CN112972675A - Use of anti-PD-1 antibodies in the treatment of tumors - Google Patents

Use of anti-PD-1 antibodies in the treatment of tumors Download PDF

Info

Publication number
CN112972675A
CN112972675A CN202110287273.9A CN202110287273A CN112972675A CN 112972675 A CN112972675 A CN 112972675A CN 202110287273 A CN202110287273 A CN 202110287273A CN 112972675 A CN112972675 A CN 112972675A
Authority
CN
China
Prior art keywords
antibody
cancer
ser
cells
treatment
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110287273.9A
Other languages
Chinese (zh)
Inventor
姚盛
冯辉
武海
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.)
Shanghai Junshi Biosciences Co Ltd
Original Assignee
Shanghai Junshi Biosciences 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 Shanghai Junshi Biosciences Co Ltd filed Critical Shanghai Junshi Biosciences Co Ltd
Priority to CN202110287273.9A priority Critical patent/CN112972675A/en
Publication of CN112972675A publication Critical patent/CN112972675A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/80Vaccine for a specifically defined cancer
    • A61K2039/876Skin, melanoma

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Immunology (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
  • Peptides Or Proteins (AREA)

Abstract

The present invention relates to the use of anti-PD-1 antibodies in the treatment of tumors. The invention also relates to the use of a reagent for detecting the status of CD8+ T cells and/or NK cells in peripheral blood or a reagent for detecting the EMSY gene in a test kit for predicting the effect of a treatment of a tumor patient against an anti-PD-1 antibody and/or an antigen-binding fragment thereof.

Description

Use of anti-PD-1 antibodies in the treatment of tumors
The application is a divisional application of an invention patent application with application date of 2018, 9 and 7, application number of 201811043430.6 and the name of 'application of anti-PD-1 antibody in treating tumor'.
Technical Field
The present invention relates to the use of anti-PD-1 antibodies in the treatment of tumors. In particular, the invention relates to the use of anti-PD-1 antibodies in the treatment of solid tumors, in particular melanoma; and the use of an anti-PD-1 antibody for the manufacture of a medicament for the treatment of solid tumors, in particular melanoma; and methods of using the biomarkers to predict the efficacy of anti-PD-1 antibodies in the treatment of solid tumors; and the use of a combination therapy or composition comprising the anti-PD-1 antibody in the treatment of solid tumors.
Background
Immune escape is one of the characteristics of cancer. Ahmadzadeh, m. et al, Blood,114:1537-44, disclose that tumor-specific T lymphocytes are frequently present in the tumor microenvironment, draining lymph nodes and peripheral Blood, but are often unable to control tumor progression due to the network of immunosuppressive mechanisms present in the tumor microenvironment. CD8+Tumor infiltrating T lymphocytes (TILs) typically express activation-induced inhibitory receptors, including CTLA-4 and PD-1, while tumor cells often express immunosuppressive ligands, including PD-1 ligand 1(PD-L1, also called B7-H1 or CD274), which inhibit T cell activation and effector functions. Among the inhibitory mechanisms, PD-1 and its ligands have become an important pathway for tumor cells to utilize it to inhibit activated T cells in the tumor microenvironment.
Programmed death receptor 1(PD-1) plays an important role in immune regulation and maintenance of peripheral tolerance. PD-1 is expressed primarily in activated T and B cells and functions to inhibit lymphocyte activation, a normal peripheral tissue tolerance mechanism of the immune system that prevents immune overstimulation. However, the activated T cells infiltrated in the tumor microenvironment highly express PD-1 molecules, and inflammatory factors secreted by the activated leukocytes can induce the tumor cells to highly express ligands PD-L1 and PD-L2 of PD-1, so that the PD-1 pathway of the activated T cells in the tumor microenvironment is continuously activated, the functions of the T cells are inhibited, and the tumor cells cannot be killed. The therapeutic PD-1 antibody can block the pathway, partially restore the function of T cells, and enable the activated T cells to continuously kill tumor cells.
In the last decade, blocking of the PD-1/PD-L1 pathway has proven to be an effective way to induce a durable anti-tumor response in various cancer indications. Monoclonal antibodies (mAbs) blocking the PD/PD-L1 pathway can enhance the activation and effector functions of tumor-specific T cells, reduce tumor burden, and improve survival rates. Between 2014 and 2017, the FDA has approved 2 anti-PD 1 monoclonal antibody (nivolumab) and 3 anti-PD-L1 monoclonal antibody (atezolizumab, avelumab, and durvalumab) for the treatment of human tumors. Melanoma is the indication that Nivolumab and Pembrolizumab were first approved in 2014.
Melanoma has long been recognized as an immunogenic cancer because of the frequently observed infiltration of lymphocytes into tumors and clinical response to high doses of IL-2 immunotherapy. Mechanistically, chronic uv radiation exposure associated with induction of DNA damage is a major cause of melanoma in western populations. Chronic ultraviolet radiation sunburn (CSD) melanoma accounts for 95% of cutaneous melanoma in the united states and other western countries. In contrast, acral freckle-like melanoma (ALM) (-50%) and Mucosal Melanoma (MM) (-20%) are the two most common subpopulations of melanoma in asian populations, a finding disclosed by Chi, z, et al, BMC Cancer (2011),11: 85. Furney, S.J. et al, Pigment Cell Melanoma Res (2014),27:835-8 disclose that both ALM and MM are not associated with chronic UV exposure and carry fewer DNA mutations. Retrospective studies by Cho, J, et al, Invest New Drugs (2016),34:677-84, have shown that immunotherapy is less effective in treating ALM and MM compared to CSD melanoma. Therefore, the uncertainty of the efficacy of immunotherapy, such as anti-PD-1 antibody, in treating melanoma, especially mucosal and acro-melanoma, which are frequent in asian population, and how to further improve the therapeutic effect thereof, is a technical problem to be solved in the art.
Disclosure of Invention
In one aspect, the invention provides a method of treating a cancer patient comprising administering to the patient a therapeutically effective amount of an anti-PD-1 antibody and/or antigen-binding fragment thereof and optionally an additional anti-cancer agent other than an anti-PD-1 antibody and/or antigen-binding fragment thereof.
In a second aspect, the invention provides the use of an anti-PD-1 antibody and/or an antigen-binding fragment thereof and optionally an additional anti-cancer agent other than an anti-PD-1 antibody and/or an antigen-binding fragment thereof in the manufacture of a medicament for the treatment of cancer.
In one or more embodiments, the additional anti-cancer agents described herein are small molecule targeted anti-cancer agents. In one embodiment, the additional anti-cancer agent of the present invention is selected from the group consisting of CDK4/6 inhibitors, FGF/FGFR inhibitors, VEGFR inhibitors and PARP inhibitors.
In one or more embodiments, the medicament comprises an anti-PD-1 antibody and a CDK4/6 inhibitor. In one or more embodiments, the medicament comprises an anti-PD-1 antibody and an FGF/FGFR inhibitor. In one or more embodiments, the medicament comprises an anti-PD-1 antibody and a VEGFR inhibitor. In one or more embodiments, the medicament comprises an anti-PD-1 antibody and a PARP inhibitor.
In one or more embodiments, the present invention provides the use of an anti-PD-1 antibody for the preparation of a medicament for use in combination with a CDK4/6 inhibitor, a FGF/FGFR inhibitor, a VEGFR inhibitor or a PARP inhibitor for the treatment of a cancer patient; and provides CDK4/6 inhibitors; use of an FGF/FGFR inhibitor, VEGFR inhibitor or PARP inhibitor for the manufacture of a medicament for the treatment of a cancer patient in combination with an anti-PD-1 antibody.
In one or more embodiments, the present invention provides the use of an anti-PD-1 antibody in combination with a CDK4/6 inhibitor, a FGF/FGFR inhibitor, a VEGFR inhibitor or a PARP inhibitor for the preparation of a medicament for treating a cancer patient.
In one or more embodiments, the cancer of the invention is a solid tumor.
In one or more embodiments, the cancer patient has activated CD8 in peripheral blood+The percentage of T cells in total CD8+ T cells in peripheral blood is greater than or equal to 65%. In one or more embodiments, the percentage of NK cells in peripheral blood of the cancer patient in peripheral blood mononuclear cells is greater than or equal to 26%. In one or more embodiments, the cancer patient has activated CD8 in peripheral blood+The percentage of T cells in total CD8+ T cells in peripheral blood is greater than or equal to 65%, and the percentage of NK cells in peripheral blood mononuclear cells is greater than or equal to 26%. In one or more embodiments, the NK cell is CD3-CD16+CD54+NK cells.
In one or more embodiments, the EMSY gene mutation is detected in tumor tissue of the cancer patient.
In some preferred embodiments, the solid tumor includes, but is not limited to, cancer of the urinary tract, bladder, breast, renal cell, lymphoma, squamous cell carcinoma of the head/neck, lung, liver, bile duct, stomach, esophagus, intestine, pancreas, neuroendocrine, melanoma, ovary, endometrium, cervix or prostate.
In one or more preferred embodiments, the cancer is melanoma. In one or more preferred embodiments, the cancer is mucosal melanoma. In one or more preferred embodiments, the cancer is acromelanoma.
In one or more embodiments, the method comprises administering to the patient having mucosal melanoma or acro-melanoma a therapeutically effective amount of an anti-PD-1 antibody and/or antigen-binding fragment thereof.
In one or more embodiments, the use is of an anti-PD-1 antibody and/or an antigen-binding fragment thereof in the manufacture of a medicament for treating mucosal melanoma or acromelanoma.
In certain embodiments, the subject has been previously treated. In certain embodiments, the subject has undergone standard systemic treatment. In one or more preferred embodiments, the standard systemic treatment is chemotherapy. In certain embodiments, the subject has no history of autoimmune disease or persistent infection. In certain embodiments, the cancer is melanoma. In other embodiments, the melanoma is advanced, metastatic, and/or refractory melanoma. In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is administered alone, or in combination with a CDK4/6 inhibitor, FGF/FGFR inhibitor, VEGFR inhibitor, and/or PARP inhibitor, to induce a durable clinical response in the individual.
In certain embodiments, administration of an anti-PD-1 antibody or antigen-binding fragment thereof alone, or in combination with a CDK4/6 inhibitor, FGF/FGFR inhibitor, VEGFR inhibitor, and/or PARP inhibitor results in increased T cell infiltration in tumor tissue or in tumors. In some embodiments, the increased T cell infiltration is characterized by activated CD8+Increased infiltration of T cells in tumor tissue or tumors. In some embodiments, the anti-PD-1 antibody or antigen binding thereof is administered aloneThe administration of anti-PD-1 antibodies to synthetic fragments, or in combination with CDK4/6 inhibitors, FGF/FGFR inhibitors, VEGFR inhibitors and/or PARP inhibitors, results in increased T cell proliferation.
In one or more embodiments, in a treatment comprising administering the anti-PD-1 antibody alone, or in combination with a CDK4/6 inhibitor, FGF/FGFR inhibitor, VEGFR inhibitor, and/or PARP inhibitor, a therapeutically effective dose of the anti-PD-1 antibody ranges from about 0.1 to about 10.0mg/kg, or a 240mg fixed dose, intravenously infused about once every 2-6 weeks. In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is administered at a fixed dose of about 1mg/kg, 3mg/kg, 10mg/kg, or 240mg once every 2-4 weeks.
A third aspect of the invention provides a kit for treating an individual having cancer, the kit comprising: (a) a monoclonal antibody or antigen-binding fragment thereof that specifically binds to and inhibits PD-1; and optionally (b) one or more inhibitors targeting CDK4/6, FGF/FGFR, VEGFR, or PARP; (c) instructions for using the anti-PD-1 antibody alone or in combination with the anti-PD-1 antibody and optionally one or more inhibitors targeting CDK4/6, FGF/FGFR, VEGFR, or PARP to treat cancer in an individual. Preferably, the cancer is melanoma. As a further preferred mode, the cancer is acromelanoma; as another further preferred mode, the cancer is mucosal melanoma.
In the use, therapy, medicament and kit of the invention, the anti-PD-1 antibody is a monoclonal antibody or an antigen-binding fragment thereof. In certain embodiments, the anti-PD-1 antibody specifically binds to PD-1, blocks binding of PD-L1 or PD-L2 to PD-1. In certain embodiments, the anti-PD-1 antibody specifically binds to PD-L1 or/and PD-L2, blocks the binding of PD-L1 and/or PD-L2 to PD-1.
In one or more embodiments, the anti-PD-1 antibody is an antibody comprising Complementarity Determining Regions (CDRs) having a light chain complementarity determining region (LCDR) as set forth in SEQ ID NO: 1.2 and 3, the heavy chain complementarity determining region (HCDR) is as shown in SEQ ID NO: 4. 5 and 6.
In one or more embodiments, the anti-PD-1 antibody comprises a light chain variable region (VL) and a heavy chain variable region (VH), wherein VL is as set forth in SEQ ID NO: 7, VH is shown as SEQ ID NO: shown in fig. 8.
In one or more embodiments, the anti-PD-1 antibody is an anti-PD-1 antibody comprising a light chain comprising the amino acid sequence set forth in SEQ ID No. 9 and a heavy chain comprising the amino acid sequence set forth in SEQ ID NO: 10, or a pharmaceutically acceptable salt thereof.
In one or more preferred embodiments, the VEGFR inhibitor in the uses, therapies, medicaments and kits described herein is N-methyl-2- [3- ((E) -2-pyridin-2-yl-vinyl) -1H-indazol-6-ylsulfanyl ] -benzamide or a pharmaceutically acceptable salt, or prodrug thereof.
In the use, therapy, medicament and kit of the invention, the individual is a human.
The use, therapy, medicament and kit of the invention, PD-L1 or CD8 of a tissue or section of cancer+Expression of one or both of the T cells was tested positive. In one or more embodiments, the tissue or section of the cancer is tested positive for PD-L1 expression. In one or more embodiments, the CD8 of a tissue or section of cancer+T cell expression was tested positive. In one or more embodiments, the PD-L1 and CD8 of a tissue or section of cancer+The expression of T cells is tested positive.
In one or more embodiments, the subject of the invention is a human and the cancer is melanoma that is tested positive for human PD-L1 or positive for CD8 +.
In one or more preferred embodiments, the subject of the invention is a human and the cancer is acromelanoma that is tested positive for human PD-L1 and/or positive for CD8 +. In a preferred embodiment, the individual of the invention is a human and the cancer is a mucosal melanoma that is tested positive for human PD-L1 and/or positive for CD8 +.
In another embodiment, the use, therapy, medicament and kit of the invention, the subject has melanoma and has not been previously treated with immunotherapy. In a preferred embodiment, the cancer is acromelanoma. In a preferred embodiment, the cancer is mucosal melanoma.
In a fourth aspect, the present invention provides a method for predicting the effectiveness of an anti-PD-1 antibody cancer treatment comprising detecting a biomarker in the peripheral blood of a patient prior to treatment, wherein said biomarker is selected from, but not limited to, CD8+T cells or NK cells.
In one or more embodiments, activated CD8 in the peripheral blood of a patient+T cells Total CD8 in peripheral blood+The percentage in T cells was 65% or more, indicating that the patient is suitable for treatment of their cancer with an anti-PD-1 antibody. In one or more embodiments, the percentage of NK cells in peripheral blood of the patient that are in peripheral blood mononuclear cells is greater than or equal to 26%, indicating that the patient is eligible for treatment of their cancer with the anti-PD-1 antibody. In one or more embodiments, activated CD8 in the peripheral blood of a patient+T cells Total CD8 in peripheral blood+The percentage of T cells is greater than or equal to 65% and the percentage of NK cells in peripheral blood mononuclear cells is greater than or equal to 26%, indicating that the patient is suitable for treating their cancer with an anti-PD-1 antibody. In one or more embodiments, the NK cell is CD3-CD16+CD54+NK cells. In one or more embodiments, the cancer is melanoma.
In a fifth aspect, the present invention provides a method for predicting the effect of a treatment with an anti-PD-1 antibody in a tumour patient, comprising detecting a mutation in the EMSY gene in the tumour tissue of the patient, wherein the presence of said EMSY gene mutation indicates that said tumour patient is suitable for treatment with an anti-PD-1 antibody.
Drawings
FIG. 1: pharmacodynamic readings of toreplatab alone were used. The PD-1 Receptor Occupancy (RO) was determined by the percentage of total PD-1 in activated lymphocytes from peripheral blood (CD3+ CD45RA-) that bound TORIPALILAMAB, as analyzed by flow cytometry.
FIG. 2: antitumor activity of torepalimab alone. The investigators evaluated clinical response every 8 weeks with (RECIST) v 1.1. (A) The percentage of the sum of target lesion diameters over the baseline measurement for each subject during the treatment of the tropicalimab is shown in the spider graph. The response in most patients is persistent, as the median duration of the response is 5.6 months (ranging from 1.8 months to 17.7+ months). (B) Waterfall plots show the optimal percentage of tumor burden reduction from baseline. Previous treatment methods were labeled with the color of each subject. 7/8 received at least two systemic treatments.
FIG. 3: progression Free Survival (PFS) of all subjects after toriplalimab treatment.
FIG. 4: overall Survival (OS) of all subjects.
FIG. 5: CD 8T cells (CD 3) were shown in clinical responders and non-responders+CD8+CD45RA-) Frequency of activation over time.
FIG. 6: PD-L1 expression by IHC staining in tumor biopsies correlates with clinical efficacy. (A) High (> 50%) (n-7), medium (1-50%) (n-9) and low (< 1%) (n-12) PD-L1 expression subpopulations as determined by anti-PD-L1 (SP142) IHC staining in tumor cells were compared to clinical responses. (B) Dual IHC staining for PD-L1 and CD8 was performed in tumor biopsies.
FIG. 7: correlation of biomarkers or subgroups with clinical efficacy.
FIG. 8: correlation of hematological Tumor Mutational Burden (TMB) with objective response to torelpalimab treatment and overall survival. (A) Correlation of TMB with clinical response. Every Mb 6 mutations were taken as classification node values. (B) Correlation of TMB with overall survival.
FIG. 9: genomic sequencing of the enrolled subjects analyzed the mutation patterns.
FIG. 10: antitumor activity of toreplilimuab in combination with axitinib. (A) The percentage of the sum of target lesion diameters above baseline measurements for each subject during treatment with the tropimalimab in combination with the axitinib is shown in the spider graph. (B) Waterfall plots of the treatment course with toreplimab and axitinib show the best percentage reduction in tumor burden from baseline.
Detailed Description
The present invention relates to a method of treating tumors. The methods of the invention comprise administering to a patient in need thereof an anti-PD-1 antibody or antigen-binding fragment thereof alone; or comprising administering to a patient in need thereof an anti-PD-1 antibody in combination with other anti-cancer agents. The invention also relates to methods of using biomarkers to predict the efficacy of anti-PD-1 antibodies in the treatment of cancer, particularly melanoma patients.
Term(s) for
In order that the invention may be more readily understood, certain technical and scientific terms are defined below. Unless otherwise defined elsewhere herein, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
By "administering," "administering," and "treating" is meant introducing a composition comprising a therapeutic agent into a subject using any of a variety of methods or delivery systems known to those skilled in the art. Routes of administration of anti-PD-1 antibodies include intravenous, intramuscular, subcutaneous, peritoneal, spinal or other parenteral routes of administration, such as injection or infusion. "parenteral administration" refers to modes of administration other than enteral or topical administration, typically by injection, including, but not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraframe, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion, and via in vivo electroporation.
An "adverse effect" (AE) as referred to herein is any adverse and often unintentional or undesirable sign, symptom or disease associated with the use of medical treatment. For example, adverse reactions may be associated with activation of the immune system or expansion of immune system cells in response to therapy. The medical treatment may have one or more related AEs, and each AE may have the same or different severity level.
The term "subject" includes any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit, etc.), and most preferably a human. The terms "subject" and "patient" are used interchangeably herein.
"antibody" as used herein refers to any form of antibody that achieves the desired biological or binding activity. It is therefore used in its broadest sense, but is not limited to, monoclonal, polyclonal, multispecific, humanized full-length human, chimeric and camelid single domain antibodies. Which specifically binds to an antigen and comprises at least two heavy (H) and two light (L) chains interconnected by disulfide bonds, or an antigen-binding fragment thereof. Each heavy chain comprises a heavy chain variable region (VH) and a heavy chain constant region comprising three constant domains CH1, CH2 and CH 3. Each light chain comprises a light chain variable region (VL) and a light chain constant region comprising a constant domain CL. The VH and VL regions may be further subdivided into hypervariable regions, termed Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, termed Framework Regions (FRs). Generally, from N-terminus to C-terminus, both light and heavy chain variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR 4. Amino acids are typically assigned to each domain according to the following definitions: sequences of Proteins of Immunological Interest, Kabat et al; national Institutes of Health, Bethesda, Md.; 5 th edition; NIH publication No. 91-3242 (1991): kabat (1978) adv.prot.chem.32: 1-75; kabat et al, (1977) J.biol.chem.252: 6609-6616; chothia et al, (1987) J mol. biol.196: 901-883 or Chothia et al, (1989) Nature 341: 878-883.
The carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function. Generally, human light chains are classified into kappa chains and lambda chains. Human heavy chains are generally classified as mu, delta, gamma, alpha, or epsilon, and define the isotype of the antibody as IgM, IgD, IgG, IgA, and IgE, respectively. The IgG subclasses are well known to those skilled in the art and include, but are not limited to, IgG1, IgG2, IgG, and IgG 4.
The term "antibody" includes: naturally occurring and non-naturally occurring abs; monoclonal and polyclonal Ab; chimeric and humanized abs; human or non-human Ab; ab is fully synthesized; and a single chain Ab. Non-human abs may be humanized by recombinant methods to reduce their immunogenicity in humans.
Unless specifically indicated otherwise, "antibody fragment" or "antigen-binding fragment" as used herein refers to an antigen-binding fragment of an antibody, i.e., an antibody fragment that retains the ability of a full-length antibody to specifically bind to an antigen, e.g., a fragment that retains one or more CDR regions. Examples of antibody binding fragments include, but are not limited to, Fab ', F (ab') 2, and Fv fragments; a diabody; a linear antibody; a single chain antibody molecule; nanobodies and multispecific antibodies formed from antibody fragments.
"chimeric antibody" refers to antibodies and fragments thereof as follows: wherein a portion of the heavy and/or light chain is identical to or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, and the remainder of the chain is identical to or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, so long as it exhibits the desired biological activity.
"human antibody" refers to an antibody comprising only human immunoglobulin sequences. A human antibody may contain murine carbohydrate chains if it is produced in a mouse, mouse cells, or a hybridoma derived from a mouse cell. Similarly, "mouse antibody" or "rat antibody" refers to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
"humanized antibody" refers to antibody forms containing sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain a minimal sequence derived from a single side of a non-human immunoglobulin. Typically, the humanized antibody will comprise substantially all of at least one and typically two variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin. The humanized antibody optionally further comprises at least a portion of an immunoglobulin constant region (Fc), typically a human immunoglobulin constant region.
The term "cancer" refers to a wide variety of diseases characterized by the uncontrolled growth of abnormal cells in the body. Unregulated cell division, growth division and growth lead to the formation of malignant tumors that invade adjacent tissues and may also metastasize to distal parts of the body through the lymphatic system or blood stream. Examples of cancer include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma. More specific examples of cancer include squamous cell carcinoma, myeloma, small-cell lung carcinoma, non-small cell lung carcinoma, glioma, hodgkin's lymphoma, non-hodgkin's lymphoma, acute myelogenous leukemia, multiple myeloma, gastrointestinal (tract) cancer, kidney cancer, ovarian cancer, liver cancer, lymphoblastic leukemia, lymphocytic leukemia, colorectal cancer, endometrial cancer, kidney cancer, prostate cancer, thyroid cancer, melanoma, chondrosarcoma, neuroblastoma, pancreatic cancer, glioblastoma multiforme, cervical cancer, brain cancer, gastric cancer, bladder cancer, hepatoma, breast cancer, colon cancer and head and neck cancer. Another embodiment of the cancer comprises melanoma. Another particular embodiment of cancer includes metastatic mucosal melanoma. Cancers of the invention include those characterized by increased expression of one or both of PD-L1 and PD-L2 in the tested tissue sample.
The term "melanoma" refers to a malignant tumor derived from melanocytes, which is usually found in the skin, mucous membrane, and choroid of the eye. Melanoma is the most malignant tumor species among skin tumors and is prone to distant metastasis. Melanoma is divided into four subtypes, including limb-end type (acral), mucosal type (mucosal), long-term sun-exposed skin type (CSD), and non-long-term positive exposure-impaired type (non-CSD), which are collectively referred to as the daylight type. McLaughlin et al, Cancer, 2005, Mar 1,103 (5): 1000- > 1007; chi z, et al, BMC Cancer, 2011; 11:85 discloses that acro-and mucosal-type melanoma is the most prominent subtype of Asian race, and its carcinogenesis is not caused by DNA mutation by ultraviolet radiation. Acro and mucosal melanomas contain only a few DNA mutations compared to the sunburn melanomas. In the united states, 95% of melanoma is of the daylight type, while in asia, especially china, more than 70% of melanoma is of the acro and mucosal type, with mucosal types reaching more than 50%. Invest New Drugs,34: 677-; cho, J, et al disclose that immunotherapy is less effective in treating limb-tip and mucosal types compared to sunburn melanoma.
The term "immunotherapy" refers to the treatment of a subject suffering from a disease or at risk of infection or relapse from a disease by a method that includes inducing, enhancing, suppressing or otherwise modifying an immune response. By "treatment" or "therapy" of a subject is meant any type of intervention or process performed on the subject, or the administration of an agent to the subject, with the purpose of reversing, alleviating, ameliorating, slowing or preventing the onset, progression, severity, or recurrence of a symptom, complication, or condition, or biochemical indicators associated with the disease.
"programmed death receptor-1 (PD-1)" refers to an immunosuppressive receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo and binds to both ligands PD-L1 and PD-L2. The term "PD-1" as used herein includes variants, isoforms, and species homologs of human PD-1(hPD-1), hPD-1, and analogs having at least one common epitope with hPD-1.
A "therapeutically effective amount" or "therapeutically effective dose" of a drug or therapeutic agent is any amount of drug that, when used alone or in combination with another therapeutic agent, protects a subject from the onset of a disease or promotes disease regression as evidenced by a reduction in the severity of disease symptoms, an increase in the frequency and duration of asymptomatic phases of the disease, or the prevention of injury or disability resulting from the affliction of the disease. The ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to those skilled in the art, such as in human subjects during clinical trials, in animal model systems that predict human efficacy, or by assaying the activity of the agent in an in vitro assay.
A therapeutically effective amount of a drug includes a "prophylactically effective amount," i.e., any amount of a drug that inhibits the development or recurrence of cancer when administered to a subject at risk for developing cancer or a subject having a recurrence of cancer, alone or in combination with an anti-neoplastic agent.
"biotherapeutic agent" refers to a biological molecule, such as an antibody or fusion protein, that blocks ligand/receptor signaling in any biological pathway that supports tumor maintenance and/or growth or inhibits an anti-tumor immune response.
As used herein, unless expressly indicated otherwise, "CDR" means that the immunoglobulin variable region is a complementarity determining region defined using the Kabat numbering system.
By "anti-cancer agent" is meant any therapeutic agent useful in the treatment of cancer. Classes of anti-cancer agents include, but are not limited to: alkylating agents, antimetabolites, kinase inhibitors, spindle poison plant alkaloids, cytotoxic/antitumor antibiotics, photosensitizers, antiestrogens and selective estrogen receptor modulators, antiprogestins, aromatase inhibitors, CDK4/6 inhibitors, FGF/FGFR inhibitors, EGFR inhibitors, VEGFR inhibitors, and antisense oligonucleotides that inhibit the expression of genes involved in abnormal cell proliferation or tumor growth.
The term "about" refers to a value or composition within an acceptable error range for the particular value or composition, as determined by one of ordinary skill in the art, depending in part on how the value or composition is measured or determined, i.e., the limitations of the measurement system. For example, "about" can mean within 1 or greater than 1 standard deviation according to practice in the art. Alternatively, "about" may refer to a range of up to 10% or 20% (i.e., ± 10% or ± 20%). For example, about 3mg/kg may include any number between 2.7mg/kg and 3.3mg/kg (relative to 10%), and between 2.4mg/kg and 3.6mg/kg (relative to 20%). Where a particular value or composition is provided herein, unless otherwise expressly stated, the meaning of "about" shall be assumed to be within an acceptable error range for that particular value or composition.
"therapeutic anti-PD-1 monoclonal antibody" refers to an antibody that specifically binds to the mature form of a particular PD-1 expressed on the surface of certain mammalian cells. Mature PD-1 lacks a pre-secretory leader sequence, or leader peptide. The terms "PD-1" and "mature PD-1" are used interchangeably herein and, unless otherwise specifically defined or clear from context, should be understood to be the same molecule.
As described herein, a therapeutic anti-human PD-1 antibody or anti-hPD-1 antibody refers to a monoclonal antibody that specifically binds to mature human PD-1.
As used herein, "framework region" or "FR" refers to immunoglobulin variable regions that do not include CDR regions.
An "isolated antibody or antigen-binding fragment thereof" refers to a molecule that is in a purified state and in this case is designated as being substantially free of other biological molecules, such as nucleic acids, proteins, lipids, carbohydrates, or other materials (such as cell debris or growth media).
By "patient," "patient," or "subject" is meant any single subject in need of medical treatment or participating in clinical trials, epidemiological studies, or as a control, including humans and mammals, such as horses, cattle, dogs, or cats.
In the following paragraphs, various aspects of the present invention are described in further detail.
anti-PD-1 antibodies
"PD-1 antibody" refers to any chemical compound or biomolecule that binds to the PD-1 receptor, blocks the binding of PD-L1 expressed on cancer cells to PD-1 expressed on immune cells (T, B, NK cells) and preferably also blocks the binding of PD-L2 expressed on cancer cells to PD-1 expressed on immune cells. Alternative nouns or synonyms for PD-1 and its ligands include: for PD-1, PDCD1, PD1, CD279, and SLEB 2; for PD-L1, there are PDCD1L1, PDL1, B7-H1, B7H1, B7-4, CD274 and B7-H; and for PD-L2 there are PDCD1L2, PDL2, B7-DC and CD 273. In any of the inventive methods of treatment, medicaments and uses for treating a human subject, the PD-1 antibody blocks the binding of human PD-L1 to human PD-1, and preferably blocks the binding of both human PD-L1 and PD-L2 to human PD 1. The human PD-1 amino acid sequence can be found at NCBI locus number: NP _ 005009. Human PD-L1 and PD-L2 amino acid sequences can be found at NCBI locus numbers: NP-054862 and NP-079515.
Herein, when referring to an "anti-PD-1 antibody," unless otherwise indicated or described, the term includes antigen-binding fragments thereof.
anti-PD-1 antibodies that can be used in any of the uses, therapies, medicaments and kits described herein include monoclonal antibodies (mabs) or antigen-binding fragments thereof that specifically bind to PD-1, and preferably specifically bind to human PD-1. The mAb may be a human, humanized or chimeric antibody and may include human constant regions. In some embodiments, the constant region is selected from the group consisting of human IgG1, IgG2, IgG3, and IgG4 constant regions, and in preferred embodiments, the constant region is a human IgG4 constant region.
In any one of the embodiments of the use, therapy, medicament and kit of the invention, the PD-1 antibody is a monoclonal antibody or antigen-binding fragment thereof comprising: (a) the light chain CDR is SEQ ID NO: 1.2 and 3, and the heavy chain CDRs are SEQ ID NO: 4. 5 and 6.
In any one of the embodiments of the use, therapy, medicament and kit of the invention, the PD-1 antibody specifically binds to human PD-1 and comprises: (a) comprises the amino acid sequence of SEQ ID NO: 7, and (b) a light chain variable region comprising SEQ ID NO: 8 in the heavy chain variable region of the antibody.
In any one of the embodiments of the use, therapy, medicament and kit of the invention, the PD-1 antibody specifically binds to human PD-1 and comprises: (a) comprises the amino acid sequence of SEQ ID NO:9, and (b) a light chain comprising SEQ ID NO: 10.
In any one embodiment of the uses, therapies, medicaments and kits described herein, the PD-1 antibody is a monoclonal antibody or antigen-binding fragment thereof, and table a below provides a list of amino acid sequences of exemplary anti-PD-1 antibody mabs for use in the uses, therapies, medicaments and kits described herein:
table a: light and heavy chain CDRs of exemplary anti-human PD-1 antibodies
Figure BDA0002980998550000111
Examples of anti-PD-1 antibodies that bind to human PD-1 and that can be used in the uses, therapies, medicaments and kits described in the present invention are described in WO 2014206107. Human PD-1 mabs that may be used as anti-PD-1 antibodies in the uses, therapies, medicaments and kits described in this invention include any of the anti-PD-1 antibodies described in WO2014206107, including: teraprimab (Torpialimab) (a humanized IgG4 mAb) having the structure described in WHO Drug Information (Vol.32, No. 2, p.372-373 (2018)) and comprising the light and heavy chain amino acid sequences shown in sequences SEQ ID NOS: 9 and 10. In certain embodiments, anti-PD-1 antibodies useful for the uses, therapies, medicaments and kits described herein further include Nivolumab and Pembrolizumab that have been approved by the FDA.
In certain embodiments, anti-PD-1 antibodies useful in the uses, therapies, medicaments and kits described herein also include anti-PD-L1 monoclonal antibodies, such as atezolizumab, avelumab and durvalumab, that specifically bind PD-L1 to block the binding of PD-L1 to PD-1.
"PD-L1" expression or "PD-L2" expression as described herein refers to any detectable expression level of a particular PD-L protein on the surface of a cell or a particular PD-L mRNA within a cell or tissue. PD-L protein expression can be detected in IHC analysis of tumor tissue sections or by flow cytometry using diagnostic PD-L antibodies. Alternatively, PD-L protein expression of tumor cells can be detected by PET imaging using a binding agent that specifically binds to a desired PD-L target (such as PD-L1 or PD-L2).
Methods for quantifying PD-L1 protein expression in IHC analysis of tumor tissue sections, see but not limited to Thompson, r.h. et al, PNAS 101 (49): 17174 and 17179 (2004); taube, j.m. et al, Sci trans Med 4, 127ra37 (2012); and Toplian, S.L. et al, New Eng.J.Med.366(26):2443-2454(2012), etc.
One method employs a simple binary endpoint of positive or negative PD-L1 expression, where positive results are defined as the percentage of tumor cells showing histological evidence of cell surface membrane staining. Counting tumor tissue sections to at least 1% of total tumor cells is defined as positive for PD-L1 expression.
In another method, PD-L1 expression in tumor tissue sections was quantified in tumor cells as well as in infiltrating immune cells. The percentages of tumor cells and infiltrating immune cells that exhibit membrane staining were quantified individually as < 1%, 1% to 50%, and then 50% up to 100%. For tumor cells, PD-L1 expression was counted as negative if the score was < 1%, and positive if the score was > 1%.
In some embodiments, the expression level of PD-L1 by malignant cells and/or by infiltrating immune cells within the tumor is determined to be "overexpressed" or "elevated" based on comparison to the expression level of PD-L1 by an appropriate control. For example, the protein or mRNA expression level of control PD-L1 can be a level quantified in non-malignant cells of the same type or in sections from matched normal tissue.
"RECIST 1.1 therapeutic criteria" as described herein refers to the definition by Eisenhauver et al, e.a. et al, eur.j Cancer45: 228-.
The term "ECOG" score is an indicator of a patient's general health and ability to tolerate treatment, as measured by their physical strength. ECOG physical performance scoring criteria score: 0 minute, 1 minute, 2 minutes, 3 minutes, 4 minutes and 5 minutes. A score of 0 means that the motility was completely normal and had no difference from the motility before onset. A score of 1 means that the person is free to walk and engage in light physical activities, including general housework or office work, but not heavy physical activities.
By "sustained response" is meant a sustained therapeutic effect following cessation of treatment with a therapeutic agent or combination therapy as described herein. In some embodiments, the sustained response has a duration that is at least the same as the duration of treatment or at least 1.5,2.0,2.5, or 3 times the duration of treatment.
"tissue section" refers to a single portion or piece of a tissue sample, such as a thin slice of tissue cut from a sample of normal tissue or a tumor.
As used herein, "treating" cancer refers to administering a treatment regimen described herein (e.g., administration of a combination therapy of an anti-PD-1 antibody and a VEGFR inhibitor) to a subject having or diagnosed with cancer to achieve at least one positive therapeutic effect (e.g., a decrease in the number of cancer cells, a decrease in tumor volume, a decrease in the rate of cancer cell infiltration into peripheral organs, or a decrease in the rate of tumor metastasis or tumor growth). Positive treatment effects in cancer can be measured in a variety of ways (see w.a. weber, j.nucl.med.,50:1S-10S (2009)). For example, for tumor growth inhibition, T/C ≦ 42% is the minimum level of anti-tumor activity according to the NCI standard. T/C (%) is considered median treated/median control tumor volume x 100. In some embodiments, the therapeutic effect achieved by the combination of the invention is any of PR, CR, OR, PFS, DFS and OS. PFS (also called "time to tumor progression") refers to the length of time during and after treatment during which cancer does not grow and includes the amount of time a patient experiences CR or PR and the amount of time a patient experiences SD. DFS refers to the length of time during and after treatment that a patient is still disease free. OS refers to an extension of life expectancy compared to an initial or untreated individual or patient. In some embodiments, the response to a combination of the invention is any of PR, CR, PFS, DFS, OR OS, assessed using RECIST 1.1 efficacy criteria. The treatment regimen for a combination of the invention effective in treating a cancer patient can vary depending on a variety of factors such as the disease state, age, weight of the patient and the ability of the therapy to elicit an anti-cancer response in the subject. Although embodiments of the invention may not achieve an effective positive therapeutic effect in each subject, a positive therapeutic effect should be effective and achieved in a statistically significant number of subjects.
The terms "mode of administration", "dosing regimen", which are used interchangeably, refer to the dosage and time of use of each therapeutic agent in the combination of the invention.
"tumor" when applied to a subject diagnosed with or suspected of having cancer refers to a malignant or potentially malignant neoplasm or tissue mass of any size and includes primary tumors and secondary neoplasms. Solid tumors typically do not contain abnormal growth or clumps of tissue from cysts or fluid areas. Different types of solid tumors are named for the cell types that form them. Examples of solid tumors are sarcomas, carcinomas and lymphomas. Hematological cancers do not typically form solid tumors.
"tumor burden" refers to the total amount of tumor mass distributed throughout the body. Tumor burden refers to the total number of cancer cells or the total size of the tumor throughout the body. Tumor burden can be determined by a variety of methods known in the art, such as measuring its size using calipers after the tumor is removed from the subject, or while in vivo using imaging techniques such as ultrasound, bone scans, Computed Tomography (CT), or Magnetic Resonance Imaging (MRI) scans.
The term "tumor size" refers to the total size of a tumor, which can be measured as the length and width of the tumor. Tumor size can be determined by a variety of methods known in the art, for example, measuring its dimensions using calipers after the tumor is removed from the subject, or while in vivo using imaging techniques such as bone scans, ultrasound, CT, or MRI scans.
The term "Tumor Mutation Burden (TMB)" refers to the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases. In some embodiments of the invention, Tumor Mutational Burden (TMB) is estimated by analysis of somatic mutations, including coding base substitutions and the megabase insertions of the panel sequences studied. In some embodiments of the invention, a subject having a Tumor Mutation Burden (TMB) of 6 mutations/Mb or greater is predictive of a better therapeutic effect of TMB <6 mutations/Mb when administered to such a subject alone or in combination with other anti-cancer agents. In some preferred embodiments, the cancer is a solid tumor. In some embodiments, the cancer is melanoma. In a preferred embodiment, the cancer is a melanoma of the limb or/and mucosa type.
The term "gene amplification" refers to a process in which the copy number of a gene encoded by a specific protein is selectively increased while other genes are not increased proportionally. Under natural conditions, gene amplification is achieved by excision of repeated sequences of the gene from the chromosome and extrachromosomal replication in a plasmid or by transcribing the entire repeated sequences of ribosomal RNA to give RNA transcripts to give additional copies of the original DNA molecule. In the present invention, gene sequencing analysis is disclosed in some embodiments. In some embodiments of the invention, the subject of the invention has some unique gene amplification. In some preferred embodiments, the subject has CDK4/6 gene amplification; in some preferred embodiments, the subject has FGF3/4/6/19 gene amplification; in some preferred embodiments, the subject has EMSY gene amplification; in some preferred embodiments, the subject has a mutation inactivating BRCA2 gene; in some preferred embodiments, the subject has PARP gene amplification; in some preferred embodiments, the subject has EGF/EGFR gene amplification; in some preferred embodiments, the subject has a TERT or TP53 gene mutation. As a preferred embodiment, said acromelanoma subject has CDK4/6 gene amplification. As a preferred embodiment, said acromelanoma subject has FGF3/4/19 gene amplification. As a preferred embodiment, the limb melanoma subject has EMSY gene amplification. In some embodiments, the tumor patient has an EMSY gene amplification that is predictive of a better therapeutic effect with the anti-PD-1 antibody of the invention.
The term "CDK", a cyclin-dependent kinase, is a group of serine/threonine protein kinases, the phosphorylation of serine/threonine proteins by CDKs by synergies with cyclin drives the cell cycle, an important factor in cell cycle regulation. The CDK family is 8 CDK species, CDK 1-8, each of which binds to a different class of cyclins to form complexes that regulate the progression of cells from G1 phase to S phase or G2 phase to M phase and out of M phase. CDK4/6 inhibitors approved by the FDA to be on the market mainly comprise Ribociclib and palbociclib, and dozens of CDK4/6 inhibitors in clinical research stage.
The term "FGF" is a family of Fibroblast growth factor (Fibroblast growth factor) proteins, the members of which are 23 in total, FGF 1-23. FGFs can be classified into three classes according to their different mechanisms of action: endocrine (FGF15/19/21/23), paracrine (FGF1-10, FGF16-18, FGF20, FGF22), and cell-secreted FGF 11/12/13/14. The process of regulating biological activity by paracrine FGFs is to use HS as a cofactor, specifically binding to FGF receptors (FGFRs) on the cell surface. Whereas FGFRs mainly include 4 types: FGFR1, FGFR2, FGFR3 and FGFR 4. At present, FGF/FGFR inhibitors are not on the market, VEGFR/PDGFR/FGFR inhibitors nintedanib of Boringer Greenham is used for treating liver cancer, non-small cell lung cancer and idiopathic fibrosis, obtains breakthrough drug qualification of FDA in 2014, and Delitinib is in clinical stage at present in China.
The term "PARP (poly ADP-ribose polymerase)" is a DNA repair enzyme. Plays a key role in the DNA repair pathway. When DNA damage is broken, PARP is activated, is used as a molecular receptor of DNA damage, has the functions of recognizing and combining to the broken position of DNA, further activates and catalyzes poly ADP ribosylation of receptor protein, and participates in the process of repairing DNA. PARP inhibitors are a cancer therapy that targets Poly ADP Ribose Polymerase (PARP). PARP inhibitors currently on the market include Olaparib of aspirin for the treatment of ovarian cancer or primary peritoneal cancer; rubraca from Clovis for the treatment of ovarian cancer; and zejua of saxidong for the treatment of ovarian and peritoneal cancer. There are several dozen other PARP inhibitors in clinical research.
The term "EMSY" is a BRCA2 inhibiting protein, the BRCA2 gene is an anti-cancer gene, and has important functions in regulating human cell replication, genetic material DNA damage repair and cell normal growth, EMSY gene expansion and protein overexpression inhibit BRCA2 function. The risk of cancer in people with BRCA2 gene mutation or function loss is greatly increased. In some embodiments, detection of EMSY may be used as an index to predict the prospects of an individual to treat cancer using an anti-PD-1 antibody alone or/and in combination with other anti-cancer agents.
VEGF (vascular endothelial growth factor) in the term "VEGF/VEGFR" is a highly specific pro-vascular endothelial cell growth factor having the effects of promoting increased vascular permeability, degeneration of extracellular matrix, migration, proliferation and vascularization of vascular endothelial cells. After VEGF is combined with an extracellular binding domain of a tyrosine kinase receptor VEGFR on a vascular endothelial cell, every two monomer receptor molecules form a dimer on a membrane, and the tyrosine residue at the tail part of the binding domain in the receptor cell is phosphorylated, so that different signal conduction is activated, and a series of biological effects are exerted. "VEGFR inhibitors" refers to small molecule inhibitors of Vascular Endothelial Growth Factor (VEGF) receptors, or monoclonal antibodies directed against Vascular Endothelial Growth Factor (VEGF). In one embodiment, a "VEGFR inhibitor" refers to a small molecule inhibitor of the Vascular Endothelial Growth Factor (VEGF) receptor. In the uses, therapies, medicaments and kits described herein, specific inhibitors useful as VEGFR inhibitors include axitinib, sunitinib, sorafenib, apatinib, revafenib and pazopanib. In one embodiment, in the uses, therapies, medicaments and kits of the present invention, specific inhibitors useful as VEGFR inhibitors include axitinib, sunitinib and apatinib.
In a particular embodiment, in the uses, therapies, medicaments and kits of the invention, the VEGFR inhibitor is a compound having the structure: n-methyl-2- [3- ((E) -2-pyridin-2-yl-vinyl) -1H-indazol-6-ylsulfanyl ] -benzamide:
Figure BDA0002980998550000151
it is called axitinib.
Axitinib is a selective inhibitor of Vascular Endothelial Growth Factor (VEGF) receptors 1, 2, 3. These receptors are involved in pathological angiogenesis, tumor growth, and cancer development. Axitinib inhibits VEGF-mediated endothelial cell proliferation and survival in vitro and in mouse models. In mouse models of tumor xenografts, axitinib was shown to have inhibitory effects on tumor growth and phosphorylation of VEGFR-2. Are currently marketed in countries and regions such as the United states, Europe, Japan, China, etc., and are clinically used mainly for adult patients who have previously failed a tyrosine kinase inhibitor or cytokine therapy for advanced Renal Cell Carcinoma (RCC).
Axitinib and pharmaceutically acceptable salts thereof are described in chinese patents CN1137884 and CN1234693, the patents listed above are incorporated herein by reference.
Small molecule targeted inhibitors, including axitinib, include salts and prodrugs thereof, unless expressly specified otherwise herein. Small molecule targeted inhibitors, including axitinib, are generally basic in nature and are capable of forming a variety of salts with various inorganic and organic acids. As used herein, "salt" refers to an acidic salt formed with an inorganic acid and/or an organic acid. Pharmaceutically acceptable salts of small molecule targeted inhibitors including axitinib may be formed by reacting a small molecule targeted inhibitor including axitinib with an amount of an acid in a medium, followed by drying.
As a specific embodiment, the acid addition salt of axitinib includes acetate, benzoate, benzenesulfonate, sulfate, borate, butyrate, citrate, camphorate, camphorsulfonate, fumarate, hydrochloride, hydrobromide, hydroiodide, lactate, maleate, methanesulfonate, nitrate, oxalate, phosphate, salicylate, succinate, sulfate, tartrate, tosylate, and the like.
For the purposes of the present invention, all of the described acid salts are pharmaceutically acceptable salts of small molecule targeted inhibitors useful in the present invention, including axitinib, and all are considered equivalent to the free form of the corresponding compound.
The term "prodrug" refers to a compound that undergoes a chemical transformation, either by metabolic or chemical processes, after administration to a subject to produce a prodrug of a small molecule targeted inhibitor, including axitinib, or a salt thereof.
The term "PD-1 receptor occupancy" or "PD-1 receptor occupancy" is equivalent in the present invention and refers to the percentage of total PD-1 antibody binding occupied by PD-1 receptor in binding to human peripheral blood lymphocytes described herein. In some embodiments of the invention, the receptor that binds to the anti-PD-1 antibody of the invention is detected by a subtype-specific antibody that recognizes the IgG4 Fc portion of the antibody. In some embodiments of the invention, cells are incubated with a saturating concentration of anti-PD-1 antibody as described herein, and then stained with PE-labeled anti-human IgG4 Fc to assess the total amount of PD-1 in human whole blood lymphocytes. PD-1 positive target cells are predominantly present in the CD45RA-T cell subset.
The term "Immunohistochemistry (IHC)" refers to the use of an antigen with an antigenThe principle of antibody specific binding is that the color developing agent (fluorescein, enzyme, metal ion and isotope) for marking antibody is developed by chemical reaction to determine the tissue cell antigen (polypeptide and protein), and the method is used for the research of positioning, qualitative and relative quantitative. In some embodiments of the invention, a tumor tissue sample from the subject is subjected to PD-L1 or/and CD8 prior to treatment with the anti-PD-1 antibody+T cell assay using Roche anti-human PD-L1 antibody SP142(Cat No: M4422), or CD8 antibody (clone 4B11, Cat # MCA 1817T) for staining experiments. In some embodiments, a tumor cell that has membrane staining intensity ≧ 1% is defined as positive for PD-L1.
Pharmaceutical compositions and dosages
The therapeutic agents of the present invention may constitute pharmaceutical compositions, such as pharmaceutical compositions containing the anti-PD-1 antibodies described herein or/and other anti-cancer agents other than the anti-PD-1 antibodies, and other pharmaceutically acceptable carriers. As used herein, "pharmaceutically acceptable carrier" includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, which are physiologically compatible. Preferably, the carriers suitable for use in the composition comprising the anti-PD-1 antibody are suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration, such as by injection or infusion, while the carriers for the composition comprising the other anti-cancer agent are suitable for parenteral administration, such as oral administration. The pharmaceutical compositions of the present invention may contain one or more pharmaceutically acceptable salts, antioxidants, water, non-aqueous carriers, and/or adjuvants such as preserving, wetting, emulsifying, and dispersing agents.
The dosage regimen is adjusted to provide the optimum desired response, such as the maximum therapeutic response and/or the minimum adverse effect. For anti-PD-1 antibodies, including administration in combination with another anti-cancer agent, the dose range may be from about 0.01 to about 20mg/kg, from about 0.1 to about 10mg/kg of the individual's body weight, or a fixed dose of 120mg, 240mg, 360mg, 480 mg. For example, the dosage may be about 0.1, about 0.3, about 1, about 2, about 3, about 5, or about 10mg/kg of the individual's body weight. Dosing regimens are generally designed to achieve such exposure, which results in sustained Receptor Occupancy (RO) based on the typical pharmacokinetic properties of abs. A representative dosing regimen may be about once per week, about once every two weeks, about once every three weeks, about once every four weeks, about once a month, or longer. In some embodiments, the anti-PD-1 antibody is administered to the individual about once every two weeks.
The dosing schedule of other anti-cancer agents varies for different drugs.
In some embodiments of the invention, the schedule of administration of the VEGFR inhibitor varies for different subtypes. For example, a typical dose of sunitinib includes a fixed dose of 50mg administered once daily to an adult with or without food, with dose adjustments made in about 12.5mg increments or increments recommended based on individual patient safety and tolerability. Whereas the starting dose of axitinib was 5mg, administered orally 2 times a day, with the dose adjusted based on individual safety and tolerability. For combination therapy of an anti-PD-1 antibody with a VEGFR inhibitor, in some embodiments, the VEGFR inhibitor is administered at its approved or recommended dose, and treatment is continued until clinical effect is observed or until unacceptable toxicity or disease progression occurs.
Method of the invention
The present invention relates to methods of treating cancer patients comprising administering to a cancer patient a therapeutically effective amount of an anti-PD-1 antibody and optionally other anti-cancer agents in addition to the anti-PD-1 antibody. In certain embodiments, the cancer patient suitable for treatment by the methods of the invention is preferably CD8 activated in peripheral blood+The percentage of T cells in total CD8+ T cells in peripheral blood is greater than or equal to 65%. In certain embodiments, cancer patients treated by the methods of the invention preferably have a percentage of NK cells in peripheral blood mononuclear cells of greater than or equal to 26%. In certain embodiments, the cancer patient suitable for treatment by the methods of the invention is preferably CD8 activated in peripheral blood+The percentage of T cells in total CD8+ T cells in peripheral blood is greater than or equal to 65%, and the percentage of NK cells in peripheral blood mononuclear cells is greater than or equal to 26%. In a preferred embodiment, the NK cell is CD3-CD16+CD54+NK cells. In thatIn certain embodiments, the cancer patient treated by the methods of the invention is preferably a cancer patient in which a mutation in the EMSY gene is detected in tumor tissue.
In certain embodiments, the present invention provides methods of treating an individual/patient having melanoma. In some embodiments, the invention includes methods of treating melanoma or individuals having melanoma. In some embodiments, the method comprises administering to the individual a therapeutically effective dose of a combination of: (a) an anti-cancer agent that is an Ab or antigen-binding fragment thereof that specifically binds to a PD-1 receptor and inhibits PD-1 activity; and (b) another anti-cancer therapy. In one embodiment, the present invention relates to a method of treating melanoma or an individual having melanoma in an individual in need thereof comprising administering to the individual an effective amount of a combination of: (i) standard therapy for melanoma, as disclosed elsewhere herein, or (ii) other anti-cancer agents. In some embodiments, the additional anti-cancer agent is selected from a CDK4/6 inhibitor, a FGF3/4/19 inhibitor, a PARP inhibitor, or a VEGFR inhibitor. Since about 50% of patients with asian melanoma are mucosal melanomas and about 20% are mucosal melanomas, in some embodiments, the melanoma is mucosal melanoma. In other embodiments, the melanoma is acro-melanoma.
anti-PD-1 antibodies suitable for use in the methods of the invention
The antibody PD-1 antibody suitable for the method of the invention is an immunosuppressive effect achieved by binding PD-1 with high specificity and affinity, blocking the binding of PD-L1/2 to PD-1, and inhibiting PD-1 signal transduction. In any of the treatment methods disclosed herein, the anti-PD-1 antibody includes an antigen-binding portion or fragment that binds to the PD-1 receptor and exhibits functional properties similar to a whole Ab in inhibiting ligand binding and upregulating the immune system. In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is an anti-PD-1 antibody or antigen-binding fragment thereof that cross-competes for binding to human PD-1 with terieprinimab. In other embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is a chimeric, humanized, or human Ab or antigen-binding fragment thereof. In certain embodiments for treating a human subject, the Ab is a humanized Ab.
In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof comprises a heavy chain constant region of human IgG1 or IgG4 isotype. In some embodiments, the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody or antigen-binding fragment thereof comprises the S228P mutation that replaces a serine residue in the hinge region with a proline residue that is typically present at the corresponding position of an IgG1 isotype antibody. In certain embodiments of any of the methods of treatment described herein comprising administering an anti-PD-1 antibody, the anti-PD-1 antibody is tereprimab. In some embodiments, the VEGFR inhibitor is axitinib. In some embodiments, the anti-PD-1 antibody is selected from the group consisting of humanized antibodies 38,39,41 and 48 described in WO 2014206107.
CDK4/6 inhibitors suitable for use in the methods of the invention
In some embodiments of the invention for use in the treatment of a tumor, the additional anti-cancer agent, i.e., the anti-cancer agent used in combination with the anti-PD-1 antibody in addition to the anti-PD-1 antibody, is a CDK4/6 inhibitor. CDK4/6 inhibitors currently approved by the FDA for marketing are mainly verzenio (abemaciciclib) from geneva, and are mainly used for treating adult patients with advanced or metastatic breast cancer who are Hormone Receptor (HR) positive and human epidermal growth factor receptor 2(HER2) negative for disease progression after endocrine therapy; and Kisqali (Ribociclib ) by nova, which in combination with an aromatase inhibitor can be used as a first line drug for the treatment of female patients with post-menopausal, advanced metastatic breast cancer that are HR positive and HER2 negative; the third is ibrane (Palbociclib ) from pfizer, which is used in combination with letrozole to treat metastatic breast cancer in postmenopausal women who are Estrogen Receptor (ER) positive and human epidermal growth factor receptor 2(HER2) negative.
FGF/FGFR inhibitors suitable for use in the methods of the invention
In some embodiments of the invention for treating tumors, the additional anti-cancer agent, i.e., the anti-cancer agent used in combination with the anti-PD-1 antibody in addition to the anti-PD-1 antibody, is an FGF/FGFR inhibitor. At present, FGF/FGFR inhibitors are not on the market, VEGFR/PDGFR/FGFR inhibitors nintedanib of Boringer Greenham is used for treating liver cancer, non-small cell lung cancer and idiopathic fibrosis, obtains breakthrough drug qualification of FDA in 2014, and Delitinib is in clinical stage at present in China.
PARP inhibitors useful in the methods of the present invention
In some embodiments of the invention for treating a tumor, the additional anti-cancer agent, i.e., the anti-cancer agent used in combination with the anti-PD-1 antibody in addition to the anti-PD-1 antibody, is a PARP inhibitor. PARP inhibitors currently on the market include Olaparib of aspirin for the treatment of ovarian cancer or primary peritoneal cancer; rubraca from Clovis for the treatment of ovarian cancer; and zejua of saxidong for the treatment of ovarian and peritoneal cancer. There are several dozen other PARP inhibitors in clinical research.
VEGFR inhibitors useful in the methods of the invention
In some embodiments of the invention for treating melanoma, the other anti-cancer agent, i.e., the anti-cancer agent used in combination with the anti-PD-1 antibody in addition to the anti-PD-1 antibody, is a VEGFR inhibitor. Approved VEGFR inhibitors include: apatinib for the treatment of advanced gastric cancer and advanced non-Lin non-small cell lung cancer; axitinib for use in the treatment of metastatic renal cell carcinoma; sorafenib is used for treating hepatocellular carcinoma, thyroid cancer and the like; pazopanib for the treatment of advanced renal cell carcinoma; sunitinib for treating gastrointestinal stromal tumor, advanced renal cell carcinoma, lenvatinib for treating thyroid cancer; regorafenib is useful for treating gastrointestinal stromal tumors. There is no approval for the treatment of melanoma, particularly for the treatment of advanced melanoma. Therefore, whether the VEGFR inhibitor and the anti-PD-1 antibody can be used for treating melanoma or not and the efficacy and the clinical safety are not foreseen, and a large number of clinical tests are needed for verification.
Use, therapy, medicament and kit
In one aspect of the invention, there is provided a method for treating cancer in an individual, the method comprising administering to the individual a combination therapy comprising an anti-PD-1 antibody and one or more of a CDK4/6 inhibitor, a FGF/FGFR inhibitor, a PARP inhibitor or a VEGFR inhibitor.
The combination therapy may also comprise one or more additional therapeutic agents. The additional therapeutic agents may be chemotherapeutic or biotherapeutic agents other than CDK4/6 inhibitors, FGF/FGFR inhibitors, PARP inhibitors or VEGFR inhibitors.
Each therapeutic agent in the combination therapies of the present invention may be administered alone or in a pharmaceutical composition comprising the therapeutic agent and one or more pharmaceutically acceptable carriers, excipients, and diluents according to standard pharmaceutical practice.
Each therapeutic agent in the combination therapy of the invention may be administered simultaneously, concurrently or sequentially in any order. The therapeutic agents in the combination therapy are administered in different dosage forms, such as one drug being a tablet or capsule and the other drug being a sterile liquid, and/or at different dosing times, such as the chemotherapeutic agent being administered at least daily and the biologic therapeutic agent being administered infrequently, such as once every week, or every two weeks or every three weeks.
In some embodiments, the CDK4/6 inhibitor, FGF/FGFR inhibitor, PARP inhibitor or VEGFR inhibitor is administered prior to the administration of the anti-PD-1 antibody, while in other embodiments, the CDK4/6 inhibitor, FGF/FGFR inhibitor, PARP inhibitor or VEGFR inhibitor is administered after the administration of the anti-PD-1 antibody.
In some embodiments, at least one of the therapeutic agents in the combination therapy is administered using the same dosing regimen (dose, frequency, duration of treatment) when the drug is used as a monotherapy to treat the same cancer.
Each small molecule therapeutic in the combination therapies described herein can be administered orally or parenterally (such as intravenous, intramuscular, intraperitoneal, subcutaneous, rectal, topical, or transdermal routes of administration).
The combination therapy described in the present invention may be administered before or after surgery and may be administered before, during or after radiotherapy.
In some embodiments, the combination therapy of the present invention is administered to a patient who has not previously been treated with a biologic or chemotherapeutic agent. In other embodiments, the combination therapy is administered to a patient who is unable to achieve a sustained response following treatment with the biologic or chemotherapeutic agent.
The combination therapies described in the present invention can be used to treat tumors found by palpation or by imaging techniques known in the art, such as MRI, ultrasound or CAT scanning.
The combination therapies described herein are preferably administered to cancer patients who are tested positive for PD-L1 expression or positive for CD8 +.
The choice of dosing regimen for the combination therapies of the invention depends on several factors including, but not limited to, serum or tissue conversion rate, degree of symptoms, immunogenicity, and accessibility to the target cells, tissues, organs of the individual being treated. Preferably, the dosing regimen will be such that the maximum amount of each therapeutic agent is delivered to the patient in conjunction with an acceptable degree of side effects. Thus, the dosage and frequency of administration of each of the biotherapeutic and chemotherapeutic agents in the combination therapy will depend on the particular therapeutic agent, the severity of the cancer being treated and the characteristics of the patient.
The anti-PD-1 antibodies and one or more of CDK4/6 inhibitors, FGF/FGFR inhibitors, PARP inhibitors, or VEGFR inhibitors described herein can be provided as a kit comprising a first container and a second container and a package insert.
The first container contains at least one dose of a medicament comprising an anti-PD-1 antibody, the second container contains at least one dose of a medicament comprising one or more of a CDK4/6 inhibitor, a FGF/FGFR inhibitor, a PARP inhibitor, or a VEGFR inhibitor, and the package insert or label contains instructions for using the medicament to treat cancer. The kit may further comprise other materials useful for administering drugs, such as diluents, filter paper, IV bags and threads, needles and syringes. As a preferred approach, the instructions may state that the drug is intended for the treatment of a cancer patient whose PD-L1 expression is positive as tested by the ICH assay.
Method for predicting effect of anti-PD-1 antibody on cancer treatment
The efficacy of anti-PD-1 antibodies, particularly of tropipalimab, of the invention for treating cancer in an individualA method for predicting fruit comprising detecting a biomarker selected from, but not limited to, CD8 in the peripheral blood of a patient prior to treatment+T cells or NK cells.
In one embodiment, the cancer of the invention is melanoma. In one embodiment, the CD8 in the prediction method of the present invention+T cells are in an activated state. In one embodiment, the activated CD8 of the present invention+The percentage of T cells in the total CD8+ T cells in the peripheral blood is not less than 65%, preferably not less than 75%. In one embodiment, the percentage of NK cells in peripheral blood mononuclear cells according to the present invention is not less than 23%, preferably not less than 26%. In certain embodiments, the NK cell is CD3-CD16+CD54+NK cells.
The invention also includes methods of using the EMSY gene to predict the effect of a tumor patient on treatment with an anti-PD-1 antibody. The presence of the EMSY gene mutation indicates that the tumor patient is suitable for treatment with an anti-PD-1 antibody.
In certain embodiments, the invention also provides biomarkers (particularly CD 8)+T cells and/or NK cells) in the preparation of a kit for predicting the effect of an anti-PD-1 antibody on the treatment of cancer. Such reagents include, for example, reagents for detecting the frequency and activation state of T lymphocytes in whole blood, such as CD45RA staining reagents and the like.
The invention also comprises the application of the reagent for detecting the EMSY gene in preparing a kit for predicting the effect of the anti-PD-1 antibody on treating the cancer. Such reagents include, but are not limited to, reagents conventionally used in assays, including, but not limited to, primers, probes, reagents required for PCR, and the like.
Abbreviations
Throughout the description and examples of the present invention, the following abbreviations are used:
BID one dose, 2 times daily
CDR complementarity determining region
Disease-free survival of DFS
FR framework regions
IgG immunoglobulin G
IHC immunohistochemistry
OR Overall response
Objective rate of remission of ORR
OS Total survival
Progression of PD disease
Progression free survival of PFS
PR partial response
CR complete response
Stabilization of SD disease
Dose limiting toxicity of DLT
Maximum tolerated dose of MTD
AE adverse events
Q2W one dose every two weeks
One daily dose of QD
Long-term exposure to CSD
non-CSD non-long-term sun exposure type
IRC independent review Committee
Adverse effects of TRAE associated with therapy
SAE Severe adverse reaction
RO receptor occupancy rate
UC urothelial carcinoma
RCC renal cell carcinoma
MM metastatic melanoma
The invention is further illustrated by the following examples, which should not be construed as limiting the invention. The contents of all references cited throughout this application are expressly incorporated herein by reference.
Examples
Example 1: clinical study of anti-PD-1 antibody alone for tumor treatment
Grouping standard: eligible subjects must (1) be between 18 and 70 years of age, (2) have metastatic melanoma or urinary cancer, (3) be refractory to standard systemic treatment, (4) have an ECOG score of 0 or 1, (5) have no history of autoimmune disease or persistent infection, (6) have not previously received any immunotherapy.
The subject must have an assessable lesion according to RECIST v1.1 criteria, not allow concurrent treatment with anti-tumor drugs, not allow treatment with systemic steroid drugs or not have been treated with anti-CTLA 4, anti-PD-1, anti-PD-L1 antibodies. Demographic data for the enrolled subjects are shown in table 1.
Table 1: demographic data of the subjects in the cohort
Figure BDA0002980998550000231
The tested drugs are: the anti-PD-1 antibody, torepalimab (WO 2014206107).
There are three anti-PD-1 antibody dose groups for this assay, which are: 1mg/kg (n-15), 3mg/kg (n-15) and 10mg/kg (n-6), intravenously every two weeks (Q2W). The detailed parameters are shown in the table 2 tumor histology parameter table.
Table 2: tumor histology parameter table
Histology of tumors 1mg/kg 3mg/kg 10mg/kg Total of
Melanoma (MEA) 10 10 2 22
Urothelial cancer 3 2 3 8
Renal cell carcinoma 2 3 1 6
Total of 15 15 6 36
And (3) clinical design:
this was a one-armed, phase I clinical, non-blind clinical trial divided into 2 groups (group a: dose escalation; group B: dose escalation). This study was conducted to evaluate the safety, tolerability, pharmacokinetics, pharmacodynamics, immunogenicity, and antitumor activity of anti-PD-1 antibodies.
Group A: inclusion of up to 18 subjects
The planned doses were 1mg/kg, 3mg/kg and 10mg/kg, Q2W.
At least 3 subjects were initially enrolled at each dose level. Subjects received one 60 minute intravenous injection of anti-PD-1 antibody sequentially for 4 weeks of toxicity assessment.
If there was no DLT in the first three subjects, it would be escalated to the next dose group, which would be expanded to a total of 6 subjects if DLT occurred.
Subjects continued to receive the predetermined dose level of anti-PD-1 antibody every two weeks 28 days after the first dose, followed by radiologic evaluation every 8 weeks. And performing corresponding evaluation by adopting RECIST v1.1 reaction evaluation standard. Subjects with disease progression or drug resistance toxicity will be excluded from the study.
Any dose not exceeding MTD for group a escalation group can be expanded to up to 12 subjects in group B to further assess safety, pharmacokinetics, pharmacodynamics and clinical activity.
Group B: subjects were evaluated by intravenous injection of anti-PD-1 antibody at 1, 3, or 10mg/kg every 2 weeks, followed by radiation every 8 weeks until disease progression, intolerable toxicity, or voluntary withdrawal from the study. Adverse events were reported in 2018, 7 and 31 days.
1.1 pharmacokinetic determination
Serum samples were taken from the subjects prior to dosing and continuously 28 days after the first anti-PD-1 antibody (tropilimumab) administration. PK was evaluated.
The PK parameter results are shown in table 3 below, and serum concentrations of tropimalimab reached Cmax 1 hour after injection and slowly exited the cycle. It shows a dose-dependent linear PK profile. The serum half-lives after intravenous injection of tropimalimab, Q2W were 9.5 + -2.0, 16.5 + -7.8 and 13.9 + -5.5 (days), respectively. The trough concentration of the tropimalimab in serum stabilized approximately after 5 consecutive doses, with the clinically steady-state trough concentrations of tropimalimab in the dose groups 1mg/kg, 3mg/kg and 10mg/kg being 8.9 + -4.4, 37.8 + -17.5 and 174.3 + -95.6 g/mL, respectively.
Table 3: PK parameters in human serum for different dose groups of anti-PD-1 antibodies (toreplamab)
Figure BDA0002980998550000251
Figure BDA0002980998550000261
1.2 pharmacodynamic Studies of PD-1 receptor-occupied anti-PD-1 antibodies
Receptors that bind anti-PD-1 antibodies (tropimalimab) are detected by subtype-specific antibodies that recognize the IgG4 Fc portion of tropimalimab. Cells were first incubated with a saturating concentration of toreplimab and then stained with PE-labeled anti-human IgG4 Fc to assess the total amount of PD-1. PD-1 positive target cells are predominantly present in the CD45RA-T cell subpopulation, which represents a non-naive or activated T cell population. As shown in table 4, most subjects in the three dose groups maintained full (> 80%) occupancy of PD-1 receptors on peripheral blood T lymphocytes during treatment. Pharmacodynamic readings of PD-1 receptor occupancy as shown in figure 1 are not associated with clinical response, with similar RO in both responder and non-responder treatments.
Table 4: PD-1 Receptor Occupancy (RO) of torepalimab in 3 dose groups
Figure BDA0002980998550000262
1.3 safety study:
from 30/3/2016 to 26/2016, 36 patients with intermediate and advanced metastatic melanoma (n-22), renal cell carcinoma (n-6) or urinary tract tumor (n-8) refractory to standard systemic therapy were enrolled in three dose escalation groups: 1mg/mL (n ═ 3), 3mg/mL (n ═ 4), 10mg/mL (n ═ 3), and three dose expansion groups: 1mg/kg (n-12), 3mg/kg (n-11), 10mg/kg (n-3).
As shown in tables 5 and 6, the anti-PD-1 antibody has an acceptable overall safety profile even at doses up to 10mg/kg Q2W, and no new safety signals are present.
Table 5: summary of treatment-related adverse reactions (TRAE) in each dose group
Figure BDA0002980998550000271
Table 6: adverse events commonly (≧ 20%) associated with tropilimumab treatment in all subjects (n ═ 36)
Figure BDA0002980998550000272
Figure BDA0002980998550000281
1.4 antitumor Activity Studies:
investigators evaluated clinical responses every 8 weeks using RECIST v 1.1. By 7/3 days 2018, 1 patient with acromelanoma in a 1mg/kg expanded cohort voluntarily dropped out of the trial on day 15 after receiving two doses of TORIPALIMAB, and was not evaluated post-treatment imaging. The subject was still included in the intended treatment population for efficacy assessment. In all 36 subjects, 1 confirmed complete response (acromelanoma), 7 confirmed partial responses (2 acromelanoma, 1 mucosal melanoma, 2 UC and 2 RCC), and 10 stable diseases (including 1 unidentified PR, UC), an Objective Response Rate (ORR) of 22.2% (95% CI,10.1-39.2) and a disease control rate of 50.0% (95% CI,32.9-67.1) were observed, with the results shown in table 7.
Clinical responses were observed at each dose level and in all three cancer types. 1. The optimal objective response rates for the 3, 10mg/kg dose groups were 21.4% (including 1 case of unconfirmed PR), 26.7%, and 33.3%, respectively, while the DCR was 64.3%, 46.7%, and 33.3%, respectively. No significant dose-related clinical efficacy was observed. 7/8 the responding subject received at least two systemic treatments. The change in tumor size (sum of target lesion diameters) and the optimal response from baseline over time are shown in the spider graph of fig. 2A and the waterfall graph of 2B.
Median time to response was 12 weeks (range 8 to 65.7 weeks). Notably, one subject with UC remained in stable condition for 460 days until PR was obtained. The response in most patients is sustained with a corresponding median time of 5.6 months (from 1.8 months to 17.7+ months).
Of the 13 acro melanoma subjects evaluated, 1 confirmed CR, 2 confirmed PR, 3 SD, with an ORR of 23.1% and a DCR of 46.2%. Meanwhile, 1PR and 1 SD were observed in 4 patients with mucosal melanoma. The duration of response was 8.0, 8.4, 10.6 and 17.7+ months for 4 melanoma patients who received 2-5 pre-line systemic treatment. Of these 1 mucosal melanoma subjects had a sustained response after 19.5 months of treatment with anti-PD-1 antibody.
Table 7: clinical response to TORIPALIMAB in RECIST v1.1 was evaluated every 8 weeks
Histology 1mg/kg(n=15) 3mg/kg(n=15) 10mg/kg(n=6) In total (n ═ 36) ORR,DCR
Melanoma (n ═ 22) 1PR,4SD 1CR,2PR,2SD 0PR 1CR,3PR,6SD 18.2%,45.5%
UC(n=8) 1PR,2SD(1uPR) 1SD 1PR 2PR,3SD 25.0%,67.5%
RCC(n=6) 1SD 1PR 1PR 2PR,1SD 33.3%,50.0%
Total 2PR,7SD 1CR,3PR,3SD 2PR 1CR,7PR,10SD 22.2%,50.0%
ORR,DCR 13.3%,60.0% 26.7%,46.7% 33.3%,33.3% 22.2%,50.0%
Progression free survival and overall survival
In the last visit of 2018, 7, 3, 21 subjects died, 4 subjects were unconnected, and 11 patients survived. The mean progression-free survival was 2.8 months (range 0.5 to 21.9+ months) (fig. 3). The mean overall survival was 12.2 months (range 1.7 to 27.5+ months) (fig. 4). Annual survival rates were 50.0% for all subjects and melanoma subgroups.
Example 2: research on correspondence between Natural Killer (NK) cells and anti-PD-1 antibody therapeutic effects on tumor
The frequency and activation status of T lymphocytes in whole blood (stained by CD45 RA) were assessed during the clinical trial described in example 1. anti-PD-1 antibody to CD8 activated in peripheral blood of individual subjects+Frequency of T lymphocytesThe influence is not great. As shown in FIG. 5, CD8 in the group with PFS greater than 250 days+Subjects with high cellular activation rates were enriched. Whereas all subjects activated CD8 prior to toriplalimab treatment+The average percentage of (c) was 65%. Finding a durable responder (PFS) of 4/5>300 days) of peripheral blood has more than 80% of activated CD8+(Total CD 8)+Cells) and more than 26% CD3-CD16+CD54+NK cells (accounting for total monocytes); in contrast CD8 activated in all subjects+The average percentages of T cells and NK cells were 65% and 20%, respectively.
Example 3: correlation study of biomarker and clinical curative effect
The correlation between tumor histology and anti-PD-1 antibody in clinical efficacy was analyzed by double IHC staining of tumor biopsy specimens from 28 patients with PD-L1 CD 8. The rabbit anti-human PD-L1 antibody SP142 and anti-CD 8 clone 4B11 from Roche were used. The positive state of PD-L1 is defined as the membrane staining intensity of the tumor cells being more than or equal to 1 percent.
As shown in fig. 6 and 7, subjects positively expressing PD-L1 benefited more from TORIPALIMAB treatment than subjects negative for PD-L1 in tumor biopsies (43.8% ORR,62.5DCR versus 0% ORR, 50% DCR). The subpopulation with high PD-1 expression (> 50%) responded best to TORIPALIMAB treatment with an ORR of 57.1% and a DCR of 71.4%. Notably, all ORR (CR + PR) subjects were PD-L1 and TIL double positive. Co-localization of TILs with PD-L1+ cells (co-localization) was found in tumor biopsies, with TILs also being present in 93.8% of PD-L1 positive tumors. Similarly, TIL-positive subjects responded better to TORIPALIMAB treatment than TIL-negative subjects (31.8% ORR, 59.1% DCR vs 0% ORR, 50% DCR) (fig. 6). However, subjects, whether PD-L1 negative or TIL negative, still benefited from PD-1 blocking therapy, and disease control was achieved in 50% of both groups following TORIPALILAMAB treatment.
Example 4: gene sequencing and tumor mutation burden determination
In the experiment of example 1, we used second generation sequencing technology to perform comprehensive genome sequencing on FFPE tumors and paired peripheral blood samples from 24 subjects using 450 cancer-associated gene panels. Tumor Mutation Burden (TMB) was assessed by analysis of somatic mutations including coding base substitutions and megabase insertions of the panel sequences studied. As shown in figure 8, all subjects had less than 10 mutations/Mb of TMB, except one acromelanoma subject (17.6 mutations/Mb). TMB levels were correlated with clinical response and overall survival (as shown in figure 8). Using 6 mutations/Mb as nodes, subjects with more than 6 mutations/Mb (n-12) achieved better clinical responses than subjects with less than 6 mutations/Mb (n-12) (41.7% ORR and 58.3% DCR versus 16.7% ORR and 33.3% DCR).
As shown in fig. 9, the genomic map also identified unique gene amplifications in limb melanoma patients (n-7), including Cyclin D1, CDK4/6 (5/7), fibroblast growth factor (FGF19/3/4) (3/7), and EMSY (3/7) (fig. 9). This aggregative gene amplification was not observed in mucosal or UV-induced cutaneous melanoma subjects.
And 2 of 7 acromelanoma subjects responded to anti-PD-1 antibody treatment (1CR and 1PR), and EMSY gene amplification was present.
In addition, as can be seen from fig. 9, DNA mutations or gene amplification were rare in mucosal melanoma subjects.
Example 5: clinical study of anti-PD-1 antibody in combination with axitinib for treating metastatic mucosal melanoma
Grouping standard: (1) mucosal melanoma; (2) with historical or fresh tumor biopsies; (3) prior to or one systemic chemotherapy; (4) ECOG score of 0-1.
The tested drugs are: commercially available axitinib tablets; anti-PD-1 antibodies (WO 2014206107).
The administration mode comprises the following steps: a total of 33 subjects were screened in the group by 2018, 4 months and 2 days.
A first part: the administration mode of 001-; axitinib was administered orally, 5mg, 2 times/day; 004 cases of 006 cases of administration are intravenous drip of anti-PD-1 antibody, 1mg/kg, 2 weeks/time; axitinib was administered orally, 5mg, 2 times/day.
Clinical results: no DLT was found and treatment-related AEs appeared in 97% of subjects, but most treatment-related AEs were grade 1 and 2, with no treatment-related AEs found on grades 4 and 5. In 6 subjects, 18.2% of treatment-related grade 3 AEs were found, including weight loss, cholesterol increase, etc., but none of the new AEs were found to appear beyond the two drugs administered alone.
The recommended dose combination was: anti-PD-1 antibody was administered by intravenous drip at 3mg/kg for 2 weeks/time; axitinib was administered orally, 5mg, 2 times/day.
A second part: the recommended dose was expanded to 30 subjects, and 27 subjects were included in the group.
007-033 cases of administration were intravenous drip of anti-PD-1 antibody at 3mg/kg for 2 weeks/time; axitinib was administered orally, 5mg, 2 times/day.
The clinical effect is as follows: clinical efficacy was assessed every 8 weeks by IRC, RECIST 1.1 therapeutic criteria. Of the 33 subjects, 20 PR and 9 SD with an ORR of 60.65% and a DCR of 87.9%, and 16 of the 20 PR subjects responded consistently without reaching a median sustained response time. The results are shown in FIG. 10.
Sequence listing
<110> Shanghai Junshi biomedical science and technology Co., Ltd
<120> use of anti-PD-1 antibodies in the treatment of tumors
<130> 187222F1
<160> 10
<170> SIPOSequenceListing 1.0
<210> 1
<211> 16
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Arg Ser Ser Gln Ser Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu
1 5 10 15
<210> 2
<211> 7
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Lys Val Ser Asn Arg Phe Ser
1 5
<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Phe Gln Gly Ser His Val Pro Leu Thr
1 5
<210> 4
<211> 5
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Asp Tyr Glu Met His
1 5
<210> 5
<211> 17
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe Lys
1 5 10 15
Gly
<210> 6
<211> 16
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe Asp Val
1 5 10 15
<210> 7
<211> 112
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Phe Gln Gly
85 90 95
Ser His Val Pro Leu Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 8
<211> 125
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Gln Gly Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Glu Met His Trp Val Arg Gln Ala Pro Ile His Gly Leu Glu Trp Ile
35 40 45
Gly Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser
115 120 125
<210> 9
<211> 219
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 9
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Phe Gln Gly
85 90 95
Ser His Val Pro Leu Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 10
<211> 452
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 10
Gln Gly Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Glu Met His Trp Val Arg Gln Ala Pro Ile His Gly Leu Glu Trp Ile
35 40 45
Gly Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr
115 120 125
Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser
130 135 140
Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu
145 150 155 160
Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His
165 170 175
Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser
180 185 190
Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys
195 200 205
Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu
210 215 220
Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu
225 230 235 240
Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu
245 250 255
Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser
260 265 270
Gln Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu
275 280 285
Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr
290 295 300
Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn
305 310 315 320
Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser
325 330 335
Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln
340 345 350
Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val
355 360 365
Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val
370 375 380
Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro
385 390 395 400
Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr
405 410 415
Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val
420 425 430
Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu
435 440 445
Ser Leu Gly Lys
450

Claims (9)

1. Use of an anti-PD-1 antibody and/or antigen-binding fragment thereof in the manufacture of a medicament for treating acro-melanoma or mucosal-type melanoma in an asian patient, wherein the light chain complementarity determining region of the anti-PD-1 antibody is set forth in SEQ ID NO: 1.2 and 3, and the heavy chain complementarity determining region is shown in SEQ ID NO: 4. 5 and 6.
2. The use of claim 1, wherein the light chain variable region of the anti-PD-1 antibody is as set forth in SEQ ID NO: 7, the heavy chain variable region is shown as SEQ ID NO: shown in fig. 8.
3. The use of claim 1, wherein the anti-PD-1 antibody is a tropilimumab.
4. Use of an anti-PD-1 antibody and/or an antigen-binding fragment thereof and a VEGFR inhibitor in the manufacture of a medicament for treating a patient with mucosal melanoma, wherein the light chain complementarity determining region of the anti-PD-1 antibody is set forth in SEQ ID NO: 1.2 and 3, and the heavy chain complementarity determining region is shown in SEQ ID NO: 4. 5 and 6.
5. The use of claim 4, wherein the light chain variable region of the anti-PD-1 antibody is as set forth in SEQ ID NO: 7, the heavy chain variable region is shown as SEQ ID NO: shown in fig. 8.
6. The use of claim 4, wherein the anti-PD-1 antibody is a tropilimumab.
7. The use of any of claims 4-6, wherein said VEGFR inhibitor is selected from the group consisting of: axitinib, sunitinib, sorafenib, apatinib, revaprenib, and pazopanib.
8. The use of any one of claims 4-6, wherein the VEGFR inhibitor is axitinib or a pharmaceutically acceptable salt thereof.
9. The use of any one of claims 4 to 6, wherein the patient is an Asian patient.
CN202110287273.9A 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors Pending CN112972675A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110287273.9A CN112972675A (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110287273.9A CN112972675A (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors
CN201811043430.6A CN110882385B (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
CN201811043430.6A Division CN110882385B (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors

Publications (1)

Publication Number Publication Date
CN112972675A true CN112972675A (en) 2021-06-18

Family

ID=69744564

Family Applications (2)

Application Number Title Priority Date Filing Date
CN202110287273.9A Pending CN112972675A (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors
CN201811043430.6A Active CN110882385B (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors

Family Applications After (1)

Application Number Title Priority Date Filing Date
CN201811043430.6A Active CN110882385B (en) 2018-09-07 2018-09-07 Use of anti-PD-1 antibodies in the treatment of tumors

Country Status (1)

Country Link
CN (2) CN112972675A (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023006055A1 (en) * 2021-07-29 2023-02-02 Shanghai Junshi Biosciences Co., Ltd. Anti-pd-1 antibody pharmaceutical composition and use thereof

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021110122A1 (en) * 2019-12-05 2021-06-10 基石药业(苏州)有限公司 Combination therapy of cdk4/6 inhibitor
CN111420060B (en) * 2020-03-30 2022-04-08 南方医科大学南方医院 Anti-tumor combined medicine composition and application thereof
US20230322930A1 (en) * 2020-08-28 2023-10-12 Shanghai Junshi Biosciences Co., Ltd. Use of an anti-pd-1 antibody and a cytotoxic anticancer drug in treatment of non-small cell lung cancer
WO2022184148A1 (en) * 2021-03-03 2022-09-09 上海君实生物医药科技股份有限公司 Il-21-anti-albumin single-domain antibody fusion protein pharmaceutical composition and use thereof
CN115227812A (en) * 2021-04-22 2022-10-25 上海君实生物医药科技股份有限公司 Use of anti-PD-1 antibody in combination with first-line chemotherapy for treating advanced NSCLC
CN115364208A (en) * 2021-05-17 2022-11-22 上海君实生物医药科技股份有限公司 Medicine and method for treating patient with completely resected mucosal melanoma
CN113325106B (en) * 2021-05-31 2022-07-26 中国医学科学院北京协和医院 Liquid chromatography tandem mass spectrometry method for detecting concentration of Terepril monoclonal antibody drug in human plasma based on characteristic peptide segment
WO2023134787A2 (en) * 2022-01-14 2023-07-20 上海君实生物医药科技股份有限公司 Use of combination of anti-pd-1 antibody and anti-vegf antibody in treatment of hepatocellular carcinoma
CN116942810A (en) * 2022-04-26 2023-10-27 上海君实生物医药科技股份有限公司 anti-PD-1 antibodies and anti-EGFR antibody combinations and their use in the treatment of head and neck squamous cell carcinoma

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1303430A (en) * 1998-04-30 2001-07-11 贝林格尔·英格海姆国际有限公司 FAP alpha specific antibody with improved producibility
CN104250302A (en) * 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-PD-1 antibody and its application
CN106390115A (en) * 2015-07-29 2017-02-15 上海君实生物医药科技股份有限公司 Stable preparation of humanized monoclonal antibody

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1303430A (en) * 1998-04-30 2001-07-11 贝林格尔·英格海姆国际有限公司 FAP alpha specific antibody with improved producibility
CN104250302A (en) * 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 Anti-PD-1 antibody and its application
CN106390115A (en) * 2015-07-29 2017-02-15 上海君实生物医药科技股份有限公司 Stable preparation of humanized monoclonal antibody

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ALEXANDER N. SHOUSHTARI ET AL.: "Efficacy of Anti-PD-1 Agents in Acral and Mucosal Melanoma", CANCER *
F STEPHEN HODI ET AL.: "Bevacizumab plus ipilimumab in patients with metastatic melanoma", CANCER IMMUNOL RES *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023006055A1 (en) * 2021-07-29 2023-02-02 Shanghai Junshi Biosciences Co., Ltd. Anti-pd-1 antibody pharmaceutical composition and use thereof

Also Published As

Publication number Publication date
CN110882385B (en) 2021-07-09
CN110882385A (en) 2020-03-17

Similar Documents

Publication Publication Date Title
CN110882385B (en) Use of anti-PD-1 antibodies in the treatment of tumors
TWI775781B (en) Therapeutic and diagnostic methods for cancer
JP6876629B2 (en) PD-L1 antagonist combination therapy
CN111492245A (en) Methods of treatment and diagnosis of cancer
KR20180008449A (en) Treatment and Diagnosis Methods for Cancer
KR20230023810A (en) Combination of a pd-1 antagonist and a vegfr inhibitor for treating cancer
JP2022068352A (en) Treatment method of lung cancer using combination of anti-pd-1 antibody and anti-ctla-4 antibody
KR20190015408A (en) Anti-PD-1 antibody for use in methods of treating tumors
US11572405B2 (en) Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
KR20220133243A (en) Method of treating cancer using anti-TIGIT antagonist antibody
JP2022058699A (en) Anti-pd-1 antibody for use in method of treatment of recurrent small cell lung cancer
EP3957326A1 (en) Use of anti-pd-1 antibody in preparation of medicament for treating solid tumors
EP3976090A1 (en) Combination therapies using cdk inhibitors
US20230212292A1 (en) Use of anti-pd-1 antibody in treating neuroendocrine tumors
CN112839644A (en) Methods of treating lung cancer with PD-1 axis binding antagonists, antimetabolites, and platinum agents
EP4104856A1 (en) Use of anti-pd-1 antibody in treatment of tumors
CN113893342B (en) Application of pharmaceutical composition containing anti-PD-1 antibody in preparation of medicines for treating advanced non-small cell lung cancer
WO2022042681A1 (en) Use of an anti-pd-1 antibody and a cytotoxic anticancer drug in treatment of non-small cell lung cancer
WO2021155840A1 (en) Use of anti-pd-1 antibody in treatment of malignant tumors
CN115364209A (en) Use of anti-PD-1 antibody in combination with chemotherapy for treating esophageal cancer
CN114099663A (en) Use of anti-PD-1 antibodies in the treatment of nasopharyngeal carcinoma
US20190211102A1 (en) Methods and combination therapy to treat 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