US20210177802A1 - Pharmaceutical composition for treating tumor - Google Patents

Pharmaceutical composition for treating tumor Download PDF

Info

Publication number
US20210177802A1
US20210177802A1 US17/180,107 US202117180107A US2021177802A1 US 20210177802 A1 US20210177802 A1 US 20210177802A1 US 202117180107 A US202117180107 A US 202117180107A US 2021177802 A1 US2021177802 A1 US 2021177802A1
Authority
US
United States
Prior art keywords
eribulin
liposomal
tumor
antibody
pharmaceutically acceptable
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
US17/180,107
Inventor
Taro Semba
Yasuhiro Funahashi
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.)
Eisai R&D Management Co Ltd
Original Assignee
Eisai R&D Management Co Ltd
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 Eisai R&D Management Co Ltd filed Critical Eisai R&D Management Co Ltd
Priority to US17/180,107 priority Critical patent/US20210177802A1/en
Assigned to EISAI R&D MANAGEMENT CO., LTD. reassignment EISAI R&D MANAGEMENT CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: FUNAHASHI, YASUHIRO, SEMBA, TARO
Publication of US20210177802A1 publication Critical patent/US20210177802A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • 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
    • A61K31/357Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having two or more oxygen atoms in the same ring, e.g. crown ethers, guanadrel
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39541Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against normal tissues, cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions
    • A61K9/127Liposomes
    • A61K9/1271Non-conventional liposomes, e.g. PEGylated liposomes, liposomes coated with polymers
    • 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2300/00Mixtures or combinations of active ingredients, wherein at least one active ingredient is fully defined in groups A61K31/00 - A61K41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/73Inducing cell death, e.g. apoptosis, necrosis or inhibition of cell proliferation

Definitions

  • the present invention relates to a pharmaceutical composition for treating tumor.
  • Eribulin represented by formula (I) is used as a therapeutic agent for breast cancer and soft tissue tumor.
  • Patent Literature 1 discloses eribulin or a pharmaceutically acceptable salt thereof and a method of producing the same.
  • Patent Literatures 2 and 3 disclose methods for producing eribulin and eribulin mesylate, which is a mesylate (methanesulfonate) thereof.
  • Patent Literature 4 discloses a method of inhibiting growth of cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient.
  • Patent Literature 5 discloses a method of treating cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient in combination with a certain second anticancer agent.
  • Patent Literature 6 discloses a method of treating cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient in combination with a second therapeutic approach.
  • Patent Literatures 7 and 8 disclose liposomal compositions comprising eribulin mesylate.
  • Patent Literature 9 discloses a method for treating breast cancer, comprising administering a combination of eribulin or a pharmaceutically acceptable salt thereof and a programmed cell death 1 protein (PD-1) antagonist.
  • PD-1 programmed cell death 1 protein
  • PD-1 is recognized as an important factor in maintenance of immunoregulation and peripheral tolerance.
  • PD-1 is moderately expressed in naive T cells, B cells, and NK T cells, and upregulated by T/B cell receptor signal transduction in lymphocytes, monocytes, and myeloid cells (Non Patent Literature 1).
  • PD-L1 is expressed in various cancer cells or T/B cells, macrophages, mDCs, plasmacytoid DCs:(pDCs), bone marrow mast cells, and the like.
  • the two known PD-1 ligands PD-L1 (B7-H1) and PD-L2 (B7-DC) are expressed in human cancer occurring in various tissues.
  • the PD-L1 expression has been shown to correlate with poor prognosis and decreased overall survival, regardless of subsequent treatment (Non Patent Literatures 2 to 13).
  • the PD-1 expression in tumor-infiltrating lymphocytes is characteristic of functionally impaired T cells in breast cancer and melanoma (Non Patent Literatures 14 to 15) and correlates with poor prognosis in kidney cancer (Non Patent Literature 16). Therefore, it has been proposed to block the immunosuppression mechanism that cancer cells bring, such as the interaction of tumor cells expressing PD-L1 with T cells expressing PD-1, and thereby bring the immune response to tumor.
  • the present invention is directed to provide a new pharmaceutical composition for treating tumor.
  • the present inventors have studied diligently and, as a result, found that the combined administration of a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist exhibits unexpected antitumor effect, thereby completing the present invention.
  • a pharmaceutical composition for treating tumor comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof, wherein the pharmaceutical composition is administered in combination with a PD-1 antagonist.
  • a pharmaceutical composition for treating tumor comprising a PD-1 antagonist, wherein the pharmaceutical composition is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
  • a therapeutic agent for tumor comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof wherein the pharmaceutical composition is administered in combination with a PD-1 antagonist.
  • a therapeutic agent for tumor comprising a PD-1 antagonist, wherein the therapeutic agent is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
  • the therapeutic agent according to any of [8] to [13] above, wherein the tumor is breast cancer.
  • a method for treating tumor comprising administering a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist to a patient in need thereof.
  • a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
  • eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
  • the PD-1 antagonist is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
  • the tumor is breast cancer.
  • a PD-1 antagonist in the manufacture of a pharmaceutical composition for treating tumor, wherein the pharmaceutical composition is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
  • a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
  • eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
  • the PD-1 antagonist is an anti-PD-1 antibody.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
  • the tumor is breast cancer.
  • a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof for use in tumor treatment wherein the liposomal composition is administered in combination with a PD-1 antagonist.
  • a PD-1 antagonist for use in tumor treatment wherein the PD-1 antagonist is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
  • the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • [34-1] The liposomal composition or PD-1 antagonist for use according to [28] to [33] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
  • [34-2] The liposomal composition or PD-1 antagonist for use according to [28] to [33] above, wherein the tumor is breast cancer.
  • a kit for treating tumor comprising a formulation comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a formulation comprising a PD-1 antagonist.
  • kits according to [38] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • the tumor is selected from the group consisting of breast cancer, gastric cancel; esophageal cancer and small cell lung cancer.
  • the tumor is breast cancer.
  • a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist exhibits unexpected antitumor effect.
  • FIG. 1 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.1 mg/kg and an anti-PD-1 antibody on tumor growth.
  • FIG. 2 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.1 mg/kg and an anti-PD-1 antibody on T ⁇ 5.
  • FIG. 3 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.3 mg/kg and an anti-PD-1 antibody on tumor growth.
  • FIG. 4 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.3 mg/kg and an anti-PD-1 antibody on T ⁇ 5.
  • the liposomal compositions in the present disclosure comprises eribulin or a pharmaceutically acceptable salt thereof (hereinafter referred to as “eribulin or the like”).
  • the “pharmaceutically acceptable salt” may be either an inorganic acid salt or an organic acid salt and is not particularly limited, as long as it forms a salt with eribulin, and examples thereof include hydrochloride, sulfate, citrate, hydrobromide, hydroiodide, nitrate, bisulfate, phosphate, superphosphate, isonicotinate, acetate, lactate, salicylate, tartrate, pantothenate, ascorbate, succinate, maleate, fumarate, gluconate, saccharinate, formate, benzoate, glutamate, mesylate (methanesulfonate), ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate.
  • the pharmaceutically acceptable salts are hydrochloride, sulfate, acetate, phosphate, citrate, and mesylate.
  • the pharmaceutically acceptable salts are hydroch
  • the pharmaceutically acceptable salt of eribulin may be a salt of eribulin and aluminum, calcium, lithium, magnesium, sodium, zinc, or diethanolamine.
  • examples of eribulin or the like include eribulin mesylate.
  • Eribulin or the like is a compound or a salt thereof described in Patent Literature 1 or U.S. Pat. No. 6,214,865 and has pharmacological activities including antitumor and antimitotic activities.
  • Patent Literature 1 discloses that eribulin or the like has, as an antitumor agent, anti-tumor activity against melanoma, fibrosarcoma, monocytic leukemia, colon cancer, ovarian cancer, breast cancel; osteosarcoma, prostate cancer, lung cancer, and ras-transformed fibroblasts. Eribulin or the like is obtained by a method of production described in Patent Literatures 1 to 3.
  • the “liposome” means a closed microvesicle having an inner phase surrounded by a lipid bilayer.
  • the liposomes include small unilamellar liposomes (SUVs: small unilamellar vesicles), large unilamellar liposomes (LUVs: large unilamellar vesicles), further large unilamellar liposomes (GUVs: giant unilamellar vesicles), multi-lamellar liposomes having a plurality of concentric membranes (MLVs: multi lamellar vesicles), liposomes having a plurality of non-concentric, irregular membranes (MVVs: multivesicular vesicles), and the like.
  • SUVs small unilamellar vesicles
  • LUVs large unilamellar vesicles
  • GUIVs giant unilamellar vesicles
  • MMVs multi lamellar vesicles
  • the “liposomal inner phase” means an aqueous region surrounded by a liposomal lipid bilayer and is used synonymously with an “inner aqueous phase” and a “liposomal inner aqueous phase”.
  • the “liposomal outer phase” means a region that is not surrounded by a liposomal lipid bilayer (that is, the region except the inner phase and the lipid bilayer) when the liposome is dispersed in a liquid.
  • the “liposomal composition” means a composition comprising a liposome and further comprising eribulin or the like in the liposomal inner phase.
  • the liposomal composition includes solid and liquid compositions.
  • the “liposomal dispersion liquid” means a composition comprising a liposome in which eribulin or the like is not yet encapsulated into the liposomal inner phase.
  • the “liposomal preparatory liquid” means a composition comprising a liposome in which an adjustment of the liposome outer phase in order to encapsulate eribulin or the like into the liposome inner phase is not yet performed.
  • the liposome preferably comprises a phospholipid and/or a phospholipid derivative as a membrane component.
  • Examples of the phospholipid and/or phospholipid derivative include phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, phosphatidylinositol, phosphatidylglycerol, cardiolipin, sphingomyelin, ceramide phosphoryl ethanolamine, ceramidephosphorylglycerol, ceramidephosphorylglycerolphosphate, 1,2-dimyristoyl-1,2-deoxyphosphatidyl choline, plasmalogen, and phosphatidate.
  • the phospholipid and/or phospholipid derivative may be one or a combination of two or more of these.
  • Fatty acid residues in the phospholipid and/or phospholipid derivative are not particularly limited, and examples thereof include saturated or unsaturated fatty acid residues having 12 to 20 carbon atoms, and specific examples thereof include acyl groups derived from fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, and linoleic acid.
  • a phospholipid derived from a natural product such as egg yolk lecithin and soy lecithin, and partially hydrogenated egg yolk lecithin, (fully) hydrogenated egg yolk lecithin, partially hydrogenated soy lecithin, and (fully) hydrogenated soybean lecithin, in which unsaturated fatty acid residues are partially or fully hydrogenated, or the like may be used.
  • the amount (molar fraction) of the phospholipid and/or phospholipid derivative, used in the preparation of the liposome, that are/is blended is not particularly limited, and is, in one embodiment, 10 to 80% and, in a particular embodiment, 30 to 60% based on the total ribosomal membrane components.
  • the liposome may comprise, as a membrane component, a sterol such as cholesterol and cholestanol and a fatty acid having a saturated or unsaturated acyl group having 8 to 22 carbon atoms as a membrane stabilizing agent, and an antioxidant such as ⁇ -tocopherol, besides the phospholipid and/or phospholipid derivative.
  • a sterol such as cholesterol and cholestanol and a fatty acid having a saturated or unsaturated acyl group having 8 to 22 carbon atoms
  • an antioxidant such as ⁇ -tocopherol
  • the amount (molar fraction) of the sterol, used in the preparation of the liposome, that is blended is not particularly limited, and is, in one embodiment, 1 to 60%, 10 to 50%, or 30 to 50% based on the total liposomal membrane components.
  • the amount (molar fraction) of the fatty acid blended is not particularly limited, and is, in one embodiment, 0 to 30% and 0 to 20% or 0 to 10% based on the total liposomal membrane components.
  • the amount (molar fraction) of the antioxidant blended is not particularly limited, as long as an amount that provides the antioxidant effect is added, and it is, in one embodiment, 0 to 15%, 0 to 10%, or 0 to 5% based on the total liposomal membrane components.
  • the liposome may comprise a functional lipid or a modified lipid as a membrane component.
  • Examples of the functional lipid include a blood-retaining lipid derivative, a temperature change-sensitive lipid derivative, and a pH-sensitive lipid derivative.
  • modified lipid examples include a PEGylated lipid, a glycolipid, an antibody-modified lipid, and a peptide-modified lipid.
  • the blood-retaining lipid derivative examples include polyethylene glycol derivatives (such as methoxy polyethylene glycol condensates) such as condensation products of phosphoethanolamine and methoxy polyethylene glycol: N- ⁇ carbonyl-methoxy polyethylene glycol-2000 ⁇ -1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine, N- ⁇ carbonyl-methoxy polyethylene glycol-5000 ⁇ -1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine, N- ⁇ carbonyl-methoxy polyethylene glycol-750 ⁇ -1,2-distearoyl-sn-glycero-3-phosphoethanolamine, N- ⁇ carbonyl-methoxy polyethylene glycol-2000 ⁇ -1,2-distearoyl-sn-glycero-3-phosphoethanolamine, (MPEG2000-distearoylphosphatidylethanolamine), and N- ⁇ carbonyl-methoxy polyethylene glycol-5000 ⁇ -1,2-distearoyl
  • the blending amount (molar fraction) of the blood-retaining lipid derivative, used in the preparation of the liposome is not particularly limited, and is, in one embodiment, 0 to 50%, 0 to 30%, or 0 to 20% based on the total liposomal membrane components.
  • Examples of the temperature change-sensitive lipid derivative include dipalmitoylphosphatidylcholine.
  • a temperature change-sensitive lipid derivative in a liposome it becomes possible to disrupt the liposome at a particular temperature, to change the surface properties of the liposome, and the like.
  • a target site such as tumor, it becomes possible to disrupt the liposome at the target site and have an active compound released at the target site, and the like.
  • Examples of the pH-sensitive lipid derivative include dioleoylphosphatidylethanolamine.
  • glycolipid, antibody-modified lipid, and peptide-modified lipid include lipids linked to a sugar, an antibody, or a peptide having an affinity for a target cell or a target tissue.
  • Using a modified lipid allows to deliver the liposome actively to the target cell or the target tissue.
  • composition of membrane components for liposome having a practically acceptable level of membrane permeability can be set by a person skilled in the art as appropriate depending on the active compound, the target tissue, and the like (see Hiroshi Kikuchi et al. “Liposome I—How to prepare and assay—(in Japanese)” Cell technology (1983) 2 (9): pp. 1136-1149 and the references cited in the reference, and the like).
  • the liposomal composition may be used not only in targeting at a target tissue such as solid cancel; but also in the delivery of an active compound to blood cancer or the like.
  • the liposomal membrane components include, in one embodiment, phospholipid, cholesterol, and a methoxy polyethylene glycol condensation product.
  • eribulin or the like is encapsulated in a liposome having a lipid membrane.
  • eribulin or the like may be distributed in the lipid bilayer.
  • the liposomal composition according to the present disclosure can be obtained by a method described in Patent Literature 7.
  • the solid liposomal composition may be dissolved or suspended in a certain solvent described below to prepare a liquid liposomal composition.
  • the frozen solid liposomal composition may be thawed by leaving the composition at room temperature or the like to prepare a liquid liposomal composition.
  • the liposomal composition according to the present disclosure is not limited, as long as it comprises (1) eribulin or the like.
  • the liposomal composition according to the present disclosure may further comprise (2) at least one ammonium salt and (3) at least one acid, salt, base, and/or amino acid.
  • the at least one ammonium salt (2) examples include ammonium chloride, ammonium borate, ammonium sulfate, ammonium formate, ammonium acetate, ammonium citrate, ammonium tartrate, ammonium succinate, and ammonium phosphate and, in one embodiment, the at least one ammonium salt (2) is ammonium sulfate, ammonium citrate, and ammonium tartrate.
  • examples of the acid include ascorbic acid, benzoic acid, succinic acid, citric acid, glutamic acid, phosphoric acid, acetic acid, propionic acid, tartaric acid, carbonic acid, lactic acid, boric acid, maleic acid, fumaric acid, malic acid, adipic acid, hydrochloric acid, and sulfuric acid;
  • examples of the salt include sodium salts of the aforementioned acids, potassium salts of the aforementioned acids, and ammonium salts of the aforementioned acids;
  • examples of the base include trishydroxymethylaminomethane, ammonia, sodium hydroxide, and potassium hydroxide; and examples of the amino acid include arginine, histidine, and glycine.
  • the acid, salt, base, and/or amino acid (3) in the liposomal inner phase is hydrochloric acid, acetic acid, lactic acid, tartaric acid, succinic acid, citric acid, and phosphoric acid, sodium salts of the aforementioned acids, and sodium hydroxide and ammonia
  • the acid, salt, base, and/or amino acid (3) is acetic acid, lactic acid, tartaric acid, citric acid, and phosphoric acid, sodium salts of the aforementioned acids, and sodium hydroxide and ammonia.
  • liposomal composition An example of the components of the liposomal composition is set forth in Table 1.
  • 96 mg/mL sucrose may be used, instead of 9 mg/mL sodium chloride, as an osmotic agent (liposomal outer phase).
  • pH adjuster Hydrogenated soy phosphatidylcholine
  • MPEG2000-distearoylphosphatidylethanolamine N- ⁇ carbonyl-methoxy polyethylene glycol-2000 ⁇ -1,2-distearoyl-sn-glycero-3-phosphoethanolamine
  • the liposomal composition according to the present disclosure may be administered by injection (intravenous injection, intraarterial injection, local injection), orally, nasally, transdermally, transpulmonarily, ophthalmically, and the like, and examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to a target cell and organ.
  • injection intravenous injection, intraarterial injection, local injection
  • examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to a target cell and organ.
  • Examples of the dosage form of the liposomal composition for oral administration include tablets, powders, granules, syrups, capsules, and oral solutions.
  • Examples of the dosage form of the liposomal composition for parenteral administration include injections, drip infusions, ophthalmic liquids, ointments, suppositories, suspensions, cataplasms, lotions, aerosols, and plasters, and, in one embodiment, the liposomal composition for parenteral administration is an injection or a drip infusion.
  • the liposomal composition according to the present disclosure may be formulated by a method, for example, described in Japanese Pharmacopoeia (JP) 17th edition, United States Pharmacopoeia (USP), or European pharmacopoeia (EP).
  • the liposomal composition may be used as it is.
  • a solvent may be injected by a physician or a patient into a vial in which a solid formulation is encapsulated to do such preparation upon use.
  • a solid formulation obtained by freezing a liquid liposomal composition may be stored in a frozen state and thawed by leaving at room temperature or thawed rapidly with heating back into a liquid upon use to be used as a liquid.
  • the dose upon administration of the liposomal composition alone vary markedly depending on the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like.
  • the liposomal composition is administered, for example, at 0.1 to 10 mg/m 2 (body surface) in terms of eribulin mesylate per day for an adult.
  • the liposomal composition is administered at a dose of 0.5 to 3 mg/m 2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks.
  • the liposomal composition is more preferably administered at a dose of 0.5 to 2 mg/m 2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks.
  • the liposomal composition is preferably administered at a dose of approximately 1.5 mg/m 2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks.
  • the liposomal composition is administered intravenously at 0.5 to 1.4 mg/m 2 on day 1 of a 21-day cycle or administered intravenously at 0.5 to 1.5 mg/m 2 on day 1 and day 15 of a 28-day cycle in terms of eribulin mesylate.
  • Eribulin or the like contained in the liposomal composition may be administered once a day or in several divided daily doses.
  • the liposomal composition may be a liposomal composition comprising, for example, 0.01 to 300 mg/mL of eribulin or the like in the liposomal inner phase.
  • the liposomal composition is formulated, for example, as an injection comprising 0.20 mg/mL eribulin mesylate (0.18 mg/mL eribulin) incorporated in a liposome having a lipid membrane consisting of HSPC, cholesterol, and MPEG2000-DSPE.
  • Such an injection may comprise sucrose or sodium chloride as an isotonizing agent, ammonium sulfate, citric acid, and L-histidine, and sodium hydroxide and hydrochloric acid to adjust pH.
  • the injection is directly administered to a patient or diluted with physiological saline to a concentration in the range of 0.0035 mg/mL or higher and lower than 0.2 mg/mL before the administration to a patient.
  • the PD-1 antagonist in the present disclosure may comprise any compound or biological molecule that blocks the binding of PD-L1 expressed in cancer cells to PD-1 expressed in immune cells (T cells, B cells, or natural killer T (NKT) cells), or that blocks the binding of PD-L2 expressed in cancer cells to PD-1 expressed in immune cells.
  • the PD-1 antagonist blocks the binding of human PD-L1 to human PD-1 and, in one embodiment, blocks the binding of both human PD-L1 and PD-L2 to human PD-1.
  • the amino acid sequence of human PD-1 can be found in NCBI Locus No.: NP_005009.
  • the amino acid sequences of human PD-L1 and PD-L2 can be found in NCBI Locus No: NP_054862 and NP_079515, respectively.
  • the PD-1 antagonist useful in the present disclosure may comprise a monoclonal antibody (mAb) or an antigen-binding fragment thereof that specifically binds to PD-1 or PD-L1 or that specifically binds to human PD-1 or human PD-L1.
  • the mAb may be a human antibody, a humanized antibody, or a chimeric antibody and may comprise a human constant region.
  • the human constant region is selected from the group consisting of IgG1, IgG2, IgG3, and IgG4 constant regions and, in one embodiment, the human constant region is an IgG1 or IgG4 constant region.
  • the antigen-binding fragment may be selected from the group consisting of Fab, Fab′-SH, F(ab) 2 , scFv, and Fv fragment.
  • an example of useful PD-1 antagonists is an anti-PD-1 antibody, which is, in one embodiment, an anti-human PD-1 antibody and, in a particular embodiment, an anti-human PD-1 monoclonal antibody (anti-human PD-1 mAb).
  • anti-human PD-1 mAb examples of the human PD-1-binding mAb binding are described in U.S. Pat. Nos. 7,488,802, 7,521,051, 8,008,449, 8,354,509, 8,168,757, WO 2004/004771, WO 2004/072286, WO 2004/056875, and US Patent Application Publication No. 2011/0271358.
  • Anti-human PD-1 monoclonal antibodies useful as the PD-1 antagonist according to the present disclosure include nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • the PD-1 antagonist according to the present disclosure may be administered by injection (intravenous injection, intraarterial injection, local injection), orally, nasally, transdermally, transpulmonarily, ophthalmically, and the like and examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to target cells and organ.
  • injection intravenous injection, intraarterial injection, local injection
  • examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to target cells and organ.
  • Examples of the dosage form of the PD-1 antagonist for oral administration include tablets, powders, granules, syrups, capsules, and oral solutions.
  • Examples of the dosage form of the PD-1 antagonist for parenteral administration include injections, drip infusions, ophthalmic liquids, ointments, suppositories, suspensions, cataplasms, lotions, aerosols, and plasters and in one embodiment, the dosage form of the PD-1 antagonist for parenteral administration is an injection or a drip infusion.
  • the PD-1 antagonist according to the present disclosure may be formulated by a method, for example, described in Japanese Pharmacopoeia (JP) 17th edition, United States Pharmacopoeia (USP), or European pharmacopoeia (EP).
  • the anti-PD-1 antibody may be provided as a liquid preparation or prepared by rehydrating freeze-drying powder with sterile water for injection before use.
  • an anti-human PD-1 mAb alone as the PD-1 antagonist Upon administration of an anti-human PD-1 mAb alone as the PD-1 antagonist to a patient, the dose thereof varies markedly depending on the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like.
  • the anti-human PD-1 mAb is administered, for example, at a dose of 1, 2, 3, 5, or 10 mg/kg at intervals of approximately 14 days ( ⁇ 2 days), approximately 21 days ( ⁇ 2 days), or approximately 30 days ( ⁇ 2 days).
  • pembrolizumab When pembrolizumab is administered as the PD-1 antagonist, pembrolizumab is, for example, administered intravenously at a dose selected from the group consisting of 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg of Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W.
  • a dose selected from the group consisting of 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg of Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W.
  • Pembrolizumab is administered as a liquid medicine, for example, comprising 25 mg/ml pembrolizumab, 7% (w/v) sucrose, and 0.02% (w/v) polysorbate 80 in a 10 mM histidine buffer, pH 5.5, and a selected dose of the medicine is administered by IV injection over a period of approximately 30 minutes.
  • a liquid medicine for example, comprising 25 mg/ml pembrolizumab, 7% (w/v) sucrose, and 0.02% (w/v) polysorbate 80 in a 10 mM histidine buffer, pH 5.5, and a selected dose of the medicine is administered by IV injection over a period of approximately 30 minutes.
  • nivolumab When nivolumab is administered as the PD-1 antagonist, nivolumab is, for example, administered intravenously at a dose selected from the group consisting of 1 mg/kg Q2W, 2 mg/kg Q2W, and 3 mg/kg Q2W.
  • the doses of the liposomal composition and the PD-1 antagonist in the combined administration of the present disclosure may usually be set at doses lower than the doses when they are administered alone. Specific doses, administration routes, administration frequencies, and administration cycles are determined as appropriate in consideration of the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like.
  • the mode of administration of the liposomal composition and the PD-1 antagonist in the present disclosure is not particularly limited, as long as the liposomal composition and the PD-1 antagonist are administered in combination when they are administered.
  • the liposomal composition and the PD-1 antagonist are administered to a patient simultaneously, separately, continuously, or at a time interval.
  • “simultaneously” means that each component is administered in the same period of time or strictly simultaneously or via the same administration route.
  • “Separately” means that each component is administered at different dose intervals or frequencies or via different administration routes.
  • Continuous means that each component is administered via the same administration route or different administration routes in any order within a certain period of time.
  • “At a time interval” means that each component is administered via the same administration route or different administration routes, with each component administered at a time interval.
  • the PD-1 antagonist is administered in a period of 1 cycle of the administration of the liposomal composition or in a period in which the cycle is repeated, it is considered that both are administered in combination.
  • Tumors to be treated in the present disclosure are, for example, breast cancer, gastric cancer, esophageal cancer, small cell lung cancer, colorectal cancer, and kidney cancer.
  • a P glycoprotein-knockout cell line produced from murine breast cancer 4T1 cells was cultured using RPMI1640 medium (SIGMA) containing 10% of FBS (fetal bovine serum), 1 mM sodium pyruvate, and antibiotics, under conditions at 37° C. in a 5% carbon dioxide gas incubator. The cells were collected using trypsin-EDTA when the cells reached to approximately 80% confluency.
  • SIGMA RPMI1640 medium
  • the medium described above (RPMI1640) was added to the collected cells, a suspension was prepared at 1.0 ⁇ 10 7 cells/mL, and 0.1 mL of the suspension was subcutaneously transplanted at the right body side into 6 mice (BALB/cAJcl, CLEA Japan, Inc.) per each group of the control group, eribulin mesylate alone administration group, eribulin mesylate liposomal formulation alone administration group, the anti-mouse PD-1 antibody (Bio X cell) alone administration group, eribulin mesylate and anti-mouse PD-1 antibody combined administration group, and eribulin mesylate liposomal formulation and anti-mouse PD-1 antibody combined administration group.
  • RPMI1640 RPMI1640
  • eribulin mesylate 0.1 mg/kg, once a week, twice in total, tail vein injection
  • eribulin mesylate liposomal formulation 0.1 mg/kg, once a week, twice in total, tail vein injection
  • anti-mouse PD-1 antibody 200 ⁇ g/mouse, once a week, twice in total, tail vein injection
  • No drug was administered to the control group.
  • the maximum tolerated dose of eribulin mesylate liposomal formulation in mice is 2.5 mg/kg and the dose in this experiment was set very low at 0.1 mg/kg, which is 1/25 of the maximum tolerated dose.
  • the liposomal composition comprising eribulin mesylate was prepared with the components set forth in Table 1 in accordance with the following method.
  • Ammonium sulfate and citric acid monohydrate were dissolved and diluted with pure water to prepare an aqueous solution of 200 mM ammonium sulfate/60 mM citric acid.
  • the aqueous solution of 200 mM ammonium sulfate/60 mM citric acid was adjusted to pH 5.5 with an aqueous ammonium solution and then diluted with pure water to obtain an aqueous solution of 100 mM ammonium sulfate/30 mM citric acid.
  • Hydrogenated soy phosphatidylcholine, cholesterol, and MPEG2000-distearoylphosphatidylethanolamine were weighted in accordance with a weight ratio of 71:23:27, respectively. These were each dissolved in chloroform and these solutions were mixed. Chloroform was then evaporated under reduced pressure in a rotary evaporator to prepare a lipid film. To the obtained lipid film, the prepared aqueous solution for liposomal inner phase heated to approximately 80° C. was added and the resulting mixture was stirred to prepare a liposomal preparatory liquid. Sizing was performed using an extruder (a product made by Lipex Biomembranes Inc.) heated to approximately 80° C. to obtain a sized liposomal preparatory liquid.
  • an extruder a product made by Lipex Biomembranes Inc.
  • the liposomal outer phase was exchanged into an aqueous solution of 0.9% sodium chloride/10 mM histidine.
  • the liquid was centrifuged at 400,000 ⁇ g for 30 minutes. After the centrifugation, re-dispersion was performed and the liquid volume was adjusted with an aqueous solution of 0.9% sodium chloride/10 mM histidine to obtain a liposomal dispersion liquid.
  • the liposomal dispersion liquid and eribulin mesylate solution were mixed in a glass container and incubated in a water bath at 60° C. for 3 minutes to obtain a liposomal composition with a liposomal inner phase in which eribulin mesylate was introduced.
  • An aqueous solution of 0.9% sodium chloride/10 mM histidine was added to the liposomal composition and filter sterilization was performed with a 0.22 ⁇ m polyvinylidene fluoride (PVDF) filter to obtain an eribulin mesylate liposomal composition.
  • PVDF polyvinylidene fluoride
  • the tumor volume was calculated in accordance with the following formula.
  • Tumor volume (mm 3 ) longest diameter (mm) ⁇ short axis 2 (mm 2 )/2
  • the results of measurement of the tumor volume in each group are illustrated as mean and standard deviation (SD) in FIG. 1 .
  • SD standard deviation
  • ANOVA repeated measures analysis of variance
  • the statistical comparison between the two groups of eribulin mesylate and anti-mouse PD-1 antibody combined administration group and eribulin mesylate liposomal formulation and anti-mouse PD-1 antibody combined administration group was conducted by repeated measures ANOVA (#: p ⁇ 0.05).
  • Pgp-KO 4T1 cells were cultured with the RPMI1640 medium containing 10% FBS, 1 mM sodium pyruvate, and antibiotics under conditions at 37° C. in a 5% carbon dioxide gas incubator.
  • the cells were collected using trypsin-EDTA when the cells reached to approximately 80% confluency.
  • the medium described above was added to the collected cells to prepare a suspension at 1.0 ⁇ 10 7 cells/mL.
  • mice BALB/cAJcl, CLEA Japan, Inc.
  • mice BALB/cAJcl, CLEA Japan, Inc.
  • mice eribulin mesylate liposomal formulation alone administration group
  • the anti-mouse PD-1 antibody Bio X cell alone administration group
  • the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody was subcutaneously transplanted at the right body side into 6 mice (BALB/cAJcl, CLEA Japan, Inc.) per each group of the control group, eribulin mesylate liposomal formulation alone administration group, the anti-mouse PD-1 antibody (Bio X cell) alone administration group
  • the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody was subcutaneously transplanted at the right body side into 6 mice (BALB/cAJcl, CLEA Japan, Inc.) per each group of the control group, e
  • eribulin mesylate liposomal formulation 0.3 mg/kg, once a week, twice in total, tail vein injection
  • the anti-mouse PD-1 antibody 200 ⁇ g/mouse, once a week, twice in total, tail vein injection
  • the liposomal composition comprising eribulin mesylate was prepared in accordance with the method as that in Example 1.
  • the tumor volume was calculated in accordance with the following formula.
  • Tumor volume (mm 3 ) longest diameter (mm) ⁇ short axis 2 (mm 2 )/2
  • (t) indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with eribulin mesylate liposomal formulation alone administration
  • (#) indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with the anti-mouse PD-1 antibody alone administration.
  • the tumor disappearance was observed in the combined administration group of eribulin mesylate liposomal formulation (0.3 mg/kg) and the anti-mouse PD-1 antibody at a frequency higher than other groups.

Landscapes

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

Abstract

Disclosed herein is a method for treating tumor, comprising administering a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist to a patient in need thereof.

Description

    CROSS-REFERENCE TO RELATED APPLICATION
  • This application claims the benefit of Japanese patent application No. 2019-138041 filed on Jul. 26, 2019, the disclosure of which is herein incorporated by reference in its entirety.
  • TECHNICAL FIELD
  • The present invention relates to a pharmaceutical composition for treating tumor.
  • BACKGROUND
  • Eribulin represented by formula (I) is used as a therapeutic agent for breast cancer and soft tissue tumor.
  • Figure US20210177802A1-20210617-C00001
  • Patent Literature 1 discloses eribulin or a pharmaceutically acceptable salt thereof and a method of producing the same. Patent Literatures 2 and 3 disclose methods for producing eribulin and eribulin mesylate, which is a mesylate (methanesulfonate) thereof. Patent Literature 4 discloses a method of inhibiting growth of cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient. Patent Literature 5 discloses a method of treating cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient in combination with a certain second anticancer agent. Patent Literature 6 discloses a method of treating cancer in a patient by administering eribulin or a pharmaceutically acceptable salt thereof to the patient in combination with a second therapeutic approach. Patent Literatures 7 and 8 disclose liposomal compositions comprising eribulin mesylate. Patent Literature 9 discloses a method for treating breast cancer, comprising administering a combination of eribulin or a pharmaceutically acceptable salt thereof and a programmed cell death 1 protein (PD-1) antagonist.
  • PD-1 is recognized as an important factor in maintenance of immunoregulation and peripheral tolerance. PD-1 is moderately expressed in naive T cells, B cells, and NK T cells, and upregulated by T/B cell receptor signal transduction in lymphocytes, monocytes, and myeloid cells (Non Patent Literature 1). Meanwhile, PD-L1 is expressed in various cancer cells or T/B cells, macrophages, mDCs, plasmacytoid DCs:(pDCs), bone marrow mast cells, and the like.
  • The two known PD-1 ligands PD-L1 (B7-H1) and PD-L2 (B7-DC) are expressed in human cancer occurring in various tissues. For example, in a large amount of sample sets of ovarian cancer, renal cancer, colorectal cancer, pancreatic cancer, liver cancer, and melanoma, the PD-L1 expression has been shown to correlate with poor prognosis and decreased overall survival, regardless of subsequent treatment (Non Patent Literatures 2 to 13). Similarly, it was found that the PD-1 expression in tumor-infiltrating lymphocytes is characteristic of functionally impaired T cells in breast cancer and melanoma (Non Patent Literatures 14 to 15) and correlates with poor prognosis in kidney cancer (Non Patent Literature 16). Therefore, it has been proposed to block the immunosuppression mechanism that cancer cells bring, such as the interaction of tumor cells expressing PD-L1 with T cells expressing PD-1, and thereby bring the immune response to tumor.
  • Several monoclonal antibodies that inhibit the interaction between PD-1 and either or both of PD-1 ligands PD-L1 and PD-L2 are under clinical development for treating cancer. It has been proposed that the efficacy of such antibodies may be increased when administered in combination with another approved or experimental cancer therapy, for example, radiation, surgery, a chemotherapeutic agent, a targeted therapy, an agent that inhibits another signaling pathway that is dysregulated in tumor, and another immunostimulant.
    • Patent Literature 1: WO 99/65894
    • Patent Literature 2: WO 2005/118565
    • Patent Literature 3: WO 2011/094339
    • Patent Literature 4: U.S. Pat. No. 6,469,182
    • Patent Literature 5: U.S. Application Publication No. 2006/104984
    • Patent Literature 6: U.S. Pat. No. 6,653,341
    • Patent Literature 7: WO 2010/113984
    • Patent Literature 8: WO 2017/188350
    • Patent Literature 9: WO 2016/141209
    • Non Patent Literature 1: Sharpe, A. H, Wherry, E. J., Ahmed R., and Freeman G. J., The function of programmed cell death 1 and its ligands in regulating autoimmunity and infection. Nature Immunology (2007); 8: 239-245.
    • Non Patent Literature 2: Dong H et al., Tumor-associated B7-H1 promotes T-cell apoptosis: a potential mechanism of immune evasion. Nat Med. 2002 August; 8(8): 793-800.
    • Non Patent Literature 3: Yang et al., PD-1 interaction contributes to the functional suppression of T-cell responses to human uveal melanoma cells in vitro. Invest Ophthalmol Vis Sci. 2008 June; 49(6 (2008): 49: 2518-2525.
    • Non Patent Literature 4: Ghebeh et al., The B7-H1 (PD-L1) T lymphocyte-inhibitory molecule is expressed in breast cancer patients with infiltrating ductal carcinoma: correlation with important high-risk prognostic factors. Neoplasia (2006) 8: 190-198.
    • Non Patent Literature 5: Hamanishi J et al., Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+T lymphocytes are prognostic factors of human ovarian cancer. Proceeding of the National Academy of Sciences (2007): 104: 3360-3365.
    • Non Patent Literature 6: Thompson R H et al., Significance of B7-H1 overexpression in kidney cancer. Clinical genitourin Cancer (2006): 5: 206-211.
    • Non Patent Literature 7: Nomi, T. Sho, M., Akahori, T., et al., Clinical significance and therapeutic potential of the programmed death-1 ligand/programmed death-1 pathway in human pancreatic cancer. Clinical Cancer Research (2007); 13: 2151-2157.
    • Non Patent Literature 8: Ohigashi Y et al., Clinical significance of programmed death-1 ligand-1 and programmed death-1 ligand 2 expression in human esophageal cancer. Clin. Cancer Research (2005): 11: 2947-2953.
    • Non Patent Literature 9: Inman et al., PD-L1 (B7-H1) expression by urothelial carcinoma of the bladder and BCG-induced granulomata: associations with localized stage progression. Cancer (2007): 109: 1499-1505.
    • Non Patent Literature 10: Shimauchi T et al., Augmented expression of programmed death-1 in both neoplasmatic and nonneoplastic CD4+ T-cells in adult T-cell Leukemia/Lymphoma. Int. J. Cancer (2007): 121: 2585-2590.
    • Non Patent Literature 11: Gao et al., Overexpression of PD-L1 significantly associates with tumor aggressiveness and postoperative recurrence in human hepatocellular carcinoma. Clinical Cancer Research (2009) 15: 971-979.
    • Non Patent Literature 12: Nakanishi J., Overexpression of B7-H1 (PD-L1) significantly associates with tumor grade and postoperative prognosis in human urothelial cancers. Cancer Immunol Immunother. (2007) 56: 1173-1182.
    • Non Patent Literature 13: Hino et al., Tumor cell expression of programmed cell death-1 is a prognostic factor for malignant melanoma. Cancer (2010): 116: 1757-1766.
    • Non Patent Literature 14: Ghebeh H., Foxp3+ tregs and B7-H1+/PD-1+T lymphocytes co-infiltrate the tumor tissues of high-risk breast cancer patients: implication for immunotherapy. BMC Cancer. 2008 Feb. 23; 8:57.
    • Non Patent Literature 15: Ahmadzadeh M. et al., Tumor antigen-specific CD8 T cells infiltrating the tumor express high levels of PD-1 and are functionally impaired. Blood (2009) 114: 1537-1544.
    • Non Patent Literature 16: Thompson R H et al., PD-1 is expressed by tumor infiltrating cells and is associated with poor outcome for patients with renal carcinoma. Clinical Cancer Research (2007) 15: 1757-1761.
    SUMMARY
  • The present invention is directed to provide a new pharmaceutical composition for treating tumor.
  • The present inventors have studied diligently and, as a result, found that the combined administration of a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist exhibits unexpected antitumor effect, thereby completing the present invention.
  • Accordingly, the present disclosure is as follows.
  • [1] A pharmaceutical composition for treating tumor, comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof, wherein the pharmaceutical composition is administered in combination with a PD-1 antagonist.
    [2] A pharmaceutical composition for treating tumor, comprising a PD-1 antagonist, wherein the pharmaceutical composition is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
    [3] The pharmaceutical composition according to [1] or [2] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [4] The pharmaceutical composition according to any of [1] to [3] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [5] The pharmaceutical composition according to any of [1] to [4] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [6] The pharmaceutical composition according to [5] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [7-1] The pharmaceutical composition according to any of [1] to [6] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
    [7-2] The pharmaceutical composition according to any of [1] to [6] above, wherein the tumor is breast cancer.
    [8] A therapeutic agent for tumor, comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof wherein the pharmaceutical composition is administered in combination with a PD-1 antagonist.
    [9] A therapeutic agent for tumor, comprising a PD-1 antagonist, wherein the therapeutic agent is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
    [10] The therapeutic agent according to [8] or [9] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [11] The therapeutic agent according to any of [8] to [10] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [12] The therapeutic agent according to any of [8] to [11] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [13] The therapeutic agent according to [12] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [14-1] The therapeutic agent according to any of [8] to [13] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
    [14-2] The therapeutic agent according to any of [8] to [13] above, wherein the tumor is breast cancer.
    [15] A method for treating tumor, comprising administering a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist to a patient in need thereof.
    [16] The method according to [15] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [17] The method according to [15] or [16] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [18] The method according to any of [15] to [17] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [19] The method according to [18] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [20-1] The method according to any of [15] to [19] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
    [20-2] The method according to any of [15] to [19] above, wherein the tumor is breast cancer.
    [21] Use of eribulin or a pharmaceutically acceptable salt thereof in the manufacture of a pharmaceutical composition for treating tumor, wherein the pharmaceutical composition is administered in combination with a PD-1 antagonist.
    [22] Use of a PD-1 antagonist in the manufacture of a pharmaceutical composition for treating tumor, wherein the pharmaceutical composition is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
    [23] The use according to [21] or [22] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [24] The use according to any of [21] to [23] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [25] The use according to any of [21] to [24] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [26] The use according to [25] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [27-1] The use according to any of [21] to [26] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
    [27-2] The use according to any of [21] to [26] above, wherein the tumor is breast cancer.
    [28] A liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof for use in tumor treatment, wherein the liposomal composition is administered in combination with a PD-1 antagonist.
    [29] A PD-1 antagonist for use in tumor treatment, wherein the PD-1 antagonist is administered in combination with a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof.
    [30] The liposomal composition or PD-1 antagonist for use according to [28] or [29] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [31] The liposomal composition or PD-1 antagonist for use according to any of [28] to [30] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [32] The liposomal composition or PD-1 antagonist for use according to any of [28] to [31] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [33] The liposomal composition or PD-1 antagonist for use according to [32] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [34-1] The liposomal composition or PD-1 antagonist for use according to [28] to [33] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancer, esophageal cancer and small cell lung cancer.
    [34-2] The liposomal composition or PD-1 antagonist for use according to [28] to [33] above, wherein the tumor is breast cancer.
    [35] A kit for treating tumor, comprising a formulation comprising a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a formulation comprising a PD-1 antagonist.
    [36] The kit according to [35] above, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously, separately, continuously, or at a time interval.
    [37] The kit according to [35] or [36] above, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
    [38] The kit according to any of [35] to [37] above, wherein the PD-1 antagonist is an anti-PD-1 antibody.
    [39] The kit according to [38] above, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
    [40-1] The kit according to any of [35] to [39] above, wherein the tumor is selected from the group consisting of breast cancer, gastric cancel; esophageal cancer and small cell lung cancer.
    [40-2] The kit according to any of [35] to [39] above, wherein the tumor is breast cancer.
    [41] The pharmaceutical composition according to any of [1] to [7-2] above or therapeutic agent according to any of [8] to [14-2] above, further comprising a pharmaceutically acceptable carrier.
  • The combined administration of a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist exhibits unexpected antitumor effect.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.1 mg/kg and an anti-PD-1 antibody on tumor growth.
  • FIG. 2 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.1 mg/kg and an anti-PD-1 antibody on T×5.
  • FIG. 3 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.3 mg/kg and an anti-PD-1 antibody on tumor growth.
  • FIG. 4 is a graph illustrating effect of treatment with a combination of a liposomal formulation comprising eribulin mesylate at a dose of 0.3 mg/kg and an anti-PD-1 antibody on T×5.
  • DETAILED DESCRIPTION
  • Embodiments of the present disclosure will be described below. The following embodiments are illustrations for the purpose of describing the present disclosure and not intended to limit the present disclosure only to these embodiments. The present disclosure can be carried out in various forms, unless they deviate from its spirit.
  • The liposomal compositions in the present disclosure comprises eribulin or a pharmaceutically acceptable salt thereof (hereinafter referred to as “eribulin or the like”).
  • In the present disclosure, the “pharmaceutically acceptable salt” may be either an inorganic acid salt or an organic acid salt and is not particularly limited, as long as it forms a salt with eribulin, and examples thereof include hydrochloride, sulfate, citrate, hydrobromide, hydroiodide, nitrate, bisulfate, phosphate, superphosphate, isonicotinate, acetate, lactate, salicylate, tartrate, pantothenate, ascorbate, succinate, maleate, fumarate, gluconate, saccharinate, formate, benzoate, glutamate, mesylate (methanesulfonate), ethanesulfonate, benzenesulfonate, p-toluenesulfonate, and pamoate. In one embodiment, the pharmaceutically acceptable salts are hydrochloride, sulfate, acetate, phosphate, citrate, and mesylate. In a particular embodiment, the pharmaceutically acceptable salt is mesylate.
  • The pharmaceutically acceptable salt of eribulin may be a salt of eribulin and aluminum, calcium, lithium, magnesium, sodium, zinc, or diethanolamine.
  • In the present disclosure, examples of eribulin or the like include eribulin mesylate.
  • Eribulin or the like is a compound or a salt thereof described in Patent Literature 1 or U.S. Pat. No. 6,214,865 and has pharmacological activities including antitumor and antimitotic activities. Patent Literature 1 discloses that eribulin or the like has, as an antitumor agent, anti-tumor activity against melanoma, fibrosarcoma, monocytic leukemia, colon cancer, ovarian cancer, breast cancel; osteosarcoma, prostate cancer, lung cancer, and ras-transformed fibroblasts. Eribulin or the like is obtained by a method of production described in Patent Literatures 1 to 3.
  • In the present disclosure, the “liposome” means a closed microvesicle having an inner phase surrounded by a lipid bilayer. The liposomes include small unilamellar liposomes (SUVs: small unilamellar vesicles), large unilamellar liposomes (LUVs: large unilamellar vesicles), further large unilamellar liposomes (GUVs: giant unilamellar vesicles), multi-lamellar liposomes having a plurality of concentric membranes (MLVs: multi lamellar vesicles), liposomes having a plurality of non-concentric, irregular membranes (MVVs: multivesicular vesicles), and the like.
  • In the present disclosure, the “liposomal inner phase” means an aqueous region surrounded by a liposomal lipid bilayer and is used synonymously with an “inner aqueous phase” and a “liposomal inner aqueous phase”. The “liposomal outer phase” means a region that is not surrounded by a liposomal lipid bilayer (that is, the region except the inner phase and the lipid bilayer) when the liposome is dispersed in a liquid.
  • In the present disclosure, the “liposomal composition” means a composition comprising a liposome and further comprising eribulin or the like in the liposomal inner phase. In the present disclosure, the liposomal composition includes solid and liquid compositions.
  • In the present disclosure, the “liposomal dispersion liquid” means a composition comprising a liposome in which eribulin or the like is not yet encapsulated into the liposomal inner phase.
  • In the present disclosure, the “liposomal preparatory liquid” means a composition comprising a liposome in which an adjustment of the liposome outer phase in order to encapsulate eribulin or the like into the liposome inner phase is not yet performed.
  • [Lipid]
  • In one embodiment, the liposome preferably comprises a phospholipid and/or a phospholipid derivative as a membrane component.
  • Examples of the phospholipid and/or phospholipid derivative include phosphatidylethanolamine, phosphatidylcholine, phosphatidylserine, phosphatidylinositol, phosphatidylglycerol, cardiolipin, sphingomyelin, ceramide phosphoryl ethanolamine, ceramidephosphorylglycerol, ceramidephosphorylglycerolphosphate, 1,2-dimyristoyl-1,2-deoxyphosphatidyl choline, plasmalogen, and phosphatidate.
  • The phospholipid and/or phospholipid derivative may be one or a combination of two or more of these.
  • Fatty acid residues in the phospholipid and/or phospholipid derivative are not particularly limited, and examples thereof include saturated or unsaturated fatty acid residues having 12 to 20 carbon atoms, and specific examples thereof include acyl groups derived from fatty acids such as lauric acid, myristic acid, palmitic acid, stearic acid, oleic acid, and linoleic acid. As the phospholipid and/or phospholipid derivative, a phospholipid derived from a natural product such as egg yolk lecithin and soy lecithin, and partially hydrogenated egg yolk lecithin, (fully) hydrogenated egg yolk lecithin, partially hydrogenated soy lecithin, and (fully) hydrogenated soybean lecithin, in which unsaturated fatty acid residues are partially or fully hydrogenated, or the like may be used.
  • The amount (molar fraction) of the phospholipid and/or phospholipid derivative, used in the preparation of the liposome, that are/is blended is not particularly limited, and is, in one embodiment, 10 to 80% and, in a particular embodiment, 30 to 60% based on the total ribosomal membrane components.
  • In the present disclosure, the liposome may comprise, as a membrane component, a sterol such as cholesterol and cholestanol and a fatty acid having a saturated or unsaturated acyl group having 8 to 22 carbon atoms as a membrane stabilizing agent, and an antioxidant such as α-tocopherol, besides the phospholipid and/or phospholipid derivative.
  • The amount (molar fraction) of the sterol, used in the preparation of the liposome, that is blended is not particularly limited, and is, in one embodiment, 1 to 60%, 10 to 50%, or 30 to 50% based on the total liposomal membrane components.
  • The amount (molar fraction) of the fatty acid blended is not particularly limited, and is, in one embodiment, 0 to 30% and 0 to 20% or 0 to 10% based on the total liposomal membrane components.
  • The amount (molar fraction) of the antioxidant blended is not particularly limited, as long as an amount that provides the antioxidant effect is added, and it is, in one embodiment, 0 to 15%, 0 to 10%, or 0 to 5% based on the total liposomal membrane components.
  • In the present disclosure, the liposome may comprise a functional lipid or a modified lipid as a membrane component.
  • Examples of the functional lipid include a blood-retaining lipid derivative, a temperature change-sensitive lipid derivative, and a pH-sensitive lipid derivative.
  • Examples of the modified lipid include a PEGylated lipid, a glycolipid, an antibody-modified lipid, and a peptide-modified lipid.
  • Examples of the blood-retaining lipid derivative include polyethylene glycol derivatives (such as methoxy polyethylene glycol condensates) such as condensation products of phosphoethanolamine and methoxy polyethylene glycol: N-{carbonyl-methoxy polyethylene glycol-2000}-1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine, N-{carbonyl-methoxy polyethylene glycol-5000}-1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine, N-{carbonyl-methoxy polyethylene glycol-750}-1,2-distearoyl-sn-glycero-3-phosphoethanolamine, N-{carbonyl-methoxy polyethylene glycol-2000}-1,2-distearoyl-sn-glycero-3-phosphoethanolamine, (MPEG2000-distearoylphosphatidylethanolamine), and N-{carbonyl-methoxy polyethylene glycol-5000}-1,2-distearoyl-sn-glycero-3-phosphoethanolamine.
  • The blending amount (molar fraction) of the blood-retaining lipid derivative, used in the preparation of the liposome, is not particularly limited, and is, in one embodiment, 0 to 50%, 0 to 30%, or 0 to 20% based on the total liposomal membrane components.
  • Examples of the temperature change-sensitive lipid derivative include dipalmitoylphosphatidylcholine. By including a temperature change-sensitive lipid derivative in a liposome, it becomes possible to disrupt the liposome at a particular temperature, to change the surface properties of the liposome, and the like. Furthermore, by combining it with heating of a target site such as tumor, it becomes possible to disrupt the liposome at the target site and have an active compound released at the target site, and the like.
  • Examples of the pH-sensitive lipid derivative include dioleoylphosphatidylethanolamine. By including a pH-sensitive lipid derivative in a liposome, it becomes possible to promote membrane fusion of the liposome and an endosome when the liposome is taken in a cell by endocytosis and improve the delivery of the active compound to the cytoplasm, and the like.
  • Examples of the glycolipid, antibody-modified lipid, and peptide-modified lipid include lipids linked to a sugar, an antibody, or a peptide having an affinity for a target cell or a target tissue. Using a modified lipid allows to deliver the liposome actively to the target cell or the target tissue.
  • The composition of membrane components for liposome having a practically acceptable level of membrane permeability can be set by a person skilled in the art as appropriate depending on the active compound, the target tissue, and the like (see Hiroshi Kikuchi et al. “Liposome I—How to prepare and assay—(in Japanese)” Cell technology (1983) 2 (9): pp. 1136-1149 and the references cited in the reference, and the like). The liposomal composition may be used not only in targeting at a target tissue such as solid cancel; but also in the delivery of an active compound to blood cancer or the like.
  • The liposomal membrane components include, in one embodiment, phospholipid, cholesterol, and a methoxy polyethylene glycol condensation product.
  • [Liposomal Composition]
  • In the liposomal composition in the present disclosure, eribulin or the like is encapsulated in a liposome having a lipid membrane. In the liposomal composition, eribulin or the like may be distributed in the lipid bilayer.
  • The liposomal composition according to the present disclosure can be obtained by a method described in Patent Literature 7.
  • If the liposomal composition is solid, the solid liposomal composition may be dissolved or suspended in a certain solvent described below to prepare a liquid liposomal composition. In case that the liposomal composition is a frozen solid, the frozen solid liposomal composition may be thawed by leaving the composition at room temperature or the like to prepare a liquid liposomal composition.
  • The liposomal composition according to the present disclosure is not limited, as long as it comprises (1) eribulin or the like. The liposomal composition according to the present disclosure may further comprise (2) at least one ammonium salt and (3) at least one acid, salt, base, and/or amino acid.
  • Examples of the at least one ammonium salt (2) include ammonium chloride, ammonium borate, ammonium sulfate, ammonium formate, ammonium acetate, ammonium citrate, ammonium tartrate, ammonium succinate, and ammonium phosphate and, in one embodiment, the at least one ammonium salt (2) is ammonium sulfate, ammonium citrate, and ammonium tartrate.
  • As for the acid, salt, base, and/or amino acid (3), examples of the acid include ascorbic acid, benzoic acid, succinic acid, citric acid, glutamic acid, phosphoric acid, acetic acid, propionic acid, tartaric acid, carbonic acid, lactic acid, boric acid, maleic acid, fumaric acid, malic acid, adipic acid, hydrochloric acid, and sulfuric acid; examples of the salt include sodium salts of the aforementioned acids, potassium salts of the aforementioned acids, and ammonium salts of the aforementioned acids; examples of the base include trishydroxymethylaminomethane, ammonia, sodium hydroxide, and potassium hydroxide; and examples of the amino acid include arginine, histidine, and glycine.
  • In one embodiment of the liposomal composition according to the present disclosure, the acid, salt, base, and/or amino acid (3) in the liposomal inner phase is hydrochloric acid, acetic acid, lactic acid, tartaric acid, succinic acid, citric acid, and phosphoric acid, sodium salts of the aforementioned acids, and sodium hydroxide and ammonia, and, in a particular embodiment, the acid, salt, base, and/or amino acid (3) is acetic acid, lactic acid, tartaric acid, citric acid, and phosphoric acid, sodium salts of the aforementioned acids, and sodium hydroxide and ammonia.
  • An example of the components of the liposomal composition is set forth in Table 1. In another specific example, 96 mg/mL sucrose may be used, instead of 9 mg/mL sodium chloride, as an osmotic agent (liposomal outer phase).
  • TABLE 1
    Component Concentration Purpose of inclusion
    Eribulin mesylate 0.2 mg/mL Drug
    HSPC[1] 7.1 mg/mL Lipid membrane component
    Cholesterol 2.3 mg/mL Lipid membrane component
    MPEG2000-DSPE[2] 2.7 mg/mL Lipid membrane component
    Ammonium sulfate
    100 mM Liposomal inner phase component
    Citric acid monohydrate 30 mM Liposomal inner phase component
    Sodium chloride 9 mg/mL Liposomal outer phase component
    L-histidine 1.6 mg/mL Liposomal outer phase component
    Sodium hydroxide/hydrochloric acid q.s. pH adjuster
    [1]Hydrogenated soy phosphatidylcholine
    [2]N-{carbonyl-methoxy polyethylene glycol-2000}-1,2-distearoyl-sn-glycero-3-phosphoethanolamine (MPEG2000-distearoylphosphatidylethanolamine)
  • The liposomal composition according to the present disclosure may be administered by injection (intravenous injection, intraarterial injection, local injection), orally, nasally, transdermally, transpulmonarily, ophthalmically, and the like, and examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to a target cell and organ. Examples of the dosage form of the liposomal composition for oral administration include tablets, powders, granules, syrups, capsules, and oral solutions. Examples of the dosage form of the liposomal composition for parenteral administration include injections, drip infusions, ophthalmic liquids, ointments, suppositories, suspensions, cataplasms, lotions, aerosols, and plasters, and, in one embodiment, the liposomal composition for parenteral administration is an injection or a drip infusion. The liposomal composition according to the present disclosure may be formulated by a method, for example, described in Japanese Pharmacopoeia (JP) 17th edition, United States Pharmacopoeia (USP), or European pharmacopoeia (EP).
  • In a case that the liposomal composition is a liquid, the liposomal composition may be used as it is. To use the liposomal composition as a medicine, for example, a solvent may be injected by a physician or a patient into a vial in which a solid formulation is encapsulated to do such preparation upon use. A solid formulation obtained by freezing a liquid liposomal composition may be stored in a frozen state and thawed by leaving at room temperature or thawed rapidly with heating back into a liquid upon use to be used as a liquid.
  • The dose upon administration of the liposomal composition alone vary markedly depending on the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like. The liposomal composition is administered, for example, at 0.1 to 10 mg/m2 (body surface) in terms of eribulin mesylate per day for an adult. In one embodiment, the liposomal composition is administered at a dose of 0.5 to 3 mg/m2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks. In a particular embodiment, the liposomal composition is more preferably administered at a dose of 0.5 to 2 mg/m2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks.
  • In another aspect, the liposomal composition is preferably administered at a dose of approximately 1.5 mg/m2 (body surface) in terms of eribulin mesylate once every 1 week, 2 weeks, or 3 weeks.
  • More specifically, the liposomal composition is administered intravenously at 0.5 to 1.4 mg/m2 on day 1 of a 21-day cycle or administered intravenously at 0.5 to 1.5 mg/m2 on day 1 and day 15 of a 28-day cycle in terms of eribulin mesylate.
  • Eribulin or the like contained in the liposomal composition may be administered once a day or in several divided daily doses.
  • The liposomal composition may be a liposomal composition comprising, for example, 0.01 to 300 mg/mL of eribulin or the like in the liposomal inner phase.
  • The liposomal composition is formulated, for example, as an injection comprising 0.20 mg/mL eribulin mesylate (0.18 mg/mL eribulin) incorporated in a liposome having a lipid membrane consisting of HSPC, cholesterol, and MPEG2000-DSPE. Such an injection may comprise sucrose or sodium chloride as an isotonizing agent, ammonium sulfate, citric acid, and L-histidine, and sodium hydroxide and hydrochloric acid to adjust pH. The injection is directly administered to a patient or diluted with physiological saline to a concentration in the range of 0.0035 mg/mL or higher and lower than 0.2 mg/mL before the administration to a patient.
  • The PD-1 antagonist in the present disclosure may comprise any compound or biological molecule that blocks the binding of PD-L1 expressed in cancer cells to PD-1 expressed in immune cells (T cells, B cells, or natural killer T (NKT) cells), or that blocks the binding of PD-L2 expressed in cancer cells to PD-1 expressed in immune cells. The PD-1 antagonist blocks the binding of human PD-L1 to human PD-1 and, in one embodiment, blocks the binding of both human PD-L1 and PD-L2 to human PD-1. The amino acid sequence of human PD-1 can be found in NCBI Locus No.: NP_005009. The amino acid sequences of human PD-L1 and PD-L2 can be found in NCBI Locus No: NP_054862 and NP_079515, respectively.
  • The PD-1 antagonist useful in the present disclosure may comprise a monoclonal antibody (mAb) or an antigen-binding fragment thereof that specifically binds to PD-1 or PD-L1 or that specifically binds to human PD-1 or human PD-L1. The mAb may be a human antibody, a humanized antibody, or a chimeric antibody and may comprise a human constant region. The human constant region is selected from the group consisting of IgG1, IgG2, IgG3, and IgG4 constant regions and, in one embodiment, the human constant region is an IgG1 or IgG4 constant region. The antigen-binding fragment may be selected from the group consisting of Fab, Fab′-SH, F(ab)2, scFv, and Fv fragment.
  • An example of useful PD-1 antagonists is an anti-PD-1 antibody, which is, in one embodiment, an anti-human PD-1 antibody and, in a particular embodiment, an anti-human PD-1 monoclonal antibody (anti-human PD-1 mAb). Examples of the human PD-1-binding mAb binding are described in U.S. Pat. Nos. 7,488,802, 7,521,051, 8,008,449, 8,354,509, 8,168,757, WO 2004/004771, WO 2004/072286, WO 2004/056875, and US Patent Application Publication No. 2011/0271358. Anti-human PD-1 monoclonal antibodies useful as the PD-1 antagonist according to the present disclosure include nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
  • The PD-1 antagonist according to the present disclosure may be administered by injection (intravenous injection, intraarterial injection, local injection), orally, nasally, transdermally, transpulmonarily, ophthalmically, and the like and examples thereof include injection such as intravenous injection, subcutaneous injection, intradermal injection, intraarterial injection, as well as local injection to target cells and organ. Examples of the dosage form of the PD-1 antagonist for oral administration include tablets, powders, granules, syrups, capsules, and oral solutions. Examples of the dosage form of the PD-1 antagonist for parenteral administration include injections, drip infusions, ophthalmic liquids, ointments, suppositories, suspensions, cataplasms, lotions, aerosols, and plasters and in one embodiment, the dosage form of the PD-1 antagonist for parenteral administration is an injection or a drip infusion. The PD-1 antagonist according to the present disclosure may be formulated by a method, for example, described in Japanese Pharmacopoeia (JP) 17th edition, United States Pharmacopoeia (USP), or European pharmacopoeia (EP).
  • If the PD-1 antagonist according to the present disclosure is an anti-PD-1 antibody, the anti-PD-1 antibody may be provided as a liquid preparation or prepared by rehydrating freeze-drying powder with sterile water for injection before use.
  • Upon administration of an anti-human PD-1 mAb alone as the PD-1 antagonist to a patient, the dose thereof varies markedly depending on the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like. The anti-human PD-1 mAb is administered, for example, at a dose of 1, 2, 3, 5, or 10 mg/kg at intervals of approximately 14 days (±2 days), approximately 21 days (±2 days), or approximately 30 days (±2 days).
  • When pembrolizumab is administered as the PD-1 antagonist, pembrolizumab is, for example, administered intravenously at a dose selected from the group consisting of 1 mg/kg Q2W, 2 mg/kg Q2W, 3 mg/kg Q2W, 5 mg/kg Q2W, 10 mg of Q2W, 1 mg/kg Q3W, 2 mg/kg Q3W, 3 mg/kg Q3W, 5 mg/kg Q3W, and 10 mg Q3W. Pembrolizumab is administered as a liquid medicine, for example, comprising 25 mg/ml pembrolizumab, 7% (w/v) sucrose, and 0.02% (w/v) polysorbate 80 in a 10 mM histidine buffer, pH 5.5, and a selected dose of the medicine is administered by IV injection over a period of approximately 30 minutes.
  • When nivolumab is administered as the PD-1 antagonist, nivolumab is, for example, administered intravenously at a dose selected from the group consisting of 1 mg/kg Q2W, 2 mg/kg Q2W, and 3 mg/kg Q2W.
  • The doses of the liposomal composition and the PD-1 antagonist in the combined administration of the present disclosure may usually be set at doses lower than the doses when they are administered alone. Specific doses, administration routes, administration frequencies, and administration cycles are determined as appropriate in consideration of the kind of the target disease, the age, sex, body weight of the patient, the severity of symptoms, and the like.
  • The mode of administration of the liposomal composition and the PD-1 antagonist in the present disclosure is not particularly limited, as long as the liposomal composition and the PD-1 antagonist are administered in combination when they are administered. For example, the liposomal composition and the PD-1 antagonist are administered to a patient simultaneously, separately, continuously, or at a time interval. Here, “simultaneously” means that each component is administered in the same period of time or strictly simultaneously or via the same administration route. “Separately” means that each component is administered at different dose intervals or frequencies or via different administration routes. “Continuously” means that each component is administered via the same administration route or different administration routes in any order within a certain period of time. “At a time interval” means that each component is administered via the same administration route or different administration routes, with each component administered at a time interval. When the PD-1 antagonist is administered in a period of 1 cycle of the administration of the liposomal composition or in a period in which the cycle is repeated, it is considered that both are administered in combination.
  • Tumors to be treated in the present disclosure are, for example, breast cancer, gastric cancer, esophageal cancer, small cell lung cancer, colorectal cancer, and kidney cancer.
  • EXAMPLES [Example 1] Antitumor Effect of Combined Administration of Low Dose of Eribulin Mesylate (0.1 mg/kg) or Low Dose of Eribulin Mesylate Liposomal Formulation (0.1 mg/kg) and Anti-Mouse PD-1 Antibody in Syngeneic Transplantation Model of Murine Breast Cancer 4T1 Cell Line (Pgp-KO 4T1) with P Glycoprotein Knock-Out
  • A P glycoprotein-knockout cell line produced from murine breast cancer 4T1 cells (purchased from ATCC) was cultured using RPMI1640 medium (SIGMA) containing 10% of FBS (fetal bovine serum), 1 mM sodium pyruvate, and antibiotics, under conditions at 37° C. in a 5% carbon dioxide gas incubator. The cells were collected using trypsin-EDTA when the cells reached to approximately 80% confluency. The medium described above (RPMI1640) was added to the collected cells, a suspension was prepared at 1.0×107 cells/mL, and 0.1 mL of the suspension was subcutaneously transplanted at the right body side into 6 mice (BALB/cAJcl, CLEA Japan, Inc.) per each group of the control group, eribulin mesylate alone administration group, eribulin mesylate liposomal formulation alone administration group, the anti-mouse PD-1 antibody (Bio X cell) alone administration group, eribulin mesylate and anti-mouse PD-1 antibody combined administration group, and eribulin mesylate liposomal formulation and anti-mouse PD-1 antibody combined administration group. From day 5 post-transplantation, eribulin mesylate (0.1 mg/kg, once a week, twice in total, tail vein injection), eribulin mesylate liposomal formulation (0.1 mg/kg, once a week, twice in total, tail vein injection), and the anti-mouse PD-1 antibody (200 μg/mouse, once a week, twice in total, tail vein injection) were each administered alone or in combination to the alone administration groups or the anti-mouse PD-1 antibody combined administration groups. No drug was administered to the control group. The maximum tolerated dose of eribulin mesylate liposomal formulation in mice is 2.5 mg/kg and the dose in this experiment was set very low at 0.1 mg/kg, which is 1/25 of the maximum tolerated dose.
  • The liposomal composition comprising eribulin mesylate was prepared with the components set forth in Table 1 in accordance with the following method.
  • <Preparation of Aqueous Solution for Liposomal Inner Phase>
  • Ammonium sulfate and citric acid monohydrate were dissolved and diluted with pure water to prepare an aqueous solution of 200 mM ammonium sulfate/60 mM citric acid. The aqueous solution of 200 mM ammonium sulfate/60 mM citric acid was adjusted to pH 5.5 with an aqueous ammonium solution and then diluted with pure water to obtain an aqueous solution of 100 mM ammonium sulfate/30 mM citric acid.
  • <Preparation of Liposomal Preparatory Liquid>
  • Hydrogenated soy phosphatidylcholine, cholesterol, and MPEG2000-distearoylphosphatidylethanolamine were weighted in accordance with a weight ratio of 71:23:27, respectively. These were each dissolved in chloroform and these solutions were mixed. Chloroform was then evaporated under reduced pressure in a rotary evaporator to prepare a lipid film. To the obtained lipid film, the prepared aqueous solution for liposomal inner phase heated to approximately 80° C. was added and the resulting mixture was stirred to prepare a liposomal preparatory liquid. Sizing was performed using an extruder (a product made by Lipex Biomembranes Inc.) heated to approximately 80° C. to obtain a sized liposomal preparatory liquid.
  • <Preparation of Liposomal Dispersion Liquid>
  • By eluting the obtained liposomal preparatory liquid through a Sephadex G-50 column with an aqueous solution of 0.9% sodium chloride/10 mM histidine (pH=7.6), the liposomal outer phase was exchanged into an aqueous solution of 0.9% sodium chloride/10 mM histidine. After exchanging the liposomal outer phase, the liquid was centrifuged at 400,000×g for 30 minutes. After the centrifugation, re-dispersion was performed and the liquid volume was adjusted with an aqueous solution of 0.9% sodium chloride/10 mM histidine to obtain a liposomal dispersion liquid.
  • <Preparation of Eribulin Mesylate Solution>
  • 0.9% of eribulin mesylate was dissolved in an aqueous solution of sodium chloride/10 mM histidine to obtain an eribulin mesylate solution.
  • <Preparation of Liposomal Composition>
  • The liposomal dispersion liquid and eribulin mesylate solution were mixed in a glass container and incubated in a water bath at 60° C. for 3 minutes to obtain a liposomal composition with a liposomal inner phase in which eribulin mesylate was introduced. An aqueous solution of 0.9% sodium chloride/10 mM histidine was added to the liposomal composition and filter sterilization was performed with a 0.22 μm polyvinylidene fluoride (PVDF) filter to obtain an eribulin mesylate liposomal composition.
  • On day 3, day 7, day 10, day 13, day 16, day 20, day 23, day 27, day 30, and day 34 after administration, with the starting date of administration being day 0, the longest diameter and the short axis of the tumor grown in each mouse were measured with a digimatic caliper (a product made by Mitutoyo Corporation).
  • The tumor volume was calculated in accordance with the following formula.

  • Tumor volume (mm3)=longest diameter (mm)×short axis2 (mm2)/2
  • The results of measurement of the tumor volume in each group are illustrated as mean and standard deviation (SD) in FIG. 1. As statistical analysis, repeated measures analysis of variance (ANOVA) followed by Dunnett's multiple comparison was conducted in comparison with the control group for tumor volumes on all measurement days in all groups (*: p<0.05, ***: p<0.001). The statistical comparison between the two groups of eribulin mesylate and anti-mouse PD-1 antibody combined administration group and eribulin mesylate liposomal formulation and anti-mouse PD-1 antibody combined administration group was conducted by repeated measures ANOVA (#: p<0.05).
  • As a result, the combined administration of a low dose of eribulin mesylate liposomal formulation (0.1 mg/kg) and an anti-mouse PD-1 antibody exhibited a remarkable antitumor effect in comparison with the control group in the Pgp-KO 4T1 syngeneic tumor transplantation model. * and *** in FIG. 1 indicate that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with the control group. # indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with the combined administration of eribulin mesylate and the anti-mouse PD-1 antibody. In contrast, no antitumor effect was observed at low doses with eribulin mesylate (0.1 mg/kg) alone administration, eribulin mesylate liposomal formulation (0.1 mg/kg) alone administration, the anti-mouse PD-1 antibody alone administration, and even the combined administration of eribulin mesylate (0.1 mg/kg) and the anti-mouse PD-1 antibody.
  • The result of comparison of the groups for the time until the tumor volume exceeds 5 times that on the starting date of administration (T×5) in the experiment is shown in FIG. 2 and the median of T×5 in each group and the percentage (%) thereof to the control group are set forth in Table 2. For statistical analysis, a log-rank test was conducted in comparison with the control group to calculate the Bonferroni-corrected p-value (*: p<0.05).
  • As a result, the combined administration of eribulin mesylate liposomal formulation (0.1 mg/kg) and the anti-mouse PD-1 antibody exhibited the effect of extending (243%) the time of suppressing tumor growth (T×5) in Pgp-KO 4T1 syngeneic tumor transplantation model. In contrast, no extension effect was observed at low doses with eribulin mesylate (0.1 mg/kg) alone administration, eribulin mesylate liposomal formulation (0.1 mg/kg) alone administration, the anti-mouse PD-1 antibody alone administration, and further the combined administration of eribulin mesylate and the anti-mouse PD-1 antibody. * in Table 2 indicates that the combined administration of eribulin mesylate liposomal formulation (0.1 mg/kg) and the anti-mouse PD-1 antibody statistically significantly extended the time of tumor growth suppression in comparison with the control group.
  • TABLE 2
    Effect of the combined administration of eribulin mesylate liposomal
    formulation (0.1 mg/kg) and anti-mouse PD-1 antibody on T × 5
    Percentage to
    Group T × 5 (days) control
    Control 10.5 100%
    Eribulin mesylate Alone 12.0 114%
    Eribulin mesylate liposomal formulation Alone 11.5 110%
    Anti-mouse PD-1 antibody Alone 14.5 138%
    Eribulin mesylate + Anti-mouse 14.5 138%
    PD-1 antibody Combined administration
    Eribulin mesylate liposomal formulation + 25.5* 243%
    Anti-mouse PD-1 antibody Combined administration
  • [Example 2] Antitumor Effect of Combined Administration of Low Dose of Eribulin Mesylate (0.3 mg/kg) or Low Dose of Eribulin Mesylate Liposomal Formulation (0.3 mg/kg) and Anti-Mouse PD-1 Antibody in Pgp-KO 4T1 Cell Line Syngeneic Transplantation Model
  • Pgp-KO 4T1 cells were cultured with the RPMI1640 medium containing 10% FBS, 1 mM sodium pyruvate, and antibiotics under conditions at 37° C. in a 5% carbon dioxide gas incubator. The cells were collected using trypsin-EDTA when the cells reached to approximately 80% confluency. The medium described above was added to the collected cells to prepare a suspension at 1.0×107 cells/mL. 0.1 mL of the cell suspension was subcutaneously transplanted at the right body side into 6 mice (BALB/cAJcl, CLEA Japan, Inc.) per each group of the control group, eribulin mesylate liposomal formulation alone administration group, the anti-mouse PD-1 antibody (Bio X cell) alone administration group, and the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody. From day 4 post-transplantation, eribulin mesylate liposomal formulation (0.3 mg/kg, once a week, twice in total, tail vein injection) and the anti-mouse PD-1 antibody (200 μg/mouse, once a week, twice in total, tail vein injection) were each administered alone or in combination to the alone administration groups or the combined administration group. No drug was administered to the control group. The liposomal composition comprising eribulin mesylate was prepared in accordance with the method as that in Example 1.
  • On day 3, day 7, day 9, day 13, day 17, day 20, day 24, day 27, day 31, day 34, day 38, day 41, day 44, day 48, and day 51 after administration, with the starting date of administration being day 0, the longest diameter and the short axis of the tumor grown in each mouse were measured with a digimatic caliper (a product made by Mitutoyo Corporation).
  • The tumor volume was calculated in accordance with the following formula.

  • Tumor volume (mm3)=longest diameter (mm)×short axis2 (mm2)/2
  • The mean and standard deviation (SD) of the results of measurement of the tumor volume in each group are illustrated in FIG. 3 and the frequencies of mice with tumor disappearance are set forth in Table 3. For statistical analysis, the statistical comparison between the two groups of eribulin mesylate liposomal formulation alone administration group or the anti-mouse PD-1 antibody alone administration group and eribulin mesylate liposomal formulation and anti-mouse PD-1 antibody combined administration group was conducted by repeated measures ANOVA (t, #: p<0.05).
  • As a result, in the Pgp-KO 4T1 syngeneic tumor transplantation model, the combined administration of a low dose of eribulin mesylate liposomal formulation (0.3 mg/kg) and the anti-mouse PD-1 antibody exhibited excellent antitumor effect in comparison with eribulin mesylate liposomal formulation alone administration group or the anti-mouse PD-1 antibody alone administration group. In FIG. 3, (t) indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with eribulin mesylate liposomal formulation alone administration and (#) indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly inhibited tumor growth in comparison with the anti-mouse PD-1 antibody alone administration. The tumor disappearance was observed in the combined administration group of eribulin mesylate liposomal formulation (0.3 mg/kg) and the anti-mouse PD-1 antibody at a frequency higher than other groups.
  • TABLE 3
    Frequency of appearance of mice with tumor disappearance in each group
    Frequency (%) of mice with tumor
    Group disappearance
    Control
    0/6 (0%)
    Eribulin mesylate liposomal formulation Alone 1/6 (17%)
    Anti-mouse PD-1 antibody Alone 0/6 (0%)
    Eribulin mesylate liposomal formulation + 4/6 (67%)
    Anti-mouse PD-1 antibody Combined administration
  • The result of comparison of the groups for the time until the tumor volume exceeds 5 times that on the starting date of administration (T×5) in the experiment is shown in FIG. 4 and the median of T×5 in each group and the percentage (%) thereof to the control group are set forth in Table 4. For statistical analysis, a log-rank test between the 2 groups of the combined administration group of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody to the anti-mouse PD-1 antibody, alone administration group was conducted (#: p<0.05).
  • As a result, in the Pgp-KO 4T1 syngeneic tumor transplantation model, the combined administration of eribulin mesylate liposomal formulation (0.3 mg/kg) and the anti-mouse PD-1 antibody exhibited the effect of extending suppression time of tumor growth (T×5) in comparison with the control group, eribulin mesylate liposomal formulation alone administration group, the anti-mouse PD-1 antibody alone administration group. # in FIG. 4 indicates that the combined administration of eribulin mesylate liposomal formulation and the anti-mouse PD-1 antibody statistically significantly extended suppression time of tumor growth in comparison with the anti-mouse PD-1 antibody alone administration.
  • TABLE 4
    Effect of combined administration of eribulin mesylate liposomal
    formulation (0.3 mg/kg) and anti-mouse PD-1 antibody to T × 5
    Group T × 5 (days) Ratio to control
    Control 11.5 100%
    Eribulin mesylate liposomal formulation Alone 33.0 287%
    Anti-mouse PD-1 antibody Alone 18.0 157%
    Eribulin mesylate liposomal formulation + >51.0# >443% 
    Anti-mouse PD-1 antibody Combined administration

Claims (19)

1-7. (canceled)
8. A method for treating a tumor, comprising administering a liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and a PD-1 antagonist to a patient in need thereof, wherein the PD-1 antagonist is an anti-PD-1 antibody, and wherein the tumor is selected from the group consisting of gastric cancer, esophageal cancer, and small cell lung cancer.
9. The method according to claim 8, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered simultaneously.
10. The method according to claim 8, wherein the liposomal composition comprising eribulin or a pharmaceutically acceptable salt thereof and the PD-1 antagonist are administered separately.
11. The method according to claim 8, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
12. The method according to claim 8, wherein the anti-PD-1 antibody is selected from the group consisting of nivolumab, pembrolizumab, cemiplimab, sintilimab, and toripalimab.
13. The method according to claim 8, wherein the tumor is gastric cancer.
14. The method according to claim 12, wherein the tumor is gastric cancer.
15. The method according to claim 8, wherein the tumor is esophageal cancer.
16. The method according to claim 12, wherein the tumor is esophageal cancer.
17. The method according to claim 8, wherein the tumor is small cell lung cancer.
18. The method according to claim 12, wherein the tumor is small cell lung cancer.
19. The method according to claim 12, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
20. The method according to claim 13, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
21. The method according to claim 14, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
22. The method according to claim 15, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
23. The method according to claim 16, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
24. The method according to claim 17, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
25. The method according to claim 18, wherein eribulin or the pharmaceutically acceptable salt thereof is eribulin mesylate.
US17/180,107 2019-07-26 2021-02-19 Pharmaceutical composition for treating tumor Abandoned US20210177802A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US17/180,107 US20210177802A1 (en) 2019-07-26 2021-02-19 Pharmaceutical composition for treating tumor

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
JP2019-138041 2019-07-26
JP2019138041 2019-07-26
US16/835,719 US11083705B2 (en) 2019-07-26 2020-03-31 Pharmaceutical composition for treating tumor
US17/180,107 US20210177802A1 (en) 2019-07-26 2021-02-19 Pharmaceutical composition for treating tumor

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US16/835,719 Continuation US11083705B2 (en) 2019-07-26 2020-03-31 Pharmaceutical composition for treating tumor

Publications (1)

Publication Number Publication Date
US20210177802A1 true US20210177802A1 (en) 2021-06-17

Family

ID=74187564

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/835,719 Active US11083705B2 (en) 2019-07-26 2020-03-31 Pharmaceutical composition for treating tumor
US17/180,107 Abandoned US20210177802A1 (en) 2019-07-26 2021-02-19 Pharmaceutical composition for treating tumor

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/835,719 Active US11083705B2 (en) 2019-07-26 2020-03-31 Pharmaceutical composition for treating tumor

Country Status (14)

Country Link
US (2) US11083705B2 (en)
EP (1) EP3970745A4 (en)
JP (1) JPWO2021020336A1 (en)
KR (1) KR20220041044A (en)
CN (1) CN113993545A (en)
AU (1) AU2020323402A1 (en)
BR (1) BR112021026170A2 (en)
CA (1) CA3143486A1 (en)
IL (1) IL289213A (en)
MA (1) MA55982A (en)
MX (1) MX2021016093A (en)
TW (1) TW202118486A (en)
WO (1) WO2021020336A1 (en)
ZA (1) ZA202110686B (en)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12029724B2 (en) 2016-04-28 2024-07-09 Eisai R&D Management Co., Ltd. Method for inhibiting tumor growth
US12036204B2 (en) 2019-07-26 2024-07-16 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating tumor
US12042560B2 (en) 2009-03-30 2024-07-23 Eisai R&D Management Co., Ltd. Liposome composition

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN116407492A (en) * 2021-12-29 2023-07-11 北京新领先医药科技发展有限公司 Eribulin mesylate injection composition and preparation method thereof

Family Cites Families (66)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JPS5551683B2 (en) 1972-10-23 1980-12-25
US5736155A (en) 1984-08-08 1998-04-07 The Liposome Company, Inc. Encapsulation of antineoplastic agents in liposomes
IL91664A (en) 1988-09-28 1993-05-13 Yissum Res Dev Co Ammonium transmembrane gradient system for efficient loading of liposomes with amphipathic drugs and their controlled release
GB9126209D0 (en) 1991-12-10 1992-02-12 Orion Yhtymae Oy Drug formulations for parenteral use
ES2188612T3 (en) 1993-04-22 2003-07-01 Skyepharma Inc MULTIVESICULAR CYCLODEXTRINE LIPOSOMES TO ENCAPSULATE PHARMACOLOGICAL COMPOUNDS AND METHODS FOR USE.
FR2736056B1 (en) 1995-06-29 1997-08-08 Commissariat Energie Atomique CYCLODEXTRIN DERIVATIVES, THEIR PREPARATION AND THEIR USE FOR INCORPORATING HYDROPHOBIC MOLECULES IN ORGANIZED SURFACTANT SYSTEMS
US6051251A (en) 1997-11-20 2000-04-18 Alza Corporation Liposome loading method using a boronic acid compound
CN1216051C (en) 1998-06-17 2005-08-24 卫材株式会社 Macrocyclic analogs and methods of their use and preparation
US6653341B1 (en) 1998-06-17 2003-11-25 Eisai Co., Ltd. Methods and compositions for use in treating cancer
US8097648B2 (en) 1998-06-17 2012-01-17 Eisai R&D Management Co., Ltd. Methods and compositions for use in treating cancer
US6593308B2 (en) 1999-12-03 2003-07-15 The Regents Of The University Of California Targeted drug delivery with a hyaluronan ligand
CN1116875C (en) 2000-10-19 2003-08-06 南京振中生物工程有限公司 Taxusol-lipid composition and its preparing process
WO2003041682A2 (en) 2001-11-13 2003-05-22 Celator Technologies, Inc. Lipid carrier compositions and methods for improved drug retention
US20050118250A1 (en) 2001-11-13 2005-06-02 Paul Tardi Lipid carrier compositions with enhanced blood stability
EP3287144A1 (en) 2002-07-03 2018-02-28 ONO Pharmaceutical Co., Ltd. Immunopotentiating compositions
DE10255106A1 (en) 2002-11-24 2004-06-09 Novosom Ag Liposomal glucocorticoids
EP1567130A2 (en) 2002-11-26 2005-08-31 Gilead Sciences, Inc. Method of drug loading in liposomes by gradient
JP4511943B2 (en) 2002-12-23 2010-07-28 ワイス エルエルシー Antibody against PD-1 and use thereof
EP1435231B8 (en) 2002-12-31 2010-03-03 Zydus BSV Pharma Private Limited Non-pegylated long-circulating liposomes
WO2004072286A1 (en) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specific to human pd-1
JPWO2004089419A1 (en) 2003-04-04 2006-07-06 国立大学法人 東京大学 Lipid membrane structure containing anti-MT-MMP monoclonal antibody
US6747011B1 (en) 2003-06-04 2004-06-08 A.P. Group, Inc. Antitumor drugs and methods
US20050129753A1 (en) 2003-11-14 2005-06-16 Gabizon Alberto A. Method for drug loading in liposomes
AU2005207786B2 (en) 2004-01-29 2010-11-25 Eisai R & D Management Co., Ltd Method of stabilizing macrolide compound
CA2821167C (en) 2004-05-03 2016-06-28 Merrimack Pharmaceuticals, Inc. Drug delivery liposomes containing anionic polyols or anionic sugars
AU2005251691A1 (en) 2004-05-17 2005-12-22 Tekmira Pharmaceuticals Corporation Liposomal formulations comprising dihydrosphingomyelin and methods of use thereof
EP2949652B1 (en) 2004-06-03 2019-08-07 Eisai R&D Management Co., Ltd. Intermediates for the preparation of halichondrin B
US8349360B2 (en) 2004-10-06 2013-01-08 Bc Cancer Agency Liposomes with improved drug retention for treatment of cancer
KR101339628B1 (en) 2005-05-09 2013-12-09 메다렉스, 인코포레이티드 Human monoclonal antibodies to programmed death 1 (pd-1) and methods for treating cancer using anti-pd-1 antibodies alone or in combination with other immunotherapeutics
DK1907424T3 (en) 2005-07-01 2015-11-09 Squibb & Sons Llc HUMAN MONOCLONAL ANTIBODIES TO PROGRAMMED death ligand 1 (PD-L1)
TWI373473B (en) 2005-09-02 2012-10-01 Otsuka Pharma Co Ltd Anticancer agent
CN101209243B (en) 2006-12-29 2010-12-08 石药集团中奇制药技术(石家庄)有限公司 Liposome medicament and preparation thereof
US20090196913A1 (en) 2007-05-11 2009-08-06 Ken Shi Kun Huang Anti-Alpha-V Immunoliposome Composition, Methods, and Uses
BRPI0812913B8 (en) 2007-06-18 2021-05-25 Merck Sharp & Dohme monoclonal antibodies or antibody fragment to human programmed death receptor pd-1, polynucleotide, method of producing said antibodies or antibody fragments, composition comprising them and use thereof
MX2010003599A (en) 2007-10-03 2010-09-10 Eisai R&D Man Co Ltd Intermediates and methods for the synthesis of halichondrin b analogs.
US20090196918A1 (en) 2008-02-01 2009-08-06 University Of Kentucky Research Foundation Liposomal formulations of hydrophobic lactone drugs in the presence of metal ions
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
CA2735006A1 (en) 2008-08-25 2010-03-11 Amplimmune, Inc. Pd-1 antagonists and methods of use thereof
ES2592216T3 (en) 2008-09-26 2016-11-28 Dana-Farber Cancer Institute, Inc. Human anti-PD-1, PD-L1 and PD-L2 antibodies and their uses
CN114835812A (en) 2008-12-09 2022-08-02 霍夫曼-拉罗奇有限公司 anti-PD-L1 antibodies and their use for enhancing T cell function
JP5622719B2 (en) 2009-03-30 2014-11-12 エーザイ・アール・アンド・ディー・マネジメント株式会社 Method for producing liposome composition
LT2415470T (en) 2009-03-30 2016-10-10 Eisai R&D Management Co., Ltd. Liposome composition
JP5388735B2 (en) 2009-07-21 2014-01-15 浜松ホトニクス株式会社 Microchannel plate
JP2013512251A (en) 2009-11-24 2013-04-11 アンプリミューン、インコーポレーテッド Simultaneous inhibition of PD-L1 / PD-L2
CA2787919C (en) 2010-01-26 2018-07-31 Eisai R&D Management Co., Ltd. Furo [3, 2 -b] pyrane derivatives useful in the synthesis of halichondrin b analogs
SG10201602147YA (en) 2011-03-18 2016-05-30 Eisai R&D Man Co Ltd Methods And Compositions For Predicting Response To Eribulin
US9220776B2 (en) 2011-03-31 2015-12-29 Merck Sharp & Dohme Corp. Stable formulations of antibodies to human programmed death receptor PD-1 and related treatments
WO2012139039A2 (en) 2011-04-08 2012-10-11 British Columbia Cancer Agency Branch Bisphenol compounds and methods for their use
EP2734191A4 (en) 2011-07-19 2015-04-29 Stc Unm Intraperitoneally-administered nanocarriers that release their therapeutic load based on the inflammatory environment of cancers
KR20190133790A (en) 2011-08-01 2019-12-03 제넨테크, 인크. Methods of treating cancer using pd-1 axis binding antagonists and mek inhibitors
RU2689977C2 (en) 2012-12-04 2019-05-30 Эйсай Ар Энд Ди Менеджмент Ко., Лтд. Using eribulin for treating breast cancer
US20160067337A1 (en) 2013-03-14 2016-03-10 Bristol-Myers Squibb Company Combination of dr5 agonist and anti-pd-1 antagonist and methods of use
WO2014193898A1 (en) 2013-05-31 2014-12-04 Merck Sharp & Dohme Corp. Combination therapies for cancer
BR112015029386B1 (en) 2013-06-26 2023-11-14 Eisai R&D Management Co., Ltd. USE OF ERIBULIN AND LENVATINIB AS COMBINATION THERAPY AND KIT
EP3046543A1 (en) 2013-09-18 2016-07-27 Stc.Unm Torroidal mesoporous silica nanoparticles (tmsnps) and related protocells
US9457019B2 (en) 2013-11-07 2016-10-04 Deciphera Pharmaceuticals, Llc Methods for inhibiting tie-2 kinase useful in the treatment of cancer
WO2015095784A1 (en) 2013-12-19 2015-06-25 Luminus Biosciences, Inc. Solid nanoparticle formulation of microtubule inhibitors with reduced ostwald ripening for oral administration
JOP20200094A1 (en) 2014-01-24 2017-06-16 Dana Farber Cancer Inst Inc Antibody molecules to pd-1 and uses thereof
CA2940983A1 (en) 2014-03-03 2015-09-11 Eisai R&D Management Co., Ltd. Use of eribulin and mtor inhibitors as combination therapy for the treatment of cancer
EP3114144A1 (en) 2014-03-05 2017-01-11 Bristol-Myers Squibb Company Treatment of renal cancer using a combination of an anti-pd-1 antibody and another anti-cancer agent
EP3148336B1 (en) 2014-05-28 2019-10-02 Eisai R&D Management Co., Ltd. Eribulin and poly (adp ribose) polymerase (parp) inhibitors as combination therapy for the treatment of cancer
CN105640935A (en) 2014-11-12 2016-06-08 天津市汉康医药生物技术有限公司 Eribulin mesylate pharmaceutical composition for injection
AU2016226157B2 (en) 2015-03-04 2022-01-27 Eisai R&D Management Co., Ltd. Combination of a PD-1 antagonist and eribulin for treating cancer
EP3449921B1 (en) 2016-04-28 2023-05-31 Eisai R&D Management Co., Ltd. Eribulin for inhibiting tumor growth
WO2018071792A1 (en) 2016-10-14 2018-04-19 Merck Sharp & Dohme Corp. Combination of a pd-1 antagonist and eribulin for treating urothelial cancer
AR112603A1 (en) * 2017-07-10 2019-11-20 Lilly Co Eli BIS SPECIFIC ANTIBODIES CONTROL POINT INHIBITORS

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US12042560B2 (en) 2009-03-30 2024-07-23 Eisai R&D Management Co., Ltd. Liposome composition
US12029724B2 (en) 2016-04-28 2024-07-09 Eisai R&D Management Co., Ltd. Method for inhibiting tumor growth
US12036204B2 (en) 2019-07-26 2024-07-16 Eisai R&D Management Co., Ltd. Pharmaceutical composition for treating tumor

Also Published As

Publication number Publication date
MA55982A (en) 2022-03-23
TW202118486A (en) 2021-05-16
JPWO2021020336A1 (en) 2021-02-04
KR20220041044A (en) 2022-03-31
US11083705B2 (en) 2021-08-10
WO2021020336A1 (en) 2021-02-04
CA3143486A1 (en) 2021-02-04
US20210023047A1 (en) 2021-01-28
BR112021026170A2 (en) 2022-02-15
IL289213A (en) 2022-02-01
EP3970745A1 (en) 2022-03-23
CN113993545A (en) 2022-01-28
EP3970745A4 (en) 2023-07-19
AU2020323402A1 (en) 2022-01-27
MX2021016093A (en) 2022-02-03
ZA202110686B (en) 2022-09-28

Similar Documents

Publication Publication Date Title
US11083705B2 (en) Pharmaceutical composition for treating tumor
AU2018269742B2 (en) Nano-enabled immunotherapy in cancer
US20230241244A1 (en) Nano-enabled immunotherapy in cancer
US12036204B2 (en) Pharmaceutical composition for treating tumor
EP3449921B1 (en) Eribulin for inhibiting tumor growth
US20240285640A1 (en) Combination therapy for treating pik3ca-mutated cancer
US20210213051A1 (en) Combined pharmaceutical formulation comprising drug-containing liposome composition and platinum preparation
EP4045054A1 (en) Nano-enabled immunotherapy in cancer
JP6560215B2 (en) Curcumphenol compounds for increasing the expression of MHC-I
CN110891944B (en) Compounds, compositions and uses thereof for the treatment of cancer
US20210100791A1 (en) Combined pharmaceutical formulation comprising drug-containing liposome composition and immune checkpoint inhibitor
EP4353223A1 (en) Application of pharmaceutical composition having specific drug-to-lipid ratio in antitumor
WO2023081514A1 (en) Cxcr4 antagonist loaded liposomes and silicasomes
US11896713B2 (en) Strategies to enhance lung cancer treatment
WO2023172300A1 (en) A drug nanocarrier system to deliver a combination of tlr agonists and/or a lipoxin plus immunogenic cell death inducing chemotherapeutic agents for cancer immunotherapy
JP2012508749A (en) Combination of bendamustine, doxorubicin and bortezomib for the treatment of multiple myeloma

Legal Events

Date Code Title Description
STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

AS Assignment

Owner name: EISAI R&D MANAGEMENT CO., LTD., JAPAN

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:SEMBA, TARO;FUNAHASHI, YASUHIRO;REEL/FRAME:055559/0780

Effective date: 20200318

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STCB Information on status: application discontinuation

Free format text: EXPRESSLY ABANDONED -- DURING EXAMINATION