WO2021164713A1 - Biomarker relating to effect of tumor immunotherapy and application thereof - Google Patents

Biomarker relating to effect of tumor immunotherapy and application thereof Download PDF

Info

Publication number
WO2021164713A1
WO2021164713A1 PCT/CN2021/076771 CN2021076771W WO2021164713A1 WO 2021164713 A1 WO2021164713 A1 WO 2021164713A1 CN 2021076771 W CN2021076771 W CN 2021076771W WO 2021164713 A1 WO2021164713 A1 WO 2021164713A1
Authority
WO
WIPO (PCT)
Prior art keywords
tumor
autoantibodies
combination
immunotherapy
trim21
Prior art date
Application number
PCT/CN2021/076771
Other languages
French (fr)
Chinese (zh)
Inventor
孙苏彭
杨盼盼
隗啸南
周静
康美华
孙立平
Original Assignee
杭州凯保罗生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 杭州凯保罗生物科技有限公司 filed Critical 杭州凯保罗生物科技有限公司
Publication of WO2021164713A1 publication Critical patent/WO2021164713A1/en

Links

Images

Classifications

    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57484Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites
    • G01N33/57488Immunoassay; Biospecific binding assay; Materials therefor for cancer involving compounds serving as markers for tumor, cancer, neoplasia, e.g. cellular determinants, receptors, heat shock/stress proteins, A-protein, oligosaccharides, metabolites involving compounds identifable in body fluids
    • GPHYSICS
    • G01MEASURING; TESTING
    • G01NINVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
    • G01N33/00Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
    • G01N33/48Biological material, e.g. blood, urine; Haemocytometers
    • G01N33/50Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
    • G01N33/53Immunoassay; Biospecific binding assay; Materials therefor
    • G01N33/574Immunoassay; Biospecific binding assay; Materials therefor for cancer
    • G01N33/57407Specifically defined cancers
    • G01N33/57423Specifically defined cancers of lung

Definitions

  • the present invention relates to the field of biotechnology. Specifically, the present invention relates to a biomarker related to the effect of tumor immunotherapy, a protein combination for detecting the biomarker, and the corresponding application in predicting the effect of tumor immunotherapy.
  • Immunotherapy is currently the most promising research direction in the field of cancer treatment.
  • One of the hot spots is the use of immune checkpoint inhibitors (immune checkpoint inhibitor, ICI) immune blocking therapy, such as blocking programmed death factor-1 (programmed death factor-1). death-1, PD-1)/programmed death ligand-1 (PD-L1) immune checkpoint pathway therapy.
  • immune checkpoint inhibitors immune checkpoint inhibitor, ICI
  • ICI immune checkpoint inhibitors
  • PD-1/PD-L1 immune checkpoint pathway blocking therapy usually refers to injecting specific antibodies against PD-1 or PD-L1 into the tumor patient so that the tumor no longer has the ability to evade the immune system. Thereby promoting the body's immune system to eliminate tumor cells.
  • This therapy has achieved a significant effect of inhibiting tumor growth and removing tumors in some patients, and has been approved for melanoma, Hodgkin’s lymphoma, lung cancer, head and neck squamous cell carcinoma, liver cancer, esophageal cancer, breast cancer, Various indications for gastric cancer, nasopharyngeal cancer, lymphoma, etc.
  • PD-L1 is highly expressed (the expression level of PD-L1 is 50% or higher), and the response rate of immune checkpoint molecular inhibitor drugs is only between 30% and 40%; but in addition, it is found that 10% of PD -L1 low expression (PD-L1 expression ⁇ 50%) or PD-L1 negative cases are response cases, and even many patients whose PD-L1 expression is so low that it is undetectable have obtained lasting clinical benefit from ICI treatment .
  • the use of PD-L1 expression as a predictor of ICI curative effect has the following drawbacks: a considerable proportion of patients with advanced tumors cannot provide enough tumor tissue for the detection of PD-L1 expression; PD-L1 expression varies in different stages of tumor development.
  • ICI efficacy include tumor mutation burden (tumor mutation burden, TMB), neoantigen burden, mismatch repair (MMR)/microsatellite instability (microsatellite instability).
  • Tumor antigen expression is also related to the cellular activity of tumor infiltrating immune cells. Therefore, autoantibodies against tumor-associated antigens may also become targets or predictive markers for highly specific immunotherapy.
  • There have been related research reports, including Sachet a. Shukla has identified a subclass of tumor testis antigen MAGE-A, which is located in a narrow 75kb region of Xq28 chromosome, which can be used to predict the efficacy of anti-CTLA-4 antibodies .
  • Other studies have shown that serum antibodies against NY-ESO-1 and/or XAGE1 tumor testis antigen can be used to predict the ICI efficacy and patient survival of initial and subsequent non-small cell lung cancer (NSCLC).
  • NSCLC non-small cell lung cancer
  • Immune checkpoint inhibitors are very expensive drugs. Unlike conventional chemotherapy, although immune checkpoint inhibitor drugs are effective for some patients, they may also cause serious adverse reactions, especially the development of systemic autoimmune diseases. Therefore, there is still a need to identify new autoantibody biomarkers that can be used to predict the efficacy of ICI, and to develop detection antigens for the autoantibody biomarkers, especially antigen combinations, in order to provide new predictive methods for tumor immunotherapy effects .
  • the present invention detects autoantibodies against different antigen targets in the blood of lung cancer patients, and finally identifies a group of immune checkpoint inhibitors (ICI) that can be used to predict or judge the therapeutic effect of tumors, especially lung cancer.
  • ICI immune checkpoint inhibitors
  • an object of the present invention is to provide a combination of autoantibody biomarkers for predicting or judging the effect of tumor immunotherapy.
  • another object of the present invention is to provide reagents for detecting the autoantibody markers, such as an antigen protein combination, which can be used to detect the self in a tumor patient sample (such as blood) Whether the antibody biomarker is positive, so as to predict or judge the immunotherapy effect of tumor patients; and provide a kit containing the detection reagent, which can be used for corresponding detection.
  • the autoantibody markers such as an antigen protein combination
  • Another object of the present invention is to provide the use of the autoantibody biomarker combination or antigen protein combination in the preparation of products for predicting or judging the effect of tumor immunotherapy.
  • Another object of the present invention is to provide a method for predicting or judging the effect of immunotherapy of tumor patients or a method for treating tumors.
  • the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy in a subject.
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes anti-tumor-related antigens selected from the group consisting of: At least one of autoantibodies: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  • the autoantibody combination may include at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY -ESO-1, P53, IMP2.
  • the biomarker can be used to predict or judge: the subject's good tumor treatment effect; the subject benefits from tumor immunotherapy; the treatment is effective; or, the subject's tumor is sensitive to immunotherapy.
  • the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Tumor-associated antigen autoantibodies: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy in a subject
  • the biomarker is a combination of autoantibodies
  • the combination of autoantibodies includes anti-tumor-associated antigen Trim21, BRCA2 , Annexin 1, HUD autoantibodies, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody and anti-HUD autoantibody.
  • the autoantibody combination may include at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, PRAME.
  • the biomarkers can be used to predict or judge: the subject's poor tumor treatment effect; the subject does not benefit from tumor immunotherapy; the treatment is ineffective; or the subject's tumor is not sensitive to immunotherapy.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a biomarker for predicting or judging a subject's immune tumor treatment effect
  • the biomarker is a combination of autoantibodies
  • the combination of autoantibodies includes anti-tumor-associated antigens HSP105, AKAP4 Anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
  • the autoantibody is the autoantibody in serum, plasma, interstitial fluid, cerebrospinal fluid or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG (e.g. IgG1, IgG2, IgG3, IgG4).
  • IgA for example, IgA1, IgA2
  • IgM for example, IgA2, IgG3, IgG4
  • the subject is a mammal, preferably a primate mammal, more preferably a human.
  • the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
  • the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy.
  • the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or
  • the CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 antibodies Or anti-PD-L1 antibody.
  • the biomarker that is, the combination of autoantibodies
  • a sample such as plasma or serum
  • the autoantibody biomarkers can be used to predict or judge whether a subject, such as a tumor patient, can benefit from immunotherapy (whether the immunotherapy effect is good or poor; whether the immunotherapy is effective; or whether the subject is effective
  • the patient’s tumor is sensitive or insensitive to immunotherapy), at least for the corresponding auxiliary judgment.
  • the level of autoantibodies in a sample can be quantified by referring to a standard curve, and then the cutoff value is used to determine the presence ( ⁇ cutoff value, positive) or absence of autoantibody biomarkers ( ⁇ cutoff value) , Is negative) judgment.
  • the cutoff value of the autoantibody level can be a reference level from a healthy person or a healthy person; for example, it can be defined as the average value of a person who is confirmed to have no cancer through a physical examination plus 2 standard deviations.
  • the autoantibody biomarker provided by the present invention can be detected by a variety of methods, for example, it can be detected by the antigen-antibody specific reaction between the tumor-associated antigen that causes the autoantibody to appear and the antigen-antibody specific reaction. Therefore, correspondingly, the present invention also provides a reagent for detecting the autoantibody biomarker.
  • the reagents may be reagents used in detection methods such as enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, immunoblotting, bead immunoassay, microfluidic immunoassay, etc.
  • ELISA enzyme-linked immunosorbent assay
  • the reagent is used to detect the autoantibody biomarker of the present invention through an antigen-antibody reaction, for example, by ELISA.
  • the reagent may be an antigen protein combination for detecting the autoantibody combination, and the antigen protein combination includes at least one selected from the following antigen proteins: Trim21, BRCA2, Annexin 1, HUD, NY- ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  • the reagent can be used to detect whether the corresponding autoantibody biomarker in a sample (such as plasma or serum) of a subject, such as a tumor patient, is positive, so as to realize the prediction or clinical effect of the administration of tumor immunotherapy as described above. judge.
  • the amino acid sequence contained in the tumor-associated antigen or antigen protein is as follows:
  • Trim21 includes the amino acid sequence shown in SEQ ID NO:1;
  • BRCA2 includes the amino acid sequence shown in SEQ ID NO: 2;
  • Annexin 1 includes the amino acid sequence shown in SEQ ID NO: 3;
  • HUD includes the amino acid sequence shown in SEQ ID NO: 4;
  • NY-ESO-1 includes the amino acid sequence shown in SEQ ID NO: 5;
  • P53 includes the amino acid sequence shown in SEQ ID NO: 6;
  • IMP2 includes the amino acid sequence shown in SEQ ID NO: 7;
  • HSP105 includes the amino acid sequence shown in SEQ ID NO: 8;
  • MAGE-A3 includes the amino acid sequence shown in SEQ ID NO: 9;
  • AKAP4 includes the amino acid sequence shown in SEQ ID NO: 10;
  • PRAME includes the amino acid sequence shown in SEQ ID NO: 11.
  • the present invention provides the use of the biomarker or reagent in the preparation of a product for predicting or judging the effect of tumor immunotherapy in a subject.
  • the tumor immunotherapy effects include good tumor immunotherapy effects and poor tumor immunotherapy effects, respectively, depending on the corresponding biomarkers or reagents.
  • the subject is a mammal, preferably a primate mammal, more preferably a human.
  • the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
  • the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy.
  • the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or
  • the CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
  • the present invention provides a kit comprising the reagent of the present invention.
  • the kit can be used for enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, western blotting, microbead immunoassay, microfluidic immunoassay, etc. to the autoantibody biomarker Kits for detection of substances.
  • ELISA enzyme-linked immunosorbent assay
  • the kit is used to detect the autoantibody biomarker of the present invention through an antigen-antibody reaction, for example, by ELISA.
  • the kit is an enzyme-linked immunosorbent assay (ELISA) detection kit. That is, using this kit, the enzyme-linked immunosorbent assay is used to detect whether the autoantibody biomarker in the subject's sample is positive.
  • the kit can also include other components required for ELISA detection of autoantibody biomarkers, all of which are well known in the art.
  • the antigen protein in the kit can be linked with a tag peptide, such as His tag, streptavidin tag, Myc tag; for another example, the kit can include a solid phase carrier, such as with immobilized antigen Microporous protein carrier, such as an enzyme-labeled plate; it may also include adsorbed protein for immobilizing antigen protein on a solid carrier, dilution of blood such as serum, washing solution, enzyme-labeled secondary antibody, color development Liquid, stop liquid, etc.
  • a tag peptide such as His tag, streptavidin tag, Myc tag
  • the kit can include a solid phase carrier, such as with immobilized antigen Microporous protein carrier, such as an enzyme-labeled plate; it may also include adsorbed protein for immobilizing antigen protein on a solid carrier, dilution of blood such as serum, washing solution, enzyme-labeled secondary antibody, color development Liquid, stop liquid, etc.
  • the present invention provides a method for predicting or judging the effect of tumor immunotherapy in a subject.
  • the present invention provides a method for predicting or judging the sensitivity of a subject's tumor to immunotherapy.
  • the above methods include testing whether the following biomarkers in a sample from a subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  • the method may include detecting whether the following biomarkers in a sample from a subject are positive:
  • the biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2.
  • the biomarker is positive, it is predicted or judged that the subject has a good tumor immunotherapy effect; the subject benefits from the tumor immunotherapy; the treatment is effective; or the subject's tumor is sensitive to the immunotherapy.
  • the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Tumor-associated antigen autoantibodies: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a method for predicting or judging the effect of a subject’s tumor immunotherapy, or a method for predicting or judging the sensitivity of a subject’s tumor to immunotherapy, the method comprising detecting from Whether the following biomarkers in the subject’s sample are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens Trim21, BRCA2, Annexin 1, and HUD, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens.
  • HUD autoantibodies namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens.
  • the method may include detecting whether the following biomarkers in a sample from the subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, and PRAME.
  • HSP105 tumor-associated antigens
  • MAGE-A3, AKAP4, and PRAME tumor-associated antigens
  • the autoantibody combination includes autoantibodies selected from the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a method for predicting or judging the effect of a subject’s tumor immunotherapy, or a method for predicting or judging the sensitivity of a subject’s tumor to immunotherapy, the method comprising detecting from Whether the following biomarkers in the subject’s sample are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens HSP105 and AKAP4, that is, anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
  • the detection can be carried out using the reagent of the present invention, for example, an antigen-protein combination or a kit containing the reagent.
  • the subject is a mammal, preferably a primate mammal, more preferably a human.
  • the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King's lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
  • the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy.
  • the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or
  • the CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
  • the sample is serum, plasma, interstitial fluid, cerebrospinal fluid, or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG ( For example, IgG1, IgG2, IgG3, IgG4).
  • IgA for example, IgA1, IgA2
  • IgM for example, IgG1, IgG2, IgG3, IgG4
  • the method includes the following steps:
  • the autoantibody biomarker in the sample is positive, predict or judge: the subject has a good or poor tumor immunotherapy effect; the subject benefits or does not benefit from tumor immunotherapy; the treatment is effective Or ineffective; or, the subject's tumor is sensitive or insensitive to immunotherapy.
  • the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  • the method may include detecting whether the following biomarkers in a sample from a subject are positive:
  • the biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2.
  • the biomarker is positive, the subject is subjected to tumor immunotherapy.
  • the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Autoantibodies against tumor-associated antigens: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens Trim21, BRCA2, Annexin 1, and HUD, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens.
  • HUD autoantibodies namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens.
  • the method may include detecting whether the following biomarkers in a sample from the subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, and PRAME.
  • HSP105 tumor-associated antigens
  • MAGE-A3, AKAP4, and PRAME tumor-associated antigens
  • the autoantibody combination includes autoantibodies selected from the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
  • the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
  • the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
  • the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens HSP105 and AKAP4, that is, anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
  • the detection can be carried out using the reagent of the present invention, for example, an antigen-protein combination or a kit containing the reagent.
  • the subject is a mammal, preferably a primate mammal, more preferably a human.
  • the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
  • the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy.
  • the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or
  • the CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  • the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
  • the sample is serum, plasma, interstitial fluid, cerebrospinal fluid, or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG ( For example, IgG1, IgG2, IgG3, IgG4).
  • IgA for example, IgA1, IgA2
  • IgM for example, IgG1, IgG2, IgG3, IgG4
  • the method includes the following steps:
  • the subject When the autoantibody biomarker in the sample is positive, the subject is allowed to undergo tumor immunotherapy, or the subject is not allowed to undergo tumor immunotherapy.
  • the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy, and the biomarker is a combination of autoantibodies.
  • the autoantibody combination of the present invention includes two combinations that predict good tumor immunotherapy effect and poor tumor immunotherapy effect.
  • the former can be called a positive prediction of tumor immunotherapy effect, and the latter can be called tumor immunity. Negative prediction of treatment effect.
  • the effective rate of immune checkpoint blocking therapy for tumor patients who test positive is more significant. Lower than the autoantibody combination test negative tumor patients (P ⁇ 0.05).
  • the autoantibody biomarkers provided by the present invention can provide accurate prediction or judgment results. Based on the prediction or judgment result, the patient or clinician can better decide whether the patient needs immunotherapy, so as to avoid excessive medical treatment, reduce treatment costs, and reduce or avoid adverse reactions.
  • the two autoantibody combinations provided by the present invention can also be used alone or in combination as required.
  • the positive conditions of the autoantibody biomarkers predicted in the positive direction and the negative conditions of the autoantibody biomarkers predicted in the negative direction can be combined.
  • Figure 1 shows the tumor response to immunotherapy after the treatment of patients who showed positive or negative autoantibody combination before treatment, where Figure 1-A: P_Ab. combination, Figure 1-B: N_Ab. combination.
  • Figure 2 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, where Figure 2-A: training set and Figure 2-B: validation set.
  • Figure 3 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, wherein Figure 3-A to Figure 3-RNP respectively show the results of corresponding autoantibody combination A to autoantibody combination RNP.
  • Figure 4 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, wherein Figure 4-K to Figure 4-P show the results of corresponding autoantibody combination K to autoantibody combination P, respectively.
  • Figure 5 shows the survival curve of patients who showed positive or negative autoantibody or autoantibody combination before treatment after treatment, where Figure 5-A: IMP2, Figure 5-B: anti-XAGE-1 and anti-NY-ESO-1.
  • Figure 6 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, where Figure 6-A: first-line immunotherapy, Figure 6-B: posterior-line immunotherapy, and Figure 6-C: immune single agent Treatment), Figure 6-D: Immunotherapy combined with chemotherapy.
  • the term "antigen” or the term “antigenic protein” can be used interchangeably.
  • the following experimental operations or definitions are involved in the present invention. It should be noted that the present invention can also be implemented using other conventional techniques in the field, and is not limited to the following experimental operations.
  • the cDNA of the tumor-associated antigen (TAA) was cloned into the 6XHis-labeled PET28(a) expression vector.
  • a streptavidin protein or the like biotin-binding tag protein
  • the obtained recombinant expression vector was transformed into Escherichia coli for expression.
  • the protein was denatured with 6M guanidine hydrochloride, and renatured in vitro according to standard methods, and then Ni-NTA affinity was carried out by 6XHis tag Column purification to obtain antigen protein.
  • Venous blood was collected in EDTA-treated or citric acid-treated blood collection tubes one week to one day before immunotherapy. Then centrifuge at 1000-2000RCF at room temperature for 15 minutes; after centrifugation, gently transfer the supernatant to another clean centrifuge tube at room temperature, and place it in a refrigerator at -80°C for long-term storage.
  • the prepared antigen protein is coated on the surface of the microwell of a 96-well solid phase plate.
  • a 96-well solid phase plate was coated overnight with 5-10ug/ml biotin-labeled bovine serum albumin; on the second day, the uncoated bovine serum albumin in the microwells of the solid phase plate was washed away.
  • Add 300uL of blocking solution containing BSA to block at room temperature for 1h; add antigen protein and incubate for 1.5h, then wash off unadsorbed antigen protein. After coating the antigen protein, add 300ul of the stabilized solution containing BSA to the microwells, incubate for 1h, and then use it or dry it under vacuum for later use.
  • the purified antigen protein is indirectly coated on the surface of the solid phase plate through the specific reaction between biotin and streptavidin.
  • the diluted plasma sample is added to the microwells coated with the antigen protein, and the autoantibodies in the plasma sample are specifically combined with the antigen protein on the surface of the solid phase plate after incubation.
  • PBS buffer prepare your own, with a pH of 7.6.
  • Stop and read Add 50ul/well of stop solution according to the order of adding color reagent, and read at 450nm in the microplate reader.
  • the cutoff value of the autoantibody level is defined as equal to the average value of the healthy control cohort in the control group (people confirmed to have no cancer through physical examination) plus 2 standard deviations (SD).
  • the results of multiple autoantibodies are combined to judge the predictive effect when analyzing the results.
  • the rule is: multiple autoantibodies are detected in a patient sample. As long as one or more of the autoantibodies are positive, the antibody combination is judged to be positive; and if all autoantibodies are negative, the antibody combination is judged to be negative .
  • the target lesion at baseline (before treatment) is evaluated, and the baseline sum of the longest diameter of the target lesion is recorded to determine the objective reaction.
  • the best curative effect refers to the record of the best curative effect from the beginning of the treatment study to the end of the treatment, which is confirmed after considering various factors.
  • PD Compared with the minimum sum of the diameters of all target lesions before treatment, the sum of the diameters of all target lesions is increased by at least 20% and the absolute value of the total increase in diameter must be greater than 5mm; or new lesions appear.
  • PR Compared with the sum of the diameters of all target lesions before treatment, the sum of the diameters of all target lesions is reduced by at least 30%.
  • SD Compared with the minimum sum of the diameters of all target lesions before treatment, the shrinkage of the target lesion does not meet the partial remission (PR), and the increase does not meet the disease progression (PD). It refers to a type between PR and PD. The state of the time.
  • PR partial remission
  • PD disease progression
  • CR All target lesions disappear, and the short axis value of any pathological lymph node (whether it is a target lesion or not) must be less than 10mm.
  • PFS Progression-free survival time, that is, the time from the beginning of randomization to the recurrence of the disease or the death of the patient due to various reasons.
  • mPFS Median progression-free survival time, that is, the median time from randomization to disease recurrence or death due to various reasons.
  • PD-L1 expression level Use immunohistochemistry to evaluate the percentage of tumor cells stained with PD-L1 membrane of any intensity in all tumor cells. The test results are divided into four groups, namely negative (less than 1%) , Low expression group (1%-49%), high expression group ( ⁇ 50%), unknown.
  • the healthy control group are those who have not been or are not diagnosed with cancer among the medical examiners.
  • the lung cancer group includes 10 patients with small cell lung cancer, 12 patients with lung squamous cell carcinoma, 19 patients with lung adenocarcinoma, and 6 patients with other subtypes of lung cancer. Plasma was collected before treatment. The information of the experimental population is shown in Table 1.
  • the detection specificity of each antigen is set to ⁇ 93.6% (in the healthy control group of 47 cases, there are ⁇ 6.4 % Positive rate); and the sensitivity in the lung cancer group (positive rate, that is, the proportion of autoantibody positive in the total number of lung cancer patients in 47 cases) is similar to that found in other literature, the positive detection rate of a single autoantibody Low, usually between 5-20%. Therefore, in the preliminary screening of lung cancer-related antigens, antigen proteins are divided into four groups based on sensitivity. The screened antigen protein and the sensitivity and specificity determined by the test are shown in Table 2.
  • this study tries to include various scenarios of immunotherapy for patients with lung cancer.
  • Thirty-eight lung cancer patients used immunotherapy as the first-line treatment, while 40 lung cancer patients received one or more treatments such as chemotherapy and targeted therapy, and then chose immunotherapy (post-line treatment).
  • the immunotherapy used by the enrolled patients included immune checkpoint inhibitors, including imported nivolumab and pembrolizumab, as well as domestic immune checkpoint inhibitors (Table 3).
  • the treatment plan for lung cancer patients includes two situations: immunotherapy is monotherapy (25 patients), or immunotherapy combined with chemotherapy (53 patients).
  • P_Ab autoantibodies against this antigen. Patients showing positive signals of such autoantibodies have basically achieved the "PR” and “SD” in BOR. “, that is, a good curative effect, and the patient population as a whole meets the percentage of "PR"/"PD” ⁇ 2.
  • This type of autoantibody is preliminarily determined as a positive predictive antibody, which has a positive predictive effect of good immunotherapy curative effect.
  • N_Ab Some tumor-associated antigens are "negatively related" antigens.
  • autoantibodies against this antigen are named "N_Ab.”
  • Patients showing positive signals of such autoantibodies all have "PD” and "SD” in BOR.
  • the patient population as a whole meets the "PD" percentage/"PR” percentage ⁇ 2.
  • Determining such autoantibodies as negative predictive antibodies has the negative predictive effect of poor immunotherapy efficacy. It is preliminarily determined that both the positive predictive antibody and the negative predictive antibody can be used to predict the effect of immunotherapy, and have good and poor predictive effects of immunotherapy respectively.
  • the treatment methods include immune monotherapy patients and immune-combined chemotherapy patients (Table 3)) as a "validation set" to verify whether the discovered autoantibodies still have the characteristics of efficacy prediction.
  • the results of multiple autoantibodies are combined to judge the predictive effect when analyzing the results.
  • the rule is: if multiple autoantibodies are detected in a patient, as long as one or more of the autoantibodies are positive, the antibody combination is judged to be positive; and if all autoantibodies are negative, the antibody combination is judged to be negative.
  • anti-Trim21 autoantibodies, anti-BRACA2 autoantibodies, anti-Annexin 1 autoantibodies and anti-HUD antibodies were selected as a combination of autoantibodies.
  • Negative refers to patients whose anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody and anti-HUD antibody are all negative (ie, the antibody combination is negative);
  • Figure 1-B N_Ab. combination
  • N_Ab. positive refers to patients whose anti-HSP105 autoantibody and anti-AKAP4 antibody are positive (that is, the combination of antibodies is positive)
  • N_Ab. negative refers to both anti-HSP105 autoantibody and anti-AKAP4 antibody negative The patient (ie, the antibody combination is negative).
  • Figure 1-A in Figure 1 shows that in the training set (first-line), 47.6% of patients with positive correlation autoantibodies have a treatment effect of "PR” after treatment, and 42% of patients have a treatment effect of "SD". , 9.5% of the patients had a treatment effect of "PD"; while the positively correlated autoantibody combination was negative, 28.6% of the patients had a treatment effect of "PR", 50% of the patients had a treatment effect of "SD", and 21.4% of the patients The therapeutic effect is "PD”.
  • 50% of the patients with positive correlation autoantibody combination had a treatment effect of "PR"
  • 37.5% of patients had a treatment effect of "SD”
  • 12.5% of patients had a treatment effect of "PR”.
  • PD for patients with a negative combination of positively correlated autoantibodies, after treatment, 50% of patients have a treatment effect of "PD” and 50% of patients have a treatment effect of "SD”.
  • Figure 1-B in Figure 1 shows that in the training set (first-line), 50% of patients with positive negative autoantibody combinations have a treatment effect of "PD” after treatment, and 50% of patients have a treatment effect of "SD” , 0% of patients have a treatment effect of "PR”.
  • the validation set after treatment, 42.9% of patients with positive negative autoantibodies have a treatment effect of "PD", 42.9% of patients have a treatment effect of "SD", and 14.3% of patients have a treatment effect of " PR”.
  • the Kaplan-Meier method was used to analyze the progression-free survival of patients in the training set and the validation set, and to draw a survival curve. The results found that the positive and negative groups of autoantibody combinations showed great differences in the progression-free survival curve.
  • the training set first-line
  • the median progression-free survival time of patients with positive antibody combinations was greater than 10 Month
  • the antibody combination negative patient is 5.52 months
  • the p-value is 0.0512, see Figure 2-A in Figure 2.
  • the validation set back line
  • the median progression-free time was 7.56 months, while the antibody combination negative patient was 2.43 months, and the p-value was less than 0.005, as shown in Figure 2-B in Figure 2.
  • the PD-L1 expression level and IMP2, XAGE-1 and NY-ESO-1 autoantibodies were used as prediction methods, and the patient's response to immunotherapy was observed. The results are shown in Table 7.
  • PD-L1 is used as a common marker for predicting the effect of immunotherapy. Because fluorescence detection of PD-L1 tissue expression requires the use of qualified tumor tissue samples from lung cancer patients, the source of the samples is not easy to obtain, and for other reasons, in the immunotherapy patients studied in the present invention, about 50% of patients There is no information on PD-L1 markers.
  • the antibody combination R-positive patients had 68.2% PR, 22.9% SD and 10% PD after treatment; the antibody combination RPN-positive patients had 72.7% PR, 28.6% SD and 15% after treatment. PD.
  • These autoantibody combinations can predict the efficacy of immunotherapy as well as PD-L1.
  • the median progression-free survival time of the IMP2 antibody positive and negative groups was 10.02 months and 5.52 months, respectively, but the P value was 0.7867, which was not statistically significant.
  • the survival curves of the positive and negative groups of anti-XAGE-1 and anti-NY-ESO-1 autoantibody combinations basically overlap, and the results are the median of the positive and negative groups of patients. There is no difference in progression-free survival time. Therefore, regardless of the combination of anti-XAGE-1 antibody and anti-NY-ESO-1 antibody or anti-IMP2 antibody, the predictive effect of the therapeutic effect of patients with immunotherapy is not particularly ideal.
  • the risk ratio of positive antibody combination (HR: Hazard Ratio) (Mantel- Haenszel) predictive value is 0.2541 (0.0684-0.8786).
  • the median progression-free survival time of the two groups of patients with antibody combination positive and negative is (>10 months) and 5.52 months (P value is 0.0309).
  • the predictive value of HR for positive antibody combination was 0.2948 (0.1409-0.6167), and the median progression-free survival time of the two groups of positive and negative antibody combination were (8.18 months) and 2.43 months (P value 0.0012) .

Abstract

Disclosed is a biomarker for predicting the effect of tumor immunotherapy. The biomarker is a group of autoantibodies against tumor-associated antigens. The biomarker comprises at least one of the following autoantibodies against the tumor-associated antigen: Trim21, BRCA2, Annexin1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, and PRAME. The biomarker can be detected in samples from patients with tumors to predict the clinical effect of immunotherapy, thereby helping to determine whether the immunotherapy benefits the patients with tumors. A combination of antigen and protein for detecting the biomarker, a kit containing the combination of antigen and protein, and a corresponding detection or diagnosis method are further provided.

Description

与肿瘤免疫治疗效果相关的生物标志物及其应用Biomarkers related to the effect of tumor immunotherapy and their applications
本专利申请要求于2020年2月21日提交的申请号为CN2020101066590的中国发明专利申请的优先权权益,在此将其全部内容引入作为参考。This patent application claims the priority rights of the Chinese invention patent application with the application number CN2020101066590 filed on February 21, 2020, and the entire content of which is hereby incorporated by reference.
技术领域Technical field
本发明涉及生物技术领域,具体而言,本发明涉及一种与肿瘤免疫治疗效果相关的生物标志物、检测其的蛋白组合以及相应的在预测肿瘤免疫治疗效果中的应用。The present invention relates to the field of biotechnology. Specifically, the present invention relates to a biomarker related to the effect of tumor immunotherapy, a protein combination for detecting the biomarker, and the corresponding application in predicting the effect of tumor immunotherapy.
背景技术Background technique
免疫疗法是目前肿瘤治疗领域中最具前景的研究方向,热点之一为采用免疫检查点抑制剂(immune checkpoint inhibitor,ICI)的免疫阻断疗法,例如基于阻断程序性死亡因子-1(programmed death-1,PD-1)/程序性死亡因子配体-1(programmed death ligand-1,PD-L1)免疫检查点通路的疗法。Immunotherapy is currently the most promising research direction in the field of cancer treatment. One of the hot spots is the use of immune checkpoint inhibitors (immune checkpoint inhibitor, ICI) immune blocking therapy, such as blocking programmed death factor-1 (programmed death factor-1). death-1, PD-1)/programmed death ligand-1 (PD-L1) immune checkpoint pathway therapy.
PD-1/PD-L1免疫检查点通路的阻断疗法通常是指,将抗PD-1或PD-L1的特异性抗体注射入肿瘤患者体内,使得肿瘤不再具备逃避免疫系统攻击的能力,从而促进机体免疫系统清除肿瘤细胞。这一疗法已经在部分患者中实现了显著抑制肿瘤生长和清除肿瘤的疗效,已获批用于黑色素瘤、霍奇金淋巴瘤、肺癌、头颈部鳞癌、肝癌、食管癌、乳腺癌、胃癌、鼻咽癌、淋巴瘤等多种适应症。PD-1/PD-L1 immune checkpoint pathway blocking therapy usually refers to injecting specific antibodies against PD-1 or PD-L1 into the tumor patient so that the tumor no longer has the ability to evade the immune system. Thereby promoting the body's immune system to eliminate tumor cells. This therapy has achieved a significant effect of inhibiting tumor growth and removing tumors in some patients, and has been approved for melanoma, Hodgkin’s lymphoma, lung cancer, head and neck squamous cell carcinoma, liver cancer, esophageal cancer, breast cancer, Various indications for gastric cancer, nasopharyngeal cancer, lymphoma, etc.
然而,虽然ICI(例如抗PD-1/PD-L1抗体)在肿瘤治疗中取得了令人瞩目的成就,但是有数据表明,大多数接受ICI的患者并没有从中获益。例如,相当比例的肿瘤患者对抗PD-1/PD-L1抗体无反应。因此,迫在眉睫的是识别和开发可预测ICI疗效的生物标志物,以精准发现ICI获益患者。However, although ICI (such as anti-PD-1/PD-L1 antibodies) has made remarkable achievements in tumor treatment, there are data showing that most patients receiving ICI do not benefit from it. For example, a considerable proportion of tumor patients do not respond to anti-PD-1/PD-L1 antibodies. Therefore, it is urgent to identify and develop biomarkers that can predict the efficacy of ICI, so as to accurately identify patients who benefit from ICI.
多项临床研究表明,PD-L1表达量升高的肿瘤患者采用ICI治疗后获益更多,因此一些研究将PD-L1表达视为主要评估目标,并且基于PD-L1表达水平在部分肿瘤中就ICI疗效预测具有的良好区分作用,其已经成为目前ICI治疗唯一的伴随诊断。但是也有研究发现,部分患者的PD-L1表达和ICI治疗响应或总生存期(overall survival,OS)之间并无关联。例如,PD-L1高表达(PD-L1的表达量为50%或更高),免疫检查点分子抑制剂药物的应答 率仅在30%至40%之间;但此外,发现10%的PD-L1低表达(PD-L1的表达量<50%)或PD-L1呈阴性的病例则是应答病例,甚至许多PD-L1表达低至无法检测到的患者从ICI治疗中获得持久的临床益处。并且,将PD-L1表达作为ICI疗效预测因素还有以下缺陷:相当比例的晚期肿瘤患者不能提供足够的肿瘤组织进行PD-L1表达的检测;PD-L1的表达在肿瘤的不同发展阶段、不同区域会存在异质性,其检测结果可能受取样时间和取样位点的影响;此外,检测方法、评估标准和阳性临界值的界定等方面的差异也会导致结果判定不同,这些都导致了PD-L1表达不能作为用于临床治疗决策的足够全面的独立生物标志物。除此之外,目前比较常用的ICI疗效预测因素还包括肿瘤突变负荷(tumor mutation burden,TMB)、新抗原负担、错配修复(mismatch repair,MMR)/微卫星不稳定性高(Microsatellite instability-high,MSI-H)、人类白细胞抗原(HLA)基因型等,但是这些预测因素均存在无法准确区分响应者和非响应者、无法提供明确的敏感性或特异性、预测性差或者检测要求如活检取样或成本过高等缺陷。A number of clinical studies have shown that patients with tumors with elevated PD-L1 expression benefit more from ICI treatment. Therefore, some studies regard PD-L1 expression as the main evaluation target and are based on PD-L1 expression levels in some tumors. Regarding the good distinguishing effect of ICI curative effect prediction, it has become the only accompanying diagnosis of ICI treatment at present. However, some studies have found that there is no correlation between PD-L1 expression in some patients and ICI treatment response or overall survival (OS). For example, PD-L1 is highly expressed (the expression level of PD-L1 is 50% or higher), and the response rate of immune checkpoint molecular inhibitor drugs is only between 30% and 40%; but in addition, it is found that 10% of PD -L1 low expression (PD-L1 expression <50%) or PD-L1 negative cases are response cases, and even many patients whose PD-L1 expression is so low that it is undetectable have obtained lasting clinical benefit from ICI treatment . In addition, the use of PD-L1 expression as a predictor of ICI curative effect has the following drawbacks: a considerable proportion of patients with advanced tumors cannot provide enough tumor tissue for the detection of PD-L1 expression; PD-L1 expression varies in different stages of tumor development. There will be heterogeneity in the region, and the detection results may be affected by the sampling time and sampling site; in addition, differences in detection methods, evaluation standards, and the definition of positive cut-off values can also lead to different results judgments, which all lead to PD -L1 expression cannot be used as a comprehensive enough independent biomarker for clinical treatment decision-making. In addition, the most commonly used predictors of ICI efficacy include tumor mutation burden (tumor mutation burden, TMB), neoantigen burden, mismatch repair (MMR)/microsatellite instability (microsatellite instability). high, MSI-H), human leukocyte antigen (HLA) genotype, etc., but these predictive factors cannot accurately distinguish between responders and non-responders, cannot provide clear sensitivity or specificity, poor predictability, or detection requirements such as biopsy Defects such as high sampling or cost.
因此,临床上用于区分对ICI治疗有无响应的患者的手段仍然是非常有限的,需要提供准确率更高、使用更简便、成本更低、更容易推广应用的生物标志物及其应用手段,以改善免疫检查点阻断疗法以及肿瘤免疫疗法的治疗效果。Therefore, the clinical methods used to distinguish patients who respond to ICI treatment are still very limited, and it is necessary to provide biomarkers and their application methods with higher accuracy, easier use, lower cost, and easier promotion and application. , To improve the therapeutic effect of immune checkpoint blocking therapy and tumor immunotherapy.
早在上世纪60年代,Robert W.Baldwin证实在肿瘤发生发展的极早期阶段,人体免疫系统已经可以产生针对肿瘤细胞的自身抗体,奠定了肿瘤相关抗原(tumor-associated antigen,TAA)研究的理论基础。人体在肿瘤免疫监视过程中产生的自身抗体已经被应用于肿瘤的早期诊断,而且被认为是预测肿瘤治疗疗效的一个极具潜力的生物标志物。例如,已有研究表明,不论EGFR是否突变,肿瘤患者中抗XAGE1(GAGED2a)抗体的存在是XAGE1(GAGED2a)抗原阳性的肿瘤患者OS延长的一个强有力的预测因子。此外,有研究提出抗p53自身抗体、抗PGP9.5自身抗体水平等可以作为预测肺癌复发的工具。As early as the 1960s, Robert W. Baldwin confirmed that in the very early stage of tumorigenesis and development, the human immune system can already produce autoantibodies against tumor cells, laying the foundation for tumor-associated antigen (TAA) research. Base. The human body's autoantibodies produced in the process of tumor immune surveillance have been used in the early diagnosis of tumors, and they are considered to be a potential biomarker for predicting the efficacy of tumor treatment. For example, existing studies have shown that the presence of anti-XAGE1 (GAGED2a) antibodies in tumor patients is a strong predictor of OS prolongation in tumor patients with XAGE1 (GAGED2a) antigen-positive regardless of EGFR mutation. In addition, some studies have proposed that the levels of anti-p53 autoantibodies and anti-PGP9.5 autoantibodies can be used as tools to predict the recurrence of lung cancer.
肿瘤抗原表达与肿瘤浸润免疫细胞的细胞活性也存在相关性。因此,抗肿瘤相关抗原的自身抗体也有可能成为高特异性免疫治疗的靶点或预测标志物。目前已有相关研究报道,包括Sachet a.Shukla鉴定了肿瘤睾丸抗原MAGE-A的一个亚类,其位于Xq28染色体的一个狭窄的75kb区域内,该区域能用于预测抗CTLA-4抗体的疗效。另有研究表明,抗NY-ESO-1和/或 XAGE1肿瘤睾丸抗原的血清抗体可用于预测初始及后线非小细胞肺癌(NSCLC)的ICI疗效和患者生存期。此外,在一项对81名采用抗PD-1/PD-L1抗体进行后线治疗的NSCLC患者的评估中,评估了抗十多种肿瘤相关抗原的自身抗体对抗PD-1治疗的预测价值,表明抗IMP2的自身抗体与肿瘤进展显著相关。但是值得注意的是,迄今为止,就针对肿瘤相关抗原的自身抗体表达和ICI响应之间的关系而言,相关报道仍然是非常有限的;并且,这些自身抗体均为单一的生物标志物,预测可能并不准确,其实际预测效果仍需要进一步的验证。Tumor antigen expression is also related to the cellular activity of tumor infiltrating immune cells. Therefore, autoantibodies against tumor-associated antigens may also become targets or predictive markers for highly specific immunotherapy. There have been related research reports, including Sachet a. Shukla has identified a subclass of tumor testis antigen MAGE-A, which is located in a narrow 75kb region of Xq28 chromosome, which can be used to predict the efficacy of anti-CTLA-4 antibodies . Other studies have shown that serum antibodies against NY-ESO-1 and/or XAGE1 tumor testis antigen can be used to predict the ICI efficacy and patient survival of initial and subsequent non-small cell lung cancer (NSCLC). In addition, in an evaluation of 81 NSCLC patients who were treated with anti-PD-1/PD-L1 antibodies for subsequent treatment, the predictive value of autoantibodies against more than ten tumor-associated antigens was evaluated for anti-PD-1 therapy. It shows that autoantibodies against IMP2 are significantly related to tumor progression. However, it is worth noting that, so far, as far as the relationship between the expression of autoantibodies against tumor-associated antigens and ICI response is concerned, relevant reports are still very limited; and these autoantibodies are all single biomarkers, which are predicted It may not be accurate, and its actual forecasting effect still needs further verification.
免疫检查点抑制剂是一种非常昂贵的药物,与常规化疗不同,虽然免疫检查点抑制剂药物对一些患者有效,但也可能导致严重的不良反应,特别是系统性自身免疫疾病的发展。因此,仍然需要识别新的可用于预测ICI疗效的自身抗体生物标志物,以及开发针对该自身抗体生物标志物的检测用抗原、特别是抗原组合,以提供针对肿瘤免疫治疗效果的新的预测手段。Immune checkpoint inhibitors are very expensive drugs. Unlike conventional chemotherapy, although immune checkpoint inhibitor drugs are effective for some patients, they may also cause serious adverse reactions, especially the development of systemic autoimmune diseases. Therefore, there is still a need to identify new autoantibody biomarkers that can be used to predict the efficacy of ICI, and to develop detection antigens for the autoantibody biomarkers, especially antigen combinations, in order to provide new predictive methods for tumor immunotherapy effects .
发明内容Summary of the invention
为了解决上述技术问题,本发明通过检测肺癌患者血液中针对不同抗原靶点的自身抗体,最终鉴定了一组可用于预测或判断肿瘤、特别是肺癌的免疫检查点抑制剂(ICI)治疗效果的自身抗体生物标志物。In order to solve the above technical problems, the present invention detects autoantibodies against different antigen targets in the blood of lung cancer patients, and finally identifies a group of immune checkpoint inhibitors (ICI) that can be used to predict or judge the therapeutic effect of tumors, especially lung cancer. Autoantibody biomarkers.
因此,本发明的一个目的是提供用于预测或判断肿瘤免疫治疗效果的自身抗体生物标志物组合。Therefore, an object of the present invention is to provide a combination of autoantibody biomarkers for predicting or judging the effect of tumor immunotherapy.
基于自身抗体生物标志物的鉴别,本发明的另一个目的是提供用于检测该自身抗体标志物的试剂,例如抗原蛋白组合,该抗原蛋白组合可用于检测肿瘤患者样本(例如血液)中该自身抗体生物标志物是否为阳性,从而预测或判断肿瘤患者的免疫治疗效果;以及提供包含该检测试剂的试剂盒,其可用于相应的检测。Based on the identification of autoantibody biomarkers, another object of the present invention is to provide reagents for detecting the autoantibody markers, such as an antigen protein combination, which can be used to detect the self in a tumor patient sample (such as blood) Whether the antibody biomarker is positive, so as to predict or judge the immunotherapy effect of tumor patients; and provide a kit containing the detection reagent, which can be used for corresponding detection.
本发明的再一个目的是提供该自身抗体生物标志物组合或抗原蛋白组合在制备用于预测或判断肿瘤免疫治疗效果的产品中的用途。Another object of the present invention is to provide the use of the autoantibody biomarker combination or antigen protein combination in the preparation of products for predicting or judging the effect of tumor immunotherapy.
本发明的还一个目的是提供一种预测或判断肿瘤患者的免疫治疗效果的方法或者一种治疗肿瘤的方法。Another object of the present invention is to provide a method for predicting or judging the effect of immunotherapy of tumor patients or a method for treating tumors.
本发明的技术方案如下。The technical scheme of the present invention is as follows.
一方面,本发明提供一种用于预测或判断受试者的肿瘤免疫治疗效果的生物标志物,所述生物标志物为自身抗体组合,所述自身抗体组合包括选自 抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。In one aspect, the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy in a subject. The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes anti-tumor-related antigens selected from the group consisting of: At least one of autoantibodies: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,所述自身抗体组合可包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2。所述生物标志物可以用于预测或判断:受试者的良好的肿瘤治疗效果;受试者受益于肿瘤免疫治疗;该治疗有效;或,受试者的肿瘤对免疫治疗敏感。With regard to the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the autoantibody combination may include at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY -ESO-1, P53, IMP2. The biomarker can be used to predict or judge: the subject's good tumor treatment effect; the subject benefits from tumor immunotherapy; the treatment is effective; or, the subject's tumor is sensitive to immunotherapy.
优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的两种、三种或四种:Trim21、BRCA2、Annexin 1、HUD;更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:Trim21和BRCA2;进一步优选地,所述自身抗体组合还包括抗以下肿瘤相关抗原的自身抗体中的一种或两种:Annexin 1、HUD。Preferably, the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Tumor-associated antigen autoantibodies: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(A)Trim21,BRCA2,IMP2;(A) Trim21, BRCA2, IMP2;
(B)Trim21,BRCA2,NY-ESO-1;(B) Trim21, BRCA2, NY-ESO-1;
(C)Trim21,BRCA2,NY-ESO-1,IMP2;(C) Trim21, BRCA2, NY-ESO-1, IMP2;
(D)Trim21,BRCA2,P53;(D) Trim21, BRCA2, P53;
(E)Trim21,BRCA2,Annexin 1;(E) Trim21, BRCA2, Annexin 1;
(F)Trim21,BRCA2,Annexin 1,P53;(F) Trim21, BRCA2, Annexin 1, P53;
(G)Trim21,BRCA2,Annexin 1,NY-ESO-1,IMP2;(G) Trim21, BRCA2, Annexin 1, NY-ESO-1, IMP2;
(H)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,IMP2;(H) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, IMP2;
(I)Trim21,BRCA2,Annexin 1,NY-ESO-1,P53,IMP2;(I) Trim21, BRCA2, Annexin 1, NY-ESO-1, P53, IMP2;
(R)Trim21,BRCA2,Annexin 1,HUD;(R) Trim21, BRCA2, Annexin 1, HUD;
(RN)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1;(RN) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1;
(RP)Trim21,BRCA2,Annexin 1,HUD,P53;或(RP) Trim21, BRCA2, Annexin 1, HUD, P53; or
(RNP)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,P53。(RNP) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53.
最优选地,本发明提供一种用于预测或判断受试者的肿瘤免疫治疗效果的生物标志物,所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原Trim21、BRCA2、Annexin 1、HUD的自身抗体,即抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD自身抗体。Most preferably, the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy in a subject, the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes anti-tumor-associated antigen Trim21, BRCA2 , Annexin 1, HUD autoantibodies, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody and anti-HUD autoantibody.
或者,就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,所 述自身抗体组合可包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME。所述生物标志物可以用于预测或判断:受试者的差的肿瘤治疗效果;受试者不受益于肿瘤免疫治疗;该治疗无效;或,受试者的肿瘤对免疫治疗不敏感。Alternatively, in terms of the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the autoantibody combination may include at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, PRAME. The biomarkers can be used to predict or judge: the subject's poor tumor treatment effect; the subject does not benefit from tumor immunotherapy; the treatment is ineffective; or the subject's tumor is not sensitive to immunotherapy.
优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:HSP105,或HSP105和AKAP4。Preferably, the autoantibody combination includes autoantibodies against the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(K)HSP105;(K)HSP105;
(L)HSP105,AKAP4;(L)HSP105, AKAP4;
(M)HSP105,MAGE-A3,AKAP4;或(M) HSP105, MAGE-A3, AKAP4; or
(P)HSP105,AKAP4,PRAME。(P) HSP105, AKAP4, PRAME.
最优选地,本发明提供一种用于预测或判断受试者的免疫肿瘤治疗效果的生物标志物,所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原HSP105、AKAP4的自身抗体,即抗HSP105自身抗体和抗AKAP4自身抗体。Most preferably, the present invention provides a biomarker for predicting or judging a subject's immune tumor treatment effect, the biomarker is a combination of autoantibodies, and the combination of autoantibodies includes anti-tumor-associated antigens HSP105, AKAP4 Anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
根据本发明,所述自身抗体为受试者接受肿瘤免疫治疗之前血清、血浆、组织间隙液、脑脊液或尿液中的自身抗体;优选地,所述自身抗体为IgA(例如IgA1、IgA2)、IgM或IgG(例如IgG1、IgG2、IgG3、IgG4)。According to the present invention, the autoantibody is the autoantibody in serum, plasma, interstitial fluid, cerebrospinal fluid or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG (e.g. IgG1, IgG2, IgG3, IgG4).
根据本发明,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人。并且,优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌。According to the present invention, the subject is a mammal, preferably a primate mammal, more preferably a human. And, preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
根据本发明,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。根据本发明的具体实施方式,所述抗体为纳武单抗、帕姆单抗、信迪利单抗、特瑞普利单抗以及国产的免疫检查点抑制剂,特别是抗PD-1抗体或抗PD-L1抗体。According to the present invention, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy. Treatment or other tumor treatment methods, wherein the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or The CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody. According to a specific embodiment of the present invention, the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 antibodies Or anti-PD-L1 antibody.
根据本发明,该生物标志物即自身抗体组合可以在受试者、例如肿瘤患者的样本(例如血浆或血清)中进行检测,以预测或判断该受试者进行肿瘤免疫治疗的疗效。数据表明,当受试者的血液中这些自身抗体生物标志物为阳性时,其更易于或更不易于从免疫治疗例如ICI治疗中获益。因此,本发明提供的自身抗体生物标志物可用于预测或判断受试者、例如肿瘤患者能否从免疫治疗中获益(该免疫治疗效果为良好或差;该免疫治疗是否有效;或受试者的肿瘤对免疫治疗敏感或不敏感),至少可用于相应的辅助判断。在本发明中,自身抗体生物标志物的“存在”或“不存在”与“阳性”或“阴性”可互换使用;对此进行判断为本领域常规技术。根据本发明的具体实施方式,可以通过参照标准曲线对样本中自身抗体的水平进行定量,进而参照cutoff值进行自身抗体生物标志物的存在(≥cutoff值,为阳性)或不存在(<cutoff值,为阴性)的判断。自身抗体水平的cutoff值可以是来自健康人或健康人群的参照水平;例如,可以被定义为经身体检查确认未患有癌症的人群的平均值加2个标准偏差。According to the present invention, the biomarker, that is, the combination of autoantibodies, can be detected in a sample (such as plasma or serum) of a subject, such as a tumor patient, to predict or judge the efficacy of tumor immunotherapy for the subject. The data indicates that when these autoantibody biomarkers are positive in the blood of a subject, they are more likely or less likely to benefit from immunotherapy such as ICI therapy. Therefore, the autoantibody biomarkers provided by the present invention can be used to predict or judge whether a subject, such as a tumor patient, can benefit from immunotherapy (whether the immunotherapy effect is good or poor; whether the immunotherapy is effective; or whether the subject is effective The patient’s tumor is sensitive or insensitive to immunotherapy), at least for the corresponding auxiliary judgment. In the present invention, "presence" or "absence" and "positive" or "negative" of autoantibody biomarkers can be used interchangeably; judging this is a conventional technique in the art. According to specific embodiments of the present invention, the level of autoantibodies in a sample can be quantified by referring to a standard curve, and then the cutoff value is used to determine the presence (≥cutoff value, positive) or absence of autoantibody biomarkers (<cutoff value) , Is negative) judgment. The cutoff value of the autoantibody level can be a reference level from a healthy person or a healthy person; for example, it can be defined as the average value of a person who is confirmed to have no cancer through a physical examination plus 2 standard deviations.
本发明提供的自身抗体生物标志物可以通过多种方法进行检测,例如可以通过导致该自身抗体出现的肿瘤相关抗原与其之间的抗原-抗体特异性反应进行检测。因此相应地,本发明还提供一种用于检测该自身抗体生物标志物的试剂。The autoantibody biomarker provided by the present invention can be detected by a variety of methods, for example, it can be detected by the antigen-antibody specific reaction between the tumor-associated antigen that causes the autoantibody to appear and the antigen-antibody specific reaction. Therefore, correspondingly, the present invention also provides a reagent for detecting the autoantibody biomarker.
取决于具体技术手段,所述试剂可以是用于酶联免疫吸附法(ELISA)、蛋白/肽段芯片检测、免疫印迹、微珠免疫检测、微流控免疫检测等检测方法的试剂。优选地,所述试剂用于通过抗原抗体反应对本发明的自身抗体生物标志物进行检测,例如通过ELISA。Depending on the specific technical means, the reagents may be reagents used in detection methods such as enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, immunoblotting, bead immunoassay, microfluidic immunoassay, etc. Preferably, the reagent is used to detect the autoantibody biomarker of the present invention through an antigen-antibody reaction, for example, by ELISA.
在这一方面,所述试剂可以是用于检测所述自身抗体组合的抗原蛋白组合,所述抗原蛋白组合包括选自以下抗原蛋白的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。In this aspect, the reagent may be an antigen protein combination for detecting the autoantibody combination, and the antigen protein combination includes at least one selected from the following antigen proteins: Trim21, BRCA2, Annexin 1, HUD, NY- ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
该试剂可用于检测受试者、例如肿瘤患者的样本(例如血浆或血清)中相应自身抗体生物标志物是否为阳性的,进而实现上文所述的对施用肿瘤免疫治疗的临床效果的预测或判断。The reagent can be used to detect whether the corresponding autoantibody biomarker in a sample (such as plasma or serum) of a subject, such as a tumor patient, is positive, so as to realize the prediction or clinical effect of the administration of tumor immunotherapy as described above. judge.
在本发明中,例如,肿瘤相关抗原或抗原蛋白包含的氨基酸序列如下:In the present invention, for example, the amino acid sequence contained in the tumor-associated antigen or antigen protein is as follows:
Trim21包含如SEQ ID NO:1所示的氨基酸序列;Trim21 includes the amino acid sequence shown in SEQ ID NO:1;
BRCA2包含如SEQ ID NO:2所示的氨基酸序列;BRCA2 includes the amino acid sequence shown in SEQ ID NO: 2;
Annexin 1包含如SEQ ID NO:3所示的氨基酸序列; Annexin 1 includes the amino acid sequence shown in SEQ ID NO: 3;
HUD包含如SEQ ID NO:4所示的氨基酸序列;HUD includes the amino acid sequence shown in SEQ ID NO: 4;
NY-ESO-1包含如SEQ ID NO:5所示的氨基酸序列;NY-ESO-1 includes the amino acid sequence shown in SEQ ID NO: 5;
P53包含如SEQ ID NO:6所示的氨基酸序列;P53 includes the amino acid sequence shown in SEQ ID NO: 6;
IMP2包含如SEQ ID NO:7所示的氨基酸序列;IMP2 includes the amino acid sequence shown in SEQ ID NO: 7;
HSP105包含如SEQ ID NO:8所示的氨基酸序列;HSP105 includes the amino acid sequence shown in SEQ ID NO: 8;
MAGE-A3包含如SEQ ID NO:9所示的氨基酸序列;MAGE-A3 includes the amino acid sequence shown in SEQ ID NO: 9;
AKAP4包含如SEQ ID NO:10所示的氨基酸序列;AKAP4 includes the amino acid sequence shown in SEQ ID NO: 10;
PRAME包含如SEQ ID NO:11所示的氨基酸序列。PRAME includes the amino acid sequence shown in SEQ ID NO: 11.
另一方面,本发明提供所述生物标志物或试剂在制备用于预测或判断受试者的肿瘤免疫治疗效果的产品中的用途。如上文所述,所述肿瘤免疫治疗效果包括分别良好的肿瘤免疫治疗效果和差的肿瘤免疫治疗效果,取决于相对应的生物标志物或试剂。In another aspect, the present invention provides the use of the biomarker or reagent in the preparation of a product for predicting or judging the effect of tumor immunotherapy in a subject. As mentioned above, the tumor immunotherapy effects include good tumor immunotherapy effects and poor tumor immunotherapy effects, respectively, depending on the corresponding biomarkers or reagents.
根据本发明,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人。并且,优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌。According to the present invention, the subject is a mammal, preferably a primate mammal, more preferably a human. And, preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
根据本发明,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。根据本发明的具体实施方式,所述抗体为纳武单抗、帕姆单抗、信迪利单抗、特瑞普利单抗,以及国产的免疫检查点抑制剂,特别是抗PD-1抗体或抗PD-L1抗体。According to the present invention, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy. Treatment or other tumor treatment methods, wherein the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or The CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody. According to a specific embodiment of the present invention, the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
又一方面,本发明提供一种试剂盒,所述试剂盒包含本发明所述的试剂。In another aspect, the present invention provides a kit comprising the reagent of the present invention.
取决于具体技术手段,所述试剂盒可以是用于酶联免疫吸附法(ELISA)、蛋白/肽段芯片检测、免疫印迹、微珠免疫检测、微流控免疫检测等对该自身抗体生物标志物进行检测的试剂盒。优选地,所述试剂盒用于通过抗原抗体反应对本发明的自身抗体生物标志物进行检测,例如通过ELISA。Depending on the specific technical means, the kit can be used for enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, western blotting, microbead immunoassay, microfluidic immunoassay, etc. to the autoantibody biomarker Kits for detection of substances. Preferably, the kit is used to detect the autoantibody biomarker of the present invention through an antigen-antibody reaction, for example, by ELISA.
因此,优选地,试剂盒为酶联免疫吸附法(ELISA)检测试剂盒。即,采用该试剂盒,通过酶联免疫吸附法来检测受试者的样本中自身抗体生物标 志物是否是阳性的。相应地,所述试剂盒还可包括用于对自身抗体生物标志物进行ELISA检测的所需其他组分,这些均是本领域公知的。出于检测目的,例如,试剂盒中的抗原蛋白可连接有标签肽,例如His标签、链霉亲和素标签、Myc标签;又如,该试剂盒可以包括固相载体,如具有可固定抗原蛋白的微孔的载体,如酶标板;还可以包括用于将抗原蛋白固定于固相载体上的吸附蛋白、血液如血清的稀释液、洗涤液、带有酶标记的二抗、显色液、终止液等。Therefore, preferably, the kit is an enzyme-linked immunosorbent assay (ELISA) detection kit. That is, using this kit, the enzyme-linked immunosorbent assay is used to detect whether the autoantibody biomarker in the subject's sample is positive. Correspondingly, the kit can also include other components required for ELISA detection of autoantibody biomarkers, all of which are well known in the art. For detection purposes, for example, the antigen protein in the kit can be linked with a tag peptide, such as His tag, streptavidin tag, Myc tag; for another example, the kit can include a solid phase carrier, such as with immobilized antigen Microporous protein carrier, such as an enzyme-labeled plate; it may also include adsorbed protein for immobilizing antigen protein on a solid carrier, dilution of blood such as serum, washing solution, enzyme-labeled secondary antibody, color development Liquid, stop liquid, etc.
还一方面,本发明提供一种用于预测或判断受试者的肿瘤免疫治疗效果的方法。或者,本发明提供一种用于预测或判断受试者的肿瘤对免疫治疗的敏感性的方法。In another aspect, the present invention provides a method for predicting or judging the effect of tumor immunotherapy in a subject. Alternatively, the present invention provides a method for predicting or judging the sensitivity of a subject's tumor to immunotherapy.
上述方法包括检测来自受试者的样本中如下生物标志物是否是阳性的:The above methods include testing whether the following biomarkers in a sample from a subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,该方法可包括检测来自受试者的样本中如下生物标志物是否是阳性的:With regard to the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the method may include detecting whether the following biomarkers in a sample from a subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2。所述生物标志物是阳性时预测或判断:受试者具有良好的肿瘤免疫治疗效果;受试者受益于肿瘤免疫治疗;该治疗有效;或,受试者的肿瘤对免疫治疗敏感。The biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2. When the biomarker is positive, it is predicted or judged that the subject has a good tumor immunotherapy effect; the subject benefits from the tumor immunotherapy; the treatment is effective; or the subject's tumor is sensitive to the immunotherapy.
优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的两种、三种或四种:Trim21、BRCA2、Annexin 1、HUD;更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:Trim21和BRCA2;进一步优选地,所述自身抗体组合还包括抗以下肿瘤相关抗原的自身抗体中的一种或两种:Annexin 1、HUD。Preferably, the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Tumor-associated antigen autoantibodies: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(A)Trim21,BRCA2,IMP2;(A) Trim21, BRCA2, IMP2;
(B)Trim21,BRCA2,NY-ESO-1;(B) Trim21, BRCA2, NY-ESO-1;
(C)Trim21,BRCA2,NY-ESO-1,IMP2;(C) Trim21, BRCA2, NY-ESO-1, IMP2;
(D)Trim21,BRCA2,P53;(D) Trim21, BRCA2, P53;
(E)Trim21,BRCA2,Annexin 1;(E) Trim21, BRCA2, Annexin 1;
(F)Trim21,BRCA2,Annexin 1,P53;(F) Trim21, BRCA2, Annexin 1, P53;
(G)Trim21,BRCA2,Annexin 1,NY-ESO-1,IMP2;(G) Trim21, BRCA2, Annexin 1, NY-ESO-1, IMP2;
(H)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,IMP2;(H) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, IMP2;
(I)Trim21,BRCA2,Annexin 1,NY-ESO-1,P53,IMP2;(I) Trim21, BRCA2, Annexin 1, NY-ESO-1, P53, IMP2;
(R)Trim21,BRCA2,Annexin 1,HUD;(R) Trim21, BRCA2, Annexin 1, HUD;
(RN)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1;(RN) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1;
(RP)Trim21,BRCA2,Annexin 1,HUD,P53;或(RP) Trim21, BRCA2, Annexin 1, HUD, P53; or
(RNP)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,P53。(RNP) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53.
最优选地,本发明提供一种预测或判断受试者的肿瘤免疫治疗效果的方法,或者一种用于预测或判断受试者的肿瘤对免疫治疗的敏感性的方法,该方法包括检测来自受试者的样本中如下生物标志物是否是阳性的:Most preferably, the present invention provides a method for predicting or judging the effect of a subject’s tumor immunotherapy, or a method for predicting or judging the sensitivity of a subject’s tumor to immunotherapy, the method comprising detecting from Whether the following biomarkers in the subject’s sample are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原Trim21、BRCA2、Annexin 1、HUD的自身抗体,即抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD自身抗体。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens Trim21, BRCA2, Annexin 1, and HUD, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens. HUD autoantibodies.
或者,就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,该方法可包括检测来自受试者的样本中如下生物标志物是否是阳性的:Alternatively, in terms of the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the method may include detecting whether the following biomarkers in a sample from the subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME。所述生物标志物是阳性时预测或判断:受试者具有差的肿瘤免疫治疗效果;受试者不受益于肿瘤免疫治疗;该治疗无效;或,受试者的肿瘤对免疫治疗不敏感。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, and PRAME. When the biomarker is positive, it is predicted or judged that the subject has poor tumor immunotherapy effect; the subject does not benefit from tumor immunotherapy; the treatment is ineffective; or the subject's tumor is not sensitive to immunotherapy.
优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体:HSP105,或HSP105和AKAP4。Preferably, the autoantibody combination includes autoantibodies selected from the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(K)HSP105;(K)HSP105;
(L)HSP105,AKAP4;(L)HSP105, AKAP4;
(M)HSP105,MAGE-A3,AKAP4;或(M) HSP105, MAGE-A3, AKAP4; or
(P)HSP105,AKAP4,PRAME。(P) HSP105, AKAP4, PRAME.
最优选地,本发明提供一种预测或判断受试者的肿瘤免疫治疗效果的方法,或者一种用于预测或判断受试者的肿瘤对免疫治疗的敏感性的方法,该 方法包括检测来自受试者的样本中如下生物标志物是否是阳性的:Most preferably, the present invention provides a method for predicting or judging the effect of a subject’s tumor immunotherapy, or a method for predicting or judging the sensitivity of a subject’s tumor to immunotherapy, the method comprising detecting from Whether the following biomarkers in the subject’s sample are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原HSP105、AKAP4的自身抗体,即抗HSP105自身抗体和抗AKAP4自身抗体。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens HSP105 and AKAP4, that is, anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
其中,所述检测可采用本发明的试剂、例如抗原蛋白组合或包含该试剂的试剂盒进行。Wherein, the detection can be carried out using the reagent of the present invention, for example, an antigen-protein combination or a kit containing the reagent.
根据本发明,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人。并且,优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌。According to the present invention, the subject is a mammal, preferably a primate mammal, more preferably a human. And, preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King's lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
根据本发明,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。根据本发明的具体实施方式,所述抗体为纳武单抗、帕姆单抗、信迪利单抗、特瑞普利单抗,以及国产的免疫检查点抑制剂,特别是抗PD-1抗体或抗PD-L1抗体。According to the present invention, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy. Treatment or other tumor treatment methods, wherein the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or The CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody. According to a specific embodiment of the present invention, the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
根据本发明,所述样本为受试者接受肿瘤免疫治疗之前的血清、血浆、组织间隙液、脑脊液或尿液;优选地,所述自身抗体为IgA(例如IgA1、IgA2)、IgM或IgG(例如IgG1、IgG2、IgG3、IgG4)。According to the present invention, the sample is serum, plasma, interstitial fluid, cerebrospinal fluid, or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG ( For example, IgG1, IgG2, IgG3, IgG4).
例如,所述方法包括以下步骤:For example, the method includes the following steps:
(1)从所述受试者获得样本;(1) Obtain a sample from the subject;
(2)检测所述样本中本发明的自身抗体生物标志物是否是阳性的;(2) Detecting whether the autoantibody biomarker of the present invention in the sample is positive;
(3)当样本中所述自身抗体生物标志物为阳性时,预测或判断:受试者具有良好或差的肿瘤免疫治疗效果;受试者受益于或不受益于肿瘤免疫治疗;该治疗有效或无效;或,受试者的肿瘤对免疫治疗敏感或不敏感。(3) When the autoantibody biomarker in the sample is positive, predict or judge: the subject has a good or poor tumor immunotherapy effect; the subject benefits or does not benefit from tumor immunotherapy; the treatment is effective Or ineffective; or, the subject's tumor is sensitive or insensitive to immunotherapy.
还一方面,本发明提供一种治疗受试者的肿瘤的方法,该方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:In yet another aspect, the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,该方法可包括检测来自受试者的样本中如下生物标志物是否是阳性的:With regard to the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the method may include detecting whether the following biomarkers in a sample from a subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2。所述生物标志物是阳性的时,使受试者进行肿瘤免疫治疗。The biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2. When the biomarker is positive, the subject is subjected to tumor immunotherapy.
优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的两种、三种或四种:Trim21、BRCA2、Annexin 1、HUD;更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:Trim21和BRCA2;进一步优选地,所述自身抗体组合还包括抗以下肿瘤相关抗原的自身抗体中的一种或两种:Annexin 1、HUD。Preferably, the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD; more preferably, the autoantibody combination includes anti- Autoantibodies against tumor-associated antigens: Trim21 and BRCA2; further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(A)Trim21,BRCA2,IMP2;(A) Trim21, BRCA2, IMP2;
(B)Trim21,BRCA2,NY-ESO-1;(B) Trim21, BRCA2, NY-ESO-1;
(C)Trim21,BRCA2,NY-ESO-1,IMP2;(C) Trim21, BRCA2, NY-ESO-1, IMP2;
(D)Trim21,BRCA2,P53;(D) Trim21, BRCA2, P53;
(E)Trim21,BRCA2,Annexin 1;(E) Trim21, BRCA2, Annexin 1;
(F)Trim21,BRCA2,Annexin 1,P53;(F) Trim21, BRCA2, Annexin 1, P53;
(G)Trim21,BRCA2,Annexin 1,NY-ESO-1,IMP2;(G) Trim21, BRCA2, Annexin 1, NY-ESO-1, IMP2;
(H)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,IMP2;(H) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, IMP2;
(I)Trim21,BRCA2,Annexin 1,NY-ESO-1,P53,IMP2;(I) Trim21, BRCA2, Annexin 1, NY-ESO-1, P53, IMP2;
(R)Trim21,BRCA2,Annexin 1,HUD;(R) Trim21, BRCA2, Annexin 1, HUD;
(RN)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1;(RN) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1;
(RP)Trim21,BRCA2,Annexin 1,HUD,P53;或(RP) Trim21, BRCA2, Annexin 1, HUD, P53; or
(RNP)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,P53。(RNP) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53.
最优选地,本发明提供一种治疗受试者的肿瘤的方法,该方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:Most preferably, the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原Trim21、BRCA2、Annexin 1、HUD的自身抗体,即抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD自身抗体。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens Trim21, BRCA2, Annexin 1, and HUD, namely anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody, and anti-tumor related antigens. HUD autoantibodies.
或者,就本发明提供的生物标志物所指示的肿瘤免疫治疗效果而言,该 方法可包括检测来自受试者的样本中如下生物标志物是否是阳性的:Alternatively, in terms of the tumor immunotherapy effect indicated by the biomarkers provided by the present invention, the method may include detecting whether the following biomarkers in a sample from the subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME。所述生物标志物是阳性的时,使受试者不进行肿瘤免疫治疗。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, and PRAME. When the biomarker is positive, the subject is prevented from undergoing tumor immunotherapy.
优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体:HSP105,或HSP105和AKAP4。Preferably, the autoantibody combination includes autoantibodies selected from the following tumor-associated antigens: HSP105, or HSP105 and AKAP4.
根据本发明的具体实施方式,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:According to a specific embodiment of the present invention, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
(K)HSP105;(K)HSP105;
(L)HSP105,AKAP4;(L)HSP105, AKAP4;
(M)HSP105,MAGE-A3,AKAP4;或(M) HSP105, MAGE-A3, AKAP4; or
(P)HSP105,AKAP4,PRAME。(P) HSP105, AKAP4, PRAME.
最优选地,本发明提供一种治疗受试者的肿瘤的方法,该方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:Most preferably, the present invention provides a method for treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
所述生物标志物为自身抗体组合,所述自身抗体组合包括抗肿瘤相关抗原HSP105、AKAP4的自身抗体,即抗HSP105自身抗体和抗AKAP4自身抗体。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes autoantibodies against tumor-associated antigens HSP105 and AKAP4, that is, anti-HSP105 autoantibodies and anti-AKAP4 autoantibodies.
其中,所述检测可采用本发明的试剂、例如抗原蛋白组合或包含该试剂的试剂盒进行。Wherein, the detection can be carried out using the reagent of the present invention, for example, an antigen-protein combination or a kit containing the reagent.
根据本发明,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人。并且,优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌。According to the present invention, the subject is a mammal, preferably a primate mammal, more preferably a human. And, preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodge King’s lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer.
根据本发明,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。根据本发明的具体实施方式,所述抗体为纳武单抗、帕姆单抗、信迪利单抗、特瑞普利单抗,以及国产的免疫检查点抑制剂,特别是抗PD-1抗体或抗PD-L1抗体。According to the present invention, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy. Treatment or other tumor treatment methods, wherein the immune checkpoint inhibitor is for PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or The CD160 immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody. According to a specific embodiment of the present invention, the antibodies are nivolumab, pambrolizumab, sintilizumab, teriprizumab, and domestic immune checkpoint inhibitors, especially anti-PD-1 Antibody or anti-PD-L1 antibody.
根据本发明,所述样本为受试者接受肿瘤免疫治疗之前的血清、血浆、 组织间隙液、脑脊液或尿液;优选地,所述自身抗体为IgA(例如IgA1、IgA2)、IgM或IgG(例如IgG1、IgG2、IgG3、IgG4)。According to the present invention, the sample is serum, plasma, interstitial fluid, cerebrospinal fluid, or urine before the subject receives tumor immunotherapy; preferably, the autoantibody is IgA (for example, IgA1, IgA2), IgM or IgG ( For example, IgG1, IgG2, IgG3, IgG4).
例如,所述方法包括以下步骤:For example, the method includes the following steps:
(1)从所述受试者获得样本;(1) Obtain a sample from the subject;
(2)检测所述样本中该自身抗体生物标志物是否是阳性的;其中优选地采用酶联免疫吸附法进行检测。(2) Detect whether the autoantibody biomarker in the sample is positive; wherein the enzyme-linked immunosorbent method is preferably used for detection.
当样本中所述自身抗体生物标志物为阳性时,使受试者进行肿瘤免疫治疗,或者使受试者不进行肿瘤免疫治疗。When the autoantibody biomarker in the sample is positive, the subject is allowed to undergo tumor immunotherapy, or the subject is not allowed to undergo tumor immunotherapy.
与现有技术相比,本发明提供了一种用于预测或判断肿瘤免疫治疗效果的生物标志物,所述生物标志物为自身抗体组合。并且本发明的自身抗体组合包括了预测良好的肿瘤免疫治疗效果和差的肿瘤免疫治疗效果的两种组合,前者可称为对肿瘤免疫治疗效果的正向预测,而后者可称为对肿瘤免疫治疗效果的负向预测。Compared with the prior art, the present invention provides a biomarker for predicting or judging the effect of tumor immunotherapy, and the biomarker is a combination of autoantibodies. And the autoantibody combination of the present invention includes two combinations that predict good tumor immunotherapy effect and poor tumor immunotherapy effect. The former can be called a positive prediction of tumor immunotherapy effect, and the latter can be called tumor immunity. Negative prediction of treatment effect.
实验表明,不论PD-L1表达水平与TMB水平,无论是免疫一线治疗还是后线治疗,就正向预测的自身抗体组合,检测阳性的肿瘤患者的免疫检查点阻断治疗的有效率要显著高于该自身抗体组合检测阴性的肿瘤患者(P<0.05);同样地,无论是免疫单药治疗还是免疫联合化疗,这些肿瘤患者的免疫检查点阻断治疗的有效率同样显著高于自身抗体组合检测阴性的肿瘤患者(P<0.05),尤其是采用免疫单药治疗时,前者的有效率更为显著。而就负向预测的自身抗体组合而言,无论是免疫一线治疗还是后线治疗,无论是免疫单药治疗还是免疫联合化疗,检测阳性的肿瘤患者的免疫检查点阻断治疗的有效率要显著低于该自身抗体组合检测阴性的肿瘤患者(P<0.05)。Experiments have shown that regardless of PD-L1 expression level and TMB level, whether it is first-line or post-line immunotherapy, the combination of autoantibodies is positively predicted, and the effective rate of immune checkpoint blocking therapy for tumor patients with positive detection is significantly higher. For tumor patients who tested negative for the autoantibody combination (P<0.05); similarly, whether it is immune monotherapy or immune combination chemotherapy, the effective rate of immune checkpoint blocking therapy in these tumor patients is also significantly higher than that of the autoantibody combination Tumor patients with negative tests (P<0.05), especially when using immune monotherapy, the former is more effective. Regarding the combination of negatively predicted autoantibodies, whether it is immune first-line therapy or post-line therapy, whether it is immune monotherapy or immune combination chemotherapy, the effective rate of immune checkpoint blocking therapy for tumor patients who test positive is more significant. Lower than the autoantibody combination test negative tumor patients (P<0.05).
因此,就肿瘤患者能否从免疫治疗、特别是免疫检查点抑制剂治疗中获益,本发明提供的自身抗体生物标志物能够提供准确的预测或判断结果。基于该预测或判断结果,患者或临床医生可以更好地决定患者是否要进行免疫治疗,从而避免过度医疗,降低治疗成本,减少或避免不良反应产生。Therefore, whether tumor patients can benefit from immunotherapy, especially immune checkpoint inhibitor therapy, the autoantibody biomarkers provided by the present invention can provide accurate prediction or judgment results. Based on the prediction or judgment result, the patient or clinician can better decide whether the patient needs immunotherapy, so as to avoid excessive medical treatment, reduce treatment costs, and reduce or avoid adverse reactions.
并且,根据需要,本发明提供的这两种自身抗体组合还可单独使用或组合使用。例如,在组合使用时,可以综合正向预测的自身抗体生物标志物的阳性情况与负向预测的自身抗体生物标志物的阴性情况。Moreover, the two autoantibody combinations provided by the present invention can also be used alone or in combination as required. For example, when used in combination, the positive conditions of the autoantibody biomarkers predicted in the positive direction and the negative conditions of the autoantibody biomarkers predicted in the negative direction can be combined.
附图说明Description of the drawings
以下,结合附图来详细说明本发明的实施方案,其中:Hereinafter, the embodiments of the present invention will be described in detail with reference to the accompanying drawings, in which:
图1显示了治疗前显示自身抗体组合阳性或阴性的患者在治疗后肿瘤对免疫治疗的响应,其中图1-A:P_Ab.组合,图1-B:N_Ab.组合。Figure 1 shows the tumor response to immunotherapy after the treatment of patients who showed positive or negative autoantibody combination before treatment, where Figure 1-A: P_Ab. combination, Figure 1-B: N_Ab. combination.
图2显示了治疗前显示自身抗体组合阳性或阴性的患者在治疗后的生存曲线,其中图2-A:训练集,图2-B:验证集。Figure 2 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, where Figure 2-A: training set and Figure 2-B: validation set.
图3显示了治疗前显示自身抗体组合阳性或阴性的患者在治疗后的生存曲线,其中图3-A至图3-RNP分别示出对应自身抗体组合A至自身抗体组合RNP的结果。Figure 3 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, wherein Figure 3-A to Figure 3-RNP respectively show the results of corresponding autoantibody combination A to autoantibody combination RNP.
图4显示了治疗前显示自身抗体组合阳性或阴性的患者在治疗后的生存曲线,其中图4-K至图4-P分别示出对应自身抗体组合K至自身抗体组合P的结果。Figure 4 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, wherein Figure 4-K to Figure 4-P show the results of corresponding autoantibody combination K to autoantibody combination P, respectively.
图5显示了治疗前显示自身抗体或自身抗体组合阳性或阴性的患者在治疗后的生存曲线,其中图5-A:IMP2,图5-B:抗XAGE-1和抗NY-ESO-1。Figure 5 shows the survival curve of patients who showed positive or negative autoantibody or autoantibody combination before treatment after treatment, where Figure 5-A: IMP2, Figure 5-B: anti-XAGE-1 and anti-NY-ESO-1.
图6显示了治疗前显示自身抗体组合阳性或阴性的患者在治疗后的生存曲线,其中图6-A:一线免疫治疗,图6-B:后线免疫治疗,图6-C:免疫单药治疗),图6-D:免疫治疗结合化疗。Figure 6 shows the survival curve of patients who showed positive or negative autoantibody combination before treatment after treatment, where Figure 6-A: first-line immunotherapy, Figure 6-B: posterior-line immunotherapy, and Figure 6-C: immune single agent Treatment), Figure 6-D: Immunotherapy combined with chemotherapy.
实施发明的最佳方式The best way to implement the invention
在本发明中,术语“抗原”或术语“抗原蛋白”可互换使用。此外,本发明中涉及以下实验操作或定义。应注意,本发明还可采用本领域其他常规技术进行实施,并不仅限于以下实验操作。In the present invention, the term "antigen" or the term "antigenic protein" can be used interchangeably. In addition, the following experimental operations or definitions are involved in the present invention. It should be noted that the present invention can also be implemented using other conventional techniques in the field, and is not limited to the following experimental operations.
(一)抗原蛋白的制备与固定(1) Preparation and fixation of antigen protein
将肿瘤相关抗原(TAA)的cDNA克隆到含6XHis标记的PET28(a)表达载体上。在抗原的N端或C端,引入链霉亲和素蛋白或类似物(结合生物素的标签蛋白)。获得的重组表达载体转化大肠杆菌进行表达,当蛋白在包涵体表达后,用6M盐酸胍对蛋白进行变性处理,并在体外按照标准方法进行复性折叠,然后通过6XHis标签进行Ni-NTA亲和柱纯化,获得抗原蛋白。The cDNA of the tumor-associated antigen (TAA) was cloned into the 6XHis-labeled PET28(a) expression vector. At the N-terminus or C-terminus of the antigen, a streptavidin protein or the like (biotin-binding tag protein) is introduced. The obtained recombinant expression vector was transformed into Escherichia coli for expression. After the protein was expressed in the inclusion body, the protein was denatured with 6M guanidine hydrochloride, and renatured in vitro according to standard methods, and then Ni-NTA affinity was carried out by 6XHis tag Column purification to obtain antigen protein.
(二)血浆的制备(2) Preparation of plasma
在免疫治疗前一周至1天内取静脉血在EDTA处理过或柠檬酸处理过的采血管中。然后室温1000-2000RCF离心15min;离心后,在室温下轻轻地把上清转移到另一个干净的离心管中,置于-80℃冰箱中长期保存。Venous blood was collected in EDTA-treated or citric acid-treated blood collection tubes one week to one day before immunotherapy. Then centrifuge at 1000-2000RCF at room temperature for 15 minutes; after centrifugation, gently transfer the supernatant to another clean centrifuge tube at room temperature, and place it in a refrigerator at -80°C for long-term storage.
(三)自身抗体的ELISA检测与定量(3) ELISA detection and quantification of autoantibodies
将制得的抗原蛋白包被到96孔固相板的微孔表面。采用间接包被:96- 孔固相板过夜包被5-10ug/ml的生物素标记的牛血清白蛋白;第2天,洗去固相板微孔中未包被的牛血清白蛋白,加入300uL含有BSA的封闭液室温封闭1h;加入抗原蛋白孵育1.5h,然后洗去未吸附的抗原蛋白。包被抗原蛋白后,微孔中加入300ul的含有BSA的稳定液,孵育1h后现用或真空干燥备用。The prepared antigen protein is coated on the surface of the microwell of a 96-well solid phase plate. Using indirect coating: a 96-well solid phase plate was coated overnight with 5-10ug/ml biotin-labeled bovine serum albumin; on the second day, the uncoated bovine serum albumin in the microwells of the solid phase plate was washed away. Add 300uL of blocking solution containing BSA to block at room temperature for 1h; add antigen protein and incubate for 1.5h, then wash off unadsorbed antigen protein. After coating the antigen protein, add 300ul of the stabilized solution containing BSA to the microwells, incubate for 1h, and then use it or dry it under vacuum for later use.
如上,纯化的抗原蛋白通过生物素与链霉亲和素之间的特异反应间接包被到固相板表面。往包被抗原蛋白的微孔中加入稀释好的血浆样本,经孵育使血浆样本中的自身抗体与固相板表面上的抗原蛋白发生特异性结合。洗掉没有结合的自身抗体,加入辣根过氧化物酶标记抗人IgG抗体,第二次孵育使该酶标记抗人IgG抗体与吸附到固相板表面上的自身抗体结合,形成抗原-抗体-酶标记抗体的复合物,继而洗掉没有结合的酶标记抗人IgG抗体,加入显色剂底物反应后用酶标仪在450nm波长下测定吸光度。最终通过与cutoff值相比较来判断自身抗体检测结果为阴性还是阳性。检测步骤如下:As above, the purified antigen protein is indirectly coated on the surface of the solid phase plate through the specific reaction between biotin and streptavidin. The diluted plasma sample is added to the microwells coated with the antigen protein, and the autoantibodies in the plasma sample are specifically combined with the antigen protein on the surface of the solid phase plate after incubation. Wash off unbound autoantibodies, add horseradish peroxidase-labeled anti-human IgG antibody, and incubate for the second time so that the enzyme-labeled anti-human IgG antibody binds to the autoantibody adsorbed on the surface of the solid phase plate to form an antigen-antibody -Enzyme-labeled antibody complex, then wash away the unbound enzyme-labeled anti-human IgG antibody, add color reagent substrate to react and measure the absorbance with a microplate reader at 450nm wavelength. Finally, it is judged whether the autoantibody test result is negative or positive by comparing with the cutoff value. The detection steps are as follows:
一、准备步骤1. Preparation steps
1.将检测用试剂放置室温至少30min使试剂恢复到室温。1. Place the reagents for detection at room temperature for at least 30 minutes to restore the reagents to room temperature.
2.稀释待测血浆样本:在1.5ml EP管内加入545ul的样本稀释液(PBS,含1%BSA),再将5ul待测血浆样本加入到样本稀释液中(可根据所需量自行调整样本量,血浆样本与样本稀释液的体积比为1∶109),上下颠倒5-6次轻轻混匀。2. Dilute the plasma sample to be tested: Add 545ul of the sample diluent (PBS, containing 1% BSA) into the 1.5ml EP tube, and then add 5ul of the plasma sample to be tested to the sample diluent (the sample can be adjusted according to the required amount The volume ratio of the plasma sample to the sample diluent is 1:109), turn it upside down 5-6 times and mix gently.
3.将各待测血浆样本稀释混匀后,转移530ul到干净的深孔槽。3. After diluting and mixing each plasma sample to be tested, transfer 530ul to a clean deep hole tank.
4.制备洗液工作液:将10倍PBST洗液,用纯化水或蒸馏水稀释10倍,配成原倍洗液,备用。4. Prepare the working solution of lotion: Dilute the 10 times PBST lotion with purified water or distilled water 10 times to make the original lotion and set aside.
5.PBS缓冲液:自备,pH值为7.6。5. PBS buffer: prepare your own, with a pH of 7.6.
二、检测步骤Second, the detection steps
1.加一抗:用PBS缓冲液270ul/孔洗酶标板1次后,向酶标板加入50ul/孔已经稀释好的待测血浆,室温在微孔振荡器上反应1h。1. Add primary antibody: After washing the microtiter plate with 270ul/well of PBS buffer once, add 50ul/well of the diluted plasma to the microtiter plate, and react on the microwell shaker for 1 hour at room temperature.
2.加二抗:使用前配制二抗(恢复到室温的辣根过氧化物酶标记抗人IgG抗体浓缩液∶酶结合物稀释液=1∶19,该稀释液为含1%BSA的PBS)。用1*PBST洗液270ul/孔洗板3次,每次均拍干,然后以50ul/孔加入二抗稀释液,贴膜,室温在微孔振荡器上反应0.5h。2. Add secondary antibody: prepare secondary antibody before use (horseradish peroxidase-labeled anti-human IgG antibody concentrated solution returned to room temperature: enzyme conjugate dilution solution = 1:19, the dilution solution is PBS containing 1% BSA ). Wash the plate 3 times with 1*PBST washing solution 270ul/well, pat dry each time, then add the secondary antibody diluent at 50ul/well, stick the membrane, and react on the microwell shaker at room temperature for 0.5h.
3.加显色剂:使用前配制显色剂(显色剂A液∶显色剂B液=1∶19)。用1*洗液270ul/孔洗板3次,每次均拍干,然后以100ul/孔加入显色剂,加第一行开始计时,贴膜,室温在微孔振荡器上反应15min。3. Add color developer: prepare color developer (liquid developer A: liquid developer B=1:19) before use. Wash the plate 3 times with 1*washing solution 270ul/well, pat dry each time, then add the color developer at 100ul/well, add the first line to start timing, stick the film, and react on the microporous shaker at room temperature for 15 minutes.
4.终止并读数:按照加入显色剂的顺序,加入终止液50ul/孔,在酶标仪450nm下读数。4. Stop and read: Add 50ul/well of stop solution according to the order of adding color reagent, and read at 450nm in the microplate reader.
5.阴阳性结果判断:OD值通过与cutoff值相比较来判断检测结果。5. Judgment of negative and positive results: The OD value is compared with the cutoff value to determine the test result.
(四)自身抗体的临界值(cutoff值)(4) Cutoff value of autoantibody (cutoff value)
自身抗体水平的cutoff值被定义为等于对照组(经身体检查确认未患有癌症的人群)中健康对照队列的平均值加2个标准偏差(SD)。The cutoff value of the autoantibody level is defined as equal to the average value of the healthy control cohort in the control group (people confirmed to have no cancer through physical examination) plus 2 standard deviations (SD).
(五)单个自身抗体的阳性判断(5) The positive judgment of a single autoantibody
对样本中自身抗体的水平进行定量后,将其与cutoff值进行比较,≥cutoff值为阳性,<cutoff值为阴性。After quantifying the level of autoantibodies in the sample, compare it with the cutoff value. ≥cutoff value is positive, and <cutoff value is negative.
(六)自身抗体组合的阳性判断(6) Positive judgment of autoantibody combination
由于单个自身抗体的阳性率低,为了增加自身抗体检出的阳性率,分析结果时联合多个自身抗体的结果来判断预测效果。规则是:在患者样本中检测多个自身抗体,只要有其中一个或者多个自身抗体显示阳性,则判断抗体组合结果为阳性;而如果所有的自身抗体均为阴性,则判断抗体组合结果为阴性。Because the positive rate of a single autoantibody is low, in order to increase the positive rate of autoantibody detection, the results of multiple autoantibodies are combined to judge the predictive effect when analyzing the results. The rule is: multiple autoantibodies are detected in a patient sample. As long as one or more of the autoantibodies are positive, the antibody combination is judged to be positive; and if all autoantibodies are negative, the antibody combination is judged to be negative .
(七)临床疗效评估指标(7) Clinical efficacy evaluation indicators
根据实体肿瘤的疗效评价标准1.1版(Response Evaluation Criteria in Solid Tumors RECIST Version 1.1,RECIST v1.1)评估基线(治疗前)时的目标病灶,记录目标病灶最长直径的基线和,用于确定客观反应。According to the response evaluation criteria for solid tumors Version 1.1 (Response Evaluation Criteria in Solid Tumors RECIST Version 1.1, RECIST v1.1), the target lesion at baseline (before treatment) is evaluated, and the baseline sum of the longest diameter of the target lesion is recorded to determine the objective reaction.
BOR:最佳疗效,是指考虑了各种因素后确认的从治疗研究开始到治疗结束的最佳疗效的记录。BOR: The best curative effect refers to the record of the best curative effect from the beginning of the treatment study to the end of the treatment, which is confirmed after considering various factors.
PD:与治疗前所有靶病灶直径和的最小值相比,所有靶病灶直径的总和至少增加20%且直径总和增加的绝对值还必须大于5mm;或者出现新的病灶。PD: Compared with the minimum sum of the diameters of all target lesions before treatment, the sum of the diameters of all target lesions is increased by at least 20% and the absolute value of the total increase in diameter must be greater than 5mm; or new lesions appear.
PR:与治疗前所有靶病灶的直径和相比,所有靶病灶直径的总和至少减小30%。PR: Compared with the sum of the diameters of all target lesions before treatment, the sum of the diameters of all target lesions is reduced by at least 30%.
SD:与治疗前所有靶病灶直径和的最小值相比,靶病灶的缩小程度不符合部分缓解(PR),增大程度不符合疾病进展(PD),是指一种介于PR和PD之间的状态。SD: Compared with the minimum sum of the diameters of all target lesions before treatment, the shrinkage of the target lesion does not meet the partial remission (PR), and the increase does not meet the disease progression (PD). It refers to a type between PR and PD. The state of the time.
CR:所有靶病灶消失,任何病理性淋巴结(无论是否为靶病灶)的短轴 值必须<10mm。CR: All target lesions disappear, and the short axis value of any pathological lymph node (whether it is a target lesion or not) must be less than 10mm.
PFS:无进展生存时间,即从随机化开始至疾病复发或由于各种原因导致患者死亡的时间。PFS: Progression-free survival time, that is, the time from the beginning of randomization to the recurrence of the disease or the death of the patient due to various reasons.
mPFS:中位无进展生存时间,即从随机化开始至疾病复发或由于各种原因导致患者死亡的中位时间。mPFS: Median progression-free survival time, that is, the median time from randomization to disease recurrence or death due to various reasons.
PD-L1表达水平:采用免疫组化法进行,评估有任何强度PD-L1膜染色的肿瘤细胞在所有肿瘤细胞中所占的百分比,将检测结果分为四组,即阴性(小于1%),低表达组(1%-49%),高表达组(≥50%),未知。PD-L1 expression level: Use immunohistochemistry to evaluate the percentage of tumor cells stained with PD-L1 membrane of any intensity in all tumor cells. The test results are divided into four groups, namely negative (less than 1%) , Low expression group (1%-49%), high expression group (≥50%), unknown.
(八)统计分析方法(8) Statistical analysis methods
使用GraphPad Prism v.6(Graphpad Prism软件,加利福尼亚州圣地亚哥)和针对Windows的IBM SPSS Statistics 23(IBM,纽约,纽约),使用Mann-Whitney U检验对两组进行统计学分析。在分析每个参数之间的关系时,执行了Spearman的相关分析。通过Kaplan-Meier方法分析了中位无进展生存(median Progression Free Survival,mPFS)。使用对数秩检验分析患者亚组之间的mPFS差异。Use GraphPad Prism v.6 (Graphpad Prism software, San Diego, California) and IBM SPSS Statistics 23 for Windows (IBM, New York, New York), and use Mann-Whitney U test to perform statistical analysis on the two groups. When analyzing the relationship between each parameter, Spearman's correlation analysis was performed. The Kaplan-Meier method was used to analyze the median Progression Free Survival (mPFS). The log-rank test was used to analyze differences in mPFS between patient subgroups.
以下参照具体的实施例来说明本发明。本领域技术人员能够理解,这些实施例仅用于说明本发明,其不以任何方式限制本发明的范围。样本采集已经患者知情同意,且获得监管部门(上海肺科医院审查委员会)的批准。Hereinafter, the present invention will be explained with reference to specific embodiments. Those skilled in the art can understand that these embodiments are only used to illustrate the present invention, and they do not limit the scope of the present invention in any way. The sample collection has been informed by the patient's consent and approved by the regulatory authority (Shanghai Pulmonary Hospital Review Committee).
下述实施例中的实验方法,如无特殊说明,均为常规方法。下述实施例中所用的药材原料、试剂材料等,如无特殊说明,均为市售购买产品。The experimental methods in the following examples, unless otherwise specified, are all conventional methods. The medicinal materials, reagent materials, etc. used in the following examples are all commercially available products unless otherwise specified.
实施例1Example 1
为了寻找能指示免疫治疗效果的自身抗体,首先用47例正常健康人(健康对照组)和47例诊断为肺癌患者(肺癌组)的血浆,检测肺癌患者是否存在针对纯化的抗原蛋白的自身抗体。健康对照组人群是体检人员中过去和现在没有被诊断为癌症的人。肺癌组人群为确诊为肺癌的患者,包括小细胞肺癌10人,肺鳞癌12人,肺腺癌19人,其他亚型肺癌6人,并在治疗前抽取血浆。实验人群信息见表1。In order to find autoantibodies that can indicate the effect of immunotherapy, first use the plasma of 47 normal healthy people (healthy control group) and 47 patients diagnosed with lung cancer (lung cancer group) to detect whether there are autoantibodies against purified antigen protein in lung cancer patients. . The healthy control group are those who have not been or are not diagnosed with cancer among the medical examiners. The lung cancer group includes 10 patients with small cell lung cancer, 12 patients with lung squamous cell carcinoma, 19 patients with lung adenocarcinoma, and 6 patients with other subtypes of lung cancer. Plasma was collected before treatment. The information of the experimental population is shown in Table 1.
表1.初筛实验人群信息Table 1. Information on the initial screening test population
Figure PCTCN2021076771-appb-000001
Figure PCTCN2021076771-appb-000001
Figure PCTCN2021076771-appb-000002
Figure PCTCN2021076771-appb-000002
为了平行对比各个抗原对应的自身抗体在健康对照组人群和肺癌组人群中的浓度,这里将每个抗原的检测特异性设定为≥93.6%(在47例的健康对照组人群中有≤6.4%的阳性率);而就在肺癌组人群中的敏感性(阳性率,即自身抗体阳性占肺癌患者总人数47例的比例),和其他文献发现的类似,单个自身抗体的阳性检出率低,通常在5-20%之间。因此,在初步筛选肺癌相关抗原中,根据敏感性将抗原蛋白分为四组。筛选的抗原蛋白以及经检测确定的灵敏度和特异性见表2。In order to compare the concentration of autoantibodies corresponding to each antigen in the healthy control group and the lung cancer group in parallel, the detection specificity of each antigen is set to ≥93.6% (in the healthy control group of 47 cases, there are ≤6.4 % Positive rate); and the sensitivity in the lung cancer group (positive rate, that is, the proportion of autoantibody positive in the total number of lung cancer patients in 47 cases) is similar to that found in other literature, the positive detection rate of a single autoantibody Low, usually between 5-20%. Therefore, in the preliminary screening of lung cancer-related antigens, antigen proteins are divided into four groups based on sensitivity. The screened antigen protein and the sensitivity and specificity determined by the test are shown in Table 2.
表2.肿瘤相关抗原的初筛结果Table 2. Results of preliminary screening of tumor-associated antigens
Figure PCTCN2021076771-appb-000003
Figure PCTCN2021076771-appb-000003
Figure PCTCN2021076771-appb-000004
Figure PCTCN2021076771-appb-000004
实施例2Example 2
在免疫治疗患者(表3)的基线(治疗前)血浆中检测自身抗体,采用实施例1表2前面三组(敏感性>5%)的抗原蛋白,筛选和肺癌患者治疗疗效相关的自身抗体。Autoantibodies were detected in the baseline (pre-treatment) plasma of immunotherapy patients (Table 3). The antigen proteins of the first three groups (sensitivity> 5%) in Example 1 and Table 2 were used to screen for autoantibodies related to the therapeutic efficacy of lung cancer patients. .
为了能筛选到适合临床应用的免疫治疗预测标志物,本研究尽量入组肺癌患者免疫治疗的各种场景。有38个肺癌患者使用免疫治疗为一线治疗,而40个肺癌患者先进行了化疗、靶向治疗等一种或多种治疗,然后再选择免疫治疗(后线治疗)。此外,入组患者使用的免疫治疗包括免疫检查点抑制剂,既包含进口的纳武单抗、派姆单抗,也包括国产的免疫检查点抑制剂(表3)。无论是一线还是后线免疫治疗,肺癌患者的治疗方案包括两种情况:免疫治疗为单一疗法(25个患者),或者免疫联合化疗(53个患者)。In order to be able to screen for predictive markers of immunotherapy suitable for clinical application, this study tries to include various scenarios of immunotherapy for patients with lung cancer. Thirty-eight lung cancer patients used immunotherapy as the first-line treatment, while 40 lung cancer patients received one or more treatments such as chemotherapy and targeted therapy, and then chose immunotherapy (post-line treatment). In addition, the immunotherapy used by the enrolled patients included immune checkpoint inhibitors, including imported nivolumab and pembrolizumab, as well as domestic immune checkpoint inhibitors (Table 3). Regardless of whether it is first-line or second-line immunotherapy, the treatment plan for lung cancer patients includes two situations: immunotherapy is monotherapy (25 patients), or immunotherapy combined with chemotherapy (53 patients).
首先,将初筛出来的肺癌相关抗原作为抗原蛋白,将一线免疫治疗患者的基线血浆作为“训练集”进行自身抗体的ELISA检测,然后将判断为阳性的自身抗体结果(参照上文所述的cutoff值)与肿瘤对免疫治疗的反应结果(BOR)进行比对。结果显示,自身抗体阳性的检测结果和免疫治疗疗效的关系归为三类(见表4):First, use the lung cancer-related antigens initially screened as the antigen protein, and use the baseline plasma of first-line immunotherapy patients as the "training set" for autoantibody ELISA testing, and then determine the positive autoantibody results (refer to the above The cutoff value) is compared with the tumor response to immunotherapy (BOR). The results show that the relationship between positive autoantibody test results and immunotherapy efficacy is classified into three categories (see Table 4):
一部分肿瘤相关抗原属于“正相关”抗原,本发明中将针对这种抗原的自身抗体命名为“P_Ab.”,显示这种自身抗体阳性信号的患者均基本实现了 BOR中“PR”和“SD”,即良好的疗效,并且患者人群整体上满足“PR”百分比/“PD”百分比≥2。将这类自身抗体初步确定为正向预测抗体,具有免疫治疗疗效良好的正向预测效果。Some tumor-associated antigens belong to "positively related" antigens. In the present invention, autoantibodies against this antigen are named "P_Ab.". Patients showing positive signals of such autoantibodies have basically achieved the "PR" and "SD" in BOR. ", that is, a good curative effect, and the patient population as a whole meets the percentage of "PR"/"PD" ≥2. This type of autoantibody is preliminarily determined as a positive predictive antibody, which has a positive predictive effect of good immunotherapy curative effect.
一部分肿瘤相关抗原属于“负相关”抗原,本发明中将针对这种抗原的自身抗体命名为“N_Ab.”,显示这种自身抗体阳性信号的患者均出现了BOR中“PD”和“SD”,并且患者人群整体上满足“PD”百分比/“PR”百分比≥2。将这类自身抗体确定为负向预测抗体,具有免疫治疗疗效差的负向预测效果。初步确定,该正向预测抗体和负向预测抗体均可用于预测免疫治疗效果,分别具有免疫治疗疗效良好和差的预测效果。Some tumor-associated antigens are "negatively related" antigens. In the present invention, autoantibodies against this antigen are named "N_Ab.". Patients showing positive signals of such autoantibodies all have "PD" and "SD" in BOR. , And the patient population as a whole meets the "PD" percentage/"PR" percentage ≥2. Determining such autoantibodies as negative predictive antibodies has the negative predictive effect of poor immunotherapy efficacy. It is preliminarily determined that both the positive predictive antibody and the negative predictive antibody can be used to predict the effect of immunotherapy, and have good and poor predictive effects of immunotherapy respectively.
另外一部分肿瘤相关抗原的自身抗体属于“相关性不显著”抗原,显示不能用来预测疗效。Another part of the autoantibodies of tumor-associated antigens are "insignificantly correlated" antigens, which show that they cannot be used to predict therapeutic effects.
表3.患者基线特征Table 3. Baseline characteristics of patients
Figure PCTCN2021076771-appb-000005
Figure PCTCN2021076771-appb-000005
Figure PCTCN2021076771-appb-000006
Figure PCTCN2021076771-appb-000006
表4.所采用的抗原蛋白以及检测到的自身抗体阳性与肿瘤对免疫疗法的响应的相关性Table 4. Correlation between antigenic proteins used and detected autoantibody positivity and tumor response to immunotherapy
Figure PCTCN2021076771-appb-000007
Figure PCTCN2021076771-appb-000007
Figure PCTCN2021076771-appb-000008
Figure PCTCN2021076771-appb-000008
在使用一线免疫治疗患者为训练集,发现了潜在可以用来指导免疫治疗的标志物之后,接着使用联合后线免疫治疗(后线治疗是指之前的治疗失败后采用免疫治疗的患者,后线治疗的方式包括免疫单药治疗患者和免疫联合化疗治疗的患者(表3))作为“验证集”,来验证已发现的自身抗体是否仍有疗效预测的特性。After using the first-line immunotherapy patients as the training set and discovering potential markers that can be used to guide immunotherapy, the combined subsequent-line immunotherapy is then used. The treatment methods include immune monotherapy patients and immune-combined chemotherapy patients (Table 3)) as a "validation set" to verify whether the discovered autoantibodies still have the characteristics of efficacy prediction.
由于单个自身抗体的阳性率低,为了增加自身抗体检出的阳性率,分析结果时联合多个自身抗体的结果来判断预测效果。规则是:在患者中检测多个自身抗体,只要有其中一个或者多个自身抗体显示阳性,则判断抗体组合结果为阳性;而如果所有的自身抗体均为阴性,则判断抗体组合结果为阴性。此次检测选择抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD抗体作为一个自身抗体组合。Because the positive rate of a single autoantibody is low, in order to increase the positive rate of autoantibody detection, the results of multiple autoantibodies are combined to judge the predictive effect when analyzing the results. The rule is: if multiple autoantibodies are detected in a patient, as long as one or more of the autoantibodies are positive, the antibody combination is judged to be positive; and if all autoantibodies are negative, the antibody combination is judged to be negative. For this test, anti-Trim21 autoantibodies, anti-BRACA2 autoantibodies, anti-Annexin 1 autoantibodies and anti-HUD antibodies were selected as a combination of autoantibodies.
在训练集和验证集中,用PD-1抑制剂治疗(所用的免疫检查点抑制剂包含进口的纳武单抗、派姆单抗,也包括国产的免疫检测点抑制剂,即为其中的任何一种)后患者显示出肿瘤对免疫治疗响应的百分比见图1。其中,图1-A(P_Ab.组合)中的“P_Ab.阳性”是指抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD抗体中有任一个阳性的患者(即抗体组合阳性),“P_Ab.阴性”是指抗Trim21自身抗体、抗BRACA2自身抗体、抗Annexin 1自身抗体和抗HUD抗体均为阴性的患者(即抗体组合阴性);图1-B(N_Ab.组合)中的“N_Ab.阳性”是指抗HSP105自身抗体和抗AKAP4抗体中有任一个阳性的患者(即抗体组合阳性),“N_Ab.阴性”是指抗HSP105自身抗体和抗AKAP4抗体均为阴性的患者(即抗体组合阴性)。In the training set and validation set, treat with PD-1 inhibitors (the immune checkpoint inhibitors used include imported nivolumab and pembrolizumab, as well as domestic immune checkpoint inhibitors, which are any of them A) The percentage of patients showing tumor response to immunotherapy is shown in Figure 1. Among them, "P_Ab. positive" in Figure 1-A (P_Ab. combination) refers to patients who are positive for any one of anti-Trim21 autoantibodies, anti-BRACA2 autoantibodies, anti-Annexin 1 autoantibodies, and anti-HUD antibodies (ie, antibody combination Positive), "P_Ab. Negative" refers to patients whose anti-Trim21 autoantibody, anti-BRACA2 autoantibody, anti-Annexin 1 autoantibody and anti-HUD antibody are all negative (ie, the antibody combination is negative); Figure 1-B (N_Ab. combination) "N_Ab. positive" refers to patients whose anti-HSP105 autoantibody and anti-AKAP4 antibody are positive (that is, the combination of antibodies is positive), "N_Ab. negative" refers to both anti-HSP105 autoantibody and anti-AKAP4 antibody negative The patient (ie, the antibody combination is negative).
图1中的图1-A显示,在训练集(一线)中,正相关自身抗体组合阳性的患者在治疗后47.6%的患者治疗效果为“PR”,42%的患者治疗效果为“SD”,9.5%的患者治疗效果为“PD”;而正相关自身抗体组合阴性的患者则出现了28.6%的患者治疗效果为“PR”,50%的患者治疗效果为“SD”,21.4%的患者治疗效果为“PD”。在验证集(后线)中,正相关自身抗体组合阳性的患者在治疗后50%的患者治疗效果为“PR”,37.5%的患者治疗效果为“SD”,12.5%的患者治疗效果为“PD”;而正相关自身抗体组合阴性的患者在治疗后则有50%的患者治疗效果为“PD”和50%的患者治疗效果为“SD”。Figure 1-A in Figure 1 shows that in the training set (first-line), 47.6% of patients with positive correlation autoantibodies have a treatment effect of "PR" after treatment, and 42% of patients have a treatment effect of "SD". , 9.5% of the patients had a treatment effect of "PD"; while the positively correlated autoantibody combination was negative, 28.6% of the patients had a treatment effect of "PR", 50% of the patients had a treatment effect of "SD", and 21.4% of the patients The therapeutic effect is "PD". In the validation set (post-line), 50% of the patients with positive correlation autoantibody combination had a treatment effect of "PR", 37.5% of patients had a treatment effect of "SD", and 12.5% of patients had a treatment effect of "PR". PD"; and for patients with a negative combination of positively correlated autoantibodies, after treatment, 50% of patients have a treatment effect of "PD" and 50% of patients have a treatment effect of "SD".
图1中的图1-B显示,在训练集(一线)中,负相关自身抗体组合阳性的患者在治疗后50%的患者治疗效果为“PD”,50%的患者治疗效果为“SD”,0%的患者治疗效果为“PR”。在验证集(后线)中,负相关自身抗体组合阳性的患者在治疗后42.9%的患者治疗效果为“PD”,42.9%的患者治疗效果为“SD”,14.3%的患者治疗效果为“PR”。Figure 1-B in Figure 1 shows that in the training set (first-line), 50% of patients with positive negative autoantibody combinations have a treatment effect of "PD" after treatment, and 50% of patients have a treatment effect of "SD" , 0% of patients have a treatment effect of "PR". In the validation set (post-line), after treatment, 42.9% of patients with positive negative autoantibodies have a treatment effect of "PD", 42.9% of patients have a treatment effect of "SD", and 14.3% of patients have a treatment effect of " PR".
因此,证明了正相关自身抗体组合的联合检测能有效预测好的免疫治疗效果,而负相关自身抗体组合的联合检测能有效预测差的免疫治疗效果。Therefore, it is proved that the combined detection of positively related autoantibody combinations can effectively predict good immunotherapy effects, while the combined detection of negatively related autoantibody combinations can effectively predict poor immunotherapy effects.
使用Kaplan-Meier方法分析训练集和验证集的患者的无进展生存,并绘制生存曲线。结果发现,正相关自身抗体组合阳性人群和阴性人群在无进展生存曲线上显示有很大的差别,其中在训练集(一线)中,抗体组合阳性的患者的中位无进展生存时间为大于10个月,而抗体组合阴性的患者为5.52个月,p-值为0.0512,见图2中的图2-A;而在验证集(后线)中,同一组正相关自身抗体组合阳性的患者的中位无进展时间为7.56个月,而抗体组合阴性的患者为2.43个月,p-值小于0.005,见图2中的图2-B。The Kaplan-Meier method was used to analyze the progression-free survival of patients in the training set and the validation set, and to draw a survival curve. The results found that the positive and negative groups of autoantibody combinations showed great differences in the progression-free survival curve. In the training set (first-line), the median progression-free survival time of patients with positive antibody combinations was greater than 10 Month, and the antibody combination negative patient is 5.52 months, the p-value is 0.0512, see Figure 2-A in Figure 2. In the validation set (back line), the same group of positively correlated autoantibody combination positive patients The median progression-free time was 7.56 months, while the antibody combination negative patient was 2.43 months, and the p-value was less than 0.005, as shown in Figure 2-B in Figure 2.
实施例3Example 3
由于同一个肺癌患者可能同时检测到几种均为阳性结果的抗体,需要确定是否联合采用数目更少的自身抗体,也能同样地预测免疫治疗的疗效,甚至达到更好的效果。Since the same lung cancer patient may detect several antibodies with positive results at the same time, it is necessary to determine whether to use a smaller number of autoantibodies in combination, which can also predict the efficacy of immunotherapy and even achieve better results.
根据单个自身抗体阳性检测结果和BOR的相关性,预测7种肿瘤相关抗原的相应自身抗体和良好的免疫治疗效果最可能相关,从而可能有最佳的肿瘤免疫治疗响应预测效果。因此,分析了七种自身抗体的不同组合,包括其在肺癌患者中的抗体组合阳性比例(敏感性),以及对应抗体组合检测结果阳性和阴性的两个不同组患者在无进展生存曲线上的差别,列出中位无进展时间和p-值,来寻找最佳的自身抗体组合。自身抗体的不同组合以及分析结果见表5。Based on the correlation between the positive test results of a single autoantibody and BOR, it is predicted that the corresponding autoantibodies of the seven tumor-associated antigens are most likely to be correlated with good immunotherapy effects, which may have the best tumor immunotherapy response prediction effect. Therefore, seven different combinations of autoantibodies were analyzed, including the positive ratio (sensitivity) of the antibody combination in lung cancer patients, and the two different groups of patients with positive and negative antibody combination test results on the progression-free survival curve. Differences, list the median progression-free time and p-value to find the best combination of autoantibodies. The different combinations of autoantibodies and the analysis results are shown in Table 5.
表5.自身抗体组合及其阳性结果以及患者的中位无进展时间Table 5. Autoantibody combinations and their positive results and the median progression-free time of patients
Figure PCTCN2021076771-appb-000009
Figure PCTCN2021076771-appb-000009
Figure PCTCN2021076771-appb-000010
Figure PCTCN2021076771-appb-000010
相应地,患者用PD-1抑制剂治疗后的中位无进展时间(Kaplan-Meier法)分别见图3中图3-A至图3-RNP。Correspondingly, the median progression-free time (Kaplan-Meier method) of patients after treatment with PD-1 inhibitors is shown in Figure 3 in Figure 3-A to Figure 3-RNP, respectively.
实施例4Example 4
根据单个自身抗体阳性检测结果和BOR的相关性,预测4种肿瘤相关抗原的相应自身抗体和差的免疫治疗效果最可能相关,从而可能有最佳的肿瘤免疫治疗响应预测效果。因此,分析了四种自身抗体的不同组合,包括其在肺癌患者中的抗体阳性比例(敏感性),以及对应抗体组合检测结果阳性和阴性的两个不同组患者在无进展生存曲线上的差别,列出中位无进展时间和p-值,来寻找最佳的自身抗体组合。自身抗体的不同组合以及分析结果见表6。Based on the correlation between the positive test results of a single autoantibody and BOR, it is predicted that the corresponding autoantibodies of the four tumor-associated antigens are most likely to be related to poor immunotherapy effects, which may have the best tumor immunotherapy response prediction effect. Therefore, four different combinations of autoantibodies were analyzed, including their positive antibody ratio (sensitivity) in lung cancer patients, and the difference in the progression-free survival curve of two different groups of patients with positive and negative antibody combination test results. , List the median progression-free time and p-value to find the best autoantibody combination. The different combinations of autoantibodies and the analysis results are shown in Table 6.
表6.自身抗体组合及其阳性结果以及患者的中位无进展时间Table 6. Autoantibody combinations and their positive results and the median progression-free time of patients
Figure PCTCN2021076771-appb-000011
Figure PCTCN2021076771-appb-000011
Figure PCTCN2021076771-appb-000012
Figure PCTCN2021076771-appb-000012
相应地,患者用PD-1抑制剂治疗后的中位无进展时间(Kaplan-Meier法)分别见图4中图4-K至图4-P。Correspondingly, the median progression-free time (Kaplan-Meier method) of patients after treatment with PD-1 inhibitors is shown in Figure 4-K to Figure 4-P in Figure 4, respectively.
结果显示,抗HSP105和AKAP4的自身抗体组合能最好地预测差的免疫治疗效果,有分析统计意义。The results show that the combination of anti-HSP105 and AKAP4 autoantibodies can best predict poor immunotherapy effects, which is statistically significant.
实施例5Example 5
用PD-L1表达水平以及采用IMP2、XAGE-1和NY-ESO-1自身抗体作为预测方式,并观察患者对免疫疗法的响应,结果见表7。The PD-L1 expression level and IMP2, XAGE-1 and NY-ESO-1 autoantibodies were used as prediction methods, and the patient's response to immunotherapy was observed. The results are shown in Table 7.
在临床应用中,PD-L1作为用于预测免疫治疗效果的常用标志物。由于荧光检测PD-L1的组织表达量时需要使用肺癌患者合格的肿瘤组织样本,样本的来源不容易得到,并且出于其他原因,在本发明研究的免疫治疗的患者中,50%左右的患者没有PD-L1标志物的信息。In clinical applications, PD-L1 is used as a common marker for predicting the effect of immunotherapy. Because fluorescence detection of PD-L1 tissue expression requires the use of qualified tumor tissue samples from lung cancer patients, the source of the samples is not easy to obtain, and for other reasons, in the immunotherapy patients studied in the present invention, about 50% of patients There is no information on PD-L1 markers.
如表7中显示,有PD-L1表达的患者中,高的PD-L1表达量(>1%)的患者在治疗后出现了45.5%的PR,14.3%的SD,和15%的PD。低的PD-L1表达量(<1%)的患者中则在治疗后出现了4.5%的PR,28.6%的SD,和30%的PD。如果把一线治疗和后线治疗区分开,数据结果类似。As shown in Table 7, among patients with PD-L1 expression, patients with high PD-L1 expression (>1%) developed 45.5% PR, 14.3% SD, and 15% PD after treatment. Patients with low PD-L1 expression (<1%) had 4.5% PR, 28.6% SD, and 30% PD after treatment. If the first-line treatment and the back-line treatment are distinguished, the data results are similar.
作为对比,抗体组合R阳性患者在治疗后出现了68.2%的PR,22.9%的SD和10%的PD;抗体组合RPN阳性患者在治疗后出现了72.7%的PR,28.6%的SD和15%的PD。这些自身抗体组合和PD-L1一样能较好地预测免疫治疗疗效。For comparison, the antibody combination R-positive patients had 68.2% PR, 22.9% SD and 10% PD after treatment; the antibody combination RPN-positive patients had 72.7% PR, 28.6% SD and 15% after treatment. PD. These autoantibody combinations can predict the efficacy of immunotherapy as well as PD-L1.
作为对比,还分析了抗XAGE-1自身抗体和抗NY-ESO-1自身抗体两者联合和抗IMP2抗体单独对免疫治疗效果的预测效果。如表7中显示,抗XAGE-1自身抗体和抗NY-ESO-1自身抗体组合阳性的患者预测了31.8%的PR,17.1%的SD,和20%的PD;IMP2抗体阳性的患者在治疗后出现了18.2%的PR,5.7%的SD,和10%的PD。同时,本研究中抗XAGE-1和抗NY-ESO-1自身抗体组合,和IMP2抗体分别进行Kaplan-Meier生存曲线分析。如图5中的图5-A显示,IMP2抗体阳性和阴性两组患者的中位无进展生存时间分别为10.02月和 5.52月,但P值为0.7867,没有统计意义。而如图5中的图5-B显示,抗XAGE-1和抗NY-ESO-1自身抗体组合结果阳性和阴性两组患者的生存曲线基本重叠,结果为阳性和阴性两组患者的中位无进展生存时间也不存在差别。因此,无论抗XAGE-1抗体和抗NY-ESO-1抗体的组合还是抗IMP2抗体,对患者采用免疫疗法的治疗效果的预测效果均并不特别理想。As a comparison, the combination of anti-XAGE-1 autoantibody and anti-NY-ESO-1 autoantibody and anti-IMP2 antibody alone are also analyzed for the predictive effect of immunotherapy. As shown in Table 7, patients with a combination of anti-XAGE-1 autoantibodies and anti-NY-ESO-1 autoantibodies were predicted to have 31.8% of PR, 17.1% of SD, and 20% of PD; patients with positive IMP2 antibodies were being treated After that, 18.2% of PR, 5.7% of SD, and 10% of PD appeared. At the same time, the combination of anti-XAGE-1 and anti-NY-ESO-1 autoantibodies, and IMP2 antibody were used for Kaplan-Meier survival curve analysis in this study. As shown in Figure 5-A in Figure 5, the median progression-free survival time of the IMP2 antibody positive and negative groups was 10.02 months and 5.52 months, respectively, but the P value was 0.7867, which was not statistically significant. As shown in Figure 5-B in Figure 5, the survival curves of the positive and negative groups of anti-XAGE-1 and anti-NY-ESO-1 autoantibody combinations basically overlap, and the results are the median of the positive and negative groups of patients. There is no difference in progression-free survival time. Therefore, regardless of the combination of anti-XAGE-1 antibody and anti-NY-ESO-1 antibody or anti-IMP2 antibody, the predictive effect of the therapeutic effect of patients with immunotherapy is not particularly ideal.
表7.肿瘤自身抗体与其他预测方式以及对免疫治疗的响应预测对比(人数(%))Table 7. Comparison of tumor autoantibodies with other prediction methods and response predictions to immunotherapy (number of people (%))
Figure PCTCN2021076771-appb-000013
Figure PCTCN2021076771-appb-000013
实施例6Example 6
以检测自身抗体组合RNP为例,综合Trim21、BRCA2、Annexin1、HUD、P53和NY-ESO-1自身抗体,其中任何一个抗体检测结果为阳性,判断结果为阳性,所有的抗体检测结果都为阴性,判断结果为阴性。分析结果见图6。Take the detection of autoantibody combination RNP as an example. Trim21, BRCA2, Annexin1, HUD, P53 and NY-ESO-1 autoantibodies are combined. If any of the antibody test results is positive, the judgment result is positive, and all antibody test results are negative. , The judgment result is negative. The analysis result is shown in Figure 6.
如图6中的图6-A(一线免疫治疗)和图6-b(后线免疫治疗)显示, 一线免疫治疗的肺癌患者中,抗体组合阳性的风险比(HR:Hazard Ratio)(Mantel-Haenszel)预测值为0.2541(0.0684-0.8786),抗体组合阳性和阴性两组患者的中位无进展生存时间分别为(>10月)和5.52月(P值为0.0309),在后线免疫治疗的肺癌患者中,抗体组合阳性的HR预测值为0.2948(0.1409-0.6167)),抗体组合阳性和阴性两组患者的中位无进展生存时间分别为(8.18月)和2.43月(P值为0.0012)。如图6中的图6-C(免疫单药治疗)和图6-D(免疫治疗结合化疗)显示,在所有进行免疫单药治疗的患者中,抗体组合阳性的HR预测值为0.1876(0.0677-0.520),抗体组合阳性和阴性两组患者的中位无进展生存时间分别为(>10月)和2.94月(P值为0.0013),而对于免疫治疗结合化疗的患者中,抗体组合阳性的HR为0.3863(0.1714-0.8705),抗体组合阳性和阴性两组患者的中位无进展生存时间分别为(9.36月)和4.27月(P值为0.0218)。As shown in Figure 6-A (first-line immunotherapy) and Figure 6-b (later-line immunotherapy), among lung cancer patients treated with first-line immunotherapy, the risk ratio of positive antibody combination (HR: Hazard Ratio) (Mantel- Haenszel) predictive value is 0.2541 (0.0684-0.8786). The median progression-free survival time of the two groups of patients with antibody combination positive and negative is (>10 months) and 5.52 months (P value is 0.0309). Among lung cancer patients, the predictive value of HR for positive antibody combination was 0.2948 (0.1409-0.6167), and the median progression-free survival time of the two groups of positive and negative antibody combination were (8.18 months) and 2.43 months (P value 0.0012) . As shown in Figure 6-C (immune monotherapy) and Figure 6-D (immunotherapy combined with chemotherapy), in all patients undergoing immune monotherapy, the predictive value of HR for a positive antibody combination was 0.1876 (0.0677 -0.520), the median progression-free survival time of the two groups of patients with antibody combination positive and negative were (>10 months) and 2.94 months (P value 0.0013), and for patients with immunotherapy combined with chemotherapy, the antibody combination was positive The HR was 0.3863 (0.1714-0.8705), and the median progression-free survival time of the antibody combination positive and negative groups were (9.36 months) and 4.27 months (P value 0.0218).
以上对本发明具体实施方式的描述并不限制本发明,本领域技术人员可以根据本发明作出各种改变或变形,只要不脱离本发明的精神,均应属于本发明所附权利要求的范围。The above description of the specific embodiments of the present invention does not limit the present invention. Those skilled in the art can make various changes or modifications according to the present invention, as long as they do not deviate from the spirit of the present invention, they shall fall within the scope of the appended claims of the present invention.

Claims (17)

  1. 一种用于预测或判断受试者的肿瘤免疫治疗效果的生物标志物,所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。A biomarker for predicting or judging the effect of tumor immunotherapy in a subject, the biomarker being a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens : Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  2. 根据权利要求1所述的生物标志物,其特征在于,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2;The biomarker of claim 1, wherein the combination of autoantibodies comprises at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1 , P53, IMP2;
    优选地,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的两种、三种或四种:Trim21、BRCA2、Annexin 1、HUD;Preferably, the autoantibody combination includes two, three or four autoantibodies selected from the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD;
    更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:Trim21和BRCA2;More preferably, the autoantibody combination includes autoantibodies against the following tumor-associated antigens: Trim21 and BRCA2;
    进一步优选地,所述自身抗体组合还包括抗以下肿瘤相关抗原的自身抗体中的一种或两种:Annexin 1、HUD;Further preferably, the autoantibody combination also includes one or two of the following autoantibodies against the following tumor-associated antigens: Annexin 1, HUD;
    再更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:Even more preferably, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
    (A)Trim21,BRCA2,IMP2;(A) Trim21, BRCA2, IMP2;
    (B)Trim21,BRCA2,NY-ESO-1;(B) Trim21, BRCA2, NY-ESO-1;
    (C)Trim21,BRCA2,NY-ESO-1,IMP2;(C) Trim21, BRCA2, NY-ESO-1, IMP2;
    (D)Trim21,BRCA2,P53;(D) Trim21, BRCA2, P53;
    (E)Trim21,BRCA2,Annexin 1;(E) Trim21, BRCA2, Annexin 1;
    (F)Trim21,BRCA2,Annexin 1,P53;(F) Trim21, BRCA2, Annexin 1, P53;
    (G)Trim21,BRCA2,Annexin 1,NY-ESO-1,IMP2;(G) Trim21, BRCA2, Annexin 1, NY-ESO-1, IMP2;
    (H)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,IMP2;(H) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, IMP2;
    (I)Trim21,BRCA2,Annexin 1,NY-ESO-1,P53,IMP2;(I) Trim21, BRCA2, Annexin 1, NY-ESO-1, P53, IMP2;
    (R)Trim21,BRCA2,Annexin 1,HUD;(R) Trim21, BRCA2, Annexin 1, HUD;
    (RN)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1;(RN) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1;
    (RP)Trim21,BRCA2,Annexin 1,HUD,P53;或(RP) Trim21, BRCA2, Annexin 1, HUD, P53; or
    (RNP)Trim21,BRCA2,Annexin 1,HUD,NY-ESO-1,P53。(RNP) Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53.
  3. 根据权利要求1或2所述的生物标志物,其特征在于,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME;The biomarker according to claim 1 or 2, wherein the combination of autoantibodies comprises at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, PRAME;
    优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:HSP105,或HSP105和AKAP4;Preferably, the autoantibody combination includes autoantibodies against the following tumor-associated antigens: HSP105, or HSP105 and AKAP4;
    更优选地,所述自身抗体组合包括抗以下肿瘤相关抗原的自身抗体:More preferably, the autoantibody combination includes autoantibodies against the following tumor-associated antigens:
    (K)HSP105;(K)HSP105;
    (L)HSP105,AKAP4;(L)HSP105, AKAP4;
    (M)HSP105,MAGE-A3,AKAP4;或(M) HSP105, MAGE-A3, AKAP4; or
    (P)HSP105,AKAP4,PRAME。(P) HSP105, AKAP4, PRAME.
  4. 根据权利要求1至3中任一项所述的生物标志物,其特征在于,所述自身抗体为受试者接受肿瘤免疫治疗之前的血清、血浆、组织间隙液、脑脊液或尿液中的自身抗体;The biomarker according to any one of claims 1 to 3, wherein the autoantibody is the autoantibody in serum, plasma, interstitial fluid, cerebrospinal fluid, or urine before the subject receives tumor immunotherapy. Antibody;
    优选地,所述自身抗体为IgA、IgM或IgG。Preferably, the autoantibody is IgA, IgM or IgG.
  5. 根据权利要求1至4中任一项所述的生物标志物,其特征在于,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人;The biomarker according to any one of claims 1 to 4, wherein the subject is a mammal, preferably a primate mammal, more preferably a human;
    优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌;Preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodgkin’s Lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer;
    优选地,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。Preferably, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy Or other tumor treatment methods, wherein the immune checkpoint inhibitor is against PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or CD160 The immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  6. 一种用于检测权利要求1至5中任一项所述的生物标志物的试剂。A reagent for detecting the biomarker of any one of claims 1 to 5.
  7. 根据权利要求6所述的试剂,其特征在于,所述试剂是用于酶联免疫吸附法(ELISA)、蛋白/肽段芯片检测、免疫印迹、微珠免疫检测、微流控免疫检测的试剂;优选地,所述试剂用于通过抗原抗体反应对所述生物标志物进行检测,例如通过ELISA;The reagent according to claim 6, wherein the reagent is a reagent for enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, immunoblotting, bead immunoassay, microfluidic immunoassay ; Preferably, the reagent is used to detect the biomarker through an antigen-antibody reaction, for example, by ELISA;
    更优选地,所述试剂为用于检测所述自身抗体组合的抗原蛋白组合,所述抗原蛋白组合包括选自以下抗原蛋白的至少一种:Trim21、BRCA2、Annexin1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。More preferably, the reagent is an antigen protein combination for detecting the autoantibody combination, and the antigen protein combination includes at least one selected from the following antigen proteins: Trim21, BRCA2, Annexin1, HUD, NY-ESO-1 , P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  8. 权利要求1至5中任一项所述的生物标志物或权利要求6或7所述 的试剂在制备用于预测或判断受试者的肿瘤免疫治疗效果的产品中的用途。Use of the biomarker according to any one of claims 1 to 5 or the reagent according to claim 6 or 7 in the preparation of a product for predicting or judging the effect of tumor immunotherapy in a subject.
  9. 根据权利要求8所述的用途,其特征在于,所述受试者为哺乳动物,优选为灵长类哺乳动物,更优选为人;The use according to claim 8, wherein the subject is a mammal, preferably a primate mammal, more preferably a human;
    优选地,所述肿瘤为肾癌、肝癌、卵巢癌、宫颈癌、头颈部鳞状细胞癌、鼻咽癌、尿路上皮癌、喉癌、胃癌、黑色素瘤、前列腺癌、霍奇金氏淋巴瘤、膀胱癌、结直肠癌、肺癌,特别是肺癌,例如小细胞肺癌、非小细胞肺癌、肺鳞癌、肺腺癌和其他亚型肺癌;Preferably, the tumor is kidney cancer, liver cancer, ovarian cancer, cervical cancer, head and neck squamous cell carcinoma, nasopharyngeal cancer, urothelial cancer, laryngeal cancer, gastric cancer, melanoma, prostate cancer, Hodgkin’s Lymphoma, bladder cancer, colorectal cancer, lung cancer, especially lung cancer, such as small cell lung cancer, non-small cell lung cancer, lung squamous cell carcinoma, lung adenocarcinoma and other subtypes of lung cancer;
    优选地,所述免疫治疗包括采用免疫检查点抑制剂治疗;优选地,所述免疫治疗为单独施用免疫检查点抑制剂治疗或免疫检查点抑制剂与化疗、放疗、抗血管治疗、靶向治疗或其他肿瘤治疗手段的联合治疗,其中所述免疫检查点抑制剂为针对PD-1、PD-L1、CTLA-4、BTLA、TIM-3、LAG-3、TIGIT、LAIR1、2B4和/或CD160的免疫检查点抑制剂,优选为抗PD-1抗体或抗PD-L1抗体。Preferably, the immunotherapy includes treatment with immune checkpoint inhibitors; preferably, the immunotherapy is the treatment of immune checkpoint inhibitors alone or immune checkpoint inhibitors combined with chemotherapy, radiotherapy, anti-vascular therapy, and targeted therapy Or other tumor treatment methods, wherein the immune checkpoint inhibitor is against PD-1, PD-L1, CTLA-4, BTLA, TIM-3, LAG-3, TIGIT, LAIR1, 2B4 and/or CD160 The immune checkpoint inhibitor is preferably an anti-PD-1 antibody or an anti-PD-L1 antibody.
  10. 一种试剂盒,其包含权利要求6或7所述的试剂。A kit comprising the reagent according to claim 6 or 7.
  11. 根据权利要求10所述的试剂盒,其特征在于,所述试剂盒是用于酶联免疫吸附法(ELISA)、蛋白/肽段芯片检测、免疫印迹、微珠免疫检测、微流控免疫检测的试剂盒;优选地,所述试剂盒用于通过抗原抗体反应对所述生物标志物进行检测;The kit according to claim 10, wherein the kit is used for enzyme-linked immunosorbent assay (ELISA), protein/peptide chip detection, western blotting, bead immunoassay, microfluidic immunoassay The kit; preferably, the kit is used to detect the biomarker through an antigen-antibody reaction;
    更优选地,所述试剂盒为ELISA检测试剂盒。More preferably, the kit is an ELISA detection kit.
  12. 一种用于预测或判断受试者的肿瘤免疫治疗效果的方法,或者,一种用于预测或判断受试者的肿瘤对免疫治疗的敏感性的方法,所述方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:A method for predicting or judging the effect of tumor immunotherapy in a subject, or, a method for predicting or judging the sensitivity of a subject’s tumor to immunotherapy, the method comprising detecting Are the following biomarkers positive in the examinee’s sample:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  13. 根据权利要求12所述的方法,其特征在于,其特征在于,所述方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:The method according to claim 12, wherein the method comprises detecting whether the following biomarkers in a sample from the subject are positive:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2;并且,所述生物标志物是阳性时预测或判断:受试者具有良好的肿瘤免疫治疗效果;受试者受益于肿瘤免疫治疗;该治疗有效;或,受试者的肿瘤对免疫治疗敏感。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2; Moreover, when the biomarker is positive, it is predicted or judged that the subject has a good tumor immunotherapy effect; the subject benefits from tumor immunotherapy; the treatment is effective; or the subject's tumor is sensitive to immunotherapy.
  14. 根据权利要求12所述的方法,其特征在于,其特征在于,所述方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:The method according to claim 12, wherein the method comprises detecting whether the following biomarkers in a sample from the subject are positive:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME;并且,所述生物标志物是阳性时预测或判断:受试者具有差的肿瘤免疫治疗效果;受试者不受益于肿瘤免疫治疗;该治疗无效;或,受试者的肿瘤对免疫治疗不敏感。The biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, PRAME; and, the biomarker is positive Time prediction or judgment: the subject has a poor tumor immunotherapy effect; the subject does not benefit from the tumor immunotherapy; the treatment is ineffective; or the subject’s tumor is not sensitive to the immunotherapy.
  15. 一种治疗受试者的肿瘤的方法,所述方法包括检测来自受试者的样本中如下生物标志物是否是阳性的:A method of treating tumors in a subject, the method comprising detecting whether the following biomarkers in a sample from the subject are positive:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2、HSP105、MAGE-A3、AKAP4、PRAME。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2, HSP105, MAGE-A3, AKAP4, PRAME.
  16. 根据权利要求15所述的方法,其特征在于,所述方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:The method according to claim 15, wherein the method comprises detecting whether the following biomarkers in a sample from the subject are positive:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:Trim21、BRCA2、Annexin 1、HUD、NY-ESO-1、P53、IMP2;并且,所述生物标志物是阳性的时,使受试者进行肿瘤免疫治疗。The biomarker is a combination of autoantibodies, and the combination of autoantibodies includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: Trim21, BRCA2, Annexin 1, HUD, NY-ESO-1, P53, IMP2; And, when the biomarker is positive, the subject is subjected to tumor immunotherapy.
  17. 根据权利要求15所述的方法,其特征在于,所述方法包括检测来自所述受试者的样本中如下生物标志物是否是阳性的:The method according to claim 15, wherein the method comprises detecting whether the following biomarkers in a sample from the subject are positive:
    所述生物标志物为自身抗体组合,所述自身抗体组合包括选自抗以下肿瘤相关抗原的自身抗体的至少一种:HSP105、MAGE-A3、AKAP4、PRAME;并且,所述生物标志物是阳性的时,使受试者不进行肿瘤免疫治疗。The biomarker is an autoantibody combination, and the autoantibody combination includes at least one selected from the group consisting of autoantibodies against the following tumor-associated antigens: HSP105, MAGE-A3, AKAP4, PRAME; and, the biomarker is positive When the subject does not undergo tumor immunotherapy.
PCT/CN2021/076771 2020-02-21 2021-02-19 Biomarker relating to effect of tumor immunotherapy and application thereof WO2021164713A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010106659.0A CN111337678B (en) 2020-02-21 2020-02-21 Biomarker related to tumor immunotherapy effect and application thereof
CN202010106659.0 2020-02-21

Publications (1)

Publication Number Publication Date
WO2021164713A1 true WO2021164713A1 (en) 2021-08-26

Family

ID=71185543

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/076771 WO2021164713A1 (en) 2020-02-21 2021-02-19 Biomarker relating to effect of tumor immunotherapy and application thereof

Country Status (2)

Country Link
CN (1) CN111337678B (en)
WO (1) WO2021164713A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113721021A (en) * 2021-09-16 2021-11-30 郑州大学 Application of PRKCZ autoantibody in auxiliary diagnosis of esophageal squamous cell carcinoma
CN114242157A (en) * 2021-12-28 2022-03-25 江苏先声医学诊断有限公司 Predicting non-small cell lung cancer immunotherapy efficacy based on bGMS

Families Citing this family (16)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111337678B (en) * 2020-02-21 2023-06-06 杭州凯保罗生物科技有限公司 Biomarker related to tumor immunotherapy effect and application thereof
WO2022017435A1 (en) * 2020-07-22 2022-01-27 信达生物制药(苏州)有限公司 SERIES OF BIOMARKERS OF CONSENSUS SEQUENCES IN CDR3 SEQUENCE OF TCR-β CHAIN AND APPLICATION THEREOF
CN112162097B (en) * 2020-07-24 2022-04-01 广州医科大学 GDF1 as biomarker for evaluating treatment effect of PD-1 monoclonal antibody
CN112345755B (en) * 2020-09-23 2023-06-16 杭州凯保罗生物科技有限公司 Biomarker for breast cancer and application thereof
WO2022099543A1 (en) * 2020-11-12 2022-05-19 深圳先进技术研究院 Application of ny-eso-1 gene inhibitor as antitumor chemotherapy drug sensitizer
CN113106158A (en) * 2021-06-16 2021-07-13 至本医疗科技(上海)有限公司 CSMD family gene prediction of advanced lung adenocarcinoma immunotherapy efficacy
CN113671180B (en) * 2021-09-16 2023-07-07 郑州大学 Application of PAIP1 autoantibody in esophageal squamous carcinoma auxiliary diagnosis
CN114167059B (en) * 2021-11-03 2023-07-14 郑州大学 Biomarker and detection kit for diagnosis of esophageal squamous carcinoma
CN114672567A (en) * 2022-04-27 2022-06-28 中国医学科学院肿瘤医院 Lung squamous carcinoma patient prognosis evaluation system based on CD47 and TIGIT double targets and application thereof
CN116159131B (en) * 2022-11-29 2024-02-13 中国人民解放军海军军医大学 Application of TRIM21 and promoter thereof in preparation of antitumor biotherapeutic drugs
CN115896290B (en) * 2022-11-29 2023-11-24 中国人民解放军海军军医大学 Application of TRIM21 gene detection in tumor diagnosis, treatment selection and prognosis evaluation
CN115877006B (en) * 2022-12-20 2023-12-15 杭州凯保罗生物科技有限公司 Ovarian cancer-related biomarker and application thereof
CN116970614A (en) * 2022-12-29 2023-10-31 达冕疫苗(广州)有限公司 Compositions and methods for ribonucleic acid vaccines encoding NY-ESO-1
CN115825441B (en) * 2023-01-30 2023-05-26 上海秤信生物科技有限公司 Autoantibody marker for predicting immune neoadjuvant therapeutic effect of patients with lung cancer in third stage
CN116298289B (en) * 2023-01-30 2023-11-03 上海秤信生物科技有限公司 Biomarker for predicting lung cancer immune new adjuvant therapy effect and application thereof
CN116773811A (en) * 2023-04-12 2023-09-19 上海秤信生物科技有限公司 Application of BRCA2 truncated protein in lung cancer screening

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101632020A (en) * 2006-09-13 2010-01-20 昂西免疫有限公司 Improved method of immunity
WO2013169971A1 (en) * 2012-05-10 2013-11-14 Bristol-Myers Squibb Company Anti-tumor antibodies as predictive or prognostic biomarkers of efficacy and survival in ipilimumab-treated patients
CN103869086A (en) * 2014-04-14 2014-06-18 杭州凯保罗生物科技有限公司 Serum autoantibody detection kit
US20150044224A1 (en) * 2012-03-02 2015-02-12 H. Lee Moffitt Cancer Center And Research Institute, Inc. Materials and methods for differential treatment of cancer
CN104427992A (en) * 2012-01-25 2015-03-18 德那翠丝有限公司 Biomarkers and combination therapies using oncolytic virus and immunomodulation
WO2018156448A1 (en) * 2017-02-21 2018-08-30 The Board Of Regents Of The Uiversity Of Texas System Prediction and treatment of immunotherapeutic toxicity
WO2019115480A1 (en) * 2017-12-12 2019-06-20 Protagen Ag Melanoma checkpoint inhibitor detection and treatment
CN110687282A (en) * 2019-08-26 2020-01-14 中国医学科学院肿瘤医院 PD-1 and/or p53 autoantibodies as markers for tumor efficacy prediction or prognosis evaluation
CN111337678A (en) * 2020-02-21 2020-06-26 杭州凯保罗生物科技有限公司 Biomarker related to tumor immunotherapy effect and application thereof

Family Cites Families (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2008532014A (en) * 2005-02-24 2008-08-14 セマインズ,インコーポレイティド Compositions and methods for classifying biological samples
WO2010065944A1 (en) * 2008-12-05 2010-06-10 Serametrix Autoantibody detection systems and methods
WO2017058827A1 (en) * 2015-09-29 2017-04-06 Essenlix Corp. Method of detecting an analyte in a sample
CN111328287A (en) * 2017-07-04 2020-06-23 库瑞瓦格股份公司 Novel nucleic acid molecules
CN110464841B (en) * 2019-08-19 2020-05-22 启辰生生物科技(珠海)有限公司 Immunity-enhancing pharmaceutical composition and application thereof

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN101632020A (en) * 2006-09-13 2010-01-20 昂西免疫有限公司 Improved method of immunity
CN104427992A (en) * 2012-01-25 2015-03-18 德那翠丝有限公司 Biomarkers and combination therapies using oncolytic virus and immunomodulation
US20150044224A1 (en) * 2012-03-02 2015-02-12 H. Lee Moffitt Cancer Center And Research Institute, Inc. Materials and methods for differential treatment of cancer
WO2013169971A1 (en) * 2012-05-10 2013-11-14 Bristol-Myers Squibb Company Anti-tumor antibodies as predictive or prognostic biomarkers of efficacy and survival in ipilimumab-treated patients
CN103869086A (en) * 2014-04-14 2014-06-18 杭州凯保罗生物科技有限公司 Serum autoantibody detection kit
WO2018156448A1 (en) * 2017-02-21 2018-08-30 The Board Of Regents Of The Uiversity Of Texas System Prediction and treatment of immunotherapeutic toxicity
WO2019115480A1 (en) * 2017-12-12 2019-06-20 Protagen Ag Melanoma checkpoint inhibitor detection and treatment
CN110687282A (en) * 2019-08-26 2020-01-14 中国医学科学院肿瘤医院 PD-1 and/or p53 autoantibodies as markers for tumor efficacy prediction or prognosis evaluation
CN111337678A (en) * 2020-02-21 2020-06-26 杭州凯保罗生物科技有限公司 Biomarker related to tumor immunotherapy effect and application thereof

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
GAO HONGJUN; ZHENG ZHAOXU; MAO YOUSHENG; WANG WEI; QIAO YUANYUAN; ZHOU LANPING; LIU FANG; HE HONGZHI; ZHAO XIAOHANG: "Identification of tumor antigens that elicit a humoral immune response in the sera of Chinese esophageal squamous cell carcinoma patients by modified serological proteome analysis", CANCER LETTERS, NEW YORK, NY, US, vol. 344, no. 1, 22 October 2013 (2013-10-22), US, pages 54 - 61, XP028826459, ISSN: 0304-3835, DOI: 10.1016/j.canlet.2013.10.007 *
HARDY-WERBIN M., ARPÍ O., TAUS A., ROCHA P., JOSEPH-PIETRAS D., NOLAN L., DANSON S., GRIFFITHS R., LOPEZ-BOTET M., ROVIRA A., ALBA: "Assessment of neuronal autoantibodies in patients with small cell lung cancer treated with chemotherapy with or without ipilimumab", ONCOIMMUNOLOGY, vol. 7, no. 2, 23 August 2017 (2017-08-23), pages e1395125, XP055839020, DOI: 10.1080/2162402X.2017.1395125 *
LEONARDO MIRANDOLA, JOSE A. FIGUEROA, TAM T. PHAN, FABIO GRIZZI, MINJI KIM, RAKHSHANDA LAYEEQUR RAHMAN, MARJORIE R. JENKINS, EVERA: "Novel antigens in non-small cell lung cancer: SP17, AKAP4, and PTTG1 are potential immunotherapeutic targets", ONCOTARGET, vol. 6, no. 5, 20 February 2015 (2015-02-20), pages 2812 - 2826, XP055428240, DOI: 10.18632/oncotarget.2802 *
YAN YUANQING, SUN NAN, WANG HONG, KOBAYASHI MAKOTO, LADD JON J., LONG JAMES P., LO KEN C., PATEL JIGAR, SULLIVAN ERIC, ALBERT THOM: "Whole Genome–Derived Tiled Peptide Arrays Detect Prediagnostic Autoantibody Signatures in Non–Small-Cell Lung Cancer", CANCER RESEARCH, AMERICAN ASSOCIATION FOR CANCER RESEARCH, vol. 79, no. 7, 1 April 2019 (2019-04-01), pages 1549 - 1557, XP055839015, ISSN: 0008-5472, DOI: 10.1158/0008-5472.CAN-18-1536 *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN113721021A (en) * 2021-09-16 2021-11-30 郑州大学 Application of PRKCZ autoantibody in auxiliary diagnosis of esophageal squamous cell carcinoma
CN113721021B (en) * 2021-09-16 2023-07-07 郑州大学 Application of PRKCZ autoantibody in esophageal squamous carcinoma auxiliary diagnosis
CN114242157A (en) * 2021-12-28 2022-03-25 江苏先声医学诊断有限公司 Predicting non-small cell lung cancer immunotherapy efficacy based on bGMS
CN114242157B (en) * 2021-12-28 2023-03-21 江苏先声医学诊断有限公司 Predicting non-small cell lung cancer immunotherapy efficacy based on bGMS

Also Published As

Publication number Publication date
CN111337678A (en) 2020-06-26
CN111337678B (en) 2023-06-06

Similar Documents

Publication Publication Date Title
WO2021164713A1 (en) Biomarker relating to effect of tumor immunotherapy and application thereof
JP6554152B2 (en) Folate receptor alpha as a diagnostic and prognostic marker for folate receptor alpha expressing cancer
US20090047689A1 (en) Autoantigen biomarkers for early diagnosis of lung adenocarcinoma
US9354233B2 (en) A+ biomarker assays
RU2016109642A (en) ANTIBODIES AND ANALYSIS METHODS FOR DETECTING THE FOLIC ACID RECEPTOR 1
WO2022083673A1 (en) Biomarker for esophageal cancer, and use thereof
WO2022063156A1 (en) Biomarker in breast cancer and application thereof
JP6691060B2 (en) Cell surface prostate cancer antigen for diagnostics
JP2020515826A (en) Antibody assay
EP3047277B1 (en) Autoantibody biomarkers of ovarian cancer
AU2017260806A1 (en) Markers of endometrial cancer
AU2010252907A1 (en) Methods for the diagnosis or prognosis of colorectal cancer
CN108738347B (en) Method, device, computer program product and kit for assisting recurrence risk prediction of hepatocellular carcinoma patients
RU2745060C2 (en) Biomarkers for combined therapy including lenvatinib and everolimus
CN107110848B (en) Method for detecting arteriosclerosis and cancer using deoxyhypusine synthase gene as index
JP7431226B2 (en) Biomarkers for combination therapy including lenvatinib and everolimus
US20200018750A1 (en) Methods and compositions for the prediction and treatment of focal segmental glomerulosclerosis
JP2022512590A (en) Biomarkers for therapy containing sorafenib compounds
KR102128251B1 (en) Biomarker Composition for Diagnosing Colorectal Cancer Specifically Binding to Arginine-methylated Dopamine Receptor D2
KR102131860B1 (en) Biomarker Composition for Diagnosing Colorectal Cancer Specifically Binding to Arginine-methylated Gamma-glutamyl Transferase 1
WO2022154037A1 (en) Prognostic biomarker for cancer
WO2023002943A1 (en) Biomarker for predicting prognosis of cancer patient, method for predicting prognosis of cancer patient, method for predicting effect of cancer therapeutic drug on cancer patient, and kit for predicting prognosis of cancer patient
WO2023202392A1 (en) Influence of receptor integrative analysis on hr+/her2+ breast cancer molecular subtypes and prognosis
US20230375550A1 (en) Method for diagnosing breast cancer by using biomarker
WO2023190820A1 (en) ANTI-CK2α ANTIBODY OR FRAGMENT THEREOF

Legal Events

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

Ref document number: 21756810

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 21756810

Country of ref document: EP

Kind code of ref document: A1