CN115364208A - Medicine and method for treating patient with completely resected mucosal melanoma - Google Patents

Medicine and method for treating patient with completely resected mucosal melanoma Download PDF

Info

Publication number
CN115364208A
CN115364208A CN202210540589.9A CN202210540589A CN115364208A CN 115364208 A CN115364208 A CN 115364208A CN 202210540589 A CN202210540589 A CN 202210540589A CN 115364208 A CN115364208 A CN 115364208A
Authority
CN
China
Prior art keywords
antibody
antigen
weeks
binding fragment
mucosal melanoma
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202210540589.9A
Other languages
Chinese (zh)
Inventor
姚盛
冯辉
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Junshi Biosciences Co Ltd
Original Assignee
Shanghai Junshi Biosciences Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Junshi Biosciences Co Ltd filed Critical Shanghai Junshi Biosciences Co Ltd
Publication of CN115364208A publication Critical patent/CN115364208A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/2803Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants against the immunoglobulin superfamily
    • C07K16/2827Immunoglobulins [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 B7 molecules, e.g. CD80, CD86
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • 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

Landscapes

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

Abstract

The invention relates to a medicine and a method for treating a patient with completely resected mucosal melanoma, in particular to application of an anti-PD-1 antibody or an antigen-binding fragment thereof in preparation of a medicine for treating the patient with completely resected mucosal melanoma or preventing recurrence or distant metastasis of the patient with the mucosal melanoma. The invention also relates to a kit for treating a patient who has had a mucosal melanoma completely resected or for preventing recurrence or distant metastasis of the mucosal melanoma in the patient. The medicine and the method can obviously prolong the RFS of a PDL1 expression positive patient and have better tolerance.

Description

Medicine and method for treating patient with completely resected mucosal melanoma
Technical Field
The invention relates to a medicine and a method for treating a patient with completely resected mucosal melanoma, in particular to application of an anti-PD-1 antibody or an antigen-binding fragment thereof in preparing a medicine for treating a patient with completely resected mucosal melanoma or preventing recurrence or distant metastasis of the patient's mucosal melanoma.
Background
Although Mucosal Melanoma is a rare Melanoma subtype in western countries, accounting for only 1.4% of all melanomas (Lian B, cui CL, zhou L, et al. The natural history and patterns of tumors from Mucosal melanals: an Analysis of 706 pro-specific-followed patterns 2017. Ang Oncol 20128-873), in asia Mucosal Melanoma ranks second in all Melanoma profiling, accounting for 22.6% of this type of cases (Cui C, lian B, zhou L, et al. Multifactorial Analysis of pathological Factors and Survival tissues Among 706 Mucosal Melanoma. Mucosal melanoma has a very poor prognosis compared to cutaneous melanoma. Previous studies have shown that the 5-year Overall Survival (OS) of cutaneous melanomas is about 50-80%, but that of Mucosal melanomas is only 25% (Lian B, cui CL, zhou L, et al. The natural history and patterns of tumors from Mucosal melanals: an Analysis of 706 generative-followed tissues. Ann Oncol 2017 868-873 Cui C, lian B, zhou L, et al. Multifactorial Analysis of pathological Factors and viral tissues Among 706 Mucosal biology. Ann Surg 2018. Also, early mucosal melanoma is prone to relapse after resection, and one previous study showed that post-operative resected mucosal melanoma patients have a median RFS of only 5.4 months (Lian B, si L, cui C, et al phase II randomised tertiary composition high-dose IFNalpha2B with a temozolomide plus as a system additional therapy for the reset mucosal tumor Cancer cutter 2013. Therefore, there is an unmet medical need for systemic adjuvant therapy for resected mucosal melanoma (restricted mucosal melanoma).
Tereprinimab (Torpillar or JS 001), an anti-programmed cell death receptor 1 (PD-1), humanized IgG4 monoclonal antibody (mAb), has been approved for second line therapy of metastatic melanoma, second line therapy of metastatic urothelial cancer, and third line therapy for recurrent or metastatic nasopharyngeal cancer in China (Mai HQ, chen QY, chen D, et al, ipaliminar or plasma plus chemotherpy as first-line therapy in advanced nasopharyngeal darcinoma: a multicenter randized phase 3 tertiary. Nat Med 2021 27 1536-3. Tereprinimab in the pre-Ib stage study, combined with vascular endothelial growth factor receptor inhibitor axitinib, showed superior anti-tumor activity and was useful as first line therapy for metastatic melanoma.
Previous clinical trials have shown that High Doses of Interferon (HDI) (IFN) - α 2b can extend the recurrence-free survival (RFS) and/or OS (Jonash E, kumar UN, linete GP, et al, additional high-dose interferon alfa-2b in tissues with high-risk melanoma. Cancer J2000: 139-145 Kirkwood JM, ibrahim JG, sosman JA, et al, high-dose interferon alfa-2b signaling mutants relax-free and overlay expressed with 2-KLH/GM-21 vaccine tissue with III-microstructure, 951J 2380, and J11, respectively, wherein. In addition, several phase III clinical trials on cytotoxic T lymphocyte-associated protein 4 (CTLA-4) and PD-1 monoclonal antibodies also demonstrated their efficacy as adjuvant therapy for the treatment of Resected high-risk cutaneous melanoma (Eggermont AMM, blank CU, mandala M, et al.Adjuvant Pembrolizumab versatilis plasma in selected Stage III Melanoma.N Engl J Med 2018. Although high doses of interferon are currently the only approved treatment for adjuvant treatment of melanoma, it is not generally accepted as a therapeutic standard. The growing evidence surrounding questionable survival benefits, high incidence of severe toxicity, and negligible benefits for patients with more disease, makes it an unattractive treatment for most patients and clinicians (Schuchter L. Adjuvant Interferon Therapy for Melanoma: high-Dose, low-Dose, no Dose, whichDose J Clin Oncol 2004. Therefore, there is a need for more effective treatments with acceptable safety profile in adjuvant therapy.
Disclosure of Invention
The invention provides an application of an anti-PD-1 antibody or an antigen-binding fragment thereof in preparing a medicament for treating a patient with completely resected mucosal melanoma or preventing recurrence or distant metastasis of the mucosal melanoma of the patient.
In yet another aspect, the present invention provides a method of aiding in the treatment of a patient having a completely resected mucosal melanoma or preventing recurrence or distant metastasis of a mucosal melanoma in the patient, comprising administering to the patient an effective amount of an anti-PD-1 antibody or antigen-binding fragment thereof of the present invention or a pharmaceutical composition thereof.
In yet another aspect, the invention provides an anti-PD-1 antibody or antigen-binding fragment thereof for use in the adjuvant treatment of a patient having a completely resected mucosal melanoma or for the prevention of recurrence of mucosal melanoma or distant metastasis in such a patient.
In one or more embodiments, an anti-PD-1 antibody or antigen-binding fragment thereof of the present invention is any antibody or antigen-binding fragment that specifically binds to PD-1 and blocks the binding of PD-1 to its ligand, PD-L1.
In one or more embodiments, the mucosal melanoma of the present invention is head and neck mucosal melanoma or non-head and neck mucosal melanoma.
In one or more embodiments, the mucosal melanoma according to the present invention is a mucosal melanoma positive for PD-L1 expression in tumor tissue section immunohistochemical staining analysis or a mucosal melanoma negative for PD-L1 expression in tumor tissue section immunohistochemical staining analysis; preferably mucosal melanoma positive for PD-L1 expression in immunohistochemical staining analysis of tumor tissue sections.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention comprises an amino acid sequence as set forth in SEQ ID NO: 1.2 and 3, and the amino acid sequence is as shown in SEQ ID NO: 4. 5 and 6.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof according to the invention comprises a light chain variable region having an amino acid sequence as set forth in SEQ ID NO. 7 and a heavy chain variable region having an amino acid sequence as set forth in SEQ ID NO. 8.
In one or more embodiments, the anti-PD-1 antibody of the invention comprises a light chain having an amino acid sequence as set forth in SEQ ID NO. 9 and a heavy chain having an amino acid sequence as set forth in SEQ ID NO. 10.
In one or more embodiments, the anti-PD-1 antibody of the invention is selected from one or more of nivolumab, pembrolizumab, toreplilimuab, sintillimuab, camrelizumab, tislelizumab, cemiplimab; preferably a toriplalimab.
In one or more embodiments, the mucosal melanoma according to the invention is head and neck mucosal melanoma, which use further comprises the combined administration of one or more therapies to an individual in need thereof; preferably, the therapy is radiation therapy.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the invention is administered at a dose of about 0.1mg/kg to about 10.0mg/kg of the individual's body weight, e.g., about 0.1mg/kg, about 0.3mg/kg, about 1mg/kg, about 2mg/kg, about 3mg/kg, about 5mg/kg, or 10mg/kg of the individual's body weight, or selected from a fixed dose of about 120mg to about 480mg, e.g., a fixed dose of about 120mg, 240mg, 360mg, or 480mg, preferably a fixed dose of about 3mg/kg of the individual's body weight or about 240 mg.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention is administered at a frequency of about once per week, once per two weeks, once per three weeks, once per four weeks, or once a month, preferably once per two weeks.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the invention is administered at a dose of 1mg/kg, 3mg/kg, 10mg/kg, or 240mg fixed dose, 480mg fixed dose once every two or three weeks.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention is administered at a dose of 3mg/kg body weight of the individual, or a fixed dose of 240mg, once every two weeks.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention is administered in a liquid dosage form, e.g., an injection, by a parenteral route, e.g., by intravenous infusion.
In one or more embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention is administered for a period of one week, two weeks, three weeks, one month, two months, three months, four months, five months, half a year, one year, or more, optionally, the time of each administration period is the same or different, and the interval between each administration period is the same or different.
In one or more embodiments, the radiation therapy dose of the present invention is CTV TB 60-64 Gy/30 times, 1-5 days per week for 6 weeks; preferably, the radiation therapy is administered concurrently within 6-8 weeks of administration of the anti-PD-1 antibody or antigen-binding fragment thereof.
In yet another aspect, the invention provides a combination therapy comprising administering to a patient in need thereof an anti-PD-1 antibody or antigen-binding fragment thereof described herein or a pharmaceutical composition thereof and one or more additional therapies.
In one or more embodiments, the additional therapy of the invention is selected from a chemotherapeutic agent, a biologic therapeutic agent, an immunogenic agent, an immunostimulatory cytokine, an encoded immunostimulatory cytokine, or radiation therapy.
In one or more embodiments, the pharmaceutical compositions of the invention comprise an anti-PD-1 antibody or antigen-binding fragment thereof described herein, and a pharmaceutically acceptable carrier or excipient.
In one or more embodiments, the methods of the present invention have fewer tertiary and above treatment-related side effects (TEAEs) than high-dose interferon alpha-2 b (HDI) adjunctive therapy methods. Preferably, tertiary and above treatment-related side effects (TEAEs) are about 60%,50%,40%,30%,20%, or 10% lower than HDI.
In one or more embodiments, the methods of the invention have fewer tertiary and above treatment-related side effects (TEAEs) than high-dose interferon alpha2b (HDI) adjunctive therapy. Preferably, tertiary and above treatment-related side effects (TEAEs) are about 10% -60%,10% -50%,10% -40%,10% -30%, or 10% -20% lower than HDI.
In one or more embodiments, the adjunctive therapy methods of the invention have superior efficacy to high dose interferon alpha-2 b (HDI) adjunctive therapy methods. Preferably, the Relapse Free Survival (RFS) of a PD-L1 expression positive patient is about 50%,40%,30%,20% or 10% higher than HDI.
In one or more embodiments, the adjunctive therapy methods of the invention may be more efficacious than high dose interferon alpha-2 b (HDI) adjunctive therapy methods. Preferably, the Relapse Free Survival (RFS) of a patient positive for PD-L1 expression is about 10-50%,10-40%,10-30%, or 10-20% higher than HDI.
In yet another aspect, the invention provides a kit comprising one or more single pharmaceutical dosage units of an anti-PD-1 antibody or antigen-binding fragment thereof, or a pharmaceutical composition thereof, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is as described herein. The kit of the invention may be used to treat a patient who has had a mucosal melanoma completely resected or to prevent recurrence or distant metastasis of a mucosal melanoma in the patient.
In yet another aspect, the present invention provides methods for predicting the efficacy of an anti-PD-1 antibody, or an antigen-binding fragment thereof, using a biomarker to treat a patient who has had mucosal melanoma completely resected to prevent recurrence or distant metastasis of mucosal melanoma in the patient.
Drawings
FIG. 1: the Kaplan-Meier curves for relapse free survival time (RFS) were analyzed for all the populations.
FIG. 2: each subgroup had no recurrence time to survival (RFS) risk ratio (HR).
FIG. 3: the overall survival time (OS) risk ratio (HR) for each subgroup.
FIG. 4: kaplan-Meier curves for subgroup relapse free survival time (RFS): a) RFS of PD-L1 positive tumor patients; b) RFS in PD-L1 negative tumor patients.
FIG. 5: the current study flow chart.
Detailed Description
The present invention relates to a method of treatment of completely resected mucosal melanoma. The methods of the invention comprise administering to a patient in need thereof an anti-PD-1 antibody or antigen-binding fragment thereof described herein.
Term(s) for
In order that the invention may be more readily understood, certain technical and scientific terms are defined below. Unless otherwise defined elsewhere herein, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs.
By "administering," "administering," and "treating" is meant introducing a composition comprising a therapeutic agent into a subject using any of a variety of methods or delivery systems known to those skilled in the art. Routes of administration of the anti-PD-1 antibody include intravenous, intramuscular, subcutaneous, peritoneal, spinal or other parenteral routes of administration, such as injection or infusion. "parenteral administration" refers to modes of administration other than enteral or topical administration, typically by injection, including, but not limited to, intravenous, intramuscular, intraarterial, intrathecal, intralymphatic, intralesional, intracapsular, intraframe, intracardiac, intradermal, intraperitoneal, transtracheal, subcutaneous, subcuticular, intraarticular, subcapsular, subarachnoid, intraspinal, epidural, and intrasternal injection and infusion, and via in vivo electroporation.
An "adverse effect" (AE) as referred to herein is any adverse and often unintentional or undesirable sign, symptom or disease associated with the use of medical treatment. For example, adverse reactions may be associated with activation of the immune system or expansion of immune system cells in response to therapy. The medical treatment may have one or more related AEs, and each AE may have the same or a different severity level.
"tumor burden" refers to the total amount of tumor mass distributed throughout the body. Tumor burden refers to the total number of cancer cells or the total size of the tumor throughout the body. Tumor burden can be determined by a variety of methods known in the art, such as measuring its size using calipers after the tumor is removed from the subject, or while in vivo using imaging techniques such as ultrasound, bone scans, computed Tomography (CT), or Magnetic Resonance Imaging (MRI) scans.
The term "tumor size" refers to the total size of a tumor, which can be measured as the length and width of the tumor. Tumor size can be determined by a variety of methods known in the art, for example, measuring its dimensions using calipers after the tumor is removed from the subject, or while in vivo using imaging techniques such as bone scans, ultrasound, CT, or MRI scans.
The terms "subject", "individual", "object" include any organism, preferably an animal, more preferably a mammal (e.g., rat, mouse, dog, cat, rabbit, etc.), and most preferably a human. The terms "subject" and "patient" are used interchangeably herein.
"antibody" as used herein refers to any form of antibody that achieves the desired biological or binding activity. It is therefore used in its broadest sense, but is not limited to, monoclonal, polyclonal, multispecific, humanized full-length human, chimeric and camel-derived single domain antibodies. An "antibody" specifically binds an antigen and comprises at least two heavy (H) chains and two light (L) chains interconnected by disulfide bonds. Each heavy chain comprises a heavy chain variable region (VH) and a heavy chain constant region comprising three constant domains CH1, CH2 and CH3. Each light chain comprises a light chain variable region (VL) and a light chain constant region comprising a constant domain CL. The VH and VL regions can be further subdivided into hypervariable regions, termed Complementarity Determining Regions (CDRs), interspersed with regions that are more conserved, termed Framework Regions (FRs). Generally, from N-terminus to C-terminus, both light and heavy chain variable domains comprise FR1, CDR1, FR2, CDR2, FR3, CDR3 and FR4. Amino acids are typically assigned to each domain according to the following definitions: sequences of Proteins of Immunological Interest, kabat et al; national Institutes of Health, bethesda, md.; version 5; NIH publication No. 91-3242 (1991): kabat (1978) adv.prot.chem.32:1-75; kabat et al, (1977) j.biol.chem.252:6609-6616; chothia et al, (1987) J mol. Biol.196:901-917 or Chothia et al, (1989) Nature 341.
The carboxy-terminal portion of the heavy chain may define a constant region primarily responsible for effector function. Generally, human light chains are classified into kappa chains and lambda chains. Human heavy chains are generally classified as mu, delta, gamma, alpha, or epsilon, and define the antibody's isotype as IgM, igD, igG, igA, and IgE, respectively. The IgG subclasses are well known to those skilled in the art and include, but are not limited to, igG1, igG2, igG, and IgG4.
The term "antibody" includes: naturally occurring and non-naturally occurring abs; monoclonal and polyclonal Ab; chimeric and humanized abs; human or non-human Ab; ab is fully synthesized; and single chain abs. Non-human abs may be humanized by recombinant methods to reduce their immunogenicity in humans.
Unless specifically indicated otherwise, "antibody fragment" or "antigen-binding fragment" as used herein refers to an antigen-binding fragment of an antibody, i.e., an antibody fragment that retains the ability of a full-length antibody to specifically bind to an antigen, e.g., a fragment that retains one or more CDR regions. Examples of antigen binding fragments include, but are not limited to, fab ', F (ab') 2, and Fv fragments; a diabody; a linear antibody; a single chain antibody molecule; nanobodies and multispecific antibodies formed from antibody fragments.
"chimeric antibody" refers to antibodies and fragments thereof as follows: wherein a portion of the heavy and/or light chain is identical to or homologous to corresponding sequences in an antibody derived from a particular species (e.g., human) or belonging to a particular antibody class or subclass, and the remainder of the chain is identical to or homologous to corresponding sequences in an antibody derived from another species (e.g., mouse) or belonging to another antibody class or subclass, so long as it exhibits the desired biological activity.
"human antibody" refers to an antibody comprising only human immunoglobulin sequences. A human antibody may contain murine carbohydrate chains if it is produced in a mouse, mouse cells, or a hybridoma derived from a mouse cell. Similarly, "mouse antibody" or "rat antibody" refers to an antibody that comprises only mouse or rat immunoglobulin sequences, respectively.
"humanized antibody" refers to antibody forms containing sequences from non-human (e.g., murine) antibodies as well as human antibodies. Such antibodies contain a minimal sequence derived from a single side of a non-human immunoglobulin. Typically, the humanized antibody will comprise substantially all of at least one and typically two variable domains, in which all or substantially all of the hypervariable loops correspond to those of a non-human immunoglobulin and all or substantially all of the FR regions are those of a human immunoglobulin. The humanized antibody optionally further comprises at least a portion of an immunoglobulin constant region (Fc), typically a human immunoglobulin constant region.
The term "immunotherapy" refers to the treatment of a subject suffering from a disease or at risk of infection or suffering from a relapse of a disease by a method that includes inducing, enhancing, suppressing or otherwise modifying an immune response. By "treatment" or "therapy" of a subject is meant any type of intervention or process performed on the subject, or the administration of an agent to the subject, with the purpose of reversing, alleviating, ameliorating, slowing or preventing the onset, progression, severity, or recurrence of a symptom, complication, or condition, or biochemical indicator associated with the disease.
"programmed death receptor-1 (PD-1)" refers to an immunosuppressive receptor belonging to the CD28 family. PD-1 is expressed predominantly on previously activated T cells in vivo and binds to both ligands PD-L1 and PD-L2. The term "PD-1" as used herein includes human PD-1 (hPD-1), variants, isoforms, and species homologs of hPD-1, and analogs having at least one common epitope with hPD-1.
A "therapeutically effective amount" or "therapeutically effective dose" of a drug or therapeutic agent is any amount of the drug that, when used alone or in combination with another therapeutic agent, protects a subject from the onset of a disease or promotes disease regression as evidenced by a decrease in the severity of disease symptoms, an increase in the frequency and duration of asymptomatic phases of a disease, or the prevention of injury or disability resulting from the affliction of a disease. The ability of a therapeutic agent to promote disease regression can be evaluated using a variety of methods known to those skilled in the art, such as in human subjects during clinical trials, in animal model systems that predict human efficacy, or by assaying the activity of the agent in an in vitro assay.
A therapeutically effective amount of a drug includes a "prophylactically effective amount," i.e., any amount of a drug that inhibits the development or recurrence of cancer when administered to a subject at risk of developing cancer or a subject having cancer recurrence, either alone or in combination with an antineoplastic agent.
"biotherapeutic agent" refers to a biological molecule, such as an antibody or fusion protein, that blocks ligand/receptor signaling in any biological pathway that supports tumor maintenance and/or growth or inhibits an anti-tumor immune response.
As used herein, unless expressly indicated otherwise, "CDR" means that the immunoglobulin variable region is a complementarity determining region defined using the Kabat numbering system.
A "therapeutic anti-PD-1 monoclonal antibody" refers to an antibody that specifically binds to the mature form of a particular PD-1 expressed on the surface of certain mammalian cells. Mature PD-1 lacks a pre-secretory leader sequence, or leader peptide. The terms "PD-1" and "mature PD-1" are used interchangeably herein and, unless otherwise specifically defined or clear from context, should be understood to be the same molecule.
As used herein, a therapeutic anti-human PD-1 antibody or anti-hPD-1 antibody refers to a monoclonal antibody that specifically binds to mature human PD-1.
As used herein, "framework region" or "FR" refers to immunoglobulin variable regions that do not include CDR regions.
An "isolated antibody or antigen-binding fragment thereof" refers to a molecule that is in a purified state and in this case is designated as containing substantially no other biological molecules, such as nucleic acids, proteins, lipids, carbohydrates, or other materials (such as cell debris or growth media).
By "patient," "patient," or "subject" is meant any single subject, typically a mammal, including humans and other mammals, such as horses, cows, dogs, or cats, in need of a medical procedure or participating in a clinical trial, epidemiological study, or as a control.
The "RECIST 1.1 efficacy standard" as described herein refers to the definition described by Eisenhauver et al, e.a. et al, eur.j Cancer 45-247 (2009) for target lesions or non-target lesions based on the background of the measured response. Prior to immunotherapy, it is the most common standard for efficacy assessment of solid tumors. However, with the advent of the immune age, many problems which have not been found in the tumor evaluation before, so that in 2016, based on the newly appeared phenomenon caused by immunotherapy, RECIST working group provides a new judgment standard after correcting the existing "RECIST v.1.1", namely the "irRECIST standard" described herein, aiming at better evaluating the curative effect of immunotherapy drugs.
The term "ECOG" score is an indicator of a patient's general health and ability to tolerate treatment, as measured by their physical strength. ECOG physical performance scoring criteria score: 0 minute, 1 minute, 2 minutes, 3 minutes, 4 minutes and 5 minutes. A score of 0 means that the motility was completely normal and had no difference from the motility before onset. A score of 1 means free to walk and engage in light physical activities, including general housework or office work, but not heavier physical activities.
By "sustained response" is meant a sustained therapeutic effect following cessation of treatment with a therapeutic agent or combination therapy as described herein. In some embodiments, the sustained response has a duration that is at least the same as the duration of treatment or at least 1.5, 2.0, 2.5, or 3 times the duration of treatment.
"tissue section" refers to a single portion or piece of a tissue sample, such as a thin slice of tissue cut from a sample of normal tissue or a tumor.
As used herein, "treating" cancer refers to administering a treatment regimen described herein (e.g., administration of an anti-PD-1 antibody) to a subject having or diagnosed with cancer to achieve at least one positive therapeutic effect (e.g., a decrease in the number of cancer cells, a decrease in tumor volume, a decrease in the rate of cancer cell infiltration into peripheral organs, or a decrease in the rate of tumor metastasis or tumor growth). Positive treatment effects in cancer can be measured in a variety of ways (see w.a.weber, j.nucl.med., 50. For example, for tumor growth inhibition, T/C ≦ 42% is the minimum level of anti-tumor activity according to the NCI standard. T/C (%) = median treated tumor volume/median control tumor volume x 100.PFS (also called "time to tumor progression") refers to the length of time during and after treatment during which cancer does not grow, and includes the amount of time a patient experiences CR or PR and the amount of time a patient experiences SD. DFS refers to the length of time during and after treatment that a patient is still disease free. OS refers to an extension of life expectancy compared to an initial or untreated individual or patient. The treatment regimen for a combination of the invention effective in treating a cancer patient can vary depending on a variety of factors such as the disease state, age, weight of the patient and the ability of the therapy to elicit an anti-cancer response in the subject. Although embodiments of the invention may not achieve an effective positive therapeutic effect in each subject, a positive therapeutic effect should be effective and achieved in a statistically significant number of subjects.
The terms "mode of administration", "dosing regimen", which are used interchangeably, refer to the dosage and time of use of each therapeutic agent in the combination of the invention.
The term "Immunohistochemistry (IHC)" refers to a method for determining tissue intracellular antigens (polypeptides and proteins) and performing localized, qualitative and relatively quantitative studies thereof by developing color-developing agents (fluorescein, enzyme, metal ion, isotope) that label antibodies through chemical reaction using the principle that antigens are specifically bound to antibodies. In some embodiments of the invention, a tumor tissue sample from a subject is subjected to PD-L1 detection using a staining experiment with Roche anti-human PD-L1 antibody SP142 (Cat No: M4422) prior to treatment with an anti-PD-1 antibody. In some embodiments, a tumor cell has a membrane staining intensity of ≧ 1% is defined as PD-L1 positive.
Herein, the term "cancer" or "malignancy" refers to a wide variety of diseases characterized by uncontrolled growth of abnormal cells in the body. Unregulated cell division, growth division and growth lead to the formation of malignant tumors that invade adjacent tissues and may also metastasize to distal parts of the body through the lymphatic system or blood stream. Examples of cancers suitable for treatment or prevention using the methods, medicaments and kits of the invention include, but are not limited to, carcinoma, lymphoma, leukemia, blastoma and sarcoma. More specific examples of cancer include squamous cell cancer, 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, nasopharyngeal cancer, cervical cancer, brain cancer, gastric cancer, bladder cancer, hepatoma, breast cancer, colon cancer, and head and neck cancer.
As used herein, the term "Tumor Mutation Burden (TMB)" refers to the total number of somatic gene coding errors, base substitutions, gene insertion or deletion errors detected per million bases. In some embodiments of the invention, tumor Mutational Burden (TMB) is estimated by analysis of somatic mutations, including coding base substitutions and the megabase insertions of the panel sequences studied.
In the following paragraphs, various aspects of the present invention are described in further detail.
anti-PD-1 antibodies
Herein, a "PD-1 antibody" refers to any chemical compound or biological molecule that binds to the PD-1 receptor, blocks the binding of PD-L1 expressed on cancer cells to PD-1 expressed on immune cells (T, B, NK cells), and preferably also blocks the binding of PD-L2 expressed on cancer cells to PD-1 expressed on immune cells. Alternative names 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, B7-4, CD274, and B7-H; and for PD-L2 there are PDCD1L2, PDL2, B7-DC and CD273. In any of the inventive methods, medicaments and uses of treatment of a human individual, the PD-1 antibody blocks the binding of human PD-L1 to human PD-1, and preferably blocks the binding of both human PD-L1 and PD-L2 to human PD 1. The human PD-1 amino acid sequence can be found at NCBI locus number: NP _005009. The amino acid sequences of human PD-L1 and PD-L2 can be found in the NCBI locus numbering, respectively: NP _054862 and NP _079515.
Herein, when referring to an "anti-PD-1 antibody," unless otherwise indicated or described, the term includes antigen-binding fragments thereof.
The anti-PD-1 antibody applicable to any application, therapy, medicament and kit disclosed by the invention is combined with PD-1 with high specificity and high affinity, blocks the combination of PD-L1/2 and PD-1 and inhibits PD-1 signal transduction, thereby achieving an immunosuppressive effect. In any of the uses, therapies, medicaments and kits disclosed herein, the anti-PD-1 antibody includes the full-length antibody itself, as well as antigen-binding portions or fragments that bind to the PD-1 receptor and exhibit functional properties similar to those of an intact Ab in inhibiting ligand binding and upregulating the immune system. In some embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is an anti-PD-1 antibody or antigen-binding fragment thereof that cross-competes for binding to human PD-1 with terieprinimab. In other embodiments, the anti-PD-1 antibody or antigen-binding fragment thereof is a chimeric, humanized, or human Ab or antigen-binding fragment thereof. In certain embodiments for treating a human subject, the Ab is a humanized Ab.
In some embodiments, the anti-PD-1 antibodies for any of the uses, therapies, medicaments and kits described herein include monoclonal antibodies (mabs) or antigen-binding fragments thereof that specifically bind to PD-1, and preferably specifically bind to human PD-1. The mAb may be a human, humanized or chimeric antibody and may include human constant regions. In some embodiments, the constant region is selected from the group consisting of human IgG1, igG2, igG3, and IgG4 constant regions; preferably, the anti-PD-1 antibodies or antigen-binding fragments thereof suitable for use in any of the uses, therapies, medicaments and kits described herein comprise a heavy chain constant region of human IgG1 or IgG4 isotype, more preferably a human IgG4 constant region. In some embodiments, the sequence of the IgG4 heavy chain constant region of the anti-PD-1 antibody or antigen-binding fragment thereof comprises a S228P mutation that replaces a serine residue in the hinge region with a proline residue that is typically present at the corresponding position of an IgG1 isotype antibody.
Preferably, in any one of the uses, therapies, medicaments and kits of the present invention, the PD-1 antibody is a monoclonal antibody or an antigen-binding fragment thereof, and the light chain CDRs thereof are SEQ ID NO: 1.2 and 3, and the heavy chain CDR is SEQ ID NO: 4. 5 and 6.
More preferably, in any one of the uses, therapies, medicaments and kits of the present invention, the PD-1 antibody is a specific binding to human PD-1 and comprises: (a) comprises SEQ ID NO:7, and (b) a light chain variable region comprising SEQ ID NO:8 in the heavy chain variable region of the antibody.
Further preferably, in any one of the uses, therapies, medicaments and kits of the present invention, the PD-1 antibody is a monoclonal antibody that specifically binds to human PD-1 and comprises: (a) comprises SEQ ID NO:9, and (b) a light chain comprising SEQ ID NO: 10.
Table a provides the amino acid sequence numbering of the light chain CDRs and heavy chain CDRs of an exemplary anti-PD-1 antibody mAb for use, therapy, medicament, and kit described herein, according to Kabat rules:
table a: light and heavy chain CDRs of exemplary anti-human PD-1 antibodies
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
VL SEQ ID NO:7
VH SEQ ID NO:8
LC SEQ ID NO:9
HC SEQ ID NO:10
Examples of anti-PD-1 antibodies that bind to human PD-1 and that can be used in the uses, therapies, medicaments and kits described in the present invention are described in WO 2014206107. Human PD-1 mabs that can be used as anti-PD-1 antibodies in the uses, therapies, medicaments and kits described in this invention include any of the anti-PD-1 antibodies described in WO2014206107, including: tereprinimab (Toriplizab), a humanized IgG4 mAb having the structure described in WHO Drug Information (Vol.32, phase 2, pp.372-373 (2018)) and comprising the light and heavy chain amino acid sequences shown in sequences SEQ ID NO. 9 and 10 in a preferred embodiment, the anti-PD-1 antibody useful in any of the uses, therapies, drugs and kits described herein is selected from the group consisting of humanized antibodies 38, 39, 41 and 48 described in WO2014206107 in a particularly preferred embodiment, the anti-PD-1 antibody useful in any of the uses, therapies, drugs and kits described herein is Tereprinimab.
anti-PD-1 antibodies useful in any of the uses, therapies, medicaments and kits described herein also include Nivolumab and Pembrolizumab, which have been approved by the FDA.
In certain embodiments, anti-PD-1 antibodies useful in any of the uses, therapies, medicaments and kits described herein also include anti-PD-L1 monoclonal antibodies that specifically bind PD-L1 to block the binding of PD-L1 to PD-1, such as nivolumab, pembrolizumab, torelizumab, sintillimumab, camrelizumab, tislelizumab, cemiplizumab.
"PD-L1" expression or "PD-L2" expression as described herein refers to any detectable expression level of a particular PD-L protein on the cell surface or a particular PD-L mRNA within a cell or tissue. PD-L protein expression can be detected in IHC analysis of tumor tissue sections or by flow cytometry using diagnostic PD-L antibodies. Alternatively, PD-L protein expression of tumor cells can be detected by PET imaging using a binding agent that specifically binds to a desired PD-L target (such as PD-L1 or PD-L2).
Methods for quantifying PD-L1 protein expression in IHC analysis of tumor tissue sections, see, but not limited to, thompson, r.h. et al, PNAS 101 (49): 17174-17179 (2004); taube, j.m. et al, sci Transl Med 4, 127ra37 (2012); and Toplian, S.L. et al, new Eng.J.Med.366 (26): 2443-2454 (2012), etc.
One method employs a simple binary endpoint of positive or negative PD-L1 expression, where positive results are defined as the percentage of tumor cells showing histological evidence of cell surface membrane staining. Counting tumor tissue sections to greater than 1% of total tumor cells is defined as positive for PD-L1 expression.
In another method, PD-L1 expression in tumor tissue sections is quantified in tumor cells as well as in infiltrating immune cells. The percentages of tumor cells and infiltrating immune cells that exhibit membrane staining were quantified individually at ≦ 1%, 1% to 50%, and then 50% up to 100%. For tumor cells, PD-L1 expression was counted as negative if the score was < 1%, and positive if the score > 1%.
In some embodiments, the PD-L1 expression level by malignant cells and/or by infiltrating immune cells within the tumor is determined to be "overexpressed" or "elevated" based on comparison to the PD-L1 expression level by an appropriate control. For example, the protein or mRNA expression level of control PD-L1 can be a level quantified in non-malignant cells of the same type or in sections from matched normal tissue.
In some embodiments, PD-L1 positive is defined as staining tumor cells CPS.gtoreq.1% with JS311 IHC. CPS represents the positive joint score. CPS criteria are: tumor cells with any intensity of membrane staining, membrane/cytoplasmic stained lymphocytes/macrophages directly associated with tumor cells, the fraction of such cells relative to tumor cells (at least one hundred), but should exclude staining of total necrotic cells, stromal cells, carcinoma in situ, and other immune cells (including but not limited to neutrophils, eosinophils, plasma cells), and the like.
Adjuvant therapy
As used herein, "adjuvant therapy" or "adjuvant therapy" is understood to be the administration of one or more drugs to a patient after surgical resection of one or more cancerous tumors in which all detectable and resectable disease (e.g., cancer) has been removed from the patient, but due to occult lesions, there is also a statistical risk of recurrence with the objective of reducing the likelihood or severity of recurrence or disease, or delaying the appearance of biological manifestations of disease recurrence.
Effective amount and excision
The term "effective amount" refers to an amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for example, by a researcher or clinician. Moreover, the term "therapeutically effective amount" refers to any amount that results in improved treatment, cure, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of progression of a disease or disorder, as compared to a corresponding subject that has not received that amount. The term also includes within its scope an amount effective to enhance normal physiological function. A therapeutically effective amount of a drug also includes a "prophylactically effective amount".
With respect to solid tumors, an effective amount includes an amount sufficient to cause tumor shrinkage and/or to reduce the rate of tumor growth (such as to inhibit tumor growth) or to delay the proliferation of other unwanted cells. In some embodiments, an effective amount is an amount sufficient to prevent or delay tumor recurrence. An effective amount may be administered in one or more administrations. The effective amount of the drug or composition may be: (ii) (i) reducing the number of cancer cells; (ii) reducing tumor size; (iii) Inhibit, delay, slow to a certain extent and can prevent cancer cells from infiltrating into peripheral organs; (iv) Inhibition, i.e. slowing down to a certain extent and may prevent tumor metastasis; (v) inhibiting tumor growth; (vi) preventing or delaying the occurrence and/or recurrence of a tumor; and/or (vii) relieving to some extent one or more symptoms associated with the cancer.
The term "resection" refers to the surgical removal of malignant tissue characteristic of melanoma from a human patient. Resection is understood to mean removal of malignant tissue such that the presence of remaining malignant tissue within the patient cannot be detected with existing methods. Resection is understood to mean the removal of melanoma such that the presence of residual melanoma in the patient cannot be detected. The resection may include a complete resection or a partial resection.
Recurrence and distant metastasis
The term "recurrence" refers to local recurrence of a tumor or melanoma, and refers to a patient who has had a tumor or melanoma excised/completely excised, with the tumor or melanoma reappearing within 2cm of the original tumor area. Local recurrence of melanoma may occur at any primary melanoma site, as well as in the lymph nodes in the area proximate to the primary melanoma.
The term "distant metastasis" refers to distant metastasis of a tumor or melanoma, and refers to a patient who has had a tumor or melanoma removed/completely removed, and which has reappeared the tumor or melanoma 2cm beyond the original tumor area, at which time the tumor or melanoma cells may spread to a distant site in the body, such as the lung, liver, etc.
Combination therapy, dosage and dosing regimen
The combination therapy of the invention may also comprise one or more additional therapies. The additional therapeutic agent can be, for example, a chemotherapeutic agent, a biotherapeutic agent, an immunogenic agent (e.g., an attenuated cancer cell, a tumor antigen, an antigen presenting cell (such as a dendritic cell pulsed with a tumor-derived antigen or nucleic acid), an immunostimulatory cytokine (e.g., IL-2, IFN-tumor, GM-CSF), a cell transfected with a gene encoding an immunostimulatory cytokine (such as, but not limited to, GM-CSF)), or radiation therapy.
The choice of a dosage regimen (also referred to herein as an administration regimen) for a medicament of the invention depends on several factors, including the rate of solid serum or tissue turnover, the level of symptoms, the overall immunogenicity, and the degree of accessibility of the target cells, tissues, or organs of the treated individual. Preferably, the dosing regimen maximizes the amount of each therapeutic agent delivered to the patient consistent with acceptable levels of side effects. Thus, the dosage and frequency of administration of each of the biotherapeutic and chemotherapeutic agents will depend in part on the particular therapeutic agent, the severity of the cancer being treated and the patient's characteristics. Guidance in selecting appropriate doses of antibodies, cytokines, and small molecules may be obtained. Determination of an appropriate dosage regimen may be made by a clinician, for example, with reference to parameters or factors known or suspected to affect treatment or expected to affect treatment in the art, and will depend on, for example, the patient's clinical history (e.g., previous treatment), the type and stage of cancer being treated, and biomarkers responsive to one or more therapeutic agents in the combination therapy.
Each therapeutic agent or therapy of the combination therapies of the invention can be administered simultaneously (i.e., in the same pharmaceutical composition), concurrently (i.e., in separate pharmaceutical formulations, one after the other in any order), or sequentially in any order. Sequential administration of therapeutic agents in combination therapy may be particularly useful when the agents may be in different dosage forms (one drug is a tablet or capsule and the other drug is a sterile liquid formulation) and/or on different dosing schedules (e.g., chemotherapeutic agents are administered at least daily and biologic therapeutic agents are administered less frequently (e.g., once per week, once every two weeks, or once every three weeks)).
In some embodiments, the therapeutic agents in at least one combination therapy are administered using the same dosage regimen (treatment dose, frequency, and duration) that is typically used when the agents are used to treat the same tumor as monotherapy. In other embodiments, the patient receives a lesser total amount of the at least one therapeutic agent in the combination therapy than when the agent is used as monotherapy, e.g., a smaller dose, less frequent dose, and/or shorter duration of treatment.
Each therapeutic agent in the combination therapy of the present invention may be administered orally or parenterally, which includes intravenous, intramuscular, intraabdominal, subcutaneous, rectal, topical and transdermal routes of administration.
The anti-PD-1 antibodies or antigen-binding fragments thereof of the invention can be administered by continuous infusion or by intermittent dosing. The single administration dose may range from about 0.01mg/kg to about 20mg/kg, from about 0.1mg/kg to about 10mg/kg of the individual's body weight, or from about 120mg to about 480mg fixed dose. For example, the dose may be about 0.1mg/kg, about 0.3mg/kg, about 1mg/kg, about 2mg/kg, about 3mg/kg, about 4mg/kg, about 5mg/kg, about 6mg/kg, about 7mg/kg, about 8mg/kg, about 9mg/kg, or about 10mg/kg of the individual's body weight. Or may be administered in a fixed dose of about 120mg, 240mg, 360mg or 480 mg. Dosing regimens are generally designed to achieve such exposure, which results in sustained Receptor Occupancy (RO) based on the typical pharmacokinetic properties of abs. A representative dosing regimen may be about once per week, about once every two weeks, about once every three weeks, about once every four weeks, about once a month, or longer. In some embodiments, the anti-PD-1 antibody is administered to the individual about once every two weeks.
In some embodiments, an anti-PD-1 antibody of the invention is tereprinimab in a single administration dose selected from about 1 to about 5mg/kg of the body weight of the individual. In some embodiments, a single administered dose of tereprinimab is selected from a dose of about 1mg/kg, 2mg/kg, 3mg/kg, 4mg/kg, and 5mg/kg body weight, or a fixed dose of about 120mg, 240mg, and 360mg, administered intravenously. In some preferred embodiments, the tereprinimab is administered as a liquid drug, and the selected dose of the drug is administered by intravenous infusion over a period of 30 to 60 minutes. In some embodiments, the tereprimab is administered by intravenous infusion at a fixed dose of about 3mg/kg or about 240mg once every two weeks (Q2W) over a period of 30 minutes.
In some embodiments, the mucosal melanoma is head and neck mucosal melanoma, and the radiation therapy is administered concurrently within 6-8 weeks of administration of the anti-PD-1 antibody or antigen-binding fragment thereof. The dose of the radiotherapy is CTVTB 60-64 Gy/30 times, and the radiotherapy is applied on the 1 st-5 th day of each week for 6 weeks.
The administration cycles of the anti-PD-1 antibody or antigen-binding fragment thereof of the present invention and chemotherapy are one week, two weeks, three weeks, one month, two months, three months, four months, five months, half a year or more, optionally, the time of each administration cycle may be the same or different, and the interval between each administration cycle may be the same or different. In some embodiments, one dosing/treatment cycle is two weeks. In some embodiments, the terieprinimab is administered at about 3mg/kg once every two weeks for a single dosing cycle. In some embodiments, one dosing/treatment cycle of radiation therapy is one week. In some embodiments, the dose of radiation therapy during a dosing cycle is CTVTB 60-64 Gy/30 times, administered 1-5 days per week. The anti-PD-1 antibody or antigen-binding fragment thereof of the invention is administered for a period of no more than 1 year. The radiation therapy of the invention is administered for 6 weeks, concurrently within 6-8 weeks from the anti-PD-1 antibody or antigen-binding fragment thereof.
Methods of treatment and uses
The invention aims to treat patients with completely resected mucosal melanoma and prevent recurrence or distant metastasis of the mucosal melanoma. The invention discovers that by administering an effective amount of any one of the anti-PD-1 antibodies or antigen-binding fragments thereof or a pharmaceutical composition containing the anti-PD-1 antibody or antigen-binding fragment thereof, recurrence or distant metastasis of mucosal melanoma can be effectively prevented in a patient who has completely resected mucosal melanoma, and the recurrence-free survival time RFS and/or the distant metastasis-free survival time DMFS of the patient can be improved; at the same time, the overall life span of the patient is improved. In some embodiments, the administration safety and tolerability of a patient is improved by administering an effective amount of any one of the anti-PD-1 antibodies or antigen-binding fragments thereof described herein or a pharmaceutical composition comprising the anti-PD-1 antibody or antigen-binding fragment thereof.
Accordingly, the present invention provides the use of an anti-PD-1 antibody or antigen-binding fragment thereof of the invention as hereinbefore described in the manufacture of a medicament for the prevention or treatment of a patient who has had mucosal melanoma completely resected or for the prevention of recurrence or distant metastasis of mucosal melanoma in such a patient.
The present invention also provides a method of preventing or treating a patient who has had mucosal melanoma completely resected or preventing recurrence or distant metastasis of mucosal melanoma in the patient, comprising administering to the patient an effective amount of an anti-PD-1 antibody or antigen-binding fragment thereof described herein. The effective amount includes a prophylactically effective amount and a therapeutically effective amount.
The present invention also provides the aforementioned anti-PD-1 antibody or antigen-binding fragment thereof of the present invention for use in treating a patient who has completely resected mucosal melanoma or preventing mucosal melanoma recurrence or distant metastasis in the patient.
The invention also provides for co-administering to a subject a therapeutically effective amount of one or more therapies (e.g., treatment modalities and/or other therapeutic agents). In some embodiments, the therapy comprises surgical treatment and/or radiation therapy.
As used herein, "completely resected mucosal melanoma" refers to complete resection of the primary lesion, with negative surgical margins.
In some embodiments, the methods or uses provided herein further comprise administering one or more therapies (e.g., treatment modalities and/or other therapeutic agents) to an individual in need thereof. The antibodies of the invention may be used alone or in combination with other therapeutic agents in therapy. For example, it may be co-administered with at least one additional therapeutic agent. In some embodiments, the methods or uses provided herein further comprise administering radiation therapy (radiotherapy) to an individual in need thereof.
In one or more embodiments, the mucosal melanoma of the present invention is head and neck mucosal melanoma or non-head and neck mucosal melanoma.
In one or more embodiments, the mucosal melanoma of the present invention is a mucosal melanoma with positive PD-L1 expression in tumor tissue section immunohistochemical staining analysis or a mucosal melanoma with negative PD-L1 expression in tumor tissue section immunohistochemical staining analysis; preferably mucosal melanoma positive for PD-L1 expression in immunohistochemical staining analysis of tumor tissue sections.
Preferably, the method, use or anti-PD-1 antibody according to any embodiment of the present invention is particularly suitable for mucosal melanoma positive for PD-L1 expression in immunohistochemical staining analysis of tumor tissue sections.
Preferred anti-PD-1 antibodies for use in treating a patient having a completely resected mucosal melanoma or preventing recurrence or distant metastasis of a mucosal melanoma in the patient may be as described in any embodiment herein, more preferably with the light chain CDRs of SEQ ID NO: 1.2 and 3, and the heavy chain CDR is the amino acid shown in SEQ ID NO: 4. 5 and 6, more preferably an antibody comprising the amino acids shown in SEQ ID NOs: 7 and the variable region of the light chain shown in SEQ ID NO:8, more preferably a monoclonal antibody comprising the heavy chain variable region of SEQ ID NO:9 and SEQ ID NO:10, more preferably the humanized antibodies 38, 39, 41 and 48 described in WO2014206107, most preferably terliplizumab.
In a particularly preferred embodiment, the present invention provides a method of treating a patient having a completely resected mucosal melanoma or preventing recurrence or distant metastasis of a mucosal melanoma in the patient, the method comprising administering to the patient a therapeutically or prophylactically effective amount of tereprinimab; preferably, the patient is positive for PD-L1 expression. In certain embodiments, the mucosal melanoma is a non-head and neck mucosal melanoma. In certain embodiments, the mucosal melanoma is head and neck mucosal melanoma, and the method further comprises administering radiation therapy concurrently within 6-8 weeks of administering tereprinimab.
In a particularly preferred embodiment, the invention provides the use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament for the treatment of a patient who has had a mucosal melanoma completely resected or for the prevention of recurrence or distant metastasis of a mucosal melanoma in such a patient. Preferably, the patients with completely resected mucosal melanoma have positive PD-L1 expression in immunohistochemical staining analysis of tumor tissue sections (CPS ≧ 1%). In certain embodiments, the mucosal melanoma is a non-head and neck mucosal melanoma. In certain embodiments, the mucosal melanoma is head and neck mucosal melanoma, and the use further comprises administering radiation therapy concurrently within 6-8 weeks of administering terliplizumab.
Medicine box
The invention also provides a kit containing one or more single pharmaceutical dosage units of an anti-PD-1 antibody or antigen-binding fragment thereof according to any one of the embodiments herein.
When present as separate formulations, each formulation may contain, in addition to the active ingredient, a pharmaceutically acceptable carrier.
The kit of the invention can be used for treating a patient who has completely resected mucosal melanoma or preventing recurrence or distant metastasis of the mucosal melanoma in the patient.
Abbreviations
Throughout the description and examples of the present invention, the following abbreviations are used:
BID one dose, 2 times daily
CDR complementarity determining region
Disease-free survival of DFS
FR framework regions
IgG immunoglobulin G
IHC immunohistochemistry
OR Overall response
Objective rate of ORR remission
DCR disease control Rate
Overall OS lifetime
mean overall survival of mOS
Progression of PD disease
PFS progression-free survival
mean progression-free survival of mPFS
PR partial response
CR complete response
Stabilization of SD disease
Dose limiting toxicity of DLT
MTD maximum tolerated dose
One dose of Q2W every two weeks
One daily dose of QD
Long-term exposure to CSD
non-CSD non-long-term sun exposure type
IRC independent review Committee
AE adverse events
Adverse events occurring during TEAE/TEAEs treatment
Adverse reactions associated with TRAE/TRAEs treatment
SAE Severe adverse reaction
RO receptor occupancy rate
Therapeutic effect evaluation standard of RECIST solid tumor
Therapeutic efficacy evaluation criteria for irRECIST immune-related solid tumors
DOR mitigation duration
TTR time to remission
BIRC Blind independent Review Committee (Blinded Industrial Review Committee)
NE cannot be evaluated
Recurrent life span of RFS
DMFS distant transfer-free lifetime
ivgtt intravenous drip
ECOG US eastern tumor cooperative group
High dose HDI interferon
HR risk ratio
PD-1 programmed death-1
PD-L1 programmed death ligand-1
CPS Joint Positive score
The invention is further illustrated by the following examples, which should not be construed as limiting the invention. The contents of all references cited throughout this application are expressly incorporated herein by reference.
Examples
Example 1: clinical research of anti-PD-1 antibody in adjuvant therapy of mucosal melanoma
This study was a multicenter, open, randomized, parallel control study comparing the efficacy and safety of treatment with recombinant humanized anti-PD-1 monoclonal antibody (tereprinimab/topalamicab/JS 001) injections or High Dose Interferon (HDI) after complete resection of mucosal melanoma patients with or without localized lymphoid disease, and evaluating the populations for optimal biomarker prediction. The clinical trial registration number: NCT03178123 (clinical trials.
1.1 study population
The main grouping criteria are as follows: eligible subjects must be (1) between 18 and 75 years of age; (2) Confirmed by pathological histology to be mucosal melanoma; (3) complete excision of the primary focus, surgical margin (-); perfecting whole-body stage examination before group entry to confirm that no region or distant metastasis exists; (4) not receiving regular adjuvant therapy; (5) an ECOG score of 0 or 1; (6) Has no treatment contraindication and has sufficient organ and bone marrow functions; the treatment was performed within 4 months after the operation.
The main exclusion criteria were: 1) Other melanoma subtypes exist simultaneously; 2) A history of autoimmune disease; 3) Non-melanoma cancers develop within 5 years; 4) In persistent infection; 5) Patients with autoimmune disease who have received systemic treatment within two years of the past;
6) A melanoma-resected patient who has received systemic adjuvant therapy; 7) Patients who have previously received anti-PD-1, anti-PD-L1 or anti-PD-L2 immunotherapy. .
1.2 clinical design
148 patients were scheduled into groups, randomized 1:1 were grouped into Teraprimab group (test group) or interferon group (control group). The random stratification factors were disease stage (I vs II vs III), primary focus site (head and neck, non-head and neck), PD-L1 expression status (positive, negative). Wherein, the disease stage, according to the definitions in the previous two studies (Lian B et al, ann Oncol 2017 868-873, cui C et al, ann Surg Oncol 2018 2184-2192): t1, mucosal or submucosal invasion; t2, invasion of the intrinsic muscle layer; t3, invasion of adventitia; t4, invading adjacent structures; n0, no region transfer node; n1, one or more regional metastatic lymph nodes; stage I, T1-2N0; stage II, T3-4/xN0; stage III, T1-4/xN1.
Patients with head and neck mucosal melanoma (mainly including nasopharynx and oropharynx), randomizedGrouping into Terepril monoclonal group or HDI group. Patients in the test group will receive a Terepril mab group infusion (3 mg/kg ivgtt q2 w) for up to 1 year (27 treatments). Patients in the control group will receive interferon alpha-2 b and some patients may choose to undergo low dose ramp therapy (poor tolerance: 3 MIU/m) as appropriate depending on patient tolerance 2 /d1、6MIU/m 2 /d2、9MIU/m 2 D3, good tolerance: 9MIU d1 or directly 15MIU/m 2 And/d). Subsequent acceptance of 15MIU/m 2 Day 1-5/week for 4 weeks; then 9MIU/m 2 D, 3 times weekly for 48 weeks. The head and neck mucous membrane melanoma patients are treated by self-initiated recombinant humanized anti-PD-1 monoclonal antibody injection or large-dose interferon within 6-8 weeks, and simultaneously initiated with auxiliary radiotherapy, reference dose: CTVTB 60-64 Gy/30 times, day 1-5/week, for 6 weeks.
Patients with non-head and neck mucosal melanoma were randomly grouped into either Teraprimab group or HDI group. Patients in the test group will receive a Terepril mab group infusion (3 mg/kg ivgtt q2 w) for up to 1 year (27 treatments). The patients in the control group will receive interferon alpha-2 b, and some patients may select appropriate low dose treatment for climbing slopes according to the tolerance of the patients (tolerance is poor: 3 MIU/m) 2 /d1、6MIU/m 2 /d2、9MIU/m 2 D3, good tolerance: 9MIU/m 2 D or direct 15MIU/m 2 D), subsequently receiving 15MIU/m 2 D, days 1-5/week for 4 weeks; then 9MIU/d, 3 times per week for 48 weeks.
Safety was evaluated during treatment. Treatment studies may be terminated when recurrent metastases of the disease occur, intolerable toxicity, withdrawal of informed consent, or other criteria for withdrawal from the study are met.
All subjects enrolled will follow up the overall survival outcome (tumor review evaluation time: 12 weeks in the first year, 16 weeks in the 2 nd year, half a year in the 3 rd to 5 th year, each year thereafter) and collect information on their subsequent anti-tumor treatment and survival. If the subject terminates study treatment without recording evidence of recurrent metastasis of the disease, the same frequency of evaluation of their tumors will continue until either the tumor recurs to metastasis or the subject begins to receive new anti-tumor therapy, whichever occurs first. The primary endpoint was RFS in the intended treatment population. Secondary endpoints include DMFS, 2 year RFS rate, OS, and security.
1.3 patient population
Between 7 months 2017 and 5 months 2019, 187 patients received screening, 145 patients were randomized to 1:1 was divided into either the tereprimab group (n =73, test group) or the interferon group (n =72, control group). Demographic data for the enrolled subjects are shown in table 1.
Table 1: demographic and baseline clinical characteristics of study subjects
Figure BDA0003648195750000201
Figure BDA0003648195750000211
Positive for PD-L1: defined as that JS311 IHC is adopted to stain the tumor cells CPS which is more than or equal to 1 percent
HDI, high dose interferon- α 2b; IQR, four-bit spacing; t, staging of the tumor; n, a node stage; PD-L1, programmed death ligand 1; CLND, complete lymph node dissection; LDH, lactate dehydrogenase; ULN, upper normal limit;
1.4 test drugs
Test drugs: the recombinant humanized anti-PD 1 monoclonal antibody injection (JS 001, common name: terepril monoclonal antibody injection, trade name: tuoyi) is freely provided by Shanghai Junshi biological medicine science and technology Limited company/Suzhou Zhonghe biological medicine science and technology Limited company; the specification is 240mg/6 ml/bottle.
Control drugs: the interferon alpha-2 b injection is provided by Moshadong (Gangle) and has a specification of 18MIU/1.2 ml/count.
1.5 disease assessment
Disease assessment is performed every 12 weeks for the first year, every 16 weeks for the second year, and every 6 months until 3-5 years, and then every year according to RECIST 1.1. Nasopharyngeal or oropharyngeal melanoma patients require additional nasopharyngeal or oropharyngeal CT or MRI. Any suspicious lymph node metastasis detected by superficial lymph node ultrasonography can be histologically verified by lymph node resection or anatomy, if necessary.
1.6 assessment of efficacy and safety
Efficacy analysis was performed for all patients randomly grouped. Primary endpoint RFS refers to the day patients randomized into the group until any recorded time of tumor recurrence, metastasis or death.
Safety assessments were performed on all patients who received at least one dose of study treatment. Adverse events were assessed and recorded according to National Cancer Institute Common Terminology criterion version 4.0 (version 4.0).
1.7 analysis of PD-L1 expression in tumor biopsy
Tumor PD-L1 IHC staining was performed in the central laboratory using JS311 antibody (Wang Z, ying J, xu J, et al Safety, anti Activity, and Pharmacokinetics of toripimab, a Programmed Cell Death 1 inhibitor, in Patents with Advanced Non-Small Cell luminescence Cancer: a Phase 1 Trial jama net Open 2020 3. Positive PD-L1 is defined as a tumor cell and/or infiltrating immune cell ratio CPS of more than or equal to 1%.
1.8 statistical analysis
Effectiveness:
recurrence-free survival time (RFS) for primary efficacy endpoint: refers to the time between the day patients were randomized into the cohort and any recorded tumor recurrence, metastasis or death.
The survival time is calculated by adopting a nonparametric Kaplan-Meier method respectively; if the influence of stratification factors such as disease stages on the survival curve is to be studied, the stratification Kaplan-Meier method can also be used for analysis. The comparison of the two sets of survival curves will use a two-sided Log-rank test.
Patients who did not observe the occurrence of an event (e.g., recurrence) will be treated as a deletion with a survival time between study initiation and the last follow-up.
In addition, a Cox proportional hazards regression was applied to perform multifactorial analyses, exploring various factors that may affect the relapse-free survival curve.
For the OS endpoint, kaplan-Meier (KM) curves and median estimates will be provided as appropriate.
Safety:
safety and tolerability will be assessed by clinical review of all relevant parameters, including Adverse Events (AEs), laboratory examinations, and vital signs.
Summary statistics (number, percentage, median, standard deviation, etc.) of the security endpoints will be provided as appropriate. Summary statistics of the number, percentage and 95% confidence intervals will be provided for any AE, any severe AE, any class 3-5 AE, any drug-related AE, any severe drug-related AE, any class 3-5 drug-related AE, suspension of drug administration due to AE, discontinuation due to AE, any immune-related AE irAE), mortality and incidence of specific AEs.
Summary statistics of the mean and standard deviation of the laboratory examination and vital sign changes from baseline results over each treatment cycle will be provided. Summary statistics of the number and percentage of laboratory values versus baseline deterioration will be provided according to CTCAE ratings.
2 results of the study
Between 7 months 2017 and 5 months 2019, 187 patients received screening, 145 patients were randomized to 1:1 was grouped into either the tereprimab group (n =73, test group) or the interferon group (n =72, control group). The mean age of the patients was 58 years; the majority of patients were female (62.8%); 80.7% of local diseases, 19.3% of local lymphatic diseases, 37.2% of local excision +/-CLND and 62.8% of wide excision +/-CLND; (ii) a PD-L1 positive 51.0% (CPS is more than or equal to 1, 22C3), PD-L1 negative 49.0%. There was no difference in baseline characteristics between the two groups.
Of 57 (39.3%) patients with primary tumors of the head and neck, 24 (85.7%) of 28 patients in the HDI group and 26 (89.7%) of 29 patients in the terlipril mab group received adjuvant radiation therapy. A total of 52 patients completed treatment for 1 year, 30 (41.1%) patients in the tereprimab group and 22 (30.6%) patients in the HDI group. The current study procedure is shown in figure 5.
By 31 days 3 months 2021, the median follow-up time for the HDI group was 26.4 months and the teriepril mab group was 25.5 months. As a clinical research of a first immunotherapy to HDI for treating mucosal melanoma, the research result shows that Terepril monoclonal antibody and HDI are used as adjuvant therapies, patients with completely resected mucosal melanoma have similar RFS, the Terepril monoclonal antibody can not only remarkably prolong RFS of a PD-L1 expression positive subgroup, but also has safety and tolerance which are obviously superior to those of HDI group, the incidence rate of adverse events (TRAEs) related to level 3 treatment is lower, and the optimal effect threshold preset as a main endpoint is reached.
The primary efficacy endpoint and each secondary efficacy endpoint results were as follows:
2.1 study of antitumor Effect
2.1.1 RFS, DMFS, OS values
By 31 days 3 months 2021, a total of 97 RFS events occurred in all 145 patients, with HDI group 46 RFS (63.9%) and terlipril mab group 51 RFS (69.9%). Median RFS values were similar for the HDI group and the terieprinimab group, 13.9 months for the HDI group (95% ci. The HDI group and the Teraprimab group had an RFS rate of 52% (95% CI: 38.61-63.76) and 52.9% (95% CI. The recurrence risk ratio HR, i.e. the ratio of RFS events in the test group to the control group, for each subgroup is shown in figure 2.
Table 2: subject median RFS, DMFS and OS data (3 months 31 days by 2021)
Figure BDA0003648195750000231
Figure BDA0003648195750000241
PD-L1+ is defined as that the CPS of the tumor cells is more than or equal to 1% by adopting JS311 IHC;
NR indicates failure to evaluate; NR: the result is not reached;
following disease recurrence or metastasis, the rates of subsequent surgical treatment were similar in both groups. However, after discontinuation of treatment, the patients receiving immunotherapy in the HDI group were more than the tereprinimab group (32.6% vs 26.0%) (table 3).
Table 3: proportion of patients receiving subsequent immunotherapy
Figure BDA0003648195750000242
HDI, high dose interferon- α 2b; PD-1, programmed cell death-1; CTLA4, cytotoxic T lymphocyte-associated protein 4.
There were 93 DMFS events occurring, including HDI group 44 (61.1%) and terliplizumab group 49 (67.1%). The median DMFS values for the HDI group and the tereprinimab group were 14.6 months (95% ci. The 1-year DMFS rates of the HDI group and the Terlipinimab group were 54.8% (95% CI.
The HDI group did not reach the median OS (95% CI. The relative risk of death (HR) was 1.11 (95% ci. HR for each subgroup OS is shown in FIG. 3.
2.1.2 subgroup effects
1) PD-L1 expression positive/negative subgroup
By 31 days 3 months 2021, tumors positive for PD-1 expression were found in 36 (50.0%) patients in the HDI group and 38 (52.1%) patients in the teripolycipimab group. In PD-L1 positive patients, median RFS was 11.1 months in HDI group (95% ci.
In tumor patients that were PD-L1 negative, the median RFS was 14.6 months in the HDI group (95% ci.
2) Other subgroups
For stage I, II and III patients with disease staging, median RFS values in HDI group were 8.3 months (95% ci; the median RFS values for the tereprimab group were 16.3 months (95% ci. In addition, neither gender, age, primary tumor site, or other baseline characteristics resulted in significant differences in RFS.
2.2 safety and tolerability Studies
By 31 days 3 months in 2021, the Terapril MAbs group TEAEs was 90.4% and HDI group was 100%. Most TEAEs in the terepril mab group were grade 1 or grade 2. No treatment-related death or infusion reactions occurred. 20 patients in the Teraprimab group (27.4%) and 63 patients in the HDI group (87.5%) developed TEAEs grade 3 or greater; 8 patients (11%) in the Tereprinimab group and 61 patients (84.7%) in the HDI group reported TRAEs of grade 3 or more (Table 4). Thus, teraprimab has better safety and tolerability than HDI group, with a lower incidence of treatment-related adverse events (TRAEs) of grade 3 or more.
Table 4: at least 15% of patients in both trials were reported to have adverse events during treatment #
Figure BDA0003648195750000251
Figure BDA0003648195750000261
Figure BDA0003648195750000271
# "indicating" a common treatment-related adverse event "is defined as occurring in greater than or equal to 20% of patients in the Teraprimab group.
Overall, the results of this randomized phase II trial comparing terepril mab and HDI in patients with completely resected mucosal melanoma for the first time, both therapies having median RFS numerically superior to the outcome of the resection surgery alone, and terepril mab significantly prolonged the RFS of the PD-L1 expression positive subgroup, highlighting the potential utility of these two interventions as adjuvant treatment for mucosal melanoma. Meanwhile, tereprinimab has superior safety and tolerability compared to HDI, which indicates that tereprinimab is a better treatment option.
Sequence listing
<110> Shanghai Junshi bio-medical science and technology GmbH
<120> medicine and method for treating patient with completely resected mucosal melanoma
<130> 213607Z11CNCN
<150> 202110532489.7
<151> 2021-05-17
<160> 10
<170> PatentIn version 3.5
<210> 1
<211> 16
<212> PRT
<213> Artificial Sequence
<220>
<223> LCDR1
<400> 1
Arg Ser Ser Gln Ser Ile Val His Ser Asn Gly Asn Thr Tyr Leu Glu
1 5 10 15
<210> 2
<211> 7
<212> PRT
<213> Artificial Sequence
<220>
<223> LCDR2
<400> 2
Lys Val Ser Asn Arg Phe Ser
1 5
<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence
<220>
<223> LCDR3
<400> 3
Phe Gln Gly Ser His Val Pro Leu Thr
1 5
<210> 4
<211> 5
<212> PRT
<213> Artificial Sequence
<220>
<223> HCDR1
<400> 4
Asp Tyr Glu Met His
1 5
<210> 5
<211> 17
<212> PRT
<213> Artificial Sequence
<220>
<223> HCDR2
<400> 5
Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe Lys
1 5 10 15
Gly
<210> 6
<211> 16
<212> PRT
<213> Artificial Sequence
<220>
<223> HCDR3
<400> 6
Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe Asp Val
1 5 10 15
<210> 7
<211> 112
<212> PRT
<213> Artificial Sequence
<220>
<223> VL
<400> 7
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Phe Gln Gly
85 90 95
Ser His Val Pro Leu Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
<210> 8
<211> 125
<212> PRT
<213> Artificial Sequence
<220>
<223> VH
<400> 8
Gln Gly Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Glu Met His Trp Val Arg Gln Ala Pro Ile His Gly Leu Glu Trp Ile
35 40 45
Gly Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser
115 120 125
<210> 9
<211> 219
<212> PRT
<213> Artificial Sequence
<220>
<223> LC
<400> 9
Asp Val Val Met Thr Gln Ser Pro Leu Ser Leu Pro Val Thr Leu Gly
1 5 10 15
Gln Pro Ala Ser Ile Ser Cys Arg Ser Ser Gln Ser Ile Val His Ser
20 25 30
Asn Gly Asn Thr Tyr Leu Glu Trp Tyr Leu Gln Lys Pro Gly Gln Ser
35 40 45
Pro Gln Leu Leu Ile Tyr Lys Val Ser Asn Arg Phe Ser Gly Val Pro
50 55 60
Asp Arg Phe Ser Gly Ser Gly Ser Gly Thr Asp Phe Thr Leu Lys Ile
65 70 75 80
Ser Arg Val Glu Ala Glu Asp Val Gly Val Tyr Tyr Cys Phe Gln Gly
85 90 95
Ser His Val Pro Leu Thr Phe Gly Gln Gly Thr Lys Leu Glu Ile Lys
100 105 110
Arg Thr Val Ala Ala Pro Ser Val Phe Ile Phe Pro Pro Ser Asp Glu
115 120 125
Gln Leu Lys Ser Gly Thr Ala Ser Val Val Cys Leu Leu Asn Asn Phe
130 135 140
Tyr Pro Arg Glu Ala Lys Val Gln Trp Lys Val Asp Asn Ala Leu Gln
145 150 155 160
Ser Gly Asn Ser Gln Glu Ser Val Thr Glu Gln Asp Ser Lys Asp Ser
165 170 175
Thr Tyr Ser Leu Ser Ser Thr Leu Thr Leu Ser Lys Ala Asp Tyr Glu
180 185 190
Lys His Lys Val Tyr Ala Cys Glu Val Thr His Gln Gly Leu Ser Ser
195 200 205
Pro Val Thr Lys Ser Phe Asn Arg Gly Glu Cys
210 215
<210> 10
<211> 452
<212> PRT
<213> Artificial Sequence
<220>
<223> HC
<400> 10
Gln Gly Gln Leu Val Gln Ser Gly Ala Glu Val Lys Lys Pro Gly Ala
1 5 10 15
Ser Val Lys Val Ser Cys Lys Ala Ser Gly Tyr Thr Phe Thr Asp Tyr
20 25 30
Glu Met His Trp Val Arg Gln Ala Pro Ile His Gly Leu Glu Trp Ile
35 40 45
Gly Val Ile Glu Ser Glu Thr Gly Gly Thr Ala Tyr Asn Gln Lys Phe
50 55 60
Lys Gly Arg Val Thr Ile Thr Ala Asp Lys Ser Thr Ser Thr Ala Tyr
65 70 75 80
Met Glu Leu Ser Ser Leu Arg Ser Glu Asp Thr Ala Val Tyr Tyr Cys
85 90 95
Ala Arg Glu Gly Ile Thr Thr Val Ala Thr Thr Tyr Tyr Trp Tyr Phe
100 105 110
Asp Val Trp Gly Gln Gly Thr Thr Val Thr Val Ser Ser Ala Ser Thr
115 120 125
Lys Gly Pro Ser Val Phe Pro Leu Ala Pro Cys Ser Arg Ser Thr Ser
130 135 140
Glu Ser Thr Ala Ala Leu Gly Cys Leu Val Lys Asp Tyr Phe Pro Glu
145 150 155 160
Pro Val Thr Val Ser Trp Asn Ser Gly Ala Leu Thr Ser Gly Val His
165 170 175
Thr Phe Pro Ala Val Leu Gln Ser Ser Gly Leu Tyr Ser Leu Ser Ser
180 185 190
Val Val Thr Val Pro Ser Ser Ser Leu Gly Thr Lys Thr Tyr Thr Cys
195 200 205
Asn Val Asp His Lys Pro Ser Asn Thr Lys Val Asp Lys Arg Val Glu
210 215 220
Ser Lys Tyr Gly Pro Pro Cys Pro Pro Cys Pro Ala Pro Glu Phe Leu
225 230 235 240
Gly Gly Pro Ser Val Phe Leu Phe Pro Pro Lys Pro Lys Asp Thr Leu
245 250 255
Met Ile Ser Arg Thr Pro Glu Val Thr Cys Val Val Val Asp Val Ser
260 265 270
Gln Glu Asp Pro Glu Val Gln Phe Asn Trp Tyr Val Asp Gly Val Glu
275 280 285
Val His Asn Ala Lys Thr Lys Pro Arg Glu Glu Gln Phe Asn Ser Thr
290 295 300
Tyr Arg Val Val Ser Val Leu Thr Val Leu His Gln Asp Trp Leu Asn
305 310 315 320
Gly Lys Glu Tyr Lys Cys Lys Val Ser Asn Lys Gly Leu Pro Ser Ser
325 330 335
Ile Glu Lys Thr Ile Ser Lys Ala Lys Gly Gln Pro Arg Glu Pro Gln
340 345 350
Val Tyr Thr Leu Pro Pro Ser Gln Glu Glu Met Thr Lys Asn Gln Val
355 360 365
Ser Leu Thr Cys Leu Val Lys Gly Phe Tyr Pro Ser Asp Ile Ala Val
370 375 380
Glu Trp Glu Ser Asn Gly Gln Pro Glu Asn Asn Tyr Lys Thr Thr Pro
385 390 395 400
Pro Val Leu Asp Ser Asp Gly Ser Phe Phe Leu Tyr Ser Arg Leu Thr
405 410 415
Val Asp Lys Ser Arg Trp Gln Glu Gly Asn Val Phe Ser Cys Ser Val
420 425 430
Met His Glu Ala Leu His Asn His Tyr Thr Gln Lys Ser Leu Ser Leu
435 440 445
Ser Leu Gly Lys
450

Claims (10)

1. Use of an anti-PD-1 antibody or antigen-binding fragment thereof in the manufacture of a medicament or kit for the adjunctive treatment of a patient having a completely resected mucosal melanoma or for the prevention of recurrence or distant metastasis of a mucosal melanoma in the patient.
2. The use according to claim 1, wherein the mucosal melanoma is head and neck or non-head and neck.
3. The use according to claim 1 or 2, wherein the mucosal melanoma is one positive for PD-L1 expression in an immunohistochemical staining assay of tumor tissue sections.
4. The use of claim 1, wherein the mucosal melanoma is head and neck mucosal melanoma, further comprising the combined administration of one or more therapies to a subject in need thereof; preferably, the therapy is a chemotherapeutic agent, a biotherapeutic agent, an immunogenic agent, an immunostimulatory cytokine, an encoded immunostimulatory cytokine, or radiation therapy.
5. The use of any one of claims 1 to 4, wherein the anti-PD-1 antibody or antigen-binding fragment thereof comprises an amino acid sequence as set forth in SEQ ID NO: 1.2 and 3, and the amino acid sequences are shown as SEQ ID NOs: 4. 5 and 6;
preferably, the anti-PD-1 antibody or an antigen-binding fragment thereof comprises a light chain variable region having an amino acid sequence as set forth in SEQ ID NO. 7 and a heavy chain variable region having an amino acid sequence as set forth in SEQ ID NO. 8;
more preferably, the anti-PD-1 antibody comprises a light chain having an amino acid sequence as set forth in SEQ ID NO. 9 and a heavy chain having an amino acid sequence as set forth in SEQ ID NO. 10.
6. The use of any one of claims 1 to 4, wherein the anti-PD-1 antibody is selected from one or more of nivolumab, pembrolizumab, toreplalimumab, sintilizumab, camrelizumab, tislelizumab, cemipilimab; preferably a toriplalimab.
7. The use of any one of claims 1 to 6, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is administered at a dose of about 0.1mg/kg to about 10.0mg/kg of the individual's body weight, such as about 0.1mg/kg, about 0.3mg/kg, about 1mg/kg, about 2mg/kg, about 3mg/kg, about 5mg/kg, or 10mg/kg of the individual's body weight, or is selected from about 120mg to about 480mg fixed dose, such as about 120mg, 240mg, 360mg or 480mg fixed dose, preferably about 3mg/kg of the individual's body weight or about 240mg fixed dose;
preferably, the anti-PD-1 antibody or antigen-binding fragment thereof is administered at a frequency of about once every week, once every two weeks, once every three weeks, once every four weeks, or once a month, preferably once every two weeks;
preferably, the anti-PD-1 antibody or an antigen-binding fragment thereof is administered at a dose of 1mg/kg body weight of the individual, 3mg/kg body weight of the individual, 10mg/kg body weight of the individual, or 240mg fixed dose, 480mg fixed dose, once every two or three weeks.
8. The use of any one of claims 1 to 7, wherein the anti-PD-1 antibody or an antigen-binding fragment thereof is administered parenterally, for example by intravenous infusion, in a liquid dosage form, for example as an injection.
9. The use of any one of claims 1 to 8, wherein the anti-PD-1 antibody or antigen-binding fragment thereof is administered for a period of one week, two weeks, three weeks, one month, two months, three months, four months, five months, half a year, one year, or longer, optionally wherein the time of each administration period is the same or different and the interval between each administration period is the same or different.
10. The use of claim 4, wherein the radiation therapy dose is CTV TB 60-64 Gy/30 times, 1-5 days per week for 6 weeks; preferably, the radiation therapy is administered concurrently within 6-8 weeks of administration of the anti-PD-1 antibody or antigen-binding fragment thereof.
CN202210540589.9A 2021-05-17 2022-05-17 Medicine and method for treating patient with completely resected mucosal melanoma Pending CN115364208A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN2021105324897 2021-05-17
CN202110532489 2021-05-17

Publications (1)

Publication Number Publication Date
CN115364208A true CN115364208A (en) 2022-11-22

Family

ID=84060086

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202210540589.9A Pending CN115364208A (en) 2021-05-17 2022-05-17 Medicine and method for treating patient with completely resected mucosal melanoma

Country Status (2)

Country Link
CN (1) CN115364208A (en)
WO (1) WO2022242621A1 (en)

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN104250302B (en) * 2013-06-26 2017-11-14 上海君实生物医药科技股份有限公司 The anti-antibody of PD 1 and its application
CN110882385B (en) * 2018-09-07 2021-07-09 上海君实生物医药科技股份有限公司 Use of anti-PD-1 antibodies in the treatment of tumors
CN113244386A (en) * 2020-02-13 2021-08-13 上海君实生物医药科技股份有限公司 Use of anti-PD-1 antibodies in the treatment of tumors

Also Published As

Publication number Publication date
WO2022242621A1 (en) 2022-11-24

Similar Documents

Publication Publication Date Title
EP3102604B1 (en) Combination of a pd-1 antagonist and a 4-1bb agonist for treating cancer
US10023649B2 (en) Method of treating cancer with a combination of an anti-CCR4 antibody and a 4-1BB agonist
US11572405B2 (en) Combination therapy with anti-IL-8 antibodies and anti-PD-1 antibodies for treating cancer
EP3957326A1 (en) Use of anti-pd-1 antibody in preparation of medicament for treating solid tumors
US20230212292A1 (en) Use of anti-pd-1 antibody in treating neuroendocrine tumors
WO2022242738A1 (en) Use of anti-pd-1 antibody in combination with chemotherapy in treating esophageal cancer
EP4327822A1 (en) Use of anti-pd-1 antibody in combination with first-line chemotherapy for treating advanced non-small cell lung cancer
WO2022042681A1 (en) Use of an anti-pd-1 antibody and a cytotoxic anticancer drug in treatment of non-small cell lung cancer
EP4104856A1 (en) Use of anti-pd-1 antibody in treatment of tumors
CN115066260A (en) Use of anti-PD-1 antibodies in the treatment of malignant tumors
WO2022242621A1 (en) Drug and method for treating patient who has undergone complete resection of mucosal melanoma
CN114099663A (en) Use of anti-PD-1 antibodies in the treatment of nasopharyngeal carcinoma
US20230365691A1 (en) Use of anti-pd-1 antibody in treatment of nasopharyngeal carcinoma
WO2023134787A2 (en) Use of combination of anti-pd-1 antibody and anti-vegf antibody in treatment of hepatocellular carcinoma
WO2023208001A1 (en) Combination of anti-pd-1 antibody and anti-egfr antibody, and use thereof in treatment of head and neck squamous cell carcinoma
KR20240028452A (en) Methods and compositions for treating cancer
CN117940452A (en) Methods and compositions for treating cancer

Legal Events

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