WO2021160151A1 - 抗pd-1抗体在***中的用途 - Google Patents

抗pd-1抗体在***中的用途 Download PDF

Info

Publication number
WO2021160151A1
WO2021160151A1 PCT/CN2021/076456 CN2021076456W WO2021160151A1 WO 2021160151 A1 WO2021160151 A1 WO 2021160151A1 CN 2021076456 W CN2021076456 W CN 2021076456W WO 2021160151 A1 WO2021160151 A1 WO 2021160151A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
cancer
melanoma
tumor
patients
Prior art date
Application number
PCT/CN2021/076456
Other languages
English (en)
French (fr)
Inventor
姚盛
冯辉
武海
Original Assignee
上海君实生物医药科技股份有限公司
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 上海君实生物医药科技股份有限公司 filed Critical 上海君实生物医药科技股份有限公司
Priority to CN202180013152.3A priority Critical patent/CN115052628A/zh
Priority to EP21753897.4A priority patent/EP4104856A1/en
Priority to US17/904,096 priority patent/US20230082898A1/en
Publication of WO2021160151A1 publication Critical patent/WO2021160151A1/zh

Links

Images

Classifications

    • 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
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2818Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily against CD28 or CD152
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6876Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes
    • C12Q1/6883Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material
    • C12Q1/6886Nucleic acid products used in the analysis of nucleic acids, e.g. primers or probes for diseases caused by alterations of genetic material for cancer
    • 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
    • A61K2039/545Medicinal preparations containing antigens or antibodies characterised by the dose, timing or administration schedule
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/56Immunoglobulins specific features characterized by immunoglobulin fragments variable (Fv) region, i.e. VH and/or VL
    • C07K2317/565Complementarity determining region [CDR]
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/70Immunoglobulins specific features characterized by effect upon binding to a cell or to an antigen
    • C07K2317/76Antagonist effect on antigen, e.g. neutralization or inhibition of binding
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q1/00Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions
    • C12Q1/68Measuring or testing processes involving enzymes, nucleic acids or microorganisms; Compositions therefor; Processes of preparing such compositions involving nucleic acids
    • C12Q1/6869Methods for sequencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/106Pharmacogenomics, i.e. genetic variability in individual responses to drugs and drug metabolism
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12QMEASURING OR TESTING PROCESSES INVOLVING ENZYMES, NUCLEIC ACIDS OR MICROORGANISMS; COMPOSITIONS OR TEST PAPERS THEREFOR; PROCESSES OF PREPARING SUCH COMPOSITIONS; CONDITION-RESPONSIVE CONTROL IN MICROBIOLOGICAL OR ENZYMOLOGICAL PROCESSES
    • C12Q2600/00Oligonucleotides characterized by their use
    • C12Q2600/156Polymorphic or mutational markers

Definitions

  • the present invention relates to the use of anti-PD-1 antibodies in the treatment of tumors. Specifically, the present invention relates to the use of anti-PD-1 antibodies in the treatment of melanoma; and the use of anti-PD-1 antibodies in the preparation of drugs for the treatment of melanoma; and the use of biomarkers to predict the use of anti-PD-1 antibodies alone Methods of efficacy in the treatment of melanoma.
  • Immune escape is one of the characteristics of cancer.
  • Ahmadzadeh, M. et al., Blood, 114:1537-44 disclosed that tumor-specific T lymphocytes often exist in the tumor microenvironment, draining lymph nodes and peripheral blood, but due to the immunosuppressive mechanism network existing in the tumor microenvironment, its It is usually impossible to control the progression of the tumor.
  • CD8 + tumor-infiltrating T lymphocytes (TIL) usually 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), this ligand inhibits T cell activation and effector functions.
  • PD-1 and its ligands have become an important way for tumor cells to use it to inhibit T cells activated in the tumor microenvironment.
  • PD-1 Programmed death receptor 1 plays an important role in immune regulation and maintenance of peripheral tolerance.
  • PD-1 is mainly expressed in activated T cells and B cells, and its function is to inhibit the activation of lymphocytes. This is a normal peripheral tissue tolerance mechanism of the immune system to prevent and treat immune overload.
  • activated T cells that infiltrate the tumor microenvironment highly express PD-1 molecules, and the inflammatory factors secreted by activated white blood cells will induce tumor cells to highly express PD-1 ligands PD-L1 and PD-L2, resulting in tumor microenvironment
  • the PD-1 pathway of activated T cells is continuously activated, and the function of T cells is inhibited, unable to kill tumor cells.
  • Therapeutic PD-1 antibody can block this pathway, partially restore the function of T cells, so that activated T cells can continue to kill tumor cells.
  • PD-1/PD-L1 pathway blockade has been proven to be an effective way to induce durable anti-tumor responses in various cancer indications.
  • Monoclonal antibodies (mAbs) that block the PD-1/PD-L1 pathway can enhance the activation and effector functions of tumor-specific T cells, reduce tumor burden, and improve survival.
  • the FDA has approved two anti-PD1 monoclonal antibodies (nivolumab and pembrolizumab) and three anti-PD-L1 monoclonal antibodies (atezolizumab, avelumab, and durvalumab) for the treatment of human tumors.
  • Melanoma is the first approved indication for Nivolumab and Pembrolizumab in 2014.
  • melanoma has long been regarded as a highly immunogenic cancer, because lymphocytes infiltrate the tumor and the clinical response to high-dose IL-2 immunotherapy is often observed.
  • chronic UV radiation exposure related to DNA damage is the main cause of melanoma.
  • Chronic ultraviolet radiation sunburn (CSD) melanomas account for 95% of skin melanomas in the United States and other Western countries.
  • acral freckle-like melanoma (ALM) ⁇ 50%)
  • MM mucosal melanoma
  • One aspect of the present invention provides a method of treating a patient with melanoma, which comprises administering to the patient a therapeutically effective amount of an anti-PD1-1 antibody.
  • the second aspect of the present invention provides a use of an anti-PD-1 antibody in the preparation of a medicine for treating patients with melanoma.
  • the melanoma is advanced or metastatic melanoma.
  • the melanoma patient has a BRAF mutation.
  • the melanoma patient is selected from acral type, mucosal type, non-acral skin type, and melanoma patients with indeterminate primary focus.
  • the melanoma patient is a non-acral melanoma patient.
  • the melanoma patient is a melanoma patient with indeterminate primary focus.
  • the PD-L1 expression of the cancer tissue or section of the melanoma patient is tested positive.
  • the tumor mutation (TMB) of the cancer biopsy sample or the matched peripheral blood sample of the melanoma patient is ⁇ 3.6Muts/Mb.
  • the PD-L1 expression of the cancer tissue or section of the melanoma patient is tested positive, and the cancer biopsy sample or paired peripheral blood sample has a tumor mutation (TMB) ⁇ 3.6Muts /Mb.
  • the third aspect of the present invention provides a method for treating a patient with melanoma, which comprises administering to the patient a therapeutically effective amount of a CDK4/6 inhibitor alone or in combination with a therapeutically effective amount of a CDK4/6 inhibitor and anti-PD-1 Antibody.
  • the melanoma patient has CDK4 or CCDN1 gene amplification.
  • the fourth aspect of the present invention provides a kit for predicting the effect of anti-PD-1 antibody treatment on a tumor by an individual suffering from a tumor, the kit comprising: (a) for detecting tumor tissue or peripheral blood of the individual And (b) instructions for using the reagent described in (a) to detect the therapeutic effect of the anti-PD-1 antibody administered to an individual.
  • the fifth aspect of the present invention provides a kit for predicting the effect of anti-PD-1 antibody treatment on tumor by an individual suffering from a tumor, the kit comprising: (a) for detecting tumor tissue or peripheral blood in an individual CDK4/CCND1 gene mutation or amplification reagent; and (b) instructions for using the reagent described in (a) to detect the therapeutic effect of an individual using a CDK4/6 inhibitor alone or in combination with an anti-PD-1 antibody.
  • the sixth aspect of the present invention provides a kit for predicting the effect of anti-PD-1 antibody treatment on a tumor by an individual suffering from a tumor, the kit comprising: (a) for detecting NRAS in individual tumor tissue or peripheral blood Gene mutation reagent; and (b) instructions for using the reagent described in (a) to detect the therapeutic effect of the anti-PD-1 antibody administered to an individual.
  • the anti-PD-1 antibody is a monoclonal antibody or an antigen-binding fragment thereof.
  • the anti-PD-1 antibody can specifically bind to PD-1 and block the binding of PD-L1 or PD-L2 to PD-1.
  • the anti-PD-1 antibody specifically binds to PD-L1 or/and PD-L2, and blocks the binding of PD-L1 and/or PD-L2 to PD-1.
  • the anti-PD-1 antibody is an antibody comprising at least one complementarity determining region (CDR), and the amino acid sequence of the complementarity determining region (CDR) is selected from the following: SEQ ID NO: 1, 2, 3 , 4, 5, or 6.
  • the anti-PD-1 antibody is an antibody comprising a complementarity determining region (CDR), and the amino acid sequence of the light chain complementarity determining region (LCDR) is shown in SEQ ID NO: 1, 2 and 3.
  • the amino acid sequence of the heavy chain complementarity determining region (HCDR) is shown in SEQ ID NO: 4, 5, and 6.
  • the anti-PD-1 antibody comprises a light chain variable region (VL) and a heavy chain variable region (VH), wherein the amino acid sequence of VL is shown in SEQ ID NO: 7, and the VH The amino acid sequence is shown in SEQ ID NO: 8.
  • the anti-PD-1 antibody is an anti-PD-1 antibody comprising a light chain and a heavy chain, and the light chain comprises the amino acid sequence shown in SEQ ID NO: 9, and the heavy chain comprises SEQ ID NO: The amino acid sequence shown in 10.
  • the anti-PD-1 antibody is selected from one or more of nivolumab, pembrolizumab, toripalimab, Sintilimab, Camrelizumab, Tislelizumab, and Cemiplimab.
  • the single administration dose of the anti-PD-1 antibody or antigen-binding fragment thereof is about 0.1 mg/kg to about 10.0 mg/kg of the individual's body weight, for example, about 0.1 mg/kg, about 0.3 mg/kg. kg, about 1mg/kg, about 2mg/kg, about 3mg/kg, about 5mg/kg or 10mg/kg of individual body weight, or selected from about 120mg to about 480mg fixed dose, such as about 120mg, 240mg, 360mg or 480mg fixed dose .
  • the dosing cycle frequency of the anti-PD-1 antibody or antigen-binding fragment thereof is about once a week, once every two weeks, once every three weeks, once every four weeks, or once a month, Preferably it is once every two weeks.
  • the single administration dose of the anti-PD-1 antibody or antigen-binding fragment thereof is 1 mg/kg individual body weight, 3 mg/kg individual body weight, 10 mg/kg individual body weight, or 240 mg fixed dose, 480 mg fixed The dose is administered once every two weeks.
  • the anti-PD-1 antibody or antigen-binding fragment thereof is administered in a liquid dosage form, such as an injection, via a parenteral route, such as intravenous infusion.
  • the administration cycle of the anti-PD-1 antibody or antigen-binding fragment thereof can be one week, two weeks, three weeks, one month, two months, three months, four months, five Months, half a year or longer, optionally, the time of each dosing cycle can be the same or different, and the interval between each dosing cycle can be the same or different.
  • the CDK4/6 inhibitor is selected from the group consisting of Pabocinil, Rebocinil, and Permazelin.
  • the individual is a human.
  • the subject has melanoma and has not received anti-PD-1 or anti-PD-L1 immunotherapy in the past.
  • the cancer is advanced melanoma.
  • the cancer is metastatic melanoma.
  • the seventh aspect of the present invention provides a method for predicting the therapeutic effect of an anti-PD-1 antibody in a tumor patient, which comprises detecting a biomarker in tumor tissue or peripheral blood of the patient before treatment, wherein the biomarker is BRAF gene mutation, wherein the presence of the BRAF gene mutation indicates that the tumor patient is suitable for treatment with an anti-PD-1 antibody.
  • the eighth aspect of the present invention provides a method for predicting the effect of anti-PD-1 antibody treatment in a tumor patient, which includes detecting a biomarker in the tumor tissue of the patient, and the biomarker is CDK4 or CCND1 gene amplification.
  • the presence of the CDK4 or CCND1 gene amplification indicates that the tumor patient is suitable for treatment with an anti-PD-1 antibody and a CDK4/6 inhibitor in combination.
  • the presence of the CDK4 or CCND1 gene amplification indicates that the tumor patient is suitable for treatment with a CDK4/6 inhibitor.
  • the ninth aspect of the present invention provides a method for predicting the therapeutic effect of an anti-PD-1 antibody in a tumor patient, which comprises detecting a biomarker in the tumor tissue or peripheral blood of the patient before treatment, and the biomarker is NRAS gene mutation.
  • the presence of the NRAS gene mutation indicates that the anti-PD-1 antibody alone has an unsatisfactory therapeutic effect or is not suitable for the anti-PD-1 antibody alone for treatment.
  • the tumor is a solid tumor.
  • the tumor is melanoma.
  • Figure 1 The relationship diagram of the clinical phase II study of toripalimab in the treatment of locally advanced or metastatic melanoma.
  • Figure 2 Evaluation of clinical response in accordance with RECIST v1.1.
  • Figure 3 The progression-free survival PFS (A) of melanoma patients in this study and the overall survival OS (B) of melanoma patients in this study.
  • FIG. 4 The overall survival OS (A) of the response subgroup and the overall survival OS (B) of the melanoma subgroup in this study.
  • Figure 6 The relationship between clinical response and tumor PD-L1 expression and TMB.
  • A PD-L1 positive is defined as any intensity of membrane staining of tumor cells or immune cells with SP142IHC staining ⁇ 1%; TMB is calculated by whole-exome sequencing of somatic mutations in the coding region; 3.6Muts/Mb is used as Cut-off value; B: percentage of PD-L1 + or TMB ⁇ 3.6Muts/Mb in the melanoma subgroup; C: progression-free survival PFS of patients with PD-L1 + and PD-L1 -; D: PD-L1 + Overall survival of patients with PD-L1 - OS; E: Progression-free survival PFS of patients with TMB ⁇ 3.6Muts/Mb and TMB ⁇ 3.6Muts/Mb; F: TMB ⁇ 3.6Muts/Mb and TMB ⁇ 3.6 The overall survival of patients with Muts/Mb is OS.
  • FIG. 7 Gene variation and frequency of 98 patients by whole exome sequencing (WES).
  • Figure 8 The relationship between clinical response and signature scores of IFN- ⁇ -related genomes, inflammation-related genomes, and angiogenesis-related genomes.
  • the present invention relates to tumor treatment methods.
  • the method of the present invention includes administering an anti-PD-1 antibody or an antigen-binding fragment thereof alone to a patient in need; or includes administering an anti-PD-1 antibody in combination with other anticancer agents to a patient in need.
  • the present invention also relates to a method for using biomarkers to predict the efficacy of anti-PD-1 antibodies in the treatment of cancer, especially melanoma patients.
  • administering refers to introducing a composition containing a therapeutic agent into a subject using any of various methods or delivery systems known to those skilled in the art.
  • Anti-PD-1 antibody administration routes include intravenous, intramuscular, subcutaneous, peritoneal, spinal, or other parenteral administration routes, such as injection or infusion.
  • Parenteral administration refers to administration methods usually by injection other than enteral or local administration, including but not limited to intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, and intrasaccular , Intra-frame, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcutaneous, intraarticular, subcapsular, subarachnoid, intraspine, intradural and intrasternal injection and infusion, and intracorporeal electroporation.
  • AE adverse reaction
  • Medical treatments can have one or more related AEs, and each AE can have the same or different severity levels.
  • Tumor burden refers to the total amount of tumor material distributed throughout the body. Tumor burden refers to the total number of cancer cells in the entire body or the total size of the tumor. Tumor burden can be measured by a variety of methods known in the prior art, such as using calipers after the tumor is removed from the subject, or using imaging techniques (such as ultrasound, bone scanning, computed tomography when the tumor is in the body) (CT) or magnetic resonance imaging (MRI) scan) to measure its size.
  • imaging techniques such as ultrasound, bone scanning, computed tomography when the tumor is in the body) (CT) or magnetic resonance imaging (MRI) scan
  • tumor size refers to the total size of the tumor, which can be measured as the length and width of the tumor.
  • the size of the tumor can be measured by a variety of methods known in the prior art, such as using a caliper after the tumor is removed from the subject, or using imaging techniques (such as bone scan, ultrasound, CT or MRI scan) while in the body. size.
  • subject include any organism, preferably animals, more preferably mammals (e.g., rats, mice, dogs, cats, rabbits, etc.), and most preferably humans.
  • mammals e.g., rats, mice, dogs, cats, rabbits, etc.
  • the “antibody” as used herein refers to any form of antibody that can achieve the desired biological activity or binding activity. Therefore, it is used in the broadest sense, but is not limited to monoclonal antibodies, polyclonal antibodies, multispecific antibodies, humanized full-length human antibodies, chimeric antibodies, and camel-derived single domain antibodies.
  • An “antibody” specifically binds to an antigen and includes at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds. Each heavy chain includes a heavy chain variable region (VH) and a heavy chain constant region, and the heavy chain constant region includes three constant domains CH1, CH2, and CH3.
  • Each light chain contains a light chain variable region (VL) and a light chain constant region, and the light chain constant region contains a constant domain CL.
  • VL light chain variable region
  • CL constant domain
  • the VH and VL regions can be further subdivided into hypervariable regions called complementarity determining regions (CDR), which are interspersed in more conservative regions called framework regions (FR).
  • CDR complementarity determining regions
  • FR framework regions
  • both the light chain and heavy chain variable domains include FR1, CDR1, FR2, CDR2, FR3, CDR3, and FR4.
  • Amino acids are usually assigned to each domain according to the following definitions: Sequences of Proteins of Immunological Interest, Kabat et al.; National Institutes of Health, Bethesda, Md.; 5th edition; NIH publication number 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: 901-917 or Chothia et al. (1989) Nature 341:878-883.
  • the carboxy terminal part of the heavy chain can define the constant region that is mainly responsible for effector functions.
  • human light chains are divided into kappa chains and lambda chains.
  • Human heavy chains are usually classified into ⁇ , ⁇ , ⁇ , ⁇ , or ⁇ , and the isotype of the antibody is defined as IgM, IgD, IgG, IgA, and IgE, respectively.
  • the IgG subclass is well known to those skilled in the art and includes but is not limited to IgG1, IgG2, IgG, and IgG4.
  • antibody includes: naturally occurring and non-naturally occurring Abs; monoclonal and polyclonal Abs; chimeric and humanized Abs; human or non-human Abs; fully synthetic Abs; and single-chain Abs.
  • Non-human Abs can be humanized by recombinant methods to reduce their immunogenicity in humans.
  • antibody fragment refers to an antigen-binding fragment of an antibody, that is, an antibody fragment that retains the ability of a full-length antibody to specifically bind to an antigen, such as retaining one or Fragments of multiple CDR regions.
  • antigen-binding fragments include, but are not limited to, Fab, Fab', F(ab')2 and Fv fragments; diabodies; linear antibodies; single-chain antibody molecules; nanobodies and multispecific antibodies formed from antibody fragments.
  • Chimeric antibody refers to an antibody and fragments thereof in which a part of the heavy chain and/or light chain is the same or homologous to the corresponding sequence in an antibody derived from a specific species (such as human) or belonging to a specific antibody class or subclass , And the rest of the chain is the same or homologous to the corresponding sequence in an antibody derived from another species (such as mouse) or belonging to another antibody class or subclass, as long as it exhibits the desired biological activity.
  • Human antibody refers to an antibody that contains only human immunoglobulin sequences. If the human antibody is produced in a mouse, mouse cell, or hybridoma derived from a mouse cell, it may contain murine carbohydrate chains. Similarly, “mouse antibody” or “rat antibody” refers to antibodies that only contain mouse or rat immunoglobulin sequences, respectively.
  • Humanized antibody refers to a form of antibody that contains sequences derived from non-human (such as murine) antibodies as well as human antibodies. Such antibodies contain the smallest sequence derived from a single side of a non-human immunoglobulin. Generally, a humanized antibody will comprise substantially all of at least one and usually two variable domains, wherein all or substantially all of the hypervariable loops correspond to the hypervariable loops of non-human immunoglobulins, and all or substantially all of the hypervariable loops The FR region is the FR region of human immunoglobulin. The humanized antibody optionally also includes at least a portion of an immunoglobulin constant region (Fc) (usually a human immunoglobulin constant region).
  • Fc immunoglobulin constant region
  • melanoma is a type of malignant tumor derived from melanocytes, commonly found in the skin, but also in mucous membranes, ocular choroids and other parts. Melanoma is the most malignant type of skin tumor, and it is prone to distant metastasis. Melanoma is divided into four subtypes, including acral type, mucosal type, skin type, and uncertain primary focus. The skin type is divided into long-term sunlight skin type (CSD) and non-long-term Positive exposure damage type (non-CSD), CSD and non-CSD are collectively referred to as daylight type.
  • CSD long-term sunlight skin type
  • non-CSD non-long-term Positive exposure damage type
  • acral and mucosal melanoma is the most important subtype of Asians. Carcinogenesis is not caused by DNA mutations caused by ultraviolet radiation. Compared with solar melanoma, acral and mucosal melanomas contain only a few DNA mutations. In the United States, 95% of melanomas are of the daylight type, while in Asia, especially China, more than 70% of melanomas are of the acral and mucosal type, of which more than 50% of the melanomas are of the mucosal type. Invest New Drugs, 34: 677-684; Cho, J. et al. disclosed that compared with solar melanoma, immunotherapy is less effective in treating acral and mucosal melanoma.
  • immunotherapy refers to the treatment of a subject who has a disease or is at risk of infection or suffers from recurrence of the disease by methods that include inducing, enhancing, suppressing or otherwise modifying the immune response.
  • Treatment or “therapy” of a subject refers to any type of intervention or process performed on the subject, or administration of an active agent to the subject, with the purpose of reversing, alleviating, ameliorating, alleviating or preventing symptoms and complications Or the onset, progression, severity or recurrence of the disease, or biochemical indicators related to the disease.
  • PD-1 Programmed death receptor-1
  • PD-1 is mainly expressed on previously activated T cells in the body, and binds to two ligands, PD-L1 and PD-L2.
  • the term "PD-1” as used herein includes human PD-1 (hPD-1), variants, isotypes and species homologs of hPD-1, and analogs that have at least one epitope in common with hPD-1 .
  • a “therapeutically effective dose” or “therapeutically effective dose” of a drug or therapeutic agent is any amount of a drug that protects the subject from the onset of disease or promotes the regression of the disease when used alone or in combination with another therapeutic agent.
  • the resolution of the disease is evidenced by a reduction in the severity of the symptoms of the disease, an increase in the frequency and duration of the asymptomatic period of the disease, or the prevention of injury or disability caused by the pain of the disease.
  • the ability of therapeutic agents 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 in vitro assays. The activity of the agent is determined in the.
  • a therapeutically effective amount of a drug includes a "prophylactically effective amount", that is, any amount that inhibits the development or recurrence of cancer when administered to a subject at risk of developing cancer or a subject suffering from recurrence of cancer, alone or in combination with an antitumor agent medicine.
  • Biotherapeutics refers to biomolecules, such as antibodies or fusion proteins, that block ligand/receptor signaling in any biological pathway that supports tumor maintenance and/or growth or inhibits anti-tumor immune responses.
  • CDR as used herein means that an immunoglobulin variable region is a complementarity determining region defined using the Kabat numbering system.
  • Therapeutic anti-PD-1 monoclonal antibody refers to an antibody that specifically binds to a mature form of a specific PD-1 expressed on the surface of certain mammalian cells. Mature PD-1 has no pre-secretory leader sequence, or leader peptide.
  • the terms “PD-1” and “mature PD-1” are used interchangeably herein, and unless clearly defined otherwise or clearly seen from the context, they should be understood as the same molecule.
  • 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.
  • framework region or "FR” refers to immunoglobulin variable regions that do not include CDR regions.
  • isolated antibody or antigen-binding fragment thereof refers to a purified state and in this case the specified molecule does not substantially contain other biological molecules, such as nucleic acids, proteins, lipids, carbohydrates or other materials (such as cell debris or growth Medium).
  • Patient refers to any single subject in need of medical treatment or participating in clinical trials, epidemiological studies or used as a control, usually mammals, including humans and other mammals, Such as horses, cows, dogs or cats.
  • the "RECIST 1.1 efficacy standard” mentioned herein refers to the definition of Eisenhauver et al., E.A. et al., Eur. J Cancer 45:228-247 (2009) for target damage or non-target damage based on the background of the measured response.
  • the RECIST working group reviewed the existing "RECIST v.1.1".
  • the "irRECIST standard” described in this article is proposed, which aims to better evaluate the efficacy of immunotherapy drugs.
  • ECOG scoring standard is an indicator of the patient's physical strength to understand its general health and tolerance to treatment.
  • ECOG physical status scoring standard score 0 points, 1 point, 2 points, 3 points, 4 points and 5 points.
  • a score of 0 means that the activity ability is completely normal, and there is no difference between the activity ability before the onset of the disease.
  • a score of 1 refers to the ability to walk around freely and engage in light physical activities, including general housework or office work, but not to engage in heavier physical activities.
  • sustained response refers to the sustained therapeutic effect after cessation of the therapeutic agent or combination therapy described herein.
  • the sustained response has a duration that is at least the same as the duration of the treatment or at least 1.5, 2.0, 2.5, or 3 times the duration of the treatment.
  • tissue section refers to a single part or piece of a tissue sample, such as a tissue slice cut from a sample of normal tissue or tumor.
  • Treatment of cancer refers to subjects suffering from cancer or having been diagnosed with cancer using the treatment regimens described herein (such as administration of anti-PD-1 antibodies or anti-PD-1 antibodies and CDK4/6 inhibitors).
  • Combination therapy to achieve at least one positive therapeutic effect (for example, a decrease in the number of cancer cells, a decrease in tumor volume, a decrease in the rate of cancer cell infiltration into surrounding organs, or a decrease in the rate of tumor metastasis or tumor growth).
  • the positive therapeutic effect in cancer can be measured in a variety of ways (see W.A. Weber, J. Nucl. Med., 50:1S-10S (2009)).
  • T/C ⁇ 42% is the minimum level of anti-tumor activity.
  • T/C (%) median value of treated tumor volume/median value of control tumor volume ⁇ 100.
  • the therapeutic effect achieved by the combination of the present invention is any one 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 that cancer does not grow, and includes the amount of time the patient experiences CR or PR and the amount of time the patient experiences SD.
  • DFS refers to the length of time that the patient remains disease-free during and after treatment.
  • OS refers to the increase in life expectancy compared to the initial or untreated individual or patient.
  • the response to the combination of the present invention is any one of PR, CR, PFS, DFS, OR, or OS, which is assessed using the RECIST 1.1 efficacy standard.
  • the treatment plan of the combination of the present invention that is effective in treating cancer patients may vary according to various factors, such as the patient's disease state, age, weight, and the ability of the therapy to stimulate the subject's anti-cancer response.
  • the embodiment of the present invention may not achieve an effective positive therapeutic effect in every subject, it should be effective and achieve a positive therapeutic effect in a statistically significant number of subjects.
  • mode of administration and “dosage regimen” are used interchangeably and refer to the dosage and time of each therapeutic agent in the combination of the present invention.
  • IHC Immunohistochemistry
  • PD-1 antibody refers to binding to PD-1 receptor, blocking the binding of PD-L1 expressed on cancer cells with PD-1 expressed on immune cells (T, B, NK cells) and preferably It can also block any chemical compound or biological molecule that binds PD-L2 expressed on cancer cells and PD-1 expressed on immune cells.
  • Alternative nouns or synonyms for PD-1 and its ligands include: for PD-1, there are PDCD1, PD1, CD279, and SLEB2; 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 CD273.
  • the PD-1 antibody blocks the binding of human PD-L1 and human PD-1, and preferably blocks both human PD-L1 and PD-L2 and human PD1 binding.
  • the amino acid sequence of human PD-1 can be found in NCBI locus number: NP_005009.
  • the amino acid sequences of human PD-L1 and PD-L2 can be found in NCBI locus numbers: NP_054862 and NP_079515, respectively.
  • anti-PD-1 antibody when referring to "anti-PD-1 antibody”, unless otherwise stated or described, the term includes antigen-binding fragments thereof.
  • the anti-PD-1 antibody suitable for any use, therapy, drug and kit of the present invention binds PD-1 with high specificity and high affinity, blocks the binding of PD-L1/2 and PD-1, and inhibits PD-1 signal transduction, so as to achieve the immunosuppressive effect.
  • the anti-PD-1 antibody includes the full-length antibody itself, as well as binding to the PD-1 receptor and behaves like a complete Ab in terms of inhibiting ligand binding and upregulating the immune system. Functional characteristic antigen-binding portion or fragment.
  • the anti-PD-1 antibody or antigen-binding fragment thereof is an anti-PD-1 antibody or antigen-binding fragment thereof that cross-competes with teriprizumab for binding to human PD-1.
  • the anti-PD-1 antibody or antigen-binding fragment thereof is a chimeric, humanized or human Ab or antigen-binding fragment thereof.
  • the Ab is a humanized Ab.
  • the anti-PD-1 antibodies used in any of the uses, therapies, drugs, and kits described in the present invention include monoclonal antibodies (mAb) or antigen-binding fragments thereof, which specifically bind to PD-1, And it preferably specifically binds to human PD-1.
  • the mAb can be a human antibody, a humanized antibody, or a chimeric antibody, and can include a human constant region.
  • the constant region is selected from the group consisting of human IgG1, IgG2, IgG3, and IgG4 constant regions; preferably, the anti-PD-1 antibody suitable for any use, therapy, drug, and kit of the present invention Or its antigen-binding fragment comprises a heavy chain constant region of human IgG1 or IgG4 isotype, more preferably a human IgG4 constant region.
  • the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody or antigen-binding fragment thereof includes the S228P mutation, which replaces the hinge region with a proline residue that is usually present at the corresponding position of an antibody of the IgG1 isotype Serine residues in.
  • the PD-1 antibody is a monoclonal antibody or an antigen-binding fragment thereof, and its light chain CDR is SEQ ID NO: 1, 2
  • the heavy chain CDRs are the amino acids shown in SEQ ID NO: 4, 5, and 6.
  • the PD-1 antibody specifically binds to human PD-1 and includes: (a) includes SEQ ID NO: 7 The variable region of the light chain shown, and (b) a monoclonal antibody comprising the variable region of the heavy chain shown in SEQ ID NO: 8.
  • the PD-1 antibody specifically binds to human PD-1 and includes: (a) includes SEQ ID NO: 9 The light chain shown, and (b) a monoclonal antibody comprising the heavy chain shown in SEQ ID NO: 10.
  • Table A below provides the amino acid sequence numbers of the light chain CDRs and heavy chain CDRs of exemplary anti-PD-1 antibody mAbs used in the uses, therapies, drugs, and kits of the present invention:
  • anti-PD-1 antibodies that bind to human PD-1 and can be used in the uses, therapies, drugs, and kits of the present invention are described in WO2014206107.
  • the human PD-1 mAb that can be used as an anti-PD-1 antibody in the uses, therapies, drugs, and kits of the present invention includes any of the anti-PD-1 antibodies described in WO2014206107, including: teriprizumab (Toripalimab) (a kind of light chain and heavy chain with the structure described in WHO Drug Information (Vol. 32, Issue 2, Pages 372-373 (2016)) and comprising the sequence SEQ ID NO: 9 and 10 A humanized IgG4 mAb with a chain amino acid sequence.
  • the anti-PD-1 antibody that can be used in any of the uses, therapies, drugs and kits of the present invention is selected from the humanized antibodies described in WO2014206107 38, 39, 41, and 48.
  • the anti-PD-1 antibody that can be used in any of the uses, therapies, drugs, and kits described in the present invention is teriprizumab.
  • Anti-PD-1 antibodies that can be used in any of the uses, therapies, drugs and kits described in the present invention also include Nivolumab and Pembrolizumab that have been approved by the FDA.
  • the anti-PD-1 antibody that can be used for any of the uses, therapies, drugs, and kits described in the present invention also includes those that specifically bind to PD-L1 to block the binding of PD-L1 to PD-1.
  • Anti-PD-L1 monoclonal antibodies such as nivolumab, pembrolizumab, toripalimab, Sintilimab, Camrelizumab, Tislelizumab, Cemiplimab.
  • PD-L1 or PD-L2 refers to any detectable expression level of a specific PD-L protein on the surface of a cell or a specific PD-L mRNA in a cell or tissue.
  • PD-L protein expression can be detected by diagnostic PD-L antibody in IHC analysis of tumor tissue sections or by flow cytometry.
  • the PD-L protein expression of tumor cells can be detected by PET imaging using a binding agent that specifically binds to the desired PD-L target (such as PD-L1 or PD-L2).
  • One method uses a simple binary endpoint where PD-L1 expression is positive or negative, where a positive result is defined by the percentage of tumor cells showing histological evidence of cell surface membrane staining. Counting tumor tissue sections as at least 1% of the total tumor cells is defined as positive for PD-L1 expression.
  • PD-L1 expression in tumor tissue sections is quantified in tumor cells and in infiltrating immune cells.
  • the percentages of tumor cells and infiltrating immune cells exhibiting membrane staining were individually quantified as ⁇ 1%, 1% to 50%, and subsequent 50% to 100%. For tumor cells, if the score is ⁇ 1%, the PD-L1 expression count is negative, and if the score is ⁇ 1%, it is positive.
  • 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 a comparison with the expression level of PD-L1 by an appropriate control.
  • the protein or mRNA expression level of the control PD-L1 can be a quantitative level in a non-malignant cell of the same type or in a section from a matched normal tissue.
  • CDK refers to cyclin-dependent kinases, which is a group of serine/threonine protein kinases. CDK drives the cell cycle through the phosphorylation of serine/threonine protein by cooperating with cyclin, which is an important factor in cell cycle regulation.
  • cyclin There are 8 types of CDK family, including CDK 1-8. Each CDK binds to different types of cyclin to form a complex, which regulates the process of cells transitioning from G1 phase to S phase or G2 phase to M phase and exiting M phase.
  • the FDA has approved the listing of CDK4/6 inhibitors, mainly Rebocinil, Pabocinil and Abemaciclib.
  • Rebocinil combined with aromatase inhibitors can be used as first-line drugs to treat HR-positive and HER2-negative postmenopausal women with advanced metastatic breast cancer;
  • pomazelin is mainly used for the treatment of hormone receptor (HR) positive and human epidermal growth factor receptor after receiving endocrine therapy 2(HER2)-negative adult patients with advanced or metastatic breast cancer.
  • CDK4/6 inhibitors in the clinical research stage, such as Milciclib, Trilaciclib, Lerociclib and Voruciclib.
  • CDK4/6 inhibitors include but are not limited to pabocinil.
  • the combined use includes but is not limited to Toripalimab and Pabocinil.
  • the present invention relates to the treatment of cancer.
  • the present invention includes administering anti-PD-1 antibodies or antigen-binding fragments thereof to patients in need alone; or including administering anti-PD-1 antibodies in combination with other anticancer agents to patients in need.
  • cancer refers to a wide range 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, which invade adjacent tissues and can also metastasize to remote parts of the body through the lymphatic system or bloodstream. Examples of cancers that are suitable for the treatment or prevention of the methods, drugs, and kits of the present invention include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma, and sarcoma.
  • cancer More specific examples include squamous cell carcinoma, myeloma, small cell lung cancer, non-small cell lung cancer, 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, stomach cancer, bladder cancer, hepatocytoma, breast cancer, colon cancer and head and neck cancer.
  • the cancer patient suitable for the present invention is a cancer patient whose PD-L1 expression is positive, or a tumor mutation burden (TMB) ⁇ 3.6Muts/Mb in peripheral blood or tumor tissue, or a PD-L1 expression is positive at the same time Tumor mutation burden (TMB) in peripheral blood or tumor tissue is ⁇ 3.6Muts/Mb.
  • cancer patients suitable for treatment by the method of the present invention are preferably cancer patients whose tumor tissues have detected BRAF gene mutations.
  • tumor mutation burden refers to the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases.
  • tumor mutational burden is estimated by analyzing somatic mutations (including coding base substitutions and macrobase insertions of the studied panel sequence).
  • TMB tumor mutation burden
  • 3.6Muts/Mb when the tumor mutation burden (TMB) of a subject is greater than or equal to 3.6Muts/Mb, it indicates that such subjects will be administered anti-PD-1 antibody alone or anti-PD-1 in combination with other anti-cancer agents. 1 Antibody can achieve better curative effect than TMB ⁇ 3.6Muts/Mb.
  • the method for treating cancer of the present invention includes administering a therapeutically effective amount of anti-PD-1 antibody, or anti-PD-1 antibody and CDK4/6 inhibitor to an individual in need.
  • the anti-PD-1 antibody may be as described in any of the embodiments herein, more preferably, the light chain CDR is the amino acid shown in SEQ ID NO: 1, 2 and 3, and the heavy chain CDR is SEQ ID NO: 4, 5 and The antibody of amino acid shown in 6, more preferably a monoclonal antibody comprising the light chain variable region shown in SEQ ID NO: 7 and the heavy chain variable region shown in SEQ ID NO: 8, and more preferably comprising SEQ ID NO
  • the monoclonal antibody of the light chain shown in: 9 and the heavy chain shown in SEQ ID NO: 10 more preferably the humanized antibodies 38, 39, 41, and 48 described in WO2014206107, and most preferably teripril anti.
  • the CDK4/6 inhibitor is Pabocinil Milciclib, Trilaciclib, Lerociclib and Voruciclib, preferably Pabocinil.
  • the present invention uses a combination of Teriprizumab and Pabocinil to treat cancer.
  • the present invention provides a method for treating melanoma, the method comprising administering to a melanoma patient a therapeutically effective amount of teriprizumab; preferably, the patient is positive for PD-L1 expression, or peripheral Tumor burden (TMB) in blood or tumor tissue is ⁇ 3.6Muts/Mb, or PD-L1 expression is positive, and tumor burden (TMB) in peripheral blood or tumor tissue is ⁇ 3.6Muts/Mb.
  • the melanoma patient is preferably a melanoma patient whose BRAF gene mutation is detected in tumor tissue, and more preferably non-acral melanoma or primary melanoma.
  • the melanoma is acral type or/and mucosal type melanoma.
  • the present invention provides a method of treating melanoma, the method comprising administering to a melanoma patient a therapeutically effective amount of teriprizumab and pabocinil; preferably, the patient's peripheral There is CDK4 or CCND1 gene amplification in blood or tumor tissue.
  • each therapeutic agent when two or more therapeutic agents are administered (ie, "combination therapy"), each therapeutic agent can be administered alone or in a pharmaceutical composition comprising the therapeutic agent and one Or multiple pharmaceutically acceptable carriers, excipients and diluents.
  • Each of the therapeutic agents in the combination therapy of the present invention can be administered simultaneously, concurrently, or sequentially in any order.
  • the therapeutic agents in combination therapy are administered in different dosage forms, such as one drug is a tablet or capsule and the other drug is a sterile liquid, and/or administered at different administration times, such as chemotherapeutic agents are administered at least daily and
  • the biotherapeutics are administered infrequently, such as once every week, or every two weeks or every three weeks.
  • the CDK4/6 inhibitor is administered before the administration of the anti-PD-1 antibody, while in other embodiments, the CDK4/6 inhibitor is administered after the administration of the anti-PD-1 antibody.
  • At least one of the therapeutic agents in the combination therapy is administered using the same dosing schedule (dose, frequency, duration of treatment).
  • Each small molecule therapeutic agent in the combination therapy of the present invention can be administered orally or parenterally (such as intravenous, intramuscular, intraperitoneal, subcutaneous, rectal, topical or transdermal administration route).
  • the combination therapy of the present invention can be administered before or after surgery, and can be administered before, during or after radiotherapy.
  • the combination therapy described in the present invention is administered to patients who have not been previously treated with biotherapeutics (usually referred to as anti-PD-1 antibodies) or chemotherapeutic agents (usually referred to as CDK4/6 inhibitors).
  • biotherapeutics usually referred to as anti-PD-1 antibodies
  • chemotherapeutic agents usually referred to as CDK4/6 inhibitors.
  • the combination therapy is administered to patients who cannot achieve a sustained response after treatment with a biotherapeutic agent or chemotherapeutic agent.
  • the combination therapy of the present invention can be used to treat tumors discovered by palpation or by imaging techniques known in the prior art, such as MRI, ultrasound or CAT scan.
  • the combination therapy of the present invention is preferentially administered to cancer patients whose PD-L1 expression is tested positive or TMB ⁇ 3.6Muts/Mb.
  • the choice of the dosing regimen of the combination therapy of the present invention depends on several factors, including but not limited to serum or tissue conversion rate, degree of symptoms, immunogenicity, and accessibility of target cells, tissues, and organs of the individual to be treated.
  • the dosage regimen should match the acceptable degree of side effects to deliver the maximum amount of various therapeutic agents to the patient. Therefore, the dosage and frequency of administration of each biotherapeutic agent and chemotherapeutic agent in the combination therapy depend on the specific therapeutic agent, the severity of the cancer being treated, and the characteristics of the patient.
  • the anti-PD-1 antibody and CDK4/6 inhibitor of the present invention can be provided as a kit including a first container, a second container and a package insert.
  • the first container contains at least one dose of the anti-PD-1 antibody-containing drug
  • the second container contains at least one dose of the CDK4/6 inhibitor
  • the package insert or label contains instructions for using the drug to treat cancer.
  • the kit may further include other materials that can be used to administer the drug, such as diluents, filter paper, IV bags and threads, needles and syringes.
  • the instructions may state that the drug is intended for the treatment of cancer patients, and its PD-L1 expression is tested positive by immunohistochemical staining (IHC) analysis.
  • IHC immunohistochemical staining
  • the therapeutic agent of the present invention can constitute a pharmaceutical composition, such as a pharmaceutical composition containing the anti-PD-1 antibody described herein or/and other anti-cancer agents other than the anti-PD-1 antibody and other pharmaceutically acceptable carriers .
  • pharmaceutically acceptable carrier includes any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, etc. that are physiologically compatible.
  • the carrier suitable for the composition containing the anti-PD-1 antibody is suitable for intravenous, intramuscular, subcutaneous, parenteral, spinal or epidermal administration, such as by injection or infusion, for containing other anticancer agents
  • the carrier of the composition is suitable for parenteral administration, such as oral administration.
  • the pharmaceutical composition of the present invention may contain one or more pharmaceutically acceptable salts, antioxidants, water, non-aqueous carriers, and/or adjuvants such as preservatives, wetting agents, emulsifying agents and dispersing agents.
  • the dosage range can be about 0.01 to about 20 mg/kg, about 0.1 to about 10 mg/kg of the individual’s body weight, or 120 mg, 240 mg, 360 mg, 480 mg fixed dose.
  • the dosage may be about 0.1, about 0.3, about 1, about 2, about 3, about 5, or about 10 mg/kg of the individual's body weight.
  • Dosing regimens are usually designed to achieve such exposures that result in sustained receptor occupancy (RO) based on the typical pharmacokinetic properties of Ab.
  • RO sustained receptor occupancy
  • a representative dosing regimen may be about once a week, about once every two weeks, about once every three weeks, about once every four weeks, about once a month, or more once.
  • the anti-PD-1 antibody is administered to the individual about once every two weeks.
  • the administration schedule of other anticancer agents varies for different drugs.
  • the administration schedule of the CDK inhibitor varies for different subtypes.
  • the present invention also includes an anti-PD-1 antibody or its combination with a CDK4/6 inhibitor for treating cancer patients, and an anti-PD-1 antibody or its combination with a CDK4/6 inhibitor in the preparation of a medicament for the treatment of cancer patients Applications.
  • the cancer patient or the cancer replaced may be as described in any of the foregoing embodiments; preferably, the cancer patient is a cancer patient with positive PD-L1 expression, or a tumor mutation burden in peripheral blood or tumor tissue ( TMB) ⁇ 3.6Muts/Mb, or PD-L1 expression is positive and tumor mutation burden (TMB) ⁇ 3.6Muts/Mb in peripheral blood or tumor tissue.
  • TMB peripheral blood or tumor tissue
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation burden
  • TMB tumor mutation
  • the melanoma patient is positive for PD-L1 expression, or the tumor burden (TMB) in the peripheral blood or tumor tissue is ⁇ 3.6Muts/Mb, or the PD-L1 expression is positive and the tumor burden in the peripheral blood or tumor tissue is (TMB) ⁇ 3.6Muts/Mb.
  • the melanoma patient is more preferably non-acral melanoma or melanoma of indeterminate primary focus.
  • the melanoma patient is a acral melanoma patient or a mucosal melanoma patient.
  • the preferred anti-PD-1 antibody for the treatment of cancer patients may be as described in any of the embodiments herein, more preferably, the light chain CDR is the amino acid shown in SEQ ID NO: 1, 2 and 3, and the heavy chain CDR is SEQ ID NO: antibodies with amino acids shown in 4, 5, and 6, more preferably monoclonal antibodies comprising the light chain variable region shown in SEQ ID NO: 7 and the heavy chain variable region shown in SEQ ID NO: 8, more preferably Preferably, it is a monoclonal antibody comprising the light chain shown in SEQ ID NO: 9 and the heavy chain shown in SEQ ID NO: 10, more preferably humanized antibodies 38, 39, 41, and 48 described in WO2014206107, most preferably It is tereprizumab.
  • the CDK4/6 inhibitor is preferably pabocinil.
  • the present invention relates to teriprizumab or a combination of teriprizumab and pabocinil for the treatment of cancer patients. More specifically, the present invention relates to a combination of teriprizumab or teriprizumab and pabocinil for the treatment of patients with melanoma; more preferably, the expression of PD-L1 in the patients with melanoma is Positive, or tumor burden (TMB) ⁇ 3.6Muts/Mb in peripheral blood or tumor tissue, or positive expression of PD-L1, and tumor burden (TMB) ⁇ 3.6Muts/Mb in peripheral blood or tumor tissue, or tumor tissue
  • TMB tumor burden
  • TMB tumor burden
  • Anti-PD-1 antibody predicts the effect of cancer treatment
  • This article can predict anti-PD by performing whole-exome sequencing on tumor biopsy samples from patients or corresponding peripheral blood samples (such as using second-generation sequencing technology) to identify whether certain genes have mutations or gene amplifications.
  • -1 antibody, CDK4/6 inhibitor or a combination of the two for the efficacy of anti-tumor therapy.
  • Gene mutation or “gene alteration” as used herein includes gene truncation, gene rearrangement/fusion, gene amplification, gene deletion and gene substitution/insertion, etc.
  • gene amplification refers to a process in which the copy number of a gene encoding a specific protein is selectively increased while other genes are not increased proportionally.
  • gene amplification is achieved by removing the repetitive sequence of the gene from the chromosome and then performing extrachromosomal replication in a plasmid, or by transcribing all the repetitive sequences of ribosomal RNA to generate RNA transcripts and then transcribing them to generate additional copies of the original DNA molecule. of.
  • the subjects of the present invention have certain unique gene mutations, for example, some subjects have CDK4 or CDDN 1 gene amplification; some subjects have BRAF gene mutations, especially Patients with non-acral melanoma or melanoma with uncertain primary foci have BRAF gene mutations; some subjects with mucosal melanoma have NF1 gene mutations; some subjects (including acral melanoma, mucosal, and mucosal melanoma) Non-acral (skin type) and primary unknown melanoma patients have NRAS gene mutations.
  • the BRAF gene mutation is selected from gene rearrangement/fusion and gene substitution/insertion.
  • the NRAS gene mutation is a gene substitution/insertion.
  • having a BRAF gene mutation usually indicates that the patient has a better therapeutic effect with the anti-PD-1 antibody of the present invention.
  • having CDK4 or CCND1 gene amplification indicates that the patient uses the anti-PD-1 antibody of the present invention in combination with a CDK4/6 inhibitor or a CDK4/6 inhibitor alone for treatment efficacy. good.
  • the presence of NRAS gene mutation indicates that the patient has unsatisfactory therapeutic effects using the anti-PD-1 antibody of the present invention alone.
  • the present invention provides a method for predicting the effect of using the anti-PD-1 antibody of the present invention, especially tereprizumab, on the treatment of cancer in an individual, including the detection of biomarkers in the patient’s peripheral blood before treatment
  • the biomarker is selected from but not limited to BRAF, NRAS, CDK4 or CCND1.
  • BRAF gene mutations indicates that the individual is suitable for treatment with anti-PD-1 antibodies
  • the presence of NRAS gene mutations indicates that the individual is not suitable for treatment with anti-PD-1 antibodies
  • the presence of CDK4 or CCND1 gene amplification indicates that the individual is suitable for treatment with anti-PD-1 antibodies.
  • the cancer is melanoma.
  • the present invention also includes a method for predicting the effect of anti-PD-1 antibody therapy in tumor patients by using BRAF gene or NRAS gene.
  • BRAF gene mutations indicates that the tumor patients are suitable for treatment with anti-PD-1 antibodies.
  • NRAS gene mutations indicates that the tumor patients are not suitable for treatment with anti-PD-1 antibodies.
  • the present invention also provides detection reagents for biomarkers (especially BRAF gene, and/or NRAS gene, and/or CDK4 gene, and/or CCND1 gene) for preparing predictive anti-PD-1 antibodies and / Or use in a kit for the effect of a CDK4/6 inhibitor on cancer.
  • biomarkers especially BRAF gene, and/or NRAS gene, and/or CDK4 gene, and/or CCND1 gene
  • Such reagents include, but are not limited to, reagents commonly used in tests, including but not limited to primers, probes, and reagents required for PCR.
  • the cancer is preferably melanoma.
  • the prediction includes: the presence of a BRAF gene mutation indicates that the individual is suitable for treatment with an anti-PD-1 antibody, the presence of a NRAS gene mutation indicates that the individual is not suitable for treatment with an anti-PD-1 antibody, and the presence of CDK4 or CCND1 gene
  • the expansion indicates that the individual is suitable for treatment with anti-PD-1 antibodies and CDK4/6 inhibitors or suitable for treatment with CDK4/6 inhibitors alone.
  • Example 1 Clinical study of anti-PD-1 antibody alone in the treatment of melanoma
  • Eligible subjects must (1) be at least 18 years old, (2) have locally advanced or metastatic melanoma, (3) be difficult to cure after standard systemic treatment, (4) ECOG score 0 or 1, (5) no history of autoimmune disease or persistent infection, (6) no anti-PD-1/or anti-PD-L1 immunotherapy before.
  • Subjects must have evaluable lesions according to the RECIST v 1.1 standard, and are not allowed to use anti-tumor drugs at the same time, not to be treated with systemic steroids, or have not been treated with anti-CTLA4, anti-PD-1, or anti-PD-L1 antibodies.
  • tested drug anti-PD-1 antibody teriprizumab (WO2014206107).
  • the dose of anti-PD-1 antibody used in this test is: 3mg/kg, intravenously injected once every two weeks (Q2W).
  • Table 1 Demographic statistics of the enrolled subjects
  • PD-L1 positive is defined as the expression of PD-L1 on tumor cells stained with SP142IHC as ⁇ 1%.
  • the average number of teriprizumab received by the patient was 10 doses (range: 1 to 73 doses).
  • 116 of 128 patients experienced treatment-related side effects. Most of them are level 1 or 2.
  • Common (>5%) treatment-related side effects are shown in Table 2.
  • Treatment-related SAEs occurred in 10 (7.8%) patients.
  • Twenty-five patients (19.5%) experienced grade 3 or higher TRAE, including 13 (10.2%) grade 3 and 12 (9.4%) grade 4 side effects. No patient experienced grade 5 side effects.
  • the TRAE of special concern was less than 5%, including 5 cases (3.9%) of vitiligo, 3 cases (2.3%) of liver injury (2 cases of grade 4), 2 cases (1.6%) of acute pancreatitis, and 2 cases of (1.6%).
  • Qualitative lung disease (1 case of grade 3), 2 cases (1.6%) with adrenal insufficiency, 2 cases (1.6%) with hypopituitarism, and 1 case (0.8%) with uveitis (grade 3).
  • ORR (CR+PR)/total ⁇ 100%
  • DCR (CR+PR+uPR+SD)/total ⁇ 100%
  • CR complete response
  • PR partial response
  • uPR unconfirmed PR
  • SD stable disease
  • PD disease progression
  • NE not assessed
  • ORR objective response rate
  • DCR disease control rate
  • CI confidence interval.
  • the target lesion size of 49 subjects decreased from baseline, as shown in Figure 2(A).
  • the median response time was 3.5 months (95%CI 1.7-3.6), the median sustained response (DOR) was not reached, and only 9 of 22 patients had disease progression after the initial response, as shown in Figure 2(B) and Figure 2(C).
  • the DCR per RECISTv1.1 is 57.5% (95% CI 48.4-66.2), and the DCR per irRECIST is 59.8% (95% CI 50.8-68.4).
  • the median PFS per RECISTv1.1 is 3.6 months (95% CI 3.7-5.5), and the median PFS per irRECIST is 3.7 months (95% CI 3.3-9.1), as shown in Figure 3(A).
  • the steady-state average trough plasma concentration of teriprizumab is 39.8 ⁇ g/mL (range 4.9-92.4 ⁇ g/mL), which is much higher than the full PD-1 blocking concentration of 1.5u g/ml (10nmol/L).
  • 128 patients were tested for anti-drug antibody (ADA).
  • ADA anti-drug antibody
  • ADA anti-drug antibody
  • PD-L1 positive status is defined as the presence of membrane staining intensity of tumor cells ⁇ 1%.
  • PD-L1 + patients also had a significant survival advantage over PD-L1- patients in progression-free survival PFS and overall survival OS.
  • TMB Tumor mutation burden
  • 26 PD-L1 positive patients only 7 had TMB ⁇ 3.6Muts/Mb( Figure 6, A).
  • Table 5 The highest 20% TMB value in each subtype is used as the cut-off value for the efficacy analysis
  • Example 1 the second-generation sequencing technology was used to perform whole-exome sequencing on the peripheral blood samples from patients undergoing tumor biopsy and pairing, and 19,278 gene mutations were identified from 98 patients, including 7964 There are three missense mutations, 509 gene deletions, 482 rearrangements, 288 alternative splicing sites, 129 frameshift truncations, and 8157 gene amplifications.
  • the most frequently changed genes ( ⁇ 10%) are BRAF (33%), TERT (32%), CDKN2A (12%), NRAS (16%) ), CDK4 (12%), APOB (11%), CCND1 (11%), AGAP2 (11%), NF1 (10%), LRP1B (10%), MDM2 (10%) and KIT (10%), See Figure 7 for details.
  • CDK4 or CCND1 (Cyclin D1) amplification was observed in 33% (13/39) acral type and 20% (3/15) mucosal type, but only in 9% (2/23) non-acral type It was found in skin type and 10% (2/21) of primary unknown melanoma.
  • ORR 0% The study showed that for patients with CCND1 amplification, CDK4/6 targeted therapy or CDK4/6 inhibitors were used with Anti-PD-1 antibody combination therapy has good potential.
  • Example 4 Analysis of messenger RNA expression profile in tumor biopsy
  • RNA sequencing and expression profile analysis were performed on mRNA extracted from tumor biopsy. Effective results were obtained from 46 patients.
  • IFN- ⁇ -related genomes IDO1, CXCL10, CXCL9, HLA-DRA, STAT1, IFNG
  • inflammation-related genomes IL-6, CXCL1, CXCL2, CXCL3, CXCL8, PTGS2
  • angiogenesis-related genomes VEGFA, KDR, ESM1, PECAM1, ANGPTL4, CD34

Abstract

本发明公开了抗PD-1抗体在治疗黑色素瘤中的用途。本发明还披露了用于检测BRAF、NRAS、CDK4/CCND1基因突变的试剂在预测黑色素瘤病人对单独施用抗PD-1抗体和/或其抗原结合片段治疗的效果的检测试剂盒中的用途。

Description

抗PD-1抗体在***中的用途 技术领域
本发明涉及抗PD-1抗体在***中的用途。具体而言,本发明涉及抗PD-1抗体在治疗黑色素瘤中的用途;以及抗PD-1抗体在制备治疗黑色素瘤的药物中的用途;以及利用生物标记物预测单独施用抗PD-1抗体在治疗黑色素瘤中的疗效的方法。
背景技术
免疫逃逸是癌症的特征之一。Ahmadzadeh,M.等,Blood,114:1537-44中公开了肿瘤特异性T淋巴细胞常存在于肿瘤微环境、引流***和外周血中,但由于肿瘤微环境中存在的免疫抑制机制网络,其通常无法控制肿瘤的进展。CD8 +肿瘤浸润T淋巴细胞(TIL)通常表达活化诱导的抑制受体,包括CTLA-4和PD-1,而肿瘤细胞经常表达免疫抑制配体,包括PD-1配体1(PD-L1,也叫B7-H1或CD274),该配体抑制T细胞激活和效应功能。在抑制机制中,PD-1及其配体已成为肿瘤细胞利用其抑制肿瘤微环境中激活的T细胞的重要途径。
程序性死亡受体1(PD-1)在免疫调节及周边耐受性维持中起重要作用。PD-1主要在激活的T细胞和B细胞中表达,功能是抑制淋巴细胞的激活,这是免疫***的一种正常的防治免疫过激的外周组织耐受机制。但是,在肿瘤微环境中浸润的活化T细胞高表达PD-1分子,活化白细胞分泌的炎症因子会诱导肿瘤细胞高表达PD-1的配体PD-L1和PD-L2,导致肿瘤微环境中活化T细胞PD-1通路持续激活,T细胞功能被抑制,无法杀伤肿瘤细胞。治疗型PD-1抗体可以阻断这一通路,部分恢复T细胞的功能,使活化T细胞能够继续杀伤肿瘤细胞。
近十年来,PD-1/PD-L1通路阻断已被证明是在各种癌症适应症中诱导持久抗肿瘤应答的有效途径。阻断PD-1/PD-L1通路的单克隆抗体(mAbs)可以增强肿瘤特异性T细胞的活化和效应功能,减轻肿瘤负荷,提高生存率。2014年至2017年之间,FDA已经批准了2款抗-PD1单克隆抗体(nivolumab和pembrolizumab)和3款抗-PD-L1单克隆抗体(atezolizumab、avelumab和durvalumab)用于治疗人类肿瘤。黑色素瘤是Nivolumab和Pembrolizumab在2014首先获批的适应症。
黑色素瘤长期以来被认为是一种免疫原性较强的癌种,因为经常观察到淋巴细胞浸润到肿瘤中,以及对高剂量IL-2免疫治疗的临床反应。从机制上讲,在西方人群中,诱导 DNA损伤相关的慢性紫外线辐射暴露是导致黑色素瘤的主要原因。慢性紫外线辐射晒伤(CSD)黑色素瘤占美国和其他西方国家皮肤黑色素瘤的95%。相比之下,肢端雀斑样黑色素瘤(ALM)(~50%)和黏膜黑色素瘤(MM)(~20%)则是在亚洲人群中最常见的两个黑色素瘤亚群,这个发现由Chi,Z.等,BMC Cancer(2011),11:85中公开。Furney,S.J.等,Pigment Cell Melanoma Res(2014),27:835-8中公开了ALM和MM都与慢性紫外线暴露无关,携带更少的DNA突变。Cho,J.等,Invest New Drugs(2016),34:677-84的回顾性研究表明,与CSD黑色素瘤相比,免疫疗法治疗ALM和MM的效果较差。因此,免疫疗法,比如抗PD-1抗体在治疗黑色素瘤,尤其是在治疗亚洲人群中常发的粘膜型和肢端型黑色素瘤中的疗效存在不确定性,以及怎样进一步提高其治疗效果,是本领域急需解决的技术问题。并且,虽然免疫检查点抑制剂疗法在过去的十年间对于转移性黑色素瘤的治疗取得了较大的改进,但是,大多数还是几种在白种人的皮肤型(也叫非肢端型)黑色素瘤上,对于亚洲以及非洲人种的研究非常有限,导致在这两种人种的非肢端以及原发型黑色素瘤的治疗尚有很大的空缺,亟待解决。
发明内容
本发明一方面提供一种治疗黑色素瘤患者的方法,其包含向所述患者施用治疗有效量的抗PD1-1抗体。
本发明第二方面提供一种抗PD-1抗体在制备用于治疗黑色素瘤患者的药物中的用途。
作为一个或多个实施方式,所述的黑色素瘤为晚期或转移性黑色素瘤。
作为一个或多个实施方式,所述的黑色素瘤患者具有BRAF突变。
作为一个或多个实施方式,所述的黑色素瘤患者选自肢端型、黏膜型、非肢端皮肤型和原发灶不确定型黑色素瘤患者。
作为一个或多个实施方式,所述的黑色素瘤患者为非肢端黑色素瘤患者。
作为一个或多个实施方式,所述的黑色素瘤患者为原发灶不确定型黑色素瘤患者。
作为一个或多个实施方式,所述的黑色素瘤患者的癌症组织或切片的PD-L1表达经测试呈阳性。在一个或多个实施方式中,所述的黑色素瘤患者的癌症的活检样本或配对的外周血样本的肿瘤突变(TMB)≥3.6Muts/Mb。在一个或多个实施方式中,所述的黑色素瘤患者的癌症的组织或切片的PD-L1表达经测试呈阳性且癌症的活检样本或配对的外周血样本的肿瘤突变(TMB)≥3.6Muts/Mb。
本发明第三方面提供一种治疗黑色素瘤患者的方法,其包含向所述患者单独施用治疗有效量的CDK4/6抑制剂,或联合施用治疗有效量的CDK4/6抑制剂和抗PD-1抗体。
作为一个或多个实施方式,所述的黑色素瘤患者具有CDK4或CCDN1基因扩增。
本发明第四方面提供了一种用于预测患有肿瘤的个体施用抗PD-1抗体的***的效果的试剂盒,所述试剂盒包括:(a)用于检测个体肿瘤组织或外周血中BRAF突变的试剂;和(b)使用(a)所述的试剂检测个体施用抗PD-1抗体治疗效果的说明书。
本发明第五方面提供了一种用于预测患有肿瘤的个体施用抗PD-1抗体***的效果的试剂盒,所述试剂盒包括:(a)用于检测个体肿瘤组织或外周血中CDK4/CCND1基因突变或扩增的试剂;和(b)使用(a)所述的试剂检测个体单独施用CDK4/6抑制剂或与抗PD-1抗体联合治疗效果的说明书。
本发明第六方面提供一种用于预测患有肿瘤的个体施用抗PD-1抗体***的效果的试剂盒,所述试剂盒包括:(a)用于检测个体肿瘤组织或外周血中NRAS基因突变的试剂;和(b)使用(a)所述的试剂检测个体施用抗PD-1抗体治疗效果的说明书。
本发明所述的用途、方法和试剂盒中,所述抗PD-1抗体是单克隆抗体或其抗原结合片段。在某些实施方案中,所述抗PD-1抗体能特异性结合PD-1,阻断PD-L1或PD-L2与PD-1结合。在某些实施方案中,所述抗PD-1抗体特异性结合PD-L1或/和PD-L2,阻断PD-L1和/或PD-L2与PD-1的结合。
在一个或多个实施方式中,抗PD-1抗体是包含至少一个互补决定区(CDR)的抗体,其互补决定区(CDR)的氨基酸序列选自以下:SEQ ID NO:1、2、3、4、5或6。
在一个或多个实施方式中,抗PD-1抗体是包含互补决定区(CDR)的抗体,其轻链互补决定区(LCDR)的氨基酸序列如SEQ ID NO:1、2和3所示,重链互补决定区(HCDR)的氨基酸序列如SEQ ID NO:4、5和6所示。
在一个或多个实施方式中,抗PD-1抗体包含轻链可变区(VL)和重链可变区(VH),其中,VL的氨基酸序列如SEQ ID NO:7所示,VH的氨基酸序列如SEQ ID NO:8所示。
在一个或多个实施方式中,抗PD-1抗体是包含轻链和重链的抗PD-1抗体,且轻链包含SEQ ID NO:9所示的氨基酸序列,重链包含SEQ ID NO:10所示的氨基酸序列。
在一个或多个实施方式中,抗PD-1抗体选自nivolumab、pembrolizumab、toripalimab、Sintilimab、Camrelizumab、Tislelizumab、Cemiplimab中的一种或几种。
在一个或多个实施方式中,抗PD-1抗体或其抗原结合片段的单次施用剂量为约0.1mg/kg至约10.0mg/kg个体体重,例如约0.1mg/kg、约0.3mg/kg、约1mg/kg、约2mg/kg、约3mg/kg、约5mg/kg或10mg/kg个体体重,或选自约120mg至约480mg固定剂量,例如约120mg、240mg、360mg或480mg固定剂量。
在一个或多个实施方式中,所述抗PD-1抗体或其抗原结合片段的给药周期频率为约每一周一次,每两周一次、每三周一次、每四周一次或一个月一次,优选为每两周一次。
在一个或多个实施方式中,抗PD-1抗体或其抗原结合片段的单次施用剂量为1mg/kg个体体重、3mg/kg个体体重、10mg/kg个体体重、或240mg固定剂量、480mg固定剂量,以每两周一次施用。
在一个或多个实施方式中,抗PD-1抗体或其抗原结合片段以液体剂型例如注射剂,经胃肠外途径例如经静脉输注施用。
在一个或多个实施方式中,抗PD-1抗体或其抗原结合片段的给药周期可以为一周、二周、三周、一个月、两个月、三个月、四个月、五个月、半年或更长时间,任选地,每个给药周期的时间可以相同或不同,且每个给药周期之间的间隔可以相同或不同。
在一个或多个实施方式中,CDK4/6抑制剂选自帕博西尼、瑞博西尼和玻玛西林。
本发明所述的用途、方法和试剂盒中,个体是人。
本发明所述的用途、方法和试剂盒在另一个或多个实施方式中,对象患有黑色素瘤且既往未接受过抗PD-1或抗-PD-L1免疫治疗。在一个优选的实施方式中,所述的癌症是晚期黑色素瘤。在一个优选的实施方式中,所述的癌症是转移性黑色素瘤。
本发明第七方面提供一种用于预测肿瘤病人对抗PD-1抗体治疗效果的方法,其包括在治疗前检测患者肿瘤组织或外周血中的生物标记物,其中,所述的生物标记物为BRAF基因突变,其中,存在所述BRAF基因突变表明所述肿瘤病人适于用抗PD-1抗体进行治疗。
本发明第八方面提供一种预测肿瘤病人对抗PD-1抗体治疗的效果的方法,其包括检测患者肿瘤组织中的生物标记物,所述的生物标记物为CDK4或CCND1基因扩增。在一个或多个实施方式中,存在所述CDK4或CCND1基因扩增表明所述肿瘤病人适于用抗PD-1抗体和CDK4/6抑制剂联合进行治疗。在另一个或多个实施方式中,存在所述CDK4或CCND1基因扩增表明所述肿瘤病人适于用CDK4/6抑制剂进行治疗。
本发明的第九方面提供一种预测肿瘤病人对抗PD-1抗体治疗效果的方法,包括在治疗前检测患者肿瘤组织或外周血中的生物标记物,所述的生物标记物为NRAS基因突变。在一个或多个实施方式中,存在所述NRAS基因突变表明单独施用抗PD-1抗体进行治疗效果不理想或不适于单独施用抗PD-1抗体进行治疗。
本发明所述的用途、方法和试剂盒中,所述的肿瘤为实体瘤。在一个或多个实施方式中,所述的肿瘤为黑色素瘤。
附图说明
图1:toripalimab治疗局部晚期或转移性黑素瘤的标准治疗的临床II期研究的关系图。
图2:按照RECIST v1.1对临床响应进行评估。A:至少一项治疗后影像学评估的患 者(n=119)相对于基线的最大肿瘤大小变化。条的长度表示靶病变的最大减少或最小增加;B:从基线开始随着时间的推移个体肿瘤负荷的变化(n=119);C:已确认应答者的暴露程度和反应持续时间(n=22)。
图3:本研究中黑色素瘤患者的无进展生存期PFS(A)和本研究中黑色素瘤患者的总生存数OS(B)。
图4:本研究中应答亚组的总生存数OS(A)和黑色素瘤亚组的总生存数OS(B)。
图5:本研究中黑色素瘤亚型的无进展生存期PFS。
图6:临床响应与肿瘤PD-L1表达和TMB之间的关系。A:PD-L1阳性定义为采用SP142IHC染色肿瘤细胞或免疫细胞的膜染色的任何强度≥1%;TMB通过对编码区域内的体细胞突变进行全外显子测序来计算;3.6Muts/Mb作为截断值;B:PD-L1 +或TMB≥3.6Muts/Mb在黑色素瘤亚组中的百分比;C:PD-L1 +与PD-L1 -的患者的无进展生存期PFS;D:PD-L1 +与PD-L1 -的患者的总体生存数OS;E:TMB≥3.6Muts/Mb和TMB<3.6Muts/Mb的患者的无进展生存期PFS;F:TMB≥3.6Muts/Mb和TMB<3.6Muts/Mb的患者的总体生存数OS。
图7:通过整体外显子组测序(WES)98位患者的基因变异和频率。
图8:临床响应和IFN-γ相关基因组、炎症相关基因组和血管生成相关基因组的签名得分之间的关系。
具体实施方式
本发明涉及肿瘤治疗方法。本发明的方法包括向有需要的患者单独施用抗PD-1抗体或其抗原结合片段;或包括向有需要的患者施用与其他抗癌剂组合的抗PD-1抗体。本发明还涉及利用生物标记物预测抗PD-1抗体在治疗癌症,尤其是黑色素瘤患者中的疗效的方法。
术语
为了更易于理解本发明,下文具体定义某些科技术语。除非本文中别处另有明确说明,否则本文所用的科技术语皆具有本发明所属技术领域的普通技术人员通常所了解的含义。
“施用”、“给与”及“处理”是指采用本领域技术人员已知的各种方法或递送***中的任意一种将包含治疗剂的组合物引入受试者。抗PD-1抗体的给药途径包括静脉内、肌内、皮下、腹膜、脊髓或其他胃肠外给药途径,比如注射或输注。“胃肠外给药”是指除了肠内或局部给药以外的通常通过注射的给药方式,包括但不限于静脉内、肌内、动脉内、鞘内、淋巴内、损伤内、囊内、框内、心内、皮内、腹膜内、经气管、皮下、表皮下、 关节内、囊下、蛛网膜下、脊柱内、硬膜内和胸骨内注射和输注以及经体内电穿孔。
本文所述的“不良反应”(AE)是与使用医学治疗相关的任何不利的和通常无意的或不期望的迹象、症状或疾病。例如,不良反应可能与在响应治疗时免疫***的激活或免疫***细胞的扩增相关。医学治疗可以具有一种或多种相关的AE,并且每种AE可以具有相同或不同的严重性水平。
“肿瘤负荷”是指分布于整个体内的肿瘤物质的总量。肿瘤负荷是指整个体内的癌细胞的总数目或肿瘤的总大小。肿瘤负荷可通过现有技术中已知的多种方法测定,比如在肿瘤自受试者移除后使用卡尺、或在体内时使用成像技术(比如超声、骨扫描、计算层析X射线照相术(CT)或磁共振成像(MRI)扫描)测量其尺寸。
术语“肿瘤大小”是指肿瘤的总大小,其可测量为肿瘤的长度及宽度。肿瘤大小可通过现有技术中已知的多种方法测定,例如在肿瘤自受试者移除后使用卡尺、或在体内时使用成像技术(比如骨扫描、超声、CT或MRI扫描)测量其尺寸。
术语“受试者”、“个体”、“对象”包括任何生物体,优选动物,更优选哺乳动物(例如大鼠、小鼠、狗、猫、兔等),且最优选的是人。术语“受试者”和“患者”在本文中可以互换使用。
本文所述的“抗体”是指能达到期望的生物活性或结合活性的任何形式的抗体。因此,它以最广泛含义使用,但不限于单克隆抗体、多克隆抗体、多特异性抗体、人源化全长人抗体、嵌合抗体及骆驼来源的单域抗体。“抗体”特异性结合抗原并包含通过二硫键互连的至少两条重(H)链和两条轻(L)链。每条重链包含重链可变区(VH)和重链恒定区,重链恒定区包含三个恒定结构域CH1、CH2和CH3。每条轻链包含轻链可变区(VL)和轻链恒定区,轻链恒定区包含一个恒定结构域CL。VH和VL区可进一步细分为称为互补决定区(CDR)的高变区,其散布于更为保守的称为框架区(FR)的区域。一般而言,自N末端至C末端,轻链及重链可变结构域二者皆包含FR1、CDR1、FR2、CDR2、FR3、CDR3及FR4。通常是根据如下的定义将氨基酸分配至每一个结构域的:Sequences of Proteins of Immunological Interest,Kabat等人;National Institutes of Health,Bethesda,Md.;第5版;NIH出版号91-3242(1991):Kabat(1978)Adv.Prot.Chem.32:1-75;Kabat等人,(1977)J.Biol.Chem.252:6609-6616;Chothia等人,(1987)J Mol.Biol.196:901-917或Chothia等人,(1989)Nature 341:878-883。
重链的羧基末端部分可定义主要负责效应子功能的恒定区。通常,人轻链分为κ链及λ链。人重链通常分为μ、δ、γ、α或ε,且将抗体的同种型分别定义为IgM、IgD、IgG、IgA及IgE。IgG亚类是本领域技术人员熟知的,包括但不限于IgG1、IgG2、IgG和IgG4。
术语“抗体”包括:天然存在的和非天然存在的Ab;单克隆和多克隆Ab;嵌合和人源化Ab;人或非人Ab;全合成Ab;和单链Ab。非人Ab可以通过重组方法人源化以降低其在人中的免疫原性。
除非另有明确表示,否则本文所述的“抗体片段”或“抗原结合片段”是指抗体的抗原结合片段,即保留了全长抗体的特异性结合至抗原能力的抗体片段,例如保留一个或多个CDR区的片段。抗原结合片段的实例包括但不限于Fab、Fab’、F(ab’)2及Fv片段;双链抗体;线形抗体;单链抗体分子;纳米抗体及由抗体片段形成的多特异性抗体。
“嵌合抗体”是指如下的抗体以及其片段:其中重链和/或轻链的一部分与源自特定物种(如人)或属于特定抗体种类或亚类的抗体中相应序列相同或同源,而链的其余部分与源自另一物种(如小鼠)或属于另一抗体种类或亚类的抗体中相应序列相同或同源,只要其表现出期望的生物活性即可。
“人抗体”是指仅包含人免疫球蛋白序列的抗体。若人抗体是在小鼠、小鼠细胞或源自小鼠细胞的杂交瘤中产生,则其可含有鼠类碳水化合物链。类似的,“小鼠抗体”或“大鼠抗体”是指仅分别包含小鼠或大鼠免疫球蛋白序列的抗体。
“人源化抗体”是指含有来自非人(如鼠类)抗体以及人抗体的序列的抗体形式。此类抗体含有源自非人免疫球单边的最小序列。通常,人源化抗体将包含实质上全部的至少一个且通常两个可变结构域,其中全部或实质上全部超变环对应于非人免疫球蛋白的超变环,且全部或实质上全部FR区为人免疫球蛋白的FR区。人源化抗体任选还包括免疫球蛋白恒定区(Fc)(通常为人免疫球蛋白恒定区)的至少一部分。
术语“黑色素瘤”是来源于黑色素细胞的一类恶性肿瘤,常见于皮肤,亦见于黏膜、眼脉络膜等部位。黑色素瘤是皮肤肿瘤中恶性程度最高的瘤种,容易出现远处转移。黑色素瘤分为四种亚型,包括肢端型(acral)、粘膜型(mucosal)、皮肤型以及原发灶不确定型,其中皮肤型又分为长期日照的皮肤型(CSD)以及非长期阳性暴露性损害型(non-CSD),CSD和non-CSD统称日光型。McLaughlin等,Cancer,2005,Mar 1,103(5):1000-1007;Chi Z.等,BMC Cancer,2011;11:85公开了肢端和粘膜型黑色素瘤是亚洲人种最主要的亚型,其癌变不是由紫外线辐射导致的DNA突变引起的。与日光型黑色素瘤相比,肢端和粘膜型黑色素瘤只含有少量DNA突变。在美国,95%的黑色素瘤是日光型的,而在亚洲,尤其是中国,70%以上的黑色素瘤是肢端和粘膜型,其中粘膜型又达到了50%以上。Invest New Drugs,34:677-684;Cho,J.等公开了与日光型黑色素瘤相比,免疫疗法治疗肢端型和粘膜型的效果较差。
术语“免疫治疗”是指通过包括诱导、增强、抑制或以其他方式修饰免疫反应的方法治疗患有疾病或具有感染或遭受疾病复发风险的受试者。受试者的“治疗”或“疗法”是 指对受试者进行的任何类型的干预或过程,或给与受试者活性剂,目的在于逆转、缓解、改善、减缓或预防症状、并发症或病症的发作、进展、严重性或复发,或与疾病相关的生化指标。
“程序性死亡受体-1(PD-1)”是指属于CD28家族的免疫抑制性受体。PD-1主要在体内先前活化的T细胞上表达,并且结合两种配体PD-L1和PD-L2。本文使用的术语“PD-1”包括人PD-1(hPD-1)、hPD-1的变体、同种型和物种同源物,以及与hPD-1具有至少一个共同表位的类似物。
药物或治疗剂的“治疗有效量”或“治疗有效剂量”是当单独使用或与另一种治疗剂组合使用时保护受试者免于疾病发作或促进疾病消退的任何量的药物,所述疾病消退通过疾病症状的严重性的降低,疾病无症状期的频率和持续时间的增加,或由疾病痛苦引起的损伤或失能的预防来证明。治疗剂促进疾病消退的能力可以使用本领域技术人员已知的多种方法来评价,比如在临床试验期间的人受试者中,在预测人类功效的动物模型***中,或通过在体外测定法中测定所述药剂的活性。
药物治疗有效量包括“预防有效量”,即当单独或与抗肿瘤剂组合给与处于发展癌症风险的受试者或患有癌症复发的受试者时,抑制癌症的发展或复发的任何量的药物。
“生物治疗剂”是指在支持肿瘤维持和/或生长或抑制抗肿瘤免疫应答的任何生物途径中阻断配体/受体信号传导的生物分子,例如抗体或融合蛋白。
除非另有明确表示,否则本文所用的“CDR”是指免疫球蛋白可变区是使用Kabat编号***定义的互补决定区。
“治疗性抗PD-1单克隆抗体”是指特异性结合至在某些哺乳动物细胞表面上表达的特定PD-1的成熟形式的抗体。成熟的PD-1无前分泌前导序列,或叫前导肽。术语“PD-1”及“成熟的PD-1”在本文中可互换使用,且除非另有明确定义,或明确能从上下文看出,否则应理解为相同分子。
如本文所述,治疗性抗人PD-1抗体或抗hPD-1抗体是指特异性结合至成熟人PD-1的单克隆抗体。
本文所述的“框架区”或“FR”是指不包括CDR区的免疫球蛋白可变区。
“分离的抗体或其抗原结合片段”是指纯化状态且在该情况下所指定的分子实质上不含有其他生物分子,诸如核酸、蛋白质、脂质、碳水化合物或其他材料(诸如细胞碎片或生长培养基)。
“患者”、“病人”或“受试者”是指需要医疗方法或参与临床试验、流行病学研究或用作对照的任意单一受试者,通常为哺乳动物,包括人及其它哺乳动物,比如马、牛、狗或猫。
本文所述的“RECIST 1.1疗效标准”是指Eisenhauver等人、E.A.等人,Eur.J Cancer45:228-247(2009)基于所测量反应的背景针对靶标损伤或非靶标损伤所述的定义。在免疫治疗之前,其是实体肿瘤疗效评估最常用的标准。但随着免疫时代的到来,出现了很多以前在肿瘤评价方面未曾出现的难题,因此基于新出现的由于免疫治疗本身带来的现象,2016年,RECIST工作组对现有的“RECIST v.1.1”进行修正后提出一个新的判断标准,即本文所述的“irRECIST标准”,旨在更好的评估免疫治疗药物的疗效。
术语“ECOG”评分标准,是从患者的体力来了解其一般健康状况和对治疗耐受能力的指标。ECOG体力状况评分标准记分:0分、1分、2分、3分、4分和5分。评分为0是指活动能力完全正常,与起病前活动能力无任何差异。评分为1是指能自由走动及从事轻体力活动,包括一般家务或办公室工作,但不能从事较重的体力活动。
“持续应答”是指在停止用本文所述治疗剂或组合疗法后的持续治疗效应。在一些实施方式中,持续应答具有至少与治疗持续时间相同或为治疗持续时间的至少1.5,2.0,2.5或3倍的持续时间。
“组织切片”是指组织样品的单一部分或片,比如从正常组织或肿瘤的样品切割的组织薄片。
本文所述的“治疗”癌症是指向患有癌症或经诊断患有癌症的受试者采用本文所述治疗方案(如施用抗PD-1抗体或抗PD-1抗体与CDK4/6抑制剂的组合疗法)以达到至少一种阳性治疗效果(比如,癌症细胞数目减少、肿瘤体积减小、癌细胞浸润至周边器官的速率降低或肿瘤转移或肿瘤生长的速率降低)。癌症中的阳性治疗效果可以多种方式测量(参见W.A.Weber,J.Nucl.Med.,50:1S-10S(2009))。比如,关于肿瘤生长抑制,根据NCI标准,T/C≦42%是抗肿瘤活性的最小水平。认为T/C(%)=经***体积中值/对照肿瘤体积中值×100。在一些实施方式中,通过本发明的组合达到的治疗效果是PR、CR、OR、PFS、DFS及OS中的任一个。PFS(也叫“至肿瘤进展的时间”)是指治疗期间及之后癌症不生长的时间长度,且包括患者经历CR或PR的时间量以及患者经历SD的时间量。DFS是指治疗期间及之后患者仍无疾病的时间长度。OS是指与初始或未经治疗的个体或患者相比预期寿命的延长。在一些实施方式中,对本发明组合的应答是PR、CR、PFS、DFS、OR或OS中的任一个,其使用RECIST 1.1疗效标准评定。有效治疗癌症患者的本发明组合的治疗方案可根据多种因素(比如患者的疾病状态、年龄、体重及疗法激发受试者的抗癌反应的能力)而变。尽管本发明的实施方式可不在每个受试者中达到有效的阳性治疗效果,但在统计学上显著数目的受试者中应有效并达到了阳性治疗效果。
术语“给药方式”、“给药方案”可互换使用,是指本发明组合中每一治疗剂的使用剂量及时间。
术语“免疫组化(IHC)”是指利用抗原与抗体特异性结合的原理,通过化学反应使标记抗体的显色剂(荧光素、酶、金属离子、同位素)显色来确定组织细胞内抗原(多肽和蛋白质),并对其进行定位、定性及相对定量的研究的方法。本发明的一些实施方式中,在利用抗PD-1抗体治疗之前,对受试者的肿瘤组织样品进行PD-L1检测,所述检测使用罗氏的抗人PD-L1抗体SP142(Cat No:M4422)进行染色实验。在一些实施方式中,肿瘤细胞的膜染色强度≥1%被定义为PD-L1阳性。
在以下段落中,进一步详细描述本发明的各个方面。
抗PD-1抗体
本文中,“PD-1抗体”是指结合PD-1受体,阻断表达于癌细胞上的PD-L1与表达于免疫细胞(T、B、NK细胞)上的PD-1结合且优选也能阻断表达于癌细胞上的PD-L2与表达于免疫细胞上的PD-1结合的任何化学化合物或生物分子。PD-1及其配体的替代名词或同义词包括:对于PD-1而言有PDCD1、PD1、CD279及SLEB2;对于PD-L1而言有PDCD1L1、PDL1、B7-H1、B7H1、B7-4、CD274及B7-H;且对于PD-L2而言有PDCD1L2、PDL2、B7-DC及CD273。在治疗人个体的任何本发明治疗方法、药物及用途中,PD-1抗体阻断人PD-L1与人PD-1的结合,且优选阻断人PD-L1和PD-L2二者与人PD1结合。人PD-1氨基酸序列可见于NCBI基因座编号:NP_005009。人PD-L1及PD-L2氨基酸序列可分别见于NCBI基因座编号:NP_054862及NP_079515。
本文中,当提及“抗PD-1抗体”时,除非另有说明或描述,否则该术语包括其抗原结合片段。
适用于本发明所述的任意用途、疗法、药物及试剂盒的抗PD-1抗体以高特异性和高亲和力结合PD-1,阻断PD-L1/2与PD-1的结合,并抑制PD-1信号转导,从而达到免疫抑制效果。本文所公开的任意用途、疗法、药物及试剂盒中,抗PD-1抗体包括全长抗体本身,以及结合PD-1受体并在抑制配体结合和上调免疫***方面表现出类似完整Ab的功能特性的抗原结合部分或片段。在一些实施方式中,抗PD-1抗体或其抗原结合片段为与特瑞普利单抗交叉竞争结合人PD-1的抗PD-1抗体或其抗原结合片段。在其他的实施方式中,抗PD-1抗体或其抗原结合片段是嵌合、人源化或人Ab或其抗原结合片段。在用于治疗人个体的某些实施方式中,所述的Ab为人源化Ab。
在一些实施方案中,用于本发明所述的任何用途、疗法、药物及试剂盒的抗PD-1抗体包括单克隆抗体(mAb)或其抗原结合片段,其特异性结合至PD-1,且优选特异性结合至人PD-1。mAb可以为人抗体、人源化抗体或嵌合抗体,且可包括人恒定区。在一些实施方式中,恒定区是选自人IgG1、IgG2、IgG3及IgG4恒定区组成的组;优选地,适 用于本发明所述的任何用途、疗法、药物及试剂盒的抗PD-1抗体或其抗原结合片段包含人IgG1或IgG4同种型的重链恒定区,更优选是人IgG4恒定区。在一些实施方式中,抗PD-1抗体或其抗原结合片段的IgG4重链恒定区的序列包含S228P突变,其用IgG1同种型抗体的相应位置处通常存在的脯氨酸残基替代铰链区中的丝氨酸残基。
优选地,在本发明所述的用途、疗法、药物及试剂盒的任意一个实施方式中,PD-1抗体是单克隆抗体或其抗原结合片段,其轻链CDR为SEQ ID NO:1、2和3所示的氨基酸,重链CDR为SEQ ID NO:4、5和6所示的氨基酸。
更优选地,在本发明所述的用途、疗法、药物及试剂盒的任意一个实施方式中,PD-1抗体是特异性结合至人PD-1且包含:(a)包含SEQ ID NO:7所示的轻链可变区,及(b)包含SEQ ID NO:8所示的重链可变区的单克隆抗体。
进一步优选地,在本发明所述的用途、疗法、药物及试剂盒的任意一个实施方式中,PD-1抗体是特异性结合至人PD-1且包含:(a)包含SEQ ID NO:9所示的轻链,及(b)包含SEQ ID NO:10所示的重链的单克隆抗体。
下表A提供了用于本发明所述的用途、疗法、药物及试剂盒中的示例性抗PD-1抗体mAb的轻链CDR和重链CDR的氨基酸序列编号:
表A:示例性抗人PD-1抗体的轻重链CDR(Kabat)
LCDR1 SEQ ID NO:1
LCDR2 SEQ ID NO:2
LCDR3 SEQ ID NO:3
HCDR1 SEQ ID NO:4
HCDR2 SEQ ID NO:5
HCDR3 SEQ ID NO:6
结合至人PD-1且可用于本发明所述的用途、疗法、药物及试剂盒的抗PD-1抗体的实施例阐述于WO2014206107中。在本发明所述的用途、疗法、药物及试剂盒中可用作抗PD-1抗体的人PD-1 mAb包括WO2014206107中描述的任意一个抗PD-1抗体,包括:特瑞普利单抗(Toripalimab)(一种具有WHO Drug Information(第32卷,第2期,第372-373页(2018))中所述的结构且包含序列SEQ ID NO:9和10所示的轻链及重链氨基酸序列的人源化IgG4 mAb。在优选的实施方案中,可用于本发明所述的任一用途、疗法、药物及试剂盒的抗PD-1抗体选自WO2014206107中描述的人源化抗体38、39、41和48。在特别优选的实施方案中,可用于本发明所述的任一用途、疗法、药物及试剂盒 的抗PD-1抗体为特瑞普利单抗。
可用于本发明所述的任一用途、疗法、药物及试剂盒的抗PD-1抗体还包括FDA已经批准的Nivolumab和Pembrolizumab。
在某些实施方案中,可用于本发明所述的任一用途、疗法、药物及试剂盒的抗PD-1抗体也包括特异性结合PD-L1以阻断PD-L1与PD-1结合的抗PD-L1单克隆抗体,如nivolumab、pembrolizumab、toripalimab、Sintilimab、Camrelizumab、Tislelizumab、Cemiplimab。
如本文所述的“PD-L1”表达或“PD-L2”表达是指细胞表面上的特定PD-L蛋白质或细胞或组织内的特定PD-L mRNA的任何可检测的表达水平。PD-L蛋白质表达可利用诊断性PD-L抗体在肿瘤组织切片的IHC分析中或通过流式细胞术检测。或者,肿瘤细胞的PD-L蛋白质表达可通过PET成像使用特异性结合至期望PD-L靶标(比如PD-L1或PD-L2)的结合剂检测。
用于在肿瘤组织切片的IHC分析中定量PD-L1蛋白质表达的方法,参见以下但不限于Thompson,R.H.等人,PNAS 101(49):17174-17179(2004);Taube,J.M.等人,Sci Transl Med 4,127ra37(2012);及Toplian,S.L.等人,New Eng.J.Med.366(26):2443-2454(2012)等。
一种方法采用PD-L1表达呈阳性或阴性的简单二元终点,其中阳性结果用显示细胞表面膜染色的组织学证据的肿瘤细胞百分比来定义。将肿瘤组织切片计数为总肿瘤细胞的至少1%定义为PD-L1表达呈阳性。
在另一方法中,在肿瘤细胞中以及在浸润免疫细胞中定量肿瘤组织切片中的PD-L1表达。将展现膜染色的肿瘤细胞及浸润免疫细胞的百分比单独的定量为<1%、1%至50%,及随后的50%直至100%。对于肿瘤细胞,若评分<1%,则将PD-L1表达计数为阴性,若评分≥1%则为阳性。
在一些实施方式中,基于与由适当对照的PD-L1表达水平的比较,由恶性细胞和/或由肿瘤内的浸润免疫细胞的PD-L1表达水平测定为“过表达”或“升高”。比如,对照PD-L1的蛋白质或mRNA表达水平可为相同类型的非恶性细胞中或来自匹配正常组织的切片中定量的水平。
CDK4/6抑制剂
本文中,术语“CDK”即周期蛋白依赖性激酶(cyclin-dependent kinases),是一组丝氨酸/苏氨酸蛋白激酶。CDK通过和周期蛋白cyclin协同作用对丝氨酸/苏氨酸蛋白的磷酸化驱动细胞周期,是细胞周期调控中的重要因子。CDK家族有CDK 1-8等8种,每种CDK结合不同类型的cyclin形成复合物,调节细胞从G1期过渡到S期或G2期过渡到M期以 及退出M期的进程。
目前FDA已经批准上市的CDK4/6抑制剂主要有瑞博西尼、帕博西尼和玻玛西林(Abemaciclib)等。瑞博西尼与芳香酶抑制剂联用可作为一线用药治疗HR阳性以及HER2阴性的绝经后晚期转移性乳腺癌女性患者;帕博西尼与来曲唑联用治疗***受体(ER)阳性、人表皮生长因子受体2(HER2)阴性的绝经后妇女转移性乳腺癌;玻玛西林主要用于治疗接受内分泌疗法后疾病进展的激素受体(HR)阳性以及人类表皮生长因子受体2(HER2)阴性的晚期或转移性乳腺癌成年患者。
而处于临床研究阶段的CDK4/6抑制剂有几十种,如Milciclib、Trilaciclib、Lerociclib和Voruciclib等。
在本发明所述的任一用途、疗法、药物及试剂盒中,CDK4/6抑制剂包括但不限于帕博西尼。
在本发明所述的任一用途、疗法、药物及试剂盒中,联用包括但不限于特瑞普利单抗(Toripalimab)和帕博西尼。
疾病及其治疗
本发明涉及癌症的治疗。本发明包括向有需要的患者单独施用抗PD-1抗体或其抗原结合片段;或包括向有需要的患者施用与其他抗癌剂组合的抗PD-1抗体。
本文中,术语“癌症”是指以身体中异常细胞不受控制的生长为特征的广泛的各种疾病。不受调节的细胞***、生长***和生长导致恶性肿瘤的形成,其侵入邻近组织并还可以通过淋巴***或血流转移至身体的远端部分。适合采用本发明的方法、药物和试剂盒来治疗或预防的癌症的实例包括但不限于癌、淋巴瘤、白血病、母细胞瘤及肉瘤。癌症的更特定的实例包括鳞状细胞癌、骨髓瘤、小细胞肺癌、非小细胞肺癌、胶质瘤、霍奇金淋巴瘤、非霍奇金淋巴瘤、急性骨髓性白血病、多发性骨髓瘤、胃肠(道)癌、肾癌、卵巢癌、肝癌、淋巴母细胞性白血病、淋巴细胞白血病、结肠直肠癌、子宫内膜癌、肾癌、***癌、甲状腺癌、黑色素瘤、软骨肉瘤、神经母细胞瘤、胰腺癌、多形性神经胶质母细胞瘤、子***、脑癌、胃癌、膀胱癌、肝细胞瘤、乳腺癌、结肠癌及头颈癌。
优选地,适用于本发明的癌症患者是PD-L1表达呈阳性的癌症患者,或者是外周血或肿瘤组织中肿瘤突变负荷(TMB)≥3.6Muts/Mb,或者是PD-L1表达呈阳性同时外周血或肿瘤组织中肿瘤突变负荷(TMB)≥3.6Muts/Mb。在某些实施方案中,适用于本发明方法进行治疗的癌症患者优选是肿瘤组织中检出BRAF基因突变的癌症患者。
本文中,术语“肿瘤突变负荷(TMB)”是指每百万碱基中被检测出的,体细胞基因编码错误、碱基替换、基因***或缺失错误的总数。在本发明的一些实施方式中,肿瘤 突变负荷(TMB)是通过分析体细胞突变(包括编码基置换和研究的面板序列的巨碱基***)来估计的。本发明中,当受试者的肿瘤突变负荷(TMB)大于等于3.6Muts/Mb,预示着对此类受试者单独施用抗PD-1抗体,或施用与其他抗癌剂组合的抗PD-1抗体相比TMB<3.6Muts/Mb能取得更好的疗效。
本发明治疗癌症的方法包括给予需要的个体治疗有效量的抗PD-1抗体,或抗PD-1抗体与CDK4/6抑制剂。所述抗PD-1抗体可如本文任一实施方案所述,更优选为轻链CDR为SEQ ID NO:1、2和3所示的氨基酸,重链CDR为SEQ ID NO:4、5和6所示的氨基酸的抗体,更优选为包含SEQ ID NO:7所示轻链可变区和SEQ ID NO:8所示的重链可变区的单克隆抗体,更优选为包含SEQ ID NO:9所示的轻链和SEQ ID NO:10所示的重链的单克隆抗体,更优选为WO2014206107中描述的人源化抗体38、39、41和48,最优选为特瑞普利单抗。所述CDK4/6抑制剂为帕博西尼Milciclib、Trilaciclib、Lerociclib和Voruciclib,优选为帕博西尼。在优选的实施方案中,本发明联用特瑞普利单抗和帕博西尼治疗癌症。
在特别优选的实施方案中,本发明提供治疗黑色素瘤的方法,所述方法包括给予黑色素瘤患者治疗有效量的特瑞普利单抗;优选地,该患者PD-L1表达呈阳性,或者外周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb,或者PD-L1表达呈阳性、同时外周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb。在某些实施方案中,该黑色素瘤患者优选是肿瘤组织中检出BRAF基因突变的黑色素瘤患者,更优选为非肢端黑色素瘤或原发型黑色素瘤。在一些实施方案中,所述黑色素瘤是肢端型或/和粘膜型黑色素瘤。在另一特别优选的实施方案中,本发明提供治疗黑色素瘤的方法,所述方法包括给予黑色素瘤患者治疗有效量的特瑞普利单抗与帕博西尼;优选地,该患者的外周血或肿瘤组织中存在CDK4或CCND1基因扩增。
根据标准医药实践,当给予两种或两种以上治疗剂(即“组合疗法”)时,每一个治疗剂均可单独施用或在药物组合物中施用,该药物组合物包含治疗剂及一种或多种药学上可接受的载体、赋形剂及稀释剂。
本发明的组合疗法中的每一个治疗剂可同时施用、并行施用或以任何次序依次施用。在组合疗法中的治疗剂以不同剂型施用,比如一种药物是片剂或胶囊且另一种药物是无菌液体,和/或以不同给药时间施用,比如化学治疗剂至少每日施用且生物治疗剂不频繁施用,比如每一周、或每两周或每三周一次。
在一些实施方式中,CDK4/6抑制剂是在施用抗PD-1抗体之前施用,而在另一些实施方式中,CDK4/6抑制剂是在施用抗PD-1抗体之后施用。
在一些实施方式中,在药物作为单一疗法用于治疗相同癌症时,组合疗法中的治疗剂 中至少一种使用相同的给药方案(剂量、频次、治疗持续时间)施用。
本发明所述的组合疗法中的每一个小分子治疗剂可经口或胃肠外(比如静脉内、肌内、腹膜内、皮下、经直肠、局部或透皮施用途径)施用。
本发明所述的组合疗法可在手术之前或之后施用,且可在放疗之前、期间或之后施用。
在一些实施方式中,将本发明所述的组合疗法施用于先前未经生物治疗剂(通常指抗PD-1抗体)或化学治疗剂(通常指CDK4/6抑制剂)治疗的患者中。在另一些实施方式中,将组合疗法施用于用生物治疗剂或化学治疗剂治疗之后而不能达到持续响应的患者中。
本发明所述的组合疗法可用于治疗通过触诊或通过现有技术中已知的成像技术发现的肿瘤中,比如MRI、超声或CAT扫描。
本发明所述的组合疗法优先施用于经测试PD-L1表达呈现阳性或TMB≥3.6Muts/Mb的癌症患者。
本发明的组合疗法的给药方案的选择取决于若干的因素,包括但不限于血清或组织转化率、症状的程度、免疫原性及所治疗个体的靶标细胞、组织、器官的可达性。优选的,给药方案要配合可接受的副作用程度来给患者递送最大量的各种治疗剂。因此,组合疗法中的每一个生物治疗剂和化学治疗剂的剂量及给药频率取决于特定的治疗剂、所治疗的癌症的严重程度及患者的特性。
本发明所述的抗PD-1抗体和CDK4/6抑制剂可作为包含第一容器及第二容器及包装插页的试剂盒提供。
第一容器含有至少一个剂量的包含抗PD-1抗体的药物,第二容器含有至少一个剂量的包含CDK4/6抑制剂,且包装插页或标签包含使用药物治疗癌症的说明。试剂盒可进一步包含可用于施用药物的其他材料,比如稀释剂、滤纸、IV袋及线、针及注射器。作为一种优选方式,说明书可陈述药物旨在用于治疗癌症患者,其PD-L1表达经免疫组化染色(IHC)分析法测试呈现阳性。
本发明所述的治疗剂可以构成药物组合物,比如含有本文所述抗PD-1抗体或/和该抗PD-1抗体以外的其他抗癌剂以及其他药学上可接受的载体的药物组合物。如本发明所述,“药学上可接受的载体”包括生理上相容的任何和所有溶剂、分散介质、包衣、抗细菌剂和抗真菌剂、等渗和吸收延迟剂等。优选的,适用于含有抗PD-1抗体的组合物的载体适合于静脉内、肌内、皮下、胃肠外、脊椎或表皮施用,比如通过注射或输注,而用于含有其他抗癌剂的组合物的载体适合于胃肠外施用,比如口服。本发明的药物组合物可以含有一种或多种药学上可接受的盐、抗氧化剂、水、非水载体、和/或佐剂比如防腐剂、润湿剂、乳化剂和分散剂。
调整给药方案以提供最佳所需的反应,比如最大治疗反应和/或最小不良效果。对于 抗PD-1抗体,包括与另一抗癌剂组合时的施用,剂量范围可为约0.01至约20mg/kg,约0.1至约10mg/kg个体体重,或120mg、240mg、360mg、480mg固定剂量。例如,剂量可以是约0.1,约0.3,约1,约2,约3,约5,或约10mg/kg个体体重。通常设计给药方案以实现这样的暴露,其导致基于Ab的典型药代动力学特性的持续受体占用(RO)。代表性的给药方案可能为约每周一次,约每两周一次,约每三周一次,约每四周一次,约一个月一次,或更长一次施用。在一些实施方式中,约每两周一次向个体施用抗PD-1抗体。
其他抗癌剂的给药时间表针对不同的药物而有所不同。本发明的一些具体实施方式中,所述的CDK抑制剂的给药时间表针对不同的亚型而变化。
用途
本发明还包括用于治疗癌症患者的抗PD-1抗体或其与CDK4/6抑制剂的组合,以及抗PD-1抗体或其与CDK4/6抑制剂的组合在制备治疗癌症患者的药物中的应用。所述癌症患者或其所换的癌症可如前述任一实施方案所述;优选地,所述癌症患者是PD-L1表达呈阳性的癌症患者,或者是外周血或肿瘤组织中肿瘤突变负荷(TMB)≥3.6Muts/Mb、或者是PD-L1表达呈阳性同时外周血或肿瘤组织中肿瘤突变负荷(TMB)≥3.6Muts/Mb。在某些实施方案中,所述癌症患者是肿瘤组织中检出BRAF基因突变的癌症患者。更优选地,所述黑色素瘤患者PD-L1表达呈阳性,或者外周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb,或者PD-L1表达呈阳性同时外周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb。在某些实施方案中,该黑色素瘤患者更优选为非肢端黑色素瘤或原发灶不确定型黑色素瘤。在一些实施方案中,该黑色素瘤患者是肢端型黑色素瘤患者或粘膜型黑色素瘤患者。
优选的用于治疗癌症患者的抗PD-1抗体可如本文任一实施方案所述,更优选为轻链CDR为SEQ ID NO:1、2和3所示的氨基酸,重链CDR为SEQ ID NO:4、5和6所示的氨基酸的抗体,更优选为包含SEQ ID NO:7所示轻链可变区和SEQ ID NO:8所示的重链可变区的单克隆抗体,更优选为包含SEQ ID NO:9所示的轻链和SEQ ID NO:10所示的重链的单克隆抗体,更优选为WO2014206107中描述的人源化抗体38、39、41和48,最优选为特瑞普利单抗。所述CDK4/6抑制剂优选为帕博西尼。
在优选的实施方案中,本发明涉及用于治疗癌症患者的特瑞普利单抗或特瑞普利单抗与帕博西尼的组合。更具体而言,本发明涉及用于治疗黑色素瘤患者的特瑞普利单抗或特瑞普利单抗与帕博西尼的组合;更优选地,所述黑色素瘤患者PD-L1表达呈阳性,或者外周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb,或者PD-L1表达呈阳性、同时外 周血或肿瘤组织中肿瘤负荷(TMB)≥3.6Muts/Mb,或肿瘤组织中检出BRAF基因突变的黑色素瘤患者,更优选为非肢端黑色素瘤或原发型黑色素瘤。在一些实施方案中,该黑色素瘤患者是肢端型黑色素瘤患者或粘膜型黑色素瘤患者。
抗PD-1抗体对治疗癌症效果的预测方法
本文可通过对来自患者的肿瘤活检样本或相应的外周血样本上进行全外显子组测序(如利用第二代测序技术),鉴定某些基因是否发生突变或基因扩增,来预测抗PD-1抗体、CDK4/6抑制剂或两者联用进行抗肿瘤治疗的疗效。
本文所述的“基因突变”或“基因改变”包括基因截短、基因重排/融合、基因扩增、基因缺失和基因取代/***等。
本文中,术语“基因扩增”是指某特异蛋白质编码的基因的拷贝数选择性地增加而其他基因并未按比例增加的过程。在自然条件下,基因扩增是通过从染色体切除基因的重复序列再在质粒中进行染色体外复制或通过将核糖体RNA的全部重复序列生成RNA转录物再转录生成原来DNA分子的额外拷贝而实现的。本发明的一些实施例中公开了基因测序分析。
本发明的一些实施方式中,本发明所述的受试者中具有某些独特的基因突变,如一些受试者具有CDK4或CDDN 1基因扩增;一些受试者则具有BRAF基因突变,尤其是非肢端黑色素瘤或原发灶不确定型黑色素瘤受试者中具有BRAF基因突变;一些粘膜黑色素瘤受试者中则具有NF1基因突变;一些受试者(包括肢端型、粘膜型、非肢端型(皮肤型)和原发性未知型黑色素瘤患者)具有NRAS基因突变。在一些实施方式中,BRAF基因突变选自基因重排/融合和基因取代/***。在一些实施方式中,NRAS基因突变为基因取代/***。
本发明的一些实施方式中,具有BRAF基因突变通常预示着该患者用本发明所述的抗PD-1抗体进行治疗疗效更好。本发明的一些实施方式中,具有CDK4或CCND1基因扩增预示着该患者用本发明所述的抗PD-1抗体与CDK4/6抑制剂联用或单独使用CDK4/6抑制剂进行治疗疗效更好。本发明的一些实施方式中,具有NRAS基因突变预示着该患者单独使用本发明所述的抗PD-1抗体进行治疗疗效不理想。
因此,本发明提供一种使用本发明所述的抗PD-1抗体,尤其是特瑞普利单抗对治疗个体中癌症的效果的预测方法,包括在治疗前检测患者外周血中的生物标记物,所述的生物标记物选自但不限于BRAF、NRAS、CDK4或CCND1。其中,存在BRAF基因突变表明所述个体适于用抗PD-1抗体进行治疗,存在NRAS基因突变表明该个体不适合用抗PD-1抗体进行治疗,存在CDK4或CCND1基因扩增表明该个体用适于用抗PD-1抗体与 CDK4/6抑制剂进行治疗或适于单独使用CDK4/6抑制剂进行治疗。优选地,所述癌症为黑色素瘤。
本发明还包括利用BRAF基因或NRAS基因预测肿瘤患者对抗PD-1抗体治疗的效果的方法。存在BRAF基因突变表明所述肿瘤患者适于用抗PD-1抗体进行治疗。存在NRAS基因突变表明所述肿瘤患者不适合用抗PD-1抗体进行治疗。
在某些实施方案中,本发明还提供生物标记物(尤其是BRAF基因,和/或NRAS基因,和/或CDK4基因,和/或CCND1基因)的检测试剂在制备预测抗PD-1抗体和/或CDK4/6抑制剂治疗癌症的效果用的试剂盒中的用途。这类试剂包括但不限于测试中常规使用到的试剂,包括但不限于引物、探针、PCR所需试剂等。所述癌症优选为黑色素瘤。优选地,所述预测包括:存在BRAF基因突变表明所述个体适于用抗PD-1抗体进行治疗,存在NRAS基因突变表明该个体不适合用抗PD-1抗体进行治疗,存在CDK4或CCND1基因扩增表明该个体用适于用抗PD-1抗体与CDK4/6抑制剂进行治疗或适于单独使用CDK4/6抑制剂进行治疗。
缩略语
贯穿于本发明的说明书及实施例中,使用以下缩略语:
BID 一个剂量,每日2次
CDR 互补决定区
DFS 无疾病生存
FR  框架区
IgG 免疫球蛋白G
IHC 免疫组织化学
OR  总体应答
ORR 客观缓解率
OS  总生存
PD  疾病进展
PFS 无进展生存
PR  部分应答
CR  完全应答
SD  疾病稳定
DLT 剂量限制性毒性
MTD 最大耐受剂量
AE      不良事件
Q2W     每两周一个剂量
QD      每天一个剂量
CSD     长期日照型
non-CSD 非长期日照型
IRC     独立审查委员会
TRAE    与治疗相关的不良反应
SAE     严重不良反应
MM      黑色素瘤
本发明通过以下实施例进一步阐述,但所述实施例不应被解释为限制本发明。整个申请中引用的所有参考文献的内容通过引用的方式明确并入本文。
实施例
实施例1:抗PD-1抗体单用治疗黑色素瘤的临床研究
入组标准:符合条件的受试者必须(1)年龄至少为18岁,(2)患有局部晚期或转移性黑素瘤,(3)经过标准全身治疗后难以治愈,(4)ECOG评分为0或1,(5)无自身免疫性疾病史或持续感染史,(6)之前未接受过任何抗-PD-1/或抗-PD-L1免疫治疗。
受试者必须是按RECIST v 1.1标准有可评估病灶,不允许同时使用抗肿瘤药物治疗、不允许全身类固醇药物治疗或未使用过抗CTLA4、抗PD-1、抗PD-L1抗体治疗。
受试药物:抗PD-1抗体特瑞普利单抗(WO2014206107)。
本试验使用的抗PD-1抗体剂量为:3mg/kg,每两周静脉注射一次(Q2W)。
临床设计:
这是一个单臂,临床II期,非盲临床试验。该研究用以评价抗PD-1抗体用于治疗晚期黑色素瘤患者的安全性和抗肿瘤活性。
从2016年12月26日到2017年9月15日,共有6个中心筛选了161名标准治疗无效的黑色素瘤患者,本研究共纳入128例患者(如图1所示)。入组受试者的人口学统计数据如表1所示。平均年龄52.5岁,男性57例(45.5%),女性71例(55.5%)。一名患者后来被诊断为没有复发的癌症,并被排除在疗效分析之外。
在黑色素瘤亚型中,50例(39.4%)为肢端型,22例(17.3%)为黏膜型,29例(22.9%)为非肢端皮肤型,26例(20.5%)为原发灶不确定型。入组的34例(26.6%)患者记录了到BRAF突变。大多数患者接受了多线的既往治疗,88名(68.7%)患者至少经过先前两种***疗法。99例(78.0%)患者接受了先前***化疗,9例(7.0%)患者曾接受过伊匹单抗的治疗。
表1:入组受试者的人口学统计数据
Figure PCTCN2021076456-appb-000001
Figure PCTCN2021076456-appb-000002
注:PD-L1阳性定义为采用SP142IHC染色肿瘤细胞PD-L1表达为≥1%。
1.1安全性研究:
截止到2019年8月15日,也就是最后一名患者登记入组的23个月后,患者接受的特瑞普利单抗的平均数为10剂(范围:1至73剂)。128位患者中有116位(97.7%)经历了治疗相关的副反应。大部分为1级或2级。常见(>5%)治疗相关的副反应见表2。治疗相关的SAE发生在10例(7.8%)患者中。25例(19.5%)患者经历了3级或以上的TRAE,包括13例(10.2%)3级和12例(9.4%)4级副作用,无患者经历5级副反应。15例(11.7%)患者因TRAE而终止治疗;由TRAE引起的剂量延迟8例(6.3%)。特殊关注的TRAE少于5%,包括5例(3.9%)白癜风,3例(2.3%)肝损伤(2例4级),2例(1.6%)急性胰腺炎,2例(1.6%)间质性肺疾病(1例3级),2例(1.6%)肾上腺功能不全,2例(1.6%)垂体功能低下,1例(0.8%)葡萄膜炎(3级)。
表2:所有受试者(n=36)中常见的(≥20%)与toripalimab治疗相关的不良事件
N(%) 所有 1级 2级 3级 4级 5级
所有不良反应 125(97.7 42(32.8) 42(32.8) 29(22.7) 12(9.4) 0
TSH上升 42(32.8 34(26.6) 8(6.3) 0 0 0
ALT上升 41(32.0) 34(26.6) 5(3.9) 2(1.6) 0 0
高血糖症 41(32.0) 35(27.3) 4(3.1) 1(0.8) 1(0.8) 0
蛋白尿 35(27.3) 33(25.8) 1(0.8) 1(0.8) 0 0
甲状腺功能减退 35(27.3) 19(14.8) 16(12.5) 0 0 0
肌酸激酶增加 33(25.8) 26(20.3) 3(2.3) 2(1.6) 2(1.6) 0
皮疹 30(23.4) 27(21.1) 3(2.3) 0 0 0
AST上升 30(23.4) 28(21.9) 1(0.8) 1(0.8) 0 0
皮肤脱色 30(23.4) 28(21.9) 2(1.6) 0 0 0
白细胞尿 29(22.7) 27(21.1) 1(0.8) 1(0.8) 0 0
血尿 29(22.7) 29(22.7) 0 0 0 0
白血球减少 28(21.9) 22(17.2) 6(4.7) 0 0 0
贫血 27(21.1) 9(7.0) 15(11.7) 3(2.3) 0 0
淀粉酶增加 25(19.5) 15(11.7) 5(3.9) 2(1.6) 3(2.3) 0
嗜中性白血球减少 24(18.8) 18(14.1) 6(4.7) 0 0 0
瘙痒 24(18.8) 20(15.6) 3(2.3) 1(0.8) 0 0
甲状腺机能亢进 23(18.0) 18(14.1) 5(3.9) 0 0 0
DBIL升高 22(17.2) 15(11.7) 7(5.5) 0 0 0
TBIL升高 22(17.2) 22(17.2) 0 0 0 0
TSH减少 19(14.8) 16(12.5) 3(2.3) 0 0 0
疲劳 19(14.8) 19(14.8) 0 0 0 0
食欲降低 19(14.8) 17(13.3) 2(1.6) 0 0 0
发烧 15(11.7) 15(11.7) 0 0 0 0
咳嗽 15(11.7) 12(9.4) 3(2.3) 0 0 0
血脂升高 14(10.9) 2(1.6) 5(3.9) 6(4.7) 1(0.8) 0
1.2抗肿瘤活性研究:
截至2019年8月15日,在意向治疗(ITT)人群中(n=127),61人(48.0%)死亡,46人(36.2%)停止治疗,20人(15.7%)仍在研究中。中位治疗时间为4.6个月(范围为0.2至33.6个月)。在经IRC/RECIST v1.1评估的127例患者中,22例患者获得了确定的客观反应(1例完全反应和21例部分反应)。每个RECIST v1.1的ORR为17.3%(95%CI 11.2-25.0),每irRECIST的ORR为18.1%(95%CI 11.8-25.9),具体的结果见表3。
表3:根据RECIST v1.1或irRECIST标准评估临床疗效
Figure PCTCN2021076456-appb-000003
注:ORR=(CR+PR)/总数×100%;DCR=(CR+PR+uPR+SD)/总数×100%。
CR:完全响应;PR:部分响应;uPR:未确认的PR;SD:疾病稳定;PD:疾病进展;NE:未评估;ORR:客观响应率;DCR:疾病控制率;CI:信赖区间。
49例(38.6%)受试者的靶病变大小相对于基线减少,具体见图2(A)。3例SD受试者最初出现部分响应,但这些响应无法证实。中位响应时间为3.5个月(95%CI 1.7-3.6),中位持续响应(DOR)未达到,22例患者中只有9例在最初反应后出现疾病进展,具体见图2(B)和图2(C)。每RECISTv1.1的DCR为57.5%(95%CI 48.4-66.2),每irRECIST的DCR为59.8%(95%CI 50.8-68.4)。每RECISTv1.1的中位PFS为3.6个月(95%CI 3.7-5.5),每irRECIST的中位PFS为3.7个月(95%CI 3.3-9.1),具体见图3(A)。
ITT人群(n=127)的总体OS中值估计为22.2个月(95%CI 15.3至未达到),具体结果见图3(B)。
经历客观响应(n=22)和疾病稳定(n=51)的受试者的中位OS未达到,在22例PR/CR患者中只有2例死亡,在51例SD患者中有17例死亡,而疾病进展的54患者的中位总生存mOS为9.7个月,具体结果见图4(A)。
在肢端、粘膜、非肢端皮肤和原发灶不确定型的四种黑色素瘤亚型中,独立审查委员会(independent review committee,IRC)评估每RECIST v1.1的ORRs分别为14.0%、0%、31.0%和23.1%,每位RECIST v1.1的PFS中值分别为3.2、1.9、5.5和7.3个月,具体结果见表4和图5。肢端和粘膜亚型的中位OS分别为16.9个月和10.3个月,而原发灶不确定型和非肢端皮肤型黑色素瘤的中位OS在截止日期前未达到,具体见图4(B)。
表4:RECIST v1.1在黑色素瘤亚组的临床疗效和生存期评估
Figure PCTCN2021076456-appb-000004
Figure PCTCN2021076456-appb-000005
NE:不可预测。
1.3药代动力学和免疫原性
特瑞普利单抗的稳态平均谷血浆浓度是39.8μg/mL(范围4.9-92.4μg/mL),远高于全PD-1阻断浓度1.5u g/ml(10nmol/L)。对128例患者进行了抗药抗体(ADA)检测。16例(12.5%)患者为ADA阳性,其中治疗前有3例为阳性,治疗后有13例(10.2%)为ADA阳性。然而,ADA阳性个体均无连续阳性样本。16例患者中只有1例降低了特瑞普利单抗的谷浓度,同时检测到ADA,表明存在中和活性。ADA阳性与ADA阴性患者在AE、SAE、3级及以上AE发生率、终止或剂量延迟、临床疗效等方面无显著差异。
实施例2:生物标记物与临床疗效相关性研究
2.1肿瘤中PD-L1的表达
通过对127例患者的肿瘤活检标本进行检测,从而分析肿瘤组织学与抗PD-1抗体在临床疗效上的相关性。采用罗氏公司的兔抗人PD-L1抗体SP142进行检测。PD-L1阳性状态被定义为肿瘤细胞存在膜染色强度≥1%。
如图6(A)所示,PD-L1阳性26例(20.5%),PD-L1阴性84例(66.1%),17例(13.4%)患者PD-L1表达情况不明。如图6(B)所示,在四种黑色素瘤亚型中,PD-L1 +在肢端型(6.8%)和粘膜型(10.5%)中的比例明显低于非肢端型(37.5%)和原发灶不确定型的黑色素瘤(52.2%)。
如图6(C)和6(D)所示,PD-L1 +患者相比PD-L1 -患者施用特瑞普利单抗治疗在ORR(38.5%对比11.9%,p=0.0065)和DCR(80.8%对比48.8%,p=0.006)中的响应更好。PD-L1 +患者在无进展生存PFS和总生存OS中也比PD-L1 -患者有显著的生存优势,中位PFS分别为7.7个月和2.7个月,HR=0.53(95%CI 0.32-0.88),p=0.013;中位OS分别为,未达到对比14.4个月,HR=0.35(95%CI 0.19-0.63),p=0.0005。
2.2肿瘤突变负担TMB测定
在实施例1的试验中,采用第二代测序技术对来自患者的肿瘤活检标本和配对的外周血样本上进行了全外显子组测序。肿瘤突变负担(TMB)是通过分析人类基因组编码区域内的体细胞突变来确定的。如图6(A)所示,98例患者获得有效结果,TMB在本研究中普遍较低,中位TMB为每百万碱基对1.5个突变(Muts/Mb)。未发现MSI高的患者。只有6例患者的TMB超过10Muts/Mb,其中3例超过20Muts/Mb。
如图6(B)所示,在四种黑色素瘤亚型中,粘膜黑色素瘤的TMB最低,中位TMB为1.6Muts/Mb,只有一名患者(6.7%)的TMB超过3.6Muts/Mb。根据Robert M.Samstein等人的建议,本研究选择TMB值前20%的截止值(3.6muts/Mb)来定义TMB高人群。以3.6Muts/Mb为界值,TMB≥3.6Muts/Mb(n=20)的患者的反应优于TMB<3.6Muts/Mb(n=78)的患者(ORR 30.0%对12.8%),但差异无统计学意义(p=0.088),具体见图6(A)和表5。TMB≥3.6Muts/Mb组PFS和OS数值上也较好,但差异也无统计学意义(图6,E和F)。
本研究中TMB≥3.6Muts/Mb(n=20)和PD-L1+(n=26)组主要为两个独立群体,26例PD-L1阳性患者中仅有7例TMB≥3.6Muts/Mb(图6,A)。
表5:每个亚型中最高20%的TMB值作为疗效分析的截止值
Figure PCTCN2021076456-appb-000006
实施例3:基因测序分析
在实施例1的试验中,采用第二代测序技术对来自患者进行肿瘤活检和配对的外周血样本上进行了全外显子组测序,从98例患者中鉴定出19278个基因突变,包括7964个错义突变、509个基因缺失、482个重排、288个选择性剪接位点、129个移码截断和8157个基因扩增。在排除了公共外显子组中经常发生突变的基因后,最频繁发生变化的基因(≥10%)是BRAF(33%)、TERT(32%)、CDKN2A(12%)、NRAS(16%)、CDK4(12%)、APOB(11%)、 CCND1(11%)、AGAP2(11%)、NF1(10%)、LRP1B(10%)、MDM2(10%)和KIT(10%),具体见图7。
如表6所示,来自98名患者的测序结果显示,在黑色素瘤亚组中,基因组的改变具有独特的模式。BRAF突变在非肢端皮肤亚型(11/23,48%)和原发未知型黑色素瘤(14/21,67%)中发生频率较高,而在肢端型(7/39,18%)和粘膜型(0/15,0%)中发生频率较低。相比之下,NF1突变在粘膜黑色素瘤中的富集程度(4/15,27%)高于其他三种亚型(8%,4%和10%)。此外,CDK4或CCND1(Cyclin D1)扩增在33%(13/39)肢端型和20%(3/15)粘膜型中被观察到,而仅在9%(2/23)非肢端皮肤型和10%(2/21)原发未知型黑色素瘤中被发现。本研究发现,70%(7/10)的CCND1拷贝数变异作为11q13基因扩增的部分在肢端亚型中被观察到。携带CCND1扩增(n=10)的受试者对于toripalimab治疗的响应低(ORR 0%),该研究显示对于具有CCND1扩增的患者,利用CDK4/6靶向治疗或CDK4/6抑制剂与抗PD-1抗体联合治疗具有较好的潜力。
本研究还发现,具有NRAS突变的受试者(n=16),其中包括7例肢端型,3例粘膜型,3例非肢端型(皮肤型),和3例原发性未知型,对于toripalimab治疗的ORR为6.3%(1/16),该研究显示NRAS突变可作为免疫治疗不良预后的预测。
表6:来自98例患者的基因突变的全外显子组测序
Figure PCTCN2021076456-appb-000007
实施例4:瘤活检中信使RNA表达谱分析
在实施例1的试验中,对从肿瘤活检中提取的mRNA进行RNA测序和表达谱分析。从46例患者中获得了有效的结果。对IFN-γ相关基因组(IDO1、CXCL10、CXCL9、HLA-DRA、STAT1、IFNG)、炎症相关基因组(IL-6、CXCL1、CXCL2、CXCL3、CXCL8、PTGS2)、血管生成相关基因组(VEGFA、KDR、ESM1、PECAM1、ANGPTL4、CD34)的基因表达特征进行评价。发现具有临床获益(CR+PR+SD)的患者和进行性疾病的患者在血管生成相关基因组,IFN相关基因组和炎症相关基因组的签名得分之间并无显着差异(图8)。

Claims (20)

  1. 抗PD-1抗体或其抗原结合片段在制备治疗癌症患者的药物中的应用,其特征在于,所述癌症患者是PD-L1表达呈阳性的癌症患者,或者是外周血或肿瘤组织中肿瘤突变负荷≥3.6Muts/Mb的癌症患者;或者是PD-L1表达呈阳性且外周血或肿瘤组织中肿瘤突变负荷(TMB)≥3.6Muts/Mb的癌症患者;或者是肿瘤组织中检出BRAF基因突变的癌症患者。
  2. 如权利要求1所述的应用,其特征在于,所述癌症患者是黑色素瘤患者。
  3. 如权利要求2所述的应用,其特征在于,所述黑色素瘤为晚期或转移性黑色素瘤。
  4. 如权利要求2所述的应用,其特征在于,所述黑色素瘤患者选自肢端型、黏膜型、非肢端皮肤型或原发灶不确定型黑色素瘤患者;优选为非肢端黑色素瘤或原发灶不确定型黑色素瘤患者。
  5. CDK4/6抑制剂单独或其与抗PD-1抗体或其抗原结合片段的组合在制备治疗癌症患者的药物中的应用,其特征在于,所述癌症患者是肿瘤组织中检出CDK4或CCND1基因扩增的癌症患者。
  6. 如权利要求1-5中任一项所述的应用,其特征在于,所述抗PD-1抗体轻链互补决定区(LCDR)的氨基酸序列如SEQ ID NO:1、2和3所示,重链互补决定区(HCDR)的氨基酸序列如SEQ ID NO:4、5和6所示。
  7. 如权利要求6所述的应用,其特征在于,所述抗PD-1抗体包含轻链可变区(VL)和重链可变区(VH),其中,VL的氨基酸序列如SEQ ID NO:7所示,VH的氨基酸序列如SEQ ID NO:8所示。
  8. 如权利要求6所述的应用,其特征在于,所述抗PD-1抗体是包含轻链和重链的抗PD-1抗体,且轻链包含SEQ ID NO:9所示的氨基酸序列,重链包含SEQ ID NO:10所示的氨基酸序列。
  9. 如权利要求1-5任一项所述的应用,其特征在于,所述抗PD-1抗体选自nivolumab、pembrolizumab、toripalimab、Sintilimab、Camrelizumab、Tislelizumab、Cemiplimab中的一种或几种;优选toripalimab。
  10. 如权利要求1-5任一项所述的应用,其特征在于,所述抗PD-1抗体是单克隆抗体或其抗原结合片段。
  11. 如权利要求5所述的应用,其特征在于,所述CDK4/6抑制剂包括帕博西尼、瑞博西尼或玻玛西林。
  12. 如权利要求1-11任一项所述的用途,其特征在于,所述抗PD-1抗体或其抗原结 合片段的单次施用剂量为约0.1mg/kg至约10.0mg/kg个体体重,例如约0.1mg/kg、约0.3mg/kg、约1mg/kg、约2mg/kg、约3mg/kg、约5mg/kg或10mg/kg个体体重,或选自约120mg至约480mg固定剂量,例如约120mg、240mg、360mg或480mg固定剂量。
  13. 如权利要求12所述的用途,其特征在于,所述抗PD-1抗体或其抗原结合片段的给药周期频率为约每一周一次、每两周一次、每三周一次、每四周一次或一个月一次,优选为每两周一次。
  14. 如权利要求12所述的用途,其特征在于,所述抗PD-1抗体或其抗原结合片段的单次施用剂量为1mg/kg个体体重、3mg/kg个体体重、10mg/kg个体体重、或240mg固定剂量、480mg固定剂量,以每两周一次施用。
  15. 如权利要求12所述的用途,其特征在于,所述抗PD-1抗体或其抗原结合片段以液体剂型例如注射剂,经胃肠外途径例如经静脉输注施用。
  16. 如权利要求12所述的用途,其特征在于,所述抗PD-1抗体或其抗原结合片段的给药周期可以为一周、二周、三周、一个月、两个月、三个月、四个月、五个月、半年或更长时间,任选地,每个给药周期的时间可以相同或不同,且每个给药周期之间的间隔可以相同或不同。
  17. 如权利要求1-8任一项所述的应用,其特征在于,所述癌症患者是肿瘤组织中检出BRAF基因突变的黑色素瘤患者,所述抗PD-1抗体是toripalimab。
  18. 如权利要求5所述的应用,其特征在于,所述应用为抗PD-1抗体或其抗原结合片段与CDK4/6抑制剂联用,且所述抗PD-1抗体为toripalimab,所述CDK4/6抑制剂包括帕博西尼、瑞博西尼或玻玛西林。
  19. 一种用于预测癌症患者单独施用抗PD-1抗体治疗效果的试剂盒,其特征在于,所述试剂盒包括:用于检测所述患者癌组织或外周血中BRAF基因突变的试剂,或用于检测所述患者癌组织或外周血中NRAS基因突变的试剂,或用于检测所述患者癌组织或外周血中CDK4或CCND1基因扩增的试剂。
  20. 用于检测患者癌组织或外周血中BRAF突变的试剂、或用于检测患者癌组织或外周血中NRAS突变的试剂或用于检测患者癌组织或外周血中CDK4或CCND1基因扩增的试剂在制备用于预测抗PD-1抗体或其抗原结合片段治疗黑色素瘤效果的试剂盒中的应用。
PCT/CN2021/076456 2020-02-13 2021-02-10 抗pd-1抗体在***中的用途 WO2021160151A1 (zh)

Priority Applications (3)

Application Number Priority Date Filing Date Title
CN202180013152.3A CN115052628A (zh) 2020-02-13 2021-02-10 抗pd-1抗体在***中的用途
EP21753897.4A EP4104856A1 (en) 2020-02-13 2021-02-10 Use of anti-pd-1 antibody in treatment of tumors
US17/904,096 US20230082898A1 (en) 2020-02-13 2021-02-10 Use of anti-pd-1 antibody in treatment of tumors

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010090829.0 2020-02-13
CN202010090829.0A CN113244386A (zh) 2020-02-13 2020-02-13 抗pd-1抗体在***中的用途

Publications (1)

Publication Number Publication Date
WO2021160151A1 true WO2021160151A1 (zh) 2021-08-19

Family

ID=77219857

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/076456 WO2021160151A1 (zh) 2020-02-13 2021-02-10 抗pd-1抗体在***中的用途

Country Status (4)

Country Link
US (1) US20230082898A1 (zh)
EP (1) EP4104856A1 (zh)
CN (2) CN113244386A (zh)
WO (1) WO2021160151A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2022242621A1 (zh) * 2021-05-17 2022-11-24 上海君实生物医药科技股份有限公司 治疗完全切除黏膜黑色素瘤的患者的药物及方法

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1200737A (zh) * 1995-09-06 1998-12-02 艾德药品公司 用于人治疗的重组抗cd4抗体
CN101137394A (zh) * 2004-10-25 2008-03-05 默克制药公司 抗addl抗体及其应用
CN104250302A (zh) * 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 抗pd-1抗体及其应用
US20190038763A1 (en) * 2016-01-28 2019-02-07 INSERM (Institute National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of cancer
US20190248774A1 (en) * 2018-02-15 2019-08-15 GiraFpharma LLC Heterocyclic compounds as kinase inhibitors

Patent Citations (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1200737A (zh) * 1995-09-06 1998-12-02 艾德药品公司 用于人治疗的重组抗cd4抗体
CN101137394A (zh) * 2004-10-25 2008-03-05 默克制药公司 抗addl抗体及其应用
CN104250302A (zh) * 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 抗pd-1抗体及其应用
WO2014206107A1 (zh) 2013-06-26 2014-12-31 上海君实生物医药科技有限公司 抗pd-1抗体及其应用
US20190038763A1 (en) * 2016-01-28 2019-02-07 INSERM (Institute National de la Santé et de la Recherche Médicale) Methods and pharmaceutical composition for the treatment of cancer
US20190248774A1 (en) * 2018-02-15 2019-08-15 GiraFpharma LLC Heterocyclic compounds as kinase inhibitors

Non-Patent Citations (19)

* Cited by examiner, † Cited by third party
Title
AHMADZADEH, M ET AL., BLOOD, vol. 114, pages 1537 - 44
CHI, Z. ET AL., BMC CANCER, vol. 11, 2011, pages 85
CHO, J ET AL., INVEST NEW DRUGS, vol. 34, 2016, pages 677 - 84
CHO, J ET AL., INVEST NEW DRUGS, vol. 34, pages 677 - 684
CHOTHIA ET AL., JMOL. BIOL., vol. 196, 1987, pages 901 - 917
CHOTHIA ET AL., NATURE, vol. 341, 1989, pages 878 - 883
E.A. ET AL., EUR. J CANCER, vol. 45, 2009, pages 228 - 247
FURNEY, S.J. ET AL., PIGMENT CELL MELANOMA RES, vol. 27, 2014, pages 835 - 8
KABAT ET AL., J. BIOL. CHEM., vol. 252, 1977, pages 6609 - 6616
KABAT ET AL.: "Sequences of Proteins of Immunological Interest", 1991, NATIONAL INSTITUTES OF HEALTH
KABAT, ADV. PROT. CHEM., vol. 32, 1978, pages 1 - 75
KATHLEEN M. MAHONEY, GORDON J. FREEMAN, DAVID F. MCDERMOTT: "The Next Immune-Checkpoint Inhibitors: PD-1/PD-L1 Blockade in Melanoma", CLINICAL THERAPEUTICS, vol. 37, no. 4, 1 April 2015 (2015-04-01), US, pages 764 - 782, XP055285031, ISSN: 0149-2918, DOI: 10.1016/j.clinthera.2015.02.018 *
KHATTAK MUHAMMAD A., REID ANNA, FREEMAN JAMES, PEREIRA MICHELLE, MCEVOY ASHLEIGH, LO JOHNNY, FRANK MARKUS H., MENIAWY TAREK, DIDAN: "PD‐L1 Expression on Circulating Tumor Cells May Be Predictive of Response to Pembrolizumab in Advanced Melanoma: Results from a Pilot Study", THE ONCOLOGIST, vol. 25, no. 3, 1 March 2020 (2020-03-01), XP055835327, ISSN: 1083-7159, DOI: 10.1634/theoncologist.2019-0557 *
MCLAUGHLIN ET AL., CANCER, vol. 103, no. 5, 1 March 2005 (2005-03-01), pages 1000 - 1007
TAUBE, J. M. ET AL., SCI TRANSL MED, vol. 4, 2012, pages 127 - 37
THOMPSON, R. H. ET AL., PNAS, vol. 101, no. 49, 2004, pages 17174 - 17179
TOPALIAN, S. L. ET AL., NEW ENG. J. MED., vol. 366, no. 26, 2012, pages 2443 - 2454
W. A. WEBER, J. NUCL. MED., vol. 50, 2009, pages 1S - 10S
WHO DRUG INFORMATION, vol. 32, no. 2, 2018, pages 372 - 373

Also Published As

Publication number Publication date
EP4104856A1 (en) 2022-12-21
CN113244386A (zh) 2021-08-13
CN115052628A (zh) 2022-09-13
US20230082898A1 (en) 2023-03-16

Similar Documents

Publication Publication Date Title
CN110882385B (zh) 抗pd-1抗体在***中的用途
JP2017508785A (ja) 癌を治療するためのpd−1アンタゴニストおよびido1阻害剤の組み合わせ
KR20200119844A (ko) 항-pd-1 항체를 사용한 암의 치료 방법
US11572405B2 (en) Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
KR20200119845A (ko) 항 pd-1 항체 및 항 ctla4 항체를 사용한 암의 치료 방법
US20230133118A1 (en) Compositions and methods for treating cancer
WO2020211804A1 (zh) 抗pd-1抗体在制备治疗实体瘤的药物中的用途
WO2021160152A1 (zh) 抗pd-1抗体在治疗神经内分泌瘤中的用途
WO2021160151A1 (zh) 抗pd-1抗体在***中的用途
WO2021155840A1 (zh) 抗pd-1抗体在治疗恶性肿瘤中的用途
US20230322930A1 (en) Use of an anti-pd-1 antibody and a cytotoxic anticancer drug in treatment of non-small cell lung cancer
CN115364209A (zh) 抗pd-1抗体联合化疗治疗食管癌的用途
CN114099663A (zh) 抗pd-1抗体在治疗鼻咽癌中的用途
US20230365691A1 (en) Use of anti-pd-1 antibody in treatment of nasopharyngeal carcinoma
WO2023208001A1 (zh) 抗pd-1抗体和抗egfr抗体组合及其在治疗头颈部鳞癌中的用途
WO2024015463A1 (en) Combination therapy

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 21753897

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021753897

Country of ref document: EP

Effective date: 20220913