CN113117055A - Polypeptide composition specifically binding with HLA-A24 typing and application - Google Patents

Polypeptide composition specifically binding with HLA-A24 typing and application Download PDF

Info

Publication number
CN113117055A
CN113117055A CN202010940084.2A CN202010940084A CN113117055A CN 113117055 A CN113117055 A CN 113117055A CN 202010940084 A CN202010940084 A CN 202010940084A CN 113117055 A CN113117055 A CN 113117055A
Authority
CN
China
Prior art keywords
amino acid
cancer
acid sequence
seq
cells
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202010940084.2A
Other languages
Chinese (zh)
Inventor
郝方元
张喜艳
刘辉
尹锋
金华君
钱其军
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Shanghai Cell Therapy Group Co Ltd
Original Assignee
Shanghai Cell Therapy Group Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Shanghai Cell Therapy Group Co Ltd filed Critical Shanghai Cell Therapy Group Co Ltd
Publication of CN113117055A publication Critical patent/CN113117055A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/0005Vertebrate antigens
    • A61K39/0011Cancer antigens
    • A61K39/001102Receptors, cell surface antigens or cell surface determinants
    • A61K39/001111Immunoglobulin superfamily
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0636T lymphocytes
    • C12N5/0638Cytotoxic T lymphocytes [CTL] or lymphokine activated killer cells [LAK]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues
    • C12N5/0602Vertebrate cells
    • C12N5/0634Cells from the blood or the immune system
    • C12N5/0639Dendritic cells, e.g. Langherhans cells in the epidermis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5154Antigen presenting cells [APCs], e.g. dendritic cells or macrophages
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/51Medicinal preparations containing antigens or antibodies comprising whole cells, viruses or DNA/RNA
    • A61K2039/515Animal cells
    • A61K2039/5156Animal cells expressing foreign proteins
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/22Colony stimulating factors (G-CSF, GM-CSF)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2302Interleukin-2 (IL-2)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/23Interleukins [IL]
    • C12N2501/2304Interleukin-4 (IL-4)
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/20Cytokines; Chemokines
    • C12N2501/24Interferons [IFN]
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2501/00Active agents used in cell culture processes, e.g. differentation
    • C12N2501/998Proteins not provided for elsewhere
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2502/00Coculture with; Conditioned medium produced by
    • C12N2502/11Coculture with; Conditioned medium produced by blood or immune system cells
    • C12N2502/1121Dendritic cells

Landscapes

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

Abstract

The invention belongs to the field of medical immunology, and particularly relates to a polypeptide composition specifically binding to HLA-A24 typing and an application thereof, wherein the polypeptide composition is selected from one or more polypeptides derived from antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3 and KIF20A, and the polypeptides derived from the antigenic proteins contain antigenic epitopes or variants of the antigenic epitopes of the antigenic proteins. The polypeptide composition achieves targeted toxicity effect on tumor cells by presenting and activating specific CD8+ cytotoxic T lymphocytes after loading antigen presenting cells. The polypeptide composition, the tumor vaccine, the DC vaccine and the pharmaceutical composition derived from the polypeptide composition can obviously activate immune effector cells, particularly T cells, obviously improve the secretion level of cytokines relevant to the activation of the T cells and the killing level of the T cells, and have potential clinical value.

Description

Polypeptide composition specifically binding with HLA-A24 typing and application
Technical Field
The invention belongs to the field of medical immunology, and particularly relates to a polypeptide composition specifically binding to HLA-A24 typing and application thereof.
Background
In recent years, advances have been made in the treatment of cancer by surgery in combination with chemoradiotherapy, which has improved patient survival, particularly in patients with invasive cancers of the breast, lung, prostate and kidney. However, most of these treatments have significant toxic side effects, which are harmful to normal cells.
Tumors can initiate both humoral and cellular immune responses in the body. The tumor antigen is processed into peptide segment in the cell, then combined with the I-type molecule of the major histocompatibility complex on the cell surface and presented to CD8+ cytotoxic lymphocyte, or firstly shed from the tumor cell, then taken up by the antigen presenting cell, processed into peptide segment, then combined with the II-type molecule of the major histocompatibility complex on the surface and presented to CD4+ helper lymphocyte, and further the anti-tumor cell immune response of the organism is induced. The increased knowledge of genetic changes during immunity to tumors and the progression of malignant tumors has enabled humans to develop more selective and safe treatments by activating the immune system to attack the developing tumor, i.e., tumor vaccines. Depending on the specific use of tumor vaccines, they can be divided into prophylactic and therapeutic vaccines. The main function of the preventive vaccine is to control the occurrence of tumors; the therapeutic vaccine is based on tumor-associated antigen and is mainly used for adjuvant therapy after chemotherapy. One of the tumor vaccines is a Dendritic Cell (DC) based vaccine. DC cells are distinct from B lymphocytes and macrophages in that they express a large amount of co-stimulatory molecules and have the ability to effectively sensitize both CD4+ helper T cells (Th) and CD8+ Cytotoxic T Cells (CTLs). DCs generate specific anti-tumor immune responses by loading with tumor antigens and inducing them into mature DCs. Based on this, various anti-tumor vaccines have been developed with DC, including DC loaded with tumor antigen peptides, DC loaded with tumor whole cell antigens, DC loaded with tumor cell RNA, DC loaded with tumor cell DNA, DC loaded with exosomes (exosomes), cytokines, and chemokine genes. DC vaccines have been tried in malignant melanoma, prostate cancer, renal cancer, and other cancer species, and have been partially successful. Various forms of DC vaccines have been tried in the beginning of immunotherapy of tumors and have shown good efficacy in preliminary clinical trials. Among them, DC vaccine Provenge produced by Dendreon corporation of usa was approved by the national food and drug administration in 2010 for advanced stage, especially for prostate cancer patients with failure of hormone therapy, and the therapeutic effect showed that it could prolong the survival time of patients by more than 4 months compared to placebo (Nature Medicine,2010,16(6): 615).
Currently, most clinical trials use autologous whole tumor lysate to load DCs by lysing the patient's own tumor tissue through multiple cycles of freeze-thawing and stimulating the DC cells with the lysate (Cancer Immunol Immunother,2006,55: 819; medical oncology,2006,23: 273.). Freeze-thaw cycles induce tumor cell necrosis, but freeze-thaw induced tumor cell necrosis is not immunogenic, and even inhibits Toll-like receptor (TLR) induced DC cell maturation and normal function (Hatfeld P, Merrick AE, West E, O' Donnell D, Selby P, Vile R, et al. optimization of dendritic cell loading with tumor cell lysis for cancer immunization. J Immunothers (2008)31(7): 620-632), and tumor tissues of patients are not always readily available. Tumor cell lysates, purified tumor-associated antigens and tumor-derived mrnas have also been demonstrated to be useful as antigen sources for loading DCs. Tumor cell lysates can provide multiple antigens for DC loading and can induce CD4+ and CD8+ T cell responses and confer different Damage-Associated Molecular Patterns (DAMPs) to the DC to ensure maturation of the DC, but also can provide immunomodulatory cytokines to the DC to induce tolerizing transformation of the DC cells (Guida M, Piscont S, Cold G.Metastatic melanoma: the new era of targeted therapy.Expert. Experin. target Targets 2012; 16Suppl 2: S61-70); purified tumor-associated antigen-loaded DCs are able to activate antigen-specific T cell responses and induce CD4+ and CD8+ T cell responses, but the number of different antigen species for a single use is limited. Tumor-derived mRNA can be transferred to tumor-associated antigens and co-stimulatory molecules, ensuring antigen presentation of MHC class I, and does not require cross-presentation (Robbins PF, Morgan RA, Feldman SA, Yang JC, Sherry RM, Dudley ME, Wunderlich JR, NaHvi AV, Helman LJ, Mackall CL, et al. tumor regression in tissues with metallic synthetic cell sarcoma and tumor using induced immune response with NY-ESO-1.J Clin Oncol; 29:917-24), but does not induce DC cell maturation, nor does it induce an effective CD4+ immune response, while the number of different antigen species used at a single time is limited.
When a short peptide fragment of a tumor-associated antigen is used as an antigen loaded with DC, the number of types of antigens that can be involved in a single use can be effectively increased, and the immune response level of CD4+ and CD8+ T cells can be improved, but it is necessary to determine the HLA haplotype of a subject and select an appropriate peptide fragment from the selected tumor-associated antigen to verify whether the peptide fragment can bind to the HLA haplotype. HLA alleles are highly polymorphic in different ethnic groups. According to the statistics of the world health organization, the number of HLA class I alleles is more than 13000 by 2018, wherein the number of HLA-A alleles is 4200, the number of HLA-B alleles is 5091 and the number of HLA-C alleles is 3854 (http:// www.hla.alleles.org/nomenclature/stats.html). Among them, the common HLA types in Asian population are HLA-A2, A3 and A24(Experimental and Therapeutic Medicine,2011,2: 109-. Three types of HLA-A2, A11 and A24 were able to cover more than 90% of the Chinese population (Immunol Today, 1996; 17: 261.). HLA-A2 belongs to HLA-A2 super type, the frequency is 45.9% at most in Chinese population, HLA-A11 belongs to HLA-A3 super type, the frequency is 37.5% at least in Caucasian (Caucasian), the frequency is 52.7% at most in Chinese; HLA-A24 belongs to HLA-A24 supertype, with a frequency of 23.9% In caucasians, 40.1% In Chinese, and 58.6% In Japanese (Curr Opinion In Immunol,1998,10: 478-482; Immunogenetics,1999,50(3-4): 201-212.). Presently, there is a lack of immunogenic compositions comprising polypeptides that can be efficiently presented by antigen presenting cells and tumor vaccines comprising such polypeptide compositions for each HLA type.
Disclosure of Invention
In tumor immunization, a polypeptide fragment of an epitope of a tumor-associated antigen is bound to HLA on the surface of an antigen presenting cell such as DC to form an HLA-tumor epitope peptide complex, which is recognized by TCR and then presented to T cells, so that the T cells capable of recognizing the corresponding tumor epitope are specifically activated and expanded. The expanded T cells become Cytotoxic T Lymphocytes (CTLs) that specifically target the tumor-associated antigen, producing a cell-mediated immune killing effect on tumor cells expressing the tumor-associated antigen.
The senescent cells can activate innate and adaptive immune responses and maintain tissue homeostasis. In addition, new findings suggest that programmatically induced cellular senescence may be important in regulating reproductive processes, in part due to immune clearance. The antigens p16, p53 and p21 have significant relation with the occurrence of senescent cells and tumors. The currently widely recognized biomarkers for senescent cells (e.g., β -galactosidase, p16INK4A), including p16, p53 and p21, have similar effects in the regulation of the cell cycle as p16, and regulatory mutations or deletions of p16 or p53 are found in a variety of tumor cells.
In order to improve the cell-mediated tumor immune killing effect, the invention screens the immunogenic polypeptide composition which is specifically combined with HLA type (A24) based on tumor-associated antigen and cell senescence-associated protein, and the method for obtaining cells loaded with the immunogenic polypeptide composition, activated immune effector cells and triggering in-vivo immune response reaction based on the immunogenic polypeptide composition. Tumor associated antigens Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF 20A. The technical scheme of the invention is as follows:
polypeptide compositions
The invention provides a polypeptide composition, which is selected from one or more polypeptides derived from antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3 and KIF20A, is part of each corresponding antigenic protein, and has the length equal to or shorter than that of the corresponding full-length antigenic protein; preferably, the polypeptide derived from each antigenic protein comprises (1) an epitope of each antigenic protein, the epitope being immunogenic; or contains an epitope having the same activity as the epitope in (1) obtained by amino acid substitution, addition or deletion, preferably having homology of 75% or more with the amino acid sequence of the epitope in (1), more preferably having homology of 85% or more with the amino acid sequence of the epitope in (1), still more preferably having homology of 95% or more with the amino acid sequence of the epitope in (1), i.e., a variant having equivalent function to the epitope in (1), having an altered amino acid sequence, such as the presence of one or more amino acid substitutions in the amino acid sequence at the epitope in (1), or one or more amino acids are added to the amino acid sequence at the antigen-presenting position in (1), or one or more amino acids are deleted from the amino acid sequence at the antigen surface position in (1), without affecting the function of the polypeptide comprising the epitope in (1) and the polypeptide composition comprising the polypeptide of the epitope in (1). As an embodiment of the present invention, 1 to 5 amino acids, preferably 1 to 3 amino acids are added to the N-terminal and/or C-terminal of the amino acid sequence at the epitope in (1). The mass ratio of each two polypeptides derived from each antigen protein in the polypeptide composition is 1-5: 1, preferably 1-3: 1, and more preferably 1: 1.
As an embodiment of the invention, the polypeptide composition is selected from any two or more polypeptides derived from the antigenic proteins Survivin, ERBB, TERT, eEF, p, WT-235, CDH, KIF20, such as polypeptides derived from Survivin and ERBB, or polypeptides derived from TERT and eEF, or polypeptides derived from p and WT-235, or polypeptides derived from CDH and KIF20, or polypeptides derived from Survivin, ERBB and any one or more selected from TERT, eEF, p, WT-235, CDH, KIF20, or polypeptides derived from p, eEF and any one or more selected from Survivin, ERBB, p, WT-235, CDH, KIF20, or polypeptides derived from p, 235 and Survivin, ERBB, TERT, eEF, CDH, KIF20, or polypeptides derived from any one or more of kh, teff, KIF20, or polypeptides derived from p, teff, WT-235, KIF20, or polypeptides derived from any one or more of kih, KIF20, teff, KIF 20; preferably any two polypeptides derived from the antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF20A, such as polypeptides derived from Survivin and ERBB2, or polypeptides derived from TERT and eEF2, or polypeptides derived from p53 and WT1-235, or polypeptides derived from CDH3 and KIF 20A; preferably, the polypeptide derived from each antigenic protein comprises (1) an epitope of each antigenic protein, the epitope being immunogenic; or contains an epitope having the same activity as the epitope in (1) obtained by amino acid substitution, addition or deletion, preferably having homology of 75% or more with the amino acid sequence of the epitope in (1), more preferably having homology of 85% or more with the amino acid sequence of the epitope in (1), and still more preferably having homology of 95% or more with the amino acid sequence of the epitope in (1). The mass ratio of any two polypeptides derived from each antigen protein in the polypeptide composition is 1-5: 1, preferably 1-3: 1, more preferably 1: 1. the mass ratio of any three polypeptides derived from each antigen protein in the polypeptide composition is 1-5: 1-5: 1, preferably 1-3: 1-3: 1, more preferably 1: 1: 1.
each polypeptide derived from each antigenic protein in the polypeptide composition is within 50 amino acids in length; preferably within 30 amino acids. Specifically, the length of the polypeptide derived from each antigenic protein may be 5-30 amino acids, 8-25 amino acids, 8-15 amino acids, or 9-12 amino acids. As an embodiment of the present invention, each polypeptide derived from each antigenic protein is 9 amino acids in length.
The antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF20A have meanings and sequences well known in the art, including the amino acid wild-type sequence and various known or potentially present variant sequences of each antigenic protein.
The polypeptide derived from the antigen protein Survivin contains an amino acid sequence shown in SEQ ID NO. 1 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 1, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 1, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 1.
The polypeptide derived from the antigen protein ERBB2 contains an amino acid sequence shown in SEQ ID NO. 2 or an amino acid sequence with the same activity obtained by amino acid substitution, addition or deletion, preferably has more than 75% homology with the amino acid sequence shown in SEQ ID NO. 2, more preferably has more than 85% homology with the amino acid sequence shown in SEQ ID NO. 2, and even more preferably has more than 95% homology with the amino acid sequence shown in SEQ ID NO. 2.
The polypeptide derived from the antigen protein TERT contains an amino acid sequence shown in SEQ ID NO. 3 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 3, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 3, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 3.
The polypeptide derived from the antigen protein eEF2 contains an amino acid sequence shown in SEQ ID NO. 4 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 4, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 4, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 4.
The polypeptide derived from the antigen protein p53 contains an amino acid sequence shown in SEQ ID NO. 5 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 5, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 5, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 5.
The polypeptide derived from the antigen protein WT1-235 contains an amino acid sequence shown in SEQ ID NO. 6 or an amino acid sequence thereof obtained by amino acid substitution, addition or deletion and having the same activity, preferably has more than 75% homology with the amino acid sequence shown in SEQ ID NO. 6, more preferably has more than 85% homology with the amino acid sequence shown in SEQ ID NO. 6, and more preferably has more than 95% homology with the amino acid sequence shown in SEQ ID NO. 6.
The polypeptide derived from the antigen protein CDH3 contains an amino acid sequence shown in SEQ ID NO. 7 or an amino acid sequence with the same activity obtained by amino acid substitution, addition or deletion, preferably has more than 75% homology with the amino acid sequence shown in SEQ ID NO. 7, more preferably has more than 85% homology with the amino acid sequence shown in SEQ ID NO. 7, and more preferably has more than 95% homology with the amino acid sequence shown in SEQ ID NO. 7.
The polypeptide derived from the antigen protein KIF20A contains an amino acid sequence shown in SEQ ID NO. 8 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75% of homology with the amino acid sequence shown in SEQ ID NO. 8, more preferably has more than 85% of homology with the amino acid sequence shown in SEQ ID NO. 8, and more preferably has more than 95% of homology with the amino acid sequence shown in SEQ ID NO. 8.
The polypeptide compositions of the present invention may be prepared by conventional methods well known in the art, including, but not limited to, chemical synthesis by solid phase synthesis followed by HPLC to separate the synthesis product from by-products, and expression of a nucleic acid encoding a polypeptide comprising an antigenic fragment of the polypeptide composition of the present invention in living cells or by purification following translation of the above-described encoding nucleic acid by an in vitro cell-free translation system to obtain an antigenic peptide fragment of the polypeptide composition of the present invention. In addition, unwanted small molecules contained in the polypeptide composition of the present invention can be removed by extensive dialysis, and the resulting product can be lyophilized and then other carriers or excipients can be added to form a desired preparation. It will also be appreciated that some amino acids, mutants, chemical modifications, etc., which may be generated in the vaccine components, may be included in the polypeptide compositions of the invention, which do not substantially interfere with the recognition of the epitope sequence by the antibody or TCR.
The polypeptide composition of the present invention binds to HLA (especially HLA-A24 subtype) on the surface of antigen presenting cells such as DC to form HLA-tumor epitope peptide complex which is recognized by TCR and then presented to T cells, so that T cells capable of recognizing the corresponding tumor epitope are specifically activated and expanded, the expanded T cells become Cytotoxic T Lymphocytes (CTL) specifically targeting the tumor associated antigen, and cell-mediated immune killing effect is exerted on tumor cells expressing the tumor associated antigen. That is, the polypeptide composition of the present invention can stimulate in vivo cell-mediated immune response and/or humoral immune response, and produce tumor prevention and/or treatment effect, and can be used for preparing a drug, or a reagent, or a kit for detecting, and/or preventing, and/or treating tumor, wherein the drug, or the reagent, or the kit includes but is not limited to biological products.
Cells loaded with polypeptide compositions
The invention also provides a cell loaded with the polypeptide composition, wherein the cell is an Antigen Presenting Cell (APC) which has an HLA type (A24 subtype) matched with the loaded polypeptide. The antigen presenting cells are professional antigen presenting cells (professional antigen presenting cells) or non-professional antigen presenting cells (non-professional antigen presenting cells). The professional antigen presenting cell is a lymphocyte (B cell), a dendritic cell DC, a macrophage, an endothelial cell or a stem cell, preferably a dendritic cell, macrophage or lymphocyte, more preferably a dendritic cell. The DC can be from an autologous source or a heterologous source, and can also be a DC cell line which is separated from an active individual or artificially constructed and has similar biological characteristics with the natural DC, the shape and/or the gene phenotype of the artificially constructed DC cell line are similar to the natural DC, for example, DC cells which are transduced with a lentiviral vector expressing a Tax gene and are described in CN108546679A have negative CD3 expression, and DC marker molecules such as CD70, CD80, CD83, CD86, CCR7, HLA-DR and the like are all expressed; GEN2.2 cell line, as described in US20050272151A1, which is a plasma cell-like DC cell line, has the phenotype of CD4+, HLA-DR +, CD123+, CD45RA +, CD11c-, CD 13-; DCs can also be differentiated from DC precursor cell lines, such as monocytes differentiated from PBMCs or a MUTZ-3 cell line, the MUTZ-3 cell line being a cell line expressing the monocyte-specific esterase, a monocyte-marker molecule, and CD14 (Santegoes SJ, van den Eertwegh AJ, van de Loosdrecht AA, Scheper RJ, de Gruijl TD. human dentritic cell line models for DC differentiation and clinical DC differentiation study. J Leukoc biol. 2008Dec; 84(6): 1364-73.). The non-professional antigen presenting cells are antigen presenting cells expressing HLA class I.
Those skilled in the art know and understand that Antigen Presenting Cells (APCs) are "pulsed" or loaded with the polypeptide composition described above in a manner that exposes them to an antigen for a time sufficient for the polypeptide composition described above to be presented on the surface of the APC, resulting in polypeptide composition loaded antigen presenting cells; the "exposure" means contacting the antigen presenting cells with the polypeptide composition described above, such as co-incubation culture, and the like. Specifically, the APC is exposed to an antigen in the form of a plurality of short polypeptide fragments, i.e., to an antigenic peptide, which is loaded directly onto the surface of the APC. In addition to short polypeptide fragments, APCs can be incubated with large fragments derived from, intact whole antigen proteins or particles containing antigen proteins, which can be engulfed by the APC into the envelope by endocytosis or the like, then processed by lysosomes or proteasomes into short polypeptide fragments and finally carried and presented to the surface of the APC, binding to HLA on the surface of the APC to form an antigen-presenting complex.
Antigen presenting cells loaded with the polypeptide composition may be prepared by contacting APCs with the polypeptide composition described above in vitro (in vitro) or in vivo (in vivo). When the APCs are loaded in vitro with the antigenic peptide of the polypeptide composition described above, the APCs can be plated for growth on a petri dish or well plate and then exposed to a sufficient amount of the polypeptide composition comprising the antigenic peptide and contacted therewith for a sufficient period of time to allow the antigenic peptide to bind to the APCs. The amount and time required for binding of the antigenic peptide to the APC can be determined by assays well known in the art. Other methods known to those skilled in the art, such as immunoassays or binding assays, can be used to detect whether an APC has an antigenic peptide loaded thereon following exposure to a polypeptide composition comprising an antigenic peptide.
The present invention provides a method for producing a cell loaded with the polypeptide composition, which comprises the steps of contacting the polypeptide composition with an antigen-presenting cell, and loading the polypeptide composition on the antigen-presenting cell to obtain the polypeptide-composition-loaded antigen-presenting cell.
The above polypeptide composition is contacted with antigen presenting cells by co-incubation using any medium conventional in the art suitable for culturing immune cells, such as any one or more of AIM-V, DMEM or RPMI-1640, preferably AIM-V medium, more preferably AIM-V medium without any serum; the incubation period is 1 to 2 days, preferably 2 days, as measured by the ability of the antigenic peptide in the polypeptide composition of the present invention to bind to HLA (subtype A11) on the surface of antigen-presenting cells to form an HLA-antigenic peptide complex; the temperature for co-incubation is room temperature to 37 ℃; the concentration of each polypeptide in the above-mentioned polypeptide composition during the co-incubation is 10 to 100. mu.g/mL, preferably 20 to 80. mu.g/mL, more preferably 40. mu.g/mL.
The antigen presenting cells are contacted with the polypeptide composition to promote the maturation of the antigen presenting cells, but other maturation promoting factors are still required to be added to contact the antigen presenting cells loaded with the polypeptide composition to further promote the maturation of the antigen presenting cells. When the antigen presenting cells loaded with the polypeptide composition are still in an immature state, the method further comprises contacting the antigen presenting cells loaded with the polypeptide composition with maturation-promoting factors to induce maturation of the antigen presenting cells, and obtaining the antigen presenting cells loaded with the polypeptide composition in a mature state.
Contacting the polypeptide-loaded antigen presenting cells with maturation-promoting factors is a co-incubation using any medium conventional in the art suitable for culturing immune cells, such as any one or more of AIM-V, DMEM or RPMI-1640, preferably AIM-V medium, more preferably AIM-V medium without any serum; the co-incubation time is 8-72 hours, preferably 24-48 hours, more preferably 24 hours; the temperature for co-incubation is room temperature to 37 ℃; the maturation promoting factor is one or more of TNF-alpha, IL-1 beta, IL-6, PGE2, IFN-gamma, poly (I: C), R848 or ATP, preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, and the working concentration of TNF-alpha is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, with a working concentration of TNF-alpha of 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably IFN-gamma, poly (I: C) and R848, or preferably IFN-gamma, poly (I: C), R848 and ATP, the working concentration of IFN-gamma is 10-1000IU/mL, preferably 100-300IU/mL, more preferably 100 IU/mL; the working concentration of poly (I: C) is 1-200. mu.g/mL, preferably 20-40. mu.g/mL, more preferably 30. mu.g/mL; the working concentration of R848 is 0.1-50. mu.g/mL, preferably 1-10. mu.g/mL, more preferably 5. mu.g/mL; the working concentration of ATP is 0.1-10mM, preferably 0.1-5mM, more preferably 1 mM; preferably IFN-gamma, poly (I: C), R848 and ATP, the working concentration of IFN-gamma is 100IU/mL, the working concentration of poly (I: C) is 30 mug/mL, and the working concentration of R848 is 5 mug/mL.
The antigen presenting cells include professional antigen presenting cells or non-professional antigen presenting cells. The professional antigen presenting cell is lymphocyte (B cell), dendritic cell DC, macrophage, endothelial cell or stem cell, etc., preferably dendritic cell, macrophage or lymphocyte, more preferably dendritic cell. The non-professional antigen presenting cells are antigen presenting cells expressing HLA class I.
The antigen presenting cell may be differentiated from a precursor cell of the antigen presenting cell, and the preparing may include: the precursor cells of the antigen-presenting cells are contacted with cytokines to induce differentiation into the antigen-presenting cells.
As an embodiment of the present invention, a method for preparing a cell loaded with the above-mentioned polypeptide composition comprises: (1) contacting the polypeptide composition with immature dendritic cells, and loading the polypeptide composition on the immature dendritic cells to obtain immature dendritic cells loaded with the polypeptide composition;
(2) contacting the immature dendritic cells loaded with the polypeptide composition with a dendritic cell maturation-promoting factor to induce the immature dendritic cells to mature, and obtaining the mature dendritic cells loaded with the polypeptide composition.
The contacting in step (1) is a co-incubation, and the culture medium used for the co-incubation is any medium conventional in the art and suitable for culturing immune cells, such as any one or more of AIM-V, DMEM or RPMI-1640, preferably AIM-V culture medium, more preferably AIM-V culture medium without any serum; the incubation period is 1 to 2 days, preferably 2 days, as measured by the ability of the antigenic peptide in the polypeptide composition of the present invention to bind to HLA (subtype A24) on the surface of antigen-presenting cells to form an HLA-antigenic peptide complex; the temperature for co-incubation is room temperature to 37 ℃; the concentration of each polypeptide in the above-mentioned polypeptide composition during the co-incubation is 10 to 100. mu.g/mL, preferably 20 to 80. mu.g/mL, more preferably 40. mu.g/mL.
The contacting in step (2) is co-incubation, and the culture medium used for co-incubation is any medium conventional in the art and suitable for culturing immune cells, such as any one or more of AIM-V, DMEM or RPMI-1640, preferably AIM-V culture medium, more preferably AIM-V culture medium without any serum; the co-incubation time is 8-72 hours, preferably 24-48 hours, more preferably 24 hours; the temperature for co-incubation is room temperature to 37 ℃; the dendritic cell maturation-promoting factor is selected from one or more of TNF-alpha, IL-1 beta, IL-6, PGE2, IFN-gamma, poly (I: C), R848 or ATP, preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, and the working concentration of TNF-alpha is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, with a working concentration of TNF-alpha of 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably IFN-gamma, poly (I: C) and R848, or preferably IFN-gamma, poly (I: C), R848 and ATP, the working concentration of IFN-gamma is 10-1000IU/mL, preferably 100-300IU/mL, more preferably 100 IU/mL; the working concentration of poly (I: C) is 1-200. mu.g/mL, preferably 20-40. mu.g/mL, more preferably 30. mu.g/mL; the working concentration of R848 is 0.1-50. mu.g/mL, preferably 1-10. mu.g/mL, more preferably 5. mu.g/mL; the working concentration of ATP is 0.1-10mM, preferably 0.1-5mM, more preferably 1 mM; preferably IFN-gamma, poly (I: C), R848 and ATP, the working concentration of IFN-gamma is 100IU/mL, the working concentration of poly (I: C) is 30 mug/mL, and the working concentration of R848 is 5 mug/mL.
In step (2), the mature dendritic cells are verified by detecting molecular markers expressed on the surface of mature DCs and/or cytokines secreted by mature DCs as known in the art, including but not limited to CD80, CD83, CD86, CCR7, HLA-ABC, HLA-DR or IL-6. The detection means used in the validation method may be any detection means in the art capable of detecting the above-mentioned molecular markers and/or cytokines, including but not limited to ELISA, Western hybridization or flow cytometry detection.
In the step (1), the immature dendritic cells are formed by differentiating precursor cells of the dendritic cells, and the steps comprise: the precursor cells of the dendritic cells are contacted with cytokines to induce differentiation into immature dendritic cells.
The precursor cells of the dendritic cells are contacted with the cytokine by co-incubation using any medium conventional in the art suitable for culturing immune cells, such as any one or more of AIM-V, DMEM and RPMI-1640, preferably AIM-V medium, more preferably AIM-V medium without any serum; the temperature for co-incubation is room temperature to 37 ℃; the length of the co-incubation is 2 to 6 days, preferably 3 to 5 days, more preferably 5 days.
The precursor cells of the dendritic cells are CD14+ DC precursor cells, preferably monocytes of PBMCs in blood; the cytokine comprises GM-CSF and IL-4, and the working concentration of the GM-CSF is 50-500ng/mL, preferably 50-100ng/mL, and more preferably 100 ng/mL; the working concentration of IL-4 is 5-100ng/mL, preferably 10-50ng/mL, more preferably 50 ng/mL.
The monocytes are obtained by culturing after isolation of PBMCs of the individual. The PBMC may be isolated by methods well known in the art, such as by drawing blood from the individual and separating by density gradient centrifugation. After the isolated PBMC are cultured, adherent cells are basically mononuclear cells. The time for culturing PBMC is preferably 2 to 8 hours, more preferably 8 hours.
The precursor cells of the dendritic cells can also be CD34+ DC precursor cells derived from hematopoietic stem cell lineages, the cytokines comprise GM-CSF and IL-4, the CD34+ DC precursor cells are separated from umbilical cord blood, are subjected to massive amplification and then are contacted with the cytokines GM-CSF and IL-4 to be induced to differentiate into immature DC. Methods for the mass expansion of CD34+ DC precursor cells can be methods known in the art, and can be found in particular in WO2010055900a 1.
The precursor cells of immature DC may also be immortalized DC precursor cell lines, such as the MUTZ-3 cell line described above, which is a cell line expressing the monocyte-marker monocyte-specific esterase and CD 14.
The cell surface of the above-mentioned polypeptide-loaded composition of the present invention forms an HLA-tumor epitope peptide complex, which is presented to T cells by TCR recognition, so that the T cells capable of recognizing the corresponding tumor epitope are specifically activated and expanded, and the expanded T cells become Cytotoxic T Lymphocytes (CTLs) specifically targeting the tumor-associated antigen, and exert a cell-mediated immune killing effect on tumor cells expressing the tumor-associated antigen. That is, the cells of the polypeptide-loaded composition of the present invention can stimulate in vivo cell-mediated immune response and/or humoral immune response, and generate tumor prevention and/or treatment effect, and can be used for preparing drugs, reagents, or kits for detecting, and/or preventing, and/or treating tumors, wherein the drugs, reagents, or kits include but are not limited to biological products.
Activated immune effector cells
The invention also provides an activated immune effector cell, which is prepared by the following method: the polypeptide composition of the invention is loaded into cells and inactivated immune effector cells. The cells such as DC loaded with the polypeptide composition provided by the invention express a certain subtype of HLA such as A24 on the surface thereof, and antigen peptide matched with the certain subtype of HLA such as tumor antigen peptide is combined with the antigen peptide to form an HLA-antigen peptide complex, and then is recognized and combined by a receptor TCR which specifically recognizes the complex on the surface of the T cell, so that the T cell expressing the TCR is stimulated and starts to proliferate.
The immune effector cell is selected from a T cell or an NK cell, preferably a T cell. The cells loaded with the polypeptide composition of the present invention are selected from lymphocytes (B cells), dendritic cell DCs, macrophages, endothelial cells or stem cells, etc., preferably lymphocytes, macrophages or dendritic cells loaded with the polypeptide composition of the present invention, and more preferably dendritic cells loaded with the polypeptide composition of the present invention. The immune effector cells are from the same individual or from a different individual, preferably from the same individual, as the cells loaded with the polypeptide composition of the invention. The ratio of the number of the polypeptide composition-loaded cells to the number of the immune effector cells is any ratio known in the art, as long as the polypeptide composition-loaded cells can effectively activate the immune effector cells recognizing the surface HLA-antigenic peptide complex thereof, and the ratio of the number of the polypeptide composition-loaded cells to the number of the immune effector cells is preferably 1: 10-50, more preferably 1: 10-30, and even more preferably 1: 10.
The contacting of the polypeptide composition-loaded cells and immune effector cells is a co-incubation. The length of the co-incubation is 2 to 48 hours, preferably 24 to 48 hours, more preferably 24 hours. The co-incubation is carried out in a medium, which is AIM-V, DMEM or RPMI1640, preferably AIM-V medium, more preferably AIM-V medium with 2% V/vbs, as in a preferred embodiment IL-2 is also included in the AIM-V medium, with a working concentration of IL-2 of 10-100U/mL, preferably 100U/mL.
The activated immune effector cells are Cytotoxic T Lymphocytes (CTL) which specifically target tumor-associated antigens, and have a cell-mediated immune killing effect on tumor cells expressing the tumor-associated antigens. That is, the activated immune effector cells of the present invention can stimulate cell-mediated immune response and/or humoral immune response in vivo to produce tumor prevention and/or treatment effect, and can be used for preparing drugs, reagents or kits for detecting, and/or preventing, and/or treating tumors, wherein the drugs, reagents or kits include but are not limited to biological products.
Medicaments, reagents or kits
The drug, reagent or kit has the activity of detecting, and/or preventing, and/or treating tumors, and comprises but is not limited to biological products, cells containing the polypeptide composition, and/or loaded with the polypeptide composition, and/or activated immune effector cells.
The medicament, reagent or kit further comprises an adjuvant and/or an immunomodulator. Adjuvants or immunomodulators are small molecules, biological macromolecules, compositions, complexes or extracts of compounds known in the art to enhance the effect of an immune response and/or reduce the adverse side effects of an immune response. The adjuvant or immunomodulator can be delivered to target cells or target organisms before the polypeptide composition, or together with the polypeptide composition or later than the polypeptide composition, so as to enhance the immune response of the cells or organisms to antigens or change the type of the immune response and reduce the toxic and side effects of the immune response.
The adjuvant or immunomodulator is selected from the group consisting of aluminum adjuvants (e.g., aluminum hydroxide, aluminum salts), freund's adjuvants (e.g., complete freund's adjuvant and incomplete freund's adjuvant), prostaglandin E2, interferon alpha, corynebacterium parvum, lipopolysaccharide, cytokines, oil-in-water emulsions, water-in-oil emulsions, nanoemulsions, microparticle delivery systems, liposomes, microspheres, biodegradable microspheres, plaque virosomes, proteoliposomes, proteasomes, immunostimulatory complexes (ISCOMs, ISCOMATRIX), microparticles, nanoparticles, biodegradable nanoparticles, silicon nanoparticles, polymeric microparticles/nanoparticles, polymeric platelet-like substrate particles (PLSP), microparticle resins, nanoliposome polymeric gels (nanolipogel), synthetic/biodegradable and biocompatible semisynthetic or natural polymers or dendrimers (e.g., PLG, PLGA, PLA, b, Polycaprolactone, silicon polymers, polyesters, polydimethylsiloxanes, sodium polystyrene sulfonate, polystyrene benzyl trimethyl ammonium chloride, polystyrene divinylbenzene resin, polyphosphazenes, poly- [ di- (carboxyacetylphenoxy) phosphazene (PCPP), poly- (methyl methacrylate), dextran, polyvinylpyrrolidone, hyaluronic acid and derivatives, chitosan and derivatives, polysaccharides, delta inulin polysaccharide, glycolipids (synthetic or natural), lipopolysaccharides, one or more polycationic compounds (e.g., polyamino acids, poly- (gamma-glutamic acid)), poly-arginine-HCl, poly-L-lysine, polypeptides, biopolymers), cationic dimethyldioctadecyl ammonium (DDA), alpha-galactosylceramide and derivatives, archaebacteria lipids and derivatives, poly (vinylogous ammonium chloride), poly (vinylogoniumyl chloride), poly, One or more of lactams, galens, glycerides, phospholipids and spirochetes.
The medicament, the reagent or the kit also contains pharmaceutically acceptable carriers and/or excipients, and the carriers or excipients do not generate any substantial influence on the properties, the biological functions and the like of the polypeptide composition, and/or cells carrying the polypeptide composition, and/or activated immune effector cells.
The medicine, reagent or kit can be prepared into any pharmaceutically acceptable dosage form with detection, and/or prevention, and/or treatment effects, such as lyophilized powder or liquid preparation, wherein the liquid preparation contains sterilized water for injection or organic solvent, such as DMSO.
The drug or agent comprising the polypeptide composition can be injected into a subject to form an HLA-antigen peptide complex by binding to an HLA-presenting cell (e.g., DC) loaded into the subject, which recognizes a corresponding subtype of an antigen peptide in the tumor vaccine, and the HLA-antigen peptide complex binds to a corresponding specific TCR to activate T cells expressing the specific TCR.
Antigen-presenting cells obtained by differentiating precursor cells (e.g., DC precursor cells) of antigen-presenting cells obtained by separation from autologous blood, such as hematopoietic precursor cells derived from cord blood CD34+ or DC obtained by differentiating mononuclear cells derived from peripheral blood CD14+, may be co-incubated with the polypeptide composition of the present invention in vitro to obtain antigen-presenting cells (particularly antigen-presenting cells in a mature state loaded with the polypeptide composition) containing the polypeptide composition and the loaded polypeptide composition of the present invention, and then introduced into the body to present the loaded antigen peptides thereof by the antigen-presenting cells and thereby activate specific T cell responses; an agent that reduces immune rejection, such as an inhibitor that inhibits endogenous TCR expression, is administered concurrently with infusion into the body.
Administration of the drug or agent may occur before or after surgical removal of the tumor, or before or after treatment of the tumor with chemoradiotherapy. The drug or agent may be administered to the patient together or in combination with other compositions or drug products. It is understood that the medicament of the present invention may be administered to an individual not having a tumor but at risk of having a tumor, in addition to an individual already having a tumor.
The medicament may be administered to the groin joint by intra-nodal injection, subcutaneously or intradermally to the extremities of the patient to be treated for cancer, or by other routes of administration, such as intramuscular injection or blood injection.
Tumors that can be treated and/or prevented by the drug administration include, but are not limited to, lung cancer, non-small cell lung cancer, ovarian cancer, colon cancer, rectal cancer, melanoma, kidney cancer, bladder cancer, breast cancer, liver cancer, lymphoma, hematological malignancies, head and neck cancer, glioma, mesothelioma, colorectal cancer, stomach cancer, nasopharyngeal cancer, laryngeal cancer, cervical cancer, uterine body tumor and osteosarcoma, bone cancer, pancreatic cancer, renal cell cancer, skin cancer, prostate cancer, cutaneous or intraocular malignant melanoma, uterine cancer, cancer of the anal region, testicular cancer, cancer of the fallopian tubes, endometrial cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, non-Hodgkin's lymphoma, esophageal cancer, small intestine cancer, cancer of the endocrine system, cancer of the bile ducts, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, urothelial cancer, penile cancer, chronic or acute leukemia (including acute myeloid leukemia, colon cancer, colon, Chronic myeloid leukemia, acute lymphoblastic leukemia, chronic lymphocytic leukemia), childhood solid tumors, lymphocytic lymphomas, renal or ureteral cancer, renal pelvis cancer, Central Nervous System (CNS) tumors, primary CNS lymphoma, tumor angiogenesis, spinal tumors, brain stem glioma, pituitary adenoma, kaposi's sarcoma, hodgkin's lymphoma, epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma, environmentally induced cancers including asbestos-induced cancers and various types of leukemia and lymphoma, and various types of precancerous lesions.
Method of eliciting an immune response in vivo
In order to achieve the effect of preventing and/or treating tumors, the invention also provides a method for inducing an immune response in vivo, wherein an effective dose of one or more of the polypeptide composition, cells loaded with the polypeptide composition and activated immune effector cells is delivered into the body.
For prophylactic purposes, one or more of the polypeptide compositions, cells loaded with the polypeptide compositions, and activated immune effector cells of the invention are administered to a subject prior to the occurrence of a tumor or neoplastic lesion. In some cases, the medicament is administered to the subject individual after the onset of one or more of the above-mentioned tumors, in order to prevent the occurrence of further symptoms or the worsening of already occurring symptoms. Prophylactic administration of one or more of the polypeptide composition of the invention, the polypeptide composition-loaded cell and the activated immune effector cell is intended to prevent or alleviate any subsequent symptoms. For therapeutic purposes, one or more of the polypeptide compositions, polypeptide composition-loaded cells, and activated immune effector cells of the invention are administered to a subject individual at or after the onset of cancer, in order to alleviate the symptoms of cancer that has already developed.
The effective dose for any particular therapeutic application will depend on various factors, such as the type of cancer, the extent of the onset of the cancer, the condition of the subject individual, such as age, sex, weight, level of various indicators of the body, and the like, as well as the particular pharmaceutical agent components being administered and the particular mode of administration, and may, for example, be administered by including, but not limited to, intravenously, intramuscularly, intradermally, transdermally, intraarterially, intraperitoneally, intralesionally, intracranially, intraarticularly, intraprostatically, intrapleurally, intratracheally, intrathecally, intranasally, intravaginally, intrarectally, parenterally, systemically, locally, intratumorally, intraperitoneally, intracerebroventricularly, subcutaneously, subconjunctivally, transmucosally, intrapericardially, intraumbilically, intraorbitally, orally, transdermally, intrapulmonary, by inhalation, by injection, by implantation, by reinfusion, by continuous reinf, The subject is administered to the individual via one or more of a catheter, via lavage, with an emulsion, and with a liposome composition. For the effective dose of the administered drug comprising one or more of the polypeptide composition of the invention, the cells loaded with the polypeptide composition, and the activated immune effector cells, one skilled in the art can empirically determine the specific components included in the drug without conducting further unnecessary experiments.
The invention has the following positive effects: the polypeptide composition provided by the invention presents and activates specific Cytotoxic T Lymphocytes (CTL) after loading DC, thereby achieving the target toxicity effect on tumor cells. The polypeptide composition, the tumor vaccine, the DC vaccine and the pharmaceutical composition derived from the polypeptide composition can obviously activate immune effector cells, particularly T cells, obviously improve the secretion level of cytokines relevant to the activation of the T cells and the killing level of the T cells, and have potential clinical value.
Some terms related to the present invention are explained below. Terms used herein are used as they are commonly used in the art, unless otherwise defined as follows.
In the present invention, the term "polypeptide" refers to a molecule consisting of monomers (amino acids) linearly linked by amide bonds (also known as peptide bonds). The term "polypeptide" refers to any chain or chains of two or more amino acids, and does not refer to a specific length of the product. Thus, peptides, dipeptides, tripeptides, oligopeptides, "proteins," "amino acid chains," or any other term used to refer to one or more chains of two or more amino acids, are included in the definition of "polypeptide," and the term "polypeptide" may be used instead of, or interchangeably with, any of these terms. The term "polypeptide" is also intended to refer to the product of a modification of a polypeptide following expression, including without limitation glycosylation, acetylation, phosphorylation, amidation, derivatization by known protecting/blocking groups, proteolytic cleavage, or modification by non-standard amino acids. The polypeptide may be derived from a natural biological source or produced by recombinant techniques, but is not necessarily translated from a specified nucleic acid sequence. It may be produced in any manner, including by chemical synthesis.
The term "… … -derived polypeptide" refers to a full-length, fragment, modification, or mutant protein or polypeptide. The term "fragment" when referring to a polypeptide can be a polypeptide of any length that retains the antigenicity of the full-length protein or polypeptide. In one or more embodiments, the fragment is an epitope of the full-length protein. In one or more embodiments, the fragment comprises up to 50, 40, 30 amino acids. For example, the fragment may be 5-30 amino acids, 8-25 amino acids, 8-15 amino acids, or 9-12 amino acids in length, or any combination of 5 of the above endpoints, such as 9-15 amino acids. In one or more embodiments, the fragment may be 9 amino acids in length.
The term "variant" or "mutant" refers to a peptide or polypeptide that has an amino acid sequence that is altered by insertion, deletion, or substitution (substitution) of one or more amino acids as compared to a reference sequence, but retains at least one biological activity. In one or more embodiments, the variant or mutant comprises an amino acid sequence that has at least 75%, preferably at least 80%, preferably at least 85%, preferably at least 90%, preferably at least 95%, preferably at least 97% sequence identity to a reference sequence (e.g., the fragments of SEQ ID NOs: 1,2, 3, 4, 5, 6, 7, 8, 9 described herein) and retains the biological activity (e.g., as an epitope) of the reference sequence. The substitution may be a non-conservative amino acid substitution or a conservative amino acid substitution. Conservative amino acid substitutions refer to substituted amino acids that have similar structural and chemical properties, similar or similar properties, and do not alter the function of the protein or polypeptide as the corresponding amino acid in the wild-type polypeptide. For example, conservative amino acid substitutions include families of amino acid residues with similar side chains, including amino acids with basic side chains (e.g., lysine, arginine, histidine), acidic side chains (e.g., aspartic acid, glutamic acid), uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), nonpolar side chains (e.g., alanine, valine, leucine, isoleucine proline, phenylalanine, methionine, tryptophan), beta-branched side chains (e.g., threonine, valine, isoleucine), and aromatic side chains (e.g., tyrosine, phenylalanine tryptophan, histidine). Thus, substitution of one or more sites with another amino acid residue from the same side chain species in the polypeptide of the invention will not substantially affect its activity.
The term "wild-type" has its meaning as understood in the art, which refers to an entity having a structure and/or activity in a "normal" (as opposed to mutant, patient, modifier, etc.) state or situation as found in nature. Those skilled in the art will appreciate that wild-type genes and polypeptides typically exist in a variety of different forms (e.g., alleles).
The term "tumor-associated antigen" or "TAA" refers to an antigen that is specifically expressed by a tumor cell or is expressed by a tumor cell at a higher frequency or density than non-tumor cells of the same tissue type. The tumor-associated antigen may be an antigen that is not normally expressed by the host; they may be aberrantly manifested by mutation, truncation, misfolding, or other means of the molecule normally expressed by the host; they may be the same as normally expressed molecules but expressed at abnormally high levels; or they may be expressed in an abnormal situation or environment. The tumor-associated antigen can be, for example, a protein or protein fragment, a complex carbohydrate, a ganglioside, a hapten, a nucleic acid, or a combination of these or other biomolecules.
The term "vaccine" refers to an immunogenic composition for administration to a mammal for eliciting an immune response in the mammal against a particular antigen. Vaccines typically comprise an agent (known as an "antigen" or "immunogen") that is similar to or derived from the target of the immune response, such as the microbe or tumor cell causing the disease. Vaccines intended for the treatment of tumours, such as cancers, typically comprise antigens derived from and capable of eliciting immunogenicity to tumour-associated antigens found on the tumour of interest.
The term "Survivin" refers to an apoptosis-inhibiting protein that is an expression product of Survivin, and may be a human apoptosis-inhibiting protein or a non-human apoptosis-inhibiting protein.
The term "ERBB 2" refers to a 185kDa cell membrane receptor encoded by the proto-oncogene erbB-2, which is one of the epidermal growth factor receptor family members.
The term "TERT" refers to the transfection of telomerase reverse transcriptase protein by the expression product of the TERT gene.
The term "eEF 2" refers to the expression product of the eEF2 gene, eukaryotic elongation factor 2.
The term "p 53" refers to the expression product p53 protein of p53 gene, which may be human p53 protein or non-human mammalian p53 protein.
The term "WT 1-235" refers to the WT1-235 peptide which is the expression product of the WT1-235 gene.
The term "CDH 3" refers to the expression product of the cadherin 3 gene.
The term "KIF 20A" refers to the expression product of KIF20A gene, kinesin family 20A.
The terms "cancer," "tumor," and "malignant" refer to or describe a physiological condition typically characterized by uncontrolled cell growth in mammals. Examples of cancers include, but are not limited to, epithelial cancers, including adenocarcinomas, lymphomas, blastomas, melanomas, sarcomas, and leukemias. More specific examples of such cancers include squamous cell cancer, small-cell lung cancer, non-small cell lung cancer, gastrointestinal cancer, hodgkin's and non-hodgkin's lymphomas, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer (such as liver cancer and hepatoma), bladder cancer, breast cancer (including hormone-mediated breast cancer), colon cancer, colorectal cancer, endometrial cancer, myeloma (such as multiple myeloma), salivary gland carcinoma, kidney cancer (such as renal cell carcinoma and wilms ' tumor), basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, blood cancer (including but not limited to Acute Myelogenous Leukemia (AML) and Multiple Myeloma (MM)), various types of head and neck cancer (including but not limited to squamous cell carcinoma), and cancers of mucinous origin (such as mucinous ovarian cancer), Cholangiocarcinoma (liver) and renal papillary carcinoma. In certain embodiments, the blood cancer is selected from the group consisting of: hodgkin's lymphoma, non-hodgkin's lymphoma, multiple myeloma, acute lymphoblastic leukemia, acute myelogenous leukemia, chronic lymphocytic leukemia, and chronic myelogenous leukemia.
The term "immunomodulator" means a substance that, when mixed with an immunogen, is capable of eliciting a stronger immune response than the immunogen alone, and/or reducing the toxic side effects of the immunity. For example, an immunopotentiator can enhance immunogenicity and provide an excellent immune response. As another example, an immunopotentiator can act by increasing the expression of costimulators on macrophages and other antigen presenting cells.
The term "adjuvant" refers to a non-specific immunopotentiator which, when delivered to the body with or in advance of an antigen, enhances the body's immune response to the antigen or alters the type of immune response, and/or reduces immunotoxic side effects. Adjuvants are of various types, including, but not limited to, aluminum adjuvants (e.g., aluminum hydroxide), Freund's adjuvants (e.g., complete Freund's adjuvant and incomplete Freund's adjuvant), Corynebacterium parvum, lipopolysaccharide, cytokines, and the like. Freund's adjuvant is currently the most commonly used adjuvant in animal testing. Aluminum hydroxide adjuvants are used more often in clinical trials.
The term "DC maturation-promoting factor" refers to any protein, nucleic acid, polypeptide, complex, extract, isolate, or composition thereof that is capable of promoting the conversion of immature DCs to mature DCs, which can be converted to mature DCs by contact with immature DCs. The mature DC may be verified by detection of molecular markers expressed on the surface of mature DCs and/or cytokines secreted by mature DCs as is well known in the art, including but not limited to CD80, CD83, CD86, CCR7, HLA-ABC, HLA-DR, and IL-6.
The term "isolated" refers to a state that is formed by artificially isolating, purifying or de novo synthesizing naturally occurring biological macromolecules such as proteins, polypeptides, nucleic acids, antibodies, or complexes thereof from a natural state in vivo.
The term "working concentration" refers to the actual concentration of an agent or active ingredient at which it is effective in a solution system. Usually, a certain reagent or a certain effective component is configured to be a mother solution or a stock solution with higher concentration before use, and is added into a final reaction system according to a certain proportion to be diluted when in use, and the final concentration obtained after dilution is usually the working concentration.
The term "loaded" refers to a protein, polypeptide, or nucleic acid molecule that binds directly to a receptor on the surface of a cell, forming a complex of the cell and the protein, polypeptide, or nucleic acid molecule. The binding may be covalent or non-covalent, including binding of the receptor to the ligand. For example, an antigen peptide-loaded DC is a DC-antigen peptide complex formed by binding an antigen peptide to an HLA molecule expressed on the surface of the DC and matching the HLA type of the antigen peptide.
The term "DC-Cytotoxic T Lymphocyte" or "DC-CTL" refers to a Cytotoxic T Lymphocyte (CTL) activated by mature DC loaded with an antigen peptide, which is capable of specifically binding to an antigen expressing the antigen containing the antigen peptide loaded by the mature DC and producing a cell killing effect on cells expressing the antigen, and is a major performer of cellular immunity mediated by DC.
The term "antigen presenting cell" (APC) refers to a class of cells capable of expressing Major Histocompatibility Complex (MHC) class I or II, a class of cells capable of activating T cells by MHC binding to an antigenic peptide to form an MHC-antigenic peptide complex and further binding to a receptor on the surface of a T cell, including but not limited to Dendritic Cells (DC), monocytes/macrophages, B cells, Langerhans cells.
The term "antigen-loaded antigen presenting cell" includes an APC that has been exposed to an antigen and activated by the antigen. For example, the APC can be loaded with antigen in vitro (e.g., during culture in the presence of antigen). APCs can also be loaded in vivo by exposure to an antigen. "antigen-loaded APCs" are typically prepared in one of two ways: (1) small fragments, called antigenic peptides, are "pulsed" directly outside the APC for binding to MHC molecules; (2) the APCs are incubated with the polypeptide large fragments, complete proteins or protein particles, which are then taken up by the APCs. These large polypeptide fragments or protein molecules are digested by the APC into small peptide fragments and eventually transported and presented on the surface of the APC. Furthermore, antigen-loaded APCs can also be generated by introducing a polynucleotide encoding an antigen into a cell.
Drawings
FIG. 1 shows the ratio of different polypeptide composition-loaded DC surface maturation markers in cytokine-induced culture of donor subjects (sample 1). Each DC group loaded with different polypeptide compositions is a schematic proportion of the mature markers CD80, CD86, CD83 from left to right in turn. Control refers to DC cells not loaded with any polypeptide.
FIG. 2 is a graph showing the ratio of DC-CTL surface markers loaded with different polypeptide compositions in flow assay donor subjects (sample 1 and sample 2).
FIG. 3 is a graph showing the killing effect of DC-CTL loaded with different polypeptide compositions on pancreatic cancer tumor cells PANC-1 from donor subjects (sample 1) in example 3.
FIG. 4 is a graph showing the killing effect of DC-CTL loaded with different polypeptide compositions on pancreatic cancer tumor cells PANC-1 from the donor subject (sample 2) in example 3.
FIG. 5 is a graph showing IFN-. gamma.secretion levels of supernatants after killing of pancreatic cancer tumor cells PANC-1 by DC-CTL loaded with different polypeptide compositions from donor subjects (sample 1) in example 3.
FIG. 6 is a graph showing IFN-. gamma.secretion levels of supernatants after killing of pancreatic cancer tumor cells PANC-1 by DC-CTL loaded with different polypeptide compositions from donor subjects (sample 2) in example 3.
The "control group" indicated in the above figures refers to the experimental group of T cells stimulated with DCs not loaded with any polypeptide, unless otherwise specified.
The labeled "peptide combination 1 group, group 1, combination 1, loaded polypeptide combination 1, peptide combination 1" in the above figures all refer to the experimental group loaded with polypeptide combination 1; "peptide combination 2 group, group 2, combination 2, polypeptide loaded combination 2, peptide combination 2" all refer to the experimental group loaded with polypeptide combination 2; "peptide combination 3 group, group 3, combination 3, polypeptide-loaded combination 3, peptide combination 3" all refer to the experimental group loaded with polypeptide composition 3; "peptide combination 4, group 4, combination 4, polypeptide loaded combination 4, peptide combination 4" all refer to the experimental group loaded with polypeptide composition 4.
Detailed Description
The invention is further illustrated by the following examples, which are not intended to limit the scope of the invention. The experimental methods without specifying specific conditions in the following examples were selected according to the conventional methods and conditions, or according to the commercial instructions.
The "room temperature" referred to in the examples refers to the temperature between the runs where the tests were performed, and is generally 25 ℃.
The "overnight" stated in the examples means more than 8 hours.
The following examples provide various polypeptide compositions comprising a plurality of tumor antigen peptides, each of which comprises at least 2 tumor-associated epitope polypeptides for HLA-A24 typing, for HLA-A24 typing, which is a high proportion of Chinese population. The polypeptides used in the following examples are shown in table 1 below:
TABLE 1
Sequence of Sequence Listing numbering Source
AYACNTSTL SEQ ID NO:1 Survivin
RWGLLLALL SEQ ID NO:2 ERBB2
VYGFVRACL SEQ ID NO:3 TERT
AYLPVNESF SEQ ID NO:4 eEF2
AIYKQSQHM SEQ ID NO:5 P53
CYTWNQMNL SEQ ID NO:6 WT1-235
DYLNEWGSRF SEQ ID NO:7 CDH3
KVYLRVRPLL SEQ ID NO:8 KIF20A
The polypeptide compositions used in the following examples are shown in table 2 below:
TABLE 2
Figure BDA0002673342600000191
The DC precursor cells and T cells in the following examples were isolated from blood PBMC of donor subjects (sample 1 and sample 2) by the following steps: mixing blood of an HLA-A24 type donor subject with equal volume of normal saline, wherein the total volume is 35mL, slowly adding the mixture into a centrifugal tube containing 15mL of Ficoll along the tube wall, so that the blood and the Ficoll are obviously layered, centrifuging for 20min at 800g, absorbing a white cell layer after centrifugation, transferring the white cell layer into another centrifugal tube, adding normal saline, centrifuging for 10min at 1500rpm, washing, discarding waste liquid, and adding normal saline for centrifugal washing once. The cleaned cells are transferred into a culture bottle to be cultured and attached overnight, suspension cells, namely T cells, are collected, counted and frozen, and the rest attached cells are mononuclear cells (DC precursor cells) which can be induced and differentiated into DC.
Unless otherwise specified in the following examples, the working concentration of each polypeptide in the polypeptide composition used is 40. mu.g/mL.
EXAMPLE 1 Effect of different HLA-A24-binding polypeptide compositions in Donor Subjects (sample 1 and sample 2) on DC maturation
The operation steps of sample 1 and sample 2 are the same, specifically as follows:
(1) culture of immature DC cells: DC precursor cells were cultured by adding serum-free AIM-V medium (purchased from Gibco), and IL-4 and GM-CSF to working concentrations of 50ng/mL and 100ng/mL, respectively. And (5) culturing to the third day, changing the liquid by half, and supplementing IL-4 and GM-CSF to respective working concentrations of 50ng/mL and 100 ng/mL.
(2) Polypeptide loading of immature DC cells: after the fifth day, the DC precursor cells were divided into 5 groups, which were control group, group 1, group 2, group 3, and group 4, respectively. The polypeptide composition 1 mixture was added to group 1, the polypeptide composition 2 mixture was added to group 2, the polypeptide composition 3 mixture was added to group 3, and the polypeptide composition 4 mixture was added to group 4. The working concentration of each peptide was 40. mu.g/mL. The cells in the control group did not have any antigenic peptide added.
(3) Maturation and detection of DC: and (4) adding IFN-gamma, poly (I: C) and R848 according to the indicated dosage until the seventh day of culture, wherein the working concentration of the IFN-gamma is 100IU/mL, the working concentration of the poly (I: C) is 30 mu g/mL, and the working concentration of the R848 is 5 mu g/mL, and continuing to culture for 24 hours to further stimulate the activated DC. The cultured DC cells were tested using CD80, CD83, CD86 flow antibodies.
As a result:
the DC cells after being stimulated by the cytokines comprise DC cells of a control group which is not loaded with the polypeptide, a group 1 loaded with the polypeptide composition, a group 2 loaded with the polypeptide composition, a group 3 loaded with the polypeptide composition and a group 4 loaded with the polypeptide composition, and as can be seen from a detection result of a sample 1 shown in figure 1, positive rates of CD80, CD86 and CD83 are all greater than 99%, and the DC maturation degree after induction is good. There was no significant difference between the control group and the experimental group loaded with different polypeptide compositions. The DC maturity of each experimental group of sample 2 was similar to the results of sample 1 and is not shown.
Example 2 detection of cell activation phenotype of different polypeptide compositions in Donor Subjects (sample 1 and sample 2) to induce DC-activated T cells
The operation steps of sample 1 and sample 2 are the same, specifically as follows:
(1) antigen peptide-loaded DCs were incubated with T cells: the DCs loaded with different antigen peptides in example 1 are co-incubated with T cells corresponding to donor according to the ratio of 1:10, and the incubation time is 72h to form DC-CTLs. The culture conditions were AIM-V medium supplemented with 2% FBS, 100U/mL IL-2 factor, with control antigen-unloaded DCs.
(2) Flow-type phenotype: the DC-CTLs were labeled with CD3, CD4, and CD8 antibodies and the proportion of CD3CD4+ and CD3CD8+ cells in the different experimental groups was analyzed by flow assay. The corresponding cell phenotype ratios were analyzed by labeling DC-CTLs with antibodies CD3+ CD4/CD8, CD3+/CD 137.
As a result:
flow assay results fig. 2 shows that, in samples 1 and 2, the ratio of CD3+ CD4+ T cells and CD3+ CD8+ T cells in each peptide-loaded group was not significantly changed compared to the control group of T cells after DC were loaded with polypeptide composition 1, polypeptide composition 2, polypeptide composition 3, and polypeptide composition 4 and co-incubated with T cells. We also examined the marker CD137 associated with T cell activation, and the results showed that neither the loaded nor unloaded polypeptide combination formed a significant difference, whether sample 1 or sample 2.
The above results show that the loaded polypeptide composition yielded DC-CTLs with less effect on the ratio of CD3+ CD4+, CD3+ CD8+, and no significant upregulation of T cell activation markers was found in both samples. It was shown that in some samples, the polypeptide-loaded DC had limited stimulation of T cells.
Example 3 Effect of different polypeptide compositions in Donor Subjects (sample 1 and sample 2) on the specific killing Effect of DC-activated T cells on pancreatic cancer cells PANC-1
The method comprises the following operation steps:
(1) tumor cell line killing experiment:
the killing effect of DC-CTLs of different experimental groups in example 2 on pancreatic cancer tumor cells PANC-1 was tested by using a real-time unlabeled cell function analyzer (RTCA). The pancreatic cancer tumor cell PANC-1 is HLA-A24 type, and is the same as the HLA type of the patient and the HLA type of the used tumor antigen peptide. The method comprises the following specific steps:
(a) zero setting: adding 50 mu L of DMEM culture solution into each hole, putting the DMEM culture solution into an instrument, selecting step 1 and zeroing;
(b) target cell plating: pancreatic cancer cells PANC-1 (purchased from American type culture Collection ATCC) were plated at 104 cells/50. mu.l/well on a plate containing a detection electrode, left for several minutes, and after the cells were stabilized, they were put into the apparatus again, step 2 was started, and the cells were cultured;
(c) adding effector cells: after the target cells are cultured for 24h, step 2 is suspended. The DC-CTLs obtained as described in example 2 using polypeptide composition 1, polypeptide composition 2 and polypeptide composition 3 were added in an amount of 50. mu.l per well, and the effective target ratio was set to 10:1, respectively, so that step 3 was started using T cells from the same donor subject without any polypeptide composition as a control, and after further 24 hours of co-culture, cell function curves recorded by the instrument were observed.
(2) Post-killer supernatant cytokine detection
And (3) detecting the contents of IL-2, IL-4, IL-6, IL-10, TNF-a and IFN-gamma cytokines in the supernatant after killing by using a cytokine kit.
As a result:
and (3) killing curves of DC-CTL (dendritic cell-cytotoxic T lymphocyte) on target cells PANC-1, which are obtained by carrying the polypeptide composition 1, the polypeptide composition 2, the polypeptide composition 3 and the polypeptide composition 4 on the DC respectively and then incubating the DC with T cells. In sample 1 (fig. 3), each experimental group loaded with the polypeptide composition had a stronger killing effect 24h after the addition of effector cells, compared to the DC-CTL group not loaded with the polypeptide composition, indicating that the loaded HLA-a24 antigen peptide can be effectively presented to T cells and enhance the killing ability of T cells. Further analysis showed that the killing curve of the loaded polypeptide composition 1 was lower than that of the polypeptide composition 2, the polypeptide composition 3 and the polypeptide composition 4, and showed better killing ability to PANC-1 cells. In sample 2 (as shown in FIG. 4), each experimental group loaded with the polypeptide composition also had a stronger killing effect after 24h addition of effector cells, compared to the DC-CTL group not loaded with the polypeptide composition, indicating that the HLA-A24 antigen peptide loaded in this sample can also be effectively presented to T cells and enhance the killing ability of T cells. Further analysis showed that the killing curves of the loaded polypeptide composition 1 and the polypeptide composition 2 were lower than those of the polypeptide composition 3 and the polypeptide composition 4, showing a better killing ability on PANC-1 cells, with combination 2 being slightly better than combination 1 in sample 2.
The results of the two representative samples show that compared with the DC-CTL group without the loaded polypeptide composition, each experimental group loaded with the polypeptide composition shows stronger killing effect after the effector cells are added for 24 hours, and the loaded HLA-A24 antigen peptide can be effectively presented to the T cells and enhances the killing capacity of the T cells. Among the four groups of HLA-A24 antigen peptides, the DC-CTL loaded with antigen peptide combination 1 and combination 2 has stronger killing capacity on pancreatic cancer cells PANC-1, and has different killing performances in different samples.
And (3) detecting the concentration of IFN-gamma cell factors in cell supernatant after the PANC-1 killing by flow. In sample 1 (as shown in fig. 5), the concentration of IFN- γ cytokine in the supernatant of the peptide composition 2-loaded experimental group was the highest and was 4701.22pg/mL, followed by peptide composition 3-loaded experimental group, peptide composition 4, and peptide composition 1 experimental group, which were 4656.65pg/mL, 4525.71pg/mL, and 4482.90pg/mL, respectively, and no significant difference between the peptide-loaded groups was higher than 3889.19pg/mL of the control group. In sample 2 (FIG. 6), the concentrations of IFN-. gamma.cytokines were highest in the supernatants of the experimental groups loaded with peptide composition 1 and polypeptide composition 2, 2699.78pg/mL and 2648.58pg/mL, respectively, followed by 1764.26pg/mL and 1270.27pg/mL of the experimental group loaded with peptide composition 3 and peptide composition 4, respectively, which were also higher than 1270.27pg/mL of the control group. The results show that the secretion level of the cell factor IFN-gamma after killing in part of samples has certain difference with the killing result.
The above results indicate that each experimental group loaded with polypeptide showed higher IFN- γ secretion level than the control group not loaded with polypeptide, which is the same as the killing result. However, the level of IFN- γ secretion after killing was partially different from the killing results in some samples.
The two representative experimental results are combined to show that the DC-CTL experimental group of each loaded polypeptide composition has higher killing capability and cytokine IFN-gamma secretion level compared with a control group without the loaded polypeptide, and the DC-CTL experimental group has higher killing capability and cytokine IFN-gamma secretion level compared with the DC-CTL experimental group without the loaded polypeptide composition. Among them, the two experimental groups loaded with the antigenic peptides of combination 1 and combination 2 had stronger killing ability than combination 3 and combination 4. These preferred antigenic compositions will have high potential medical value in the future in the treatment of tumors typified by PANC-1 tumor cells.
All documents referred to herein are incorporated by reference into this application as if each were individually incorporated by reference. Furthermore, it should be understood that various changes and modifications can be made by those skilled in the art after reading the above disclosure, and equivalents also fall within the scope of the invention as defined by the appended claims.
Sequence listing
<110> Shanghai cell therapy group Co., Ltd
<120> polypeptide composition specifically binding HLA-A24 typing and application
<130> 19A884z1
<150> CN 201911405274.8
<151> 2019-12-31
<160> 8
<170> SIPOSequenceListing 1.0
<210> 1
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 1
Ala Tyr Ala Cys Asn Thr Ser Thr Leu
1 5
<210> 2
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 2
Arg Trp Gly Leu Leu Leu Ala Leu Leu
1 5
<210> 3
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 3
Val Tyr Gly Phe Val Arg Ala Cys Leu
1 5
<210> 4
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 4
Ala Tyr Leu Pro Val Asn Glu Ser Phe
1 5
<210> 5
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 5
Ala Ile Tyr Lys Gln Ser Gln His Met
1 5
<210> 6
<211> 9
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 6
Cys Tyr Thr Trp Asn Gln Met Asn Leu
1 5
<210> 7
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 7
Asp Tyr Leu Asn Glu Trp Gly Ser Arg Phe
1 5 10
<210> 8
<211> 10
<212> PRT
<213> Artificial Sequence (Artificial Sequence)
<400> 8
Lys Val Tyr Leu Arg Val Arg Pro Leu Leu
1 5 10

Claims (17)

1. A polypeptide composition that specifically binds to HLA-a24 typing, selected from one or more polypeptides derived from the antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF20A, preferably from any two or more of the antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF20A, p16, p21, more preferably from any two of the polypeptides derived from the antigenic proteins Survivin, ERBB2, TERT, eEF2, p53, WT1-235, CDH3, KIF20A, p16, p 21; the mass ratio of polypeptides derived from each antigen protein is preferably 1-5: 1, preferably 1-3: 1, and more preferably 1: 1.
2. The polypeptide composition of claim 1, wherein the polypeptide derived from each antigenic protein comprises (1) an epitope of each antigenic protein, or an epitope having the same activity as the epitope in (1) obtained by amino acid substitution, addition or deletion, preferably has 75% or more homology with the amino acid sequence of the epitope in (1), more preferably 85% or more homology with the amino acid sequence of the epitope in (1), and still more preferably 95% or more homology with the amino acid sequence of the epitope in (1).
3. The polypeptide composition of claim 1, wherein the polypeptide derived from the antigenic protein Survivin comprises the amino acid sequence shown in SEQ ID No. 1 or an amino acid sequence thereof obtained by amino acid substitution, addition or deletion and having the same activity, preferably has more than 75% homology with the amino acid sequence shown in SEQ ID No. 1, more preferably has more than 85% homology with the amino acid sequence shown in SEQ ID No. 1, and more preferably has more than 95% homology with the amino acid sequence shown in SEQ ID No. 1;
the polypeptide derived from the antigen protein ERBB2 contains an amino acid sequence shown in SEQ ID NO. 2 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75% of homology with the amino acid sequence shown in SEQ ID NO. 2, more preferably has more than 85% of homology with the amino acid sequence shown in SEQ ID NO. 2, and more preferably has more than 95% of homology with the amino acid sequence shown in SEQ ID NO. 2;
the polypeptide derived from the antigen protein TERT contains an amino acid sequence shown in SEQ ID NO. 3 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 3, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 3, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 3;
the polypeptide derived from the antigen protein eEF2 contains an amino acid sequence shown by SEQ ID NO. 4 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown by SEQ ID NO. 4, more preferably has more than 85 percent of homology with the amino acid sequence shown by SEQ ID NO. 4, and more preferably has more than 95 percent of homology with the amino acid sequence shown by SEQ ID NO. 4;
the polypeptide derived from the antigen protein p53 contains an amino acid sequence shown in SEQ ID NO. 5 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown in SEQ ID NO. 5, more preferably has more than 85 percent of homology with the amino acid sequence shown in SEQ ID NO. 5, and more preferably has more than 95 percent of homology with the amino acid sequence shown in SEQ ID NO. 5;
the polypeptide derived from the antigen protein WT1-235 contains an amino acid sequence shown in SEQ ID NO. 6 or an amino acid sequence with the same activity obtained by amino acid substitution, addition or deletion, preferably has more than 75% homology with the amino acid sequence shown in SEQ ID NO. 6, more preferably has more than 85% homology with the amino acid sequence shown in SEQ ID NO. 6, and more preferably has more than 95% homology with the amino acid sequence shown in SEQ ID NO. 6;
the polypeptide derived from the antigen protein CDH3 contains an amino acid sequence shown by SEQ ID NO. 7 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75 percent of homology with the amino acid sequence shown by SEQ ID NO. 7, more preferably has more than 85 percent of homology with the amino acid sequence shown by SEQ ID NO. 7, and more preferably has more than 95 percent of homology with the amino acid sequence shown by SEQ ID NO. 7;
the polypeptide derived from the antigen protein KIF20A contains an amino acid sequence shown in SEQ ID NO. 8 or an amino acid sequence which is obtained by amino acid substitution, addition or deletion and has the same activity, preferably has more than 75% of homology with the amino acid sequence shown in SEQ ID NO. 8, more preferably has more than 85% of homology with the amino acid sequence shown in SEQ ID NO. 8, and more preferably has more than 95% of homology with the amino acid sequence shown in SEQ ID NO. 8.
4. A cell loaded with the polypeptide composition of any one of claims 1-3.
5. The cell according to claim 4, wherein the cell is an antigen presenting cell, including professional antigen presenting cells or non-professional antigen presenting cells, professional antigen presenting cells including lymphocytes, dendritic cell DCs, macrophages, endothelial cells or stem cells, preferably dendritic cells, macrophages or lymphocytes, more preferably dendritic cells; non-professional antigen presenting cells are antigen presenting cells that express HLA class I.
6. A method for producing the cell according to claim 4 or 5, characterized by comprising the steps of: contacting the polypeptide composition of any one of claims 1 to 3 with antigen presenting cells, and loading the polypeptide composition on the antigen presenting cells to obtain the polypeptide composition-loaded antigen presenting cells.
7. The method according to claim 6, wherein the contacting is a co-incubation in which the medium used is any one or more of AIM-V, DMEM or RPMI-1640, preferably AIM-V medium, more preferably AIM-V medium without any serum; the co-incubation time is 1-2 days, preferably 2 days; the temperature for co-incubation is room temperature to 37 ℃; the concentration of each polypeptide in the polypeptide composition according to any of claims 1-3 during co-incubation is 10-100 μ g/mL, preferably 20-80 μ g/mL, more preferably 40 μ g/mL.
8. The method of claim 6, further comprising contacting the antigen presenting cells loaded with the polypeptide composition with a maturation-promoting factor to induce maturation of the antigen presenting cells, thereby obtaining mature antigen presenting cells loaded with the polypeptide composition.
9. The method according to claim 8, wherein the contacting is a co-incubation in which the medium used is any one or more of AIM-V, DMEM or RPMI-1640, preferably an AIM-V medium, more preferably an AIM-V medium without any serum; the co-incubation time is 8-72 hours, preferably 24-48 hours, more preferably 24 hours; the temperature for co-incubation is room temperature to 37 ℃; the maturation promoting factor is one or more of TNF-alpha, IL-1 beta, IL-6, PGE2, IFN-gamma, poly (I: C), R848 or ATP, preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, and the working concentration of TNF-alpha is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably TNF-alpha, IL-1 beta, IL-6 and PGE2, with a working concentration of TNF-alpha of 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-1 beta is 5-50ng/mL, preferably 10-30 ng/mL; the working concentration of IL-6 is 800-1500U/mL, preferably 800-1200U/mL; the working concentration of PGE2 is 0.5-3 mug/mL, preferably 0.5-1.5 mug/mL; or preferably IFN-gamma, poly (I: C) and R848, or preferably IFN-gamma, poly (I: C), R848 and ATP, the working concentration of IFN-gamma is 10-1000IU/mL, preferably 100-300IU/mL, more preferably 100 IU/mL; the working concentration of poly (I: C) is 1-200. mu.g/mL, preferably 20-40. mu.g/mL, more preferably 30. mu.g/mL; the working concentration of R848 is 0.1-50. mu.g/mL, preferably 1-10. mu.g/mL, more preferably 5. mu.g/mL; the working concentration of ATP is 0.1-10mM, preferably 0.1-5mM, more preferably 1 mM.
10. An activated immune cell obtained by contacting the cell of claim 4 or 5 with an inactivated immune effector cell.
11. The immune cell of claim 10, wherein the contacting is a co-incubation for a period of 2-48 hours, preferably 24-48 hours, more preferably 24 hours; the co-incubation medium is AIM-V, DMEM or RPMI1640, preferably AIM-V medium, more preferably AIM-V medium containing 2% V/vFBS; the ratio of the number of the cells of claim 4 or 5 to the number of the immune effector cells is 1: 10-50, preferably 1: 10-30, more preferably 1: 10.
12. The immune cell of claim 11, wherein the AIM-V medium further comprises IL-2, and wherein the working concentration of IL-2 is 10-100U/mL, preferably 100U/mL.
13. Use of the polypeptide composition of any one of claims 1-3, and/or the cell of claim 4 or 5, and/or the immune cell of any one of claims 10-12 for the preparation of a medicament, or a reagent, or a kit for detecting, and/or preventing, and/or treating a tumor, including but not limited to biologicals.
14. A drug, a reagent, or a kit for detecting, and/or preventing, and/or treating a tumor, comprising the polypeptide composition according to any one of claims 1 to 3, and/or the cell according to claim 4 or 5, and/or the immune cell according to any one of claims 10 to 12.
15. The medicament, or the agent, or the kit according to claim 14, further comprising an adjuvant, and/or an immunomodulator, and/or a carrier, and/or an excipient.
16. The medicament of claim 14, wherein the tumor includes, but is not limited to, lung cancer, non-small cell lung cancer, ovarian cancer, colon cancer, rectal cancer, melanoma, kidney cancer, bladder cancer, breast cancer, liver cancer, lymphoma, hematologic malignancies, head and neck cancer, glioma, mesothelioma, colorectal cancer, stomach cancer, nasopharyngeal cancer, laryngeal cancer, cervical cancer, uterine body tumor and osteosarcoma, bone cancer, pancreatic cancer, renal cell cancer, skin cancer, prostate cancer, cutaneous or intraocular malignant melanoma, uterine cancer, cancer of the anal region, testicular cancer, carcinoma of the fallopian tubes, endometrial cancer, vaginal cancer, vulvar cancer, hodgkin's disease, non-hodgkin's lymphoma, esophageal cancer, small intestine cancer, cancer of the endocrine system, cancer of the bile ducts, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, cancer of the urethra, urothelial cancer, penile cancer, chronic or acute leukemia, cervical cancer, A solid tumor of a child, a lymphocytic lymphoma, a renal or ureteral cancer, a renal pelvis cancer, a tumor of the central nervous system, a primary CNS lymphoma, a tumor angiogenesis, a spinal tumor, a brain stem glioma, a pituitary adenoma, a kaposi's sarcoma, a hodgkin's lymphoma, an epidermoid carcinoma, a squamous cell carcinoma, a T-cell lymphoma, or an environmentally induced cancer.
17. A method of eliciting an immune response in vivo by administering an effective amount of the polypeptide composition of any one of claims 1-3, and/or the cell of claim 4 or 5, and/or the immune cell of any one of claims 10-12 to the body.
CN202010940084.2A 2019-12-31 2020-09-09 Polypeptide composition specifically binding with HLA-A24 typing and application Pending CN113117055A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201911405274 2019-12-31
CN2019114052748 2019-12-31

Publications (1)

Publication Number Publication Date
CN113117055A true CN113117055A (en) 2021-07-16

Family

ID=76772020

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202010940084.2A Pending CN113117055A (en) 2019-12-31 2020-09-09 Polypeptide composition specifically binding with HLA-A24 typing and application

Country Status (1)

Country Link
CN (1) CN113117055A (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115785208A (en) * 2022-06-10 2023-03-14 河北博海生物工程开发有限公司 Lung cancer specific molecular target 01 and application thereof
CN115785207A (en) * 2022-06-10 2023-03-14 河北博海生物工程开发有限公司 Lung cancer specific molecular target 02 and application thereof

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115785208A (en) * 2022-06-10 2023-03-14 河北博海生物工程开发有限公司 Lung cancer specific molecular target 01 and application thereof
CN115785207A (en) * 2022-06-10 2023-03-14 河北博海生物工程开发有限公司 Lung cancer specific molecular target 02 and application thereof

Similar Documents

Publication Publication Date Title
JP6435286B2 (en) Cancer vaccine composition
Salgaller et al. Dendritic cell-based immunotherapy of prostate cancer
RU2464275C2 (en) Peptide cancer vaccines expressing tumour-specific antigens
EP2547691B1 (en) Composition of tumor-associated peptides and related anti-cancer vaccine for the treatment of gastric cancer and other cancers
KR101284237B1 (en) PROTEINS BELONGING TO THE Bcl-2 FAMILY AND FRAGMENTS THEREOF, AND THEIR USE IN CANCER PATIENTS
CN106220721B (en) PRAME derived peptides and immunogenic compositions comprising the same
Toes et al. Protective antitumor immunity induced by immunization with completely allogeneic tumor cells
CN115558030A (en) Novel antigens and methods of use thereof
US8735357B2 (en) Method of inducing antigen-specific T cells
JP5087925B2 (en) Novel cancer antigen peptides and uses thereof
CN112584852A (en) Novel antigens and uses thereof
CN113117055A (en) Polypeptide composition specifically binding with HLA-A24 typing and application
CN117083081A (en) Tissue specific antigens for cancer immunotherapy
US20050048646A1 (en) Method for inducing cytotoxic T lymphocyte
CN111848733B (en) Polypeptide composition and vaccine
CN111848732B (en) Polypeptide composition and vaccine
CN113045635A (en) Polypeptide composition specifically binding with HLA-A2 typing and application
CN112402596B (en) Polypeptide composition and vaccine
US20220313803A1 (en) Novel neoantigens and cancer immunotherapy using same
Soler et al. Triiodothyronine-stimulated dendritic cell vaccination boosts antitumor immunity against murine colon cancer
CN111518216B (en) Polypeptide, composition containing polypeptide and application of composition in tumor immunity
JP7181676B2 (en) Immunity inducer
JP2005245430A (en) Method for inducing ctl
Koide et al. Efficient CTL productivity of modified fusion cells by increase of heat shock protein 70
US20230381291A1 (en) Tumor-associated peptides and uses thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
CB02 Change of applicant information

Country or region after: China

Address after: 201805 No. 1585 Garden National Road, Anting Town, Jiading District, Shanghai

Applicant after: Shanghai Cell Therapy Group Co.,Ltd.

Address before: 200000 building 6, no.1585 Yuanguo Road, Anting Town, Jiading District, Shanghai

Applicant before: SHANGHAI CELL THERAPY GROUP Co.,Ltd.

Country or region before: China

CB02 Change of applicant information