufficient to result in a complete response in the individual. In some embodiments, the amount of the taxane nanoparticle composition is sufficient to result in a partial response in the individual. In some embodiments, the amount of the taxane nanoparticle composition administered (for example when administered alone) is sufficient to produce an overall response rate of more than about any of 40%, 50%, 60%, or 64% among a population of individuals treated with the taxane nanoparticle composition. Responses of an individual to the treatment of the methods described herein can be determined, for example, based on RECIST or CA-125 level. For example, when CA-125 is used, a complete response can be defined as a return to a normal range value of at least 28 days from the pretreatment value. A particle response can be defined as a sustained over 50% reduction from the pretreatment value.
  • In some embodiments, the amount of the composition is sufficient to prolong progress-free survival of the individual (for example as measured by RECIST or CA-125 changes). In some embodiments, the amount of the composition is sufficient to prolong overall survival of the individual. In some embodiments, the amount of the composition (for example when administered along) is sufficient to produce clinical benefit of more than about any of 50%, 60%, 70%, or 77% among a population of individuals treated with the taxane nanoparticle composition.
  • In some embodiments, the amount of the taxane (e.g., paclitaxel) in the composition is below the level that induces a toxicological effect (i.e., an effect above a clinically acceptable level of toxicity) or is at a level where a potential side effect can be controlled or tolerated when the composition is administered to the individual. In some embodiments, the amount of the composition is close to a maximum tolerated dose (MTD) of the composition following the same dosing regime. In some embodiments, the amount of the composition is more than about any of 80%, 90%, 95%, or 98% of the MTD.
  • In some embodiments, the amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 0.5 to about 5 mg, about 5 to about 10 mg, about 10 to about 15 mg, about 15 to about 20 mg, about 20 to about 25 mg, about 20 to about 50 mg, about 25 to about 50 mg, about 50 to about 75 mg, about 50 to about 100 mg, about 75 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg. In some embodiments, the amount of a taxane (e.g., paclitaxel) or derivative thereof in the effective amount of the composition (e.g., a unit dosage form) is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg. In some embodiments, the concentration of the taxane (e.g., paclitaxel) in the composition is dilute (about 0.1 mg/ml) or concentrated (about 100 mg/ml), including for example any of about 0.1 to about 50 mg/ml, about 0.1 to about 20 mg/ml, about 1 to about 10 mg/ml, about 2 mg/ml to about 8 mg/ml, about 4 to about 6 mg/ml, about 5 mg/ml. In some embodiments, the concentration of the taxane (e.g., paclitaxel) is at least about any of 0.5 mg/ml, 1.3 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, or 50 mg/ml.
  • Exemplary effective amounts of a taxane (e.g., paclitaxel) in the nanoparticle composition include, but are not limited to, about any of 25 mg/m2, 30 mg/m2, 50 mg/m2, 60 mg/m2, 75 mg/m2, 80 mg/m2, 90 mg/m2, 100 mg/m2, 120 mg/m2, 125 mg/m2, 150 mg/m2, 160 mg/m2, 175 mg/m2, 180 mg/m2, 200 mg/m2, 210 mg/m2, 220 mg/m2, 250 mg/m2, 260 mg/m2, 300 mg/m2, 350 mg/m2, 400 mg/m2, 500 mg/m2, 540 mg/m2, 750 mg/m2, 1000 mg/m2, or 1080 mg/m2 of a taxane (e.g., paclitaxel). In various embodiments, the composition includes less than about any of 350 mg/m2, 300 mg/m2, 250 mg/m2, 200 mg/m2, 150 mg/m2, 120 mg/m2, 100 mg/m2, 90 mg/m2, 50 mg/m2, or 30 mg/m2 of a taxane (e.g., paclitaxel). In some embodiments, the amount of the taxane (e.g., paclitaxel) per administration is less than about any of 25 mg/m2, 22 mg/m2, 20 mg/m2, 18 mg/m2, 15 mg/m2, 14 mg/m2, 13 mg/m2, 12 mg/m2, 11 mg/m2, 10 mg/m2, 9 mg/m2, 8 mg/m2, 7 mg/m2, 6 mg/m2, 5 mg/m2, 4 mg/m2, 3 mg/m2, 2 mg/m2, or 1 mg/m2. In some embodiments, the effective amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 1 to about 5 mg/m2, about 5 to about 10 mg/m2, about 10 to about 25 mg/m2, about 25 to about 50 mg/m2, about 50 to about 75 mg/m2, about 75 to about 100 mg/m2, about 100 to about 125 mg/m2, about 125 to about 150 mg/m2, about 150 to about 175 mg/m2, about 175 to about 200 mg/m2, about 200 to about 225 mg/m2, about 225 to about 250 mg/m2, about 250 to about 300 mg/m2, about 300 to about 350 mg/m2, or about 350 to about 400 mg/m2. Preferably, the effective amount of a taxane (e.g., paclitaxel) in the composition is about 5 to about 300 mg/m2, such as about 100 to about 150 mg/m2, about 120 mg/m2, about 130 mg/m2, or about 140 mg/m2.
  • In some embodiments of any of the above aspects, the effective amount of a taxane (e.g., paclitaxel) in the composition includes at least about any of 1 mg/kg, 2.5 mg/kg, 3.5 mg/kg, 5 mg/kg, 6.5 mg/kg, 7.5 mg/kg, 10 mg/kg, 15 mg/kg, or 20 mg/kg. In various embodiments, the effective amount of a taxane (e.g., paclitaxel) in the composition includes less than about any of 350 mg/kg, 300 mg/kg, 250 mg/kg, 200 mg/kg, 150 mg/kg, 100 mg/kg, 50 mg/kg, 25 mg/kg, 20 mg/kg, 10 mg/kg, 7.5 mg/kg, 6.5 mg/kg, 5 mg/kg, 3.5 mg/kg, 2.5 mg/kg, or 1 mg/kg of a taxane (e.g., paclitaxel).
  • Exemplary dosing frequencies include, but are not limited to, weekly without break; weekly, three out of four weeks; once every three weeks; once every two weeks; weekly, two out of three weeks. In some embodiments, the composition is administered about once every 2 weeks, once every 3 weeks, once every 4 weeks, once every 6 weeks, or once every 8 weeks. In some embodiments, the composition is administered at least about any of 1×, 2×, 3×, 4×, 5×, 6×, or 7× (i.e., daily) a week. In some embodiments, the intervals between each administration are less than about any of 6 months, 3 months, 1 month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some embodiments, the intervals between each administration are more than about any of 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, or 12 months. In some embodiments, there is no break in the dosing schedule. In some embodiments, the interval between each administration is no more than about a week.
  • The administration of the composition can be extended over an extended period of time, such as from about a month up to about seven years. In some embodiments, the composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, 36, 48, 60, 72, or 84 months. In some embodiments, the taxane (e.g., paclitaxel) or derivative thereof is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 0.25 mg/m2 to about 75 mg/m2, such as about 0.25 mg/m2 to about 25 mg/m2 or about 25 mg/m2 to about 50 mg/m2.
  • In some embodiments, the dosage of a taxane (e.g., paclitaxel) in a nanoparticle composition can be in the range of 5-400 mg/m2 when given on a 3 week schedule, or 5-250 mg/m2 when given on a weekly schedule. For example, the amount of a taxane (e.g., paclitaxel) is about 60 to about 300 mg/m2 (e.g., about 260 mg/m2).
  • Other exemplary dosing schedules for the administration of the nanoparticle composition (e.g., paclitaxel/albumin nanoparticle composition) include, but are not limited to, 100 mg/m2, weekly, without break; 75 mg/m2 weekly, 3 out of four weeks; 100 mg/m2, weekly, 3 out of 4 weeks; 125 mg/m2, weekly, 3 out of 4 weeks; 125 mg/m2, weekly, 2 out of 3 weeks; 130 mg/m2, weekly, without break; 175 mg/m2, once every 2 weeks; 260 mg/m2, once every 2 weeks; 260 mg/m2, once every 3 weeks; 180-300 mg/m2, every three weeks; 60-175 mg/m2, weekly, without break; 20-150 mg/m2 twice a week; and 150-250 mg/m2 twice a week. The dosing frequency of the composition may be adjusted over the course of the treatment based on the judgment of the administering physician.
  • In some embodiments, the individual is treated for at least about any of one, two, three, four, five, six, seven, eight, nine, or ten treatment cycles.
  • The compositions described herein allow infusion of the composition to an individual over an infusion time that is shorter than about 24 hours. For example, in some embodiments, the composition is administered over an infusion period of less than about any of 24 hours, 12 hours, 8 hours, 5 hours, 3 hours, 2 hours, 1 hour, 30 minutes, 20 minutes, or 10 minutes. In some embodiments, the composition is administered over an infusion period of about 30 minutes.
  • In some embodiments, the invention provides a method of treating cancer in an individual by parenterally administering to the individual (e.g., a human) an effective amount of a composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin). The invention also provides a method of treating cancer in an individual by intravenous, intra-arterial, intramuscular, subcutaneous, inhalation, oral, intraperitoneal, nasally, or intra-tracheal administering to the individual (e.g., a human) an effective amount of a composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin). In some embodiments, the route of administration is intraperitoneal. In some embodiments, the route of administration is intravenous, intra-arterial, intramuscular, or subcutaneous. In various embodiments, about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 to about 500 mg, of the taxane (e.g., paclitaxel) or derivative thereof is administered per dose. In some embodiments, the taxane (e.g., paclitaxel) or derivative thereof is the only pharmaceutically active agent for the treatment of cancer that is contained in the composition. In some embodiments, the method comprises intraperitoneally administering composition comprising nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein (e.g., albumin such as human serum albumin) at the dose of any of about 60 mg/m2, 80 mg/m2, 100 mg/m2, 125 mg/m2, and 150 mg/m2.
  • Any of the compositions described herein can be administered to an individual (such as human) via various routes, including, for example, intravenous, intra-arterial, intraperitoneal, intrapulmonary, oral, inhalation, intravesicular, intramuscular, intra-tracheal, subcutaneous, intraocular, intrathecal, transmucosal, and transdermal. In some embodiments, sustained continuous release formulation of the composition may be used. In one variation of the invention, nanoparticles (such as albumin nanoparticles) of the inventive compounds can be administered by any acceptable route including, but not limited to, orally, intramuscularly, transdermally, intravenously, through an inhaler or other air borne delivery systems and the like.
  • In some embodiments, paclitaxel-containing nanoparticles composition may be administered with a second therapeutic compound and/or a second therapy. The dosing frequency of the paclitaxel-containing nanoparticles composition and the second compound may be adjusted over the course of the treatment based on the judgment of the administering physician. In some embodiments, the first and second therapies are administered simultaneously, sequentially, or concurrently. When administered separately, the taxane (e.g., paclitaxel)-containing nanoparticles composition and the second compound can be administered at different dosing frequency or intervals. For example, the taxane (e.g., paclitaxel)-containing nanoparticle composition can be administered weekly, while a second compound can be administered more or less frequently. In some embodiments, sustained continuous release formulation of the taxane (e.g., paclitaxel)-containing nanoparticle and/or second compound may be used. Various formulations and devices for achieving sustained release are known in the art. A combination of the administration configurations described herein can be used.
  • When the taxane nanoparticle compositions are administered in conjunction with another chemotherapeutic agent, the nanoparticle composition and the other chemotherapeutic agent can be administered using the same route of administration or different routes of administration. In some embodiments (for both simultaneous and sequential administrations), the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered at a predetermined ratio. For example, in some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is about 1 to 1. In some embodiments, the weight ratio may be between about 0.001 to about 1 and about 1000 to about 1, or between about 0.01 to about 1 and 100 to about 1. In some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is less than about any of 100:1, 50:1, 30:1, 10:1, 9:1, 8:1, 7:1, 6:1, 5:1, 4:1, 3:1, 2:1, and 1:1 In some embodiments, the ratio by weight of the taxane in the nanoparticle composition and the other chemotherapeutic agent is more than about any of 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, 9:1, 30:1, 50:1, 100:1. Other ratios are contemplated.
  • The doses required for the taxane and/or the other chemotherapeutic agent may (but not necessarily) be lower than what is normally required when each agent is administered alone. Thus, in some embodiments, a subtherapeutic amount of the drug in the nanoparticle composition and/or the other chemotherapeutic agent are administered. “Subtherapeutic amount” or “subtherapeutic level” refer to an amount that is less than the therapeutic amount, that is, less than the amount normally used when the drug in the nanoparticle composition and/or the other chemotherapeutic agent are administered alone. The reduction may be reflected in terms of the amount administered at a given administration and/or the amount administered over a given period of time (reduced frequency).
  • In some embodiments, enough chemotherapeutic agent is administered so as to allow reduction of the normal dose of the drug in the nanoparticle composition required to effect the same degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more. In some embodiments, enough drug in the nanoparticle composition is administered so as to allow reduction of the normal dose of the other chemotherapeutic agent required to effect the same degree of treatment by at least about any of 5%, 10%, 20%, 30%, 50%, 60%, 70%, 80%, 90%, or more.
  • In some embodiments, the dose of both the taxane in the nanoparticle composition and the other chemotherapeutic agent are reduced as compared to the corresponding normal dose of each when administered alone. In some embodiments, both the taxane in the nanoparticle composition and the other chemotherapeutic agent are administered at a subtherapeutic, i.e., reduced, level. In some embodiments, the dose of the nanoparticle composition and/or the other chemotherapeutic agent is substantially less than the established maximum toxic dose (MTD). For example, the dose of the nanoparticle composition and/or the other chemotherapeutic agent is less than about 50%, 40%, 30%, 20%, or 10% of the MTD.
  • Metronomic Therapy Regimes
  • The present invention also provides metronomic therapy regimes for any of the methods of treatment and methods of administration described herein. Exemplary metronomic therapy regimes and embodiments for the use of metronomic therapy regimes are discussed below and disclosed in U.S. Ser. No. 11/359,286, filed Feb. 21, 2006, published as U.S. Pub. No. 2006/0263434 (such as those described in paragraphs [0138] to [0157]), which is hereby incorporated by reference in its entirety. In some embodiments, the nanoparticle composition is administered over a period of at least one month, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 0.25% to about 25% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the nanoparticle composition is administered over a period of at least two months, wherein the interval between each administration is no more than about a week, and wherein the dose of the taxane (e.g., paclitaxel) at each administration is about 1% to about 20% of its maximum tolerated dose following a traditional dosing regime. In some embodiments, the dose of a taxane (e.g., paclitaxel) per administration is less than about any of 25%, 24%, 23%, 22%, 20%, 18%, 15%, 14%, 13%, 12%, 11%, 10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, or 1% of the maximum tolerated dose. In some embodiments, any nanoparticle composition is administered at least about any of 1×, 2×, 3×, 4×, 5×, 6×, or 7× (i.e., daily) a week. In some embodiments, the intervals between each administration are less than about any of 6 months, 3 months, 1 month, 20 days, 15, days, 12 days, 10 days, 9 days, 8 days, 7 days, 6 days, 5 days, 4 days, 3 days, 2 days, or 1 day. In some embodiments, the intervals between each administration are more than about any of 1 month, 2 months, 3 months, 4 months, 5 months, 6 months, 8 months, or 12 months. In some embodiments, the composition is administered over a period of at least about any of 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 18, 24, 30, 36, 48, 60, 72, or 84 months.
  • Carrier Proteins
  • Provided herein are also compositions comprising nanoparticles that comprise a taxane and a carrier protein for use in methods of treatment, methods of administration, and dosage regimes described herein. In some embodiments, the carrier protein is albumin. In some embodiments, the carrier protein is human serum albumin.
  • Examples of suitable carrier proteins include proteins normally found in blood or plasma, which include, but are not limited to, albumin, immunoglobulin including IgA, lipoproteins, apolipoprotein B, α-acid glycoprotein, β-2-macroglobulin, thyroglobulin, transferin, fibronectin, factor VII, factor VIII, factor IX, factor X, and the like. In some embodiments, the carrier protein is a non-blood protein, such as casein, α-lactalbumin, or β-lactoglobulin. The carrier proteins may either be natural in origin or synthetically prepared. In some embodiments, the pharmaceutical acceptable carrier comprises albumin, such as human serum albumin (HSA). HSA is a highly soluble globular protein of Mr 65K and consists of 585 amino acids. HSA is the most abundant protein in the plasma and accounts for 70-80% of the colloid osmotic pressure of human plasma. The amino acid sequence of HSA contains a total of 17 disulphide bridges, one free thiol (Cys 34), and a single tryptophan (Trp 214). Other albumins are contemplated, such as bovine serum albumin. Use of such non-human albumins could be appropriate, for example, in the context of use of these compositions in non-human mammals, such as the veterinary animals (including domestic pets and agricultural animals).
  • Human serum albumin (HSA) has multiple hydrophobic binding sites (a total of eight for fatty acids, an endogenous ligand of HSA) and binds a diverse set of drugs, especially neutral and negatively charged hydrophobic compounds (Goodman et al., The Pharmacological Basis of Therapeutics, 9th ed, McGraw-Hill New York (1996)). Two high affinity binding sites have been proposed in subdomains IIA and IIIA of HSA, which are highly elongated hydrophobic pockets with charged lysine and arginine residues near the surface which function as attachment points for polar ligand features (see, e.g., Fehske et al., Biochem. Pharmcol., 30, 687-92 (1981), Vorum, Dan. Med. Bull., 46, 379-99 (1999), Kragh-Hansen, Dan. Med. Bull., 1441, 131-40 (1990), Curry et al., Nat. Struct. Biol., 5, 827-35 (1998), Sugio et al., Protein. Eng., 12, 439-46 (1999), He et al., Nature, 358, 209-15 (1992), and Carter et al., Adv. Protein. Chem., 45, 153-203 (1994)).
  • The carrier protein (e.g., albumin) in the composition generally serves as a carrier for the taxane (e.g., paclitaxel) or derivative thereof, i.e., the carrier protein in the composition makes the taxane (e.g., paclitaxel) or derivative thereof more readily suspendable in an aqueous medium or helps maintain the suspension as compared to compositions not comprising a carrier protein. This can avoid the use of toxic solvents for solubilizing of the taxane (e.g., paclitaxel), and thereby can reduce one or more side effects of administration of the taxane (e.g., paclitaxel) into an individual (e.g., human). In some embodiments, the composition is substantially free (e.g. free) of organic solvents or surfactants. A composition is “substantially free of organic solvent” or “substantially free of surfactant” if the amount of organic solvent or surfactant in the composition is not sufficient to cause one or more side effect(s) in an individual when the composition is administered to the individual. In some embodiments, the nanoparticles in the composition have a solid core. In some embodiments, the nanoparticles in the composition have a core that is not aqueous (i.e., other than aqueous core). In some embodiments, the nanoparticles of the composition lack a polymeric matrix. In some embodiments, the nanoparticles of the composition are filter sterilizable. In some embodiments, the nanoparticles in the composition comprise at least one cross-linked carrier protein. In some embodiments, the nanoparticles in the composition comprise at least ten-percent of carrier protein that is cross-linked.
  • The taxane (e.g., paclitaxel) is “stabilized” in an aqueous suspension if it remains suspended in an aqueous medium (e.g., without visible precipitation or sedimentation) for an extended period of time, such as for at least about any of 0.1, 0.2, 0.25, 0.5, 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 24, 36, 48, 60, or 72 hours. The suspension is generally, but not necessarily, suitable for administration to an individual (e.g., human). Stability of the suspension is generally (but not necessarily) evaluated at storage temperature, such as room temperature (e.g., 20-25° C.) or refrigerated conditions (e.g., 4° C.). For example, a suspension is stable at a storage temperature if it exhibits no flocculation or particle agglomeration visible to the naked eye or when viewed under the optical microscope at 1000 times, at about fifteen minutes after preparation of the suspension. Stability can also be evaluated under accelerated testing conditions, such as at a temperature that is higher than about 40° C.
  • In some embodiments, the composition comprises nanoparticles comprising (in various embodiments consisting essentially of) a taxane (e.g., paclitaxel) and a carrier protein. When the taxane (e.g., paclitaxel) is in a liquid form, the particles or nanoparticles are also referred to as droplets or nanodroplets. In some embodiments, taxane is coated with the carrier protein. Particles (such as nanoparticles) of poorly water soluble pharmaceutical agents have been disclosed in, for example, U.S. Pat. Nos. 5,916,596; 6,506,405; and 6,537,579 and also in U.S. Pat. App. Pub. No. 2005/0004002A1.
  • The amount of carrier protein in the composition described herein will vary depending on the taxane (e.g., paclitaxel) or derivative thereof, and other components in the composition. In some embodiments, the composition comprises a carrier protein in an amount that is sufficient to stabilize the taxane (e.g., paclitaxel) in an aqueous suspension, for example, in the form of a stable colloidal suspension (e.g., a stable suspension of nanoparticles). In some embodiments, the carrier protein is in an amount that reduces the sedimentation rate of the taxane (e.g., paclitaxel) in an aqueous medium. The amount of the carrier protein also depends on the size and density of particles of the taxane (e.g., paclitaxel).
  • In some embodiments of any of the aspects of the invention, the taxanes in the nanoparticles are coated with a carrier protein, such as albumin (e.g., human serum albumin). In some embodiments, the taxane is paclitaxel. In various embodiments, the composition comprises more than about any of 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the taxane or derivative thereof in nanoparticle form. In some embodiments, the taxane or derivative thereof constitutes more than about any of 50%, 60%, 70%, 80%, 90%, 95%, or 99% of the nanoparticles by weight. In some embodiments, the nanoparticles are substantially free of polymeric core materials. In some embodiments, the taxane in the nanoparticles is amorphous. In some embodiments, the taxane used for making the nanoparticle compositions is in anhydrous form. In some embodiments, the carrier protein (such as albumin) to a taxane weight ratio in the taxane nanoparticle composition is about any of 18:1 or less, 15:1 or less, 14:1 or less, 13:1 or less, 12:1 or less, 11:1 or less, 10:1 or less, 9:1 or less, 8:1 or less, 7.5:1 or less, 7:1 or less, 6:1 or less, 5:1 or less, 4:1 or less, or 3:1 or less. In some embodiments, the albumin to taxane weight ratio is between about any of 18:1 to 1:18, 10:1 to 1:10, 9:1 to 1:1, 8:1 to 1:1, 7.5:1 to 1.1, 7.1:1 to 1:1, 6:1 to 1:1, 5:1 to 1:1, 4:1 to 1:1, or 3:1 to 1:1. In some embodiments, the composition comprises a stable aqueous suspension of particles (e.g., nanoparticles) comprising a taxane and carrier protein (such as albumin, e.g., particles of paclitaxel coated with albumin).
  • In some embodiments, the composition comprises nanoparticles of any shape (e.g., a spherical or non-spherical shape) with an average or mean diameter of no greater than about 1000 nanometers (nm), such as no greater than about any of 900 nm, 800 nm, 700 nm, 600 nm, 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm, or 50 nm. In some embodiments, the average or mean diameter of the particles is no greater than about 200 nm. In some embodiments, the composition is sterile filterable. In some embodiments, the composition is sterile filtered. In some embodiments, the average or mean diameter of the particles is no greater than about 100 nm. In some embodiments, the average or mean diameter of the particles is between about 20 to about 400 run. In some embodiments, the average or mean diameter of the particles is between about 40 to about 200 nm. In some embodiments, the particles are sterile-filterable. In some embodiments, the composition comprises nanoparticles with a diameter of about any of 1000 nm, 900 nm, 800 nm, 700 nm, 600 nm, 500 nm, 400 nm, 300 nm, 200 nm, 100 nm, 90 nm, 80 nm, 70 nm, 60 nm, or 50 nm. In some embodiments, the composition comprises nanoparticles with a diameter of between about any of 50 nm and 150 nm, 50 nm and 75 nm, 50 nm and 100 nm, 75 nm and 100 nm, 100 nm and 125 nm, 125 nm and 150 nm, 100 nm and 150 nm, 150 nm and 175 nm, or 175 nm and 200 nm. In some embodiments, the diameter of about any of 50% or more, 65% or more, 75% or more, 80% or more, 90% or more, 95% or more, 98% or more, 99% or more, 99.5% or more, or 99.9% or more of the nanoparticles can fall within the range specified. Average or mean diameters of nanoparticles can be determined by methods known in the art, for example laser light scattering.
  • The nanoparticles described herein may be present in a dry formulation (e.g., lyophilized composition) or suspended in a biocompatible medium. Suitable biocompatible media include, but are not limited to, water, buffered aqueous media, saline, buffered saline, optionally buffered solutions of amino acids, optionally buffered solutions of proteins, optionally buffered solutions of sugars, optionally buffered solutions of vitamins, optionally buffered solutions of synthetic polymers, lipid-containing emulsions, and the like.
  • In some embodiments, the nanoparticles do not comprise a blood-insoluble gas or do not comprise gas-filled microbubbles.
  • In some embodiments, the carrier protein is present in an effective amount to reduce one or more side effects associated with administration of taxane to a human compared to compositions without carrier protein. These side effects include, but are not limited to, myelosuppression, neurotoxicity, hypersensitivity, inflammation, venous irritation, phlebitis, pain, skin irritation, neutropenic fever, anaphylactic reaction, hematologic toxicity, and cerebral or neurologic toxicity, and combinations thereof. In some embodiments, there is provided a method of reducing hypersensitivity reactions associated with administration of the taxane (e.g., paclitaxel), including, for example, severe skin rashes, hives, flushing, dyspnea, tachycardia, pulmonary hypertension (e.g., lymphoma); chest pain; black, tarry stools; general feeling of illness, shortness of breath; swollen glands; weight loss; yellow skin and eyes, abdominal pain; unexplained anxiousness; bloody or cloudy urine; bone pain; chills; confusion; convulsions (seizures); cough; decreased urge to urinate; fast, slow, or irregular heartbeat; fever; frequent urge to urinate; increased thirst; loss of appetite; lower back or side pain; mood changes; muscle pain or cramps; nausea or vomiting; numbness or tingling around lips, hands, or feet; painful or difficult urination; rash; sore throat; sores or white spots on lips or in mouth; swelling of hands, ankles, feet, or lower legs; swollen glands; trouble breathing; unusual bleeding or bruising; unusual tiredness or weakness; weakness or heaviness of legs, skin ulcer or sores, weight gain, acne; constipation; diarrhea; difficulty in moving; headache; loss of energy or weakness; muscle pain or stiffness; pain; shaking or trembling; trouble sleeping; nosebleed; and/or swelling of the face. These side effects, however, are merely exemplary and other side effects, or combination of side effects, associated with the taxane (e.g., paclitaxel) can be reduced. The side effects may be immediate or delayed (such as not occurring for a few days, weeks, months, or years after treatment begins).
  • Antimicrobial Agents in Compositions
  • In some embodiments, the compositions of the invention also includes an antimicrobial agent (e.g., an agent in addition to the taxane (e.g., paclitaxel)) in an amount sufficient to significantly inhibit (e.g., delay, reduce, slow, and/or prevent) microbial growth in the composition for use in the methods of treatment, methods of administration, and dosage regimes described herein. Exemplary microbial agents and variations for the use of microbial agents are disclosed in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006 (such as those described in paragraphs [0036] to [0058]). In some embodiments, the antimicrobial agent is a chelating agent, such as EDTA, edetate, citrate, pentetate, tromethamine, sorbate, ascorbate, derivatives thereof; or mixtures thereof. In some embodiments, the antimicrobial agent is a polydentate chelating agent. In some embodiments, the antimicrobial agent is a non-chelating agent, such as any of sulfites, benzoic acid, benzyl alcohol, chlorobutanol, and paraben. In some embodiments, an antimicrobial other than the taxane discussed above is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • Sugar Containing Compositions
  • In some embodiments, the compositions of the invention include a sugar for use in the methods of treatment described herein. In some embodiments, the compositions of the invention include both a sugar and an antimicrobial agent for use in the methods of treatment described herein. Exemplary sugars and variations for the use of sugars are disclosed in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006 (such as those described in paragraphs [0084] to [0090]). In some embodiments, the sugar serves as a reconstitution enhancer which causes a lyophilized composition to dissolve or suspend in water and/or aqueous solution more quickly than the lyophilized composition would dissolve without the sugar. In some embodiments, the composition is a liquid (e.g., aqueous) composition obtained by reconstituting or resuspending a dry composition. In some embodiments, the concentration of sugar in the composition is greater than about 50 mg/ml. In some embodiments, the sugar is in an amount that is effective to increase the stability of the taxane (e.g., paclitaxel) or derivative thereof in the composition as compared to a composition without the sugar. In some embodiments, the sugar is in an amount that is effective to improve filterability of the composition as compared to a composition without the sugar.
  • The sugar-containing compositions described herein may further comprise one or more antimicrobial agents, such as the antimicrobial agents described herein or in U.S. Ser. No. 11/514,030, filed Aug. 30, 2006. In addition to one or more sugars, other reconstitution enhancers (such as those described in U.S. Pat. App. Publication No. 2005/0152979, which is hereby incorporated by reference in its entirety) can also be added to the compositions. In some embodiments, a sugar is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • Stabilizing Agents in Compositions
  • In some embodiments, the compositions of the invention also include a stabilizing agent for use in the methods of treatment, methods of administration, and dosage regimes described herein. In some embodiments, the compositions of the invention include an antimicrobial agent and/or a sugar and/or a stabilizing agent for use in the methods of treatment, methods of administration, and dosage regimes described herein. Exemplary stabilizing agents and variations for the use of stabilizing agents are disclosed in U.S. Ser. No. 11/513,756, filed Aug. 30, 2006 (such as those described in paragraphs [0038] to [0083] and [0107] to [0114]). The present invention in another variation provides for compositions and methods of preparation of a taxane (e.g., paclitaxel) which retain the desirable therapeutic effects and remain physically and/or chemically stable upon exposure to certain conditions such as prolonged storage, elevated temperature, or dilution for parenteral administration. The stabilizing agent includes, for example, chelating agents (e.g., citrate, malic acid, edetate, or pentetate), sodium pyrophosphate, and sodium gluconate. In some embodiments, the invention provides pharmaceutical formulations of a taxane (e.g., paclitaxel) comprising citrate, sodium pyrophosphate, EDTA, sodium gluconate, citrate and sodium chloride, and/. In another variation, the invention provides a composition of a taxane, wherein the taxane (e.g., paclitaxel) used for preparing the formulation is in an anhydrous form prior to being incorporated into the composition.
  • In some embodiments, a stabilizing agent is not contained or used in the methods of treatment, methods of administration, and dosage regimes described herein.
  • Pharmaceutical Compositions and Formulations
  • The compositions described herein may be used in the preparation of a formulation, such as a pharmaceutical formulation, by combining the nanoparticle composition(s) described with a pharmaceutical acceptable carrier, excipients, stabilizing agents or other agent, which are known in the art, for use in the methods of treatment, methods of administration, and dosage regimes described herein. To increase stability by increasing the negative zeta potential of nanoparticles, certain negatively charged components may be added. Such negatively charged components include, but are not limited to bile salts, bile acids, glycocholic acid, cholic acid, chenodeoxycholic acid, taurocholic acid, glycochenodeoxycholic acid, taurochenodeoxycholic acid, litocholic acid, ursodeoxycholic acid, dehydrocholic acid, and others; phospholipids including lecithin (egg yolk) based phospholipids which include the following phosphatidylcholines: palmitoyloleoylphosphatidylcholine, palmitoyllinoleoylphosphatidylcholine, stearoyllinoleoylphosphatidylcholine, stearoyloleoylphosphatidylcholine, stearoylarachidoylphosphatidylcholine, and dipalmitoylphosphatidylcholine. Other phospholipids including L-α-dimyristoylphosphatidylcholine (DMPC), dioleoylphosphatidylcholine (DOPC), distearoylphosphatidylcholine (DSPC), hydrogenated soy phosphatidylcholine (HSPC), and other related compounds. Negatively charged surfactants or emulsifiers are also suitable as additives, e.g., sodium cholesteryl sulfate and the like.
  • In some embodiments, the composition is suitable for administration to a human. There are a wide variety of suitable formulations of the inventive composition (see, e.g., U.S. Pat. Nos. 5,916,596 and 6,096,331, which are hereby incorporated by reference in their entireties). The following formulations and methods are merely exemplary and are in no way limiting. Formulations suitable for oral administration can comprise (a) liquid solutions, such as an effective amount of the compound dissolved in diluents, such as water, saline, or orange juice, (b) capsules, sachets or tablets, each containing a predetermined amount of the active ingredient, as solids or granules, (c) suspensions in an appropriate liquid, (d) suitable emulsions, and (e) powders. Tablet forms can include one or more of lactose, mannitol, corn starch, potato starch, microcrystalline cellulose, acacia, gelatin, colloidal silicon dioxide, croscarmellose sodium, talc, magnesium stearate, stearic acid, and other excipients, colorants, diluents, buffering agents, moistening agents, preservatives, flavoring agents, and pharmacologically compatible excipients. Lozenge forms can comprise the active ingredient in a flavor, usually sucrose and acacia or tragacanth, as well as pastilles comprising the active ingredient in an inert base, such as gelatin and glycerin, or sucrose and acacia, emulsions, gels, and the like containing, in addition to the active ingredient, such excipients as are known in the art.
  • Formulations suitable for parenteral administration include aqueous and non-aqueous, isotonic sterile injection solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes that render the formulation compatible with the blood of the intended recipient, and aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizing agents, and preservatives. The formulations can be presented in unit-dose or multi-dose sealed containers, such as ampules and vials, and can be stored in a freeze-dried (lyophilized) condition requiring only the addition of the sterile liquid excipient methods of treatment, methods of administration, and dosage regimes described herein (i.e., water) for injection, immediately prior to use. Extemporaneous injection solutions and suspensions can be prepared from sterile powders, granules, and tablets of the kind previously described. Injectable formulations are preferred.
  • The invention also includes formulations of nanoparticle compositions comprising the taxane (e.g., paclitaxel) or derivative thereof and a carrier suitable for administration by inhalation for use in the methods of the invention. Formulations suitable for aerosol administration comprise the inventive composition include aqueous and non-aqueous, isotonic sterile solutions, which can contain anti-oxidants, buffers, bacteriostats, and solutes, as well as aqueous and non-aqueous sterile suspensions that can include suspending agents, solubilizers, thickening agents, stabilizing agents, and preservatives, alone or in combination with other suitable components, which can be made into aerosol formulations to be administered via inhalation. These aerosol formulations can be placed into pressurized acceptable propellants, such as dichlorodifluoromethane, propane, nitrogen, and the like. They also can be formulated as pharmaceuticals for non-pressured preparations, such as in a nebulizer or an atomizer.
  • In some embodiments, the composition is formulated to have a pH in the range of about 4.5 to about 9.0, including for example pH ranges of any of about 5.0 to about 8.0, about 6.5 to about 7.5, and about 6.5 to about 7.0. In some embodiments, the pH of the composition is formulated to no less than about 6, including for example no less than about any of 6.5, 7, or 8 (e.g., about 8). The composition can also be made to be isotonic with blood by the addition of a suitable tonicity modifier, such as glycerol.
  • The nanoparticles of this invention can be enclosed in a hard or soft capsule, can be compressed into tablets, or can be incorporated with beverages or food or otherwise incorporated into the diet. Capsules can be formulated by mixing the nanoparticles with an inert pharmaceutical diluent and inserting the mixture into a hard gelatin capsule of the appropriate size. If soft capsules are desired, a slurry of the nanoparticles with an acceptable vegetable oil, light petroleum or other inert oil can be encapsulated by machine into a gelatin capsule.
  • Also provided are unit dosage forms comprising the compositions and formulations described herein. These unit dosage forms can be stored in a suitable packaging in single or multiple unit dosages and may also be further sterilized and sealed. For example, the pharmaceutical composition (e.g., a dosage or unit dosage form of a pharmaceutical composition) may include (i) nanoparticles that comprise a taxane (e.g., paclitaxel) and a carrier protein and (ii) a pharmaceutically acceptable carrier. In some embodiments, the pharmaceutical composition also includes one or more other compounds (or pharmaceutically acceptable salts thereof) that are useful for treating cancer. In various embodiments, the amount of a taxane (e.g., paclitaxel) in the composition is included in any of the following ranges: about 5 to about 50 mg, about 20 to about 50 mg, about 50 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg. In some embodiments, the amount of a taxane (e.g., paclitaxel) or derivative thereof in the composition (e.g., a dosage or unit dosage form) is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg, of the taxane (e.g., paclitaxel) or derivative thereof. In some embodiments, the carrier is suitable for parental administration (e.g., intravenous administration). In some embodiments, the taxane (e.g., paclitaxel) or derivative thereof is the only pharmaceutically active agent for the treatment of cancer that is contained in the composition.
  • In some embodiments, the invention features a dosage form (e.g., a unit dosage form) for the treatment of cancer comprising (i) nanoparticles that comprise a carrier protein and a taxane (e.g., paclitaxel), wherein the amount of a taxane (e.g., paclitaxel) or derivative thereof in the unit dosage from is in the range of about 5 mg to about 500 mg, and (ii) a pharmaceutically acceptable carrier. In some embodiments, the amount of the taxane (e.g., paclitaxel) or derivative thereof in the unit dosage form includes about 30 mg to about 300 mg.
  • Also provided are articles of manufacture comprising the compositions, formulations, and unit dosages described herein in suitable packaging for use in the methods of treatment, methods of administration, and dosage regimes described herein. Suitable packaging for compositions described herein are known in the art, and include, for example, vials (such as sealed vials), vessels (such as sealed vessels), ampules, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. These articles of manufacture may further be sterilized and/or sealed.
  • Kits
  • The invention also provides kits comprising the compositions, formulations, unit dosages, and articles of manufacture described herein for use in the methods of treatment, methods of administration, and dosage regimes described herein. Kits of the invention include one or more containers comprising a taxane (e.g., paclitaxel)-containing nanoparticle compositions (formulations or unit dosage forms and/or articles of manufacture), and in some embodiments, further comprise instructions for use in accordance with any of the methods of treatment described herein. In some embodiments, the kit comprises i) a composition comprising nanoparticles comprising a taxane (e.g., paclitaxel) and a carrier protein (such as albumin) and ii) instructions for administering the nanoparticles and the other chemotherapeutic agents simultaneously and/or sequentially, for treatment of cancer. In various embodiments, the amount of a taxane (e.g., paclitaxel) in the kit is included in any of the following ranges: about 5 mg to about 20 mg, about 20 to about 50 mg, about 50 to about 100 mg, about 100 to about 125 mg, about 125 to about 150 mg, about 150 to about 175 mg, about 175 to about 200 mg, about 200 to about 225 mg, about 225 to about 250 mg, about 250 to about 300 mg, about 300 to about 350 mg, about 350 to about 400 mg, about 400 to about 450 mg, or about 450 to about 500 mg. In some embodiments, the amount of a taxane (e.g., paclitaxel) in the kit is in the range of about 5 mg to about 500 mg, such as about 30 mg to about 300 mg or about 50 mg to about 200 mg. In some embodiments, the kit includes one or more other compounds (i.e., one or more compounds other than a taxane) that are useful for cancer.
  • Instructions supplied in the kits of the invention are typically written instructions on a label or package insert (e.g., a paper sheet included in the kit), but machine-readable instructions (e.g., instructions carried on a magnetic or optical storage disk) are also acceptable. The instructions relating to the use of the nanoparticle compositions generally include information as to dosage, dosing schedule, and route of administration for the intended treatment. The kit may further comprise a description of selecting an individual suitable or treatment.
  • The present invention also provides kits comprising compositions (or unit dosages forms and/or articles of manufacture) described herein and may further comprise instruction(s) on methods of using the composition, such as uses further described herein. In some embodiments, the kit of the invention comprises the packaging described above. In other embodiments, the kit of the invention comprises the packaging described above and a second packaging comprising a buffer. It may further include other materials desirable from a commercial and user standpoint, including other buffers, diluents, filters, needles, syringes, and package inserts with instructions for performing any methods described herein.
  • For combination therapies of the invention, the kit may contain instructions for administering the first and second therapies simultaneously and/or sequentially for the effective treatment of cancer. The first and second therapies can be present in separate containers or in a single container. It is understood that the kit may comprise one distinct composition or two or more compositions wherein one composition comprises a first therapy and one composition comprises a second therapy.
  • Kits may also be provided that contain sufficient dosages of the taxane (e.g., paclitaxel) as disclosed herein to provide effective treatment for an individual for an extended period, such as any of a week, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 3 months, 4 months, 5 months, 6 months, 7 months, 8 months, 9 months or more. Kits may also include multiple unit doses of the taxane (e.g., paclitaxel) compositions, pharmaceutical compositions, and formulations described herein and instructions for use and packaged in quantities sufficient for storage and use in pharmacies, for example, hospital pharmacies and compounding pharmacies. In some embodiments, the kit comprises a dry (e.g., lyophilized) composition that can be reconstituted, resuspended, or rehydrated to form generally a stable aqueous suspension of nanoparticles comprising a taxane (e.g., paclitaxel) and albumin (e.g., a taxane (e.g., paclitaxel) coated with albumin).
  • The kits of the invention are in suitable packaging. Suitable packaging include, but is not limited to, vials, bottles, jars, flexible packaging (e.g., sealed Mylar or plastic bags), and the like. Kits may optionally provide additional components such as buffers and interpretative information.
  • Methods of Making the Compositions
  • Methods of making compositions containing carrier proteins and poorly water soluble pharmaceutical agents are known in the art. For example, nanoparticles containing poorly water soluble pharmaceutical agents and carrier proteins (e.g., albumin) can be prepared under conditions of high shear forces (e.g., sonication, high pressure homogenization, or the like). These methods are disclosed in, for example, U.S. Pat. Nos. 5,916,596; 6,506,405; and 6,537,579 and also in U.S. Pat. Pub. No. 2005/0004002A1, which are each hereby incorporated by reference in their entireties.
  • Briefly, the taxane (e.g., paclitaxel) is dissolved in an organic solvent. Suitable organic solvents include, for example, ketones, esters, ethers, chlorinated solvents, and other solvents known in the art. For example, the organic solvent can be methylene chloride, chloroform/ethanol, or chloroform/t-butanol (for example with a ratio of about any of 1:9, 1:8, 1:7, 1:6, 1:5, 1:4, 1:3, 1:2, 1:1, 2:1, 3:1, 4:1, 5:1, 6:1, 7:1, 8:1, or 9:1 or with a ratio of about any of 3:7, 5:7, 4:6, 5:5, 6:5, 8:5, 9:5, 9.5:5, 5:3, 7:3, 6:4, or 9.5:0.5). The solution is added to a carrier protein (e.g., human serum albumin). The mixture is subjected to high pressure homogenization (e.g., using an Avestin, APV Gaulin, Microfluidizer™ such as a Microfluidizer™ Processor M-110EH from Microfluidics, Stansted, or Ultra Turrax homogenizer). The emulsion may be cycled through the high pressure homogenizer for between about 2 to about 100 cycles, such as about 5 to about 50 cycles or about 8 to about 20 cycles (e.g., about any of 8, 10, 12, 14, 16, 18 or 20 cycles). The organic solvent can then be removed by evaporation utilizing suitable equipment known for this purpose, including, but not limited to, rotary evaporators, falling film evaporators, wiped film evaporators, spray driers, and the like that can be operated in batch mode or in continuous operation. The solvent may be removed at reduced pressure (such as at about any of 25 mm Hg, 30 mm Hg, 40 mm Hg, 50 mm Hg, 100 mm Hg, 200 mm Hg, or 300 mm Hg). The amount of time used to remove the solvent under reduced pressure may be adjusted based on the volume of the formulation. For example, for a formulation produced on a 300 mL scale, the solvent can be removed at about 1 to about 300 mm Hg (e.g., about any of 5-100 mm Hg, 10-50 mm Hg, 20-40 mm Hg, or 25 mm Hg) for about 5 to about 60 minutes (e.g., about any of 7, 8, 9, 10, 11, 12, 13, 14, 15 16, 18, 20, 25, or 30 minutes).
  • If desired, human albumin solution may be added to the dispersion to adjust the human serum albumin to the taxane (e.g., paclitaxel) ratio or to adjust the concentration of the taxane (e.g., paclitaxel) in the dispersion. For example, human serum albumin solution (e.g., 25% w/v) can be added to adjust the human serum albumin to a taxane (e.g., paclitaxel) ratio to about any of 18:1, 15, :1 14:1, 13:1, 12:1, 11:1, 10:1, 9:1, 8:1, 7.5:1, 7:1, 6:1, 5:1, 4:1, or 3:1. For example, human serum albumin solution (e.g., 25% w/v) or another solution is added to adjust the concentration of a taxane (e.g., paclitaxel) in the dispersion to about any of 0.5 mg/ml, 1.3 mg/ml, 1.5 mg/ml, 2 mg/ml, 3 mg/ml, 4 mg/ml, 5 mg/ml, 6 mg/ml, 7 mg/ml, 8 mg/ml, 9 mg/ml, 10 mg/ml, 15 mg/ml, 20 mg/ml, 25 mg/ml, 30 mg/ml, 40 mg/ml, or 50 mg/ml. The dispersion may be serially filtered through multiple filters, such as a combination of 1.2 μm and 0.8/0.2 μm filters; the combination of 1.2 μm, 0.8 μm, 0.45 μm, and 0.22 μm filters; or the combination of any other filters known in the art. The dispersion obtained can be further lyophilized. The nanoparticle compositions may be made using a batch process or a continuous process (e.g., the production of a composition on a large scale).
  • If desired, a second therapy (e.g., one or more compounds useful for treating cancer), an antimicrobial agent, sugar, and/or stabilizing agent can also be included in the composition. This additional agent can either be admixed with the taxane (e.g., paclitaxel) and/or the carrier protein during preparation of the taxane (e.g., paclitaxel)/carrier protein composition, or added after the taxane (e.g., paclitaxel)/carrier protein composition is prepared. In some embodiments, the agent is admixed with the taxane (e.g., paclitaxel)/carrier protein composition prior to lyophilization. In some embodiments, the agent is added to the lyophilized pharmaceutical agent/carrier protein composition. In some embodiments when the addition of the agent changes the pH of the composition, the pH in the composition are generally (but not necessarily) adjusted to a desired pH. Exemplary pH values of the compositions include, for example, in the range of about 5 to about 8.5. In some embodiments, the pH of the composition is adjusted to no less than about 6, including for example no less than any of about 6.5, 7, or 8 (e.g., about 8).
  • Unless defined otherwise, the meanings of all technical and scientific terms used herein are those commonly understood by one of skill in the art to which this invention belongs. One of skill in the art will also appreciate that any methods and materials similar or equivalent to those described herein can also be used to practice or test the invention.
  • The following Examples are provided to illustrate, but not limit, the invention.
  • EXAMPLES
  • The examples, which are intended to be purely exemplary of the invention and should therefore not be considered to limit the invention in any way, also describe and detail aspects and variations of the invention discussed above. Efforts have been made to ensure accuracy with respect to numbers used (for example, amounts, temperature, etc.) but some experimental errors and deviations should be accounted for. Unless indicated otherwise, parts are parts by weight, molecular weight is weight average molecular weight, temperature is in degrees Centigrade, and pressure is at or near atmospheric.
  • Example 1 Exemplary Method for the Formation of Nanoparticle Compositions with Paclitaxel and Albumin
  • This example provides formulations of paclitaxel/albumin. The compositions were prepared essentially as described in U.S. Pat. Nos. 5,439,686 and 5,916,596. Briefly, paclitaxel was dissolved in an organic solvent (such as methylene chloride or a chloroform/ethanol mixture), and the solution was added to a human serum albumin solution. The mixture was homogenized for 5 minutes at low RPM to form a crude emulsion, and then transferred into a high pressure homogenizer. The emulsification was performed at 9000-40,000 psi while recycling the emulsion for at least 5 cycles. The resulting system was transferred into a rotary evaporator, and the organic solvent was rapidly removed at 40° C., at reduced pressure (30 mm Hg) for 20-30 minutes. The dispersion was then further lyophilized for 48 hours. The resulting cake was reconstituted to the original dispersion by addition of sterile water or saline, which may optionally contain additional antimicrobial agent(s).
  • Example 2 Evaluation of Efficacy of Nanoparticle Albumin Bound Paclitaxel (Nab-Paclitaxel) in Treating Platinum-Sensitive Patients with Recurrent Ovarian, Peritoneal, or Fallopian Tube Cancer
  • The primary objective of this study was to determine the objective response rate in platinum-sensitive patients with recurrent ovarian, peritoneal, or fallopian tube cancer. The study also evaluated the progression free survival, overall survival, quality of life during treatment, and the safety and toxicity of the treatment in this patient population.
  • Patient Population Evaluated
  • Patients were eligible for inclusion for inclusion in this study if she met all of the following criteria: had histologically or cytologically confirmed epithelial cancer of the ovary, fallopian tube or peritoneum (any stage, grade 2-3 if stage 1), had measurable disease by RECIST or elevated CA-125 (>70) in the absence of measurable disease, had received prior platinum-based chemotherapy, was considered platinum sensitive (i.e., having a treatment-free interval >6 months since completion of platinum base chemotherapy), had an ECOG Performance Status (PS) 0-2, had a present, existing peripheral neuropathy which was less than or equal to Grade 1, and had signed an Institutional Review Board approved informed consent. Patients were excluded from this study if she met any of the following criteria: had previously untreated stage 1, Grade 1 disease, was chemotherapy-naïve, had received more than one prior regimen or prior regimen that was not platinum-based, had non-epithelial disease, had nonmeasurable disease with CA-125 less than or equal to 70, had received a taxane within 6 months of registration or any prior treatment with Nab-paclitaxel.
  • Treatment Schedule
  • Premedication was administered at the discretion of the treating physician. Patients received 260 mg/m2 of Nab-paclitaxel (Abraxane®) administered IV during a 30-minute period on Day 1 of each 21-day cycle. Successive cycles were initiated every 3 weeks and were continued until there was evidence of disease progression, unacceptable toxicity, or until 6 cycles. In patients whose only disease measure was an elevated CA-125, up to 3 cycles of treatment were to be administered at the discretion of the treating physician before response was assessed. Patients who achieved a CR could have received an additional 2 cycles at the discretion of the treating physician therefore, CR patients were eligible to receive a maximum of 8 cycles.
  • Assessments
  • Baseline: Complete medical history and physical exam, assessment of ECOG performance status and peripheral neuropathy, CBC, CMP, CA-125, pregnancy test (if appropriate), clinical and radiological assessment of the sites of disease, urinalysis, ECG, and completion FACT-O quality of life questionnaire was determined.
  • During treatment (prior to the start of each cycle): brief medical history and physical exam, assessment of ECOG performance status and peripheral neuropathy, CBC, CMP, CA-125, pregnancy test (if appropriate), clinical and radiological assessment of tumor response, assessment of other sites of disease, toxicity assessments, and completion FACT-O quality of life questionnaire. These same assessments were made at the time that patients went off study treatment and every 3 months for a total of 18 months after the last dose. Responses were assessed by RECIST criteria, CA-125 criteria, or both. See Therasse P. et al., J. NCI 95:205-16 (2000). Response assessment using RECIST followed the commonly applied definitions.
  • Using CA-125, a complete response was defined as a return to a normal range value for at least 28 days from the pretreatment value. See Rustin et al. Clin. Cancer Res. 10(11):3919-26 (2004). A partial response was defined as a sustained over 50% reduction from the pretreatment value. Stable disease was defined as a sustained greater than 50% increase in CA-125 over 28 days in the absence of any new clinically measurable disease, after an adequate trial of therapy. Progressive disease was defined as a sustained over 50% increase in CA-125 or development of new clinically measurable disease after an adequate trial of therapy. An increase was measured from the nadir (the lowest CA-125 value since enrollment).
  • Patients who discontinued due to adverse events, PD, noncompliance, etc. were followed only for survival, additional therapy, site and date of relapse or progression. Adverse event data were collected for 30 days following last dose.
  • Patient Characteristics
  • The majority of enrolled patients had recurrence of disease great than 12 months since prior platinum based chemotherapy.
  • TABLE 1
    Patient characteristics.
    Number of Subjects Enrolled 47
    Age (Years)
    Median 65.4  
    Range 42-84
    Number and Percentage
    (%) of Subjects
    Race
    White 41 (87.2)
    Black 1 (2.1)
    Hispanic 3 (6.4)
    Asian 1 (2.1)
    Indian 1 (2.1)
    ECOG Performance Status*
    0 38 (80.8)
    1  9 (19.2)
    Prior Therapy**
    Prior Chemotherapy greater than or equal to 43 (91.5)
    2 months
    Prior Chemotherapy less than or equal to 12 4 (8.5)
    months
    Surgery 44 (93.6)
    Site of Primary Disease
    Epithelial ovarian 37 (78.7)
    Fallopian tube 1 (2.1)
    Peritoneum  9 (19.2)
    Histological Grade
    GX (can not be assessed) 1 (2.1)
    G1 (well differentiated) 4 (8.5)
    G2 (moderately differentiated)  9 (19.2)
    G3 (poorly differentiated) 23 (48.9)
    Missing/Unknown 10 (21.3)
    *Assessed prior to first treatment
    **Subjects may not have more than one prior chemotherapy regimen.
  • Status
  • TABLE 2
    Patient Status.
    Total Number of Subjects 47
    Number and Percentage (%)
    of Subjects
    Status
    Alive 44 (93.6)
    Dead 3 (6.4)
    Cause of Death
    Progressive disease
    3  
    Reason for Discontinuation
    Normal Completion 30 (63.8)
    Toxicity 1 (2.1)
    Patient Request/Withdrew Consent 2 (4.2)
    Investigator decision 3 (6.4)
    Recurrence 10 (21.3)
    Other (Coronary artery bypass surgery) 1 (2.1)
    Total Cycles Received
    Median 6.0
    Range 1-8
  • Response to Treatment
  • Overall response rates were calculated using either RECIST or CA-125 or both, the ORR (CP+PR) was 63.2%. Three patients were not evaluable, two were found to be ineligible after start of treatment, and one patient was never treated. Overall survival and progression free survival were plotted in terms of months (x-axis) to proportion of survival or proportion not progressed (y-axis), respectively. See FIGS. 1 a and 1 b.
  • TABLE 3
    Patient Response
    Total Number of Eligible/Treated Subjects 44
    Best Response N (%) 95% Cl
    Complete Response 14 (31.8) (18.1, 45.6)
    Partial Response 14 (31.8) (18.1, 45.6)
    Stable disease 14 (31.8) (18.1, 45.6)
    SD greater than or equal to 6 6
    months
    SD less than 6 moths 8
    Progressive disease   2 (4.5%)   (0, 10.7)
    Clinical Benefit 34 (77.3)
    Non-evaluable 3
    Time to Response (months)
    Median 1.8
    Range 0.6-3.4 
    Duration of Response
    (months)
    Median 6.5
    Range 2.7-13.2
    95% Cl 6.6, NA
  • TABLE 4
    Best Response.
    By RECIST
    Best Response By RECIST By CA-125 and CA-125
    Complete Response 1 6 7
    Partial Response 4 3 7
    Stable disease 5 2 7
    SD greater than or 3 0 3
    equal to 6 months
    SD less than 6 moths 2 2 4
    Progressive disease 1 0 1
    Clinical Benefit 8 9 17
  • Treatment-Related Toxicity >Grade 3
  • There were 56 patients treated. Grade 3 neuropathy occurred in 8.7% of patients.
  • TABLE 5
    Treatment-related Grade 3-4 Toxicity in greater
    than or equal to 2% of patients
    Grade Grade
    Adverse Event 3 4 Total Total %
    Hematologic
    Leukopenia
    6 0 6 13.0%
    Neutropenia
    6 5 11 23.9%
    Nonhematologic
    Abdominal pain 1 0 1 2.2%
    Diarrhea
    1 0 1 2.2%
    Fatigue
    1 0 1 2.2%
    Neuropathy 4 0 4 8.7%
    Pneumonia
    1 0 1 2.2%
    Upper
    1 0 1 2.2%
    respiratory tract
    infection
    Generalized 1 0 1 2.2%
    weakness
  • TABLE 6
    Incidences of Alopecia
    Alopecia
    Grade
    1  5 (10.8%)
    Grade 2 35 (76.1%)
  • Estimated Quality of Life
  • The Quality of Life (FACT-O) questionnaire was completed at baseline and before each cycle. Overall quality of life measured decreased from baseline at the initiation of therapy. As measured by the questionnaire, the major contributors to the initial decline in quality of life were the functional, social, and physical well being assessments. All measure improved at the completion of therapy and returned to based line.
  • Conclusion
  • Nab-paclitaxel was very active as a single agent in patients with platinum sensitive recurrent ovarian, peritoneal, or fallopian tube cancer. The ORR was 64% and the clinical benefit rate was 77%. Toxicities were tolerable and manageable.
  • Example 3 Evaluation of Efficacy of Nab-Paclitaxel Plus Carboplatin in Treating Patients with Recurrent Platinum-Sensitive Ovarian or Primary Peritoneal Cancer
  • This open-label, non-randomized study was designed to determine the efficacy and safety of nab-paclitaxel (a 130-nm albumin-bound particle form of paclitaxel) plus carboplatin in patients with metastatic ovarian or primary peritoneal carcinoma following platinum-based chemotherapy.
  • Methods
  • Eligible patients had to have either measurable disease (based on RECIST criteria) or pretreatment CA-125 levels >2 times the upper limit of normal in the absence of measurable disease. Patients also had to have good performance status, adequate hepatic/renal function, peripheral neuropathy <grade 1, and life expectancy >6 months. Patients may have received prior chemotherapy for ovarian cancer, including taxane-containing regimens, provided the treatment was completed at least 6 months before enrollment. Nab-Paclitaxel 100 mg/m2 was administered IV over 30 minutes on days 1, 8, and 15 every 28 days. Carboplatin AUC 6 was administered IV over 1-2 hours on day 1 every 28 days. Treatment continued for 6 cycles (or longer in the absence of unacceptable toxicity). Planned sample size was 43 patients (39 evaluable). Efficacy was determined by changes in tumor size for patients with measurable disease or changes in CA-125 levels for those with non-measurable disease.
  • Results
  • To date, 10 patients have been enrolled, 2 of whom completed 2 treatment cycles; both patients had a 50% reduction in their disease (partial response). One patient had grade 4 neutropenia; grade 3 hematologic events were neutropenia (2 patients), thrombocytopenia (3), and anemia (1). Severe headache and severe nausea were reported for 2 patients each; all 4 events required medication.
  • Conclusions
  • Preliminary results indicated that nab-paclitaxel 100 mg/m2 plus carboplatin AUC 6 will have antitumor activity in patients with recurrent platinum-sensitive ovarian or primary peritoneal cancer and that treatment appears to be well tolerated.
  • Example 3B Abraxane® Plus Carboplatin in Patients with Recurrent Platinum-Sensitive Ovarian or Primary Peritoneal Cancer: Evaluation of the Response and Survival and Progression-Free Survival
  • The aim of this study was to evaluate the effectiveness of Abraxane® plus carboplatin in the treatment of patients with recurrent platinum-sensitive ovarian or primary peritoneal cancer.
  • Methods
  • Patients with recurrent platinum-sensitive ovarian or primary peritoneal carcinoma with measurable or biological evidence of disease had received Abraxane® 100 mg/m2 on days 1, 8, and 15 and carboplatin AUC 5 on day 1, every 28 days, intravenously for six cycles. In this study of patients with advanced ovarian cancer, an interim data analysis was conducted of the first 29 of 43 patients who had completed six cycles of treatment. Patients were evaluated with respect to the safety, tolerability, and antitumor effect of intravenous Abraxane® plus carboplatin. Efficacy was measured as changes from baseline in tumor size measured by CT using RECIST Criteria, survival, progression-free survival, and recurrence-free survival during treatment and post study. Safety and tolerability were monitored through adverse events and clinical laboratory values, as well as physical examinations during study drug dosing.
  • Results
  • Twenty-six of 29 patients completed six cycles of chemotherapy. Complete response was attained by 20 of 29 (68.9%) patients. Partial response based on CT evaluation was achieved by four of 29 (13.8%) patients. In one of 29 (3.4%) patients, tumor size increased after the second cycle and then decreased. Two of 29 (6.8%) patients progressed and discontinued treatment: one (1/29, 3.4%) discontinued treatment secondary to carboplatin toxicity and the other (1/29, 3.4%) patient discontinued treatment secondary to severe thrombocytopenia. Abraxane® plus carboplatin can be used to treat recurrent platinum-sensitive ovarian or primary peritoneal cancer effectively.
  • Example 4 Comparison of Pharmacokinetics of Intravenous and Intraperitoneal
  • The purpose of this study was to compare the pharmacokinetics of intravenous and intraperitoneal administration of Nab-paclitaxel. For intravenous administration, 20 rats were dosed with Abraxane® at 50 mg/kg. Dose volume of 10 ml/kg was delivered intravenously via the tail vein. Blood was drawn at the following intervals for LC/MS/MS analysis of paclitaxel: 0.0, 0.083, 0.25, 0.5, 1, 2, 4, 8, 24, 48, 72 hr. For intraperitoneal administration, 3 rats were dosed with Abraxane® at 50 mg/kg. Dose volume of 10 ml/kg was delivered intraperitoneally. Blood was drawn at the following intervals for LC/MS/MS analysis of paclitaxel: 0.0, 0.083, 0.25, 0.5, 1, 2, 4, 8, 24, 48, 72 hr.
  • Results
  • The plasma level of paclitaxel was measured by LC/MS/MS analysis (FIG. 2 and FIG. 3). As indicated in Table 7, intraperitoneal delivery leaded to a slow absorption of paclitaxel into the circulation with tmax occurring at ˜3 hrs. The terminal half-lives of paclitaxel when administered intraperitoneally or intravenously were about the same. Systemic exposure after IP administration was ˜50% of IV (over 72 hrs), indicating substantial local intraperitoneal exposure of drug, especially in the early phase (0-4 hrs) after administration. There may be sustained exposure beyond 72 hrs; however, this study was terminated at 72 hrs.
  • TABLE 7
    Pharmacokinetics Parameters of Intravenous and Intraperitoneal
    Cmax AUC last AUC inf % AUC F
    HL (h) Tmax (hr) (ng/ml) (hr*ng/ml) (hr*ng/ml) Extrapolate (%)
    IP (N = 3) 6.31 ± 0.20 3.33 ± 1.16 3207 ± 717 26233 ± 3848 26770 ± 3904 2.0 ± 1.2 53
    IV (N = 20) 7.96 ± 4.05 0.083 ± 0.000 25060 ± 3573 50038 ± 6134 50146 ± 6137 0.2 ± 0.1
  • Example 5 Intraperitoneal Administration of Abraxane® in Recurrent Mullerian Cancer
  • Intraperitoneal therapy for recurrent diseases is an area that requires further study. The purpose of this experiment is to determine the safety, efficacy, and maximum tolerable dose of Nab-paclitaxel when administered intraperitoneally.
  • Methods
  • A Phase I dose-escalation trial is conducted with 6 patients per cohort to provide sufficient pharmacokinetic data points. Every 4 weeks dosages are escalated so that the cumulative toxicity of weekly treatment may be evaluated. When the MTD is reached, an additional 10 patients are treated to obtain further information about the tolerability at this dose continuously (or ¾ weeks) as well as preliminary information regarding efficacy.
  • Intraperitoneal dosing of Abraxane® begins at 60 mg/m2. Doses is escalated as follows: Dose level 1: 60 mg/m2, Dose level 2: 80 mg/m2, Dose level 3: 100 mg/m2, Dose level 4: 125 mg/m2, and Dose level 5: 150 mg/m2. Ten patients are added at the DLT, and the study is expected to accrue approximately 30-40 patients.
  • Patient Population
  • Patients are included in the study if they meet the following criteria: having recurrent Mullerian cancer (ovarian, peritoneal, fallopian tube, malignant mixed mullerian tumor, serous endo), having disease <1 cm by CT/MRI or SLO, displaying adequate heme/hepatic/renal function, and PS 0-1. Patients are excluded based on meeting the following criteria: having a bowel obstruction (or impending), existing intra-abdominal infection, having significant loculations/adhesions or other contraindications to IP port placement, existing neuropathy >grade 1, having extraperitoneal disease, or displaying an inability to tolerate IV paclitaxel and docetaxel, suggestive of taxane allergy.

Claims (21)

1. A method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein.
2. The method of claim 1, wherein the recurrent ovarian, peritoneal, or fallopian cancer is platinum-sensitive.
3. The method of claim 1, wherein the individual has received a prior platinum-based chemotherapy and has a treatment free interval of more than about 12 months since the completion of the platinum-based chemotherapy.
4. The method of claim 1, wherein the composition comprising nanoparticles comprising a taxane and a carrier protein is administered alone.
5. The method of claim 1, wherein the taxane is paclitaxel.
6. The method of claim 1, wherein the carrier protein is albumin.
7. The method of claim 1, wherein the nanoparticles in the composition have an average or mean diameter of less than about 200 nm.
8. The method of claim 7, wherein the composition comprising nanoparticles comprising a taxane and a carrier protein is Nab-paclitaxel.
9. The method of claim 1, wherein the individual has received a prior chemotherapy and has a treatment free interval for more about 6 months prior to the initiation of the treatment.
10. The method of claim 1, wherein the individual does not show a symptom of hypersensitivity prior to the initiation of the treatment.
11. The method of claim 1, wherein the individual does not show a symptom resulting from the recurrent cancer upon completion of the treatment.
12. The method of claim 1, wherein the individual has a reduced CA-125 level upon completion of the treatment.
13. A method of treating a recurrent ovarian, peritoneal, or fallopian tube cancer in an individual, comprising administering to the individual: a) an effective amount of a composition comprising nanoparticles comprising a taxane and a carrier protein, and b) an effective amount of a platinum-based agent.
14. The method of claim 13, wherein the composition comprising nanoparticles and the platinum-based agent are concurrently administered.
15. The method of claim 13, wherein the platinum-based agent is carboplatin.
16. The method of claim 13, wherein the individual has received a prior platinum-based chemotherapy and has a treatment free interval of more than about 12 months since the completion of the platinum-based chemotherapy.
17. The method of claim 13, wherein the taxane is paclitaxel.
18. The method of claim 13, wherein the carrier protein is albumin.
19. The method of claim 13, wherein the nanoparticles in the composition have an average or mean diameter of less than about 200 nm.
20. The method of claim 19, wherein the composition comprising nanoparticles comprising a taxane and a carrier protein is Nab-paclitaxel.
21-24. (canceled)
US13/781,482 2007-06-01 2013-02-28 Methods and compositions for treating recurrent cancer Abandoned US20140017316A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US13/781,482 US20140017316A1 (en) 2007-06-01 2013-02-28 Methods and compositions for treating recurrent cancer

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
US93275007P 2007-06-01 2007-06-01
PCT/US2008/007024 WO2008150532A1 (en) 2007-06-01 2008-06-02 Methods and compositions for treating recurrent cancer
US60099110A 2010-05-10 2010-05-10
US13/781,482 US20140017316A1 (en) 2007-06-01 2013-02-28 Methods and compositions for treating recurrent cancer

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2008/007024 Continuation WO2008150532A1 (en) 2007-06-01 2008-06-02 Methods and compositions for treating recurrent cancer
US60099110A Continuation 2007-06-01 2010-05-10

Publications (1)

Publication Number Publication Date
US20140017316A1 true US20140017316A1 (en) 2014-01-16

Family

ID=39733105

Family Applications (3)

Application Number Title Priority Date Filing Date
US12/600,991 Active 2030-05-11 US8927019B2 (en) 2007-06-01 2008-06-02 Methods and compositions for treating recurrent cancer
US13/781,482 Abandoned US20140017316A1 (en) 2007-06-01 2013-02-28 Methods and compositions for treating recurrent cancer
US14/550,509 Abandoned US20160158365A9 (en) 2007-06-01 2014-11-21 Methods and compositions for treating recurrent cancer

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US12/600,991 Active 2030-05-11 US8927019B2 (en) 2007-06-01 2008-06-02 Methods and compositions for treating recurrent cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
US14/550,509 Abandoned US20160158365A9 (en) 2007-06-01 2014-11-21 Methods and compositions for treating recurrent cancer

Country Status (12)

Country Link
US (3) US8927019B2 (en)
EP (1) EP2155188B1 (en)
JP (3) JP5579057B2 (en)
AU (1) AU2008260447B2 (en)
CA (1) CA2689914C (en)
DK (1) DK2155188T3 (en)
ES (1) ES2435452T3 (en)
HK (1) HK1140944A1 (en)
HR (1) HRP20131142T1 (en)
PL (1) PL2155188T3 (en)
PT (1) PT2155188E (en)
WO (1) WO2008150532A1 (en)

Cited By (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100048499A1 (en) * 2006-12-14 2010-02-25 Desai Neil P Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US20100215751A1 (en) * 2007-06-01 2010-08-26 Desai Neil P Methods and compositions for treating recurrent cancer
US20110118342A1 (en) * 2005-08-31 2011-05-19 Tapas De Compositions and methods for preparation of poorly water soluble drugs with increased stability
US8846771B2 (en) 2002-12-09 2014-09-30 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US8911786B2 (en) 2007-03-07 2014-12-16 Abraxis Bioscience, Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
US9061014B2 (en) 2011-04-28 2015-06-23 Abraxis Bioscience, Llc Intravascular delivery of nanoparticle compositions and uses thereof
US9101543B2 (en) 2005-02-18 2015-08-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9149455B2 (en) 2012-11-09 2015-10-06 Abraxis Bioscience, Llc Methods of treating melanoma
US9393318B2 (en) 2010-03-29 2016-07-19 Abraxis Bioscience, Llc Methods of treating cancer
US9399071B2 (en) 2010-06-04 2016-07-26 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9446003B2 (en) 2009-04-15 2016-09-20 Abraxis Bioscience, Llc Prion free nanoparticle compositions and methods of making thereof
US9511046B2 (en) 2013-01-11 2016-12-06 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US9561288B2 (en) 2005-02-18 2017-02-07 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9585960B2 (en) 2011-12-14 2017-03-07 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
US9962373B2 (en) 2013-03-14 2018-05-08 Abraxis Bioscience, Llc Methods of treating bladder cancer
US10527604B1 (en) 2015-03-05 2020-01-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and paclitaxel
US10660965B2 (en) 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
US10705070B1 (en) 2015-03-05 2020-07-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and poorly water soluble drug
US10744110B2 (en) 2013-03-12 2020-08-18 Abraxis Bioscience, Llc Methods of treating lung cancer
US10973806B2 (en) 2015-06-29 2021-04-13 Abraxis Bioscience, Llc Methods of treating epithelioid cell tumors comprising administering a composition comprising nanoparticles comprising an mTOR inhibitor and an albumin
US11497737B2 (en) 2019-10-28 2022-11-15 Abraxis Bioscience, Llc Pharmaceutical compositions of albumin and rapamycin
US11944708B2 (en) 2018-03-20 2024-04-02 Abraxis Bioscience, Llc Methods of treating central nervous system disorders via administration of nanoparticles of an mTOR inhibitor and an albumin

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5439686A (en) * 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US8853260B2 (en) 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030199425A1 (en) * 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
US20100166869A1 (en) * 2007-05-03 2010-07-01 Desai Neil P Methods and compositions for treating pulmonary hypertension
AU2009234127B2 (en) * 2008-04-10 2015-04-30 Abraxis Bioscience, Llc Compositions of hydrophobic taxane derivatives and uses thereof
CA2774550A1 (en) * 2009-09-18 2011-03-24 Abraxis Bioscience, Llc Use of the sparc microenvironment signature in the treatment of cancer
MX359413B (en) 2010-03-26 2018-09-27 Abraxis Bioscience Llc Methods of treatment of hepatocellular carcinoma.
US9193782B2 (en) 2010-06-03 2015-11-24 Abraxis Bioscience, Llc Use of the SPARC microenvironment signature in the treatment of cancer
LT2846809T (en) 2012-05-09 2021-01-25 Cantex Pharmaceuticals, Inc. Treatment of myelosuppression
WO2017161387A1 (en) * 2016-03-18 2017-09-21 Reid Christopher Brian Compositions of natural extracts and use thereof in methods for preventing or treating diseases
CN107205979A (en) 2014-12-02 2017-09-26 细胞基因公司 Combination treatment
EP3258941A4 (en) 2015-02-17 2018-09-26 Cantex Pharmaceuticals, Inc. Treatment of cancers and hematopoietic stem cell disorders privileged by cxcl12-cxcr4 interaction
CN108778244B (en) * 2015-09-16 2022-04-01 Dfb索里亚有限责任公司 Delivery of drug nanoparticles and methods of use thereof
CN105412024B (en) 2015-12-14 2018-03-30 广州帝奇医药技术有限公司 Target hydrophobic anticancer drug nanometer formulation and preparation method thereof
US20210069122A1 (en) * 2016-01-27 2021-03-11 Instar Technologies A.S. Oromucosal nanofiber carriers for therapeutic treatment
US11585805B2 (en) 2016-02-19 2023-02-21 Nantcell, Inc. Methods of immunogenic modulation
WO2018038253A1 (en) * 2016-08-26 2018-03-01 哲治 奥野 Fine nano-sized medicinal agent and use thereof
ES2955884T3 (en) 2017-03-15 2023-12-07 Dfb Soria Llc Topical therapy for the treatment of skin malignancies with taxane nanoparticles
WO2019178024A1 (en) 2018-03-16 2019-09-19 Dfb Soria, Llc Topical therapy for the treatment of cervical intraepithelial neoplasia (cin) and cervical cancer using nanoparticles of taxanes
WO2023070023A1 (en) * 2021-10-21 2023-04-27 Faeth Therapeutics, Inc. Combination therapy for pik3ca-associated diseases or disorders

Family Cites Families (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5206018A (en) 1978-11-03 1993-04-27 Ayerst, Mckenna & Harrison, Inc. Use of rapamycin in treatment of tumors
IT1197485B (en) * 1986-10-07 1988-11-30 Boehringer Biochemia Srl PHARMACEUTICAL COMPOSITIONS WITH ANTINEOPLASTIC ACTIVITY
US5540931A (en) 1989-03-03 1996-07-30 Charles W. Hewitt Methods for inducing site-specific immunosuppression and compositions of site specific immunosuppressants
US6753006B1 (en) 1993-02-22 2004-06-22 American Bioscience, Inc. Paclitaxel-containing formulations
DE69433723T3 (en) 1993-02-22 2008-10-30 Abraxis Bioscience, Inc., Los Angeles PROCESS FOR IN VIVO ADMINISTRATION OF BIOLOGICAL SUBSTANCES AND COMPOSITIONS USED THEREFROM
US6749868B1 (en) 1993-02-22 2004-06-15 American Bioscience, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US20070117862A1 (en) 1993-02-22 2007-05-24 Desai Neil P Novel formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US5916596A (en) 1993-02-22 1999-06-29 Vivorx Pharmaceuticals, Inc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
US5665382A (en) 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of pharmaceutically active agents for in vivo delivery
US6096331A (en) 1993-02-22 2000-08-01 Vivorx Pharmaceuticals, Inc. Methods and compositions useful for administration of chemotherapeutic agents
US20030068362A1 (en) 1993-02-22 2003-04-10 American Bioscience, Inc. Methods and formulations for the delivery of pharmacologically active agents
US5650156A (en) 1993-02-22 1997-07-22 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of nutriceuticals and compositions useful therefor
US5665383A (en) 1993-02-22 1997-09-09 Vivorx Pharmaceuticals, Inc. Methods for the preparation of immunostimulating agents for in vivo delivery
US5439686A (en) 1993-02-22 1995-08-08 Vivorx Pharmaceuticals, Inc. Methods for in vivo delivery of substantially water insoluble pharmacologically active agents and compositions useful therefor
US5997904A (en) 1993-02-22 1999-12-07 American Bioscience, Inc. Total nutrient admixtures as stable multicomponent liquids or dry powders and methods for the preparation thereof
US6537579B1 (en) 1993-02-22 2003-03-25 American Bioscience, Inc. Compositions and methods for administration of pharmacologically active compounds
US6528067B1 (en) 1993-02-22 2003-03-04 American Bioscience, Inc. Total nutrient admixtures as stable multicomponent liquids or dry powders and methods for the preparation thereof
US5362478A (en) 1993-03-26 1994-11-08 Vivorx Pharmaceuticals, Inc. Magnetic resonance imaging with fluorocarbons encapsulated in a cross-linked polymeric shell
EP1683517A1 (en) 1996-08-19 2006-07-26 American Bioscience, Inc. Methods for the production of protein particles useful for delivery of pharmacological agents
US8137684B2 (en) 1996-10-01 2012-03-20 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
AU744095B2 (en) * 1997-03-27 2002-02-14 Baker Norton Pharmaceuticals, Inc. Methods and compositions for treatment of ovarian cancer
US8853260B2 (en) 1997-06-27 2014-10-07 Abraxis Bioscience, Llc Formulations of pharmacological agents, methods for the preparation thereof and methods for the use thereof
US20030199425A1 (en) 1997-06-27 2003-10-23 Desai Neil P. Compositions and methods for treatment of hyperplasia
KR100789008B1 (en) 1997-06-27 2007-12-26 아브락시스 바이오사이언스 인크. Novel Formulations of Pharmacological Agents
CA2369740A1 (en) 1999-04-22 2000-11-02 American Biosciences, Inc. Long term administration of pharmacologically active agents
CA2684454A1 (en) 1999-05-21 2000-11-18 American Bioscience, Llc. Protein stabilized pharmacologically active agents, methods for the preparation thereof and methods for the use thereof
ITMI20001107A1 (en) 2000-05-18 2001-11-18 Acs Dobfar Spa METHOD FOR TREATMENT OF SOLIC TUMORS BY INCORPORATING PACLITAXEL MICROPARTICLES OF ALBUMIN
BR0208627A (en) 2001-04-06 2004-03-09 Wyeth Corp Antineoplastic combinations such as rapamycin together with gemcitabine or fluorouracil
US20030054042A1 (en) 2001-09-14 2003-03-20 Elaine Liversidge Stabilization of chemical compounds using nanoparticulate formulations
EP1494699A4 (en) 2002-04-11 2009-07-22 Childrens Medical Center Methods for inhibiting vascular hyperpermeability
US20040126400A1 (en) 2002-05-03 2004-07-01 Iversen Patrick L. Delivery of therapeutic compounds via microparticles or microbubbles
DK1585548T3 (en) 2002-12-09 2018-09-03 Abraxis Bioscience Llc COMPOSITIONS AND PROCEDURES FOR THE DELIVERY OF PHARMACOLOGICAL AGENTS
KR20050095826A (en) 2002-12-09 2005-10-04 아메리칸 바이오사이언스, 인크. Compositions and methods of delivery of pharmacological agents
AU2003293529A1 (en) 2002-12-16 2004-07-29 Nitromed, Inc. Nitrosated and nitrosylated rapamycin compounds, compositions and methods of use
US20050119330A1 (en) 2003-03-17 2005-06-02 Kao Peter N. Use of antiproliferative agents in the treatment and prevention of pulmonary proliferative vascular diseases
US20050038498A1 (en) 2003-04-17 2005-02-17 Nanosys, Inc. Medical device applications of nanostructured surfaces
US20050152979A1 (en) 2003-09-05 2005-07-14 Cell Therapeutics, Inc. Hydrophobic drug compositions containing reconstitution enhancer
DE10347994A1 (en) 2003-10-15 2005-06-16 Pari GmbH Spezialisten für effektive Inhalation Aqueous aerosol preparation
CA2566571A1 (en) 2004-05-14 2005-12-15 Abraxis Bioscience, Inc. Treatment methods utilizing albumin-binding proteins as targets
EP3173072A1 (en) 2004-10-01 2017-05-31 Ramscor, Inc. Conveniently implantable sustained release drug compositions
WO2006053754A1 (en) 2004-11-19 2006-05-26 Novartis Ag COMBINATIONS OF ANTI-ATHEROSCLEROTIC PEPTIDES AND AN mTOR INHIBITING AGENT AND THEIR METHODS OF USE
MX339142B (en) 2005-02-18 2016-05-13 Abraxis Bioscience Llc Combinations and modes of administration of therapeutic agents and combination therapy.
US20090130163A1 (en) 2005-02-18 2009-05-21 Abraxis Bio Scoence, Inc. Drugs With Improved Hydrophobicity For Incorporation in Medical Devices
US20070166388A1 (en) 2005-02-18 2007-07-19 Desai Neil P Combinations and modes of administration of therapeutic agents and combination therapy
US8735394B2 (en) 2005-02-18 2014-05-27 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
EA200701998A1 (en) 2005-03-17 2008-02-28 Элан Фарма Интернэшнл Лтд. COMPOSITIONS FOR INJECTION OF NANOPARTICLES IMMUNE DEPRESSIVE COMPOUNDS
WO2006113579A2 (en) 2005-04-16 2006-10-26 Michigan State University Tumor inhibition by modulating sprouty expression or activity
CA2607325A1 (en) 2005-05-16 2006-11-23 Novartis Ag The use of rapamycin derivatives for the treatment and/or prevention of cardiovascular disorders
TWI429452B (en) 2005-08-31 2014-03-11 Abraxis Bioscience Llc Compositions comprising poorly water soluble pharmaceutical agents and antimicrobial agents
KR101457834B1 (en) 2005-08-31 2014-11-05 아브락시스 바이오사이언스, 엘엘씨 Compositions and methods for preparation of poorly water soluble drugs with increased stability
AU2006347740A1 (en) 2006-08-31 2008-03-06 Abraxis Bioscience, Llc Methods of inhibiting angiogenesis and treating angiogenesis-associated diseases
US20080280987A1 (en) 2006-08-31 2008-11-13 Desai Neil P Methods of inhibiting angiogenesis and treating angiogenesis-associated diseases
US20100112077A1 (en) 2006-11-06 2010-05-06 Abraxis Bioscience, Llc Nanoparticles of paclitaxel and albumin in combination with bevacizumab against cancer
ITFI20060277A1 (en) * 2006-11-08 2008-05-09 Giuliano Giustini LIFT FOR MOTORCYCLES AND SIMILAR, OFFERS MOBILITY OF OMNIDIRECTIONAL MOVEMENTS
EP3470071A1 (en) 2006-12-14 2019-04-17 Abraxis BioScience, LLC Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
KR20150002886A (en) 2007-03-07 2015-01-07 아브락시스 바이오사이언스, 엘엘씨 Nanoparticle comprising rapamycin and albumin as anticancer agent
US20100166869A1 (en) 2007-05-03 2010-07-01 Desai Neil P Methods and compositions for treating pulmonary hypertension
PL2155188T3 (en) 2007-06-01 2014-03-31 Abraxis Bioscience Llc Methods and compositions for treating recurrent cancer
AU2009234127B2 (en) 2008-04-10 2015-04-30 Abraxis Bioscience, Llc Compositions of hydrophobic taxane derivatives and uses thereof
NZ593343A (en) 2008-12-11 2013-04-26 Abraxis Bioscience Llc Combination cancer treatment conprising Abraxane and an inflamatory inhibitor
US20120189701A1 (en) 2009-03-13 2012-07-26 Desai Neil P Combination therapy with thiocolchicine derivatives
CN102458112A (en) 2009-04-10 2012-05-16 阿布拉科斯生物科学有限公司 Nanoparticle formulations and uses therof
MY175260A (en) 2009-04-15 2020-06-17 Abraxis Bioscience Llc Prion free nanoparticle compositions and methods of making thereof
PT2470173E (en) 2009-08-25 2016-06-15 Abraxis Bioscience Llc Combination therapy with nanoparticle compositions of taxane and hedgehog inhibitors
US9775819B2 (en) 2009-09-16 2017-10-03 R.P. Scherer Technologies, Llc Oral solid dosage form containing nanoparticles and process of formulating the same using fish gelatin
US8269348B2 (en) * 2010-02-22 2012-09-18 Texas Instruments Incorporated IC die including RDL capture pads with notch having bonding connectors or its UBM pad over the notch
MX359413B (en) 2010-03-26 2018-09-27 Abraxis Bioscience Llc Methods of treatment of hepatocellular carcinoma.
NZ703047A (en) 2010-03-29 2016-11-25 Abraxis Bioscience Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
CA3087813A1 (en) 2010-03-29 2011-10-06 Abraxis Bioscience, Llc Methods of treating cancer
MX343671B (en) 2010-06-02 2016-11-16 Abraxis Bioscience Llc * Methods of treating bladder cancer.
NZ604031A (en) 2010-06-04 2015-05-29 Abraxis Bioscience Llc Methods of treatment of pancreatic cancer
CN103037846B (en) 2010-06-07 2016-03-02 阿布拉科斯生物科学有限公司 The combinational therapeutic methods for the treatment of proliferative disease
US8871256B2 (en) * 2012-09-19 2014-10-28 Transdermal Biotechnology, Inc. Methods and systems for treatment of inflammatory diseases with nitric oxide
US9149455B2 (en) 2012-11-09 2015-10-06 Abraxis Bioscience, Llc Methods of treating melanoma
US20140186447A1 (en) 2012-12-28 2014-07-03 Abraxis Bioscience, Llc Nanoparticle compositions of albumin and paclitaxel
US9511046B2 (en) 2013-01-11 2016-12-06 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US20140199405A1 (en) 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on mutation status of k-ras
US20140199404A1 (en) 2013-01-11 2014-07-17 Abraxis Bioscience, Llc Method for treating cancer based on level of a nucleoside transporter

Cited By (42)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US8846771B2 (en) 2002-12-09 2014-09-30 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US9012518B2 (en) 2002-12-09 2015-04-21 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US9012519B2 (en) 2002-12-09 2015-04-21 Abraxis Bioscience, Llc Compositions and methods of delivery of pharmacological agents
US9561288B2 (en) 2005-02-18 2017-02-07 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US9101543B2 (en) 2005-02-18 2015-08-11 Abraxis Bioscience, Llc Combinations and modes of administration of therapeutic agents and combination therapy
US20110151012A1 (en) * 2005-08-31 2011-06-23 Desai Neil P Compositions comprising poorly water soluble pharmaceutical agents and antimicrobial agents
US9308180B2 (en) 2005-08-31 2016-04-12 Abraxis Bioscience, Llc Compositions and methods for preparation of poorly water soluble drugs with increased stability
US20110118342A1 (en) * 2005-08-31 2011-05-19 Tapas De Compositions and methods for preparation of poorly water soluble drugs with increased stability
US9724323B2 (en) 2006-12-14 2017-08-08 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US8999396B2 (en) 2006-12-14 2015-04-07 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US9675578B2 (en) 2006-12-14 2017-06-13 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US20100048499A1 (en) * 2006-12-14 2010-02-25 Desai Neil P Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US10682420B2 (en) 2006-12-14 2020-06-16 Abraxis Bioscience, Llc Breast cancer therapy based on hormone receptor status with nanoparticles comprising taxane
US8911786B2 (en) 2007-03-07 2014-12-16 Abraxis Bioscience, Llc Nanoparticle comprising rapamycin and albumin as anticancer agent
US8927019B2 (en) 2007-06-01 2015-01-06 Abraxis Bioscience, Llc Methods and compositions for treating recurrent cancer
US20100215751A1 (en) * 2007-06-01 2010-08-26 Desai Neil P Methods and compositions for treating recurrent cancer
US9446003B2 (en) 2009-04-15 2016-09-20 Abraxis Bioscience, Llc Prion free nanoparticle compositions and methods of making thereof
US10206887B2 (en) 2009-04-15 2019-02-19 Abraxis Bioscience, Llc Prion free nanoparticle compositions and methods of making thereof
US9393318B2 (en) 2010-03-29 2016-07-19 Abraxis Bioscience, Llc Methods of treating cancer
US10660965B2 (en) 2010-03-29 2020-05-26 Abraxis Bioscience, Llc Methods of enhancing drug delivery and effectiveness of therapeutic agents
US9597409B2 (en) 2010-03-29 2017-03-21 Abraxis Bioscience, Llc Methods of treating cancer
US9399071B2 (en) 2010-06-04 2016-07-26 Abraxis Bioscience, Llc Methods of treatment of pancreatic cancer
US9061014B2 (en) 2011-04-28 2015-06-23 Abraxis Bioscience, Llc Intravascular delivery of nanoparticle compositions and uses thereof
US9884013B2 (en) 2011-04-28 2018-02-06 Abraxis Bioscience, Llc Intravascular delivery of nanoparticle compositions and uses thereof
US10258565B2 (en) 2011-04-28 2019-04-16 Abraxis Bioscience, Llc Intravascular delivery of nanoparticle compositions and uses thereof
US10076501B2 (en) 2011-12-14 2018-09-18 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
US9585960B2 (en) 2011-12-14 2017-03-07 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
US10555912B2 (en) 2011-12-14 2020-02-11 Abraxis Bioscience, Llc Use of polymeric excipients for lyophilization or freezing of particles
US9149455B2 (en) 2012-11-09 2015-10-06 Abraxis Bioscience, Llc Methods of treating melanoma
US10328031B2 (en) 2013-01-11 2019-06-25 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US9855220B2 (en) 2013-01-11 2018-01-02 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US9511046B2 (en) 2013-01-11 2016-12-06 Abraxis Bioscience, Llc Methods of treating pancreatic cancer
US10744110B2 (en) 2013-03-12 2020-08-18 Abraxis Bioscience, Llc Methods of treating lung cancer
US9962373B2 (en) 2013-03-14 2018-05-08 Abraxis Bioscience, Llc Methods of treating bladder cancer
US10413531B2 (en) 2013-03-14 2019-09-17 Abraxis Bioscience, Llc Methods of treating bladder cancer
US10527604B1 (en) 2015-03-05 2020-01-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and paclitaxel
US10705070B1 (en) 2015-03-05 2020-07-07 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and poorly water soluble drug
US10900951B1 (en) 2015-03-05 2021-01-26 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and paclitaxel
US11320416B1 (en) 2015-03-05 2022-05-03 Abraxis Bioscience, Llc Methods of assessing suitability of use of pharmaceutical compositions of albumin and poorly water soluble drug
US10973806B2 (en) 2015-06-29 2021-04-13 Abraxis Bioscience, Llc Methods of treating epithelioid cell tumors comprising administering a composition comprising nanoparticles comprising an mTOR inhibitor and an albumin
US11944708B2 (en) 2018-03-20 2024-04-02 Abraxis Bioscience, Llc Methods of treating central nervous system disorders via administration of nanoparticles of an mTOR inhibitor and an albumin
US11497737B2 (en) 2019-10-28 2022-11-15 Abraxis Bioscience, Llc Pharmaceutical compositions of albumin and rapamycin

Also Published As

Publication number Publication date
DK2155188T3 (en) 2013-11-04
JP2010529025A (en) 2010-08-26
PL2155188T3 (en) 2014-03-31
JP2014185186A (en) 2014-10-02
JP2016172777A (en) 2016-09-29
AU2008260447A1 (en) 2008-12-11
US20150079181A1 (en) 2015-03-19
HK1140944A1 (en) 2010-10-29
US20160158365A9 (en) 2016-06-09
US20100215751A1 (en) 2010-08-26
EP2155188A1 (en) 2010-02-24
JP5579057B2 (en) 2014-08-27
US8927019B2 (en) 2015-01-06
PT2155188E (en) 2013-12-19
CA2689914C (en) 2016-08-16
CA2689914A1 (en) 2008-12-11
EP2155188B1 (en) 2013-10-09
AU2008260447B2 (en) 2013-10-10
HRP20131142T1 (en) 2014-01-03
ES2435452T3 (en) 2013-12-19
WO2008150532A1 (en) 2008-12-11

Similar Documents

Publication Publication Date Title
US8927019B2 (en) Methods and compositions for treating recurrent cancer
AU2018200347B2 (en) Methods of treatment of pancreatic cancer
AU2014228386B2 (en) Methods of treatment of pediatric solid tumor
AU2011261684B2 (en) Methods of treating bladder cancer
JP2018087235A (en) Methods of treating pancreatic cancer
WO2001089522A1 (en) Compositions and methods for administration of pharmacologically active compounds

Legal Events

Date Code Title Description
STCB Information on status: application discontinuation

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