CN115607678A - Combination therapy for the treatment of cancer - Google Patents

Combination therapy for the treatment of cancer Download PDF

Info

Publication number
CN115607678A
CN115607678A CN202110791354.2A CN202110791354A CN115607678A CN 115607678 A CN115607678 A CN 115607678A CN 202110791354 A CN202110791354 A CN 202110791354A CN 115607678 A CN115607678 A CN 115607678A
Authority
CN
China
Prior art keywords
cancer
virus
inhibitor
pi3k
dosage form
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.)
Pending
Application number
CN202110791354.2A
Other languages
Chinese (zh)
Inventor
何南海
邱庆崇
路杨
杨东晖
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.)
Hangzhou Arnold Biomedical Technology Co ltd
Original Assignee
Hangzhou Arnold Biomedical Technology 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 Hangzhou Arnold Biomedical Technology Co ltd filed Critical Hangzhou Arnold Biomedical Technology Co ltd
Priority to CN202110791354.2A priority Critical patent/CN115607678A/en
Priority to CN202280013623.5A priority patent/CN116887837A/en
Priority to PCT/CN2022/104040 priority patent/WO2023284597A1/en
Priority to TW111125796A priority patent/TW202317134A/en
Publication of CN115607678A publication Critical patent/CN115607678A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/761Adenovirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/763Herpes virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/765Reovirus; Rotavirus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/766Rhabdovirus, e.g. vesicular stomatitis virus
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/66Microorganisms or materials therefrom
    • A61K35/76Viruses; Subviral particles; Bacteriophages
    • A61K35/768Oncolytic viruses not provided for in groups A61K35/761 - A61K35/766
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Virology (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Oncology (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Biomedical Technology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present application provides a method of treating a tumor/cancer and/or generating a memory immune response against a tumor/cancer in a subject in need thereof, comprising administering to the subject an effective amount of a PI3K inhibitor, an oncolytic virus, and a PD-1 inhibitor, wherein the PI3K inhibitor, the oncolytic virus, and the PD-1 inhibitor can be administered simultaneously, separately, or sequentially. The present application also provides a pharmaceutical composition comprising a PI3K inhibitor, an oncolytic virus and a PD-1 inhibitor, and a pharmaceutically acceptable carrier, and a kit comprising the pharmaceutical composition and instructions for use.

Description

Combination therapy for the treatment of cancer
Technical Field
The present invention relates to a combination therapeutic method and pharmaceutical composition comprising administering a PI3K inhibitor, an oncolytic virus and a PD-1 inhibitor to a subject in need thereof.
Background
Immune Checkpoint Inhibitors (ICI) using anti-CTLA 4 antibodies and anti-PD (L) 1 antibodies have revolutionized cancer immunotherapy. Since the first Immune Checkpoint Inhibitor (ICI) approval in 2001, this series of drugs has attracted extensive attention due to the long-lasting clinical effects on complex cancers. However, there are still a large number of cancer patients who do not respond to ICI therapy. The response rate of cancer patients treated by ICI to date is about 20%, so it is very necessary to find a more effective immunotherapy approach to further treat cancer with the immune system.
pan-I type P13K inhibitors
The occurrence rate of the PI3K-AKT-mTOR signaling pathway disorder is high in cancer patients, and the disorder mainly regulates key cellular processes such as cell growth, proliferation and metabolism, and can promote the survival, expansion and spread of cancer cells in the cancer patients. PI3K isoform p110 α encoded by the PIK3CA gene is the second most easily mutated proto-oncogene in human cancers.
In the PI3K family, PI3K β is also involved in tumorigenesis, but PI3K α plays a major role, and in addition, two other isoforms, PI3K γ and PI3K δ, play important roles in regulating the immune system. PI3K δ is able to regulate and maintain the function of regulatory T cells (tregs), and PI3K γ is able not only to recruit inhibitory myeloid cells into the cancer microenvironment, but also to enhance their suppressive capacity for the anti-cancer effects of T cells.
The compound with the structure shown in the formula I (AN 2025) and pharmaceutically acceptable salts thereof are oral pan-I PI3K inhibitors, and are currently in the third clinical development stage, and the indication is head and neck squamous cell carcinoma. It can not only inhibit wild PI3 alpha, but also inhibit mutant P13K alpha, and the mutation sites include (H1047R, E542K and E545K). At the same time, it also has inhibitory activity against other PI3K isomers (PI 3K β, PI3K γ, PI3K δ). AN2025 has been clinically proven effective for the development of breast cancer caused by PI3K/PIK3CA mutation, and the patients with the highest response rate all had PIK3CA mutation (Campone M et al Eur J cancer.2018; 103. AN2025 can also down-regulate Tregs and inhibitory myeloid cell function and its ability to enter the tumor microenvironment by inhibiting PI3K δ and PI3K γ (O' Donnell JS et al Semin Cancer biol.2018; 48-103 Borazanci et al the Oncol.2020.
Figure BDA0003161060360000021
Oncolytic virus
Oncolytic viruses are a class of replication competent tumor-killing viruses. Besides tumor cell lysis caused by selective infection of tumor cells, the oncolytic virus can further kill tumors by activating immune reaction. On one hand, tumor cells infected by oncolytic virus can release a large amount of inflammatory factors, activate natural killer cells (NK cells), and start innate immune response to kill tumors. The inflammatory environment can promote migration of natural killer cells, dendritic cells, and T cells into the tumor microenvironment, turning "cold" tumors "hot. On the other hand, oncolytic viruses lyse tumor cells to produce large amounts of tumor-associated antigens, and antigen presenting cells kill tumor cells by processing and presenting tumor-associated antigens and virus-associated antigens to T cells, activating adaptive immune responses.
Pelareorecep is a non-pathogenic, naturally occurring unmodified reovirus type 3. At present, the cancer is about to enter the clinical third stage of the global multicenter, and the indication is advanced metastatic breast cancer. Good experimental results have been obtained in a previous second phase clinical study with pelareoreep in combination with paclitaxel for the treatment of advanced/metastatic breast cancer.
anti-PD (L) 1 antibodies
The PD1 and PDL1 signaling pathways play a key role in regulating T cell activation processes. PDL1 is expressed primarily in immune cells as well as in most human cancer cells. In the tumor microenvironment, cancer PDL1 interacts with PD1 on T cells, eventually inhibiting the cancer cell killing activity of T cells. The large number of successful cases of anti-PD (L) 1 therapy makes it the cornerstone of modern tumor immunotherapy.
Pelareorecep can kill tumor cells in a specific mode to generate an inflammatory tumor microenvironment, promote immune cells to infiltrate into tumor tissues and change cold tumors into hot tumors; AN2025 is able to systematically modulate the immunosuppressive tumor microenvironment, both of which are complementary to the mechanisms of anti-PD (L) 1 therapy to kill cancer. This triple combination provides an ideal treatment option for most patients with advanced solid tumors.
Disclosure of Invention
The technical problem to be solved by the invention is to solve the problem of low response rate of cancer patients treated by ICI in the prior art. It is an object of the present invention to provide a more effective immunotherapy regime to further utilize the immune system to treat cancer.
In view of this, the present invention provides a method of treating cancer using a combination of a PI3K inhibitor, an oncolytic virus, and a PD-1 inhibitor, wherein the method has an anti-tumor activity significantly superior to any combination of two.
Specifically, the invention provides the following technical scheme:
a method of treating a tumor/cancer and/or generating a memory immune response against a tumor/cancer in a subject in need thereof, comprising administering to the subject an effective amount of a PI3K inhibitor, an oncolytic virus, and a PD-1 inhibitor, wherein the PI3K inhibitor, oncolytic virus, and PD-1 inhibitor can be administered simultaneously, separately, or sequentially.
In one embodiment of the present invention, wherein the PI3K inhibitor is selected from PI3K α, PI3K β, PI3K γ, and PI3K δ subtype inhibitors.
In one embodiment of the invention, wherein the PI3K inhibitor is selected from the group consisting of Idelalisib, copalisib, duvelisib, alpelisib, seletilisib, gedatolisib, rigoserinib sodium, lenilisib, umbralisib, buparlisib (AN 2025), AMG-319, GM-604, acalisib, bimiralisb, GDC-0084, ACP-319, tenalisib, serabelisib, SF-1126, nemiralisb, fimepinostat, LY-3024, voxtalisib, daxtalisib, parsaclisib, GSK-2636771, AZD-8186, ASN-003, and any combination thereof.
In one embodiment of the present invention, wherein the PI3K inhibitor is a compound having formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0003161060360000031
in one embodiment of the invention, wherein the oncolytic virus comprises a naturally occurring, modified or recombinant virus having replication capacity and capable of infecting and lysing tumor cells.
In one embodiment of the invention, wherein the oncolytic virus is selected from the group consisting of reovirus (reovirus), newcastle disease virus (Newcastle disease virus), vesicular stomatitis virus (vesicular stomatis virus), adenovirus (adeno virus), vaccinia virus (vaccinia virus), filterable virus (paradox orf virus), sindbis virus (Sindbis virus) and herpes simplex virus (herpes simplex virus).
In one embodiment of the present invention, wherein the PD-1 inhibitor is selected from: anti-PD-1, anti-PD-L1, and anti-PD-L2 antibodies; preferably, wherein the PD-1 inhibitor is selected from the group consisting of Nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, terieprinimab, certolizumab, temeprizumab, tirlizumab, carprilizumab, and any combination thereof.
In one embodiment of the invention, the PI3K inhibitor, oncolytic virus and PD-1 inhibitor may be formulated in a dosage form for oral, buccal or parenteral routes, e.g. the oral route may be in the form of tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the parenteral route may be in the form of intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal penetration, transmucosal administration, e.g. may be in the form of solutions, powder injections or lyophilisates.
In one embodiment of the invention, wherein the PI3K inhibitor is administered in a dosage form of oral, buccal or parenteral route, e.g. the dosage form of the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form of the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal.
In one embodiment of the invention, wherein the PI3K inhibitor is administered 1 or 2 times per day; or 1 administration of the PI3K inhibitor every 2, 3, 4,5, 6, 7, 8, 9, 10 days or every 1,2 or 3 weeks; or 1 administration of the PI3K inhibitor daily for 5 consecutive days per week followed by 2 days intervals.
In one embodiment of the invention, wherein the PI3K inhibitor is administered in a dosage range of about 20 mg/day to about 200 mg/day, about 30 mg/day to about 160 mg/day, about 60 mg/day to about 120 mg/day in an adult human.
In one embodiment of the invention, wherein the PI3K inhibitor is administered to the subject at an effective dose of about 0.5 to about 250mg/kg, about 1 to about 250mg/kg, about 2 to about 200mg/kg, about 3 to about 120mg/kg, about 5 to about 250mg/kg, about 10 to about 200mg/kg, or about 20 to about 120mg/kg.
In one embodiment of the invention, wherein the oncolytic virus is administered in a dosage form of oral, buccal or parenteral route, e.g. the dosage form of the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form of the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration.
In one embodiment of the invention, wherein the oncolytic virus is administered at least once daily, for repeated administrations on days 2, 3, 4,5, 6, 7, 8, 9, 10, 14, 21 or 28, or for any period of 2-28 days, or longer, for continuous or intermittent administrations of the oncolytic virus per day for 1,2, 3, 4,5, 6, 7, 8, 9, 10, 12 or 24 hours, or for any period of 1-24 hours; preferably, the duration of administration is 5, 15, 30, 60, 90, 120, 150 or 180 minutes or any time from 5 to 180 minutes or longer.
In one embodiment of the present invention, wherein the dose of the oncolytic virus administered in an adult human differs according to individual differences (age, weight, sex), administration of the virus, for example, at about 10 per human 2 -10 17 PFU (colony Forming Unit) or TCID 50 (half the tissue culture infectious dose) administration. In one embodiment of the invention, wherein the amount of PFU/kg is from about 1.0 to about 10 15 Administering an effective dose of PFU/kg to the subject the oncolytic virus.
In one embodiment of the invention, wherein the PD-1 inhibitor is administered by an parenteral route, for example, the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration.
In one embodiment of the invention, the PD-1 inhibitor is prepared into solution, lyophilizate or powder injection.
In one embodiment of the invention, wherein the PD-1 inhibitor is administered at an effective dose of 0.05mg/kg, 0.1mg/kg, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg or 8 mg/kg.
In one embodiment of the present invention, wherein said tumor/cancer comprises a primary tumor/cancer, a recurrent tumor/cancer, or a metastatic tumor/cancer, including solid tumors and hematological tumors.
In one embodiment of the present invention, wherein said tumor/cancer is selected from the group consisting of: head and neck squamous cell carcinoma, head and neck cancer, brain cancer, glioma, glioblastoma multiforme, neuroblastoma, central nervous system cancer, neuroendocrine tumor, laryngeal cancer, nasopharyngeal cancer, esophageal cancer, thyroid cancer, malignant pleural mesothelioma, lung cancer, breast cancer, liver cancer, hepatoma, hepatobiliary cancer, pancreatic cancer, gastric cancer, gastrointestinal cancer, intestinal cancer, colon cancer, colorectal cancer, kidney cancer, clear cell renal cell cancer, ovarian cancer, endometrial cancer, cervical cancer, bladder cancer, prostate cancer, testicular cancer, skin cancer, melanoma, leukemia, lymphoma, bone cancer, chondrosarcoma, myeloma, multiple myeloma, myelodysplastic syndrome, myeloproliferative tumors, squamous cell carcinoma, ewing's sarcoma, systemic light chain amyloidosis, and merkel cell carcinoma; more preferably, the lymphoma is selected from: hodgkin's lymphoma, non-hodgkin's lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, primary mediastinal large B-cell lymphoma, mantle cell lymphoma, small lymphocytic lymphoma, large B-cell lymphoma enriched in T-cells/histiocytes, and lymphoplasmacytic lymphoma, said lung cancer being selected from the group consisting of: non-small cell lung cancer and small cell lung cancer, said leukemia being selected from: chronic myeloid leukemia, acute myeloid leukemia, lymphocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, and myeloid leukemia.
In addition, the invention provides a pharmaceutical composition and/or a pharmaceutical combination comprising a PI3K inhibitor, an oncolytic virus and a PD-1 inhibitor, and a pharmaceutically acceptable carrier.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PI3K inhibitor is selected from PI3K α, PI3K β, PI3K γ, PI3K δ subtype inhibitors.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PI3K inhibitor is selected from the group consisting of Idelalisib, copalisib, duvelisib, alpelisib, seletalisib, gedatolisib, rigoserinib sodium, lenilisib, umbralisib, buparlisib (AN 2025), AMG-319, GM-604, acalisib, bimiralisib, GDC-0084, ACP-319, tenalisib, serabelisib, SF-1126, nemiralisib, fimepinostat, LY-3024, voxtalisib, dactolisib, parsaclisib, GSK-2636771, AZD-8186, ASN-003, and any combination thereof.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PI3K inhibitor is a compound having the formula (I) or a pharmaceutically acceptable salt thereof,
Figure BDA0003161060360000061
in the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the oncolytic virus comprises a naturally occurring, modified or recombinant virus having replication capacity and capable of infecting and lysing tumor cells.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, the oncolytic virus is selected from the group consisting of reovirus (reovirus), newcastle disease virus (Newcastle disease virus), vesicular stomatitis virus (vesicular stomatis virus), adenovirus (adenovirus), vaccinia virus (vaccinia virus), filtering virus (parapox orf virus), sindbis virus (Sindbis virus) and herpes simplex virus (herpes simplex virus).
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PD-1 inhibitor is selected from: anti-PD-1 antibodies, anti-PD-L1 antibodies, and anti-PD-L2 antibodies; preferably, wherein the PD-1 inhibitor is selected from the group consisting of Nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, terepril mab, cedilizumab, terralizumab, cerlizumab, cerilizumab, and any combination thereof.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PI3K inhibitor, the oncolytic virus and the PD-1 inhibitor may be in the same and/or separate dosage form (dosage form).
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the pharmaceutical composition and/or pharmaceutical combination may be formulated in a dosage form for oral, buccal or parenteral routes, for example, the dosage form for the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, for example, the dosage form for the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal, for example, may be a solution, powder injection or lyophilized formulation.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, the PI3K inhibitor may be formulated in a dosage form for oral, buccal or parenteral routes, for example, the dosage form for the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, for example, the dosage form for the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal permeation, transmucosal administration, for example, may be a solution, powder injection or lyophilizate.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the oncolytic virus may be formulated into a dosage form for oral, buccal or parenteral routes, for example, the dosage form for oral routes may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, for example, the dosage form for parenteral routes may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein the PD-1 inhibitor may be formulated into a dosage form for administration in parenteral route, for example, the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration, for example, may be a solution, powder injection or lyophilizate.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, a PI3K inhibitor is included in a dosage of 1-1000mg per unit dosage form (unit dosage form), for example, the dosage is a value between 1,2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, 150, 160, 170, 175, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 750, 800, 900, 1000mg or any two of the above values.
In the pharmaceutical composition and/or pharmaceutical combination of the present invention, wherein about 10 is contained in each unit dosage form (unit dosage form) 2 PFU to about 10 17 Oncolytic virus at PFU dose, e.g. 10 2 、10 3 、10 4 、10 5 、10 6 、10 7 、10 8 、10 9 、10 10 、10 11 、10 12 、10 13 、10 14 、10 15 、10 16 、10 17 PFU or TCID 50 Or a value between any two of the above values.
In a pharmaceutical composition and/or pharmaceutical combination according to the invention, a dose of 1 to 5000mg of the PD-1 inhibitor is comprised per unit dosage form, e.g. a dose of 1,2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, 150, 160, 170, 175, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 750, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 1900, 2000, 2200, 2400, 2500, 2600, 2700, 2750, 2800, 3000, 3500, 4000, 4500, 5000mg or a value between any two of the above values.
The present invention provides a pharmaceutical composition and/or a pharmaceutical combination for use in the treatment of tumors/cancers and/or for generating a memory immune response against tumors/cancers.
The invention provides the use of a pharmaceutical composition and/or a pharmaceutical combination for the manufacture of a medicament for the treatment of a tumor/cancer and/or for generating a memory immune response against a tumor/cancer.
In addition, the present invention provides a kit comprising a pharmaceutical composition and/or a pharmaceutical combination according to the present invention, together with instructions for use, wherein the PI3K inhibitor, the oncolytic virus and the PD-1 inhibitor may be in the same and/or separate containers.
Drawings
FIG. 1 tumor growth curves for a single EMT6 tumor treated with different drug combinations of AN2025, pelareore, and anti-PD-1 antibody. Each group contained 10 mice. Administration of AN 2025: 30mg/kg, orally (PO) once daily. Pelareoreep administration: 2 x 10 8 TCID 50 Once daily for 2 consecutive days, i.v. administration was followed by 4 days off. anti-PD-1 antibody administration: 10mg/kg, intravenous (IV) once every 6 days.
Detailed Description
Definition of terms
Unless otherwise indicated herein, the terms used herein have the ordinary meaning as is understood in the art to which they pertain.
The term "PI3K inhibitor" refers herein to phosphatidylinositol 3-kinase, a highly conserved family of enzymes that are an important component of the intracellular PI3K-Akt-mTOR signaling axis. Examples of PI3K inhibitors include, but are not limited to, PI3K α, PI3K β, PI3K γ, PI3K δ subtype inhibitors. Specific examples include, but are not limited to Idelalisib, copalisib, duvelisib, alpelisib, seletalisib, gedatolisib, rigoserinib sodium, lenilisib, umbralisib, buparlisib (AN 2025), AMG-319, GM-604, acalisib, bimiralisib, GDC-0084, ACP-319, tenalisib, serabelisib, SF-1126, nemirassib, filepistat, LY-3023414, voxtalisib, dactolisib, parsaclisib, GSK-2636771, AZD-8186, ASN-003, and any combination thereof. Preferred examples include, but are not limited to, compounds of formula I (also referred to herein as AN 2025) as taught herein
Figure BDA0003161060360000091
The term "oncolytic virus" refers herein to a virus that is capable of replicating in and killing tumor cells. Including reovirus (reovirus), newcastle disease virus (Newcastle disease virus), vesicular stomatitis virus (vesicular stomatis virus), adenovirus (adenovirus), vaccinia virus (vaccinia virus), filtering virus (parapox orf virus), sindbis virus (Sindbis virus) and herpes simplex virus (herpes simplex virus). Wherein the oncolytic virus includes, but is not limited to, oncolytic viruses of members of the following virus families: myoviridae (myoviridae), leptoviridae (sipoviridae), breptoviridae (podoviridae), cochlamyridae (teciviridae), coticoviridae (corticoidae), blastomyridae (plasmaviridae), lipofencidae (lipothrixviridae), microfusiformes (fuselloviridae), poxviridae (poxyiridae) iridoviridae (iridoviridae), alphaviridae (phycodnaviridae), baculoviridae (baculoviridae), herpesviridae (herpesviridae), adenoviridae (adenoviridae), papovaviridae (papovaviridae), polyvidae (polynaviridae), filoviridae (inoviridae), miniviridae (microviviridae), geminiviridae (geminiviridae), and combinations thereof circoviridae (circoviridae), parvoviridae (parvoviridae), hepadnaviridae (hepadnaviridae), retroviridae (retroviridae), capsoviridae (cyctoviridae), reoviridae (reoviridae), biscnaviridae (birnaviridae), paramyxoviridae (paramyxoviridae), rhabdoviridae (rhabdoviridae), filoviridae (filoviridae) Orthomyxoviridae (Orthomyxoviridae), bunyaviridae (bunyaviridae), arenaviridae (arenaviridae), leviviridae (leviviridae), picornaviridae (picornaviridae), companion viridae (sequisviridae), comoviridae (comoviridae), potyviridae (potyviridae), caliciviridae (caliciviridae), astroviridae (astroviridae), nodaviridae (nodaviridae), tetraviridae (tetraviridae), bushy stunviridae (tombusviridae), coronaviridae (coronaviridae), flaviviridae (glaviviridae), togaviridae (togaviridae) and baculoviridae (barnaviridae). In addition, methods are provided that include immunoprotective viruses as well as recombinant viruses of these viruses. In addition, methods are provided that include the use of a combination of at least two oncolytic viruses. Preferably, the oncolytic virus provided by the present method is reovirus (reovirus). More preferably, the oncolytic virus provided by the method is a serogroup 3 virus-free reovirus (serotype-3 detaching strain). Examples of oncolytic viruses include, but are not limited to, onyx-015, DNX-2401, coloAdl, oncos102, prostAtak, CG0070, pexavec, GLONC1, HF10, HSV1716, reolysin, and Cavatak.
The term "PD-1 inhibitor" refers to an "anti-PD 1/PDL1 antibody," which is an antibody directed against programmed death protein 1 (PD-1)/programmed death protein ligand 1 (PD-L1). Exemplary antibodies include, but are not limited to, antibodies as shown in patent nos. US7,029,674, US7,488,802, US7,521,051, US8,008,449, US8,354,509, US8,617,546, and US8,709,417. Specific examples of PD-1/PD-L1 inhibitors also include Nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, terepril mab, cedilizumab, terralizumab, cerilizumab, rilizumab, and any combination thereof.
"treating," "treatment," and "treating" refer to reducing, inhibiting, and/or reversing the progression of a disease (e.g., tumor/cancer) in a subject in need thereof. The term "treatment" includes any sign of successful treatment or amelioration of a disease, including any objective or subjective parameter, such as remission; moderating; reduced symptoms or making the subject more tolerant to injury, pathology or condition; delay or slow the rate of development, etc. The measurement of treatment or improvement may be based on the results of physical examination, pathological examination, and/or diagnostic examination, for example, as known in the art. For example, in one embodiment, the present invention relates to "treating" cancer by administering a PI3K inhibitor, oncolytic virus, and PD-1 inhibitor of the present invention in combination therapy to a cancer or a subject diagnosed with cancer, to achieve at least one positive therapeutic result, such as a reduction in the number of cancer cells, a reduction in the size of the tumor, a reduction in the rate of tumor cell infiltration into peripheral organs, or a reduction in the rate of tumor metastasis or tumor growth.
Treatment may also refer to reducing the risk of onset or onset of disease, or reducing disease recurrence (e.g., prolonging time to recurrence) as compared to what would occur in the absence of such measures. In the medical field, such treatment is also referred to as "prevention".
The term "effective amount" or "therapeutically effective amount" refers to an amount effective to treat a disease, as noted by clinical testing and evaluation, patient observation, and the like. An "effective amount" may further refer to an amount that causes a detectable change in a biological or chemical activity. Detectable changes can be detected and/or further quantified by one of ordinary skill in the art familiar with relevant mechanisms or methods. Furthermore, an "effective amount" may refer to an amount that maintains a desired physiological state (i.e., reduces or prevents significant deterioration and/or promotes improvement of a condition). An "effective amount" may further refer to a therapeutically effective amount.
In some embodiments, the PI3K inhibitor is administered to the subject in an effective amount. An effective amount is typically 0.01mg/kg body weight to 500mg/kg body weight per day. In some embodiments, pharmaceutically acceptable compositions can be formulated such that a dose of the compound of 0.01mg/kg body weight to 200mg/kg body weight or 0.01mg/kg body weight to 100mg/kg body weight per day can be administered to a subject receiving these compositions (e.g., a dose of 0.75mg to 7.5g or 15g based on a 75kg human). In certain embodiments, the active pharmaceutical ingredient of the present invention is formulated to provide a dose of 0.01mg/kg to 70mg/kg (e.g., a dose of 0.75mg to 5.25g based on a 75kg human).
In some embodiments, an effective dose of a PI3K inhibitor is about 0.5 to about 250mg/kg, about 1 to about 250mg/kg, about 2 to about 200mg/kg, about 3 to about 120mg/kg, about 5 to about 250mg/kg, about 10 to about 200mg/kg, or about 20 to about 120mg/kg. In some embodiments, an effective dose comprises about 0.5mg/kg, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 8mg/kg, 10mg/kg, 20mg/kg, 25mg/kg, 40mg/kg, 50mg/kg, 60mg/kg, 75mg/kg, 100mg/kg, 120mg/kg, 150mg/kg, 175mg/kg, 200mg/kg, 225mg/kg, 250mg/kg, and 300mg/kg. The dosage form may take a variety of suitable forms, such as a tablet or capsule, and an effective dose may be provided in one or more unit dosage forms (e.g., tablets, capsules) and provided 1,2 or 3 times per day, or throughout the day at intervals of, for example, 4, 8 or 12 hours. Tablets or capsules, for example, may contain, for example, 10, 25, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, 1,000, 1,100, or 1,250mg of the compound. For example, administration of a PI3K inhibitor to a human subject in some embodiments may include a daily dose of PI3K inhibitor in the range of 100-1,250, 150-1,000, 200-800, or 250-750mg, which may be administered all once a day, or in multiple portions at regular intervals throughout the day. Liquid formulations may also be prepared so that any dose can be easily and conveniently dispensed.
In some embodiments, the oncolytic virus is administered to the subject in an effective dose. The effective dose is determined according to individual differences, and its influence factors include, but are not limited to, age, sex, body weight and tumor characteristics, such as tumor type and size, etc. For administration to a subject, the effective dose is typically 10 3 -10 12 PFU or TCID 50 . The effective dose may also be 1.0PFU/kg body weight to about 10 15 PFU/kg body weight, e.g. from 10 2 PFU/kg body weight to 10 13 PFU/kg body weight. Preferably, the effective dose is about 1X 10 8 To 1 × 10 12 PFU or TCID 50 . For example, for administration to a subject, the effective dose is about 10 2 To 10 17 PFU。
In some embodiments, the oncolytic virus may be administered to the subject in a single or multiple administrations, which may occur simultaneously or sequentially. For example, administration may last from several days to several weeks. Oncolytic viruses can be administered to one or more tumor sites in the same individual. Dosage forms may take a variety of suitable forms, for example, administration of the oncolytic virus in a dosage form for oral, buccal or parenteral routes, and an effective dose may be provided in one or more unit dosage forms (e.g., tablets and capsules) and provided at least once daily. Tablets and capsules, for example, may contain about 10 2 PFU to about 10 17 PFU dose of oncolytic virus. For example, the dose is 10 2 、10 3 、10 4 、10 5 、10 6 、10 7 、10 8 、10 9 、10 10 、10 11 、10 12 、10 13 、10 14 、10 15 、10 16 、10 17 PFU or TCID 50 Or a value between any two of the above values. The antibody will typically be mixed with a pharmaceutically acceptable non-toxic carrier substance (e.g., physiological saline or phosphate buffered saline) prior to administration, and may be administered using any medically appropriate procedure, including, for example, but not limited to, intravenous or intra-arterial administration and injection into the cerebrospinal fluid. In certain instances, intraperitoneal, intradermal, intracavity, intrathecal, or direct administration to the tumor or to the artery supplying the tumor may be advantageous.
In some embodiments, an effective dose of the antibody is about 5 to about 250mg/kg, about 10 to about 200mg/kg, or about 20 to about 120mg/kg. In some embodiments, an effective dose comprises 5mg/kg, 10mg/kg, 20mg/kg, 25mg/kg, 40mg/kg, 50mg/kg, 60mg/kg, 75mg/kg, 100mg/kg, 120mg/kg, 150mg/kg, 175mg/kg, 200mg/kg, 225mg/kg, 250mg/kg, and 300mg/kg. The dosage form may take the form of, for example, a tablet or capsule, and an effective dose may be provided in one or more tablets, capsules, or the like, and provided once per day or throughout the day at intervals of, for example, 4, 8, or 12 hours. The tablet or capsule may contain, for example, 10, 25, 50, 75, 100, 150, 200, 250, 300, 350, 400, 450, 500, 600, 700, 800, 900, or 1,000mg of antibody. Liquid formulations can also be prepared so that any dose can be easily and conveniently dispensed.
In some embodiments, the antibody is administered to the subject in an effective amount. An effective amount is typically 0.01mg/kg body weight to 500mg/kg body weight per day. In some embodiments, pharmaceutically acceptable compositions may be formulated such that a dose of the compound of 0.01mg/kg body weight to 200mg/kg body weight or 0.01mg/kg body weight to 100mg/kg body weight per day (e.g., a dose of 0.75mg to 7.5g or 15g based on 75kg of human) may be administered to a patient receiving these compositions. In certain embodiments, the compositions of the invention are formulated to provide a dose of 0.01mg/kg to 70mg/kg (e.g., a dose of 0.75mg to 5.25g based on a 75kg human).
An effective amount of the antibody may be, for example, 0.05mg/kg, 0.1mg/kg, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg or 8mg/kg per dose (e.g., a dose of 3.75mg to 600mg based on 75kg of human).
The dose of the antibody may be administered once, twice, three times, four times, five or more times per week, once per two weeks, or even once per three weeks during the course of treatment. The time of administration may be once daily, once every two days, once every three days, once every four days, once every five days, once a week, once every two weeks, or once every three weeks. The formulation comprising the antibody may be prepared such that any dose may be easily and conveniently dispensed.
The term "subject" refers to a mammalian subject, and in particular a human subject, including male or female subjects, and including neonatal, infant, toddler, adolescent, adult or elderly subjects, and further including various ethnic and racial groups, such as the caucasian, african and asian ethnic groups. Herein, the subject suffers from a tumor/cancer. In one embodiment, the tumor patient is not considered to express PD-L1. In another embodiment, the tumor patient is a tumor patient positive for PD-L1 expression.
The term "pharmaceutically acceptable salts" refers to relatively non-toxic inorganic or organic acid salts of the compounds of formula I or formula II of the present invention. These salts can be prepared in situ during the final isolation and purification of the compounds or by reacting the purified compound in free form with a suitable organic or inorganic acid, respectively, and isolating the salt thus formed. Representative acid salts include, but are not limited to, acetate, adipate, aspartate, benzoate, benzenesulfonate, bicarbonate/carbonate, bisulfate/sulfate, borate, camsylate, citrate, cyclamate, edisylate, ethanesulfonate, formate, fumarate, glucoheptonate, gluconate, glucuronate, hexafluorophosphate, salicylate, hydrochloride/chloride, hydrobromide/bromide, hydroiodide, isethionate, lactate, malate, maleate, malonate, methanesulfonate, methylsulfate, napthalate (naphylate), 2-naphthalenesulfonate, nicotinate, nitrate, orotate, oxalate, palmitate, pamoate, phosphate/biphosphate/dihydrogen phosphate, pyroglutamate, gluconate, stearate, succinate, tannate, tartrate, tosylate, trifluoroacetate, and xinafoate. In one embodiment, the pharmaceutically acceptable salt is a hydrochloride/chloride salt.
The terms "tumor" and "cancer" are used interchangeably herein to refer to uncontrolled, abnormal proliferation of local tissues of the body. If the medical intervention is not performed in time, the tumor/cancer can grow uncontrollably and may metastasize to other locations in the body, eventually leading to death of the body.
The term "cancer" may include cancers caused by genetically inherited mutations. Examples of such cancers include, but are not limited to, breast cancer, cancers that may be associated with lyverine syndrome (e.g., childhood sarcoma, leukemia, and brain cancer), cancers that may be associated with lindie syndrome such as colon cancer, cholangiocarcinoma, brain cancer, endometrial cancer, kidney cancer, ovarian cancer, pancreatic cancer, small bowel cancer, stomach cancer, and ureteral cancer, lung cancer, melanoma, prostate cancer, retinoblastoma, thyroid cancer, and uterine cancer.
In addition, cancer can be the result of acquired mutations (e.g., mutations caused by diet, environment, and/or lifestyle) or somatic mutations. <xnotran> () , , , , , , , , , ( , ), , , , , , , , , ( , ), ( B , T , , ), , , , , , , , ( ), , , ( , ), , , , , , , , , , , , , , , , , , , , , , , , , , , , , , </xnotran> Hodgkin's disease, non-hodgkin's lymphoma, soft tissue sarcoma, osteogenic sarcoma, primary macroglobulinemia, seminoma, teratocarcinoma, osteosarcoma, xeroderma pigmentosum, keratoacanthoma, and retinoblastoma.
"metastatic cancer" refers to a cancer in which cancer cells from one organ or body part have spread (by "metastasis") to another non-adjacent organ or body part. Cancers at non-adjacent organs or body parts ("secondary tumors" or "metastatic tumors") include cancer cells derived from organs or body parts from which the cancer or cancer cells have spread. Sites where secondary tumors may occur include, but are not limited to, lymph nodes, lung, liver, brain and/or bone.
The terms "pharmaceutically acceptable" and "pharmaceutically acceptable" are used interchangeably herein and refer to the type generally accepted by those skilled in the pharmaceutical arts. Such as pharmaceutically acceptable salts, pharmaceutically acceptable carriers, and the like.
By "oral dosage form" is meant a pharmaceutical formulation prepared for administration to an individual by the oral route of administration. Examples of known oral dosage forms include, but are not limited to, tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, and the like. In some methods, powders, pills, granules, capsules, and tablets may be coated with suitable polymers or commonly used coating materials to achieve, for example, greater stability in the gastrointestinal tract or to achieve a desired release rate. In addition, the capsule shell of the powder, pill or granule is further coated. The tablets may be scored to facilitate division of the dosage.
When administered as an oral formulation, the compounds of formula I according to the invention are preferably coated with a film. Suitable membranes are known in the art and are commercially available or can be made according to known methods. Typically, the membrane coating material is a hydrophilic polymer such as polyethylene glycol, polyvinylpyrrolidone, polyvinyl alcohol, hydroxypropyl cellulose, hydroxymethyl cellulose, hydroxypropyl methylcellulose, and the like. The film coating composition ingredients may include conventional amounts of plasticizers such as polyethylene glycol, triethyl citrate, diethyl phthalate, propylene glycol, glycerin; and opacifiers such as titanium dioxide and colorants such as iron oxide, aluminum lakes, and the like. Typically, the film coating material is applied in an amount that provides a film coating in the range of, for example, 1% to 6% of the solid oral dosage form.
When the oncolytic virus of the invention is administered as an intravenous drip formulation, two commonly used fluids can be selected: crystalline fluids and colloidal fluids. The crystals are aqueous solutions of mineral salts or other water-soluble molecules. Colloids contain larger insoluble molecules, such as gelatin; blood itself is a colloid. The most commonly used crystal fluid is physiological saline, i.e. a 0.9% sodium chloride solution, which is near to concentration in blood (isotonic). Ringer's lactic acid or ringer's acetic acid is another isotonic solution commonly used for displacement of large volumes of fluid. Other pharmaceutically acceptable carriers include phosphate buffered saline or other physiologically acceptable buffers, lactose, dextrose, sucrose, sorbitol, mannitol, starch, gum powder, calcium phosphate, alginates, scutellaria, gelatin, calcium silicate, microcrystals, cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methylcellulose. The pharmaceutical compositions may also include, but are not limited to, lubricants such as talc, magnesium stearate, and mineral oil; a wetting agent; emulsifying and suspending agents; preservatives, such as methyl and propyl hydroxybenzoate; a sweetener; and a flavoring agent.
The terms "comprising" and "including," as used herein, have an open and non-limiting meaning unless otherwise indicated. For example, in addition to the pharmaceutical combination of a PI3K inhibitor, an oncolytic virus and a PD-1 inhibitor of the present invention, other therapeutic agents, such as "biotherapeutic agents", "chemotherapeutic agents", may be included.
As used herein, "biotherapeutic agent" refers to a biomolecule, such as an antibody or fusion protein, that blocks ligand/receptor activity in any biological pathway that supports tumor maintenance and/or growth or suppresses tumor immune responses. For example, "biotherapeutic agents" include, but are not limited to: alemtuzumab, bevacizumab, bernetuzumab, cetuximab, denosumab, gemtuzumab, ipilimumab, nimotuzumab, ofatumumab, panitumumab, rituximab, tositumomab, trastuzumab, nivaletuzumab, amituzumab, eptizumab, DOVAMAb, and avizumab.
Among these, "chemotherapeutic agents" are chemical compounds that may be used in the treatment of cancer, including, but not limited to, alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytotoxic/antineoplastic biotins, topoisomerase inhibitors, photosensitizers, antiestrogens and selective estrogen receptor modulators, aromatase inhibitors, EGFR inhibitors, VEGF inhibitors, antisense oligonucleotides that inhibit the expression of genes associated with abnormal cell proliferation or tumor growth. <xnotran> , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , , C, , , , , , , , , , α, , , , , , alisertib, amuvatinib, , , , , , , , crenolanib, , , , , , , , foretinib, ganetespib, , , , , iniparib, , lenvatinib, linifanib, linsitinib, , momelotinib, , , , niraparib, oprozomib, olaparib, , pictilisib, , quizartinib, , rigosertib, rucaparib, , , saridegib, , , , tivantinib, , , , , , , , volasertib. </xnotran>
The choice of dosage regimen for the combination therapy of the invention depends on a number of factors, such as: the subject treated has a solid serum or tissue turnover rate, a symptom level, overall immunogenicity, and a degree of accessibility of the target cells, tissues, or organs. Preferably, the dosage regimen maximizes the amount of each therapeutic agent delivered to the patient, consistent with an acceptable level of side effects. Thus, the dosage and frequency of administration of each of the biotherapeutic and chemotherapeutic agents in the combination therapy will depend in part on the particular therapeutic agent, the severity of the cancer being treated and the patient's characteristics.
The combination therapy of the present invention may be administered before or after surgery to remove the tumor, and may be administered before, during, or after radiation therapy.
WO2007084786 specifically describes the compounds of formula I of the present invention, which compounds or their pharmaceutically acceptable salts and processes for their preparation are disclosed in the examples of WO2007084786, which is incorporated herein by reference in its entirety.
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention in any way.
In some embodiments, administration of a PI3K inhibitor, an oncolytic virus in combination with a PD-1 inhibitor results in an enhancement of the PD-1 inhibitor such that, for example, a smaller dose or longer time interval of the PD-1 inhibitor may be effective for treatment.
"antibodies" include all types of immunoglobulins, including IgG, igM, igA, igD, and IgE or fragments thereof, which may be suitable for the medical uses disclosed herein. The antibody may be monoclonal or polyclonal and may be of any species origin, including, for example, mouse, rat, rabbit, horse or human. Antibody fragments that retain specific binding to the protein or epitope (PDL 1 or PD 1) to which the antibody used in the present invention binds are included within the term "antibody". Such fragments may be generated by known techniques. The antibody may be chimeric or humanized, particularly when it is used for therapeutic purposes. Antibodies can be obtained or prepared using methods known in the art.
In order that the invention described herein may be more fully understood, the following examples are set forth. It should be understood that these examples are for illustrative purposes only and are not to be construed as limiting the invention in any way.
DETAILED DESCRIPTION OF EMBODIMENT (S) OF INVENTION
Example 1: combination therapy of a Compound of formula I (AN 2025), AN oncolytic Virus (Pelareore) and AN anti-PD-1 antibody for the treatment of mouse EMT6 tumors
Materials and methods
Reagents and instrumentation: pelareoreep is provided by the company; AN2025 is purchased from MedChem Express. Antibodies against PD-1 from mice (clone number: RMP 1-14) were purchased from BioXcell.
Cell line: mouse breast cancer EMT6 cells (CRL-2755) were purchased from the american tissue culture center. The cells were cultured in DMEM medium supplemented with 10% fetal bovine serum at 37 ℃ and 5% carbon dioxide in an incubator and subcultured twice per week until the necessary number of cells for inoculating the mice was obtained.
Animals: balB/c female mice from 6-8 weeks were purchased from The Jackson Laboratory. Animals were housed in miniature isolation cages and a 12h light/dark cycle was performed. The cages were replaced twice a week. Animals were observed once a week and clinical signs were recorded.
Animal study: EMT6 cells cultured in vitro were harvested at 2X 10 6 The cell concentration of cells/ml was suspended in 100 μ l serum-free medium and the mice were injected subcutaneously (sc) in the lower back on the right side using a syringe. Several days after cell implantation, mice were randomly assigned based on tumor size such that the average tumor volume of each group of mice was 76.82mm 3 . Administration of AN 2025: 30mg/kg, orally (PO) once daily. Pelareorecep administration: 2 x 10 8 TCID 50 The subsequent administration was performed as one administration cycle by intravenous administration once a day for 2 consecutive days and then discontinuation for 4 days. anti-PD-1 antibody administration: 10mg/kg administered by Intravenous (IV) once every 6 days. Tumor size was measured 2-3 times per week with digital caliper and using the formula (l × w) 2 )/2=mm 3 The volume is calculated, where l and w refer to the larger and smaller orthogonal dimensions collected in each measurement. Group tumor size (mean ± SEM) versus time was plotted using GraphPad Prism 9 software. Statistical analysis was performed using the Reproduced Measure (RM) two-way ANOVA test for analysis.
Results of the experiment
As shown in FIG. 1, although any two combinations of Pelareore, anti-PD-1 antibody and AN2025 showed some anti-tumor activity in the mouse breast cancer EMT6 model. However, the combined use of the Pelareoreep, the anti-PD-1 antibody and the AN2025 brings about AN anti-tumor activity superior to that of any two combinations, and is significantly superior to that of the combined use of the AN2025 and the anti-PD-1 antibody (G5 Vs G3, p is less than 0.05).

Claims (37)

1. A method of treating a tumor/cancer and/or generating a memory immune response against a tumor/cancer in a subject in need thereof, comprising administering to the subject an effective amount of a PI3K inhibitor, an oncolytic virus, and a PD-1 inhibitor, wherein the PI3K inhibitor, oncolytic virus, and PD-1 inhibitor can be administered simultaneously, separately, or sequentially.
2. The method of claim 1, wherein the PI3K inhibitor is selected from PI3K α, PI3K β, PI3K γ, PI3K δ subtype inhibitors.
3. The method of claim 1 or 2, wherein the PI3K inhibitor is selected from the group consisting of Idelalisib, copanlisib, duvelisib, alepliisib, seletalisib, gedatolisib, rigosertib sodium, leniolisib, umbralisib, buparlisb (AN 2025), AMG-319, GM-604, acalisib, bimariisib, GDC-0084, ACP-319, tenalisib, serabelisib, SF-1126, nemralisib, fimepinostat, LY-3023414, voxtalisib, dactulisib, parsaclisib, GSK-2636771, AZD-8186, ASN-003, and any combination thereof.
4. The method of any one of claims 1-3, wherein the PI3K inhibitor is a compound having formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003161060350000011
5. the method of any one of claims 1-4, wherein the oncolytic virus comprises a naturally occurring, modified or recombinant virus that is replication competent and capable of infecting and lysing tumor cells.
6. The method of any one of claims 1 to 5, wherein the oncolytic virus is selected from the group consisting of reovirus (reovirus), newcastle disease virus (Newcastle disease virus), vesicular stomatitis virus (vesicular stomatis virus), adenovirus (adenovirus), vaccinia virus (vaccinia virus), filtering virus (parapox orf virus), sindbis virus (Sindbis virus) and herpes simplex virus (herpes simplex virus).
7. The method of any one of claims 1-6, wherein the PD-1 inhibitor is selected from the group consisting of: anti-PD-1, anti-PD-L1, and anti-PD-L2 antibodies; preferably, wherein the PD-1 inhibitor is selected from the group consisting of Nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, terieprinimab, certolizumab, temeprizumab, tirlizumab, carprilizumab, and any combination thereof.
8. The method of any one of claims 1-7, wherein the PI3K inhibitor is administered in a dosage form of oral, buccal or parenteral route, e.g. the dosage form of the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form of the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal permeation, transmucosal administration.
9. The method of any one of claims 1-8, wherein the PI3K inhibitor is administered 1 or 2 times per day; or 1 administration of the PI3K inhibitor every 2, 3, 4,5, 6, 7, 8, 9, or 10 days or every 1,2, or 3 weeks; or 1 administration of the PI3K inhibitor daily 1,2, 3, 4,5, or 6 consecutive days per week followed by 6, 5, 4, 3, 2, or 1 day intervals.
10. The method of any one of claims 1-9, wherein the PI3K inhibitor is administered in a dosage range of about 20 mg/day to about 200 mg/day, about 30 mg/day to about 160 mg/day, about 60 mg/day to about 120 mg/day in an adult human.
11. The method of any one of claims 1-10, wherein the PI3K inhibitor is administered to the subject at an effective dose of about 0.5 to about 250mg/kg, about 1 to about 250mg/kg, about 2 to about 200mg/kg, about 3 to about 120mg/kg, about 5 to about 250mg/kg, about 10 to about 200mg/kg, or about 20 to about 120mg/kg.
12. The method according to any one of claims 1-11, wherein the oncolytic virus is administered in a dosage form of oral, buccal or parenteral route, e.g. the dosage form of the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form of the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration.
13. The method of any of claims 1-12, wherein the oncolytic virus is administered at least once daily, repeatedly on days 2, 3, 4,5, 6, 7, 8, 9, 10, 14, 21 or 28, or for any period of 2-28 days, or longer, continuously or intermittently daily for 1,2, 3, 4,5, 6, 7, 8, 9, 10, 12 or 24 hours, or for any period of 1-24 hours; preferably, the duration of administration is 5, 15, 30, 60, 90, 120, 150 or 180 minutes or any time from 5 to 180 minutes or longer.
14. The method according to any one of claims 1 to 13, wherein the oncolytic virus is administered at a dose in an adult human that varies according to individual differences (age, weight, sex), differences in administered virus, e.g. at about 10 per human 2 -10 17 PFU (colony Forming Unit) or TCID 50 (half the tissue culture infectious dose) administration.
15. The method of any one of claims 1-14, wherein the amount of PFU per kg is from about 1.0 to about 10 15 Administering an effective dose of PFU/kg to the subject the oncolytic virus.
16. The method of any one of claims 1-15, wherein the PD-1 inhibitor is administered by an parenteral route, e.g., intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration.
17. The method of any one of claims 1-16, wherein the PD-1 inhibitor is formulated as a solution, a lyophilizate, a powder injection.
18. The method of any one of claims 1-17, wherein the PD-1 inhibitor is administered at an effective dose of 0.05mg/kg, 0.1mg/kg, 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, 5mg/kg, 6mg/kg, 7mg/kg, or 8 mg/kg.
19. The method of any one of claims 1-18, wherein the tumor/cancer comprises a primary tumor/cancer, a recurrent tumor/cancer or a metastatic tumor/cancer, including solid tumors and hematologic tumors.
20. The method of any one of claims 1-19, wherein the tumor/cancer is selected from the group consisting of: head and neck squamous cell carcinoma, head and neck cancer, brain cancer, glioma, glioblastoma multiforme, neuroblastoma, central nervous system cancer, neuroendocrine tumor, laryngeal cancer, nasopharyngeal cancer, esophageal cancer, thyroid cancer, malignant pleural mesothelioma, lung cancer, breast cancer, liver cancer, hepatoma, hepatobiliary cancer, pancreatic cancer, gastric cancer, gastrointestinal cancer, intestinal cancer, colon cancer, colorectal cancer, kidney cancer, clear cell renal cell cancer, ovarian cancer, endometrial cancer, cervical cancer, bladder cancer, prostate cancer, testicular cancer, skin cancer, melanoma, leukemia, lymphoma, bone cancer, chondrosarcoma, myeloma, multiple myeloma, myelodysplastic syndrome, myeloproliferative tumors, squamous cell carcinoma, ewing's sarcoma, systemic light chain amyloidosis, and merkel cell carcinoma; more preferably, the lymphoma is selected from: hodgkin's lymphoma, non-hodgkin's lymphoma, diffuse large B-cell lymphoma, follicular lymphoma, primary mediastinal large B-cell lymphoma, mantle cell lymphoma, small lymphocytic lymphoma, large B-cell lymphoma enriched in T-cells/histiocytes, and lymphoplasmacytic lymphoma, said lung cancer being selected from the group consisting of: non-small cell lung cancer and small cell lung cancer, said leukemia being selected from: chronic myeloid leukemia, acute myeloid leukemia, lymphocytic leukemia, lymphoblastic leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia, and myeloid leukemia.
21. A pharmaceutical composition and/or a pharmaceutical combination comprising a PI3K inhibitor, an oncolytic virus and a PD-1 inhibitor, and a pharmaceutically acceptable carrier.
22. The pharmaceutical composition and/or pharmaceutical combination of claim 21, wherein the PI3K inhibitor is selected from PI3K α, PI3K β, PI3K γ, and PI3K δ subtype inhibitors.
23. The pharmaceutical composition and/or combination of drugs of claim 21 or 22, wherein the PI3K inhibitor is selected from the group consisting of Idelalisib, copanlisib, duvelisib, alpelisib, seletalisib, gedatolisib, rigosertib sodium, leniolisib, umbralisib, buparlisb (AN 2025), AMG-319, GM-604, acalisib, bimarilisib, GDC-0084, ACP-319, tenalisb, serabelisib, SF-1126, nemralisib, fimepinostat, LY-3023414, voxtalisib, dacalcolisib, parsalclisib, GSK-2636771, AZD-8186, ASN-003, and any combination thereof.
24. The pharmaceutical composition and/or pharmaceutical combination of any one of claims 21-23, wherein the PI3K inhibitor is a compound having formula (I) or a pharmaceutically acceptable salt thereof,
Figure FDA0003161060350000041
25. the pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21 to 24, wherein the oncolytic virus comprises a naturally occurring, modified and recombinant virus having replication capacity and capable of infecting and lysing tumor cells.
26. The pharmaceutical composition and/or pharmaceutical combination of any one of claims 21-25, wherein the oncolytic virus is selected from the group consisting of reovirus (reovirus), newcastle disease virus (newcastle disease virus), vesicular stomatitis virus (vesicular stomatis virus), adenovirus (adenovirus), vaccinia virus (vaccinia virus), filtering virus (parapox orf virus), sindbis virus (Sindbis virus) and herpes simplex virus (herpes simplex virus).
27. The pharmaceutical composition and/or pharmaceutical combination of any one of claims 21-26, wherein the PD-1 inhibitor is selected from the group consisting of: anti-PD-1, anti-PD-L1, and anti-PD-L2 antibodies; preferably, wherein the PD-1 inhibitor is selected from the group consisting of Nivolumab, pembrolizumab, atezolizumab, avelumab, durvalumab, tremelimumab, terieprinimab, certolizumab, temeprizumab, tirlizumab, carprilizumab, and any combination thereof.
28. The pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21 to 27, wherein the PI3K inhibitor, the oncolytic virus and the PD-1 inhibitor may be in the same and/or separate dosage form (dosage form).
29. The pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21 to 28, wherein the PI3K inhibitor may be formulated in a dosage form for oral, buccal or parenteral route, e.g. the dosage form for the oral route may be tablets, capsules, powders, pills, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form for the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal penetration, transmucosal administration, e.g. may be solutions, powder injections or lyophilisates.
30. The pharmaceutical composition and/or pharmaceutical combination according to any of claims 21-29, wherein the oncolytic virus is administered in a dosage form of oral, buccal or parenteral route, e.g. the dosage form of the oral route may be tablets, capsules, powders, pellets, granules, suspensions, solutions and solution preconcentrates, emulsions and emulsion preconcentrates, e.g. the dosage form of the parenteral route may be intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal penetration, transmucosal administration.
31. The pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21 to 30, wherein the PD-1 inhibitor is formulated for administration by an parenteral route, e.g. intravenous, intraperitoneal, intradermal, subcutaneous, intramuscular, intracranial, intrathecal, intratumoral, transdermal, transmucosal administration, e.g. as a solution, powder injection or lyophilisate.
32. The pharmaceutical composition and/or pharmaceutical combination according to any of claims 21 to 31, wherein a dose of 1 to 1000mg of PI3K inhibitor is contained per unit dosage form (unit dosage form), e.g. a dose of 1,2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, 150, 160, 170, 175, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 750, 800, 900, 1000mg or a value between any two of the above.
33. The pharmaceutical composition and/or pharmaceutical combination according to any of claims 21 to 32, comprising about 10 units per unit dosage form (unit dose form) 2 PFU to about 10 17 Oncolytic virus at PFU dose, e.g. 10 2 、10 3 、10 4 、10 5 、10 6 、10 7 、10 8 、10 9 、10 10 、10 11 、10 12 、10 13 、10 14 、10 15 、10 16 、10 17 PFU or a value between any two of the above values.
34. The pharmaceutical composition and/or pharmaceutical combination of any one of claims 21-33, wherein a dose of 1-5000mg of PD-1 inhibitor is contained per unit dosage form, e.g., a dose of 1,2, 3, 4,5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30, 35, 40, 50, 60, 70, 75, 80, 90, 100, 110, 120, 125, 130, 140, 150, 160, 170, 175, 180, 190, 200, 250, 300, 350, 400, 450, 500, 600, 700, 750, 800, 900, 1000, 1100, 1200, 1300, 1400, 1500, 1600, 1700, 1800, 2000, 2200, 2400, 2500, 2600, 2700, 2750, 2800, 3000, 3500, 4000, 4500, 2500 mg, or a value between any two of the foregoing.
35. The pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21 to 34 for use in the treatment of tumors/cancers and/or for generating a memory immune response against tumors/cancers.
36. Use of a pharmaceutical composition and/or a pharmaceutical combination according to any one of claims 21-34 for the manufacture of a medicament for the treatment of a tumor/cancer and/or for generating a memory immune response against a tumor/cancer.
37. A kit comprising the pharmaceutical composition and/or pharmaceutical combination according to any one of claims 21-35, together with instructions for use, wherein the PI3K inhibitor, the oncolytic virus and the PD-1 inhibitor may be in the same and/or separate containers.
CN202110791354.2A 2021-07-13 2021-07-13 Combination therapy for the treatment of cancer Pending CN115607678A (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
CN202110791354.2A CN115607678A (en) 2021-07-13 2021-07-13 Combination therapy for the treatment of cancer
CN202280013623.5A CN116887837A (en) 2021-07-13 2022-07-06 Combination therapy for the treatment of cancer
PCT/CN2022/104040 WO2023284597A1 (en) 2021-07-13 2022-07-06 Combined therapy for treating cancer
TW111125796A TW202317134A (en) 2021-07-13 2022-07-08 Combination therapy for treating cancer

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110791354.2A CN115607678A (en) 2021-07-13 2021-07-13 Combination therapy for the treatment of cancer

Publications (1)

Publication Number Publication Date
CN115607678A true CN115607678A (en) 2023-01-17

Family

ID=84856157

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202110791354.2A Pending CN115607678A (en) 2021-07-13 2021-07-13 Combination therapy for the treatment of cancer
CN202280013623.5A Pending CN116887837A (en) 2021-07-13 2022-07-06 Combination therapy for the treatment of cancer

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN202280013623.5A Pending CN116887837A (en) 2021-07-13 2022-07-06 Combination therapy for the treatment of cancer

Country Status (3)

Country Link
CN (2) CN115607678A (en)
TW (1) TW202317134A (en)
WO (1) WO2023284597A1 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024207425A1 (en) * 2023-04-07 2024-10-10 Virogin Biotech (Shanghai) Ltd. Combination of recombinant oncolytic virus and checkpoint inhibitor for the treatment of cancer

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
MA40035A (en) * 2014-10-14 2016-04-21 Dana Farber Cancer Inst Inc PD-L1 ANTIBODY MOLECULES AND THEIR USES
EP3328418A1 (en) * 2015-07-29 2018-06-06 Novartis AG Combination therapies comprising antibody molecules to pd-1
WO2017106656A1 (en) * 2015-12-17 2017-06-22 Novartis Ag Antibody molecules to pd-1 and uses thereof
CN114668847A (en) * 2018-07-25 2022-06-28 广州威溶特医药科技有限公司 Application of combination of anti-tumor drug and oncolytic virus in preparation of drug for treating tumor
BR112022011158A2 (en) * 2019-12-12 2022-08-30 Ignite Immunotherapy Inc VARIANT ONCOLYTIC VACCINIA VIRUS AND METHODS OF USE THEREOF

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2024207425A1 (en) * 2023-04-07 2024-10-10 Virogin Biotech (Shanghai) Ltd. Combination of recombinant oncolytic virus and checkpoint inhibitor for the treatment of cancer

Also Published As

Publication number Publication date
TW202317134A (en) 2023-05-01
CN116887837A (en) 2023-10-13
WO2023284597A1 (en) 2023-01-19

Similar Documents

Publication Publication Date Title
AU2012321110B2 (en) Combination treatment
DK2605764T3 (en) Compositions for the treatment of cancer
KR102392441B1 (en) Ferric citrate for reducing mortality and morbidity related to adverse cardiac events in chronic kidney disease patients
EA034512B1 (en) Treatment of cancer with tor kinase inhibitors
US20230181565A1 (en) Crenolanib combination therapy
US20240285640A1 (en) Combination therapy for treating pik3ca-mutated cancer
JP2023542093A (en) Use of thiauranib in combination with immune checkpoint inhibitors in antitumor therapy
JP6090836B2 (en) Anti-tumor activity enhancer of chemotherapeutic agent
SG187828A1 (en) Novel combination therapy for the treatment of cancer
EA020046B1 (en) Pharmaceutical combination
WO2019142490A1 (en) Urine alkali agent useful for treatment of cancer patient
JPWO2020080499A1 (en) New pharmaceutical composition
TWI316405B (en) A pharmaceutical composition for reducing pain in a mammal suffering from a neoplasm
WO2023284597A1 (en) Combined therapy for treating cancer
WO2006118212A1 (en) Agent for preventing and treating pancreatitis
US20090148538A1 (en) Use of adsorbent carbon microspheres to treat pouchitis
CN114728003A (en) Therapeutic combinations of acatinib and caspasertinib for the treatment of B-cell malignancies
IL268224B1 (en) Pharmaceutical composition for preventing and treating pancreatic cancer, containing gossypol and phenformin as active ingredients
CN105916517A (en) Pharmaceutical composition for treatment or palliation of elderly or end-stage cancer patient
JP7381115B2 (en) Compositions and their application in the preparation of medicines for cancer treatment
JP7357386B2 (en) Application of the compound or its pharmaceutically acceptable salt, dimer or trimer in the preparation of drugs for cancer treatment
TW202408524A (en) Use of btk inhibitors for the treatment of diseases
JP2023065622A (en) Treatment method for cancer patient with severe renal dysfunction
BR112021008606A2 (en) use of tivozanib to treat individuals with refractory cancer
RU2827402C2 (en) Pharmaceutical composition containing pyridine compound and use thereof in ros1-positive non-small cell lung cancer

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination