WO2022166947A1 - 肿瘤浸润淋巴细胞的制备方法及其用途 - Google Patents

肿瘤浸润淋巴细胞的制备方法及其用途 Download PDF

Info

Publication number
WO2022166947A1
WO2022166947A1 PCT/CN2022/075341 CN2022075341W WO2022166947A1 WO 2022166947 A1 WO2022166947 A1 WO 2022166947A1 CN 2022075341 W CN2022075341 W CN 2022075341W WO 2022166947 A1 WO2022166947 A1 WO 2022166947A1
Authority
WO
WIPO (PCT)
Prior art keywords
til
cell
present application
cells
immune checkpoint
Prior art date
Application number
PCT/CN2022/075341
Other languages
English (en)
French (fr)
Inventor
刘雅容
赵佩佩
Original Assignee
苏州沙砾生物科技有限公司
上海沙砾生物科技有限公司
珠海拓域生物科技有限公司
珠海沙砾生物科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 苏州沙砾生物科技有限公司, 上海沙砾生物科技有限公司, 珠海拓域生物科技有限公司, 珠海沙砾生物科技有限公司 filed Critical 苏州沙砾生物科技有限公司
Priority to CN202280003156.8A priority Critical patent/CN115315509B/zh
Publication of WO2022166947A1 publication Critical patent/WO2022166947A1/zh

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration

Definitions

  • the present application relates to the field of biomedicine, in particular to a method for preparing tumor-infiltrating lymphocytes and uses thereof.
  • Tumor therapy using adoptively autologously transferred tumor-infiltrating lymphocytes is an effective approach to treat patients with poor prognosis.
  • adoptive autologous transfer of tumor-infiltrating lymphocytes to treat tumors requires a large number of tumor-infiltrating lymphocytes. Therefore, how to provide a robust and reliable method for culturing tumor-infiltrating lymphocytes is an urgent problem to be solved.
  • the present application provides a method for culturing tumor-infiltrating lymphocytes, the culturing method has one or more effects selected from the group consisting of: improving the number of TIL cells, improving the secretory ability of TIL cells, making TIL cells
  • the killing ability increases, the proportion of NK cells increases, the proportion of TIL cells is changed, the proportion of CD4 + cells increases, the proportion of CD8 + cells decreases, the proportion of central memory T cells increases, and the proportion of regulatory T cells decreases, Increase the proportion of activated T cells, increase the proportion of tumor-specific T cells, and increase the proportion of stem-like T cells.
  • the present application provides a method for culturing tumor-infiltrating lymphocytes (TILs), comprising: subjecting TILs derived from tumor tissue and not expanded in vitro through at least one stage of in vitro expansion, and in at least one stage of the in vitro expansion In addition, the TIL is contacted with one or more T cell activators and one or more immune checkpoint inhibitors.
  • TILs tumor-infiltrating lymphocytes
  • the TIL derived from tumor tissue and not expanded in vitro is subjected to a first stage of in vitro expansion, a second stage of in vitro expansion and a third stage of in vitro expansion, and in the third stage of in vitro expansion
  • the TILs expanded in the second stage in vitro are contacted with the one or more T cell activators and the immune checkpoint inhibitor.
  • the TIL is combined with the one or more T cell activators and the one or more immune checkpoint inhibitors substantially simultaneously in a single stage of the in vitro expansion touch.
  • the third stage in vitro expansion is performed for up to about 24 hours.
  • the third stage in vitro expansion is performed for about 12 hours to about 24 hours.
  • in at least one in vitro expansion phase compared to a corresponding TIL that has not been contacted with said T cell activator and/or said immune checkpoint inhibitor during the in vitro expansion phase TILs to which the one or more T cell activators and the one or more immune checkpoint inhibitors have been exposed show improved expansion.
  • the improved expansion effect comprises one or more selected from the group consisting of an increased number of TIL cells, an improved proportion of T cell subsets, an increased cytokine secretion capacity, and an increased tumor cell killing ability.
  • the improved proportion of T cell subsets comprises one or more selected from the group consisting of: increased proportion of central memory T cells, decreased proportion of regulatory T cells, increased activated T cells ratio, increased proportion of tumor-specific T cells, and increased proportion of stem-like T cells.
  • the method further comprises contacting the TIL with the one or more T cell activators during at least one stage of the in vitro expansion.
  • the one or more T cell activators comprise one or more selected from the group consisting of CD80, CD86, B7-H3, 4-1BBL, CD27, CD30, CD134, B7h, CD40, LIGHT, and their functionally active fragments.
  • the one or more T cell activators comprise agonists of one or more targets selected from the group consisting of CD3, CD28, HVEM, CD40L, OX40, and 4-1BB.
  • the one or more T cell activators comprise a CD3 agonist and/or a CD28 agonist.
  • the one or more T cell activators comprise a CD3 agonist.
  • the one or more T cell activators comprise anti-CD3 antibodies and/or antigen-binding fragments thereof.
  • the one or more T cell activators comprise a CD28 agonist.
  • the one or more T cell activators comprise an anti-CD28 antibody and/or antigen-binding fragment thereof.
  • the contacting of the TIL with the one or more T cell activators comprises one or more means selected from the group consisting of: (1) contacting the one or more T cells with adding an activator to the cell culture medium of the TIL; (2) adding engineered cells expressing the one or more T cell activators to the cell culture medium of the TIL; (3) adding the A solid phase medium of the one or more T cell activators is added to the cell culture medium of the TIL.
  • the initial concentration of each of the T cell activators in the cell culture medium of the TIL is each independently at least about 30 ng/mL.
  • the initial concentration of each of the T cell activators in the cell culture medium of the TIL is each independently about 30 ng/mL to about 300 ng/mL.
  • the solid phase medium has a diameter of about 500 nanometers to about 10 micrometers.
  • the solid phase medium has a diameter of from about 1 nanometer to about 500 nanometers.
  • the diameter of the solid medium is measured by transmission electron microscopy.
  • the solid phase medium comprises a polymer
  • the amount of each of the T cell activators contained in the solid medium is each independently at least about 25 ⁇ g per mg of the solid medium.
  • the solid phase medium comprising the one or more T cell activators is added to the in the cell culture medium of the TIL.
  • the solid phase medium comprising the one or more T cell activators is added to the solid phase medium in a ratio of about 1:100 to about 1:2000 of the solid phase medium to the TIL in the cell culture medium of the TIL.
  • the immune checkpoint inhibitor comprises a substance that inhibits the interaction of PD-1 with PD-L1 and/or PD-L2.
  • the immune checkpoint inhibitor comprises a PD-1 inhibitor.
  • the immune checkpoint inhibitor comprises an antibody to PD-1 and/or an antigen-binding fragment thereof.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to bind PD -1 with a KD value of about 100 pM or less.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to bind PD-1 with an EC50 value of about 100 pM or less.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to inhibit the binding of PD-1 to PD-L1 and/or PD-L2 with an IC50 value of about 1 nM or less.
  • the immune checkpoint inhibitor comprises HCDR3, and the HCDR3 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 3 and 17.
  • the immune checkpoint inhibitor comprises HCDR2, and the HCDR2 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 2 and 16.
  • the immune checkpoint inhibitor comprises HCDR1, and the HCDR1 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 1 and 15.
  • the immune checkpoint inhibitor comprises LCDR3, and the LCDR3 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 6 and 20.
  • the immune checkpoint inhibitor comprises LCDR2, and the LCDR2 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 5 and 19.
  • the immune checkpoint inhibitor comprises LCDR1, and the LCDR1 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 4 and 18.
  • the immune checkpoint inhibitor comprises VH, and the VH comprises the amino acid sequence set forth in any one of SEQ ID NOs: 7, 11, and 21.
  • the immune checkpoint inhibitor comprises VL, and the VL comprises the amino acid sequence set forth in any one of SEQ ID NOs: 8, 12, and 22.
  • the immune checkpoint inhibitor is selected from the group consisting of (1) an antibody or antigen-binding fragment thereof comprising the following heavy and light chains, the heavy chain comprising an amino acid sequence such as SEQ ID NO: HCDR1 shown in any one of 1 and 15, HCDR2 with amino acid sequence shown in any one of SEQ ID NOs: 2 and 16, and HCDR3 with amino acid sequence shown in any one of SEQ ID NOs: 3 and 17 , and the light chain comprises an LCDR1 whose amino acid sequence is shown in any one of SEQ ID NOs: 4 and 18, an LCDR2 whose amino acid sequence is shown in any one of SEQ ID NOs: 5 and 19, and an amino acid sequence such as SEQ ID NO: 5 and 19.
  • an antibody or antigen-binding fragment thereof comprising the following heavy and light chains
  • the heavy chain comprising an amino acid sequence such as SEQ ID NO: HCDR1 shown in any one of 1 and 15, HCDR2 with amino acid sequence shown in any one of SEQ ID NOs: 2 and 16, and HCDR3 with
  • LCDR3 set forth in any one of ID NOs: 6 and 20; (2) an antibody or antigen-binding fragment thereof comprising a heavy chain and a light chain comprising amino acid sequences such as SEQ ID NOs: 7, 11 and 21, and the light chain comprises a VL whose amino acid sequence is shown in any one of SEQ ID NOs: 8, 12, and 22.
  • the antibody is selected from the group consisting of chimeric antibodies, humanized antibodies and fully human antibodies.
  • the antigen binding fragment is selected from the group consisting of: Fab, Fab', Fv fragments, F(ab') 2 , F(ab) 2 , scFv, di-scFv, VHH and dAb.
  • the initial concentration of each of the immune checkpoint inhibitors in the cell culture medium of the TIL is each independently at least about 0.1 ⁇ g/mL.
  • the initial concentration of each of the immune checkpoint inhibitors in the cell culture medium of the TIL is each independently about 0.1 ⁇ g/mL to about 20 ⁇ g/mL.
  • it further comprises: in at least one stage of said in vitro expansion, contacting said TIL with one or more T cell growth factors.
  • the TIL is contacted with the one or more T cell activators and with the one or more T cell growth factors in a single stage of the in vitro expansion.
  • the TIL is contacted with the one or more T cell activators and the one or more T cell growth factors substantially simultaneously in a single stage of the in vitro expansion .
  • the one or more T cell growth factors are selected from one or more of the following group: IL-2, IL-7, IL-12, IL-15, IL-21, gamma Interferons, and their functionally active fragments.
  • the one or more T cell growth factors comprise IL-2 and/or functionally active fragments thereof.
  • contacting the TIL with the one or more T cell growth factors comprises adding the T cell growth factors to the cell culture medium of the TIL.
  • the initial concentration of each of the T cell growth factors in the cell culture medium of the TIL is each independently at least about 300 IU/mL.
  • it further comprises: co-culturing the TIL with feeder cells in at least one stage of the in vitro expansion.
  • the TIL is contacted with the one or more T cell activators and/or the one or more T cell growth factors in a single stage of the in vitro expansion and Co-cultured with the feeder cells.
  • the TIL is contacted with the one or more T cell activators and/or the one or more T cell growth factors in a single stage of the in vitro expansion After a period of time, it is then co-cultured with the feeder cells.
  • the certain period of time is at least about 2 hours.
  • the certain period of time is from about 6 hours to about 72 hours.
  • the certain period of time is from about 12 hours to about 48 hours.
  • the certain period of time is about 6 hours, about 12 hours, about 24 hours, about 48 hours, or about 72 hours.
  • the feeder cells comprise antigen presenting cells.
  • the feeder cells comprise one or more selected from the group consisting of peripheral mononuclear cells, dendritic cells, and artificial antigen presenting cells.
  • the feeder cells are peripheral mononuclear cells.
  • the feeder cells are irradiated feeder cells.
  • co-culturing the TIL with the feeder cell comprises contacting the surface of the feeder cell with the surface of the TIL.
  • the co-cultivation of the TIL with the feeder cells comprises adding the feeder cells to the cell culture medium of the TIL.
  • the feeder cells are added to the cell culture medium of the TIL at a ratio of the feeder cells to the TIL from about 40:1 to about 400:1.
  • the TIL derived from tumor tissue and not expanded in vitro is TIL derived from fragments of the tumor tissue.
  • the fragments have a volume of about 1 cubic millimeter to about 27 cubic millimeters.
  • TILs tumor-infiltrating lymphocytes
  • step (A) contacting a first TIL population derived from tumor tissue and not expanded in vitro with one or more T cell growth factors; obtaining a second TIL population through the step (A);
  • step (B) contacting said second TIL population with said one or more T cell growth factors and/or one or more T cell activators; obtaining a third TIL population through said step (B);
  • step (C) the TIL is contacted with the one or more T cell activators and the one or more immune checkpoint inhibitors substantially simultaneously.
  • the step (C) is carried out for up to about 24 hours.
  • the step (C) is carried out for about 12 hours to about 24 hours.
  • step (C) with said one compared to a corresponding TIL that has not been contacted with said T cell activator and/or said immune checkpoint inhibitor in step (C) TILs to which the one or more T cell activators and the one or more immune checkpoint inhibitors have been exposed show improved expansion.
  • the improved expansion effect comprises one or more selected from the group consisting of an increased number of TIL cells, an improved proportion of T cell subsets, an increased cytokine secretion capacity, and an increased tumor cell killing ability.
  • the improved proportion of T cell subsets comprises one or more selected from the group consisting of: increased proportion of central memory T cells, decreased proportion of regulatory T cells, increased activated T cells ratio, increased proportion of tumor-specific T cells, and increased proportion of stem-like T cells.
  • the one or more T cell activators comprise one or more selected from the group consisting of CD80, CD86, B7-H3, 4-1BBL, CD27, CD30, CD134, B7h, CD40, LIGHT, and their functionally active fragments.
  • the one or more T cell activators comprise agonists of one or more targets selected from the group consisting of CD3, CD28, HVEM, CD40L, OX40, and 4-1BB.
  • the one or more T cell activators comprise a CD3 agonist and/or a CD28 agonist.
  • the one or more T cell activators comprise a CD3 agonist.
  • the one or more T cell activators comprise anti-CD3 antibodies and/or antigen-binding fragments thereof.
  • the one or more T cell activators comprise a CD28 agonist.
  • the one or more T cell activators comprise an anti-CD28 antibody and/or antigen-binding fragment thereof.
  • the contacting of the TIL with the one or more T cell activators comprises one or more means selected from the group consisting of: (1) contacting the one or more T cells with adding an activator to the cell culture medium of the TIL; (2) adding engineered cells expressing the one or more T cell activators to the cell culture medium of the TIL; (3) adding the A solid phase medium of the one or more T cell activators is added to the cell culture medium of the TIL.
  • the initial concentration of each of the T cell activators in the cell culture medium of the TIL is each independently at least about 30 ng/mL.
  • the initial concentration of each of the T cell activators in the cell culture medium of the TIL is each independently about 30 ng/mL to about 300 ng/mL.
  • the solid phase medium has a diameter of about 500 nanometers to about 10 micrometers.
  • the solid phase medium has a diameter of from about 1 nanometer to about 500 nanometers.
  • the diameter of the solid medium is measured by transmission electron microscopy.
  • the solid phase medium comprises a polymer
  • the amount of each of the T cell activators contained in the solid medium is each independently at least about 25 ⁇ g per mg of the solid medium.
  • the solid phase medium comprising the one or more T cell activators is added to the in the cell culture medium of the TIL.
  • the solid phase medium comprising the one or more T cell activators is added to the solid phase medium in a ratio of about 1:100 to about 1:2000 of the solid phase medium to the TIL in the cell culture medium of the TIL.
  • the immune checkpoint inhibitor comprises a substance that inhibits the interaction of PD-1 with PD-L1 and/or PD-L2.
  • the immune checkpoint inhibitor comprises a PD-1 inhibitor.
  • the immune checkpoint inhibitor comprises an antibody to PD-1 and/or an antigen-binding fragment thereof.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to bind PD -1 with a KD value of about 100 pM or less.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to bind PD-1 with an EC50 value of about 100 pM or less.
  • the immune checkpoint inhibitor comprises an immune checkpoint inhibitor having the ability to inhibit the binding of PD-1 to PD-L1 and/or PD-L2 with an IC50 value of about 1 nM or less.
  • the immune checkpoint inhibitor comprises HCDR3, and the HCDR3 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 3 and 17.
  • the immune checkpoint inhibitor comprises HCDR2, and the HCDR2 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 2 and 16.
  • the immune checkpoint inhibitor comprises HCDR1, and the HCDR1 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 1 and 15.
  • the immune checkpoint inhibitor comprises LCDR3, and the LCDR3 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 6 and 20.
  • the immune checkpoint inhibitor comprises LCDR2, and the LCDR2 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 5 and 19.
  • the immune checkpoint inhibitor comprises LCDR1, and the LCDR1 comprises the amino acid sequence set forth in any one of SEQ ID NOs: 4 and 18.
  • the immune checkpoint inhibitor comprises VH, and the VH comprises the amino acid sequence set forth in any one of SEQ ID NOs: 7, 11, and 21.
  • the immune checkpoint inhibitor comprises VL, and the VL comprises the amino acid sequence set forth in any one of SEQ ID NOs: 8, 12, and 22.
  • the immune checkpoint inhibitor is selected from the group consisting of (1) an antibody or antigen-binding fragment thereof comprising the following heavy and light chains, the heavy chain comprising an amino acid sequence such as SEQ ID NO: HCDR1 shown in any one of 1 and 15, HCDR2 with amino acid sequence shown in any one of SEQ ID NOs: 2 and 16, and HCDR3 with amino acid sequence shown in any one of SEQ ID NOs: 3 and 17 , and the light chain comprises an LCDR1 whose amino acid sequence is shown in any one of SEQ ID NOs: 4 and 18, an LCDR2 whose amino acid sequence is shown in any one of SEQ ID NOs: 5 and 19, and an amino acid sequence such as SEQ ID NO: 5 and 19.
  • an antibody or antigen-binding fragment thereof comprising the following heavy and light chains
  • the heavy chain comprising an amino acid sequence such as SEQ ID NO: HCDR1 shown in any one of 1 and 15, HCDR2 with amino acid sequence shown in any one of SEQ ID NOs: 2 and 16, and HCDR3 with
  • LCDR3 set forth in any one of ID NOs: 6 and 20; (2) an antibody or antigen-binding fragment thereof comprising a heavy chain and a light chain comprising amino acid sequences such as SEQ ID NOs: 7, 11 and 21, and the light chain comprises a VL whose amino acid sequence is shown in any one of SEQ ID NOs: 8, 12, and 22.
  • the antibody is selected from the group consisting of chimeric antibodies, humanized antibodies and fully human antibodies.
  • the antigen binding fragment is selected from the group consisting of: Fab, Fab', Fv fragments, F(ab') 2 , F(ab) 2 , scFv, di-scFv, VHH and dAb.
  • the initial concentration of each of the immune checkpoint inhibitors in the cell culture medium of the TIL is each independently at least about 0.1 ⁇ g/mL.
  • the initial concentration of each of the immune checkpoint inhibitors in the cell culture medium of the TIL is each independently about 0.1 ⁇ g/mL to about 20 ⁇ g/mL.
  • step (B) the TIL is contacted with the one or more T cell activators and the one or more T cell growth factors substantially simultaneously.
  • the one or more T cell growth factors are selected from one or more of the following group: IL-2, IL-7, IL-12, IL-15, IL-21, gamma Interferons, and their functionally active fragments.
  • the one or more T cell growth factors comprise IL-2 and/or functionally active fragments thereof.
  • contacting the TIL with the one or more T cell growth factors comprises adding the T cell growth factors to the cell culture medium of the TIL.
  • the initial concentration of each of the T cell growth factors in the cell culture medium of the TIL is each independently at least about 300 IU/mL.
  • it further comprises: in step (A), step (B) and/or step (C), co-culturing the TIL with feeder cells.
  • step (B) the TIL is contacted with the one or more T cell activators and/or the one or more T cell growth factors and with the feeder Cell co-culture.
  • step (B) after contacting the TIL with the one or more T cell activators and/or the one or more T cell growth factors for a certain period of time, Co-cultured with the feeder cells.
  • the certain period of time is at least about 2 hours.
  • the certain period of time is from about 6 hours to about 72 hours.
  • the certain period of time is from about 12 hours to about 48 hours.
  • the certain period of time is about 6 hours, about 12 hours, about 24 hours, about 48 hours, or about 72 hours.
  • the feeder cells comprise antigen presenting cells.
  • the feeder cells comprise one or more selected from the group consisting of peripheral mononuclear cells, dendritic cells, and artificial antigen presenting cells.
  • the feeder cells are peripheral mononuclear cells.
  • the feeder cells are irradiated feeder cells.
  • co-culturing the TIL with the feeder cell comprises contacting the surface of the feeder cell with the surface of the TIL.
  • the co-cultivation of the TIL with the feeder cells comprises adding the feeder cells to the cell culture medium of the TIL.
  • the feeder cells are added to the cell culture medium of the TIL at a ratio of the feeder cells to the TIL from about 40:1 to about 400:1.
  • the TIL derived from tumor tissue and not expanded in vitro is TIL derived from fragments of the tumor tissue.
  • the fragments have a volume of about 1 cubic millimeter to about 27 cubic millimeters.
  • the present application also provides a tumor-infiltrating lymphocyte (TIL) obtained by the method of the present application.
  • TIL tumor-infiltrating lymphocyte
  • the application also provides a composition comprising the TIL of the application.
  • the application also provides a pharmaceutical composition comprising the TIL of the application and/or the composition of the application, and optionally a pharmaceutically acceptable carrier.
  • the present application also provides a method of affecting tumor cell growth, comprising administering to a subject a TIL of the present application, a composition of the present application, and/or a pharmaceutical composition of the present application.
  • the present application also provides the use of the TIL of the present application, the composition of the present application and/or the pharmaceutical composition of the present application in preparing a medicament for preventing and/or treating tumors.
  • the tumor is a solid tumor.
  • the tumor is one or more selected from the group consisting of melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, stomach cancer, colorectal cancer cancer, and kidney cancer.
  • the present application also provides the TIL of the present application, the composition of the present application and/or the pharmaceutical composition of the present application, which are used for preventing and/or treating tumors.
  • the tumor is a solid tumor.
  • the tumor is one or more selected from the group consisting of melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, stomach cancer, colorectal cancer cancer, and kidney cancer.
  • the present application also provides a method of preventing and/or treating tumors, comprising administering the TIL of the present application, the composition of the present application and/or the pharmaceutical composition of the present application to a subject.
  • the tumor is a solid tumor.
  • the tumor is one or more selected from the group consisting of melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, stomach cancer, colorectal cancer cancer, and kidney cancer.
  • Figures 1A-1B show the intracellular factor expression ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • Figures 2A-2B show the intracellular factor expression ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • Figures 3A-3C show the detection results of IL-2 secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • FIGS 4A-4C show the detection results of TNF secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • FIGS 5A-5C show the detection results of IFN ⁇ secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • Figures 6A-6B show the detection results of the cell killing ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • Figures 7A-7C show, for different donors, the proliferation of TIL cells with the addition of CD3 antibody in the second phase of in vitro expansion (REP phase) when the CD3 antibody was added, compared to the TIL cells that were only added with the CD3 antibody.
  • Figures 8A-8B show, for different donors, the proliferation of TIL cells with the addition of CD3 antibody in the second stage of in vitro expansion (REP stage) when the CD3 antibody was added, compared to the TIL cells that were only added with the CD3 antibody.
  • Figures 9A-9B show the cell viability and cell typing of TILs with CD3 antibody added in the second stage of in vitro expansion (REP stage) with the addition of CD3 antibody, compared to TILs with only CD3 antibody.
  • Figures 10A-10B show the proportion of activation-related (41BB + ) cells in TILs with CD3 antibody added when CD3 antibody was added in the second stage of in vitro expansion (REP stage).
  • Figures 11A-11C show that the addition of PD-1 antibody when CD3 antibody is added during the second phase of in vitro expansion (REP phase) correlates with activation (CD25 + and/or CD27 + ) in TIL compared to CD3 antibody only cell ratio.
  • Figures 12A-12B show the proportion of depletion-related (TIM3 + ) cells in TILs with CD3 antibody added when CD3 antibody was added in the second stage of in vitro expansion (REP stage).
  • FIGS 13A-13B show the proportion of TNF- ⁇ secreting cells in TILs with only CD3 antibody added when the CD3 antibody was added in the second stage of in vitro expansion (REP stage).
  • Figures 14A-14D show the proportion of IFN- ⁇ -secreting cells in TILs with CD3 antibody added in the second stage of in vitro expansion (REP stage) when the CD3 antibody was added additionally, compared to the TIL with only CD3 antibody added.
  • Figures 15A-15C show that when CD3 antibody was added in the second phase of in vitro expansion (REP phase), the addition of PD-1 antibody compared to the cytokines (IL-2 and/or IL-2 and/or IL- 4) The amount of secretion.
  • Figures 16A-16C show that when CD3 antibody was added in the second phase of in vitro expansion (REP phase), the addition of PD-1 antibody compared to the cytokines (IL-17 and/or IL-17 and/or IL- 6) The amount of secretion.
  • Figures 17A-17D show that the addition of PD-1 antibody when CD3 antibody was added during the second phase of in vitro expansion (REP phase) compared to cytokines (TNF- ⁇ and/or IFN- ⁇ ) The amount of secretion.
  • Figures 18A-18D show the secretion of cytokines (IL-4 and/or TNF- ⁇ ) of TILs stimulated by adding only transACT after the addition of PD-1 antibody for stimulation at the end of the REP stage .
  • Figures 18E-18F show the secretion of cytokines (IL-2 and/or IL-6) of TILs stimulated by adding only transACT after the addition of PD-1 antibody for stimulation at the end of the REP stage .
  • Figures 19A-19D show the secretion of cytokines (IL-6 and/or TNF- ⁇ ) of TILs stimulated by adding PD-1 antibody only after the REP stage culture .
  • Figures 20A-20C show that after the end of the REP phase of culture, additional PD-1 antibody for stimulation, compared to only transACT stimulation of TIL cytokines (IL-2, IL-4 and/or IFN- ⁇ ) ) secretion volume.
  • TIL cytokines IL-2, IL-4 and/or IFN- ⁇
  • the term "expression” generally refers to the process of transcription and/or translation that occurs within a cell of a gene encoding a polypeptide of interest.
  • the level of transcription of the gene encoding the polypeptide of interest in the host cell can be determined by measuring the amount of the corresponding mRNA present in the cell. For example, quantitative measurement of mRNA transcribed from a gene encoding a polypeptide of interest can be performed by PCR or by RNA hybridization (see Sambrook et al., Molecular Cloning: A Laboratory Manual, Cold Spring Harbor Laboratory Press (1989)).
  • the level of translation of a gene encoding a polypeptide of interest can be measured by a variety of methods, such as by ELISA, by polypeptide biological activity assays, or by Western blotting or radioimmunoassays (see Sambrook et al., supra).
  • stage in the terms “one stage in vitro expansion”, “single stage in vitro expansion”, or “first stage in vitro expansion” etc. generally refers to a period of expansion that TILs undergo in vitro process.
  • each stage can be divided by the change in the number of TIL cells, in one embodiment, when the number of TIL cells increases by at least about 1-fold, it can be considered that TIL cells have entered the next stage.
  • TIL cells when the number of TIL cells increases by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times At least about 50-fold, TIL cells can be considered to have entered the next stage of in vitro expansion. In one embodiment, each stage can also be divided by the conditions of TIL cell culture.
  • TIL cells can be considered to enter the next stage of in vitro expansion when T cell activators and/or T cell growth factors are added or supplemented to the cell culture medium. In one embodiment, after the TIL cells are centrifuged and/or cell washed, the TIL cells can be considered to have entered the next stage of in vitro expansion. In one embodiment, each stage can also be divided by the number of days in which the TIL cells are cultured.
  • TIL cells when TIL cells are cultured in vitro for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 30 days, about 40 days, After about 50 days or about 100 days, the TIL cells can be considered to have entered the next stage of in vitro expansion.
  • first stage in vitro expansion generally refers to the stage of expansion using T cell growth factors after primary TILs are obtained from tissue.
  • the tissue of the present application may be selected from the group consisting of tumor tissue and pleural effusion, and the pleural effusion of the present application may be pleural effusion of a patient with metastatic cancer.
  • the amplification of the present application may be autologous or allogeneic in vivo amplification, or may be in vitro amplification.
  • the first phase of in vitro expansion of the present application may also be referred to as the preREP (pre-rapid expansion) phase.
  • the term "second-stage in vitro expansion” generally refers to a stage in which tissue is removed from a subject and expanded, and then expanded again.
  • the number of TIL cells expanded in vitro in the second stage of the present application is increased, eg, at least about 10-fold (or at least about 20, 30 fold), compared to the TILs expanded in vitro in the first stage. , 40, 50, 60, 70, 80 or 90-fold), or in one embodiment the number of cells can be increased by at least about 100-fold.
  • the culture conditions of the second stage in vitro expansion may be different from that of the first stage in vitro expansion, eg, the added culture substance may be different.
  • the second stage of in vitro expansion of the present application may also be referred to as the REP (rapid expansion) stage.
  • in vivo generally refers to an event that occurs in a subject.
  • in vitro generally refers to events that occur outside the body of a subject.
  • ex vivo generally refers to events involving treatment or surgery on cells, tissues and/or organs that have been removed from a subject.
  • the cells, tissues and/or organs can be returned to the subject's body by surgery or treatment.
  • secretion generally refers to the transfer of an expressed polypeptide or protein by a cell to the extracellular environment.
  • secretory capacity generally refers to the ability of a cell to express a polypeptide or protein and to transfer the polypeptide or protein of the present application to the extracellular environment.
  • irradiation generally refers to the treatment of a substance by means of radiation.
  • irradiating may refer to irradiating a substance with X-rays, alpha rays, beta rays, or gamma rays.
  • engineered cell generally refers to a cell that has been genetically modified by adding additional genetic material in the form of DNA or RNA to the total genetic material of the cell.
  • the engineered cells can be genetically modified to express TILs of the T cell activators and/or T cell growth factors of the present application.
  • co-culture generally refers to the culturing of two or more different populations of cells with some degree of contact between them.
  • Contacting of two or more different populations of cells of the present application may, in one embodiment, be by direct contact, ie, in which cells of one population are in direct physical contact with cells of another population.
  • indirect contact mediated by shared culture medium may be used.
  • the shared medium of the present application may contain metabolites produced and released by at least one population of co-cultured cells and used to culture cells of another population.
  • the term "contacting" generally means that two or more substances of different types are brought into contact together in any order, in any manner, and for any length of time.
  • one or more feeder cells, T cell activators and/or T cell growth factors can be added to the medium of TIL cells by direct contact, eg, one or more feeder cells can be added.
  • the medium of cells, T cell activators and/or T cell growth factors is added to and/or replaced with the medium of TIL cells, for example, one or more feeder cells, T cell activators and/or T cell growth factors may be added to The medium of factors is used for the cultivation of TIL cells; in one embodiment, the metabolites produced and released by the feeder cells can be used for the cultivation of TIL cells by indirect contact, for example.
  • the term "mixture” generally refers to a combination of two or more different substances.
  • the CD28 antibody or antigen-binding fragment thereof of the present application and the CD3 antibody or antigen-binding fragment thereof can be added to the cell culture medium as a mixture after mixing.
  • the terms “concurrently contacting”, “co-contacting”, “concurrently in contact with”, “simultaneously” and “co-contacting” generally refer to the administration of two or more substances to a subject and/or cell such that the substances both in the subject and/or in the context of the cell culture.
  • Simultaneous contacting can include simultaneous administration in different compositions, administration in different compositions at different times, or administration in a composition in which two or more active pharmaceutical ingredients are present.
  • “simultaneously contacting” as used herein may generally refer to substantially simultaneous contacting.
  • the term “expansion” generally refers to a several-fold increase in the number of cells over a period of time.
  • the number of cells can be increased by at least about 3-fold (or 4, 5, 6, 7, 8 or 9-fold), in one embodiment the number of cells can be increased by at least about 10-fold (or 20, 30, 40, 50, 60, 70, 80 or 90-fold), or in one embodiment the number of cells can be increased by at least about 100-fold.
  • the term “expanded” generally means that the cells of the application have been expanded by one or more of the above.
  • polymer generally refers to a molecule consisting of separate chemical moieties linked together, which may be the same or different in this application.
  • the term “polymer” may refer to individual chemical moieties linked tail to tail to form a linear molecule, as well as individual chemical moieties linked together in branched (eg, "multi-armed” or "star") structures .
  • the polymer may include, for example, polysaccharides, dextran, hydrogels, polyethylene glycol, or poloxamers.
  • Poloxamers are nonionic triblock copolymers with a central hydrophobic chain of polyoxypropylene (poly(propylene oxide)) and two pendant hydrophilic chains of polyoxyethylene (poly(ethylene oxide)).
  • the materials encompassed by this application can be formulated with, or administered with, any of the polymers described herein or known in the art.
  • inhibitor is generally meant to include partial or complete blocking, inhibition or neutralization of any molecule, such as a protein, described herein.
  • molecules inhibited by the present application include, but are not limited to, PD-1.
  • Suitable inhibitor molecules may include antagonist antibodies or antibody fragments, fragments or derivatives of small molecules, peptides, antisense oligonucleotides, small organic molecules, and the like.
  • Methods of identifying an inhibitor of the present application include contacting a cell expressing the molecule inhibited by the application with a candidate inhibitor molecule, and detecting a detectable change in one or more biological activities associated with the molecule inhibited by the application.
  • an inhibitor of the present application can be a PD-1 inhibitor, which can block the binding of PD-1 to its ligand.
  • a suitable PD-1 inhibitor can be a PD-1 antibody.
  • immune checkpoint inhibitor generally refers to a molecule that reduces, inhibits, interferes with, or modulates one or more checkpoint proteins, in whole or in part.
  • checkpoint proteins are known to contain, such as PD-1 and its ligands PD-L1 and PD-L2.
  • Immune checkpoint inhibitors can include antibodies or polypeptides derived from antibodies.
  • IC50 value or “IC50 value” generally refers to the concentration of a target that is required to obtain 50% inhibition of a biological process. IC50 values can be converted to absolute inhibition constants (Ki) using the Cheng-Prusoff equation (Biochem. Pharmacol. (1973) 22:3099).
  • EC50 value generally refers to the half-maximal effective concentration of a binding substance (eg, an antibody) in the context of an in vitro or in vivo assay that induces a response of 50% of the response between the baseline value and the maximum value.
  • a reduced EC50 value may indicate higher drug affinity and efficacy.
  • K D value or “KD value” generally refers to the dissociation constant, which can be determined by surface plasmon resonance.
  • surface plasmon resonance analysis uses the BIAcore system (Pharmacia Biosensor, Piscataway, NJ) to measure ligands (substances immobilized on the biosensor matrix) and analytes (substances in solution) by surface plasmon resonance (SPR) ) in real-time binding interactions.
  • SPR surface plasmon resonance
  • Surface plasmon analysis can also be performed by immobilizing analytes (substances on a biosensor matrix) and presenting ligands.
  • PD-1 or "PD1” generally refers to apoptosis protein 1, a 288 amino acid type I membrane protein first described in 1992 (Ishida et al., EMBO J., 11 (1992), 3887-3895).
  • PD-1 is a member of the expanded CD28/CTLA-4 T cell regulator family and has two ligands, PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273).
  • the structure of the protein includes an extracellular IgV domain followed by a transmembrane region and an intracellular tail.
  • the intracellular tail contains two phosphorylation sites located in the immunoreceptor tyrosine-based inhibitory motif and the immunoreceptor tyrosine-based switch motif, suggesting that PD-1 negatively regulates TCR signaling. This is consistent with the binding of SHP-1 and SHP-2 phosphatases to the cytoplasmic tail of PD-1 upon ligand binding.
  • the terms "apoptotic 1”, “apoptotic cell death 1”, “protein PD-1”, “PD-1”, PD1”, PDCD1", “hPD-1” and “hPD-I” are used interchangeably , and also includes variants, isoforms, species homologs, and analogs of human PD-1 that share at least one epitope with PD-1.
  • the amino acid sequence of human PD1 can be shown in UniProt (www.uniprot.org) accession number Q15116.
  • the term “PD-L1” or “PDL1” generally refers to programmed cell death 1 ligand 1, which may also be referred to as B7 homolog 1, B7-H1, cluster of differentiation 274, (3)274 or CD274, which binds to PD-1 downregulates T cell activation and cytokine secretion.
  • P-L1 includes any native PD-L1 from any vertebrate source, including mammals, such as primates (eg, humans and cynomolgus monkeys) and rodents (eg, mice and rats) ).
  • the term encompasses "full-length", unprocessed PD-L1 as well as any form of PD-L1 produced by cellular processing.
  • PD-L1 can exist as a transmembrane protein or as a soluble protein.
  • "PD-L1" includes complete PD-L1 and fragments thereof, as well as functional variants, isoforms, species homologues, derivatives, analogs of PD-L1, and functional variants, isoforms, derivatives, and analogs of PD-L1, as well as those having at least one in common with PD-L1 Epitope analogs.
  • the basic structure of PD-L1 includes four domains: extracellular Ig-like V-type domain and Ig-like C2-type domain, transmembrane domain and cytoplasmic domain.
  • Exemplary human PD-L1 amino acid sequences can be found under NCBI Accession No. NP_001254653 or UniProt Accession No. Q9NZQ7.
  • antibody generally refers to an immunoglobulin reactive against a specified protein or peptide or fragment thereof.
  • Such antibodies include, but are not limited to, human antibodies, primatized antibodies, chimeric antibodies, monoclonal antibodies, monospecific antibodies, polyclonal antibodies, multispecific antibodies, nonspecific antibodies, bispecific antibodies, multispecific antibodies
  • Antibodies can be from any class of antibodies, including but not limited to IgG, IgA, IgM, IgD, and IgE, and from any subclass (eg, IgGl, IgG2, IgG3, and IgG4).
  • the antibody may have a heavy chain constant region selected from, eg, IgGl, IgG2, IgG3, or IgG4.
  • the antibody may also have a light chain selected from, for example, kappa ( ⁇ ) or lambda ( ⁇ ).
  • the antibodies of the present application can be derived from any species, including but not limited to mouse, human, camel, llama, fish, shark, goat, rabbit, chicken, and bovine.
  • the constant region of the antibody can be altered, eg, mutated, to modify the properties of the antibody (eg, to increase or decrease one or more of the following: Fc receptor binding, antibody glycosylation, number of cysteine residues, effect organ cell function, or complement function).
  • the antibody specifically binds to a predetermined antigen, eg, an antigen associated with a disorder, eg, an inflammatory, immune, autoimmune, neurodegenerative, metabolic and/or malignant disorder.
  • chimeric antibody generally refers to an antibody in which the variable region of a murine antibody is fused with the constant region of a human antibody, which can alleviate the immune response induced by the murine antibody.
  • a hybridoma that secretes a mouse-specific monoclonal antibody can be established, and then the variable region gene can be cloned from the mouse hybridoma cell, and the constant region gene of the human antibody can be cloned according to the needs.
  • the human constant region gene is connected into a chimeric gene and inserted into an expression vector, and the chimeric antibody molecule can be expressed in a eukaryotic system or a prokaryotic system.
  • humanized antibody also known as CDR-grafted antibody
  • CDR-grafted antibody generally refers to the grafting of murine CDR sequences into the framework of human antibody variable regions, i.e. different Types of human germline antibody framework sequences produced in antibodies.
  • the heterologous reaction induced by chimeric antibodies can be overcome because they carry a large amount of murine protein components.
  • framework sequences can be obtained from public DNA databases or published references that include germline antibody gene sequences.
  • the germline DNA sequences of human heavy and light chain variable region genes can be found in the "VBase" human germline sequence database.
  • monoclonal antibodies has gone through four stages, namely: murine monoclonal antibodies, chimeric monoclonal antibodies, humanized monoclonal antibodies and fully human monoclonal antibodies.
  • the antibodies or ligands described herein may be fully human monoclonal antibodies.
  • Related technologies for the preparation of fully human antibodies include: human hybridoma technology, EBV transformation of B lymphocytes, phage display technology, transgenic mouse antibody preparation technology, and single B cell antibody preparation technology.
  • CDR generally refers to one of the six hypervariable regions within the variable domain of an antibody that primarily contribute to antigen binding.
  • 6 CDRs are provided by Kabat E.A. et al., (1991) Sequences of proteins of immunological interest. NIH Publication 91-3242), Chothia et al., “Canonical Structures For the Hypervariable Regions of Immunoglobulins, "J. Mol. Biol. 196:901 (1987); and MacCallum et al., “Antibody-Antigen Interactions: Contact Analysis and Binding Site Topography,” J. Mol. Biol. 262:732 (1996)).
  • CDR L1, CDR L2, CDR L3 or L1, L2, L3 the Kabat definition of CDRs can be applied to CDR1, CDR2 and CDR3 (CDR L1, CDR L2, CDR L3 or L1, L2, L3) of the light chain variable domains, as well as the heavy chain variable domains of CDR1, CDR2 and CDR3 (CDR H1, CDR H2, CDR H3 or H1, H2, H3).
  • the term "antigen-binding fragment” generally refers to one or more polypeptide fragments that have the ability to specifically bind an antigen (eg, PD-L1).
  • the antigen-binding fragment may include Fab, Fab', F(ab) 2 , Fv fragment, F(ab') 2 , scFv, di-scFv and/or dAb.
  • variable region generally refers to a region where certain segments of the variable domain may differ significantly in sequence between antibodies.
  • a “variable region” in a light chain may comprise a light chain variable region VL; a “variable region” in a heavy chain may comprise the heavy chain variable region VH.
  • the variable domains mediate antigen binding and determine the specificity of a particular antibody for its particular antigen.
  • the variability is not evenly distributed across the variable domains. It is usually concentrated in three segments called hypervariable regions (CDRs or HVRs) in the light and heavy chain variable domains.
  • CDRs or HVRs hypervariable regions
  • the more highly conserved portions of variable domains are referred to as framework regions (FRs).
  • variable domains of native heavy and light chains each comprise four FR regions, most adopting a ⁇ -sheet configuration, connected by three CDRs that form loops connecting, and in some cases forming part of, the ⁇ -sheet structure .
  • the CDRs in each chain are held in close proximity by the FR regions, and the CDRs from the other chain together contribute to the formation of the antigen-binding site of the antibody (see Kabat et al, Sequences of Immunological Interest, Fifth Edition, National Institute of Health, Bethesda, Md. (1991)).
  • Fab generally refers to an antigen-binding fragment of an antibody.
  • Intact antibodies can be digested with papain as described above. Papain digestion of the antibody yields two identical antigen-binding fragments, the "Fab” fragment, and a residual "Fc” fragment (ie, the Fc region, supra).
  • Fab fragments may consist of a complete L chain with the variable region of a heavy chain and the first constant region (CH1) of the H chain (VH).
  • Fab' or "Fab' fragment” generally refers to a monovalent antigen-binding fragment of a human monoclonal antibody, which fragment may be slightly larger than a Fab fragment.
  • a Fab' fragment can include all of the light chain, all of the variable regions of the heavy chain, and all or part of the first and second constant regions of the heavy chain.
  • a Fab' fragment may also include part or all of the 220-330 amino acid residues of the heavy chain.
  • (Fab')2 generally refers to antibody fragments produced by pepsin digestion of whole antibodies.
  • the F(ab')2 fragment contains two Fab fragments and part of the hinge region held together by disulfide bonds.
  • F(ab')2 fragments have bivalent antigen-binding activity and are capable of cross-linking antigens.
  • Fv or “Fv fragment” generally refers to a monovalent antigen-binding fragment of a human monoclonal antibody, comprising all or part of the heavy and light chain variable regions, and lacking the heavy chain constant regions and light chain constant region.
  • Heavy chain variable regions and light chain variable regions include, for example, CDRs.
  • Fv fragments include all or part of the amino-terminal variable regions of the heavy and light chains of about 110 amino acids.
  • the term "scFv” generally refers to a fusion protein comprising at least one antibody fragment comprising a variable region of a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein the light and heavy chains can be The variable regions are contiguous (eg, via synthetic linkers such as short flexible polypeptide linkers) and can be expressed as a single-chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived.
  • a scFv may have the VL and VH variable regions described in any order (eg, with respect to the N- and C-termini of the polypeptide), and the scFv may include a VL-linker-VH or VH-linker-VL can be included.
  • dAb generally refers to an antigen-binding fragment consisting of a VH domain or a VL domain, see for example Ward et al. (Nature, 1989 Oct 12; 341(6242): 544-6), see Holt et al., Trends Biotechnol., 2003, 21(11): 484-490.
  • VHH generally refers to an antibody comprising the variable antigen binding domain of a heavy chain antibody (see Vanlandschoot P. et al., 2011, Antiviral Research 92, 389-407). VHHs may also be referred to as Nanobodies (Nb).
  • anti-CD3 antibody generally refers to an antibody or variant thereof that targets CD3, such as a monoclonal antibody, including human, humanized, chimeric or murine antibodies, directed against T cells of mature T cells CD3 receptor among antigen receptors.
  • Anti-CD3 antibodies can include OKT3.
  • Anti-CD3 antibodies can include SP34.
  • Anti-CD3 antibodies can also include other anti-CD3 antibodies including, for example, in one embodiment otelixizumab, teplizumab, and visilizumab.
  • IL-2 or "IL2” generally refers to the T cell growth factor known as interleukin-2, and includes all forms of IL-2, which in one embodiment may include humans and mammals forms, conservative amino acid substitutions, glycoform modifications or variants, or active fragments thereof.
  • the GeneID encoding the IL-2 gene may be 3558.
  • the term "antigen-presenting cell”, “antigen-presenting cell”, or “APC” generally refers to an immunizing agent that displays on its surface a foreign antigen complexed with the major histocompatibility complex (MHC).
  • Systemic cells such as helper cells (eg, B cells, dendritic cells, etc.). T cells can recognize these complexes using their T cell receptors (TCRs).
  • TCRs T cell receptors
  • APCs can process and present antigens to T cells.
  • the antigen presenting cells may comprise selected from the group consisting of peripheral mononuclear cells, dendritic cells, and artificial antigen presenting cells.
  • expansion effect generally refers to the effect that occurs after a cell has been expanded. Changes in the effect of expansion can include changes in the number and/or ratio of cells, changes in secretion capacity, changes in killing capacity, or changes in expression capacity, or any combination thereof. Variations of the present application may be increased or decreased.
  • nanoparticle generally refers to at least one microscopic particle having a size of less than 100 nm.
  • nanoparticles typically have diameters in the range of 50 nm to 500 nm (ie, 0.05 ⁇ m to 0.5 ⁇ m); are structurally stable in physiological environments; and can accommodate smaller molecules (such as drugs or other bioactive agents), which can then be delivered to the desired site.
  • the nanoparticles of the present application may comprise a CD28 antibody or antigen-binding fragment thereof.
  • the nanoparticles of the present application may comprise a CD28 antibody or antigen-binding fragment thereof and a CD3 antibody or antigen-binding fragment thereof.
  • an anti-CD28 antibody can include OKT3.
  • an anti-CD28 antibody can include 15E8.
  • an artificial antigen-presenting cell generally refers to an artificially constructed immune cell for presenting exogenous antigens.
  • the complex of the histocompatibility complex MHC.
  • an isolated artificial antigen presenting cell may be included, which may comprise expression of HLA-A/B/C (the gene encoding the GeneID of which may be 3105, 3106 or 3107), CD64 (the gene encoding it GeneID can be 2209), CD80 (GeneID encoding it can be 941), ICOS-L (GeneID encoding it can be 23308) and CD58 (GeneID encoding it can be 965) cells, and can be modified To express more than one T cell activator, the above of the present application may include this number.
  • fusion protein generally refers to an amino acid sequence comprising a first polypeptide or protein or a fragment, analog or derivative thereof and a heterologous polypeptide or protein (ie, different from the first polypeptide or protein or of the amino acid sequence of a second polypeptide or protein or a fragment, analog or derivative thereof, or generally not part of the first polypeptide or protein or fragment, analog or derivative thereof) polypeptide or protein.
  • a fusion protein can comprise a prophylactic or therapeutic drug fused to a heterologous protein, polypeptide or peptide.
  • the heterologous proteins, polypeptides or peptides of the present application may or may not be different types of prophylactic or therapeutic drugs.
  • fusion protein may retain or increase the activity of the heterologous protein, polypeptide, or the original polypeptide or protein prior to protein fusion.
  • the fusion protein of the present application can be a fusion protein fused with a CD28 antibody or an antigen-binding fragment thereof, and a CD3 antibody or an antigen-binding fragment thereof.
  • the term "killing ability" generally refers to killing target cells by contacting the cells of the present application with an effective amount of a substance.
  • the agents of the present application may be TIL cells. Killing of the present application may include killing cells by themselves or by promoting CDC, apoptosis, ADCC, and/or phagocytosis of other cells or substances, or by a combination of two or more of these mechanisms.
  • administration generally refers to the delivery of a substance to a subject in need thereof by any route known in the art.
  • Pharmaceutically acceptable carriers and formulations or compositions are also well known in the art. Routes of administration may include: intravenous, intramuscular, intradermal, subcutaneous, transdermal, mucosal, intratumoral and/or mucosal.
  • kit generally refers to two or more components packaged together in a container, receptacle or other container, one of which corresponds to the substance of this application.
  • TIL cells of the present application are included.
  • the term "subject” generally refers to cells or animals, which may be mammals such as humans, non-human primates (apes, gibbons, gorillas, chimpanzees, orangutans, macaques), domestic animals (dogs and cats), farm animals (poultry such as chickens and ducks, horses, cattle, goats, sheep, pigs) and laboratory animals (mice, rats, rabbits, guinea pigs).
  • Human subjects include fetal, neonatal, infant, adolescent and adult subjects.
  • Subjects include animal disease models, such as tumor animal models, and other animal models known to those of skill in the art.
  • the term "feeder” generally refers to a cultured cell that grows in vitro and secretes at least one factor into the culture medium and can be used to support the growth of another cell of interest.
  • the feeder cells may comprise antigen presenting cells.
  • the term "specifically binds” generally refers to an antibody that recognizes a specific antigen, but does not substantially recognize or bind to other molecules in a sample.
  • an antibody can specifically bind a specific antigen of the present application from one species, the antibody of the present application can also specifically bind an antigen of the present application or a cognate antigen from another species or species. Such interspecies reactivity may not by itself alter the classification of the antibody as specific.
  • antibodies that specifically bind to an antigen can also bind to different allelic forms of the antigen.
  • complete culture process generally refers to the complete process of starting cells from isolated tumor tissue from a patient, going through one or more expansions, and finally obtaining cells that can be administered to a subject. .
  • cell culture medium generally refers to a nutrient solution in which cells, such as mammalian cells, are grown.
  • the formulation of cell culture media is well known in the art.
  • cell culture media includes buffers, salts, carbohydrates, amino acids, vitamins, and necessary trace elements.
  • the cell culture medium may or may not contain serum, peptone, and/or protein.
  • Cell culture media can be supplemented with additional components or increased concentrations of components such as amino acids, salts, sugars, vitamins, hormones, growth factors, buffers, antibiotics, lipids, trace elements, etc., depending on the cells to be cultured requirements and/or desired cell culture parameters.
  • the term "pharmaceutical composition” or “pharmaceutical formulation” generally refers to a preparation which may allow the biological activity of the active ingredient to be effective and which may be free of receptors for which the formulation will be administered. test subjects unacceptably toxic additional components. Such formulations are sterile. "Pharmaceutically acceptable” excipients (carriers, additives) are those that can reasonably be administered to a subject mammal to provide an effective dose of the active ingredient used.
  • TIL tumor infiltrating lymphocytes
  • TILs can include, but are not limited to, CD8 + cytotoxic T cells (lymphocytes), Th1 and Th17 CD4 + T cells, natural killer cells, dendritic cells, and M1 macrophages.
  • TILs can include primary TILs and secondary TILs.
  • Primary TILs can be those TIL cells obtained from a subject tissue sample, and "secondary TILs” can be any TIL cell population that has been expanded or expanded in the present application.
  • the tumor-infiltrating lymphocytes of the present application may not be isolated and purified, or may be mutually infiltrated with tumor cells.
  • TIL of the present application may refer to a population of TIL cells.
  • central memory T cells generally refers to T cells that have long-term memory and are able to receive antigenic restimulation.
  • Central memory T cells can have a CD45RA - CCR7 + phenotype, for example, central memory T cells can be identified by CD45RA- and CCR7 + .
  • Central memory T cells can have stronger anti-tumor growth ability than ordinary T cells.
  • regulatory T cells generally refers to a subset of T cells that control autoimmune reactivity in the body. Regulatory T cells can have the phenotype of CD4 + CD25 + Foxp3 + , eg, regulatory T cells can be identified by CD4 + , CD25 + , and Foxp3 + . Regulatory T cells may have the ability to suppress the antitumor growth of T cells.
  • activated T cells generally refers to T cells that have been activated to have the ability to resist tumor growth.
  • Activated T cells may have the phenotype of PD-1 + , LAG3 + or CD28 + , eg, activated T cells may be identified by PD-1 + , LAG3 + or CD28 + .
  • Activated T cells may have the ability to resist tumor growth.
  • tumor-specific T cells generally refers to T cells that can specifically fight tumor growth.
  • Tumor-specific T cells can have a CD103 + CD39 + phenotype, eg, tumor-specific T cells can be identified by CD103 + and CD39 + .
  • Tumor-specific T cells may have a more specific anti-tumor growth ability than ordinary T cells.
  • stem-like T cells generally refers to a class of T cells that can have the potential to self-proliferate and/or differentiate.
  • Stem-like T cells can have a TCF1 + phenotype, for example, stem-like T cells can be identified by TCF1 + .
  • Tumor-specific T cells may have stronger and/or longer-term anti-tumor growth ability than normal T cells.
  • tumor fragments generally refers to tumor fragments that can be formed by mechanical disruption, enzymatic hydrolysis, and/or other disruption methods after tumor tissue is removed from a subject.
  • composition or “pharmaceutical composition” generally refers to at least one cell and at least one and optionally more than one other pharmaceutically acceptable chemical components such as carriers, stabilizers , a mixture of diluents, dispersing agents, suspending agents, thickening agents and/or excipients.
  • the term "pharmaceutically acceptable carrier” generally refers to one or more non-toxic materials that do not interfere with the active ingredient.
  • a pharmaceutically acceptable carrier may not interfere with the biological activity of the active ingredient; for example, a pharmaceutically acceptable carrier may not interfere with the effectiveness of the biological activity possessed by the active ingredient.
  • Such formulations may conventionally contain salts, buffers, preservatives, compatible carriers, and optionally other therapeutic agents.
  • Such pharmaceutically acceptable formulations may also contain compatible solid or liquid fillers, diluents or encapsulating substances suitable for administration to humans.
  • contemplated carriers, excipients, and/or additives may include, for example, flavoring agents, antimicrobial agents, sweeteners, antioxidants, antistatic agents, lipids , protein excipients (such as serum albumin, gelatin, casein), salt-forming counterions (such as sodium), and the like.
  • flavoring agents such as peppermint, peppermint, peppermint, trehalose, trehalose, trehalose, trehalose, glycerin, trehalose, glycerin, glycerin, sorbitol, sorbitol, and the like.
  • antimicrobial agents such as sweeteners, antioxidants, antistatic agents, lipids , protein excipients (such as serum albumin, gelatin, casein), salt-forming counterions (such as sodium), and the like.
  • salt-forming counterions such as sodium
  • the term "functionally active fragment” generally refers to a fragment that has a partial region of a full-length protein or nucleic acid, but retains or partially retains the biological activity or function of the full-length protein or nucleic acid.
  • a functionally active fragment may retain or partially retain the ability of the full-length protein to bind another molecule.
  • a functionally active fragment of the growth factor IL-2 may retain or partially retain the biologically active function of full-length IL-2 that causes cell proliferation.
  • T cell activator generally refers to a substance that binds to the corresponding binding receptor on T cells and mediates T cell co-stimulatory responses.
  • T cell activators can be substances other than antigen receptors that are required by T cells to generate an effective immune response.
  • T cell activators may refer to T cell costimulatory molecules.
  • T cell activators may include, but are not limited to, MHC class I molecules, TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activation molecules (SLAM proteins), NK cell activation receptors, BTLA (the gene encoding it can be 151888), Toll ligand receptor, OX40 (the gene encoding it can be 7293), CD2 (the gene encoding it can be 914), CD7 (the gene encoding it can be GeneID 914) is 924), CD27 (the gene encoding it can be 939), CD28 (the gene encoding it can be 940), CD30 (the gene encoding it can be 943), CD40 (the gene encoding it can be 958) ), CDS, ICAM-1 (the GeneID encoding the gene can be 3383), LFA-1 (CD11a/CD18) (the GeneID encoding the gene can be 3689), 4-1BB (CD137) (the GeneID encoding the
  • the term "T cell growth factor” generally refers to a biologically active polypeptide or small molecule compound that causes cell proliferation.
  • the T cell growth factor may be selected from one or more of the following group: IL-2 (the gene encoding it may be GeneID 3558), IL-4 (the gene encoding it may be 3565) , IL-7 (the gene GeneID encoding it can be 3574), IL-10 (the gene encoding it can be 3586), IL-12 (the gene encoding it can be 3592 or 3593), IL-15 (the gene encoding it can be 3592 or 3593) Its gene GeneID may be 3600), and gamma interferon (the gene encoding it may be 3458).
  • IL-2 the gene encoding it may be GeneID 3558
  • IL-4 the gene encoding it may be 3565
  • IL-7 the gene GeneID encoding it can be 3574
  • IL-10 the gene encoding it can be 3586
  • IL-12 the gene en
  • substantially simultaneously generally means that the TIL can be in contact with two or more substances simultaneously during a period of time during the contacting process, but may not be limited to the fact that the TIL is always simultaneously with two or more substances during the entire contacting process touch. In one embodiment, substantially simultaneously can mean that TIL can interact with at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 75%, 80%, 85%, 90%, 95% of each of the two or more substances are in contact at the same time.
  • solid phase medium generally refers to a solid phase material that has a binding function.
  • the solid-phase medium in the present application may refer to a material that binds one or more substances in the medium and/or on the surface of the medium through covalent binding and/or non-covalent binding.
  • the solid phase medium of the present application can bind one or more T cell activators.
  • the solid-phase medium of the present application may refer to the binding of CD28 antibody or its antigen-binding fragment and CD3 antibody or its antigen-binding fragment in and/or on the surface of the medium through covalent binding and/or non-covalent binding.
  • the solid phase medium of the present application may be microspheres of about 500 nanometers to about 10 micrometers in diameter comprising the OKT3 antibody and the 15E8 antibody.
  • the solid phase medium of the present application may be a polymeric material.
  • the solid phase medium of the present application can be microspheres having a diameter of at least about 500 nanometers.
  • the solid phase medium of the present application may be a nanomatrix.
  • the solid phase medium of the present application may be a nanomatrix having a diameter of about 1 nanometer to about 500 nanometers comprising the OKT3 antibody and the 15E8 antibody.
  • the term "nanomatrix" generally refers to a material having a diameter of from about 1 nanometer to about 500 nanometers.
  • the nanomatrix can have a binding function, for example, the nanomatrix of the present application can bind one or more T cell activators.
  • the nanomatrix may comprise a polymer, for example, the nanomatrix of the present application may comprise a degradable polymer.
  • the nanomatrix may comprise polysaccharides, and/or dextran.
  • gene editing generally refers to a type of genetic engineering that utilizes one or more nucleases and/or nickases to insert, replace or remove DNA from a target DNA (eg, the TCR ⁇ genome of a cell).
  • a target DNA eg, the TCR ⁇ genome of a cell
  • gene knockout generally refers to genetic engineering means that silence a gene and/or disable the expression of the protein it encodes.
  • gene knockout can refer to the targeted disruption of a gene in a cell or in vivo, resulting in a complete loss of its function.
  • the gene knockout of the present application can use site-specific nucleases.
  • the gene knockout of the present application can use zinc finger nucleases (ZFNs), TAL effector nucleases (TALENs), and/or CRISPR/Cas-based systems.
  • ZFNs zinc finger nucleases
  • TALENs TAL effector nucleases
  • CRISPR/Cas-based systems for example, the gene knockout of the present application can use the CRISPR/Cas9 system.
  • dendritic cells generally refers to antigen-presenting cells that are present in vivo, in vitro, ex vivo or in a host or subject or which can be derived from hematopoietic stem cells or monocytes. Dendritic cells and their precursors can be isolated from various lymphoid organs such as spleen, lymph nodes, as well as bone marrow and peripheral blood. The dendritic cells of the present application may have characteristic morphologies such as lamellae (lamellipodia) extending in multiple directions of the dendritic cell body. In general, dendritic cells can express high levels of MHC and co-stimulatory (eg B7-1 and B7-2) molecules. Dendritic cells can induce antigen-specific differentiation of T cells in vitro and are able to elicit primary T cell responses in vitro and in vivo.
  • MHC co-stimulatory
  • in vitro expansion generally refers to culturing to produce changes in the number of cells, and the expanded cells can also produce changes in the number and/or ratio of cells, changes in secretion capacity, changes in killing capacity or expression Changes in abilities, or any combination of them. Variations of the present application may be increased or decreased.
  • in vitro expansion can be for the purpose of expansion; in order to detect the function of TIL cells, such as detecting the ability of TIL cells to release cytokines, the operation steps performed on TIL cells (such as adding a one or more substances to detect the ability of TIL cells to release cytokines), which may not belong to the in vitro expansion of the present application.
  • peripheral mononuclear cells or “peripheral blood mononuclear cells” generally refers to cells in peripheral blood that have a single nucleus.
  • the peripheral blood mononuclear cells of the present application may include lymphocytes, monocytes and/or dendritic cells.
  • cytokine generally refers to a protein released by one cell population that acts as an intercellular regulator of another cell.
  • Cytokines of the present application may be lymphokines, monokines and polypeptide hormones.
  • Cytokines of the present application may include interleukins (ILs) such as IL-1, IL-1 ⁇ , IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL-21, and/or IL-12.
  • ILs interleukins
  • the term cytokine may include proteins from natural sources or from recombinant cell culture, biologically active equivalents of native sequence cytokines, and functionally active fragments thereof.
  • the term “diameter” generally refers to the diameter of a cross-section of the substance of the application.
  • the term “diameter” generally refers to the largest diameter and/or the mean diameter of the largest cross-section of the substance of the present application.
  • the method of determining the diameter of the substance may be a method commonly used in the art, such as transmission electron microscopy.
  • tumor generally refers to any new pathological tissue proliferation.
  • the tumors of the present application may be benign or malignant.
  • the tumor of the present application may be solid or hematological.
  • the term “tumor” may be selected from one or more of the following group: melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, stomach cancer, colorectal cancer, and kidney cancer .
  • tumor tissue generally refers to a sample from a tumor in a subject, including any solid tumor and/or any tissue that is not a solid tumor in a subject.
  • CD28 agonist generally refers to a compound that binds to the cell surface CD28 protein and elicits a response in the cell.
  • a CD28 agonist of the present application can be a small molecule preparation that binds CD28.
  • a CD28 agonist of the present application can be an antibody or antigen-binding fragment thereof that binds CD28.
  • the term "proportion of T cell subsets" generally refers to the proportion of TIL cells or TIL cell populations according to different T cell subsets.
  • different T cell subsets of the present application have different immune activity and/or differentiation capacity.
  • T cell subsets of the present application can be differentiated based on T cell surface markers.
  • central memory T cells can have a CD45RA - CCR7 + phenotype.
  • regulatory T cells can have a CD4 + CD25 + Foxp3 + phenotype.
  • activated T cells can have the phenotype of CD25 + , CD28 + , TIM3 + , PD-1 + , or 41BB + .
  • tumor-specific T cells can have a CD103 + CD39 + phenotype.
  • stem-like T cells can have a TCF1 + phenotype.
  • the term "number of TIL cells” generally refers to the number of cells in the TIL cells of the present application.
  • the number of TIL cells may refer to the number of cells in the TIL population obtained at any stage of the present application.
  • the number of TIL cells can refer to the number of cells of the first TIL population derived from tumor tissue and not expanded in vitro.
  • the number of TIL cells can refer to the number of cells of the second TIL population expanded in vitro through the first stage.
  • the number of TIL cells can refer to the number of cells of the third TIL population expanded in vitro through the second stage.
  • the number of TIL cells can refer to the cells of TIL finally obtained by any one of the culturing methods of the present application.
  • the number of TIL cells can be measured by methods commonly used in the art, such as, but not limited to, manual cell counting with a cytometer and/or automated cell counter counting.
  • the terms “about” and “approximately” generally refer to within a statistically significant range of values. Such a range may be within an order of magnitude of a given value or range, may be within 50%, may be within 20%, may be within 10%, may be within 5%. The permissible variation encompassed by the term “about” or “approximately” may depend on the particular system under study, and can be readily understood by one of ordinary skill in the art. The terms “above”, “below”, “at most” and “at least” may include the present number.
  • the present application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: subjecting TILs derived from tumor tissue and not expanded in vitro through at least one stage of in vitro expansion, wherein in at least one In the in vitro expansion phase, the TIL is contacted with one or more T cell activators and one or more immune checkpoint inhibitors.
  • TILs tumor-infiltrating lymphocytes
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) combining a first population of TILs derived from tumor tissue and not expanded in vitro with one or more Contacting T cell growth factors; wherein, a second TIL cell population is obtained through the step (A); (B) contacting the second TIL population with one or more T cell growth factors of the present application and/or one contacting with one or more T cell activators; wherein, the third TIL cell population is obtained through the step (B); (C) the third TIL population is made to contact one or more T cell activators of the present application and a or exposure to multiple immune checkpoint inhibitors.
  • TILs tumor-infiltrating lymphocytes
  • the first stage in vitro amplification of the present application can be used in any substitution with step (A) in the method of the above aspect.
  • the second stage in vitro amplification of the present application can be used arbitrarily alternatively to step (B) in the method of the above aspect.
  • the TILs expanded in vitro in the first stage of the present application can be arbitrarily replaced with the second population of TILs obtained through step (A) of the method of the above aspect.
  • the TILs expanded in vitro in the second stage of the present application can be arbitrarily replaced with the third population of TILs obtained through step (B) of the method of the above aspect.
  • the third-stage in vitro amplification of the present application can be optionally used in place of any additional step (C) in the method of the above aspect, if necessary.
  • the TILs expanded in vitro in the third stage of the present application can be arbitrarily replaced with the fourth TIL population obtained through any additional step (C) in the method of the above aspect, if desired. .
  • the tumor tissue-derived TILs of the present application that have not been expanded in vitro can be subjected to a first-stage in vitro expansion, a second-stage in vitro expansion, and a third-stage in vitro expansion, and the present application
  • the TILs expanded in the second phase of the present application can be contacted with one or more T cell activators of the present application and/or immune checkpoint inhibitors of the present application.
  • the tumor tissue-derived TILs of the present application that have not been expanded in vitro can be subjected to a first-stage in vitro expansion, a second-stage in vitro expansion, and a third-stage in vitro expansion, and the present application
  • the TIL derived from the tumor tissue of the present application and not expanded in vitro can be combined with one or more T cell activators of the present application and/or the immune checkpoint inhibitor of the present application.
  • agent contact, and in the third stage in vitro expansion of the present application the TILs expanded in vitro by the second stage of the present application can be subjected to one or more T cell activators of the present application and/or the immunization of the present application Checkpoint inhibitor exposure.
  • the tumor tissue-derived TILs of the present application that have not been expanded in vitro can be subjected to a first-stage in vitro expansion, a second-stage in vitro expansion, and a third-stage in vitro expansion, and the present application
  • the TILs expanded in vitro in the first stage of the present application can be contacted with one or more T cell activators of the present application and/or immune checkpoint inhibitors of the present application
  • the TILs expanded in the second stage of the present application can be combined with one or more T cell activators of the present application and/or immune checkpoint inhibitors of the present application touch.
  • the tumor tissue-derived TILs of the present application that have not been expanded in vitro can be subjected to a first-stage in vitro expansion, a second-stage in vitro expansion, and a third-stage in vitro expansion, and the present application
  • the TIL derived from the tumor tissue of the present application and not expanded in vitro can be combined with one or more T cell activators of the present application and/or the immune checkpoint inhibitor of the present application.
  • the TILs expanded in vitro by the first stage of the present application can be subjected to one or more T cell activators of the present application and/or the immunization of the present application
  • the checkpoint inhibitor is contacted, and in the third stage of the in vitro expansion of the present application, the TILs expanded by the second stage of the present application in vitro can be contacted with one or more T cell activators of the present application and/or the present Apply for an immune checkpoint inhibitor exposure.
  • each stage of in vitro expansion can be divided by the change in the number of TIL cells, in one embodiment, TIL cells can be considered when the number of TIL cells increases by at least about 1-fold Entered the next stage of in vitro expansion. In some embodiments, when the number of TIL cells increases by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times At least about 50 times, at least about 100 times, at least about 200 times, at least about 500 times, or at least about 1000 times, TIL cells can be considered to have entered the next stage of in vitro expansion.
  • each stage of in vitro expansion may also be demarcated by changes in the conditions of TIL cell culture.
  • TIL cells can be considered to enter the next stage of in vitro expansion when T cell activators and/or T cell growth factors are added or supplemented to the cell culture medium.
  • TIL cells can be considered to have entered the next stage of in vitro expansion.
  • TIL cells can be considered to enter the next stage of in vitro expansion.
  • TIL cells can be considered to enter the next stage of in vitro expansion when immune checkpoint inhibitors are added or supplemented to the cell culture medium.
  • TIL cells when feeder cells are added or supplemented to the cell culture medium, TIL cells can be considered to have entered the next stage of in vitro expansion. In one embodiment, after the TIL cells are subjected to centrifugation and/or cell washing, it can be considered that the TIL cells have entered the next stage of in vitro expansion. In one embodiment, each stage can also be divided by the number of days in which the TIL cells are cultured.
  • TIL cells when TIL cells are cultured in vitro for about 1 day, about 2 days, about 3 days, about 4 days, about 5 days, about 6 days, about 7 days, about 8 days, about 9 days, about 10 days days, about 11 days, about 12 days, about 13 days, about 14 days, about 15 days, about 16 days, about 17 days, about 18 days, about 19 days, about 20 days, about 30 days, about 40 days, After about 50 days or about 100 days, the TIL cells can be considered to have entered the next stage of in vitro expansion.
  • the second stage in vitro expansion of the present application can be performed for up to about 13 days.
  • the number of days during which the second-stage in vitro expansion of the present application is performed can be calculated from the start time of the second-stage in vitro expansion. For example, when the second-stage in vitro expansion starts, it can be considered that the second-stage in vitro expansion has been carried out for about 0 days. For example, about 24 hours after the start of the second-stage in vitro expansion, it can be considered that the second-stage in vitro expansion has been carried out for about one day. For example, on the day when the second-stage in vitro expansion starts, it can be considered that the second-stage in vitro expansion has been carried out for about 0 days.
  • the number of days for the second-stage in vitro expansion of the present application may be calculated by the number of days for the second-stage in vitro expansion.
  • the second-stage in vitro expansion of the present application can be performed for up to about 13 days, up to about 12 days, up to about 11 days, up to about 10 days, up to about 9 days, up to about 8 days, up to about 7 days, up to about 6 days, up to about 5 days, up to about 4 days, up to about 3 days, up to about 2 days, or up to about 1 day.
  • the second stage in vitro expansion of the present application can be performed for about 3 days to about 13 days. In one embodiment, the second stage in vitro expansion of the present application can be performed for about 1 day to about 13 days. For example, the second stage in vitro expansion of the present application can be performed for about 2 days to about 13 days, about 3 days to about 13 days, about 4 days to about 13 days, about 5 days to about 13 days, about 6 days to about 6 days 13 days, about 7 days to about 13 days, about 8 days to about 13 days, about 9 days to about 13 days, about 10 days to about 13 days, about 11 days to about 13 days, or about 12 days to about 13 days sky.
  • the second stage in vitro expansion of the present application can be performed for about 2 days to about 3 days, about 2 days to about 4 days, about 2 days to about 5 days, about 2 days to about 6 days, about 2 days to about 2 days 7 days, about 2 days to about 8 days, about 2 days to about 9 days, about 2 days to about 10 days, about 2 days to about 11 days, about 2 days to about 12 days, or about 2 days to about 13 days sky.
  • the second stage in vitro expansion of the present application can be performed for about 3 days to about 4 days, about 3 days to about 5 days, about 3 days to about 6 days, about 3 days to about 7 days, about 3 days to about 3 days 8 days, about 3 days to about 9 days, about 3 days to about 10 days, about 3 days to about 11 days, about 3 days to about 12 days, or about 3 days to about 13 days.
  • the second stage in vitro expansion of the present application can be performed for about 13 days, about 12 days, about 11 days, about 10 days, about 9 days, about 8 days, about 7 days, about 6 days, about 5 days, about 4 days, about 3 days, about 2 days, or about 1 day.
  • the second stage of in vitro expansion of the present application can be considered as the REP (rapid expansion protocol) stage.
  • the first stage of in vitro expansion of the present application can be considered the preREP stage.
  • the third stage in vitro expansion of the present application can be performed for up to about 24 hours.
  • the number of days for the third-stage in vitro expansion of the present application can be calculated from the start of the third-stage in vitro expansion. For example, when the third-stage in vitro expansion starts, it can be considered that the third-stage in vitro expansion has been carried out for about 0 days. For example, it can be considered that the third-stage in vitro expansion has been carried out for about 24 hours after the start of the third-stage in vitro expansion for about 1 day. For example, on the day when the third-stage in vitro expansion starts, it can be considered that the third-stage in vitro expansion has been carried out for about 0 days.
  • the third-stage in vitro expansion has been carried out for about one day.
  • the third stage in vitro amplification of the present application can be performed for up to about 24 hours, up to about 23 hours, up to about 22 hours, up to about 21 hours, up to about 20 hours, up to about 19 hours, up to about 18 hours, up to about 17 hours, up to about 16 hours, up to about 15 hours, up to about 14 hours, up to about 13 hours, or up to about 12 hours.
  • the third stage in vitro expansion of the present application can be performed for about 12 hours to about 24 hours.
  • the third stage in vitro expansion of the present application can be performed for about 12 hours to about 24 hours, about 13 hours to about 24 hours, about 14 hours to about 24 hours, about 15 hours to about 24 hours, about 16 hours to about 24 hours, about 17 hours to about 24 hours, about 18 hours to about 24 hours, about 19 hours to about 24 hours, about 20 hours to about 24 hours, about 21 hours to about 24 hours, about 22 hours to about 24 hours , about 23 hours to about 24 hours, about 12 hours to about 22 hours, about 13 hours to about 22 hours, about 14 hours to about 22 hours, about 15 hours to about 22 hours, about 16 hours to about 22 hours, about 17 hours to about 22 hours, about 18 hours to about 22 hours, about 19 hours to about 22 hours, about 20 hours to about 22 hours, about 21 hours to about 22 hours, about 12 hours to about 20 hours, about 13 hours to about 20 hours, about 14 hours to about 20 hours, about 15 hours to about 20 hours, about 16 hours to about 20 hours, about 17 hours to about 20 hours, about 18 hours to about 20 hours, about 19 hours to about 22
  • the T-cell activator of the present application and/or the immune checkpoint inhibitor of the present application is compared to the corresponding TIL that has not been contacted with the T-cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion phase.
  • the cell activator and the TIL of the present application to which the immune checkpoint inhibitor of the present application has been contacted can show improved expansion effects.
  • the corresponding TIL that has not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to a TIL derived from the same donor and has not been contacted with the T cell activator of the present application and/or TIL cells contacted by an immune checkpoint inhibitor of the present application.
  • the corresponding TILs that have not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to the T cells from the same donor that have been isolated in the same way and have not been contacted with the T cells of the present application.
  • TIL cells contacted by the activator and/or the immune checkpoint inhibitor of the present application may refer to the same tumor source from the same donor and has not been activated with the T cell of the present application.
  • the corresponding TIL that has not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to the same tumor source from the same donor that has been isolated in the same way and has not been contacted with the immune checkpoint inhibitor of the present application.
  • TIL cells contacted by the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to the same tumor source from the same donor that has been isolated in the same way and has not been contacted with the immune checkpoint inhibitor of the present application.
  • the corresponding TILs that have not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to dividing TIL cells derived from the same donor into two groups, one of which has not been
  • the TIL cells contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may be corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application.
  • the corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to dividing TIL cells from the same donor and isolated in the same way into two groups , wherein a group of TIL cells that have not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application can be the corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application .
  • the corresponding TIL that has not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to dividing TIL cells derived from the same tumor from the same donor into two groups, One group of TIL cells that have not been in contact with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application can be corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application.
  • the corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application may refer to TIL cells isolated in the same way from the same tumor source from the same donor Divided into two groups, one group of TIL cells that have not been in contact with the T cell activator of the application and/or the immune checkpoint inhibitor of the application can be the T cell activator of the application and/or the immune checkpoint inhibitor of the application. Contact the corresponding TIL.
  • the improved expansion effect of the present application may comprise one or more selected from the group consisting of increased number of TIL cells, improved proportion of T cell subsets, increased cytokine secretion capacity, and Enhanced tumor cell killing ability.
  • the increased number of TIL cells of the present application may refer to the increase in the number of TIL cells in the The number of cells of the TIL of the present application that has been contacted with the T cell activator of the present application and the immune checkpoint inhibitor of the present application in at least one in vitro expansion stage can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased number of TIL cells of the present application may refer to the increase in the number of TIL cells in the The number of cells of the TIL of the present application that has been contacted with the T cell activator of the present application and the immune checkpoint inhibitor of the present application in at least one in vitro expansion stage can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1%, at least about 0.5%, at least about 0.4%, at least about 0.3%,
  • the increased cytokine secretion capacity of the present application may refer to the increased cytokine secretion capacity of TIL cells selected from the group consisting of CD107a, GZMB, IL-4, IL-17, IL-6, IL -2. TNF and IFN ⁇ .
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the cytokine secretion capacity of the TIL of the present application that has been contacted with the T cell activator of the present application and the immune checkpoint inhibitor of the present application can be increased at least about 1-fold, at least about 2-fold, at least about 3-fold during at least one in vitro expansion stage , at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times , at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the cytokine secretion capacity of the TILs of the present application that has been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application can be increased by at least about 100%, at least about 90%, at least about 80% during at least one phase of in vitro expansion , at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16% , at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6% , at least about
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the CD107a secretory capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application in at least one in vitro expansion stage can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, At least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the CD107a secretion capacity of the TILs of the present application that has been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application in at least one in vitro expansion stage can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the GZMB secretion capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application in at least one in vitro expansion stage can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, At least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the GZMB secretion capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage, IL-4, IL-17, IL-6, and/or IL-2 secretion capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application during at least one in vitro expansion phase can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold, at least about 5-fold, at least about 6-fold, at least about 7-fold, at least about 8-fold, at least about 9-fold, at least about 10-fold times, at least about 11 times, at least about 12 times, at least about 13 times, at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage, IL-4, IL-17, IL-6, and/or IL-2 secretion capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application during at least one in vitro expansion phase may increase by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19% %, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9% %, at least about 8%, at least about
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the TNF secretion capacity of the TILs of the present application that have been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application in at least one in vitro expansion stage can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, At least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the TNF secretion capacity of the TILs of the present application that has been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application may be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%,
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the IFN ⁇ secretory capacity of the TILs of the present application that has been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application can be increased at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 2-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times, At least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the increased cytokine secretion capacity of the present application may refer to a corresponding TIL that has not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the IFN ⁇ secretion capacity of the TILs of the present application that has been contacted with the T cell activators of the present application and the immune checkpoint inhibitors of the present application may be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%
  • the determination of the cytokine secretion capacity of the TILs of the present application may be by measuring the cytokine expression capacity of TIL cells.
  • the cytokine secretion ability of the immune cells of the present application can be determined by cytometry.
  • the cytokine secretion capacity of the TILs of the present application is determined by measuring the cytokine release capacity of TIL cells.
  • the cytokine secretion ability of the TIL of the present application is determined by the CBA method (Cytometric Bead Array).
  • the improved tumor cell killing ability of the present application may refer to the comparison with corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage,
  • the tumor cell killing rate of the TIL of the present application that has been contacted with the T cell activator of the present application and the immune checkpoint inhibitor of the present application can be increased at least about 1-fold, at least about 2-fold, at least about 3-fold during at least one in vitro expansion stage , at least about 4 times, at least about 5 times, at least about 6 times, at least about 7 times, at least about 8 times, at least about 9 times, at least about 10 times, at least about 11 times, at least about 12 times, at least about 13 times , at least about 14 times, at least about 15 times, at least about 20 times, at least about 30 times, at least about 40 times, or at least about 50 times.
  • the improved tumor cell killing ability of the present application may refer to the comparison with corresponding TILs that have not been contacted with the T cell activator of the present application and/or the immune checkpoint inhibitor of the present application during the in vitro expansion stage
  • the tumor cell killing rate of the TIL of the present application that has been contacted with the T cell activator of the present application and the immune checkpoint inhibitor of the present application can be increased by at least about 100%, at least about 90%, at least about 80% in at least one in vitro expansion stage , at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16% , at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6% , at least about 5%, at least about 4%,
  • the tumor cell killing rate of the TILs of the present application can be measured by CFSE and DAPI staining.
  • tumor cell killing of TILs of the present application may refer to the ability of TILs to kill solid tumor cells.
  • the tumor cell killing of TILs of the present application may refer to the ability of TILs to kill cervical cancer cells.
  • the tumor cell killing of TILs of the present application may refer to the ability of TILs to kill HeLa cells.
  • the improved proportion of T cell subsets of the present application may comprise one or more selected from the group consisting of: increased proportion of central memory T cells, decreased proportion of regulatory T cells, decreased depletion Proportion of T cells, increased proportion of activated T cells, increased proportion of tumor-specific T cells, and increased proportion of stem-like T cells.
  • the increased proportion of central memory T cells of the present application may be an increase in the proportion of CD45RA ⁇ CCR7 + cells in TIL cells.
  • the proportion of central memory T cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1%, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%.
  • the reduced proportion of regulatory T cells of the present application may be a reduction in the proportion of CD4 + CD25 + Foxp3 + cells in TIL cells.
  • the proportion of regulatory T cells in TIL cells can be reduced by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1%, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%.
  • the reduced proportion of exhausted T cells of the present application may be a reduction in the proportion of TIM3 + cells in TIL cells.
  • the proportion of depleted T cells in TIL cells can be reduced by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%.
  • the increased proportion of activated T cells of the present application may be an increase in the proportion of CD25 + , CD28 + , CD27 + , PD-1 + or 41BB + cells in TIL cells.
  • the proportion of activated T cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least
  • the proportion of CD25 + cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%, or can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold , at least about 5 times, at least about 6 times, or can
  • the proportion of CD28 + cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%, or can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold , at least about 5 times, at least about 6 times, or can
  • the proportion of CD27 + cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%, or can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold , at least about 5 times, at least about 6 times, or can
  • the proportion of PD-1 + cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about about 30%, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about about 11%, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about about about 1%, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%, or can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4 times, at least about 5 times, at least about 6 times, at least about
  • the proportion of 41BB + cells in TIL cells can be increased by at least about 100%, at least about 90%, at least about 80%, at least about 70%, at least about 60%, at least about 50%, at least about 40%, at least about 30% %, at least about 20%, at least about 19%, at least about 18%, at least about 17%, at least about 16%, at least about 15%, at least about 14%, at least about 13%, at least about 12%, at least about 11% %, at least about 10%, at least about 9%, at least about 8%, at least about 7%, at least about 6%, at least about 5%, at least about 4%, at least about 3%, at least about 2%, at least about 1% %, at least about 0.5%, at least about 0.4%, at least about 0.3%, at least about 0.2%, or at least about 0.1%, or can be increased by at least about 1-fold, at least about 2-fold, at least about 3-fold, at least about 4-fold , at least about 5 times, at least about 6 times, or can
  • the methods of the present application may further comprise: in at least one stage of the in vitro expansion of the present application, contacting the TIL of the present application with one or more T cell activators of the present application.
  • the T cell activator may comprise an agonist of one or more targets selected from the group consisting of CD3, CD28, HVEM, CD40L, OX40 and 4-1BB.
  • the TILs of the present application are contacted with one or more immune checkpoint inhibitors of the present application and with one or more T cell activators of the present application.
  • the TILs of the present application in the first stage of the in vitro expansion of the present application, can be contacted with one or more immune checkpoint inhibitors of the present application and with one or more of the TILs of the present application Cell Activator Contact.
  • the TILs of the present application in the second stage of the in vitro expansion of the present application, can be contacted with one or more immune checkpoint inhibitors of the present application and with one or more TILs of the present application Cell Activator Contact. In one embodiment, in the third stage of the in vitro expansion of the present application, the TILs of the present application can be contacted with one or more immune checkpoint inhibitors of the present application and with one or more of the TILs of the present application Cell Activator Contact.
  • the TIL of the present application in a single stage of in vitro expansion of the present application, can be made substantially simultaneously with one or more immune checkpoint inhibitors of the present application and one or more of the present application exposure to T cell activators.
  • the TILs of the present application in the first stage of the in vitro expansion of the present application, can be made substantially simultaneously with the one or more immune checkpoint inhibitors of the present application and one or more of the present application T cell activator contacts.
  • the TIL of the present application in the second phase of the in vitro expansion of the present application, can be substantially simultaneously combined with the one or more immune checkpoint inhibitors of the present application and one or more of the present application T cell activator contacts.
  • the TIL of the present application in the third stage of the in vitro expansion of the present application, can be substantially simultaneously combined with the one or more immune checkpoint inhibitors of the present application and one or more of the present application T cell activator contacts.
  • the T cell activator of the present application may comprise one or more selected from the group consisting of CD80, CD86, B7-H3, 4-1BBL, CD27, CD30, CD134, B7h, CD40, LIGHT , and their functionally active fragments.
  • the T cell activator of the present application may comprise an agonist of one or more targets selected from the group consisting of CD3, CD28, HVEM, CD40L, OX40 and 4-1BB.
  • the T cell activator of the present application may comprise antibodies selected from the group consisting of CD3, CD28, HVEM, CD40L, OX40, and 4-1BB, and antigen-binding fragments thereof.
  • the T cell activator of the present application may comprise a CD3 agonist. In one embodiment, the T cell activator of the present application may comprise an anti-CD3 antibody and/or an antigen-binding fragment thereof, such as OKT3 from Miltenyi Biotech and SP34 from BD.
  • the T cell activator of the present application may comprise an anti-CD3 antibody and/or an antigen-binding fragment thereof, for example, may comprise HCDR1-3 of Miltenyi Biotech's OKT3 and/or LCDR1-3 of Miltenyi Biotech's OKT3 3, for example can be the HCDR1-3 of SP34 comprising BD and/or LCDR1-3 of OKT3 of Miltenyi Biotech, for example can be the VH of OKT3 that comprises Miltenyi Biotech and/or the VL of OKT3 of Miltenyi Biotech, for example can be comprising BD VH of SP34 and/or VL of OKT3 from Miltenyi Biotech.
  • the T cell activator of the present application may comprise a CD28 agonist.
  • the T cell activator of the present application may comprise an anti-CD28 antibody and/or an antigen-binding fragment thereof, such as Sigma-Aldrich's 15E8.
  • the T cell activator of the present application may comprise an anti-CD28 antibody and/or an antigen-binding fragment thereof, for example, may comprise HCDR1-3 of Sigma-Aldrich's 15E8 and/or Sigma-Aldrich's 15E8 LCDR1-3, for example, may comprise the VH of Sigma-Aldrich's 15E8 and/or the VL of Sigma-Aldrich's 15E8.
  • contacting the TILs of the present application with one or more T cell activators of the present application may comprise one or more means selected from the group consisting of: (1) activating the T cells of the present application (2) adding the engineered cells expressing the T cell activator of the present application to the cell culture medium of the TIL of the present application; (3) adding the T-cell activator of the present application The solid phase medium of the cell activator is added to the cell culture medium of the TIL of the present application.
  • contacting a TIL of the present application with one or more T cell activators of the present application may comprise adding a solid phase medium comprising the T cell activator of the present application to a cell culture of the TIL of the present application base.
  • contacting a TIL of the present application with one or more T cell activators of the present application may comprise adding a solid phase medium comprising the CD28 antibody of the present application and the CD3 antibody of the present application to the cells of the TIL of the present application in the culture medium.
  • the initial concentration of the T cell activator in the cell culture medium of the TIL of the present application may be at least about 30 ng/mL.
  • the initial concentration of the CD28 antibody of the present application in the cell culture medium of the TIL of the present application can be at least about 30 ng/mL;
  • the initial concentration of the CD3 antibody of the present application in the cell culture medium of the TIL of the present application can be at least about 30ng/mL.
  • the selection of the initial concentration of the CD28 antibody of the present application can be independent of the selection of the initial concentration of the CD3 antibody of the present application; for example, the initial concentration of the CD28 antibody of the present application and the CD3 antibody of the present application in the cell culture medium of the TIL of the present application Any combination is possible.
  • the initial concentration of the CD28 antibody of the present application in the cell culture medium of the TIL of the present application can be arbitrarily selected from about 30 ng/mL to about 300 ng/mL.
  • the initial concentration of the CD3 antibody of the present application in the cell culture medium of the TIL of the present application can be arbitrarily selected from about 30 ng/mL to about 300 ng/mL.
  • the initial concentration of the CD28 antibody of the present application in the cell culture medium of the TIL of the present application can be arbitrarily selected from about 30 ng/mL to about 300 ng/mL, and the initial concentration of the CD3 antibody of the present application in the cell culture medium of the TIL of the present application
  • the concentration can be arbitrarily selected from about 30 ng/mL to about 300 ng/mL, and the selection of the initial concentration of the CD28 antibody of the present application can be independent of the selection of the initial concentration of the CD3 antibody of the present application.
  • the solid phase medium of the present application may have a diameter of about 500 nanometers to about 10 micrometers.
  • the diameter of the solid phase medium of the present application can be measured by transmission electron microscopy.
  • the solid phase medium of the present application may have a diameter of about 1 nanometer to about 500 nanometers.
  • the solid phase medium of the present application may have a diameter of about 100 nanometers to about 500 nanometers.
  • the solid phase medium of the present application may have a diameter of about 200 nanometers to about 500 nanometers.
  • the diameter of the solid phase medium of the present application can be measured by transmission electron microscopy.
  • the solid phase medium of the present application may comprise a polymer. In one embodiment, the solid phase medium of the present application may comprise dextran.
  • the solid phase medium of the present application comprises at least about 25 ⁇ g of the T cell activator of the present application per mg of the present application.
  • the amount of each T cell activating agent contained per mg of the solid phase medium of the present application may be independent of each other.
  • the solid phase medium comprising one or more T cell activators of the present application is added to the present application at a ratio of about 1:100 to about 1:2000 of the present application solid phase medium to the present application TIL Apply TIL in the cell culture medium. In one embodiment, the solid phase medium comprising one or more T cell activators of the present application is added to the present application at a ratio of about 2:1 to about 1:2 of the present application solid phase medium to the present application TIL Apply TIL in the cell culture medium.
  • the ratio of the solid phase medium of the present application to the TIL of the present application may be about 2:1 to about 1:2, which will contain a A solid-phase medium of one or more T cell activators is added to the cell culture medium of the TILs of the present application.
  • the diameter of the solid phase medium of the present application is about 100 nanometers to about 500 nanometers, it may be about 2:1 to about 1:2, about 2:1 to about 1:1, or about 1:1 - a ratio of about 1:2 of the application's solid phase medium to the application's TILs, the solid phase medium comprising the application's one or more T cell activators, such as a CD3 agonist and/or a CD28 agonist, is added to the application TIL in cell culture medium.
  • T cell activators such as a CD3 agonist and/or a CD28 agonist
  • the ratio of the solid phase medium of the present application to the TIL of the present application may be about 1:100 to about 1:2000, which will contain a A solid-phase medium of one or more T cell activators is added to the cell culture medium of the TILs of the present application.
  • the diameter of the solid phase medium of the present application when the diameter of the solid phase medium of the present application is about 100 nanometers to about 500 nanometers, it can be about 1:100-about 1:2000, about 1:200-about 1:2000, about 1:300- About 1:2000, about 1:400-about 1:2000, about 1:500-about 1:2000, about 1:600-about 1:2000, about 1:700-about 1:2000, about About 1:800-about 1:2000, about 1:900-about 1:2000, about 1:1000-about 1:2000, about 1:1200-about 1:2000, about 1:1400-about 1 :2000, at about 1:1600-about 1:2000, or at a ratio of about 1:1800-about 1:2000 of the application's solid phase medium to the application's TIL, will comprise one or more T-cell activation of the application
  • a solid phase medium of an agent such as a CD3 agonist and/or a CD28 agonist, is added to the cell culture medium of the TILs of the present application.
  • the immune checkpoint inhibitor of the present application comprises a substance that inhibits the interaction of PD-1 with PD-L1 and/or PD-L2.
  • the immune checkpoint inhibitor of the present application may comprise a PD-1 inhibitor; for example, the immune checkpoint inhibitor of the present application may comprise an antibody and/or antigen-binding fragment thereof of PD-1.
  • the immune checkpoint inhibitor of the present application comprises a substance having the ability to bind PD -1 with a KD value of about 100 pM or less, eg, a KD value of about 100 pM or less, about 90 pM A KD value of about 80 pM or less, a KD value of about 50 pM or less, a KD value of about 30 pM or less, or a KD value of about 10 pM or less.
  • the immune checkpoint inhibitor of the present application comprises a substance having the ability to bind PD-1 with an EC 50 value of about 100 pM or less, eg, an EC 50 value of about 100 pM or less, about 90 pM An EC50 value of about 80 pM or less, an EC50 value of about 50 pM or less, an EC50 value of about 30 pM or less, or an EC50 value of about 10 pM or less.
  • the immune checkpoint inhibitor of the present application comprises a substance having the ability to inhibit the binding of PD-1 to PD-L1 and/or PD-L2 with an IC50 value of about 1 nM or less, eg, about IC50 value of 1 nM or less, IC50 value of about 0.9nM or less, IC50 value of about 0.8nM or less, IC50 value of about 0.5nM or less, IC50 value of about 0.3nM or less 50 value, or an IC50 value of about 0.1 nM or less.
  • an IC50 value of about 1 nM or less eg, about IC50 value of 1 nM or less, IC50 value of about 0.9nM or less, IC50 value of about 0.8nM or less, IC50 value of about 0.5nM or less, IC50 value of about 0.3nM or less 50 value, or an IC50 value of about 0.1 nM or less.
  • the immune checkpoint inhibitor of the present application may be a PD-1 antibody or an antigen-binding protein thereof.
  • the antibody or antigen-binding protein thereof of the present application comprises at least one CDR in the VH of the variable region of the antibody heavy chain.
  • the application CDRs may be defined according to IMGT nomenclature, the application CDRs may be defined according to Chothia, or the application CDRs may be defined according to Kabat.
  • the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody or antigen-binding protein thereof of the present application comprises at least one CDR in the VH of the variable region of the antibody heavy chain.
  • the present CDRs may be defined according to IMGT nomenclature, or the present CDRs may be defined according to Kabat.
  • the antibody of the present application or its antigen-binding protein may comprise HCDR1, and the HCDR1 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 1 and 15; the CDRs of the present application may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise HCDR2, and the HCDR2 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 2 and 16; the CDRs of the present application may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise HCDR3, and the HCDR3 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 3 and 17; the CDRs of the present application may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody or antigen-binding protein thereof of the present application may comprise HCDR1-3, wherein the HCDR1 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 1 and 15, and the HCDR2 of the present application may comprise SEQ ID NO: 2
  • the amino acid sequence shown in any one of and 16, and the application HCDR3 may comprise the amino acid sequence shown in any one of SEQ ID NO: 3 and 17; the application CDR may be defined according to Kabat;
  • the antigen binding protein may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein can comprise the same HCDR1-3 as 6H6 or Hu-6H6, wherein the HCDR1 of the present application can comprise the amino acid sequence shown in SEQ ID NO: 1, and the HCDR2 of the present application can comprise SEQ ID NO: 2
  • the amino acid sequence shown, and the HCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 3; the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have PD-1 binding ability .
  • the antibody of the present application or its antigen-binding protein may comprise the same HCDR1-3 as Pembrolizumab, wherein the HCDR1 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 15, and the HCDR2 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 16
  • the amino acid sequence of HCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 17; the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody or antigen-binding protein thereof of the present application comprises at least one CDR in the variable region VL of the antibody light chain.
  • the present CDRs may be defined according to IMGT nomenclature, or the present CDRs may be defined according to Kabat.
  • the antibody of the present application or its antigen-binding protein may comprise LCDR1, and LCDR1 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 4 and 18; the CDRs of the present application may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody or antigen-binding protein thereof of the present application may comprise LCDR2, and LCDR2 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 5 and 19; the present application CDRs may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise LCDR3, and LCDR3 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 6 and 20; the CDRs of the present application may be defined according to Kabat; for example, The antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise LCDR1-3, wherein the LCDR1 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 4 and 18, and the LCDR2 of the present application may comprise SEQ ID NO: 5
  • the amino acid sequence shown in any one of and 19, and the application LCDR3 may comprise the amino acid sequence shown in any one of SEQ ID NOs: 6 and 20;
  • the application CDRs may be defined according to the IMGT nomenclature;
  • the application CDRs Can be as defined by Kabat; for example, the antigen binding proteins of the present application can have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein can comprise the same LCDR1-3 as 6H6 or Hu-6H6, wherein the LCDR1 of the present application can comprise the amino acid sequence shown in SEQ ID NO: 4, and the LCDR2 of the present application can comprise SEQ ID NO: 5
  • the amino acid sequence shown, and LCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 6; the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have PD-1 binding ability .
  • the antibody of the present application or its antigen-binding protein may comprise the same LCDR1-3 as Pembrolizumab, wherein the LCDR1 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 18, and the LCDR2 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 19
  • the amino acid sequence of the present application, and LCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 20; the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody of the present application or its antigen-binding protein may comprise HCDR1-3 and LCDR1-3, wherein the HCDR1 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 1 and 15, and the HCDR2 of the present application may comprise the SEQ ID NO: 1 and 15.
  • the HCDR3 of the present application may comprise the amino acid sequence shown in any one of SEQ ID NO: 3 and 17, and the LCDR1 of the present application may comprise the amino acid sequence of SEQ ID NO: 4 and 18
  • the amino acid sequence shown in any one, LCDR2 of the present application can comprise the amino acid sequence shown in any one of SEQ ID NO: 5 and 19, and LCDR3 of the present application can comprise the amino acid sequence shown in any one of SEQ ID NO: 6 and 20.
  • the amino acid sequence shown in the present application; the CDRs of the present application may be defined according to the IMGT nomenclature; the CDRs of the present application may be defined according to Kabat; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody of the present application or its antigen-binding protein can comprise the same HCDR1-3 and LCDR1-3 as 6H6 or Hu-6H6, wherein the HCDR1 of the present application can comprise the amino acid sequence shown in SEQ ID NO: 1, and the HCDR2 of the present application can comprise SEQ ID NO: 1
  • the amino acid sequence shown in ID NO: 2 can comprise the amino acid sequence shown in SEQ ID NO: 3
  • the LCDR1 of the present application can comprise the amino acid sequence shown in SEQ ID NO: 4
  • the LCDR2 of the present application can comprise the amino acid sequence of SEQ ID NO: 4 : the amino acid sequence shown in 5
  • LCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 6;
  • the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein can comprise the same HCDR1-3 and LCDR1-3 as Pembrolizumab, wherein the HCDR1 of the present application can comprise the amino acid sequence shown in SEQ ID NO: 15, and the HCDR2 of the present application can comprise SEQ ID NO: : the amino acid sequence shown in 16, the application HCDR3 can include the amino acid sequence shown in SEQ ID NO: 17, the application LCDR1 can include the amino acid sequence shown in SEQ ID NO: 18, the application LCDR2 can include SEQ ID NO: 19
  • the amino acid sequence shown, and LCDR3 of the present application may comprise the amino acid sequence shown in SEQ ID NO: 20; the CDRs of the present application may be defined according to the Kabat nomenclature; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1 .
  • the antibody or antigen-binding protein thereof of the present application may comprise a heavy chain variable region VH, and the VH of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 7, 11 and 21;
  • the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH as 6H6, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 7; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH as Hu_6H6, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 11; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH as Pembrolizumab, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 21; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody or antigen-binding protein thereof of the present application may comprise a light chain variable region VL, and the VL of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 8, 12 and 22;
  • the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VL as 6H6, and the VL of the present application may comprise the amino acid sequence shown in SEQ ID NO: 8; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VL as Hu-6H6, and the VL of the present application may comprise the amino acid sequence shown in SEQ ID NO: 12; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VL as Pembrolizumab, and the VL of the present application may comprise the amino acid sequence shown in SEQ ID NO: 22; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody or antigen-binding protein thereof of the present application may comprise a heavy chain variable region VH and a light chain variable region VL, and the application VH may comprise any one of SEQ ID NOs: 7, 11 and 21
  • the amino acid sequence shown in the item, the VL of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 8, 12 and 22; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH and VL as a 6H6, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 7, and the VL of the present application may comprise SEQ ID NO: 8
  • the indicated amino acid sequence; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH and VL as Hu-6H6, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 11, and the VL of the present application may comprise the amino acid sequence shown in SEQ ID NO: 12
  • the amino acid sequence of for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody of the present application or its antigen-binding protein may comprise the same VH and VL as Pembrolizumab, and the VH of the present application may comprise the amino acid sequence shown in SEQ ID NO: 21, and the VL of the present application may comprise the amino acid sequence shown in SEQ ID NO: 22
  • the amino acid sequence of for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody or antigen-binding protein thereof of the present application may comprise a heavy chain, and the heavy chain of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 9, 13 and 23;
  • the antigen-binding protein of the application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain as 6H6, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 9; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain as Hu-6H6, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 13; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain as Pembrolizumab, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 23; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody or antigen-binding protein thereof of the present application may comprise a light chain, and the light chain of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 10, 14 and 24;
  • the antigen-binding protein of the application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same light chain as 6H6, and the light chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 10; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody of the present application or its antigen-binding protein may comprise the same light chain as Hu-6H6, and the light chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 14; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody of the present application or its antigen-binding protein may comprise the same light chain as Pembrolizumab, and the light chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 24; for example, the antigen-binding protein of the present application may have PD- 1 binding capacity.
  • the antibody or antigen-binding protein thereof of the present application may comprise a heavy chain and a light chain
  • the heavy chain of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 9, 13 and 23
  • the light chain of the present application may comprise the amino acid sequence shown in any one of SEQ ID NOs: 10, 14 and 24; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain and light chain as 6H6, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 9, and the light chain of the present application may comprise SEQ ID NO : the amino acid sequence shown in 10; for example, the antigen-binding protein of the present application may have PD-1 binding ability.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain and light chain as Hu-6H6, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 13, and the light chain of the present application may comprise SEQ ID NO : the amino acid sequence shown in 14; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibody of the present application or its antigen-binding protein may comprise the same heavy chain and light chain as Pembrolizumab, and the heavy chain of the present application may comprise the amino acid sequence shown in SEQ ID NO: 23, and the light chain of the present application may comprise SEQ ID NO: : the amino acid sequence shown in 24; for example, the antigen-binding protein of the present application may have the ability to bind to PD-1.
  • the antibodies of the present application are selected from the group consisting of chimeric antibodies, humanized antibodies, and fully human antibodies.
  • the antigen binding fragments of the present application are selected from the group consisting of Fab, Fab', Fv fragments, F(ab') 2 , F(ab) 2 , scFv, di-scFv, VHH and dAb.
  • the initial concentration of each immune checkpoint inhibitor of the present application in the cell culture medium of the TIL of the present application is each independently at least about 0.1 ⁇ g/mL.
  • the initial concentration of the PD-1 antibody in the cell culture medium of the TILs of the present application can be at least about 0.1 ⁇ g/mL, at least about 0.2 ⁇ g/mL, at least about 0.3 ⁇ g/mL, at least about 0.4 ⁇ g/mL, at least about 0.4 ⁇ g/mL about 0.5 ⁇ g/mL, at least about 0.6 ⁇ g/mL, at least about 0.7 ⁇ g/mL, at least about 0.8 ⁇ g/mL, at least about 0.9 ⁇ g/mL, at least about 1 ⁇ g/mL, at least about 2 ⁇ g/mL, at least about 3 ⁇ g/mL mL, at least about 4 ⁇ g/mL, at least about 5 ⁇ g/mL, at least about 6 ⁇ g
  • the initial concentration of each immune checkpoint inhibitor of the present application in the cell culture medium of the TIL of the present application is each independently at least about 0.1 ⁇ g/mL to about 20 ⁇ g/mL.
  • the initial concentration of PD-1 antibody in the cell culture medium of the TILs of the present application can be about 0.1 ⁇ g/mL to about 20 ⁇ g/mL, about 0.2 ⁇ g/mL to about 20 ⁇ g/mL, about 0.3 ⁇ g/mL to about 20 ⁇ g/mL, about 0.5 ⁇ g/mL to about 20 ⁇ g/mL, about 0.7 ⁇ g/mL to about 20 ⁇ g/mL, about 1 ⁇ g/mL to about 20 ⁇ g/mL, about 3 ⁇ g/mL to about 20 ⁇ g/mL, about 5 ⁇ g/mL - about 20 ⁇ g/mL, about 7 ⁇ g/mL to about 20 ⁇ g/mL, about 10 ⁇ g/
  • the methods of the present application may further comprise: in at least one stage of the in vitro expansion of the present application, contacting the TIL of the present application with one or more T cell growth factors.
  • a TIL of the present application can be contacted with one or more T cell activators of the present application and with one or more T cells of the present application in a single stage of in vitro expansion of the present application growth factor exposure.
  • the TILs of the present application can be contacted with one or more T cell activators of the present application and with one or more T cell growth factors of the present application.
  • the TIL of the present application can be contacted with one or more T cell activators of the present application and with one or more T cell growth factors of the present application.
  • the TIL of the present application in the third stage in vitro expansion of the present application, can be contacted with one or more T cell activators of the present application and with one or more T cell growth factors of the present application.
  • the TILs of the present application are contacted with the one or more T cell activators of the present application and the one or more T cell growth factors of the present application at substantially the same time in a single stage of the present in vitro expansion .
  • the TIL of the present application in the first stage of the in vitro expansion of the present application, is contacted with the one or more T cell activators of the present application and the one or more T cell growth factors of the present application at substantially the same time.
  • the TIL of the present application is contacted with the one or more T cell activators of the present application and the one or more T cell growth factors of the present application at substantially the same time.
  • the TIL of the present application is contacted with the one or more T cell activators of the present application and the one or more T cell growth factors of the present application at substantially the same time.
  • the T cell growth factor of the present application may be selected from one or more of the following group: IL-2, IL-7, IL-12, IL-15, IL-21, gamma interferon, and their functionally active fragments.
  • the T cell growth factors of the present application may comprise IL-2 and/or functionally active fragments thereof.
  • functionally active fragments of IL-2 can comprise fragments of IL-2 known in the art that can bind to the IL-2 receptor of T cells.
  • contacting a TIL of the present application with one or more T cell growth factors of the present application may comprise adding the T cell growth factors of the present application to the cell culture medium of the TIL of the present application.
  • the initial concentration of the T cell growth factor of the present application in the cell culture medium of the TIL of the present application may be at least about 300 IU/mL.
  • the initial concentration of the IL-2 of the present application in the cell culture medium of the TIL of the present application may be at least about 350 IU/mL, at least about 400 IU/mL, at least about 500 IU/mL, at least about 600 IU/mL, at least about 700 IU/mL, at least about 800 IU/mL, at least about 900 IU/mL, at least about 1000 IU/mL, at least about 1100 IU/mL, at least about 1200 IU/mL, at least about 1300 IU/mL, at least about 1400 IU/mL, at least about About 1500IU/mL, at least about 2000IU/mL, at least about 2500IU/mL, at least about 2600IU/mL, at least about 2700IU/mL, at least about 2800IU/mL, at least about 2900IU/mL, at least about 3000IU/mL, at least about 3100IU /mL, at least about 3200IU/mL
  • the method of the present application may further comprise: in at least one stage of the in vitro expansion of the present application, the TIL of the present application may be co-cultured with feeder cells.
  • the TIL of the application in a single stage of the in vitro expansion of the application, can be contacted with one or more T cell activators and/or one or more T cell growth factors of the application and with The feeder cells of the present application are co-cultured.
  • the in vitro expansion of the present application in a single stage may refer to the in vitro expansion of the present application at the same stage, for example, the in vitro expansion of the present application may be performed at the same stage , can be amplified in vitro in the second stage of the present application, or can be amplified in vitro in the third stage of the present application.
  • the TIL of the present application in the first stage of the in vitro expansion of the present application, can be combined with one or more T cell activators and/or one or more T cell growth factors of the present application contacted and co-cultured with the feeder cells of the present application. In one embodiment, in the second stage of the in vitro expansion of the present application, the TIL of the present application can be combined with one or more T cell activators and/or one or more T cell growth factors of the present application contacted and co-cultured with the feeder cells of the present application.
  • the TIL of the present application in the third stage of the in vitro expansion of the present application, can be combined with one or more T cell activators and/or one or more T cell growth factors of the present application contacted and co-cultured with the feeder cells of the present application.
  • the TILs of the present application may be contacted with one or more T cell activators and/or one or more T cell growth factors of the present application for a period of time in a single stage of the in vitro expansion of the present application After that, it is co-cultured with the feeder cells of the present application.
  • the TIL of the present application in the first stage of the in vitro expansion of the present application, may be contacted with one or more T cell activators and/or one or more T cell growth factors of the present application for a certain period of time After that, it is co-cultured with the feeder cells of the present application.
  • the TILs of the present application may be contacted with one or more T cell activators and/or one or more T cell growth factors of the present application for a certain period of time After that, it is co-cultured with the feeder cells of the present application.
  • the TILs of the present application in the third stage of the in vitro expansion of the present application, may be contacted with one or more T cell activators and/or one or more T cell growth factors of the present application for a certain period of time After that, it is co-cultured with the feeder cells of the present application.
  • the TILs of the present application may be contacted with one or more T cell activators and/or one or more T cell growth factors of the present application for a period of time in a single stage of the in vitro expansion of the present application After that, it is co-cultured with the feeder cells of the present application.
  • the certain time of the present application may be at least about 2 hours.
  • the certain time of the present application can be at least about 1 hour, at least about 2 hours, at least about 3 hours, at least about 4 hours, at least about 5 hours, at least about 6 hours, at least about 7 hours, at least about about 8 hours, at least about 9 hours, at least about 10 hours, at least about 11 hours, at least about 12 hours, at least about 13 hours, at least about 14 hours, at least about 15 hours, at least about 16 hours, at least about 17 hours, at least about about 18 hours, at least about 19 hours, at least about 20 hours, at least about 21 hours, at least about 22 hours, at least about 23 hours, at least about 24 hours, at least about 36 hours, at least about 48 hours, at least about 60 hours, or at least about About 72 hours.
  • the certain period of time of the present application may be from about 6 hours to about 72 hours. In one embodiment, the certain time of the present application may be about 6 hours to about 7 hours, about 6 hours to about 8 hours, about 6 hours to about 9 hours, about 6 hours to about 10 hours, about 6 hours to about 6 hours to About 11 hours, about 6 hours to about 12 hours, about 6 hours to about 13 hours, about 6 hours to about 14 hours, about 6 hours to about 15 hours, about 6 hours to about 16 hours, about 6 hours to about 17 hours hours hours, about 6 hours to about 18 hours, about 6 hours to about 19 hours, about 6 hours to about 20 hours, about 6 hours to about 21 hours, about 6 hours to about 22 hours, about 6 hours to about 23 hours, About 6 hours to about 24 hours, about 6 hours to about 36 hours, about 6 hours to about 48 hours, about 6 hours to about 60 hours, or about 6 hours to about 72 hours.
  • the certain time of the present application may be about 12 hours to about 13 hours, about 12 hours to about 14 hours, about 12 hours to about 15 hours, about 12 hours to about 16 hours, about 12 hours to about 12 hours to About 17 hours, about 12 hours to about 18 hours, about 12 hours to about 19 hours, about 12 hours to about 20 hours, about 12 hours to about 21 hours, about 12 hours to about 22 hours, about 12 hours to about 23 hours hours, about 12 hours to about 24 hours, about 12 hours to about 36 hours, about 12 hours to about 48 hours, about 12 hours to about 60 hours, or about 12 hours to about 72 hours.
  • the certain time of the present application may be about 1 hour, about 2 hours, about 3 hours, about 4 hours, about 5 hours, about 6 hours, about 7 hours, about 8 hours, about 9 hours, about 10 hours, about 11 hours, about 12 hours, about 13 hours, about 14 hours, about 15 hours, about 16 hours, about 17 hours, about 18 hours, about 19 hours, about 20 hours, about 21 hours, about 22 hours hours, about 23 hours, about 24 hours, about 36 hours, about 48 hours, about 60 hours, or about 72 hours.
  • the feeder cells of the present application may comprise antigen presenting cells. In one embodiment, the feeder cells of the present application may comprise one or more selected from the group consisting of peripheral mononuclear cells, dendritic cells, and artificial antigen presenting cells. In one embodiment, the feeder cells of the present application may be peripheral mononuclear cells. In one embodiment, the feeder cells of the present application may be irradiated feeder cells.
  • the feeder cells of the present application can be isolated artificial antigen-presenting cells (aAPCs), and the artificial antigen-presenting cells of the present application can comprise cells expressing HLA-A/B/C, CD64, CD80, ICOS-L and/or CD58 , and can be modified to express more than one T cell activator of the present application.
  • the feeder cells of the present application can be irradiated, for example, can be gamma irradiated, or can be X-ray irradiated.
  • co-culturing the TIL of the present application with the feeder cell of the present application may comprise contacting the surface of the feeder cell of the present application with the surface of the TIL of the present application. In one embodiment, co-culturing the TILs of the present application with the feeder cells of the present application comprises adding the feeder cells of the present application to the cell culture medium of the TILs of the present application.
  • the feeder cells of the present application may be added to the cell culture medium of the TILs of the present application at a ratio of the feeder cells of the present application to the TIL of the present application at a ratio of about 40:1 to about 400:1.
  • the present application may be at about 40:1 to about 400:1, at about 40:1 to about 300:1, at about 40:1 to about 200:1, at about 40:1 to about 100:1, about 40:1-about 90:1, about 40:1-about 80:1, about 40:1-about 70:1, about 40:1-about 60:1, about 40:1-about 50:1, about 50:1-about 400:1, about 60:1-about 400:1, about 70:1-about 400:1, about 80:1-about 400 :1, at about 90:1 to about 400:1, at about 100:1 to about 400:1, at about 200:1 to about 400:1, or at about 300:1 to about 400:1 of the present application
  • the TILs of the present application may be TILs derived from fragments of tumor tissue. In one embodiment, the TILs of the present application can be obtained by processing tumor tissue into tumor fragments. In one embodiment, the tumor fragments of the present application are about 1-27 cubic millimeters in volume.
  • the tumor fragments of the present application have a volume of about 1 cubic millimeter, about 2 cubic millimeters, about 3 cubic millimeters, about 4 cubic millimeters, about 5 cubic millimeters, about 6 cubic millimeters, about 7 cubic millimeters , about 8 mm3, about 9 mm3, about 10 mm3, about 11 mm3, about 12 mm3, about 13 mm3, about 15 mm3, about 17 mm3, about 19 mm3, about 20 mm3 , about 21 mm3, about 23 mm3, about 24 mm3, about 25 mm3, about 26 mm3 or 27 mm3.
  • the present application provides a method for culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) a first population of TILs derived from tumor tissue and not expanded in vitro and T cell growth factors contact; wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be contacted with a T cell growth factor, a T cell activator, and the TIL and feeder cells can be combined Culturing; wherein, the third TIL population is obtained through the step (B); (C) the third TIL population can be contacted with a T cell activator and an immune checkpoint inhibitor, and step (C) can be performed at most About 24 hours.
  • TILs tumor-infiltrating lymphocytes
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 wherein, a second group of TILs is obtained through the step (A); (B) the second group of TILs can be contacted with IL-2, contacted with a T cell activator, and co-cultured with feeder cells; Wherein, the third TIL population is obtained through the step (B); (C) the third TIL population can be contacted with a T cell activator and an immune checkpoint inhibitor, and the step (C) can be performed for up to about 24 Hour.
  • TILs tumor-infiltrating lymphocytes
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be contacted with IL-2, a nanomatrix that can be used with a T cell activator (for example, a CD3 antibody and a CD28 antibody can be included) , CD3 antibody or a mixture of CD3 antibody and CD28 antibody) contact and co-culture the TIL with feeder cells; wherein, the third TIL population is obtained through the step (B); (C) can make the third TIL population Contacting with a T cell activator (eg, a nanomatrix, a CD3 antibody, or a mixture of CD3 and CD28 antibodies, which may comprise CD3 and CD28 antibodies) and with a PD-1 antibody, the heavy chain of which may comprise amino acids
  • TILs
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be contacted with IL-2, a nanomatrix that can be used with a T cell activator (for example, a CD3 antibody and a CD28 antibody can be included) , CD3 antibody or a mixture of CD3 antibody and CD28 antibody) contact and co-culture the TIL with feeder cells; wherein, the third TIL population is obtained through the step (B); (C) can make the third TIL population Contacting with a T cell activator (eg, a nanomatrix, a CD3 antibody, or a mixture of CD3 and CD28 antibodies, which may comprise CD3 and CD28 antibodies) and with a PD-1 antibody, the heavy chain of which may comprise amino acids
  • TILs
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be contacted with IL-2, contacted with a nanomatrix comprising CD3 antibody and CD28 antibody, and the TIL can be contacted with Feeder cell co-cultivation; wherein, a third TIL population is obtained through the step (B); (C) the third TIL population can be contacted with a nanomatrix comprising CD3 antibody and CD28 antibody and with PD-1 antibody,
  • the heavy chain of the PD-1 antibody may comprise the VH of the amino acid sequence shown in any one of SEQ ID NOs: 7, 11 and 21, and the light chain of the PD-1 antibody may comprise the amino acid sequence of SEQ ID NO: : the VL shown in any one of 8, 12 and 22, the initial
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be contacted with IL-2, contacted with a nanomatrix comprising CD3 antibody and CD28 antibody, and in step (B) Co-culturing the TIL with feeder cells after at least about 2 hours, the nanomatrix may have a diameter of about 1 nanometer to about 500 nanometers, and each mg of the nanomatrix may comprise about 25 ⁇ g of each of the CD3 antibody and the CD28 antibody, respectively , the feeder cells may comprise peripheral mononuclear cells; wherein, a third TIL population is obtained through the step (B); (C) the third TIL population may be contacted with a nanomatrix comprising CD3 antibody and CD28 antibody and In contact with the TILs (
  • the chain may comprise HCDR1 having the amino acid sequence set forth in any one of SEQ ID NOs: 1 and 15, HCDR2 having the amino acid sequence set forth in any one of SEQ ID NOs: 2 and 16, and HCDR2 having the amino acid sequence set forth in any one of SEQ ID NO: 3 and the HCDR3 shown in any one of 17, and the light chain of the PD-1 antibody may comprise an amino acid sequence such as the LCDR1 shown in any one of SEQ ID NO: 4 and 18, and the amino acid sequence is such as SEQ ID NO: 5 and LCDR2 shown in any one of 19, and LCDR3 whose amino acid sequence is shown in any one of SEQ ID NOs: 6 and 20,
  • the initial concentration of the PD-1 antibody in the cell culture medium of the TIL can be At least about 0.1 ⁇ g/mL
  • step (C) can be carried out for up to about 24 hours.
  • the application provides a method of culturing tumor-infiltrating lymphocytes (TILs), which may comprise: (A) contacting a first population of TILs derived from tumor tissue and not expanded in vitro with IL-2 , the initial concentration of the IL-2 in the cell culture medium of the TIL can be at least about 300 IU/mL; wherein, the second TIL population is obtained through the step (A); (B) the second TIL population can be made The population of TILs is contacted with IL-2, the TILs are co-cultured with feeder cells, and the TILs are co-cultured with feeder cells after step (B) for at least about 2 hours, with the nanomatrix comprising the CD3 antibody and the CD28 antibody.
  • TILs tumor-infiltrating lymphocytes
  • the initial concentration in the cell culture medium can be at least about 300 IU/mL
  • the diameter of the nanomatrix can be from about 1 nanometer to about 500 nanometers
  • each mg of the nanomatrix can contain about 25 ⁇ g of CD3 antibody and CD28 antibody, respectively can be added to the cell culture medium of the TIL at a ratio of about 1:100 to about 1:2000 of the nanomatrix to the TIL
  • the feeder cells can comprise peripheral mononuclear cells, and can be added at a ratio of about 40:
  • the third population of TILs can be contacted with a nanomatrix comprising CD3 antibody and CD28 antibody and with PD-1 antibody, the nanomatrix can have a diameter of about 1 nanometer to about 500 nanometers, per mg of the nanometer
  • the matrix may comprise about 25 ⁇
  • the present application provides a method of culturing tumor-infiltrating lymphocytes (TILs).
  • TILs tumor-infiltrating lymphocytes
  • the method for obtaining TIL cells from the subject's tissue sample can be that the patient obtains an in situ tumor sample or a metastatic tumor sample through surgery, and the weight can be at least about 1 g, or multiple pieces of tissue can be combined.
  • Tumor tissue is transported in a sample transport solution, such as a commercially commonly used tumor tissue transport solution, tumor tissue preservation solution or tumor tissue transport solution, at about 2-8 degrees, and processed within 48 hours.
  • Tissue pieces can be mechanically disrupted to a size of approximately 1-27 mm3 each, transferred into a gas permeable culture bag or Grex, and added with T cell serum-free medium at a concentration of 300-9000 IU/mL (for example, it can be 1000-9000 IU/mL, For example, 6000 IU/mL) of IL-2 can be cultured for about 3-14 days. Collect the cells in the medium and transfer them into a gas-permeable culture bag, or Grex, or Xuri equipment.
  • the T cell serum-free medium can be supplemented with CD28 antibody, CD3 antibody and CD28 antibody of the present application, and magnetic beads containing CD3 antibody and CD28 antibody (such as Dynabeads) and/or nanomatrixes comprising CD3 and CD28 antibodies (e.g. transACT) and IL-2 at a concentration of 300-9000 IU/mL (e.g. can be 1000-9000 IU/mL, e.g. can be 6000 IU/mL), activate the After a certain period of application, irradiated PBMCs (TIL and PBMCs in a ratio of about 1:40 to about 1:400) were added, and the culture was expanded for about 3-14 days.
  • CD28 antibody CD3 antibody and CD28 antibody of the present application
  • magnetic beads containing CD3 antibody and CD28 antibody such as Dynabeads
  • nanomatrixes comprising CD3 and CD28 antibodies (e.g. transACT) and IL-2 at a concentration of 300-9000 IU/
  • the cells in the medium can be collected using a cell processing system, and the TIL cells of the present application can be cultured with the CD3 antibody of the present application and the PD-1 antibody (eg, can be at least about 0.1 ⁇ g/mL), and optionally the CD28 antibody,
  • the incubation can be carried out for about 12 to about 24 hours. After washing, cryopreservation, and testing.
  • the CD3 ratio of the final product can be greater than 80%, the cell viability can be greater than 70%, and the T cells greater than 80% can be memory effector T cells and effector T cells. IFNy may be secreted upon stimulation, and/or may be characterized by an up-regulated proportion of activated T cells.
  • the present application provides a tumor-infiltrating lymphocyte (TIL), and the TIL of the present application can be cultured according to the culture method of the present application.
  • TIL tumor-infiltrating lymphocyte
  • the TIL provided by the present application may comprise one or one batch of the culturing method of the present application to obtain TIL.
  • the TILs provided by the present application may comprise multiple or multiple batches of TILs cultured by the culturing method of the present application and combined in any ratio.
  • TILs expanded using the methods of the present application can be administered to a patient as a pharmaceutical composition.
  • the pharmaceutical composition may be a suspension of TIL in sterile buffer.
  • TILs expanded using the PBMCs of the present application can be administered by any suitable route known in the art.
  • T cells can be administered as a single intra-arterial or intravenous infusion, which can last about 30 to 60 minutes. Other suitable routes of administration may include intraperitoneal, intrathecal and intralymphatic administration.
  • any suitable dose of TIL can be administered.
  • the tumor is a melanoma
  • from about 2.3 x 109 to about 13.7 x 1010 TILs can be administered.
  • about 1 ⁇ 10 9 to about 12 ⁇ 10 10 TILs can be administered.
  • about 1.2 ⁇ 10 10 to about 4.3 ⁇ 10 10 TILs can be administered.
  • about 3 ⁇ 10 10 to about 12 ⁇ 10 10 TILs can be administered.
  • about 4 ⁇ 10 10 to about 10 ⁇ 10 10 TILs can be administered.
  • about 5 ⁇ 10 10 to about 8 ⁇ 10 10 TILs can be administered.
  • about 6 ⁇ 10 10 to about 8 ⁇ 10 10 TILs can be administered. In some embodiments, about 7 ⁇ 10 10 to about 8 ⁇ 10 10 TILs can be administered. In some embodiments, the therapeutically effective dose may be from about 2.3 ⁇ 10 9 to about 13.7 ⁇ 10 10 . In some embodiments, a therapeutically effective dose may be from about 1 x 109 to about 12 x 1010 TILs. In some embodiments, the therapeutically effective dose may be from about 1.2 ⁇ 10 10 to about 4.3 ⁇ 10 10 TILs. In some embodiments, the therapeutically effective dose may be from about 3 ⁇ 10 10 to about 12 ⁇ 10 10 TILs. In some embodiments, a therapeutically effective dose may be from about 4 ⁇ 10 10 to about 10 ⁇ 10 10 TILs.
  • the therapeutically effective dose may be from about 5x1010 to about 8x1010 TILs. In some embodiments, a therapeutically effective dose may be from about 6 ⁇ 10 10 to about 8 ⁇ 10 10 TILs. In some embodiments, the therapeutically effective dose may be from about 7 ⁇ 10 10 to about 8 ⁇ 10 10 TILs.
  • the amount of TIL provided in the compositions of the present application can be about 1 ⁇ 10 6 , about 2 ⁇ 10 6 , about 3 ⁇ 10 6 , about 4 ⁇ 10 6 , about 5 ⁇ 10 6 , about 6 ⁇ 10 6 , approx. 7 ⁇ 10 6 , approx. 8 ⁇ 10 6 , approx. 9 ⁇ 10 6 , approx. 1 ⁇ 10 7 , approx. 2 ⁇ 10 7 , approx. 3 ⁇ 10 7 , approx. 4 ⁇ 10 7 , approx. 5 ⁇ 10 7 , approx. 6 ⁇ 10 7 , approx. 7 ⁇ 10 7 , approx. 8 ⁇ 10 7 , approx. 9 ⁇ 10 7 , approx. 1 ⁇ 10 8 , approx. 2 ⁇ 10 8 , approx.
  • the amount of TIL provided in the compositions of the present application may range from about 1 ⁇ 10 6 to 5 ⁇ 10 6 , about 5 ⁇ 10 6 to 1 ⁇ 10 7 , about 1 ⁇ 10 7 to 5 ⁇ 10 7 , approx. 5 ⁇ 10 7 to 1 ⁇ 10 8 , approx. 1 ⁇ 10 8 to 5 ⁇ 10 8 , approx. 5 ⁇ 10 8 to 1 ⁇ 10 9 , approx.
  • the concentration of TIL provided in the compositions of the present application may be less than, for example, about 100%, about 90%, about 80%, about 70%, about 60%, about 50%, about 40% of the composition , about 30%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, about 0.5%, about 0.4%, about 0.3%, about 0.2% , about 0.1%, about 0.09%, about 0.08%, about 0.07%, about 0.06%, about 0.05%, about 0.04%, about 0.03%, about 0.02%, about 0.01%, about 0.009%, about 0.008%, about 0.007%, about 0.006%, about 0.005%, about 0.004%, about 0.003%, about 0.002%, about 0.001%, about 0.0009%, about 0.0008%, about
  • the concentration of TIL provided in the composition of the present application can be greater than about 90%, about 80%, about 70%, about 60%, about 50%, about 40%, about 30%, about 20%, about 19.75%, about 19.50%, about 19.25%, about 19%, about 18.75%, about 18.50%, about 18.25%, about 18%, about 17.75%, about 17.50%, about 17.25%, about 17 %, about 16.75%, about 16.50%, about 16.25%, about 16%, about 15.75%, about 15.50%, about 15.25%, about 15%, about 14.75%, about 14.50%, about 14.25%, about 14%, about 13.75%, about 13.50%, about 13.25%, about 13%, about 12.75%, about 12.50%, about 12.25%, about 12%, about 11.75%, about 11.50%, about 11.25%, about 11%, about 10.75 %, about 10.50%, about 10.25%, about 10%, about 9.75%, about 9.50%, about 9.25%, about 9%
  • the TILs provided in the compositions of the present application may be provided in concentrations ranging from about 0.0001% to about 50%, about 0.001% to about 40%, about 0.01% to about 30%, about 0.02% of the composition to about 29%, about 0.03% to about 28%, about 0.04% to about 27%, about 0.05% to about 26%, about 0.06% to about 25%, about 0.07% to about 24%, about 0.08% to about 23%, about 0.09% to about 22%, about 0.1% to about 21%, about 0.2% to about 20%, about 0.3% to about 19%, about 0.4% to about 18%, about 0.5% to about 17% , about 0.6% to about 16%, about 0.7% to about 15%, about 0.8% to about 14%, about 0.9% to about 12%, or about 1% to about 10% w/w, w/v, or v /v.
  • the TILs provided in the compositions of the present application may be provided in concentrations ranging from about 0.001% to about 10%, about 0.01% to about 5%, about 0.02% to about 4.5%, about 0.03% of the composition to about 4%, about 0.04% to about 3.5%, about 0.05% to about 3%, about 0.06% to about 2.5%, about 0.07% to about 2%, about 0.08% to about 1.5%, about 0.09% to about 1%, or from about 0.1% to about 0.9% w/w, w/v or v/v.
  • the amount of TIL provided in the compositions of the present application may be equal to or less than about 10 g, about 9.5 g, about 9.0 g, about 8.5 g, about 8.0 g, about 7.5 g, about 7.0 g, about 6.5 g g, about 6.0g, about 5.5g, about 5.0g, about 4.5g, about 4.0g, about 3.5g, about 3.0g, about 2.5g, about 2.0g, about 1.5g, about 1.0g, about 0.95g, about 0.9g, about 0.85g, about 0.8g, about 0.75g, about 0.7g, about 0.65g, about 0.6g, about 0.55g, about 0.5g, about 0.45g, about 0.4g, about 0.35g, about 0.3 g, about 0.25g, about 0.2g, about 0.15g, about 0.1g, about 0.09g, about 0.08g, about 0.07g, about 0.06g, about 0.05g, about 0.04g, about 0.03g, about 0.
  • the amount of TIL provided in the compositions of the present application can be greater than about 0.0001 g, about 0.0002 g, about 0.0003 g, about 0.0004 g, about 0.0005 g, about 0.0006 g, about 0.0007 g, about 0.0008 g , about 0.0009g, about 0.001g, about 0.0015g, about 0.002g, about 0.0025g, about 0.003g, about 0.0035g, about 0.004g, about 0.0045g, about 0.005g, about 0.0055g, about 0.006g, about 0.0065g, about 0.007g, about 0.0075g, about 0.008g, about 0.0085g, about 0.009g, about 0.0095g, about 0.01g, about 0.015g, about 0.02g, about 0.025g, about 0.03g, about 0.035g , about 0.04g, about 0.045g, about 0.05g, about 0.055g, about 0.06g,
  • the TIL can be administered in a single dose. Such administration can be by injection, eg, intravenous injection. In some embodiments, the TIL can be administered in multiple doses. The dosage can be one, two, three, four, five, six or more than six times per year. Dosing can be once a month, once every two weeks, once a week or once every 2 days. In some embodiments, the administration of TIL can be continuous.
  • the application provides a pharmaceutical composition.
  • it may comprise a TIL of the present application and/or a composition of the present application, together with a pharmaceutically acceptable carrier.
  • the present application provides a kit, which can include T cell activators, T cell growth factors and/or feeder cells used in the method for culturing tumor-infiltrating lymphocytes (TILs) of the present application and a method for culturing tumors described in the present application. Instructions for the steps of the infiltrating lymphocyte (TIL) method.
  • the present application provides a kit, and the kit of the present application may comprise the TIL of the present application and/or the pharmaceutical composition of the present application.
  • the present application provides a method of affecting tumor cell growth, which can include administering to a subject a TIL of the present application and/or a pharmaceutical composition of the present application.
  • affecting tumor growth can comprise a reduction in the volume of the tumor to, for example, about 99%, about 95%, about 90%, about 80%, about 70%, about 60%, about 50%, about 40% of the volume before administration %, about 30%, about 20%, about 19%, about 18%, about 17%, about 16%, about 15%, about 14%, about 13%, about 12%, about 11%, about 10%, about 9%, about 8%, about 7%, about 6%, about 5%, about 4%, about 3%, about 2%, about 1%, about 0.5%, about 0.4%, about 0.3%, about 0.2 % or about 0.1%.
  • the present application provides the application of the TIL of the present application and/or the pharmaceutical composition of the present application in the preparation of a medicament, and the medicament of the present application can be used for the prevention and/or treatment of tumors.
  • the tumors of the present application are selected from solid tumors.
  • the tumor of the present application can be selected from one or more of the following group: melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, gastric cancer, Rectal cancer, and kidney cancer.
  • the present application provides a method of preventing and/or treating tumors, which may include administering the TIL of the present application and/or the pharmaceutical composition of the present application to a subject.
  • the tumors of the present application are selected from solid tumors.
  • the tumor of the present application can be selected from one or more of the following group: melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, gastric cancer, Rectal cancer, and kidney cancer.
  • the present application provides a TIL of the present application and/or a pharmaceutical composition of the present application, which can be used for preventing and/or treating tumors.
  • the tumors of the present application are selected from solid tumors.
  • the tumor of the present application can be selected from one or more of the following group: melanoma, ovarian cancer, cervical cancer, lung cancer, bladder cancer, breast cancer, head and neck cancer, pancreatic cancer, liver cancer, gastric cancer, Rectal cancer, and kidney cancer.
  • TIL tumor infiltrating lymphocyte
  • PBMC peripheral blood mononuclear cells
  • erythrocytes can be split after centrifugation. Add erythrocyte lysate at 1:2 to 1:3 of the cell sediment volume and erythrocyte lysate, mix well, lyse at room temperature for 10 minutes, and gently mix the centrifuge tube for 2- 3 times to ensure the lysis effect. After the lysis is completed, add PBS or normal saline to wash the cells. After splitting, the cells were washed twice, centrifuged at 400g for 6 minutes, and the samples were counted before the last centrifugation.
  • a pre-concentration step can be performed to concentrate the blood volume to within 120mL.
  • the neatcell program can be used to separate and wash PBMC.
  • the washing solution is normal saline, and the intermediate volume is 20 mL; the resuspended solution is the basal medium, and 80 mL/batch is added.
  • each donor PBMC is a bag of 100mL.
  • the liquid level can be no more than 1 cm, and the X-ray irradiation is 50-200Gy.
  • the cells were sampled and counted, and the cells were collected and washed three times using the culture wash program.
  • the washing solution was normal saline; the intermediate volume and final volume were set so that each 1 ⁇ 10 9 cells was no less than 2 mL; add an equal amount to 2 times for cryopreservation liquid mix. Adjust the cell density to about 1 ⁇ 10 7 cells/mL to 2 ⁇ 10 8 cells/mL with 1-fold freezing solution, divide into 20 mL/bags, freeze in a programmed cooling device, and store in liquid nitrogen.
  • the PBMC cells in the blood samples were isolated and cryopreserved according to the above procedures for manual isolation and cryopreservation of PBMC. Take a culture bottle or culture bag with a gas permeable surface, such as a culture bag (Origen), and add 300 mL of rewarmed complete medium.
  • the complete medium can optionally be X-vivo15 medium or other commercial T cell culture medium.
  • T cell culture medium such as Stem Cell, Lonza, Thermo, Miltenyi and other brands of T cell culture medium, and can add essential amino acids and antibiotics, and add a concentration of 300-9000IU/mL (for example, it can be 1000-9000IU/mL, for example, it can be 6000 IU/mL) of IL-2.
  • 300-9000IU/mL for example, it can be 1000-9000IU/mL, for example, it can be 6000 IU/mL
  • IL-2 a concentration of 300-9000IU/mL (for example, it can be 1000-9000IU/mL, for example, it can be 6000 IU/mL) of IL-2.
  • ophthalmic scissors and ophthalmic forceps to perform preliminary cutting to remove fatty tissue and necrotic tissue, and continue to cut each tissue block to a size of about 27 cubic millimeters.
  • take the non-suspended tumor tissue block use a 20 mL syringe to remove the inner piston, connect it to the culture bag, and use a pipette to transfer about 1 g of the tissue block into the culture bag through the syringe. Place the culture bag in a carbon dioxide incubator for cultivation.
  • the scissors and tweezers were cleaned, and after preliminary disinfection with 75% alcohol, ultrasonic cleaning was performed, and the first TIL group was obtained.
  • preREP stage The first stage of in vitro expansion and harvest
  • the medium is replenished or half-changed every 3-7 days to ensure cell nutrition.
  • Use complete medium X-vivo 15 medium or other commercial T cell medium, such as Stem Cell, Lonza, Thermo, Miltenyi and other brands of T cell medium, can be optionally added to the complete medium.
  • samples can be counted on the 3rd, 4th, 5th, 6th, 7th, 8th, 9th, 10th, 11th, 12th, 13th or 14th day, if the number of cells is 5 ⁇ 10 5 When it reaches 5 ⁇ 10 8 , the following first-stage in vitro amplification harvesting step is entered.
  • TIL second TIL population
  • sample and count about 5 ⁇ 10 5 to 2 ⁇ 10 8 cells enter the following first-stage in vitro expansion step; about 5 ⁇ 10 5 cells can be taken for quality control testing; the rest of the cells are added to cryopreservation solution for cryopreservation.
  • T cell medium Take 5 ⁇ 10 5 to 2 ⁇ 10 8 cells expanded in vitro in the first stage, and use complete medium, X-vivo 15 medium or other commercial T cell medium, such as Stem, can be used as the complete medium.
  • Cell, Lonza, Thermo, Miltenyi and other brands of T cell culture medium, and essential amino acids and antibiotics can be added to adjust the cell density to 5 ⁇ 10 5 to 2 ⁇ 10 6 cells/mL, suspended in a 24-well culture plate , 1mL/well, add IL-2 at a concentration of 300-9000IU/mL (for example, it can be 1000-9000IU/mL, for example, it can be 6000IU/mL).
  • a CD3 agonist and/or a CD28 agonist can be added, eg, OKT3 at about 30 ng/mL, CD28 antibody at about 30 ng/mL, magnetic beads at about 1:2-2:1 Magnetic beads (approximately 1 to 10 ⁇ m in diameter Dynabeads, Thermo Fisher) and/or transACT (approximately 100 to 500 nm in diameter, Miltenyi) to TIL were added at a ratio to TIL and/or at a ratio of approximately 1:100-1:2000.
  • Tn can be 0 hours to 14 days, such as 24 hours or 48 hours
  • Tn can be 0 hours to 14 days, such as 24 hours or 48 hours
  • Donor mixed feeder cells transfer activated TIL cells and feeder cells into G-Rex100 culture flasks or gas-permeable bags, supplement complete medium, sample and count every 1-3 days, and refill or change the medium in half amount according to the cell status until When the total number of cells is greater than 1 ⁇ 10 9 or the second-stage in vitro expansion culture reaches 13 days, the second-stage in vitro expansion culture is terminated.
  • TIL third TIL population expanded in vitro in the second stage.
  • Samples were counted during the three washings. According to the counting results, the supernatant was discarded after the last centrifugation, and 3 ⁇ 10 6 cells were taken for quality control testing; all the remaining cells were added to the freezing solution, and the cell density was adjusted to 1-3 ⁇ 10 8 cells/ mL cryopreserved.
  • the resuscitated TILs can optionally be relieved for a certain period of time, such as about 1 hour, about 2 hours, about 6 hours, about 12 hours. hours, about 1 day, about 2 days, or about 4 days.
  • Use complete medium to adjust cell density X-vivo 15 medium or other commercial T cell medium, such as Stem Cell, Lonza, Thermo, Miltenyi and other brands of T cell medium, can be used as the complete medium.
  • Control group add PBS or normal saline
  • PD-1 Antibody Panel Add at least about 0.1 ⁇ g/mL (eg, about 1 ⁇ g/mL or about 10 ⁇ g/mL) of PD-1 antibody (eg, pembrolizumab, MSD, or 6H6, full-length heavy chain as SEQ ID NO: 9) and 13, and the full length of the light chain is shown in any of SEQ ID NOs: 10 and 14);
  • PD-1 antibody eg, pembrolizumab, MSD, or 6H6, full-length heavy chain as SEQ ID NO: 9 and 13
  • SEQ ID NOs: 10 and 14 Add at least about 0.1 ⁇ g/mL (eg, about 1 ⁇ g/mL or about 10 ⁇ g/mL) of PD-1 antibody (eg, pembrolizumab, MSD, or 6H6, full-length heavy chain as SEQ ID NO: 9) and 13, and the full length of the light chain is shown in any of SEQ ID NOs: 10 and 14);
  • Nanomatrix group transACT (about 100 to 500 nm in diameter, Miltenyi) was added at a ratio of transACT to TIL of about 1:100-1:2000;
  • Nanomatrix + PD-1 antibody group add transACT at a ratio of transACT to TIL of about 1:100-1:2000, and add PD at least about 0.1 ⁇ g/mL (eg, about 1 ⁇ g/mL or about 10 ⁇ g/mL) -1 antibody (e.g., pembrolizumab, MSD, or 6H6, the full length of the heavy chain is set forth in any one of SEQ ID NOs: 9 and 13, and the full length of the light chain is set forth in any one of SEQ ID NOs: 10 and 14 );
  • Nanomatrix + PD-1 Antibody Group A Add transACT at a ratio of transACT to TIL of about 1:100-1:2000, and add at least about 0.1 ⁇ g/mL (eg, about 1 ⁇ g/mL or about 10 ⁇ g/mL) PD-1 antibody A (pembrolizumab, MSD);
  • Nanomatrix + PD-1 Antibody Group B TransACT is added at a ratio of transACT to TIL of about 1:100-1:2000, and at least about 0.1 ⁇ g/mL (eg, about 1 ⁇ g/mL or about 10 ⁇ g/mL) PD-1 Antibody B (full length of the heavy chain is set forth in any one of SEQ ID NOs: 9 and 13, and full length of the light chain is set forth in any one of SEQ ID NOs: 10 and 14);
  • the fourth TIL population was harvested and counted at about 12 hours to about 24 hours in the third stage of in vitro expansion, and the culture of the third stage of in vitro expansion was terminated.
  • the harvested therapeutic tumor-infiltrating lymphocytes can be administered to the subject by intravenous infusion.
  • Figures 1A-1B show the intracellular factor expression ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results showed that compared with the third stage of in vitro expansion culture, no PD-1 antibody and T cell activator (control group), only PD-1 antibody (PD-1 antibody group) and only T cell activator were added.
  • PD-1 antibody and T cell activator no PD-1 antibody and T cell activator (Nanomatrix group)
  • adding PD-1 antibody and T cell activator nanomatrix + PD-1 antibody group
  • PD-1 antibody and T cell activator had a significant synergistic effect on the enhancement of intracellular factor expression capacity.
  • Figures 2A-2B show the intracellular factor expression ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results show that, compared with the third stage of in vitro expansion culture without the addition of PD-1 antibody and T cell activator (control group), different types of PD-1 antibodies and T cells were added at the same time in the third stage of in vitro expansion culture.
  • Activators Nanomatrix + PD-1 Antibody Group A or Nanomatrix + PD-1 Antibody Group B
  • had similar increases in intracellular factor expression eg, higher CD107a expression.
  • different combinations of PD-1 antibodies and T cell activators have a significant enhancement effect on the expression of intracellular factors.
  • Cytokine secretion detection was performed on the fourth TIL population obtained by the third-stage in vitro expansion culture of each test group in different ways in Example 1.
  • BD Human Th1/Th2/Th17 cytokine standard lyophilized powder
  • 2mL Assay Diluent Diluent (BD) the concentration of each cytokine in the standard solution was 5000pg/mL
  • 1:2, 1 :4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1024 serial dilution then mixed with Capture Beads (BD) and detected by PE Detection Reagent Reagents (BD) were mixed and transferred to a 15mL conical bottom centrifuge tube, labeled "Standard Tube". Take 1 tube containing Assay Diluent Diluent only as a negative control.
  • Figures 3A-3C show the detection results of IL-2 secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results show that, compared with the third stage of in vitro expansion culture without adding PD-1 antibody and T cell activator (control group) and only adding T cell activator (nanomatrix group), in the third stage of in vitro expansion culture Simultaneous addition of PD-1 antibody and T cell activator (nanomatrix + PD-1 antibody group) has higher cytokine secretion capacity, for example, higher IL-2 secretion capacity.
  • Figures 4A-4C show the detection results of TNF secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results show that, compared with the third stage of in vitro expansion culture without adding PD-1 antibody and T cell activator (control group) and only adding T cell activator (nanomatrix group), in the third stage of in vitro expansion culture Simultaneous addition of PD-1 antibody and T cell activator (nanomatrix + PD-1 antibody group) has higher cytokine secretion capacity, for example, higher TNF secretion capacity.
  • Figures 5A-5C show the detection results of IFN ⁇ secretion of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results show that, compared with the third stage of in vitro expansion culture without adding PD-1 antibody and T cell activator (control group) and only adding T cell activator (nanomatrix group), in the third stage of in vitro expansion culture Simultaneous addition of PD-1 antibody and T cell activator (nanomatrix + PD-1 antibody group) has higher cytokine secretion capacity, for example, higher IFN ⁇ secretion capacity.
  • the cell killing ability was tested for the fourth TIL population obtained by the third-stage in vitro expansion culture of each test group in different ways in Example 1.
  • TIL obtained from each experimental group for assay and target cells (eg, Hela tumor cells) for co-culture.
  • target cells eg, Hela tumor cells
  • CFSE (6(6)-Carboxyfluorescein diacetate N-succinimidyl ester, Sigma, 21888-25MG-F): wash tumor cells with PBS, resuspend tumor cells in 500 ⁇ L of PBS; add CFSE to 500 ⁇ L of PBS , mixed with 500 ⁇ L of tumor cell PBS resuspension to a final concentration of CFSE of 0.5 ⁇ mol/L. After incubation at 37°C for 6 minutes, add medium containing 10% FBS to wash, centrifuge at 600g for 5 minutes, use X-vivo 15 medium or other commercial T cell medium, such as Stem Cell, Lonza, Thermo, Miltenyi, etc.
  • TIL cells Resuspend tumor cells in branded T cell medium at a concentration of 5 x 10 5 cells/mL.
  • the TIL cells in each test group were centrifuged at 600 g for 5 minutes, and the TIL cells were resuspended according to the effect-target ratio (ratio of TIL cells to tumor cells) 3:1 (that is, the concentration of resuspended TIL cells was 1.5 ⁇ 10 6 cells/mL).
  • 100 ⁇ L each of tumor cells and TIL cells were added to a U-bottom 96-well plate (Corning), and three replicate wells were set in each group.
  • a control group containing only tumor cells was set up, and different reagents were added according to different groups in the experiment. Centrifuge the plate at 200g for 1 minute and incubate at 37°C for 4 hours to overnight.
  • Killing rate % Dapi + CFSE + cell number/total CFSE + ⁇ 100%.
  • Figures 6A-6B show the detection results of the cell killing ability of the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways for different donors.
  • the results show that, compared with the third stage of in vitro expansion culture without the addition of PD-1 antibody and T cell activator (control group), different types of PD-1 antibodies and T cells were added at the same time in the third stage of in vitro expansion culture.
  • the activators nanomatrix + PD-1 antibody group A or nanomatrix + PD-1 antibody group B
  • different combinations of PD-1 antibodies and T cell activators all had a significant enhancement effect on cell killing.
  • CD3 antibody group add CD3 antibody, such as OKT3 about 30ng/mL;
  • CD3 antibody + PD-1 antibody group M add CD3 antibody, such as OKT3 about 30ng/mL, and about 1 ⁇ g/mL PD-1 antibody (such as pembrolizumab, MSD, or 6H6, heavy chain full length as SEQ ID NO: 9 and 13, and the full length of the light chain is shown in any of SEQ ID NOs: 10 and 14);
  • CD3 antibody + PD-1 antibody group N Add CD3 antibody, such as OKT3 about 30 ng/mL, and about 10 ⁇ g/mL PD-1 antibody (such as pembrolizumab, MSD, or 6H6, heavy chain full length such as SEQ ID NO: 9 and 13, and the full length of the light chain is shown in any of SEQ ID NOs: 10 and 14);
  • CD3 antibody such as OKT3 about 30 ng/mL, and about 10 ⁇ g/mL PD-1 antibody (such as pembrolizumab, MSD, or 6H6, heavy chain full length such as SEQ ID NO: 9 and 13, and the full length of the light chain is shown in any of SEQ ID NOs: 10 and 14);
  • Figures 7A-7C show that, for different donors, when CD3 antibody was added in the second stage of in vitro expansion (REP stage), different concentrations of PD-1 antibody were additionally added, compared with the proliferation of TIL cells with only CD3 antibody added.
  • the numerical values of the ordinates in Figures 7A-7C indicate that the number of TIL cells in the TIL cells at each time point obtained by the second stage of in vitro expansion in each experimental group was expanded compared to the second TIL population before the start of the second stage of in vitro expansion. to the amplification fold.
  • the results showed that by adding different concentrations of PD-1 antibody in the second stage of in vitro expansion (REP stage), there was no obvious advantage in the proliferative capacity of TILs obtained.
  • CD3 antibody group add CD3 antibody, such as OKT3 about 30ng/mL;
  • CD3 antibody + PD1 (PD-1) antibody group add CD3 antibody, such as OKT3 about 30 ng/mL, and about 10 ⁇ g/mL PD-1 antibody (such as pembrolizumab, MSD, or 6H6, full-length heavy chain such as SEQ ID NO : shown in any one of 9 and 13, and the full length of the light chain is shown in any one of SEQ ID NOs: 10 and 14); each of the above groups, on the 6th day after the start of the second stage in vitro amplification, Harvested and sampled on days 10 and 13.
  • CD3 antibody such as OKT3 about 30 ng/mL
  • 10 ⁇ g/mL PD-1 antibody such as pembrolizumab, MSD, or 6H6, full-length heavy chain such as SEQ ID NO : shown in any one of 9 and 13, and the full length of the light chain is shown in any one of SEQ ID NOs: 10 and 14
  • each of the above groups on the 6th day after the start of the second stage in vitro
  • Figures 8A-8B show, for different donors, the proliferation of TIL cells with the addition of CD3 antibody in the second stage of in vitro expansion (REP stage) when the CD3 antibody was added, compared to the TIL cells that were only added with the CD3 antibody.
  • the results showed that the additional addition of PD-1 antibody in the second stage of in vitro expansion (REP stage) did not have a significant advantage in the proliferative capacity of TILs obtained.
  • FIGS 9A-9B show the cell viability and cell typing of TILs with CD3 antibody added in the second stage of in vitro expansion (REP stage) with the addition of CD3 antibody, compared to TILs with only CD3 antibody.
  • the results showed that the addition of PD-1 antibody in the second stage of in vitro expansion (REP stage) had no obvious advantage in the cell viability and CD3 + ratio of the obtained TILs.
  • Figures 10A-10B show the proportion of activation-related (41BB + ) cells in TILs with CD3 antibody added when CD3 antibody was added in the second stage of in vitro expansion (REP stage).
  • Figures 11A-11C show that the addition of PD-1 antibody when CD3 antibody is added during the second phase of in vitro expansion (REP phase) correlates with activation (CD25 + and/or CD27 + ) in TIL compared to CD3 antibody only cell ratio.
  • Figures 12A-12B show the proportion of depletion-related (TIM3 + ) cells in TILs with CD3 antibody added when CD3 antibody was added in the second stage of in vitro expansion (REP stage). The results showed that the addition of PD-1 antibody in the second stage of in vitro expansion (REP stage) had no obvious advantage in the proportion of depletion-related cells obtained by TIL.
  • FIGS 13A-13B show the proportion of TNF- ⁇ secreting cells in TILs with only CD3 antibody added when the CD3 antibody was added in the second stage of in vitro expansion (REP stage).
  • Figures 14A-14D show the proportion of IFN- ⁇ -secreting cells in TILs with CD3 antibody added in the second stage of in vitro expansion (REP stage) when the CD3 antibody was added additionally, compared to the TIL with only CD3 antibody added.
  • Figures 15A-15C show that when CD3 antibody was added in the second phase of in vitro expansion (REP phase), the addition of PD-1 antibody compared to the cytokines (IL-2 and/or IL-2 and/or IL- 4) The amount of secretion.
  • Figures 16A-16C show that when CD3 antibody was added in the second phase of in vitro expansion (REP phase), the addition of PD-1 antibody compared to the cytokines (IL-17 and/or IL-17 and/or IL- 6) The amount of secretion.
  • Figures 17A-17D show that the addition of PD-1 antibody when CD3 antibody was added during the second phase of in vitro expansion (REP phase) compared to cytokines (TNF- ⁇ and/or IFN- ⁇ ) The amount of secretion.
  • Example 7 In vitro amplification and detection of TIL culture effect with PD1 antibody addition in the third stage
  • the fourth TIL population obtained by the third-stage in vitro expansion culture in different ways of each test group in Example 1 was detected.
  • transACT Miltenyi
  • an additional 20 ⁇ g/mL of PD-1 antibody was used for stimulation to detect cytokine secretion capacity.
  • Figures 18A-18D show the secretion of cytokines (IL-4 and/or TNF- ⁇ ) of TILs stimulated by adding only transACT after the addition of PD-1 antibody for stimulation at the end of the REP stage .
  • Figures 18E-18F show the secretion of cytokines (IL-2 and/or IL-6) of TILs stimulated by adding only transACT after the addition of PD-1 antibody for stimulation at the end of the REP stage .
  • Example 8 Detection of the effect of TIL culture with the addition of PD1 antibody in the second stage of in vitro expansion (REP stage) and the addition of PD1 antibody in the third stage of in vitro expansion
  • the PD1 (PD-1) antibody was added to culture, and after the second stage in vitro expansion (REP stage) culture, nanomatrix (transACT, Miltenyi) or Additional PD-1 antibody (2 ⁇ g/mL or 20 ⁇ g/mL) was added for stimulation to detect cytokine secretion ability.
  • Figures 19A-19D show the secretion of cytokines (IL-6 and/or TNF- ⁇ ) of TILs stimulated by adding PD-1 antibody only after the REP stage culture .
  • Figures 20A-20C show that after the end of the REP phase of culture, additional PD-1 antibody for stimulation, compared to only transACT stimulation of TIL cytokines (IL-2, IL-4 and/or IFN- ⁇ ) ) secretion volume.
  • TIL cytokines IL-2, IL-4 and/or IFN- ⁇

Abstract

涉及肿瘤浸润淋巴细胞(TIL)的制备方法及其用途,具体涉及一种培养肿瘤浸润淋巴细胞的方法,其包含:使源自肿瘤组织且未经体外扩增的TIL经过至少一个阶段的体外扩增,其中,在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。还涉及使用肿瘤浸润淋巴细胞预防和/或***的方法。

Description

肿瘤浸润淋巴细胞的制备方法及其用途 技术领域
本申请涉及生物医药领域,具体涉及肿瘤浸润淋巴细胞的制备方法及其用途。
背景技术
使用过继性自体转移肿瘤浸润淋巴细胞***是一种治疗预后不良患者的有效方法。但是过继性自体转移肿瘤浸润淋巴细胞***需要大量的肿瘤浸润淋巴细胞。因此如何提供一种稳健可靠的肿瘤浸润淋巴细胞的培养方法是亟待解决的问题。
发明内容
本申请提供了一种肿瘤浸润淋巴细胞的培养方法,所述培养方法具有选自以下组的一种或多种的效果:使TIL细胞的数量改善,使TIL细胞的分泌能力提高,使TIL细胞的杀伤能力提高,使NK细胞比例增加,改变TIL细胞的比例,使CD4 +细胞的比例增加,使CD8 +细胞的比例降低,使中心记忆T细胞比例增加,使调节性T细胞的比例降低,使活化T细胞比例增加,使肿瘤特异性T细胞比例增加,和使干细胞样T细胞比例增加。
本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其包含:使源自肿瘤组织且未经体外扩增的TIL经过至少一个阶段的体外扩增,在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
在一种实施方式中,使所述源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在所述第三阶段体外扩增中,使经所述第二阶段体外扩增的TIL与所述一种或多种T细胞激活剂以及所述免疫检查点抑制剂接触。
在一种实施方式中,在单个阶段的所述体外扩增中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触。
在一种实施方式中,所述第三阶段体外扩增进行至多约24小时。
在一种实施方式中,所述第三阶段体外扩增进行约12小时至约24小时。
在一种实施方式中,与在体外扩增阶段未曾与所述T细胞激活剂和/或所述免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与所述一种或多种T细胞激活剂以及 所述一种或多种免疫检查点抑制剂接触过的TIL显示出改善的扩增效果。
在一种实施方式中,所述改善的扩增效果包含选自以下组的一种或多种:增加的TIL细胞数量,改善的T细胞亚群比例,提高的细胞因子分泌能力,和提高的肿瘤细胞杀伤能力。
在一种实施方式中,所述改善的T细胞亚群比例包含选自以下组的一种或多种:增加的中心记忆T细胞比例,降低的调节性T细胞的比例,增加的活化T细胞比例,增加的肿瘤特异性T细胞比例,和增加的干细胞样T细胞比例。
在一种实施方式中,所述方法还包含:在至少一个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂接触。
在一种实施方式中,所述一种或多种T细胞激活剂包含选自以下组的一种或多种:CD80、CD86、B7-H3、4-1BBL、CD27、CD30、CD134、B7h、CD40、LIGHT、以及它们的功能活性片段。
在一种实施方式中,所述一种或多种T细胞激活剂包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD3激动剂和/或CD28激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD3激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含抗CD3的抗体和/或其抗原结合片段。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD28激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含抗CD28的抗体和/或其抗原结合片段。
在一种实施方式中,所述使TIL与所述一种或多种T细胞激活剂接触包含选自以下组的一种或多种方式:(1)将所述一种或多种T细胞激活剂添加至所述TIL的细胞培养基中;(2)将表达所述一种或多种T细胞激活剂的工程化细胞添加至所述TIL的细胞培养基中;(3)将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为至少约30ng/mL。
在一种实施方式中,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为约30ng/mL-约300ng/mL。
在一种实施方式中,所述固相介质的直径为约500纳米至约10微米。
在一种实施方式中,所述固相介质的直径为约1纳米至约500纳米。
在一种实施方式中,所述固相介质的直径通过透射电子显微镜测量。
在一种实施方式中,所述固相介质包含聚合物。
在一种实施方式中,每mg所述固相介质中包含的每一种所述T细胞激活剂的量各自独立地至少为约25μg。
在一种实施方式中,以约2:1-约1:2的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,以约1:100-约1:2000的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,所述免疫检查点抑制剂包含抑制PD-1与PD-L1和/或PD-L2相互作用的物质。
在一种实施方式中,所述免疫检查点抑制剂包含PD-1抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含PD-1的抗体和/或其抗原结合片段。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约100pM或更低的K D值结合PD-1的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约100pM或更低的EC 50值结合PD-1的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约1nM或更低的IC 50值抑制PD-1与PD-L1和/或PD-L2结合的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR3,且所述HCDR3包含SEQ ID NO:3和17中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR2,且所述HCDR2包含SEQ ID NO:2和16中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR1,且所述HCDR1包含SEQ ID NO:1和15中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR3,且所述LCDR3包含SEQ ID NO:6和20中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR2,且所述LCDR2包含SEQ ID NO:5和19中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR1,且所述LCDR1包含SEQ ID NO:4和18中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含VH,且所述VH包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含VL,且所述VL包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂选自以下组:(1)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:1和15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述轻链包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3;(2)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述轻链包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL。
在一种实施方式中,其中所述抗体选自:嵌合抗体,人源化抗体和全人源抗体。
在一种实施方式中,其中所述抗原结合片段选自:Fab,Fab′,Fv片段,F(ab') 2,F(ab) 2,scFv,di-scFv,VHH和dAb。
在一种实施方式中,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL。
在一种实施方式中,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地为约0.1μg/mL-约20μg/mL。
在一种实施方式中,其还包含:在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞生长因子接触。
在一种实施方式中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂且与所述一种或多种T细胞生长因子接触。
在一种实施方式中,在单个阶段的所述体外扩增中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种T细胞生长因子接触。
在一种实施方式中,所述一种或多种T细胞生长因子选自以下组的一种或多种:IL-2、IL-7、IL-12、IL-15、IL-21、γ干扰素、以及它们的功能活性片段。
在一种实施方式中,所述一种或多种T细胞生长因子包含IL-2和/或其功能活性片段。
在一种实施方式中,所述TIL与所述一种或多种T细胞生长因子接触包含将所述T细胞生长因子添加至所述TIL的细胞培养基中。
在一种实施方式中,每一种所述T细胞生长因子在所述TIL的细胞培养基中的初始浓度各自独立地为至少约300IU/mL。
在一种实施方式中,其还包含:在至少一个阶段的所述体外扩增中,使所述TIL与饲养细胞共培养。
在一种实施方式中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触且与所述饲养细胞共培养。
在一种实施方式中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触一定时间之后,再与所述饲养细胞共培养。
在一种实施方式中,所述一定时间为至少约2小时。
在一种实施方式中,所述一定时间为约6小时至约72小时。
在一种实施方式中,所述一定时间为约12小时至约48小时。
在一种实施方式中,所述一定时间为约6小时、约12小时、约24小时、约48小时、或约72小时。
在一种实施方式中,所述饲养细胞包含抗原呈递细胞。
在一种实施方式中,所述饲养细胞包含选自以下组的一种或多种:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。
在一种实施方式中,所述饲养细胞为外周单个核细胞。
在一种实施方式中,所述饲养细胞为经过辐照的饲养细胞。
在一种实施方式中,所述TIL与所述饲养细胞共培养包含使所述饲养细胞的表面与所述TIL的表面相接触。
在一种实施方式中,所述TIL与所述饲养细胞共培养包含将所述饲养细胞添加至所述TIL的细胞培养基中。
在一种实施方式中,以约40:1-约400:1的所述饲养细胞与所述TIL的比例,将所述饲养细胞添加至所述TIL的细胞培养基中。
在一种实施方式中,所述源自肿瘤组织且未经体外扩增的TIL为源自所述肿瘤组织的碎片的TIL。
在一种实施方式中,所述碎片的体积为约1立方毫米至约27立方毫米。
本申请还提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其包含:
(A)使源自肿瘤组织且未经体外扩增的第一TIL群与一种或多种T细胞生长因子接触;经所述步骤(A)得到第二TIL群;
(B)使所述第二TIL群与所述一种或多种T细胞生长因子接触和/或一种或多种T细胞激活剂接触;经所述步骤(B)得到第三TIL群;
(C)使所述第三TIL群与所述一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
在一种实施方式中,在步骤(C)中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触。
在一种实施方式中,所述步骤(C)进行至多约24小时。
在一种实施方式中,所述步骤(C)进行约12小时至约24小时。
在一种实施方式中,与在步骤(C)中未曾与所述T细胞激活剂和/或所述免疫检查点抑制剂接触的相应TIL相比,在步骤(C)中与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触过的TIL显示出改善的扩增效果。
在一种实施方式中,所述改善的扩增效果包含选自以下组的一种或多种:增加的TIL细胞数量,改善的T细胞亚群比例,提高的细胞因子分泌能力,和提高的肿瘤细胞杀伤能力。
在一种实施方式中,所述改善的T细胞亚群比例包含选自以下组的一种或多种:增加的中心记忆T细胞比例,降低的调节性T细胞的比例,增加的活化T细胞比例,增加的肿瘤特异性T细胞比例,和增加的干细胞样T细胞比例。
在一种实施方式中,所述一种或多种T细胞激活剂包含选自以下组的一种或多种:CD80、CD86、B7-H3、4-1BBL、CD27、CD30、CD134、B7h、CD40、LIGHT、以及它们的功能活性片段。
在一种实施方式中,所述一种或多种T细胞激活剂包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD3激动剂和/或CD28激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD3激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含抗CD3的抗体和/或其抗原结合片段。
在一种实施方式中,所述一种或多种T细胞激活剂包含CD28激动剂。
在一种实施方式中,所述一种或多种T细胞激活剂包含抗CD28的抗体和/或其抗原结合片段。
在一种实施方式中,所述使TIL与所述一种或多种T细胞激活剂接触包含选自以下组的一种或多种方式:(1)将所述一种或多种T细胞激活剂添加至所述TIL的细胞培养基中;(2) 将表达所述一种或多种T细胞激活剂的工程化细胞添加至所述TIL的细胞培养基中;(3)将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为至少约30ng/mL。
在一种实施方式中,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为约30ng/mL-约300ng/mL。
在一种实施方式中,所述固相介质的直径为约500纳米至约10微米。
在一种实施方式中,所述固相介质的直径为约1纳米至约500纳米。
在一种实施方式中,所述固相介质的直径通过透射电子显微镜测量。
在一种实施方式中,所述固相介质包含聚合物。
在一种实施方式中,每mg所述固相介质中包含的每一种所述T细胞激活剂的量各自独立地至少为约25μg。
在一种实施方式中,以约2:1-约1:2的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,以约1:100-约1:2000的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
在一种实施方式中,所述免疫检查点抑制剂包含抑制PD-1与PD-L1和/或PD-L2相互作用的物质。
在一种实施方式中,所述免疫检查点抑制剂包含PD-1抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含PD-1的抗体和/或其抗原结合片段。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约100pM或更低的K D值结合PD-1的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约100pM或更低的EC 50值结合PD-1的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含具有以约1nM或更低的IC 50值抑制PD-1与PD-L1和/或PD-L2结合的能力的免疫检查点抑制剂。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR3,且所述HCDR3包含SEQ ID NO:3和17中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR2,且所述HCDR2包含SEQ ID NO:2和16中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含HCDR1,且所述HCDR1包含SEQ ID NO:1和15中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR3,且所述LCDR3包含SEQ ID NO:6和20中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR2,且所述LCDR2包含SEQ ID NO:5和19中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含LCDR1,且所述LCDR1包含SEQ ID NO:4和18中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含VH,且所述VH包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂包含VL,且所述VL包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列。
在一种实施方式中,所述免疫检查点抑制剂选自以下组:(1)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:1和15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述轻链包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3;(2)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述轻链包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL。
在一种实施方式中,其中所述抗体选自:嵌合抗体,人源化抗体和全人源抗体。
在一种实施方式中,其中所述抗原结合片段选自:Fab,Fab′,Fv片段,F(ab') 2,F(ab) 2,scFv,di-scFv,VHH和dAb。
在一种实施方式中,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL。
在一种实施方式中,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地为约0.1μg/mL-约20μg/mL。
在一种实施方式中,在步骤(B)中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种T细胞生长因子接触。
在一种实施方式中,所述一种或多种T细胞生长因子选自以下组的一种或多种:IL-2、 IL-7、IL-12、IL-15、IL-21、γ干扰素、以及它们的功能活性片段。
在一种实施方式中,所述一种或多种T细胞生长因子包含IL-2和/或其功能活性片段。
在一种实施方式中,所述TIL与所述一种或多种T细胞生长因子接触包含将所述T细胞生长因子添加至所述TIL的细胞培养基中。
在一种实施方式中,每一种所述T细胞生长因子在所述TIL的细胞培养基中的初始浓度各自独立地为至少约300IU/mL。
在一种实施方式中,其还包含:在步骤(A)、步骤(B)和/或步骤(C)中,使所述TIL与饲养细胞共培养。
在一种实施方式中,在步骤(B)中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触且与所述饲养细胞共培养。
在一种实施方式中,在步骤(B)中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触一定时间之后,再与所述饲养细胞共培养。
在一种实施方式中,所述一定时间为至少约2小时。
在一种实施方式中,所述一定时间为约6小时至约72小时。
在一种实施方式中,所述一定时间为约12小时至约48小时。
在一种实施方式中,所述一定时间为约6小时、约12小时、约24小时、约48小时、或约72小时。
在一种实施方式中,所述饲养细胞包含抗原呈递细胞。
在一种实施方式中,所述饲养细胞包含选自以下组的一种或多种:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。
在一种实施方式中,所述饲养细胞为外周单个核细胞。
在一种实施方式中,所述饲养细胞为经过辐照的饲养细胞。
在一种实施方式中,所述TIL与所述饲养细胞共培养包含使所述饲养细胞的表面与所述TIL的表面相接触。
在一种实施方式中,所述TIL与所述饲养细胞共培养包含将所述饲养细胞添加至所述TIL的细胞培养基中。
在一种实施方式中,以约40:1-约400:1的所述饲养细胞与所述TIL的比例,将所述饲养细胞添加至所述TIL的细胞培养基中。
在一种实施方式中,所述源自肿瘤组织且未经体外扩增的TIL为源自所述肿瘤组织的碎片的TIL。
在一种实施方式中,所述碎片的体积为约1立方毫米至约27立方毫米。
本申请还提供一种肿瘤浸润淋巴细胞(TIL),所述TIL经过本申请的方法获得。
本申请还提供一种组合物,其包含本申请的TIL。
本申请还提供一种药物组合物,其包含本申请的TIL和/或本申请的组合物,以及任选地药学上可接受的载体。
本申请还提供一种影响肿瘤细胞生长的方法,包含向受试者施用本申请的TIL、本申请的组合物和/或本申请的药物组合物。
本申请还提供了本申请的TIL、本申请的组合物和/或本申请的药物组合物在制备药物中的应用,所述药物用于预防和/或***。
在一种实施方式中,所述肿瘤为实体瘤。
在一种实施方式中,所述肿瘤选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
本申请还提供了本申请的TIL、本申请的组合物和/或本申请的药物组合物,其用于预防和/或***。
在一种实施方式中,所述肿瘤为实体瘤。
在一种实施方式中,所述肿瘤选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
本申请还提供了一种预防和/或***的方法,其包含向受试者施用本申请的TIL、本申请的组合物和/或本申请的药物组合物。
在一种实施方式中,所述肿瘤为实体瘤。
在一种实施方式中,所述肿瘤选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
本领域技术人员能够从下文的详细描述中容易地洞察到本申请的其它方面和优势。下文的详细描述中仅显示和描述了本申请的示例性实施方式。如本领域技术人员将认识到的,本申请的内容使得本领域技术人员能够对所公开的具体实施方式进行改动而不脱离本申请所涉及发明的精神和范围。相应地,本申请的附图和说明书中的描述仅仅是示例性的,而非为限制性的。
附图说明
本申请所涉及的发明的具体特征如所附权利要求书所显示。通过参考下文中详细描述的 示例性实施方式和附图能够更好地理解本申请所涉及发明的特点和优势。对附图简要说明如下:
图1A-1B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的胞内因子表达能力。
图2A-2B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的胞内因子表达能力。
图3A-3C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的IL-2分泌检测结果。
图4A-4C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的TNF分泌检测结果。
图5A-5C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的IFNγ分泌检测结果。
图6A-6B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的细胞杀伤能力检测结果。
图7A-7C显示的是,对于不同供者,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL细胞增殖情况。
图8A-8B显示的是,对于不同供者,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL细胞增殖情况。
图9A-9B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞活率和细胞分型情况。
图10A-10B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中活化相关(41BB +)细胞比例情况。
图11A-11C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中活化相关(CD25 +和/或CD27 +)细胞比例情况。
图12A-12B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中耗竭相关(TIM3 +)细胞比例情况。
图13A-13B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中分泌TNF-α细胞比例情况。
图14A-14D显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中分泌IFN-γ细胞比例情况。
图15A-15C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞因子(IL-2和/或IL-4)分泌量情况。
图16A-16C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞因子(IL-17和/或IL-6)分泌量情况。
图17A-17D显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞因子(TNF-α和/或IFN-γ)分泌量情况。
图18A-18D显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-4和/或TNF-α)分泌量情况。图18E-18F显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-2和/或IL-6)分泌量情况。
图19A-19D显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-6和/或TNF-α)分泌量情况。
图20A-20C显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-2、IL-4和/或IFN-γ)分泌量情况。
具体实施方式
以下由特定的具体实施例说明本申请发明的实施方式,熟悉此技术的人士可由本说明书所公开的内容容易地了解本申请发明的其他优点及效果。
术语定义
在本申请中,术语“表达”通常是指编码目标多肽的基因在细胞内发生的转录和/或翻译过程。可以通过测量存在于细胞中的相应mRNA的量来确定宿主细胞中编码目标多肽的基因的转录水平。例如,可通过PCR或通过RNA杂交对编码目标多肽的基因转录的mRNA进行定量测量(参见Sambrook等,分子克隆:实验手册,Cold Spring Harbor Laboratory Press(1989))。可以通过多种方法测量编码目标多肽的基因的翻译水平,例如通过ELISA,通过多肽生物活性测试,或通过蛋白质印迹或放射免疫测试法(参见Sambrook等,同上)。
在本申请中,术语“一个阶段的体外扩增”、“单个阶段的体外扩增”、或“第一阶段体外扩增”等中的“阶段”通常是指TIL在体外经过的一段扩增过程。在一种实施方式中,每一个阶段之间可以是通过TIL细胞数量的变化来划分的,在一种实施方式中,当TIL细胞的数量增加至少约1倍时,可以认为TIL细胞进入了下一个阶段的体外扩增。在一些实施方式中,当TIL细胞的数量增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约 6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍时,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,每一个阶段之间也可以是通过TIL细胞培养的条件来划分的。在一种实施方式中,当细胞培养基中添加了或补充添加了T细胞激活剂和/或T细胞生长因子后,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,当TIL细胞进行了离心和/或细胞洗涤后,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,每一个阶段之间也可以是通过TIL细胞培养的天数来划分的。在一种实施方式中,当TIL细胞体外培养约1天、约2天、约3天、约4天、约5天、约6天、约7天、约8天、约9天、约10天、约11天、约12天、约13天、约14天、约15天、约16天、约17天、约18天、约19天、约20天、约30天、约40天、约50天或约100天后,可以认为TIL细胞进入了下一个阶段的体外扩增。
在本申请中,术语“第一阶段体外扩增”通常是指从组织中获得初级TIL后,使用T细胞生长因子进行扩增的阶段。在一种实施方式中,本申请的组织可以选自以下组:肿瘤组织和胸腔积液,本申请的胸腔积液可以是有转移癌的患者的胸腔积液。在一种实施方式中,本申请的扩增可以是自体或者异体进行的体内扩增,或者可以是体外扩增。本申请的第一阶段体外扩增也可以称为preREP(快速扩增前)阶段。
在本申请中,术语“第二阶段体外扩增”通常是指从受试者体内取出的组织并进行扩增后,再次进行扩增的阶段。在一种实施方式中,与经第一阶段体外扩增的TIL相比,本申请的经第二阶段体外扩增的TIL细胞数量增加,例如可以增加至少约10倍(或至少约20、30、40、50、60、70、80或90倍),或者在一种实施方式中细胞的数量可以增加至少约100倍。在一种实施方式中,第二阶段体外扩增可以与第一阶段体外扩增的培养条件不同,例如加入的培养物质可以不同。本申请的第二阶段体外扩增也可以称为REP(快速扩增)阶段。
在本申请中,术语“体内”通常是指发生在受试者体内的事件。
在本申请中,术语“体外”通常是指在受试者体外发生的事件。
在本申请中,术语“离体”通常是指涉及对已从受试者体内移除的细胞、组织和/或器官进行治疗或进行手术的事件。在一种实施方式中,该细胞、组织和/或器官可以通过手术或治疗方法返回到受试者的身体。
在本申请中,术语“分泌”通常是指细胞将表达的多肽或蛋白转移到细胞外环境。
在本申请中,术语“分泌能力”通常是指细胞表达多肽或蛋白并将本申请的多肽或蛋白转移到细胞外环境的能力。
在本申请中,术语“辐照”通常是指通过射线对物质进行的处理。例如,在一种实施方式中,辐照可以是指通过X射线、α射线、β射线或γ射线对物质进行辐照。
在本申请中,术语“工程化细胞”通常是指将DNA或RNA形式的额外遗传物质加入细胞的总遗传物质而被基因修饰的细胞。在一种实施方式中,工程化细胞可以经过基因修饰以表达本申请的T细胞激活剂和/或T细胞生长因子的TIL。
在本申请中,术语“共培养”通常是指将两个或更多个不同群体的细胞在它们之间有一定程度的接触的情况下培养。本申请的两个或更多个不同群体的细胞的“接触”,在一种实施方式中可以通过直接接触,即其中一个群体的细胞与另一个群体的细胞直接物理接触。或者在一种实施方式中可以通过共用培养基所介导的间接接触。本申请的共用的培养基可以含有由共培养细胞的至少一个群体所产生和释放的代谢产物,并用于培养另一个群体的细胞。
在本申请中,术语“接触”通常是指两个或更多个不同类型的物质以任何顺序、任何方式以及任何时长接触在一起。在一种实施方式中可以通过直接接触,例如可以将一种或多种饲养细胞、T细胞激活剂和/或T细胞生长因子加入TIL细胞的培养基,例如可以将包含一种或多种饲养细胞、T细胞激活剂和/或T细胞生长因子的培养基加入和/或替换TIL细胞的培养基,例如,可以将包含一种或多种饲养细胞、T细胞激活剂和/或T细胞生长因子的培养基用于TIL细胞的培养;在一种实施方式中可以通过间接接触,例如可以将饲养细胞产生和释放的代谢产物,用于培养TIL细胞。
在本申请中,术语“混合物”通常是指两个或更多个不同物质的组合。例如,本申请的CD28抗体或其抗原结合片段以及CD3抗体或其抗原结合片段可以在混合后作为混合物加入细胞培养基。
在本申请中,术语“同时接触”、“共同接触”、“与...接触同时”、“同时”和“共同”通常是指向受试者和/或细胞施用两种以上物质,使得物质同时存在于受试者和/或细胞培养的环境中。同时接触可以包括以不同的组合物同时施用、以不同的组合物在不同时间施用,或以其中存在两种以上活性药物成分的组合物施用。例如,本申请中“同时接触”通常可以是指基本上同时接触。
在本申请中,术语“扩增”通常是指在一段时间内细胞的数量增加若干倍。在一种实施方式中细胞的数量可以增加至少约3倍(或4、5、6、7、8或9倍),在一种实施方式中细胞的数量可以增加至少约10倍(或20、30、40、50、60、70、80或90倍),或者在一种实施方式中细胞的数量可以增加至少约100倍。在本申请中,术语“经扩增”通常是指本申请的细胞经过上述一种或多种扩增。
在本申请中,术语“聚合物”通常是指由连接在一起的单独化学部分组成的分子,本申请的聚合物部分可相同或不同。在一种实施方式中,术语“聚合物”可以指尾尾相连而形成线性分子的单独化学部分,以及以分支(如“多臂”或“星型”)结构形式连接在一起的单独化学部分。在一种实施方式中聚合物可以包括例如多糖、葡聚糖、水凝胶、聚乙二醇、或泊洛沙姆。泊洛沙姆是非离子三嵌段共聚物,其具有聚氧丙烯(聚(环氧丙烷))中央疏水链,侧连两条聚氧乙烯(聚(环氧乙烷))亲水链。本申请包含的物质可以与本文所描述的或本领域已知的任何聚合物一起配制,或与它们一起给予。
在本申请中,术语“抑制剂”通常是指包括部分或完全阻断、抑制或中和本文所述的任何分子,例如蛋白质。例如,本申请所抑制的分子包含但不限于PD-1。合适的抑制剂分子可以包括拮抗剂抗体或抗体片段、小分子的片段或衍生物、肽、反义寡核苷酸、小的有机分子等。鉴定本申请的抑制剂的方法包括使表达本申请所抑制的分子的细胞与候选抑制剂分子接触,检测本申请所抑制的分子相关的一种或多种生物学活性可检测的变化。例如,本申请的抑制剂可以是PD-1抑制剂,该抑制剂可以阻断PD-1与其配体结合。例如,合适的PD-1抑制剂可以是PD-1抗体。
在本申请中,术语“免疫检查点抑制剂”通常是指整体或部分减少、抑制、干扰或调节一种或多种检查点蛋白质的分子。已知多种检查点蛋白质可以包含,如PD-1及其配体PD-L1和PD-L2。免疫检查点抑制剂可以包括抗体或源自于抗体的多肽。
在本申请中,术语“IC 50值”或“IC50值”通常是指目标物获得生物学过程50%抑制需要的浓度。可以使用Cheng-Prusoff方程(Biochem.Pharmacol.(1973)22:3099)将IC50值换算成绝对抑制常数(Ki)。
在本申请中,术语“EC 50值”通常是指结合物质(例如抗体)在体外或体内分析背景下诱导的应答在基线值和最大值之间的50%的反应的半最大有效浓度。减小的EC 50值可以表示较高的药物亲和力和功效。
在本申请中,术语“K D值”或“KD值”通常是指解离常数,其可通过表面等离子体共振进行测定。通常,表面等离子体共振分析使用BIAcore***(Pharmacia Biosensor,Piscataway,NJ),通过表面等离子体共振(SPR),测量配体(固定化于生物传感器基质上的物质)和分析物(溶液中的物质)之间的实时结合相互作用。也可以通过固定化分析物(生物传感器基质上的物质)和呈递配体,进行表面等离子体分析。
在本申请中,术语“PD-1”或“PD1”通常是指编程性细胞死亡蛋白1,是一种288个氨基酸的I型膜蛋白,首次在1992年有描述(Ishida et al.,EMBO J.,11(1992),3887-3895)。PD-1 是扩大的CD28/CTLA-4T细胞调节物家族的一个成员且具有两种配体,PD-L1(B7-H1,CD274)和PD-L2(B7-DC,CD273)。该蛋白质的结构包括一个胞外IgV域,接着是一个跨膜区和一个胞内尾。胞内尾含有位于免疫受体基于酪氨酸的抑制性基序和免疫受体基于酪氨酸的转换基序中的两个磷酸化位点,提示PD-1阴性负调节TCR信号。这与配体结合后SHP-1和SHP-2磷酸酶对PD-1的胞质尾的结合一致。术语“编程性死亡1”,“编程性细胞死亡1”,“蛋白PD-1”,“PD-1”,PD1”,PDCD1”,“hPD-1”和“hPD-I”可以互换使用,而且包括人PD-1的变体,同等型,物种同系物,和与PD-1具有至少一个共同表位的类似物。人PD1的氨基酸序列可以在UniProt(www.uniprot.org)登录号Q15116中显示。
在本申请中,术语“PD-L1”或“PDL1”通常是指程序性细胞死亡1配体1,也可称为B7同源物1、B7-H1、分化簇274、(3)274或CD274,其与PD-1结合后下调T细胞活化和细胞因子分泌。“PD-L1”包括任何脊椎动物来源的任何天然PD-L1,所述任何脊椎动物来源包括哺乳动物,诸如灵长类(例如,人和食蟹猴)和啮齿类(例如,小鼠和大鼠)。所述术语涵盖“全长”、未加工的PD-L1以及由细胞加工所产生的任何形式的PD-L1。PD-L1可作为跨膜蛋白或作为可溶性蛋白存在。“PD-L1”包括完整的PD-L1及其片段,还包括PD-L1的功能性变体、同工型、物种同源物、衍生物、类似物,以及具有至少一个与PD-L1共同表位的类似物。PD-L1的基本结构包括4个结构域:胞外Ig样V型结构域和Ig样C2型结构域、跨膜结构域以及细胞质结构域。示例性的人PD-L1氨基酸序列可在NCBI登录号NP_001254653或UniProt登录号Q9NZQ7下找到。
在本申请中,术语“抗体”通常是指对指定蛋白质或肽或其片段有反应性的免疫球蛋白。此类抗体包括但不限于人抗体、灵长类化抗体、嵌合抗体、单克隆抗体、单特异性抗体、多克隆抗体、多特异性抗体、非特异性抗体、双特异性抗体、多特异性抗体、人源化抗体、合成抗体、重组抗体、杂合抗体、突变型抗体、嫁接偶联抗体(即偶联或融合至其它蛋白质、放射性标记物、细胞毒素的抗体)、和体外生成的抗体。抗体可来自任何类的抗体,包括但不限于IgG、IgA、IgM、IgD、和IgE,及来自任何亚类(例如IgG1、IgG2、IgG3、和IgG4)的抗体。抗体可具有选自例如IgG1、IgG2、IgG3、或IgG4的重链恒定区。抗体还可具有选自例如卡帕(κ)或拉姆达(λ)的轻链。本申请的抗体可衍生自任何物种,包括但不限于小鼠、人、骆驼、美洲驼、鱼、鲨鱼、山羊、家兔、鸡、和牛。抗体的恒定区可进行改变,例如突变,以修饰抗体的特性(例如以提高或降低下述一项或多项:Fc受体结合、抗体糖基化、半胱氨酸残基的数目、效应器细胞功能、或补体功能)。通常,抗体特异性结合预定抗原,例如与病症有关的抗原,病症例如炎性的、免疫性的、自身免疫性的、神经变性性的、代谢的和/或恶性的病症。
在本申请中,术语“嵌合抗体(chimeric antibody)”通常是指鼠源性抗体的可变区与人抗体的恒定区融合而成的抗体,可以减轻鼠源性抗体诱发的免疫应答反应。建立嵌合抗体,可以建立分泌鼠源性特异性单抗的杂交瘤,然后从鼠杂交瘤细胞中克隆可变区基因,可以根据需要克隆人抗体的恒定区基因,将鼠可变区基因与人恒定区基因连接成嵌合基因后***表达载体中,可以在真核***或原核***中表达嵌合抗体分子。
在本申请中,术语“人源化抗体(humanized antibody)”,也称为CDR移植抗体(CDR-grafted antibody),通常是指将鼠的CDR序列移植到人的抗体可变区框架,即不同类型的人种系抗体框架序列中产生的抗体。可以克服嵌合抗体由于携带大量鼠蛋白成分,从而诱导的异源性反应。此类构架序列可以从包括种系抗体基因序列的公共DNA数据库或公开的参考文献获得。如人重链和轻链可变区基因的种系DNA序列可以在“VBase”人种系序列数据库。
在本申请中,术语“全人源抗体”、“全人抗体”或“完全人源抗体”,也称“全人源单克隆抗体”,其抗体的可变区和恒定区可以都是人源的,去除免疫原性和毒副作用。单克隆抗体的发展经历了四个阶段,分别为:鼠源性单克隆抗体、嵌合性单克隆抗体、人源化单克隆抗体和全人源单克隆抗体。本申请所述抗体或配体可以为全人源单克隆抗体。全人抗体制备的相关技术可以为:人杂交瘤技术、EBV转化B淋巴细胞技术、噬菌体显示技术(phage display)、转基因小鼠抗体制备技术(transgenic mouse)和单个B细胞抗体制备技术等。
在本申请中,术语“CDR”通常是指抗体的可变结构域内主要促成抗原结合的6个高变区之一。所述6个CDR的最常用的定义之一由Kabat E.A.等人,(1991)Sequences of proteins of immunological interest.NIH Publication 91-3242)提供,Chothia等人,“Canonical Structures For the Hypervariable Regions of Immunoglobulins,”J.Mol.Biol.196:901(1987);和MacCallum等人,“Antibody-Antigen Interactions:Contact Analysis and Binding Site Topography,”J.Mol.Biol.262:732(1996))。如本申请中使用的,CDR的Kabat定义可以应用于轻链可变结构域的CDR1、CDR2和CDR3(CDR L1、CDR L2、CDR L3或L1、L2、L3),以及重链可变结构域的CDR1、CDR2和CDR3(CDR H1、CDR H2、CDR H3或H1、H2、H3)。
在本申请中,术语“抗原结合片段”通常是指具有特异结合抗原(例如,PD-L1)能力的一个或多个多肽片段。在本申请中,所述抗原结合片段可以包括Fab,Fab’,F(ab) 2、Fv片段、F(ab’) 2,scFv,di-scFv和/或dAb。
在本申请中,术语“可变区”通常是指在抗体之间可变结构域的某些区段在序列上可能存在较大差异的区域。轻链中的“可变区”可以包含轻链可变区VL;重链中的“可变区”可以包含重链可变区VH。可变结构域介导抗原结合并决定特定抗体对其特定抗原的特异性。然而,可 变性并非在整个可变结构域范围内均匀分布。它通常集中在轻链和重链可变结构域中称为高变区(CDR或HVR)的三个区段中。可变结构域的更高度保守的部分称为框架区(FR)。天然重链和轻链的可变结构域各自包含四个FR区,大多数采用β-折叠构型,通过三个CDR连接,其形成环形连接,并且在一些情况下形成β-折叠结构的一部分。每条链中的CDR通过FR区紧密靠近地保持在一起,并且来自另一条链的CDR一同促进抗体的抗原结合位点的形成(参见Kabat et al,Sequences of Immunological Interest,Fifth Edition,National Institute of Health,Bethesda,Md.(1991))。
在本申请中,术语“Fab”通常是指抗体的抗原结合片段。如上所述,可以使用木瓜蛋白酶消化完整的抗体。抗体经木瓜蛋白酶消化后产生两个相同的抗原结合片段,即“Fab”片段,和残余的“Fc”片段(即Fc区,同上)。Fab片段可以由一条完整的L链与一条重链的可变区和该H链(VH)的第一恒定区(CH1)组成。
在本申请中,术语“Fab’”或“Fab′片段”通常是指人单克隆抗体的单价抗原结合片段,该片段可以比Fab片段稍大。例如,Fab′片段可以包括所有轻链,所有重链可变区以及重链的所有或部分第一和第二恒定区。例如,Fab′片段还可以包括重链的部分或所有的220-330个氨基酸残基。
在本申请中,术语“(Fab’)2”通常是指通过胃蛋白酶消化完整抗体所产生的抗体片段。F(ab')2片段含有由二硫键维持在一起的两个Fab片段和部分铰链区。F(ab')2片段具有二价抗原结合活性并且能够交联抗原。
在本申请中,术语“Fv”或“Fv片段”通常是指人单克隆抗体的单价抗原结合片段,包括所有或部分重链可变区和轻链可变区,并且缺乏重链恒定区和轻链恒定区。重链可变区和轻链可变区包括例如CDR。例如,Fv片段包括重链和轻链的约110个氨基酸的所有或部分氨基端可变区。
在本申请中,术语“scFv”通常是指包含至少一个包括轻链的可变区抗体片段和至少一个包括重链的可变区的抗体片段的融合蛋白,其中所述轻链和重链可变区是邻接的(例如经由合成接头例如短的柔性多肽接头),并且能够以单链多肽形式表达,且其中所述scFv保留其所来源的完整抗体的特异性。除非特别说明,否则如本申请中使用的那样,scFv可以以任何顺序(例如相对于多肽的N末端和C末端)具有所述的VL和VH可变区,scFv可以包括VL-接头-VH或可以包括VH-接头-VL。
在本申请中,术语“dAb”通常是指具有VH域或VL域组成的抗原结合片段,参考例如Ward等人(Nature,1989Oct 12;341(6242):544-6),参考Holt等人,Trends Biotechnol.,2003,21(11):484-490。
在本申请中,术语“VHH”通常是指包含重链抗体的可变抗原结合结构域的抗体(参见Vanlandschoot P.等人,2011,Antiviral Research 92,389-407)。VHH也可称为纳米抗体(Nanobody)(Nb)。
在本申请中,术语“抗CD3抗体”通常是指靶向CD3的抗体或其变体,例如单克隆抗体,包括人、人源化、嵌合或鼠抗体,其针对成熟T细胞的T细胞抗原受体中的CD3受体。抗CD3抗体可以包括OKT3。抗CD3抗体可以包括SP34。抗CD3抗体还可以包括其他抗CD3抗体包括例如在一种实施方式中otelixizumab、teplizumab和visilizumab。
在本申请中,术语“IL-2”或“IL2”通常是指称为白细胞介素2的T细胞生长因子,并包括所有形式的IL-2,可以包括在一种实施方式中人和哺乳动物形式、保守性氨基酸取代、糖型修饰或变体,或其活性片段。编码IL-2基因的GeneID可以为3558。
在本申请中,术语“抗原呈递细胞”、“抗原递呈细胞”、或“APC”通常是指,在其表面上展示与主要组织相容性复合物(MHC)复合的外源抗原的免疫***细胞,如辅助细胞(例如,B细胞、树突细胞等)。T细胞可以使用其T细胞受体(TCR)识别这些复合物。APC可以加工抗原并将其递呈至T细胞。在一种实施方式中,抗原呈递细胞可以包括选自以下组:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。
在本申请中,术语“扩增效果”通常是指细胞经过扩增后出现的效果。扩增效果的变化可以包括,细胞的数量和/或比例变化,分泌能力变化,杀伤能力变化或表达能力的变化,或它们的任何组合。本申请的变化可以是提高或者降低。
在本申请中,术语“纳米颗粒”通常是指至少一个尺寸小于100nm的微观颗粒。通常,纳米颗粒具有50nm至500nm(即0.05μm至0.5μm)范围内的直径;在生理环境中结构稳定;且可以容纳更小的分子(如药物或其他生物活性剂),然后可以将该分子递送至希望的部位。例如,本申请的纳米颗粒可以包含CD28抗体或其抗原结合片段。例如,本申请的纳米颗粒可以包含CD28抗体或其抗原结合片段以及CD3抗体或其抗原结合片段。例如,抗CD28抗体可以包括OKT3。例如,抗CD28抗体可以包括15E8。
在本申请中,术语“人工抗原呈递细胞”通常是指人工构建的用于呈递外源抗原的免疫细胞,例如,呈递外源抗原的方式可以是人工抗原呈递细胞的表面包含外源抗原与主要组织相容性复合物(MHC)的复合物。在一个实施方案中,可以包括分离的人工抗原呈递细胞(aAPC), 其可以包含表达HLA-A/B/C(编码其的基因GeneID可以为3105、3106或3107)、CD64(编码其的基因GeneID可以为2209)、CD80(编码其的基因GeneID可以为941)、ICOS-L(编码其的基因GeneID可以为23308)和CD58(编码其的基因GeneID可以为965)的细胞,并可以被修饰以表达一种以上T细胞激活剂,本申请的以上可以包含本数。
在本申请中,术语“融合蛋白”通常是指含有第一多肽或蛋白质或其片段、类似物或衍生物的氨基酸序列和异源多肽或蛋白质(即,不同于第一多肽或蛋白质或其片段、类似物或衍生物的第二多肽或蛋白质或其片段、类似物或衍生物,或者通常不是第一多肽或蛋白质或其片段、类似物或衍生物的一部分)的氨基酸序列的多肽或蛋白质。在某些情形中,融合蛋白可以包含与异源蛋白、多肽或肽融合的预防性或治疗性药物。其中,本申请的异源蛋白、多肽或肽可以是或不是不同类型的预防性或治疗性药物。例如,可将具有免疫调节活性的两种不同蛋白质、多肽或肽融合到一起形成融合蛋白。在某些情形中,与异源蛋白、多肽或蛋白质融合前的初始多肽或蛋白质的活性相比,融合蛋白可以保留或提高了活性。例如,本申请的融合蛋白可以是融合了CD28抗体或其抗原结合片段,以及CD3抗体或其抗原结合片段的融合蛋白。
在本申请中,术语“杀伤能力”通常是指通过使本申请的细胞接触有效量的物质从而杀伤靶细胞来实现。在一个实施方案中,本申请的物质可以是TIL细胞。本申请的杀伤可以包括通过自身或者促进其它细胞或物质的CDC、凋亡、ADCC、和/或吞噬作用,或通过两种或更多种这些机制的组合以杀伤细胞。
在本申请中,术语“施用”通常是指通过本领域已知的任意途径,将物质递送给有此需要的受试者。药用载体和制剂或组合物也是本领域众所周知的。给药途径可以包括:静脉内的、肌肉内的、真皮内的、皮下的、透皮的、粘膜的、瘤内的和/或粘膜的。
在本申请中,术语“试剂盒”通常是指一起被包装在容器、接受器或其它容器中的两种或更多种组分,其中一种对应于本申请的物质。例如,包含本申请的TIL细胞。
在本申请中,术语“受试者”通常是指细胞或动物,可以是哺乳动物,诸如人、非人灵长类动物(猿、长臂猿、大猩猩、黑猩猩、猩猩、猕猴)、家畜(狗和猫)、农场动物(家禽如鸡和鸭、马、牛、山羊、绵羊、猪)和实验动物(小鼠、大鼠、兔、豚鼠)。人受试者包括胎儿、新生儿、婴儿、青少年和成人受试者。受试者包括动物疾病模型,例如肿瘤动物模型,和本领域技术人员已知的其它动物模型。
在本申请中,术语“饲养细胞(feeder)”通常是指体外生长和分泌至少一种因子至培养基并且可以用于支持培养另一种所关注的细胞生长的培养细胞。在一种实施方式中,饲养细胞可以包括抗原呈递细胞。
在本申请中,术语“特异性结合”通常是指识别特异性抗原,但是基本不识别或结合样品中其它分子的抗体。例如,如果一种抗体可以特异性结合来自一个物种的本申请的特异性抗原,则本申请的抗体还可以特异性结合来自其它的一个或多个物种的本申请的抗原或同源抗原。这种种间反应性本身可以不会改变抗体作为特异性的分类。在某些情形中,特异性结合至抗原的抗体还可以结合至抗原的不同等位形式。
在本申请中,术语“完整的培养过程”通常是指将细胞从患者体内分离的肿瘤组织中分离开始,经过一次或一次以上的扩增,最终获得可以施用于受试者的细胞的完整过程。
在本申请中,术语“细胞培养基”通常是指细胞例如哺乳动物细胞在其中生长的营养液。细胞培养基的配制在本领域中是熟知的。典型地,细胞培养基包括缓冲液、盐、碳水化合物、氨基酸、维生素以及必要的微量元素。细胞培养基可以含有或不含有血清、蛋白胨、和/或蛋白质。细胞培养基可以补充有另外的组分或浓度增加的组分,如氨基酸、盐、糖、维生素、激素、生长因子、缓冲液、抗生素、脂质、微量元素等,这取决于有待培养的细胞的要求和/或所希望的细胞培养参数。
在本申请中,术语“药物组合物”或“药物制剂”通常是指一种制备物,本申请的制备物可以允许有效成分的生物活性有效,并且可以不含有对于将会施用该制剂的受试者不可接受地有毒的额外组分。这类制剂是无菌的。“可药用的”赋形剂(载体、添加物)是可以合理地施用至受试哺乳动物以提供有效剂量的所用有效成分的那些赋形剂。
在本申请中,术语“肿瘤浸润淋巴细胞”或“TIL”通常是指最初作为白细胞获得的细胞群,本申请的细胞已经离开受试者的血流并迁移到肿瘤中。TIL可以包括但不限于CD8 +细胞毒性T细胞(淋巴细胞)、Th1和Th17 CD4 +T细胞、天然杀伤细胞、树突细胞和M1巨噬细胞。TIL可以包括初级TIL和次级TIL。“初级TIL”可以是从受试者组织样品获得的那些TIL细胞,“次级TIL”可以是本申请中已扩增或经扩增的任何TIL细胞群。在一些实施方式中,本申请的肿瘤浸润淋巴细胞可以是未经分离纯化的,或者可以是与肿瘤细胞相互浸润的。在一种实施方式中,本申请的TIL可以是指TIL细胞群。
在本申请中,术语“中心记忆T细胞”通常是指具有长期记忆性的,并能够接受抗原再刺激的T细胞。中心记忆T细胞可以具有CD45RA -CCR7 +的表型,例如可以是通过CD45RA -和 CCR7 +来鉴定中心记忆T细胞。中心记忆T细胞可以相比普通T细胞具有更强的抗肿瘤生长的能力。
在本申请中,术语“调节性T细胞”通常是指一类控制体内自身免疫反应性的T细胞亚群。调节性T细胞可以具有CD4 +CD25 +Foxp3 +的表型,例如可以是通过CD4 +、CD25 +和Foxp3 +来鉴定调节性T细胞。调节性T细胞可以具有抑制T细胞的抗肿瘤生长的能力。
在本申请中,术语“活化T细胞”通常是指经过活化而可以具有抗肿瘤生长的能力的T细胞。活化T细胞可以具有PD-1 +、LAG3 +或CD28 +的表型,例如可以是通过PD-1 +、LAG3 +或CD28 +来鉴定活化T细胞。活化T细胞可以具有抗肿瘤生长的能力。
在本申请中,术语“肿瘤特异性T细胞”通常是指可以特异性抗肿瘤生长的T细胞。肿瘤特异性T细胞可以具有CD103 +CD39 +的表型,例如,可以是通过CD103 +和CD39 +来鉴定肿瘤特异性T细胞。肿瘤特异性T细胞可以相比普通T细胞具有更特异性的抗肿瘤生长的能力。
在本申请中,术语“干细胞样T细胞”通常是指可以具有自我增殖和/或分化的潜能的一类T细胞。干细胞样T细胞可以具有TCF1 +的表型,例如可以是通过TCF1 +来鉴定干细胞样T细胞。肿瘤特异性T细胞可以相比普通T细胞具有更强和/或更长期的抗肿瘤生长的能力。
在本申请中,术语肿瘤“碎片”通常是指从受试者体内取出肿瘤组织后,可以通过机械破碎、酶解和/或其它破碎方法,形成的肿瘤碎片。
在本申请中,术语“组合物”或“药物组合物”通常是指至少一种细胞以及至少一种和任选多于一种的其他药学上可接受的化学组分如运载体、稳定剂、稀释剂、分散剂、助悬剂、增稠剂和/或赋形剂的混合物。
在本申请中,术语“药学上可接受的载体”通常是指不干扰活性成分的一种或多种非毒性材料。例如,药学上可接受的载体可以不干扰扰活性成分的生物活性;例如,药学上可接受的载体可以不干扰扰活性成分所具有的生物活性的有效性。这类制剂常规地可以含有盐、缓冲剂、防腐剂、相容的载体、以及任选地其他治疗剂。这类药学上可接受的制剂还可以含有适合于给予人的相容的固体或液体填料、稀释剂或包封物质。可以用于在此所描述的配制品中的其他设想的载体、赋形剂、和/或添加剂可以包括:例如,调味剂、抗微生物剂、增甜剂、抗氧化剂、抗静电剂、脂质、蛋白质赋形剂(如血清白蛋白、明胶、酪蛋白)、成盐平衡离子(如钠)等等。适合用于在此所描述的配制品中的这些和另外已知的药物载体、赋形剂和/或添加剂是本领域中已知的。
在本申请中,术语“功能活性片段”通常是指具有全长蛋白质或核酸的部分区域,但保留或部分保留全长蛋白质或核酸的生物活性或功能的片段。例如,功能活性片段可以保留或部分保留全长蛋白质结合另一种分子的能力。例如,生长因子IL-2的功能活性片段,可以保留或部分保留全长IL-2的引起细胞增殖的生物活性功能。
在本申请中,术语“T细胞激活剂”通常是指与T细胞上的相应结合受体结合,并介导T细胞共刺激反应的物质。T细胞激活剂可以是T细胞产生有效免疫应答所需的除抗原受体之外的物质。T细胞激活剂可以是指T细胞共刺激分子。T细胞激活剂可以包括但不限于MHC I类分子、TNF受体蛋白、免疫球蛋白样蛋白、细胞因子受体、整联蛋白、信号淋巴细胞活化分子(SLAM蛋白)、NK细胞活化受体、BTLA(编码其的基因GeneID可以为151888)、Toll配体受体、OX40(编码其的基因GeneID可以为7293)、CD2(编码其的基因GeneID可以为914)、CD7(编码其的基因GeneID可以为924)、CD27(编码其的基因GeneID可以为939)、CD28(编码其的基因GeneID可以为940)、CD30(编码其的基因GeneID可以为943)、CD40(编码其的基因GeneID可以为958)、CDS、ICAM-1(编码其的基因GeneID可以为3383)、LFA-1(CD11a/CD18)(编码其的基因GeneID可以为3689)、4-1BB(CD137)(编码其的基因GeneID可以为3604)、B7-H3(编码其的基因GeneID可以为80381)、ICOS(CD278)(编码其的基因GeneID可以为29851)、GITR(编码其的基因GeneID可以为8784)、BAFFR(编码其的基因GeneID可以为115650)、LIGHT(编码其的基因GeneID可以为8740)、HVEM(LIGHTR)(编码其的基因GeneID可以为8764)、KIRDS2、SLAMF7(编码其的基因GeneID可以为57823)、NKp80(KLRF1)(编码其的基因GeneID可以为51348)、NKp44(编码其的基因GeneID可以为9436)、NKp30(编码其的基因GeneID可以为259197)、NKp46(编码其的基因GeneID可以为9437)、CD19(编码其的基因GeneID可以为930)、CD4(编码其的基因GeneID可以为920)、CD8α(编码其的基因GeneID可以为925)、CD8β(编码其的基因GeneID可以为926)、IL-2Rβ、IL-2Rγ、IL7Rα(编码其的基因GeneID可以为)、ITGA4(编码其的基因GeneID可以为3676)、VLA1(编码其的基因GeneID可以为3672)、CD49a(编码其的基因GeneID可以为3672)、IA4(编码其的基因GeneID可以为3732)、CD49D(编码其的基因GeneID可以为3676)、ITGA6(编码其的基因GeneID可以为3655)、VLA-6(编码其的基因GeneID可以为3655)、CD49f(编码其的基因GeneID可以为3655)、ITGAD(编码其的基因GeneID可以为3681)、CD11d(编码其的基因GeneID可以为3681)、ITGAE(编码其的基因GeneID可以为3682)、CD103(编码其的基因GeneID可以为3682)、ITGAL(编码其的基因GeneID可以为3683)、CD11a(编码其的基因GeneID可以为3683)、LFA-1(编码其的基因GeneID可以为3683)、ITGAM(编码其的基因 GeneID可以为3684)、CD11b(编码其的基因GeneID可以为3684)、ITGAX(编码其的基因GeneID可以为3687)、CD11c(编码其的基因GeneID可以为3687)、ITGB1(编码其的基因GeneID可以为3688)、CD29(编码其的基因GeneID可以为3688)、ITGB2(编码其的基因GeneID可以为3689)、CD18(编码其的基因GeneID可以为3689)、LFA-1(编码其的基因GeneID可以为3689)、ITGB7(编码其的基因GeneID可以为3695)、NKG2D(编码其的基因GeneID可以为22914)、NKG2C(编码其的基因GeneID可以为3822)、TNFR2(编码其的基因GeneID可以为7133)、TRANCE/RANKL(编码其的基因GeneID可以为8600)、DNAM1(CD226)(编码其的基因GeneID可以为10666)、SLAMF4(CD244、2B4)(编码其的基因GeneID可以为51744)、CD84(编码其的基因GeneID可以为8832)、CD96(Tactile)(编码其的基因GeneID可以为10225)、CEACAM1(编码其的基因GeneID可以为634)、CRTAM(编码其的基因GeneID可以为56253)、Ly9(CD229)(编码其的基因GeneID可以为4063)、CD160(BY55)(编码其的基因GeneID可以为11126)、PSGL1(编码其的基因GeneID可以为6404)、CD100(SEMA4D)(编码其的基因GeneID可以为10507)、CD69(编码其的基因GeneID可以为969)、SLAMF6(NTB-A、Ly108)(编码其的基因GeneID可以为114836)、SLAM(SLAMF1、CD150、IPO-3)(编码其的基因GeneID可以为6504)、BLAME(SLAMF8)(编码其的基因GeneID可以为56833)、SELPLG(CD162)(编码其的基因GeneID可以为6404)、LTBR(编码其的基因GeneID可以为4055)、LAT(编码其的基因GeneID可以为27040)、GADS(编码其的基因GeneID可以为9402)、SLP-76(编码其的基因GeneID可以为3937)、PAG/Cbp(编码其的基因GeneID可以为55824)、CD19a、和特异性结合CD3的配体、特异性结合CD28的配体、特异性结合HVEM的配体、特异性结合CD40L的配体、特异性结合OX40的配体、和特异性结合4-1BB的配体。共刺激胞内信号传导结构域可以是指T细胞激活剂的胞内部分。胞内信号传导结构域可以包含从中衍生的分子的完整胞内部分或完整天然胞内信号传导结构域或其功能性片段。
在本申请中,术语“T细胞生长因子”通常是指引起细胞增殖的生物活性多肽或小分子化合物。在一种实施方式中,T细胞生长因子可以选自以下组的一种或多种:IL-2(编码其的基因GeneID可以为3558)、IL-4(编码其的基因GeneID可以为3565)、IL-7(编码其的基因GeneID可以为3574)、IL-10(编码其的基因GeneID可以为3586)、IL-12(编码其的基因GeneID可以为3592或3593)、IL-15(编码其的基因GeneID可以为3600)、和γ干扰素(编码其的基因GeneID可以为3458)。
在本申请中,术语“基本上同时”通常是指接触过程的一段时间内TIL可以与两种以上的物质同时接触,但是可以不限于在整个接触过程中TIL总是与两种以上的物质同时接触。在 一种实施方式中,基本上同时可以是指一段时间内TIL可以与至少10%、20%、30%、40%、50%、60%、70%、75%、80%、85%、90%、95%的两种以上的物质的每种物质同时接触。
在本申请中,术语“固相介质”或“介质”通常是指具有结合功能的固相材料。例如,本申请固相介质可以是指通过共价结合和/或非共价结合的作用,将一种或一种以上的物质结合在介质内和/或介质表面的材料。例如,本申请的固相介质可以结合一种或一种以上的T细胞激活剂。例如,本申请的固相介质可以是指通过共价结合和/或非共价结合的作用将CD28抗体或其抗原结合片段以及CD3抗体或其抗原结合片段结合在介质内和/或介质表面的材料。例如,本申请的固相介质可以是包含OKT3抗体和15E8抗体的直径为约500纳米至约10微米的微球。例如,本申请的固相介质可以是聚合物材料。例如,本申请的固相介质可以是直径至少约500纳米的微球。例如,本申请的固相介质可以是纳米基质。例如,本申请的固相介质可以是包含OKT3抗体和15E8抗体的直径为约1纳米至约500纳米的纳米基质。
在本申请中,术语“纳米基质”通常是指一种直径在约1纳米到约500纳米的材料。在本申请中,纳米基质可以具有结合功能,例如,本申请的纳米基质可以结合一种或一种以上的T细胞激活剂。在本申请中,纳米基质可以包含聚合物,例如,本申请的纳米基质可以包含可降解聚合物。在本申请中,纳米基质可以包含多糖、和/或葡聚糖。
在本申请中,术语“基因编辑”通常是指利用一个或多个核酸酶和/或切口酶,将靶DNA(例如细胞的TCRβ基因组)***、替换或去除DNA的一种基因工程。
在本申请中,术语“基因敲除”通常是指使基因沉默和/或无法表达出其编码的蛋白质的基因工程手段。例如,基因敲除可以是指细胞内或体内基因的定向破坏,导致其功能完全丧失。例如,本申请的基因敲除可以使用位点特异性核酸酶。例如,本申请的基因敲除可以使用锌指核酸酶(ZFN)、TAL效应物核酸酶(TALEN)和/或基于CRISPR/Cas的***。例如,本申请的基因敲除可以使用CRISPR/Cas9的***。
在本申请中,术语“树突状细胞”通常是指存在于体内、体外、离体或宿主或受试者内的或可衍生自造血干细胞或单核细胞的抗原递呈细胞。树突状细胞及其前体可以从各种淋巴器官例如脾脏、***以及骨髓和外周血分离。本申请的树突状细胞可以具有特征形态,例如在树突细胞体的多个方向上延伸的薄层(板状伪足)。通常,树突细胞可以表达高水平的MHC和共刺激(例如B7-1和B7-2)分子。树突状细胞可以在体外诱导T细胞的抗原特异性分化,并且能够在体外和体内引发原代T细胞应答。
在本申请中,术语“体外扩增”通常是指经过培养以产生细胞的数量的变化,经扩增的细胞也可以产生细胞的数量和/或比例变化,分泌能力变化,杀伤能力变化或表达能力的变化, 或它们的任何组合。本申请的变化可以是提高或者降低。在本申请中,体外扩增可以是为了扩增目的;为了检测TIL细胞的功能,例如检测TIL细胞释放细胞因子能力,而对TIL细胞进行的操作步骤(例如向TIL细胞的培养基中加入一种或一种以上物质以检测TIL细胞释放细胞因子能力),可以不属于本申请的体外扩增。
在本申请中,术语“外周单个核细胞”或“外周血单个核细胞”通常是指外周血中具有单个核的细胞。例如,在本申请中,本申请的外周血单个核细胞可以包括淋巴细胞、单核细胞和/或树突状细胞。
在本申请中,术语“细胞因子”通常是指由一个细胞群释放的对另一个细胞起细胞间调节剂作用的蛋白。本申请的细胞因子可以是淋巴细胞因子(lymphokines)、单核细胞因子(monokines)和多肽激素。本申请的细胞因子可以包括白细胞介素(ILs)如IL-1、IL-1α、IL-2、IL-3、IL-4、IL-5、IL-6、IL-7、IL-8、IL-9、IL-10、IL-11、IL-21、和/或IL-12。在本申请中,术语细胞因子可以包括来自天然来源或来自重组细胞培养物的蛋白,天然序列细胞因子的生物活性等价物,以及其功能活性片段。
在本申请中,术语“直径”通常是指本申请物质的截面的直径。例如,当本申请的物质不是球形时,则术语“直径”通常是指本申请物质的最大截面的最大直径和/或平均直径。确定物质的直径的方法可以是本领域通用的方法,例如透射电子显微镜。
在本申请中,术语“肿瘤”通常是指任何新的病理性的组织增生。本申请的肿瘤可能是良性的,也可能是恶性的。本申请的肿瘤可能是实体的,也可能是血液的。术语“肿瘤”可以选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
在本申请中,术语“肿瘤组织”通常是指来自对象中的肿瘤,包括对象中的任何实体肿瘤和/或非实体肿瘤的任何组织的样品。
在本申请中,术语“CD28激动剂”通常是指结合细胞表面CD28蛋白并且在细胞中引发应答的化合物。例如,本申请的CD28激动剂可以是结合CD28的小分子制剂。例如,本申请的CD28激动剂可以是结合CD28的抗体或其抗原结合片段。
在本申请中,术语“T细胞亚群比例”通常是指根据不同T细胞亚群占TIL细胞或TIL细胞群中的比例。例如,本申请不同的T细胞亚群具有不同的免疫活性和/或分化能力。例如,本申请的T细胞亚群可以根据T细胞表面标志物进行区分。例如,中心记忆T细胞可以具有CD45RA -CCR7 +的表型。例如,调节性T细胞可以具有CD4 +CD25 +Foxp3 +的表型。例如,活化T细胞可以具有CD25 +、CD28 +、TIM3 +、PD-1 +或41BB +的表型。例如,肿瘤特异性T细 胞可以具有CD103 +CD39 +的表型。例如,干细胞样T细胞可以具有TCF1 +的表型。
在本申请中,术语“TIL细胞数量”通常是指本申请的TIL细胞中细胞数量。在本申请中,TIL细胞数量可以是指本申请任一阶段获得的TIL群中的细胞数量。例如,TIL细胞数量可以是指源自肿瘤组织且未经体外扩增的第一TIL群的细胞数量。例如,TIL细胞数量可以是指经第一阶段体外扩增的第二TIL群的细胞数量。例如,TIL细胞数量可以是指经第二阶段体外扩增的第三TIL群的细胞数量。例如,TIL细胞数量可以是指本申请任意一种培养方法最终获得的TIL的细胞。在本申请中,TIL细胞数量可以通过本领域常用的方法测量,例如可以包括但不限于细胞计数板手动细胞计数和/或自动细胞计数器计数。
在本申请中,术语“约”和“大约”通常是指在统计上有意义的数值范围内。这样的范围可以在给定值或范围的一个数量级内,可以包括在50%内,可以包括在20%内,可以包括在10%内,可以包括在5%内。术语“约”或“大约”所包含的可允许变化可以取决于所研究的特定***,并且本领域普通技术人员可以容易地理解。术语“以上”、“以下”、“至多”和“至少”可以包括本数。
发明详述
一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:使源自肿瘤组织且未经体外扩增的TIL经过至少一个阶段的体外扩增,其中,在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)使源自肿瘤组织且未经体外扩增的第一TIL群与一种或多种T细胞生长因子接触;其中,经所述步骤(A)得到第二TIL细胞群;(B)使所述第二TIL群与本申请一种或多种T细胞生长因子接触和/或一种或多种T细胞激活剂接触;其中,经所述步骤(B)得到第三TIL细胞群;(C)使所述第三TIL群与本申请一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
在一种实施方式中的术语中,本申请的第一阶段体外扩增可以与以上方面的方法中的步骤(A)任意替换使用。在一种实施方式中的术语中,本申请的第二阶段体外扩增可以与以上方面的方法中的步骤(B)任意替换使用。在一种实施方式中的术语中,本申请的经第一阶段体外扩增的TIL可以与经以上方面的方法中步骤(A)得到的第二TIL群任意替换使用。在一种实施方式中的术语中,本申请的经第二阶段体外扩增的TIL可以与经以上方面的方法中步骤(B)得到的第三TIL群任意替换使用。在一种实施方式中的术语中,如有需要,本申请的第三阶段体外扩增可以与以上方面的方法中任意增加的步骤(C)任意替换使用。在一种实 施方式中的术语中,如有需要,本申请的经第三阶段体外扩增的TIL可以与经以上方面的方法中任意增加的步骤(C)得到的第四TIL群任意替换使用。
在一种实施方式中,可以使本申请的源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在本申请的第三阶段体外扩增中,可以使经本申请的第二阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触。
在一种实施方式中,可以使本申请的源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在本申请的第一阶段体外扩增中,可以使经本申请的源自肿瘤组织且未经体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触,且在本申请的第三阶段体外扩增中,可以使经本申请的第二阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触。
在一种实施方式中,可以使本申请的源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在本申请的第二阶段体外扩增中,可以使经本申请的第一阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触,且在本申请的第三阶段体外扩增中,可以使经本申请的第二阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触。
在一种实施方式中,可以使本申请的源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在本申请的第一阶段体外扩增中,可以使经本申请的源自肿瘤组织且未经体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触,且在本申请的第二阶段体外扩增中,可以使经本申请的第一阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触,且在本申请的第三阶段体外扩增中,可以使经本申请的第二阶段体外扩增的TIL与本申请的一种或多种T细胞激活剂和/或本申请的免疫检查点抑制剂接触。
在一种实施方式中,每一个阶段体外扩增之间可以是通过TIL细胞数量的变化来划分的,在一种实施方式中,当TIL细胞的数量增加至少约1倍时,可以认为TIL细胞进入了下一个阶段的体外扩增。在一些实施方式中,当TIL细胞的数量增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、至少约50倍、至少约100倍、至少约200倍、至 少约500倍、或者至少约1000倍时,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,每一个阶段的体外扩增之间也可以是通过TIL细胞培养的条件的变化来划分的。在一种实施方式中,当细胞培养基中添加了或补充添加了T细胞激活剂和/或T细胞生长因子后,可以认为TIL细胞进入了下一个阶段的体外扩增。例如,当细胞培养基中添加了或补充添加了IL-2后,可以认为TIL细胞进入了下一个阶段的体外扩增。例如,当细胞培养基中添加了或补充添加了CD28激动剂后,可以认为TIL细胞进入了下一个阶段的体外扩增。例如,当细胞培养基中添加了或补充添加了免疫检查点抑制剂后,可以认为TIL细胞进入了下一个阶段的体外扩增。例如,当细胞培养基中添加了或补充添加了饲养细胞后,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,当TIL细胞进行了离心和/或细胞洗涤的操作后,可以认为TIL细胞进入了下一个阶段的体外扩增。在一种实施方式中,每一个阶段之间也可以是通过TIL细胞培养的天数来划分的。在一种实施方式中,当TIL细胞体外培养约1天、约2天、约3天、约4天、约5天、约6天、约7天、约8天、约9天、约10天、约11天、约12天、约13天、约14天、约15天、约16天、约17天、约18天、约19天、约20天、约30天、约40天、约50天或约100天后,可以认为TIL细胞进入了下一个阶段的体外扩增。
在一种实施方式中,本申请的第二阶段体外扩增可以进行至多约13天。在一种实施方式中,本申请第二阶段体外扩增进行的天数可以是从第二阶段体外扩增的开始时刻进行计算。例如,第二阶段体外扩增开始的当时,可以认为是第二阶段体外扩增进行了约0天。例如,第二阶段体外扩增开始后进行了约24小时,可以认为是第二阶段体外扩增进行了约1天。例如,第二阶段体外扩增开始的当天,可以认为是第二阶段体外扩增进行了约0天。在一种实施方式中,本申请第二阶段体外扩增进行的天数可以是通过第二阶段体外扩增进行的天数进行计算。例如,第二阶段体外扩增开始后的第二天,可以认为是第二阶段体外扩增进行了约1天。例如,本申请的第二阶段体外扩增可以进行至多约13天、至多约12天、至多约11天、至多约10天、至多约9天、至多约8天、至多约7天、至多约6天、至多约5天、至多约4天、至多约3天、至多约2天或至多约1天。在一种实施方式中,本申请的第二阶段体外扩增可以进行约3天至约13天。在一种实施方式中,本申请的第二阶段体外扩增可以进行约1天至约13天。例如,本申请的第二阶段体外扩增可以进行约2天至约13天、约3天至约13天、约4天至约13天、约5天至约13天、约6天至约13天、约7天至约13天、约8天至约13天、约9天至约13天、约10天至约13天、约11天至约13天、或约12天至约13天。例如,本申请的第二阶段体外扩增可以进行约2天至约3天、约2天至约4天、约2天至约 5天、约2天至约6天、约2天至约7天、约2天至约8天、约2天至约9天、约2天至约10天、约2天至约11天、约2天至约12天、或约2天至约13天。例如,本申请的第二阶段体外扩增可以进行约3天至约4天、约3天至约5天、约3天至约6天、约3天至约7天、约3天至约8天、约3天至约9天、约3天至约10天、约3天至约11天、约3天至约12天、或约3天至约13天。例如,本申请的第二阶段体外扩增可以进行约13天、约12天、约11天、约10天、约9天、约8天、约7天、约6天、约5天、约4天、约3天、约2天或约1天。在一种实施方式中,本申请的第二阶段体外扩增可以认为是REP(rapid expansion protocol)阶段。在一种实施方式中,本申请的第一阶段体外扩增可以认为是preREP阶段。
在一种实施方式中,本申请的第三阶段体外扩增可以进行至多约24小时。在一种实施方式中,本申请第三阶段体外扩增进行的天数可以是从第三阶段体外扩增开始进行计算。例如,第三阶段体外扩增开始的当时,可以认为是第三阶段体外扩增进行了约0天。例如,第三阶段体外扩增开始后进行了约24小时,可以认为是第三阶段体外扩增进行了约1天。例如,第三阶段体外扩增开始的当天,可以认为是第三阶段体外扩增进行了约0天。例如,第三阶段体外扩增开始后的第二天,可以认为是第三阶段体外扩增进行了约1天。例如,本申请的第三阶段体外扩增可以进行至多约24小时、至多约23小时、至多约22小时、至多约21小时、至多约20小时、至多约19小时、至多约18小时、至多约17小时、至多约16小时、至多约15小时、至多约14小时、至多约13小时、或至多约12小时。在一种实施方式中,本申请的第三阶段体外扩增可以进行约12小时至约24小时。例如,本申请的第三阶段体外扩增可以进行约12小时至约24小时、约13小时至约24小时、约14小时至约24小时、约15小时至约24小时、约16小时至约24小时、约17小时至约24小时、约18小时至约24小时、约19小时至约24小时、约20小时至约24小时、约21小时至约24小时、约22小时至约24小时、约23小时至约24小时、约12小时至约22小时、约13小时至约22小时、约14小时至约22小时、约15小时至约22小时、约16小时至约22小时、约17小时至约22小时、约18小时至约22小时、约19小时至约22小时、约20小时至约22小时、约21小时至约22小时、约12小时至约20小时、约13小时至约20小时、约14小时至约20小时、约15小时至约20小时、约16小时至约20小时、约17小时至约20小时、约18小时至约20小时、约19小时至约20小时、约12小时至约18小时、约13小时至约18小时、约14小时至约18小时、约15小时至约18小时、约16小时至约18小时、约17小时至约18小时、约12小时至约16小时、约13小时至约16小时、约14小时至约16小时、约15小时至约16小时、约12小时至约14小时、或约13小时至约14小时。在一种实施方式中,本申请的第二阶段体外扩 增可以认为是REP(rapid expansion protocol)阶段。在一种实施方式中,本申请的第三阶段体外扩增可以认为是reREP阶段。
在一种实施方式中,与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL可以显示出改善的扩增效果。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指源自同一供体的且未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指源自同一供体的经过同样方式分离的且未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指源自同一供体的同一肿瘤来源的且未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指源自同一供体的同一肿瘤来源的经过同样方式分离的且未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指将源自同一供体的TIL细胞分为两组,其中一组未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞可以为未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指将源自同一供体的经过同样方式分离的TIL细胞分为两组,其中一组未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞可以为未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指将源自同一供体的同一肿瘤来源的TIL细胞分为两组,其中一组未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞可以为未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL。在一种实施方式中,未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL可以是指将源自同一供体的同一肿瘤来源的经过同样方式分离的TIL细胞分为两组,其中一组未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的TIL细胞可以为未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL。
在一种实施方式中,本申请的改善的扩增效果可以包含选自以下组的一种或多种:增加 的TIL细胞数量,改善的T细胞亚群比例,提高的细胞因子分泌能力,和提高的肿瘤细胞杀伤能力。
在一种实施方式中,本申请的增加的TIL细胞数量可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的细胞数量可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的增加的TIL细胞数量可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的细胞数量可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。
在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指TIL细胞的选自以下组的细胞因子分泌能力提高:CD107a、GZMB、IL-4、IL-17、IL-6、IL-2、TNF和IFNγ。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的细胞因子分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的细胞因子分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约 18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的CD107a分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的CD107a分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的GZMB分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的GZMB分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约 10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的IL-4、IL-17、IL-6、和/或IL-2分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的IL-4、IL-17、IL-6、和/或IL-2分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的TNF分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的TNF分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少 约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的IFNγ分泌能力可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的细胞因子分泌能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的IFNγ分泌能力可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的TIL的细胞因子分泌能力的测定可以是通过测量TIL细胞的细胞因子表达能力。例如,本申请的免疫细胞的细胞因子分泌能力可以通过细胞流式检测的方法测定。在一种实施方式中,本申请的TIL的细胞因子分泌能力通过测量TIL细胞的细胞因子释放能力测定。在一种实施方式中,本申请的TIL的细胞因子分泌能力是通过CBA法(Cytometric Bead Array)测定。
在一种实施方式中,本申请的提高的肿瘤细胞杀伤能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申请TIL的肿瘤细胞杀伤率可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。在一种实施方式中,本申请的提高的肿瘤细胞杀伤能力可以是指与在体外扩增阶段未曾与本申请T细胞激活剂和/或本申请免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与本申请T细胞激活剂以及本申请免疫检查点抑制剂接触过的本申 请TIL的肿瘤细胞杀伤率可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。在一种实施方式中,本申请的TIL的肿瘤细胞杀伤率可以通过CFSE和DAPI染色法测量。在一种实施方式中,本申请的TIL的肿瘤细胞杀伤可以是指TIL杀伤实体瘤细胞的能力。在一种实施方式中,本申请的TIL的肿瘤细胞杀伤可以是指TIL杀伤***细胞的能力。在一种实施方式中,本申请的TIL的肿瘤细胞杀伤可以是指TIL杀伤Hela细胞的能力。
在一种实施方式中,本申请的改善的T细胞亚群比例可以包含选自以下组的一种或多种:增加的中心记忆T细胞比例,降低的调节性T细胞的比例,降低的耗竭T细胞的比例,增加的活化T细胞比例,增加的肿瘤特异性T细胞比例,和增加的干细胞样T细胞比例。
在一种实施方式中,本申请的增加的中心记忆T细胞比例可以是TIL细胞中CD45RA -CCR7 +细胞的比例的增加。例如,在TIL细胞中中心记忆T细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。
在一种实施方式中,本申请的减少的调节性T细胞的比例可以是TIL细胞中CD4 +CD25 +Foxp3 +细胞的比例的减少。例如,在TIL细胞中调节性T细胞比例可以减少至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。
在一种实施方式中,本申请的减少的耗竭T细胞的比例可以是TIL细胞中TIM3 +细胞的比例的减少。例如,在TIL细胞中耗竭T细胞比例可以减少至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约 20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%。
在一种实施方式中,本申请的增加的活化T细胞比例可以是TIL细胞中CD25 +、CD28 +、CD27 +、PD-1 +或41BB +细胞的比例的增加。例如,在TIL细胞中活化T细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。例如,在TIL细胞中CD25 +细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。例如,在TIL细胞中CD28 +细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至 少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。例如,在TIL细胞中CD27 +细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。例如,在TIL细胞中PD-1 +细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。例如,在TIL细胞中41BB +细胞比例可以增加至少约100%、至少约90%、至少约80%、至少约70%、至少约60%、至少约50%、至少约40%、至少约30%、至少约20%、至少约19%、至少约18%、至少约17%、至少约16%、至少约15%、至少约14%、至少约13%、至少约12%、至少约11%、至少约10%、至少约9%、至少约8%、至少约7%、至少约6%、至少约5%、至少约4%、至少约3%、至少约2%、至少约1%、至少约0.5%、至少约0.4%、至少约0.3%、至少约0.2%、或至少约0.1%,或可以增加至少约1倍、至少约2倍、至少约3倍、至少约4倍、至少约5倍、至少约6倍、至少约7倍、至少约8倍、至少约9倍、至少约10倍、至少约11倍、至少约12倍、至少约13倍、至少约14倍、至少约15倍、至少约20倍、至少约30倍、至少约40倍、或者至少约50倍。
在一种实施方式中,本申请的方法还可以包含:在至少一个阶段的本申请体外扩增中,使本申请TIL与本申请一种或多种T细胞激活剂接触。
在一种实施方式中,在单个阶段的本申请的体外扩增中,T细胞激活剂可以包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。使本申请的TIL与本申请的一种或多种免疫检查点抑制剂接触且与本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第一阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种免疫检查点抑制剂接触且与本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第二阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种免疫检查点抑制剂接触且与本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第三阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种免疫检查点抑制剂接触且与本申请的一种或多种T细胞激活剂接触。
在一种实施方式中,在单个阶段的本申请的体外扩增中,可以使本申请的TIL基本上同时与本申请的一种或多种免疫检查点抑制剂以及本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第一阶段体外扩增中,可以使本申请的TIL基本上同时与本申请的一种或多种免疫检查点抑制剂以及本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第二阶段体外扩增中,可以使本申请的TIL基本上同时与本申请的一种或多种免疫检查点抑制剂以及本申请的一种或多种T细胞激活剂接触。在一种实施方式中,在本申请第三阶段体外扩增中,可以使本申请的TIL基本上同时与本申请的一种或多种免疫检查点抑制剂以及本申请的一种或多种T细胞激活剂接触。
在一种实施方式中,本申请的T细胞激活剂可以包含选自以下组的一种或多种:CD80、CD86、B7-H3、4-1BBL、CD27、CD30、CD134、B7h、CD40、LIGHT、以及它们的功能活性片段。在一种实施方式中,本申请的T细胞激活剂可以包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。在一种实施方式中,本申请的T细胞激活剂可以包含选自以下组:CD3、CD28、HVEM、CD40L、OX40和4-1BB的抗体以及它们的抗原结合片段。在一种实施方式中,本申请的T细胞激活剂可以包含CD3激动剂。在一种实施方式中,本申请的T细胞激活剂可以包含抗CD3的抗体和/或其抗原结合片段,例如可以是Miltenyi Biotech的OKT3、可以是BD的SP34。在一种实施方式中,本申请的T细胞激活剂可以包含抗CD3的抗体和/或其抗原结合片段,例如可以是包含Miltenyi Biotech的OKT3的HCDR1-3和/或Miltenyi Biotech的OKT3的LCDR1-3,例如可以是包含BD的SP34的HCDR1-3和/或Miltenyi Biotech的OKT3的LCDR1-3,例如可以是包含Miltenyi Biotech的OKT3的VH和/或Miltenyi Biotech的OKT3的VL,例如可以是包含BD的SP34的VH和/或Miltenyi Biotech的OKT3的VL。在一种实施方式中,本申请的T细胞激活剂可 以包含CD28激动剂。在一种实施方式中,本申请的T细胞激活剂可以包含抗CD28的抗体和/或其抗原结合片段,例如可以是Sigma-Aldrich的15E8。在一种实施方式中,本申请的T细胞激活剂可以包含抗CD28的抗体和/或其抗原结合片段,例如可以是包含Sigma-Aldrich的15E8的HCDR1-3和/或Sigma-Aldrich的15E8的LCDR1-3,例如可以是包含Sigma-Aldrich的15E8的VH和/或Sigma-Aldrich的15E8的VL。
在一种实施方式中,使本申请的TIL与本申请的一种或多种T细胞激活剂接触可以包含选自以下组的一种或多种方式:(1)将本申请的T细胞激活剂添加至本申请的TIL的细胞培养基中;(2)将表达本申请的T细胞激活剂的工程化细胞添加至本申请的TIL的细胞培养基中;(3)将包含本申请的T细胞激活剂的固相介质添加至本申请的TIL的细胞培养基中。在一种实施方式中,使本申请的TIL与本申请的一种或多种T细胞激活剂接触可以包含将包含本申请的T细胞激活剂的固相介质添加至本申请的TIL的细胞培养基中。在一种实施方式中,使本申请的TIL与本申请的一种或多种T细胞激活剂接触可以包含将包含本申请的CD28抗体与CD3抗体的固相介质添加至本申请的TIL的细胞培养基中。
在一种实施方式中,所述T细胞激活剂在本申请TIL的细胞培养基中的初始浓度可以为至少约30ng/mL。例如,本申请的CD28抗体在本申请TIL的细胞培养基中的初始浓度可以为至少约30ng/mL;例如,本申请的CD3抗体在本申请TIL的细胞培养基中的初始浓度可以为至少约30ng/mL。例如,本申请的CD28抗体初始浓度的选择可以与本申请的CD3抗体初始浓度的选择相互独立;例如,本申请的CD28抗体与本申请的CD3抗体在本申请TIL的细胞培养基中的初始浓度可以任意组合。例如,本申请的CD28抗体在本申请TIL的细胞培养基中的初始浓度可以任意选自约30ng/mL-约300ng/mL。例如,本申请的CD3抗体在本申请TIL的细胞培养基中的初始浓度可以任意选自约30ng/mL-约300ng/mL。例如,本申请的CD28抗体在本申请TIL的细胞培养基中的初始浓度可以任意选自约30ng/mL-约300ng/mL,且本申请的CD3抗体在本申请TIL的细胞培养基中的初始浓度可以任意选自约30ng/mL-约300ng/mL,本申请的CD28抗体初始浓度的选择可以与本申请的CD3抗体初始浓度的选择相互独立。在一种实施方式中,本申请的固相介质的直径可以为约500纳米至约10微米。在一种实施方式中,本申请的固相介质的直径可以通过透射电子显微镜测量。在一种实施方式中,本申请的固相介质的直径可以为约1纳米至约500纳米。在一种实施方式中,本申请的固相介质的直径可以为约100纳米至约500纳米。在一种实施方式中,本申请的固相介质的直径可以为约200纳米至约500纳米。在一种实施方式中,本申请的固相介质的直径可以通过透射电子显微镜测量。
在一种实施方式中,本申请的固相介质可以包含聚合物。在一种实施方式中,本申请的固相介质可以包含葡聚糖。
在一种实施方式中,每mg本申请的固相介质包含至少约25μg的本申请的T细胞激活剂。例如,每mg本申请的固相介质包含的每一种的T细胞激活剂的量可以是相互独立的。
在一种实施方式中,以约1:100-约1:2000的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂的固相介质添加至本申请TIL的细胞培养基中。在一种实施方式中,以约2:1-约1:2的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂的固相介质添加至本申请TIL的细胞培养基中。
例如,当本申请的固相介质的直径为约100纳米至约500纳米时,可以以约2:1-约1:2的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂的固相介质添加至本申请TIL的细胞培养基中。例如,当本申请的固相介质的直径为约100纳米至约500纳米时,可以以约2:1-约1:2、以约2:1-约1:1、或以约1:1-约1:2的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂,例如CD3激动剂和/或CD28激动剂的固相介质添加至本申请TIL的细胞培养基中。
例如,当本申请的固相介质的直径为约100纳米至约500纳米时,可以以约1:100-约1:2000的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂的固相介质添加至本申请TIL的细胞培养基中。例如,当本申请的固相介质的直径为约100纳米至约500纳米时,可以以约1:100-约1:2000、以约1:200-约1:2000、以约1:300-约1:2000、以约1:400-约1:2000、以约1:500-约1:2000、以约1:600-约1:2000、以约1:700-约1:2000、以约1:800-约1:2000、以约1:900-约1:2000、以约1:1000-约1:2000、以约1:1200-约1:2000以约1:1400-约1:2000、以约1:1600-约1:2000、或以约1:1800-约1:2000的本申请固相介质与本申请TIL的比例,将包含本申请一种或多种T细胞激活剂,例如CD3激动剂和/或CD28激动剂的固相介质添加至本申请TIL的细胞培养基中。
在一种实施方式中,本申请的免疫检查点抑制剂包含抑制PD-1与PD-L1和/或PD-L2相互作用的物质。例如,本申请的免疫检查点抑制剂可以包含PD-1抑制剂;例如,本申请的免疫检查点抑制剂可以包含PD-1的抗体和/或其抗原结合片段。在一种实施方式中,本申请的免疫检查点抑制剂包含具有以约100pM或更低的K D值结合PD-1的能力的物质,例如,约100pM或更低的K D值、约90pM或更低的K D值、约80pM或更低的K D值、约50pM或更低的K D值、约30pM或更低的K D值、或约10pM或更低的K D值。在一种实施方式中,本申请的免疫检查点抑制剂包含具有以约100pM或更低的EC 50值结合PD-1的能力的物质, 例如,约100pM或更低的EC 50值、约90pM或更低的EC 50值、约80pM或更低的EC 50值、约50pM或更低的EC 50值、约30pM或更低的EC 50值、或约10pM或更低的EC 50值。在一种实施方式中,本申请的免疫检查点抑制剂包含具有以约1nM或更低的IC 50值抑制PD-1与PD-L1和/或PD-L2结合的能力的物质,例如,约1nM或更低的IC 50值、约0.9nM或更低的IC 50值、约0.8nM或更低的IC 50值、约0.5nM或更低的IC 50值、约0.3nM或更低的IC 50值、或约0.1nM或更低的IC 50值。
在一种实施方式中,本申请的免疫检查点抑制剂可以为PD-1抗体或其抗原结合蛋白。
在本申请中,本申请抗体或其抗原结合蛋白包含抗体重链可变区VH中的至少一个CDR。本申请CDR可以是根据IMGT命名法定义的,本申请CDR可以是根据Chothia定义的,或本申请CDR可以是根据Kabat定义的。例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在本申请中,本申请抗体或其抗原结合蛋白包含抗体重链可变区VH中的至少一个CDR。本申请CDR可以是根据IMGT命名法定义的,或本申请CDR可以是根据Kabat定义的。
例如,本申请的抗体或其抗原结合蛋白可以包含HCDR1,且本申请HCDR1可以包含SEQ ID NO:1和15中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含HCDR2,且本申请HCDR2可以包含SEQ ID NO:2和16中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含HCDR3,且本申请HCDR3可以包含SEQ ID NO:3和17中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含HCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:1和15中任一项所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:2和16中任一项所示的氨基酸序列,且本申请HCDR3可以包含SEQ ID NO:3和17中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6或Hu_6H6相同的HCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:1所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:2所示的氨基酸序列,且本申请HCDR3可以包含SEQ ID NO:3所示的氨基酸序列; 本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的HCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:15所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:16所示的氨基酸序列,且本申请HCDR3可以包含SEQ ID NO:17所示的氨基酸序列;本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在本申请中,本申请抗体或其抗原结合蛋白包含抗体轻链可变区VL中的至少一个CDR。本申请CDR可以是根据IMGT命名法定义的,或本申请CDR可以是根据Kabat定义的。
例如,本申请的抗体或其抗原结合蛋白可以包含LCDR1,且本申请LCDR1可以包含SEQ ID NO:4和18中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含LCDR2,且本申请LCDR2可以包含SEQ ID NO:5和19中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含LCDR3,且本申请LCDR3可以包含SEQ ID NO:6和20中任一项所示的氨基酸序列;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含LCDR1-3,其中本申请LCDR1可以包含SEQ ID NO:4和18中任一项所示的氨基酸序列,本申请LCDR2可以包含SEQ ID NO:5和19中任一项所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:6和20中任一项所示的氨基酸序列;本申请CDR可以是根据IMGT命名法定义的;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6或Hu_6H6相同的LCDR1-3,其中本申请LCDR1可以包含SEQ ID NO:4所示的氨基酸序列,本申请LCDR2可以包含SEQ ID NO:5所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:6所示的氨基酸序列;本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的LCDR1-3,其中本申请LCDR1可以包含SEQ ID NO:18所示的氨基酸序列,本申请LCDR2可以包含SEQ  ID NO:19所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:20所示的氨基酸序列;本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含HCDR1-3和LCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:1和15中任一项所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:2和16中任一项所示的氨基酸序列,本申请HCDR3可以包含SEQ ID NO:3和17中任一项所示的氨基酸序列,本申请LCDR1可以包含SEQ ID NO:4和18中任一项所示的氨基酸序列,本申请LCDR2可以包含SEQ ID NO:5和19中任一项所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:6和20中任一项所示的氨基酸序列;本申请CDR可以是根据IMGT命名法定义的;本申请CDR可以是根据Kabat定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6或Hu_6H6相同的HCDR1-3和LCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:1所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:2所示的氨基酸序列,本申请HCDR3可以包含SEQ ID NO:3所示的氨基酸序列,本申请LCDR1可以包含SEQ ID NO:4所示的氨基酸序列,本申请LCDR2可以包含SEQ ID NO:5所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:6所示的氨基酸序列;本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的HCDR1-3和LCDR1-3,其中本申请HCDR1可以包含SEQ ID NO:15所示的氨基酸序列,本申请HCDR2可以包含SEQ ID NO:16所示的氨基酸序列,本申请HCDR3可以包含SEQ ID NO:17所示的氨基酸序列,本申请LCDR1可以包含SEQ ID NO:18所示的氨基酸序列,本申请LCDR2可以包含SEQ ID NO:19所示的氨基酸序列,且本申请LCDR3可以包含SEQ ID NO:20所示的氨基酸序列;本申请CDR可以是根据Kabat命名法定义的;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含重链可变区VH,且本申请VH可包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6相同的VH,且本申请VH可包含SEQ ID NO:7所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能 力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的VH,且本申请VH可包含SEQ ID NO:11所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的VH,且本申请VH可包含SEQ ID NO:21所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含轻链可变区VL,且本申请VL可包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6相同的VL,且本申请VL可包含SEQ ID NO:8所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的VL,且本申请VL可包含SEQ ID NO:12所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的VL,且本申请VL可包含SEQ ID NO:22所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含重链可变区VH和轻链可变区VL,且本申请VH可包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列,本申请VL可包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与一种6H6相同的VH和VL,且本申请VH可包含SEQ ID NO:7所示的氨基酸序列,本申请VL可包含SEQ ID NO:8所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的VH和VL,且本申请VH可包含SEQ ID NO:11所示的氨基酸序列,本申请VL可包含SEQ ID NO:12所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的VH和VL,且本申请VH可包含SEQ ID NO:21所示的氨基酸序列,本申请VL可包含SEQ ID NO:22所 示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含重链,且本申请重链可包含SEQ ID NO:9、13和23中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6相同的重链,且本申请重链可包含SEQ ID NO:9所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的重链,且本申请重链可包含SEQ ID NO:13所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的重链,且本申请重链可包含SEQ ID NO:23所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含轻链,且本申请轻链可包含SEQ ID NO:10、14和24中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6相同的轻链,且本申请轻链可包含SEQ ID NO:10所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的轻链,且本申请轻链可包含SEQ ID NO:14所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的轻链,且本申请轻链可包含SEQ ID NO:24所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体或其抗原结合蛋白可以包含重链和轻链,且本申请重链可包含SEQ ID NO:9、13和23中任一项所示的氨基酸序列,本申请轻链可包含SEQ ID NO:10、14和24中任一项所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与6H6相同的重链和轻链,且本申请重 链可包含SEQ ID NO:9所示的氨基酸序列,本申请轻链可包含SEQ ID NO:10所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Hu_6H6相同的重链和轻链,且本申请重链可包含SEQ ID NO:13所示的氨基酸序列,本申请轻链可包含SEQ ID NO:14所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
例如,本申请的抗体或其抗原结合蛋白可以包含与Pembrolizumab相同的重链和轻链,且本申请重链可包含SEQ ID NO:23所示的氨基酸序列,本申请轻链可包含SEQ ID NO:24所示的氨基酸序列;例如,本申请的抗原结合蛋白可以具有PD-1结合能力。
在一种实施方式中,本申请的抗体选自以下组:嵌合抗体,人源化抗体和全人源抗体。在一种实施方式中,本申请的抗原结合片段选自以下组:Fab,Fab′,Fv片段,F(ab') 2,F(ab) 2,scFv,di-scFv,VHH和dAb。
在一种实施方式中,每一种本申请的免疫检查点抑制剂在本申请的TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL。例如,PD-1抗体在本申请的TIL的细胞培养基中的初始浓度可以为至少约0.1μg/mL、至少约0.2μg/mL、至少约0.3μg/mL、至少约0.4μg/mL、至少约0.5μg/mL、至少约0.6μg/mL、至少约0.7μg/mL、至少约0.8μg/mL、至少约0.9μg/mL、至少约1μg/mL、至少约2μg/mL、至少约3μg/mL、至少约4μg/mL、至少约5μg/mL、至少约6μg/mL、至少约7μg/mL、至少约8μg/mL、至少约9μg/mL、至少约10μg/mL、至少约11μg/mL、至少约12μg/mL、至少约13μg/mL、至少约14μg/mL、至少约15μg/mL、至少约16μg/mL、至少约17μg/mL、至少约18μg/mL、至少约19μg/mL或至少约20μg/mL。在一种实施方式中,每一种本申请的免疫检查点抑制剂在本申请的TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL-约20μg/mL。例如,PD-1抗体在本申请的TIL的细胞培养基中的初始浓度可以为约0.1μg/mL-约20μg/mL、约0.2μg/mL-约20μg/mL、约0.3μg/mL-约20μg/mL、约0.5μg/mL-约20μg/mL、约0.7μg/mL-约20μg/mL、约1μg/mL-约20μg/mL、约3μg/mL-约20μg/mL、约5μg/mL-约20μg/mL、约7μg/mL-约20μg/mL、约10μg/mL-约20μg/mL、约15μg/mL-约20μg/mL、约17μg/mL-约20μg/mL、约19μg/mL-约20μg/mL、约0.1μg/mL-约15μg/mL、约0.2μg/mL-约15μg/mL、约0.3μg/mL-约15μg/mL、约0.5μg/mL-约15μg/mL、约0.7μg/mL-约15μg/mL、约1μg/mL-约15μg/mL、约3μg/mL-约15μg/mL、约5μg/mL-约15μg/mL、约7μg/mL-约15μg/mL、约10μg/mL-约15μg/mL、约0.1μg/mL-约10μg/mL、约0.2μg/mL-约10μg/mL、约0.3μg/mL-约10μg/mL、约0.5μg/mL-约10μg/mL、约0.7μg/mL-约10μg/mL、约1μg/mL-约10μg/mL、约3μg/mL-约10μg/mL、 约5μg/mL-约10μg/mL、约7μg/mL-约10μg/mL、约0.1μg/mL-约5μg/mL、约0.2μg/mL-约5μg/mL、约0.3μg/mL-约5μg/mL、约0.5μg/mL-约5μg/mL、约0.7μg/mL-约5μg/mL、约1μg/mL-约5μg/mL、约3μg/mL-约5μg/mL、约0.1μg/mL-约1μg/mL、约0.2μg/mL-约1μg/mL、约0.3μg/mL-约1μg/mL、约0.5μg/mL-约1μg/mL、约0.7μg/mL-约1μg/mL、约0.1μg/mL-约0.5μg/mL、约0.2μg/mL-约0.5μg/mL、或约0.3μg/mL-约0.5μg/mL。
在一种实施方式中,本申请的方法还可以包含:在至少一个阶段的本申请体外扩增中,使本申请TIL与一种或多种T细胞生长因子接触。
在一种实施方式中,在单个阶段的本申请体外扩增中,可以使本申请的TIL与本申请的一种或多种T细胞激活剂接触且与本申请的一种或多种T细胞生长因子接触。例如,在本申请第一阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种T细胞激活剂接触且与本申请的一种或多种T细胞生长因子接触。例如,在本申请第二阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种T细胞激活剂接触且与本申请的一种或多种T细胞生长因子接触。例如,在本申请第三阶段体外扩增中,可以使本申请的TIL与本申请的一种或多种T细胞激活剂接触且与本申请的一种或多种T细胞生长因子接触。
在一种实施方式中,在单个阶段的本申请体外扩增中,使本申请TIL基本上同时与本申请一种或多种T细胞激活剂以及本申请一种或多种T细胞生长因子接触。例如,在本申请第一阶段体外扩增中,使本申请TIL基本上同时与本申请一种或多种T细胞激活剂以及本申请一种或多种T细胞生长因子接触。例如,在本申请第二阶段体外扩增中,使本申请TIL基本上同时与本申请一种或多种T细胞激活剂以及本申请一种或多种T细胞生长因子接触。例如,在本申请第三阶段体外扩增中,使本申请TIL基本上同时与本申请一种或多种T细胞激活剂以及本申请一种或多种T细胞生长因子接触。
在一种实施方式中,本申请的T细胞生长因子可以选自以下组的一种或多种:IL-2、IL-7、IL-12、IL-15、IL-21、γ干扰素、以及它们的功能活性片段。在一种实施方式中,本申请的T细胞生长因子可以包含IL-2和/或其功能活性片段。例如,IL-2的功能活性片段可以包含本领域已知的可以与T细胞的IL-2受体结合的IL-2的片段。
在一种实施方式中,本申请的TIL与本申请一种或多种T细胞生长因子接触可以包含将本申请T细胞生长因子添加至本申请TIL的细胞培养基中。在一种实施方式中,本申请的T细胞生长因子在本申请TIL的细胞培养基中的初始浓度可以为至少约300IU/mL。在一种实施方式中,本申请IL-2在本申请TIL的细胞培养基中的初始浓度可以为至少约350IU/mL、至少约400IU/mL、至少约500IU/mL、至少约600IU/mL、至少约700IU/mL、至少约800 IU/mL、至少约900IU/mL、至少约1000IU/mL、至少约1100IU/mL、至少约1200IU/mL、至少约1300IU/mL、至少约1400IU/mL、至少约1500IU/mL、至少约2000IU/mL、至少约2500IU/mL、至少约2600IU/mL、至少约2700IU/mL、至少约2800IU/mL、至少约2900IU/mL、至少约3000IU/mL、至少约3100IU/mL、至少约3200IU/mL、至少约3300IU/mL、至少约3400IU/mL、至少约3500IU/mL、至少约4000IU/mL、至少约4500IU/mL、至少约5000IU/mL、至少约5500IU/mL、至少约6000IU/mL、至少约6500IU/mL、至少约7000IU/mL、至少约7500IU/mL、至少约8000IU/mL、至少约8500IU/mL、或至少约9000IU/mL。
在一种实施方式中,本申请的方法还可以包含:在至少一个阶段的本申请体外扩增中,可以使本申请TIL与饲养细胞共培养。
在一种实施方式中,在单个阶段的本申请体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触且与本申请饲养细胞共培养,在一种实施方式中,单个阶段的本申请体外扩增可以指在同一个阶段的本申请的体外扩增,例如,可以同在本申请的第一阶段体外扩增、可以同在本申请的第二阶段体外扩增、或可以同在本申请的第三阶段体外扩增等。
在一种实施方式中,本申请的在本申请第一阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触且与本申请饲养细胞共培养。在一种实施方式中,本申请的在本申请第二阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触且与本申请饲养细胞共培养。在一种实施方式中,本申请的在本申请第三阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触且与本申请饲养细胞共培养。
在一种实施方式中,在单个阶段的本申请体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触一定时间之后,再与本申请饲养细胞共培养。在一种实施方式中,在本申请第一阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触一定时间之后,再与本申请饲养细胞共培养。在一种实施方式中,在本申请第二阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触一定时间之后,再与本申请饲养细胞共培养。在一种实施方式中,在本申请第三阶段体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触一定时间之后,再与本申请饲养细胞共培养。
在一种实施方式中,在单个阶段的本申请体外扩增中,可以使本申请TIL与本申请一种或多种T细胞激活剂和/或一种或多种T细胞生长因子接触一定时间之后,再与本申请饲养细胞共培养。在一种实施方式中,本申请的一定时间可以为至少约2小时。在一种实施方式中,本申请的一定时间可以为至少约1小时、至少约2小时、至少约3小时、至少约4小时、至少约5小时、至少约6小时、至少约7小时、至少约8小时、至少约9小时、至少约10小时、至少约11小时、至少约12小时、至少约13小时、至少约14小时、至少约15小时、至少约16小时、至少约17小时、至少约18小时、至少约19小时、至少约20小时、至少约21小时、至少约22小时、至少约23小时、至少约24小时、至少约36小时、至少约48小时、至少约60小时或至少约72小时。在一种实施方式中,本申请的一定时间可以为约6小时至约72小时。在一种实施方式中,本申请的一定时间可以为约6小时到约7小时、约6小时到约8小时、约6小时到约9小时、约6小时到约10小时、约6小时到约11小时、约6小时到约12小时、约6小时到约13小时、约6小时到约14小时、约6小时到约15小时、约6小时到约16小时、约6小时到约17小时、约6小时到约18小时、约6小时到约19小时、约6小时到约20小时、约6小时到约21小时、约6小时到约22小时、约6小时到约23小时、约6小时到约24小时、约6小时到约36小时、约6小时到约48小时、约6小时到约60小时或约6小时到约72小时。在一种实施方式中,本申请的一定时间可以为约12小时到约13小时、约12小时到约14小时、约12小时到约15小时、约12小时到约16小时、约12小时到约17小时、约12小时到约18小时、约12小时到约19小时、约12小时到约20小时、约12小时到约21小时、约12小时到约22小时、约12小时到约23小时、约12小时到约24小时、约12小时到约36小时、约12小时到约48小时、约12小时到约60小时或约12小时到约72小时。在一种实施方式中,本申请的一定时间可以为约1小时、约2小时、约3小时、约4小时、约5小时、约6小时、约7小时、约8小时、约9小时、约10小时、约11小时、约12小时、约13小时、约14小时、约15小时、约16小时、约17小时、约18小时、约19小时、约20小时、约21小时、约22小时、约23小时、约24小时、约36小时、约48小时、约60小时或约72小时。
在一种实施方式中,本申请的饲养细胞可以包含抗原呈递细胞。在一种实施方式中,本申请的饲养细胞可以包含选自以下组的一种或多种:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。在一种实施方式中,本申请的饲养细胞可以为外周单个核细胞。在一种实施方式中,本申请的饲养细胞可以为经过辐照的饲养细胞。例如,本申请的饲养细胞可以为分离的人工抗原呈递细胞(aAPC),本申请的人工抗原呈递细胞可以包含表达HLA-A/B/C、CD64、 CD80、ICOS-L和/或CD58的细胞,并可以被修饰以表达一种以上本申请的T细胞激活剂。在一种实施方式中,本申请的饲养细胞可以经过辐照,例如,可以经过伽马射线辐照,或可以经过X射线辐照。
在一种实施方式中,本申请的TIL与本申请的饲养细胞共培养可以包含使本申请的饲养细胞的表面与本申请的TIL的表面相接触。在一种实施方式中,本申请的TIL与本申请的饲养细胞共培养包含将本申请的饲养细胞添加至本申请的TIL的细胞培养基中。
在一种实施方式中,本申请可以以约40:1-约400:1的本申请饲养细胞与本申请TIL的比例,将本申请饲养细胞添加至本申请TIL的细胞培养基中。在一种实施方式中,本申请可以以约40:1-约400:1、以约40:1-约300:1、以约40:1-约200:1、以约40:1-约100:1、以约40:1-约90:1、以约40:1-约80:1、以约40:1-约70:1、以约40:1-约60:1、以约40:1-约50:1、以约50:1-约400:1、以约60:1-约400:1、以约70:1-约400:1、以约80:1-约400:1、以约90:1-约400:1、以约100:1-约400:1、以约200:1-约400:1、或以约300:1-约400:1的本申请饲养细胞与本申请TIL的比例,将本申请饲养细胞添加至本申请TIL的细胞培养基中。
在一种实施方式中,本申请的TIL可以为源自肿瘤组织的碎片的TIL。在一种实施方式中,可以通过将肿瘤组织处理成肿瘤碎片获得本申请的TIL。在一种实施方式中,本申请的肿瘤碎片的体积约为1-27立方毫米。在一种实施方式中,本申请的肿瘤碎片的体积约为约1立方毫米、约2立方毫米、约3立方毫米、约4立方毫米、约5立方毫米、约6立方毫米、约7立方毫米、约8立方毫米、约9立方毫米、约10立方毫米、约11立方毫米、约12立方毫米、约13立方毫米、约15立方毫米、约17立方毫米、约19立方毫米、约20立方毫米、约21立方毫米、约23立方毫米、约24立方毫米、约25立方毫米、约26立方毫米或27立方毫米。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与T细胞生长因子接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与T细胞生长因子接触、与T细胞激活剂接触且使所述TIL与饲养细胞共培养;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与T细胞激活剂接触以及与免疫检查点抑制剂接触,步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与T细胞激活剂接触且使所述TIL 与饲养细胞共培养;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与T细胞激活剂接触以及与免疫检查点抑制剂接触,步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与T细胞激活剂(例如可以包含CD3抗体以及CD28抗体的纳米基质、CD3抗体或CD3抗体和CD28抗体的混合物)接触且使所述TIL与饲养细胞共培养;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与T细胞激活剂(例如可以包含CD3抗体以及CD28抗体的纳米基质、CD3抗体或CD3抗体和CD28抗体的混合物)接触以及与PD-1抗体接触,所述PD-1抗体的重链可以包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述PD-1抗体的轻链可以包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL,步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与T细胞激活剂(例如可以包含CD3抗体以及CD28抗体的纳米基质、CD3抗体或CD3抗体和CD28抗体的混合物)接触且使所述TIL与饲养细胞共培养;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与T细胞激活剂(例如可以包含CD3抗体以及CD28抗体的纳米基质、CD3抗体或CD3抗体和CD28抗体的混合物)接触以及与PD-1抗体接触,所述PD-1抗体的重链可以包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述PD-1抗体的轻链可以包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL,所述PD-1抗体在所述TIL的细胞培养基中的初始浓度可以至少为约0.1μg/mL,步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与包含CD3抗体以及CD28抗体的纳米基质接触且使所述TIL与饲养细胞共培养;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与包含CD3抗体以及CD28抗体的纳米基质接触以及与PD-1抗体接触,所述PD-1抗体的重链可以包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述PD-1抗体的轻链可以包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL,所述PD-1抗体在所述TIL的细胞培养基中的初始浓度可以至少为约0.1μg/mL, 步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与包含CD3抗体以及CD28抗体的纳米基质接触且在步骤(B)进行至少约2小时之后使所述TIL与饲养细胞共培养,所述纳米基质的直径可以为约1纳米至约500纳米,每mg所述纳米基质可以分别包含CD3抗体以及CD28抗体各为约25μg,所述饲养细胞可以包含外周单个核细胞;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与包含CD3抗体以及CD28抗体的纳米基质接触以及与PD-1抗体接触,所述纳米基质的直径可以为约1纳米至约500纳米,每mg所述纳米基质可以分别包含CD3抗体以及CD28抗体各为约25μg,所述PD-1抗体的重链可以包含氨基酸序列如SEQ ID NO:1和15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述PD-1抗体的轻链可以包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3,所述PD-1抗体在所述TIL的细胞培养基中的初始浓度可以至少为约0.1μg/mL,步骤(C)可以进行至多约24小时。
在另一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法,其可以包含:(A)可以使源自肿瘤组织且未经体外扩增的第一TIL群与IL-2接触,所述IL-2在所述TIL的细胞培养基中的初始浓度可以为至少约300IU/mL;其中,经所述步骤(A)得到第二TIL群;(B)可以使所述第二TIL群与IL-2接触、与包含CD3抗体以及CD28抗体的纳米基质接触且在步骤(B)进行至少约2小时之后使所述TIL与饲养细胞共培养,所述IL-2在所述TIL的细胞培养基中的初始浓度可以为至少约300IU/mL,所述纳米基质的直径可以为约1纳米至约500纳米,每mg所述纳米基质可以分别包含CD3抗体以及CD28抗体各为约25μg,可以以约1:100-约1:2000的所述纳米基质与所述TIL的比例添加至所述TIL的细胞培养基中,所述饲养细胞可以包含外周单个核细胞,可以以约40:1-约400:1的所述饲养细胞与所述TIL的比例,将所述饲养细胞添加至所述TIL的细胞培养基中;其中,经所述步骤(B)得到第三TIL群;(C)可以使所述第三TIL群与包含CD3抗体以及CD28抗体的纳米基质接触以及与PD-1抗体接触,所述纳米基质的直径可以为约1纳米至约500纳米,每mg所述纳米基质可以分别包含CD3抗体以及CD28抗体各为约25μg,可以以约1:100-约1:2000的所述纳米基质与所述TIL的比例添加至所述TIL的细胞培养基中,所述PD-1抗体的重链可以包含氨基 酸序列如SEQ ID NO:1和15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述PD-1抗体的轻链可以包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3,所述PD-1抗体在所述TIL的细胞培养基中的初始浓度可以至少为约0.1μg/mL,步骤(C)可以进行至多约24小时。
一方面,本申请提供一种培养肿瘤浸润淋巴细胞(TIL)的方法。从受试者组织样品获得的TIL细胞的方法可以是患者手术取得原位肿瘤样本或转移肿瘤样本,重量可以至少约1g,也可以多块组织合并。肿瘤组织在样本运输液,例如可以是商业常用的肿瘤组织运输液、肿瘤组织保存液或肿瘤组织转运液,内约2-8度运输,48小时内处理。组织块可以机械破碎至每块约1-27立方毫米大小,转移入透气培养袋或Grex中,加入T细胞无血清培养基和浓度为300-9000IU/mL(例如可以是1000-9000IU/mL,例如可以是6000IU/mL)的IL-2培养约3-14天。收集培养基中细胞,转移入透气培养袋、或Grex、或Xuri设备,T细胞无血清培养基可以添加本申请的CD28抗体、CD3抗体以及CD28抗体、包含CD3抗体以及CD28抗体的磁珠(例如Dynabeads)和/或包含CD3抗体以及CD28抗体的纳米基质(例如transACT)以及浓度为300-9000IU/mL(例如可以是1000-9000IU/mL,例如可以是6000IU/mL)的IL-2,活化本申请的一定时间后,添加辐照PBMC(TIL与PBMC按照比率约1:40-约1:400),扩增培养约3-14天。可以使用细胞处理***收集培养基中细胞,并且可以使用本申请的CD3抗体和PD-1抗体(例如可以是至少为约0.1μg/mL),以及任选地CD28抗体培养本申请的TIL细胞,例如可以培养约12至约24小时。之后清洗冻存,并检测。最终产品CD3比例可以大于80%,细胞活率可以大于70%,大于80%的T细胞可以为记忆效应T细胞和效应T细胞。经刺激后可以分泌IFNγ,和/或可以具有活化T细胞比例上调的特征。
一方面,本申请提供一种肿瘤浸润淋巴细胞(TIL),本申请的TIL可以根据本申请的培养方法培养得到。在一种实施方式中,本申请提供的TIL可以包含一种或一个批次的本申请的培养方法培养得到TIL。在一种实施方式中,本申请提供的TIL可以包含多种或多个批次的本申请的培养方法培养得到并以任意比例组合的TIL。
在一些实施方式中,可以将使用本申请方法扩增的TIL作为药物组合物施用于患者。在一些实施方式中,药物组合物可以是TIL在无菌缓冲液中的悬液。使用本申请的PBMC扩增的TIL可以通过本领域已知的任何合适途径施用。在一些实施方式中,T细胞可以以单次动脉内或静脉内输注施用,输注可以持续约30至60分钟。其他合适的施用途径可以包括腹膜 内、鞘内和***内施用。
在一些实施方式中,可以施用任何合适剂量的TIL。在一些实施方式中,例如当肿瘤是黑色素瘤时,可以施用约2.3×10 9至约13.7×10 10个TIL。在一些实施方式中,可以施用约1×10 9至约12×10 10个TIL。在一些实施方式中,可以施用约1.2×10 10至约4.3×10 10个TIL。在一些实施方式中,可以施用约3×10 10至约12×10 10个TIL。在一些实施方式中,可以施用约4×10 10至约10×10 10个TIL。在一些实施方式中,可以施用约5×10 10至约8×10 10个TIL。在一些实施方式中,可以施用约6×10 10至约8×10 10个TIL。在一些实施方式中,可以施用约7×10 10至约8×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约2.3×10 9至约13.7×10 10。在一些实施方式中,治疗有效剂量可以为约1×10 9至约12×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约1.2×10 10至约4.3×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约3×10 10至约12×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约4×10 10至约10×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约5×10 10至约8×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约6×10 10至约8×10 10个TIL。在一些实施方式中,治疗有效剂量可以为约7×10 10至约8×10 10个TIL。
在一些实施方式中,本申请的组合物中提供的TIL的数量可以为约1×10 6、约2×10 6、约3×10 6、约4×10 6、约5×10 6、约6×10 6、约7×10 6、约8×10 6、约9×10 6、约1×10 7、约2×10 7、约3×10 7、约4×10 7、约5×10 7、约6×10 7、约7×10 7、约8×10 7、约9×10 7、约1×10 8、约2×10 8、约3×10 8、约4×10 8、约5×10 8、约6×10 8、约7×10 8、约8×10 8、约9×10 8、约1×10 9、约2×10 9、约3×10 9、约4×10 9、约5×10 9、约6×10 9、约7×10 9、约8×10 9、约9×10 9、约1×10 10、约2×10 10、约3×10 10、约4×10 10、约5×10 10、约6×10 10、约7×10 10、约8×10 10、约9×10 10、约1×10 11、约2×10 11、约3×10 11、约4×10 11、约5×10 11、约6×10 11、约7×10 11、约8×10 11、约9×10 11、约1×10 12、约2×10 12、约3×10 12、约4×10 12、约5×10 12、约6×10 12、约7×10 12、约8×10 12、约9×10 12、约1×10 13、约2×10 13、约3×10 13、约4×10 13、约5×10 13、约6×10 13、约7×10 13、约8×10 13,或约9×10 13。在一些实施方式中,本申请的组合物中提供的TIL数量的范围可以为约1×10 6至5×10 6、约5×10 6至1×10 7、约1×10 7至5×10 7、约5×10 7至1×10 8、约1×10 8至5×10 8、约5×10 8至1×10 9、约1×10 9至5×10 9、约5×10 9至1×10 10、约1×10 10至5×10 10、约5×10 10至1×10 11、约5×10 11至1×10 12、约1×10 12至5×10 12,或约5×10 12至1×10 13
在一些实施方式中,本申请的组合物中提供的TIL的浓度可以小于组合物的例如约100%、约90%、约80%、约70%、约60%、约50%、约40%、约30%、约20%、约19%、约18%、约17%、约16%、约15%、约14%、约13%、约12%、约11%、约10%、约9%、 约8%、约7%、约6%、约5%、约4%、约3%、约2%、约1%、约0.5%、约0.4%、约0.3%、约0.2%、约0.1%、约0.09%、约0.08%、约0.07%、约0.06%、约0.05%、约0.04%、约0.03%、约0.02%、约0.01%、约0.009%、约0.008%、约0.007%、约0.006%、约0.005%、约0.004%、约0.003%、约0.002%、约0.001%、约0.0009%、约0.0008%、约0.0007%、约0.0006%、约0.0005%、约0.0004%、约0.0003%、约0.0002%,或约0.0001%w/w、w/v或者v/v。
在一些实施方式中,本申请的组合物中提供的TIL的浓度可以大于组合物的约90%、约80%、约70%、约60%、约50%、约40%、约30%、约20%、约19.75%、约19.50%、约19.25%、约19%、约18.75%、约18.50%、约18.25%、约18%、约17.75%、约17.50%、约17.25%、约17%、约16.75%、约16.50%、约16.25%、约16%、约15.75%、约15.50%、约15.25%、约15%、约14.75%、约14.50%、约14.25%、约14%、约13.75%、约13.50%、约13.25%、约13%、约12.75%、约12.50%、约12.25%、约12%、约11.75%、约11.50%、约11.25%、约11%、约10.75%、约10.50%、约10.25%、约10%、约9.75%、约9.50%、约9.25%、约9%、约8.75%、约8.50%、约8.25%、约8%、约7.75%、约7.50%、约7.25%、约7%、约6.75%、约6.50%、约6.25%、约6%、约5.75%、约5.50%、约5.25%、约5%、约4.75%、约4.50%、约4.25%、约4%、约3.75%、约3.50%、约3.25%、约3%、约2.75%、约2.50%、约2.25%、约2%、约1.75%、约1.50%、约125%、约1%、约0.5%、约0.4%、约0.3%、约0.2%、约0.1%、约0.09%、约0.08%、约0.07%、约0.06%、约0.05%、约0.04%、约0.03%、约0.02%、约0.01%、约0.009%、约0.008%、约0.007%、约0.006%、约0.005%、约0.004%、约0.003%、约0.002%、约0.001%、约0.0009%、约0.0008%、约0.0007%、约0.0006%、约0.0005%、约0.0004%、约0.0003%、约或0.0002%,或者约0.0001%w/w、w/v或v/v。
在一些实施方式中,本申请的组合物中提供的TIL的浓度范围可以为组合物的约0.0001%至约50%、约0.001%至约40%、约0.01%至约30%、约0.02%至约29%、约0.03%至约28%、约0.04%至约27%、约0.05%至约26%、约0.06%至约25%、约0.07%至约24%、约0.08%至约23%、约0.09%至约22%、约0.1%至约21%、约0.2%至约20%、约0.3%至约19%、约0.4%至约18%、约0.5%至约17%、约0.6%至约16%、约0.7%至约15%、约0.8%至约14%、约0.9%至约12%,或约1%至约10%w/w、w/v或者v/v。
在一些实施方式中,本申请的组合物中提供的TIL的浓度范围可以为组合物的约0.001%至约10%、约0.01%至约5%、约0.02%至约4.5%、约0.03%至约4%、约0.04%至约3.5%、 约0.05%至约3%、约0.06%至约2.5%、约0.07%至约2%、约0.08%至约1.5%、约0.09%至约1%、或约0.1%至约0.9%w/w、w/v或者v/v。
在一些实施方式中,本申请的组合物中提供的TIL的量可以等于或小于约10g、约9.5g、约9.0g、约8.5g、约8.0g、约7.5g、约7.0g、约6.5g、约6.0g、约5.5g、约5.0g、约4.5g、约4.0g、约3.5g、约3.0g、约2.5g、约2.0g、约1.5g、约1.0g、约0.95g、约0.9g、约0.85g、约0.8g、约0.75g、约0.7g、约0.65g、约0.6g、约0.55g、约0.5g、约0.45g、约0.4g、约0.35g、约0.3g、约0.25g、约0.2g、约0.15g、约0.1g、约0.09g、约0.08g、约0.07g、约0.06g、约0.05g、约0.04g、约0.03g、约0.02g、约0.01g、约0.009g、约0.008g、约0.007g、约0.006g、约0.005g、约0.004g、约0.003g、约0.002g、约0.001g、约0.0009g、约0.0008g、约0.0007g、约0.0006g、约0.0005g、约0.0004g、约0.0003g、约0.0002g,或者约0.0001g。
在一些实施方式中,本申请的组合物中提供的TIL的量可以大于约0.0001g、约0.0002g、约0.0003g、约0.0004g、约0.0005g、约0.0006g、约0.0007g、约0.0008g、约0.0009g、约0.001g、约0.0015g、约0.002g、约0.0025g、约0.003g、约0.0035g、约0.004g、约0.0045g、约0.005g、约0.0055g、约0.006g、约0.0065g、约0.007g、约0.0075g、约0.008g、约0.0085g、约0.009g、约0.0095g、约0.01g、约0.015g、约0.02g、约0.025g、约0.03g、约0.035g、约0.04g、约0.045g、约0.05g、约0.055g、约0.06g、约0.065g、约0.07g、约0.075g、约0.08g、约0.085g、约0.09g、约0.095g、约0.1g、约0.15g、约0.2g、约0.25g、约0.3g、约0.35g、约0.4g、约0.45g、约0.5g、约0.55g、约0.6g、约0.65g、约0.7g、约0.75g、约0.8g、约0.85g、约0.9g、约0.95g、约1g、约1.5g、约2g、约2.5g、约3g、约3.5g、约4g、约4.5g、约5g、约5.5g、约6g、约6.5g、约7g、约7.5g、约8g、约8.5g、约9g、约9.5g,或者约10g。
在一些实施方式中,TIL可以单剂量施用。此种施用可以通过注射,例如可以静脉内注射。在一些实施方式中,TIL可以多剂量施用。剂量可以是每年一次、两次、三次、四次、五次、六次或超过六次。剂量可以是每月一次、每两周一次、每周一次或每2天一次。在一些实施方式中,TIL的施用可以连续施用。
一方面,本申请提供一种药物组合物。在一些实施方式中,其可以包含本申请的TIL和/或本申请的组合物,与药学上可接受的载体。
一方面,本申请提供一种试剂盒,本申请的试剂盒可以包含本申请培养肿瘤浸润淋巴细胞(TIL)方法的T细胞激活剂、T细胞生长因子和/或饲养细胞与记载本申请培养肿瘤浸润淋巴细胞(TIL)方法的步骤的说明书。一方面,本申请提供一种试剂盒,本申请试剂盒可以包含本申请的TIL和/或本申请的药物组合物。
方法和制药用途
一方面,本申请提供一种影响肿瘤细胞生长的方法,可以包括向受试者施用本申请的TIL和/或本申请的药物组合物。在一些实施方式中,影响肿瘤生长可以包含肿瘤的体积减少到施用前的例如约99%、约95%、约90%、约80%、约70%、约60%、约50%、约40%、约30%、约20%、约19%、约18%、约17%、约16%、约15%、约14%、约13%、约12%、约11%、约10%、约9%、约8%、约7%、约6%、约5%、约4%、约3%、约2%、约1%、约0.5%、约0.4%、约0.3%、约0.2%或约0.1%。
一方面,本申请提供本申请的TIL和/或本申请的药物组合物在制备药物中的应用,本申请的药物可以用于预防和/或***。在一些实施方式中,本申请的肿瘤选自实体瘤。在一些实施方式中,本申请的肿瘤可以选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
一方面,本申请提供一种预防和/或***的方法,可以包括向受试者施用本申请的TIL和/或本申请的药物组合物。在一些实施方式中,本申请的肿瘤选自实体瘤。在一些实施方式中,本申请的肿瘤可以选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
一方面,本申请提供一种本申请的TIL和/或本申请的药物组合物,其可以用于预防和/或***。在一些实施方式中,本申请的肿瘤选自实体瘤。在一些实施方式中,本申请的肿瘤可以选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
不欲被任何理论所限,下文中的实施例仅仅是为了阐释本申请的融合蛋白、制备方法和用途等,而不用于限制本申请发明的范围。
实施例
实施例1 肿瘤浸润淋巴细胞(TIL)细胞的培养方法
饲养细胞接收及制备
1.1.1单采血接收
记录单采血信息,批号及体积,并复温至室温。
1.1.2 PBMC(外周血单个核细胞)手动分离及冻存
使用75%酒精消毒血袋,转移至生物安全柜内。使用无菌剪刀剪开血袋后,将单采血转移至50mL离心管内,使用20mL注射器注入20mL PBS或生理盐水清洗血袋,将洗涤液一 并转入50mL离心管内。每个50mL离心管内液体体积可以不超过30mL。将单采血3000g离心10分钟。离心过程中准备6-8支50mL离心管,加入已复温的淋巴细胞分离液(天津灏洋Ficoll),20mL/支。离心结束后,弃掉上层血浆,使用PBS或生理盐水稀释细胞沉淀,将稀释后的血细胞混合液缓慢滴加上淋巴细胞分离液上层,可以不破坏界面,每管约加25mL样品,可以不超过28mL。
离心使用水平转子,500-600g离心15-30分钟,温度18-22℃,离心结束后得到的白膜层将处于生理盐水及淋巴细胞分离液Ficoll的分界面处。吸弃上层血浆及生理盐水,用移液管吸取中间白膜层至另一干净的50mL离心管内。使用PBS或生理盐水稀释收集到的白膜层,600g离心10分钟,室温。离心结束后弃上清,PBS或生理盐水清洗细胞一次,500g离心5分钟,室温。
如红细胞较多,离心结束后可以进行裂红,按照细胞沉淀体积与红细胞裂解液1:2至1:3加入红细胞裂解液,混匀,室温裂解10分钟中,中间轻柔混匀离心管2-3次,保证裂解效果,裂解完成后加入PBS或生理盐水清洗细胞。裂红后清洗细胞两次,400g离心6分钟,最后一次离心前取样计数。
弃上清,基础培养基重悬细胞,调整细胞密度约2-3×10 7个细胞/mL,液面高度可以不超过1厘米,每T225培养瓶中体积可以低于200mL;平铺状态下,X射线辐照50-200Gy。离心弃上清,根据计数结果冻存细胞,约1-2×10 8个细胞/mL,1-2mL/支;将细胞放入程序降温盒内转移至﹣80℃冰箱内冻存。
1.1.3 PBMC自动分离及冻存
将血袋的管路与cpro分离套件(Cytiva)输入端无菌接管。若血量大于120mL,进行预浓缩步骤,可以将血液体积浓缩至120mL以内。可以使用neatcell程序进行PBMC分离及洗涤,洗涤液为生理盐水,中间体积20mL;重悬液为基础培养基,添加80mL/批。分离后每供者PBMC为一袋100mL,在平铺状态下,液面高度可以不超过1厘米,X射线辐照50-200Gy。辐照后取样计数,使用culture wash程序收集细胞并洗涤三次,洗涤液为生理盐水;设置中间体积及终体积,使得每1×10 9个细胞不少于2mL;加入等量至2倍冻存液混匀。使用1倍冻存液调整细胞密度约为1×10 7个细胞/mL至2×10 8个细胞/mL,分装20mL/袋,程序降温仪内冻存,液氮保存。
1.2肿瘤组织接收及处理
1.2.1组织接收
接收供者的肿瘤组织及血样,核对样品信息并记录,打印相应样品标签。
1.2.2组织处理及培养
使用75%酒精消毒样品管及采血管,转移至生物安全柜内。根据上述PBMC手动分离及冻存操作程序分离血样中PBMC细胞并进行冻存。取一种具有透气表面的培养瓶或培养袋,例如培养袋(Origen),加入300mL已复温的完全培养基,完全培养基可以任意地选用X-vivo15培养基或其它商用的T细胞培养基,例如Stem Cell,Lonza,Thermo,美天旎等品牌的T细胞培养基,并可以添加必须氨基酸及抗生素,并添加浓度为300-9000IU/mL(例如可以是1000-9000IU/mL,例如可以是6000IU/mL)的IL-2。取数个10厘米培养皿,加入适量培养基,使用无菌眼科镊从样品管中取出肿瘤组织于10厘米培养皿中,培养基量以刚没过肿瘤组织为准,观察组织形态并记录。洗涤组织并更换培养皿。使用眼科剪及眼科镊将进行初步剪切,去除脂肪组织及坏死组织,每块组织块继续剪碎至约27立方毫米大小。取非悬浮肿瘤组织块,使用20mL注射器去除内部活塞后,与培养袋连接,使用移液管将约1g组织块通过注射器转入培养袋内。将培养袋放入二氧化碳培养箱内进行培养。清理剪刀及镊子,并用75%酒精进行初步消毒后,超声清洗后进行灭菌,得到第一TIL群。
1.3第一阶段体外扩增及收获(preREP阶段)
1.3.1第一阶段体外扩增
根据细胞生长状态,每3-7天补液或半量换液,保证细胞营养。使用完全培养基,完全培养基可以任意地选用X-vivo 15培养基或其它商用的T细胞培养基,例如Stem Cell,Lonza,Thermo,美天旎等品牌的T细胞培养基,并可以添加必须氨基酸及抗生素,并添加浓度为300-9000IU/mL(例如可以是1000-9000IU/mL,例如可以是6000IU/mL)的IL-2,例如6000IU/mL的IL-2。第一阶段体外扩增的3-14天,例如可以第3、4、5、6、7、8、9、10、11、12、13或14天时取样计数,若细胞数目处于5×10 5至5×10 8之间时进入下述第一阶段体外扩增的收获步骤。
1.3.2第一阶段体外扩增的收获
收集第一阶段体外扩增结束细胞,离心,弃去培养基,使用PBS或生理盐水洗涤细胞一次,获得经第一阶段体外扩增的TIL(第二TIL群),并取样计数留取约5×10 5至2×10 8个细胞进入下述第一阶段体外扩增步骤;取约5×10 5个细胞可以进行质量控制检测;其余细胞加入冻存液冻存。
1.4第二阶段体外扩增(REP阶段)
1.4.1第二阶段体外扩增的TIL活化
取5×10 5至2×10 8的第一阶段体外扩增的细胞量,使用完全培养基,完全培养基可以任意 地选用X-vivo 15培养基或其它商用的T细胞培养基,例如Stem Cell,Lonza,Thermo,美天旎等品牌的T细胞培养基,并可以添加必须氨基酸及抗生素,调整细胞密度为5×10 5至2×10 6个细胞/mL,于悬浮24孔培养板内,1mL/孔,添加浓度为300-9000IU/mL(例如可以是1000-9000IU/mL,例如可以是6000IU/mL)的IL-2。各TIL加入IL-2的同时,可以添加CD3激动剂和/或CD28激动剂,例如,约30ng/mL的OKT3、约30ng/mL的CD28抗体、以约1:2-2:1的磁珠与TIL的比例加入磁珠(直径约1至10μm Dynabeads,Thermo Fisher)和/或以约1:100-1:2000的transACT(直径约100至500nm,Miltenyi)与TIL的比例加入transACT。
1.4.2第二阶段体外扩增的扩大培养
在第二阶段体外扩增加入IL-2与不同形式的T细胞激活剂后的若干时间T n以后(T n可以取0小时到14天,例如24小时或48小时),复苏1-5名供者混合的饲养细胞;将活化的TIL细胞,饲养细胞转入G-Rex100培养瓶或者透气袋内,补充完全培养基,每1-3天取样计数,并根据细胞状态补液或半量换液直至细胞总数大于1×10 9或第二阶段体外扩增培养达13天,终止第二阶段体外扩增的培养。
1.4.3肿瘤浸润淋巴细胞的收获
取第二阶段体外扩增的细胞,离心后弃去培养基上清,并使用PBS或生理盐水或复方电解质溶液清洗三次,获得经第二阶段体外扩增的TIL(第三TIL群),第三次清洗时取样计数,根据计数结果,最后一次离心后弃上清,取3×10 6细胞送质量控制检测;其余全部细胞加入冻存液,调整细胞密度1-3×10 8个细胞/mL冻存。
1.5第三阶段体外扩增
收集第二阶段体外扩增结束的TIL,或取冻存的第三TIL群并复苏,复苏后的TIL可以任选地缓解若干时间,例如约1小时、约2小时、约6小时、约12小时、约1天、约2天或约4天。使用完全培养基调整细胞密度,完全培养基可以任意地选用X-vivo 15培养基或其它商用的T细胞培养基,例如Stem Cell,Lonza,Thermo,美天旎等品牌的T细胞培养基,并可以添加必须氨基酸及抗生素;调整细胞密度为5×10 5至2×10 6个细胞/mL,于悬浮24孔培养板内,1mL/孔。各个试验组进行不同方式的刺激:
对照组:加入PBS或生理盐水;
PD-1抗体组:加入至少为约0.1μg/mL(例如约1μg/mL或约10μg/mL)的PD-1抗体(例如pembrolizumab,MSD,或6H6,重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);
纳米基质组:以约1:100-1:2000的transACT与TIL的比例加入transACT(直径约100至 500nm,Miltenyi);
纳米基质+PD-1抗体组:以约1:100-1:2000的transACT与TIL的比例加入transACT,以及加入至少为约0.1μg/mL(例如约1μg/mL或约10μg/mL)的PD-1抗体(例如pembrolizumab,MSD,或6H6,重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);
纳米基质+PD-1抗体组A:以约1:100-1:2000的transACT与TIL的比例加入transACT,以及加入至少为约0.1μg/mL(例如约1μg/mL或约10μg/mL)的PD-1抗体A(pembrolizumab,MSD);
纳米基质+PD-1抗体组B:以约1:100-1:2000的transACT与TIL的比例加入transACT,以及加入至少为约0.1μg/mL(例如约1μg/mL或约10μg/mL)的PD-1抗体B(重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);
以上各个组,在第三阶段体外扩增的约12小时至约24小时时收获第四TIL群并取样计数,终止第三阶段体外扩增的培养。
1.6肿瘤浸润淋巴细胞的应用
可以将收获的治疗性肿瘤浸润淋巴细胞给予受试者静脉滴注。
实施例2 TIL胞内因子表达检测
对于实施例1中各个试验组不同方式的第三阶段体外扩增培养获得的第四TIL群进行胞内因子表达检测。
试验准备
配制胞内因子表达检测所需培养基:取T细胞培养基,按照体积比1:500添加CD107a抗体(BD)。
检测步骤
取各个试验组的TIL离心后,使用600μL上述胞内因子表达检测所需培养基重悬为1×10 6个细胞/mL,加入96孔板内,100μL/孔,置于37℃培养箱孵育过夜。
孵育结束后,200μL/孔PBS洗涤一次,600g离心3分钟,弃上清。配制抗体混合工作液进行细胞表面染色CD3/CD4/CD8(BD),抗体浓度为1:100,viability(1:10000),50μL/组染色,2-8℃避光孵育30分钟。染色结束后清洗细胞,使用PBS重悬,进行流式上机检测。
图1A-1B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的胞内因子表达能力。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组)、仅添加PD-1抗体(PD-1抗体组)和仅添加T细胞激活剂(纳米基质 组),在第三阶段体外扩增培养同时添加PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组)具有更高的胞内因子表达能力,例如,更高的CD107a表达能力。在第三阶段体外扩增中,PD-1抗体和T细胞激活剂在胞内因子表达能力的提高方面具有显著的协同作用。
图2A-2B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的胞内因子表达能力。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组),在第三阶段体外扩增培养同时添加不同种类的PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组A或纳米基质+PD-1抗体组B)具有相近的胞内因子表达能力的提高,例如,更高的CD107a表达能力。在第三阶段体外扩增中,不同的PD-1抗体与T细胞激活剂组合对于胞内因子表达能力都具有显著的增强作用。
实施例3 TIL细胞因子分泌检测
对于实施例1中各个试验组不同方式的第三阶段体外扩增培养获得的第四TIL群进行细胞因子分泌检测。
标准品准备
人Th1/Th2/Th17细胞因子标准品冻干粉(BD)使用2mL Assay Diluent稀释液(BD)复溶(标准品原液各细胞因子浓度均为5000pg/mL)并按顺序:1:2,1:4,1:8,1:16,1:32,1:64,1:128,1:256,1:512,1:1024梯度稀释,随后与Capture Beads(BD)混合及PE Detection Reagent检测试剂(BD)混合,转移至15mL锥底离心管,标记为“标准品管”。取1管仅含有Assay Diluent稀释液作为阴性对照。
检测步骤
按照2μL/Beads/孔与每种Capture Beads(BD)混合,然后按照10μL/孔与PE Detection Reagent检测试剂(BD)混合,配制为混合物(mix),按照22μL/孔加入V底96孔板内,随后按照10μL/孔加入各实验组培养上清混合,室温下避光孵育3小时。
孵育结束,每孔加入200μL Wash Buffer(BD),500g离心3分钟。离心结束,每孔加入100μL Wash Buffer(BD)重悬,进行流式分析。
图3A-3C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的IL-2分泌检测结果。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组)以及仅添加T细胞激活剂(纳米基质组),在第三阶段体外扩增培养同时添加PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组)具有更高的细胞因子分泌能力,例如,更高的IL-2分泌能力。
图4A-4C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL 群的TNF分泌检测结果。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组)以及仅添加T细胞激活剂(纳米基质组),在第三阶段体外扩增培养同时添加PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组)具有更高的细胞因子分泌能力,例如,更高的TNF分泌能力。
图5A-5C显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL群的IFNγ分泌检测结果。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组)以及仅添加T细胞激活剂(纳米基质组),在第三阶段体外扩增培养同时添加PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组)具有更高的细胞因子分泌能力,例如,更高的IFNγ分泌能力。
实施例4 TIL细胞杀伤能力检测
对于实施例1中各个试验组不同方式的第三阶段体外扩增培养获得的第四TIL群进行细胞杀伤能力检测。
细胞准备
准备用于检测的各个试验组获得的TIL和用于共培养的靶细胞(例如Hela肿瘤细胞)。
检测步骤
用CFSE(5(6)-Carboxyfluorescein diacetate N-succinimidyl ester,Sigma,21888-25MG-F)标记肿瘤细胞:用PBS清洗肿瘤细胞,重悬肿瘤细胞于500μL的PBS中;将CFSE加入500μL的PBS中,与500μL的肿瘤细胞PBS重悬液混合,至CFSE的终浓度为0.5μmol/L。37℃孵育6分钟后,加含10%FBS的培养基清洗,600g离心5分钟,用X-vivo 15培养基或其它商用的T细胞培养基,例如Stem Cell,Lonza,Thermo,美天旎等品牌的T细胞培养基重悬肿瘤细胞浓度为5×10 5个细胞/mL。对各个试验组的TIL细胞600g离心5分钟,按照效靶比(TIL细胞与肿瘤细胞的比例)3:1重悬TIL细胞(即重悬TIL细胞浓度为1.5×10 6个细胞/mL)。于U底96孔板(Corning)中加入肿瘤细胞和TIL细胞各100μL,每组设置三个复孔。同时设置一组只包含肿瘤细胞的对照组并按照实验不同分组加入不同试剂。将孔板200g离心1分钟,置于37℃孵育4小时至过夜。
孵育完成后,600g离心3分钟,弃上清,每孔加入20μL胰酶,37℃培养箱内孵育3-5分钟消化肿瘤细胞,消化完成后加入180μL含10%FBS的培养基终止消化。将Dapi(碧云天,C0060)用1:100稀释,然后每孔加入20μL稀释后的Dapi。进行流式上机检测。
杀伤率%=Dapi +CFSE +细胞数/总CFSE +×100%。
图6A-6B显示的是,对于不同供者,不同方式的第三阶段体外扩增培养获得的第四TIL 群的细胞杀伤能力检测结果。结果显示,相比于第三阶段体外扩增培养时不添加PD-1抗体和T细胞激活剂(对照组),在第三阶段体外扩增培养同时添加不同种类的PD-1抗体和T细胞激活剂(纳米基质+PD-1抗体组A或纳米基质+PD-1抗体组B)具有相近的细胞杀伤能力的提高。在第三阶段体外扩增中,不同的PD-1抗体与T细胞激活剂组合对于细胞杀伤能力都具有显著的增强作用。
实施例5 TIL增殖能力检测
对于实施例1中第二阶段体外扩增(REP阶段)的不同培养方法进行探究试验。各个试验组在第二阶段体外扩增(REP阶段)进行不同方式的刺激:
CD3抗体组:加入CD3抗体,例如OKT3约30ng/mL;
CD3抗体+PD-1抗体组M:加入CD3抗体,例如OKT3约30ng/mL,以及约1μg/mL的PD-1抗体(例如pembrolizumab,MSD,或6H6,重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);
CD3抗体+PD-1抗体组N:加入CD3抗体,例如OKT3约30ng/mL,以及约10μg/mL的PD-1抗体(例如pembrolizumab,MSD,或6H6,重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);
以上各个组,在第二阶段体外扩增开始后的第7天、第10天和第17天时收获并取样计数。
图7A-7C显示的是,对于不同供者,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入不同浓度的PD-1抗体,相比仅加入CD3抗体的TIL细胞增殖情况。图7A-7C中纵坐标的数值表示,各个试验组第二阶段体外扩增获得的各个时间点的TIL细胞相比于第二阶段体外扩增开始前的第二TIL群,TIL细胞数量扩增至的扩增倍数。结果显示,在第二阶段体外扩增(REP阶段)额外加入不同浓度的PD-1抗体,获得的TIL增殖能力没有明显优势。
实施例6 第二阶段体外扩增(REP阶段)添加PD1抗体的TIL培养效果检测
对于实施例1中第二阶段体外扩增(REP阶段)的不同培养方法进行探究试验。各个试验组在第二阶段体外扩增(REP阶段)进行不同方式的刺激:
CD3抗体组:加入CD3抗体,例如OKT3约30ng/mL;
CD3抗体+PD1(PD-1)抗体组:加入CD3抗体,例如OKT3约30ng/mL,以及约10μg/mL的PD-1抗体(例如pembrolizumab,MSD,或6H6,重链全长如SEQ ID NO:9和13中任一项所示,且轻链全长如SEQ ID NO:10和14中任一项所示);以上各个组,在第二阶 段体外扩增开始后的第6天、第10天和第13天时收获并取样计数。
图8A-8B显示的是,对于不同供者,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL细胞增殖情况。结果显示,在第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL增殖能力没有明显优势。
培养7天(7D)及13天(13D)进行流式检测细胞分型,并取样添加transACT(Miltenyi)进行刺激,流式检测胞内因子分泌比例(2E5个细胞/孔)及细胞因子分泌量(1E5个细胞/孔)。
图9A-9B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞活率和细胞分型情况。结果显示,第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL的细胞活率和CD3 +比例没有明显优势。
图10A-10B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中活化相关(41BB +)细胞比例情况。
图11A-11C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中活化相关(CD25 +和/或CD27 +)细胞比例情况。
结果显示,第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL的活化相关细胞比例没有明显优势。
图12A-12B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中耗竭相关(TIM3 +)细胞比例情况。结果显示,第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL的耗竭相关细胞比例没有明显优势。
图13A-13B显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中分泌TNF-α细胞比例情况。
图14A-14D显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL中分泌IFN-γ细胞比例情况。
结果显示,第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL的细胞因子分泌细胞比例没有明显优势。
图15A-15C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞因子(IL-2和/或IL-4)分泌量情况。
图16A-16C显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD-1抗体,相比仅加入CD3抗体的TIL的细胞因子(IL-17和/或IL-6)分泌量情况。
图17A-17D显示的是,在第二阶段体外扩增(REP阶段)加入CD3抗体时额外加入PD- 1抗体,相比仅加入CD3抗体的TIL的细胞因子(TNF-α和/或IFN-γ)分泌量情况。
结果显示,第二阶段体外扩增(REP阶段)额外加入PD-1抗体,获得的TIL的细胞因子分泌能力没有明显优势。
实施例7 第三阶段体外扩增添加PD1抗体的TIL培养效果检测
对于实施例1中各个试验组不同方式的第三阶段体外扩增培养获得的第四TIL群进行检测。第二阶段体外扩增(REP阶段)培养结束后,采用transACT(Miltenyi)或额外加入20μg/mL的PD-1抗体进行刺激,检测细胞因子分泌能力。
图18A-18D显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-4和/或TNF-α)分泌量情况。图18E-18F显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-2和/或IL-6)分泌量情况。
结果显示,在REP阶段培养结束后额外加入PD-1抗体,获得的TIL的细胞因子分泌能力明显增强。
实施例8 第二阶段体外扩增(REP阶段)添加PD1抗体以及第三阶段体外扩增添加PD1抗体的TIL培养效果检测
对于实施例1中第二阶段体外扩增(REP阶段)加入PD1(PD-1)抗体培养,且在第二阶段体外扩增(REP阶段)培养结束后,采用纳米基质(transACT,Miltenyi)或额外加入PD-1抗体(2μg/mL或20μg/mL)进行刺激,检测细胞因子分泌能力。
图19A-19D显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-6和/或TNF-α)分泌量情况。
图20A-20C显示的是,在REP阶段培养结束后,额外加入PD-1抗体进行刺激,相比仅加入transACT进行刺激的TIL的细胞因子(IL-2、IL-4和/或IFN-γ)分泌量情况。
结果显示,即使在第二阶段体外扩增(REP阶段)加入PD1(PD-1)抗体对TIL进行培养,在REP阶段培养结束后额外加入PD-1抗体,获得的TIL的细胞因子分泌能力也可以明显增强。
前述详细说明是以解释和举例的方式提供的,并非要限制所附权利要求的范围。目前本文所列举的实施方式的多种变化对本领域普通技术人员来说是显而易见的,且保留在所附的权利要求和其等同方案的范围内。

Claims (139)

  1. 一种培养肿瘤浸润淋巴细胞(TIL)的方法,其包含:使源自肿瘤组织且未经体外扩增的TIL经过至少一个阶段的体外扩增,其中,在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
  2. 根据权利要求1所述的方法,其中,使所述源自肿瘤组织且未经体外扩增的TIL经过第一阶段体外扩增、第二阶段体外扩增和第三阶段体外扩增,且在所述第三阶段体外扩增中,使经所述第二阶段体外扩增的TIL与所述一种或多种T细胞激活剂以及所述免疫检查点抑制剂接触。
  3. 根据权利要求1-2中任一项所述的方法,其中,在单个阶段的所述体外扩增中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触。
  4. 根据权利要求2-3中任一项所述的方法,所述第三阶段体外扩增进行至多约24小时。
  5. 根据权利要求2-4中任一项所述的方法,所述第三阶段体外扩增进行约12小时至约24小时。
  6. 根据权利要求1-5中任一项所述的方法,与在体外扩增阶段未曾与所述T细胞激活剂和/或所述免疫检查点抑制剂接触的相应TIL相比,在至少一个体外扩增阶段中与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触过的TIL显示出改善的扩增效果。
  7. 根据权利要求6所述的方法,所述改善的扩增效果包含选自以下组的一种或多种:增加的TIL细胞数量,改善的T细胞亚群比例,提高的细胞因子分泌能力,和提高的肿瘤细胞杀伤能力。
  8. 根据权利要求7所述的方法,所述改善的T细胞亚群比例包含选自以下组的一种或多种:增加的中心记忆T细胞比例,降低的调节性T细胞的比例,增加的活化T细胞比例,增加的肿瘤特异性T细胞比例,和增加的干细胞样T细胞比例。
  9. 根据权利要求1-8中任一项所述的方法,所述方法还包含:在至少一个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂接触。
  10. 根据权利要求1-9中任一项所述的方法,所述一种或多种T细胞激活剂包含选自以下组的一种或多种:CD80、CD86、B7-H3、4-1BBL、CD27、CD30、CD134、B7h、CD40、LIGHT、以及它们的功能活性片段。
  11. 根据权利要求1-10中任一项所述的方法,所述一种或多种T细胞激活剂包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。
  12. 根据权利要求1-11中任一项所述的方法,所述一种或多种T细胞激活剂包含CD3激动剂 和/或CD28激动剂。
  13. 根据权利要求1-12中任一项所述的方法,所述一种或多种T细胞激活剂包含CD3激动剂。
  14. 根据权利要求1-13中任一项所述的方法,所述一种或多种T细胞激活剂包含抗CD3的抗体和/或其抗原结合片段。
  15. 根据权利要求1-14中任一项所述的方法,所述一种或多种T细胞激活剂包含CD28激动剂。
  16. 根据权利要求1-15中任一项所述的方法,所述一种或多种T细胞激活剂包含抗CD28的抗体和/或其抗原结合片段。
  17. 根据权利要求1-16中任一项所述的方法,所述使TIL与所述一种或多种T细胞激活剂接触包含选自以下组的一种或多种方式:(1)将所述一种或多种T细胞激活剂添加至所述TIL的细胞培养基中;(2)将表达所述一种或多种T细胞激活剂的工程化细胞添加至所述TIL的细胞培养基中;(3)将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
  18. 根据权利要求17所述的方法,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为至少约30ng/mL。
  19. 根据权利要求17-18中任一项所述的方法,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为约30ng/mL-约300ng/mL。
  20. 根据权利要求17-19中任一项所述的方法,所述固相介质的直径为约500纳米至约10微米。
  21. 根据权利要求17-19中任一项所述的方法,所述固相介质的直径为约1纳米至约500纳米。
  22. 根据权利要求17-21中任一项所述的方法,所述固相介质的直径通过透射电子显微镜测量。
  23. 根据权利要求17-22中任一项所述的方法,所述固相介质包含聚合物。
  24. 根据权利要求17-23中任一项所述的方法,每mg所述固相介质中包含的每一种所述T细胞激活剂的量各自独立地至少为约25μg。
  25. 根据权利要求17-24中任一项所述的方法,以约2:1-约1:2的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
  26. 根据权利要求17-25中任一项所述的方法,以约1:100-约1:2000的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养 基中。
  27. 根据权利要求1-26中任一项所述的方法,所述免疫检查点抑制剂包含抑制PD-1与PD-L1和/或PD-L2相互作用的物质。
  28. 根据权利要求1-27中任一项所述的方法,所述免疫检查点抑制剂包含PD-1抑制剂。
  29. 根据权利要求1-28中任一项所述的方法,所述免疫检查点抑制剂包含PD-1的抗体和/或其抗原结合片段。
  30. 根据权利要求1-29中任一项所述的方法,所述免疫检查点抑制剂包含具有以约100pM或更低的K D值结合PD-1的能力的免疫检查点抑制剂。
  31. 根据权利要求1-30中任一项所述的方法,所述免疫检查点抑制剂包含具有以约100pM或更低的EC 50值结合PD-1的能力的免疫检查点抑制剂。
  32. 根据权利要求1-31中任一项所述的方法,所述免疫检查点抑制剂包含具有以约1nM或更低的IC 50值抑制PD-1与PD-L1和/或PD-L2结合的能力的免疫检查点抑制剂。
  33. 根据权利要求1-32中任一项所述的方法,所述免疫检查点抑制剂包含HCDR3,且所述HCDR3包含SEQ ID NO:3和17中任一项所示的氨基酸序列。
  34. 根据权利要求1-33中任一项所述的方法,所述免疫检查点抑制剂包含HCDR2,且所述HCDR2包含SEQ ID NO:2和16中任一项所示的氨基酸序列。
  35. 根据权利要求1-34中任一项所述的方法,所述免疫检查点抑制剂包含HCDR1,且所述HCDR1包含SEQ ID NO:1和15中任一项所示的氨基酸序列。
  36. 根据权利要求1-35中任一项所述的方法,所述免疫检查点抑制剂包含LCDR3,且所述LCDR3包含SEQ ID NO:6和20中任一项所示的氨基酸序列。
  37. 根据权利要求1-36中任一项所述的方法,所述免疫检查点抑制剂包含LCDR2,且所述LCDR2包含SEQ ID NO:5和19中任一项所示的氨基酸序列。
  38. 根据权利要求1-37中任一项所述的方法,所述免疫检查点抑制剂包含LCDR1,且所述LCDR1包含SEQ ID NO:4和18中任一项所示的氨基酸序列。
  39. 根据权利要求1-38中任一项所述的方法,所述免疫检查点抑制剂包含VH,且所述VH包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列。
  40. 根据权利要求1-39中任一项所述的方法,所述免疫检查点抑制剂包含VL,且所述VL包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列。
  41. 根据权利要求1-40中任一项所述的方法,所述免疫检查点抑制剂选自以下组:(1)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:1和 15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述轻链包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3;(2)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述轻链包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL。
  42. 根据权利要求29-41中任一项所述的方法,其中所述抗体选自:嵌合抗体,人源化抗体和全人源抗体。
  43. 根据权利要求29-42中任一项所述的方法,其中所述抗原结合片段选自:Fab,Fab′,Fv片段,F(ab') 2,F(ab) 2,scFv,di-scFv,VHH和dAb。
  44. 根据权利要求1-43中任一项所述的方法,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL。
  45. 根据权利要求1-44中任一项所述的方法,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地为约0.1μg/mL-约20μg/mL。
  46. 根据权利要求1-45中任一项所述的方法,其还包含:在至少一个阶段的所述体外扩增中,使所述TIL与一种或多种T细胞生长因子接触。
  47. 根据权利要求46所述的方法,其中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂且与所述一种或多种T细胞生长因子接触。
  48. 根据权利要求46-47中任一项所述的方法,其中,在单个阶段的所述体外扩增中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种T细胞生长因子接触。
  49. 根据权利要求46-48中任一项所述的方法,所述一种或多种T细胞生长因子选自以下组的一种或多种:IL-2、IL-7、IL-12、IL-15、IL-21、γ干扰素、以及它们的功能活性片段。
  50. 根据权利要求46-49中任一项所述的方法,所述一种或多种T细胞生长因子包含IL-2和/或其功能活性片段。
  51. 根据权利要求46-50中任一项所述的方法,所述TIL与所述一种或多种T细胞生长因子接触包含将所述T细胞生长因子添加至所述TIL的细胞培养基中。
  52. 根据权利要求46-51中任一项所述的方法,每一种所述T细胞生长因子在所述TIL的细胞培养基中的初始浓度各自独立地为至少约300IU/mL。
  53. 根据权利要求1-52中任一项所述的方法,其还包含:在至少一个阶段的所述体外扩增中,使所述TIL与饲养细胞共培养。
  54. 根据权利要求53所述的方法,其中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触且与所述饲养细胞共培养。
  55. 根据权利要求53-54中任一项所述的方法,其中,在单个阶段的所述体外扩增中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触一定时间之后,再与所述饲养细胞共培养。
  56. 根据权利要求55所述的方法,所述一定时间为至少约2小时。
  57. 根据权利要求55-56中任一项所述的方法,所述一定时间为约6小时至约72小时。
  58. 根据权利要求55-57中任一项所述的方法,所述一定时间为约12小时至约48小时。
  59. 根据权利要求55-57中任一项所述的方法,所述一定时间为约6小时、约12小时、约24小时、约48小时、或约72小时。
  60. 根据权利要求53-59中任一项所述的方法,所述饲养细胞包含抗原呈递细胞。
  61. 根据权利要求53-60中任一项所述的方法,所述饲养细胞包含选自以下组的一种或多种:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。
  62. 根据权利要求53-61中任一项所述的方法,所述饲养细胞为外周单个核细胞。
  63. 根据权利要求53-62中任一项所述的方法,所述饲养细胞为经过辐照的饲养细胞。
  64. 根据权利要求53-63中任一项所述的方法,所述TIL与所述饲养细胞共培养包含使所述饲养细胞的表面与所述TIL的表面相接触。
  65. 根据权利要求53-64中任一项所述的方法,所述TIL与所述饲养细胞共培养包含将所述饲养细胞添加至所述TIL的细胞培养基中。
  66. 根据权利要求53-65中任一项所述的方法,以约40:1-约400:1的所述饲养细胞与所述TIL的比例,将所述饲养细胞添加至所述TIL的细胞培养基中。
  67. 根据权利要求1-66中任一项所述的方法,所述源自肿瘤组织且未经体外扩增的TIL为源自所述肿瘤组织的碎片的TIL。
  68. 根据权利要求67所述的方法,所述碎片的体积为约1立方毫米至约27立方毫米。
  69. 一种培养肿瘤浸润淋巴细胞(TIL)的方法,其包含:
    (A)使源自肿瘤组织且未经体外扩增的第一TIL群与一种或多种T细胞生长因子接触;其中,经所述步骤(A)得到第二TIL群;
    (B)使所述第二TIL群与所述一种或多种T细胞生长因子接触和/或一种或多种T细胞激活剂接触;其中,经所述步骤(B)得到第三TIL群;
    (C)使所述第三TIL群与所述一种或多种T细胞激活剂以及一种或多种免疫检查点抑制剂接触。
  70. 根据权利要求69所述的方法,其中,在步骤(C)中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触。
  71. 根据权利要求69-70中任一项所述的方法,所述步骤(C)进行至多约24小时。
  72. 根据权利要求69-71中任一项所述的方法,所述步骤(C)进行约12小时至约24小时。
  73. 根据权利要求69-72中任一项所述的方法,与在步骤(C)中未曾与所述T细胞激活剂和/或所述免疫检查点抑制剂接触的相应TIL相比,在步骤(C)中与所述一种或多种T细胞激活剂以及所述一种或多种免疫检查点抑制剂接触过的TIL显示出改善的扩增效果。
  74. 根据权利要求73所述的方法,所述改善的扩增效果包含选自以下组的一种或多种:增加的TIL细胞数量,改善的T细胞亚群比例,提高的细胞因子分泌能力,和提高的肿瘤细胞杀伤能力。
  75. 根据权利要求74所述的方法,所述改善的T细胞亚群比例包含选自以下组的一种或多种:增加的中心记忆T细胞比例,降低的调节性T细胞的比例,增加的活化T细胞比例,增加的肿瘤特异性T细胞比例,和增加的干细胞样T细胞比例。
  76. 根据权利要求69-75中任一项所述的方法,所述一种或多种T细胞激活剂包含选自以下组的一种或多种:CD80、CD86、B7-H3、4-1BBL、CD27、CD30、CD134、B7h、CD40、LIGHT、以及它们的功能活性片段。
  77. 根据权利要求69-76中任一项所述的方法,所述一种或多种T细胞激活剂包含选自以下组的一种或多种靶点的激动剂:CD3、CD28、HVEM、CD40L、OX40和4-1BB。
  78. 根据权利要求69-77中任一项所述的方法,所述一种或多种T细胞激活剂包含CD3激动剂和/或CD28激动剂。
  79. 根据权利要求69-78中任一项所述的方法,所述一种或多种T细胞激活剂包含CD3激动剂。
  80. 根据权利要求69-79中任一项所述的方法,所述一种或多种T细胞激活剂包含抗CD3的抗体和/或其抗原结合片段。
  81. 根据权利要求69-80中任一项所述的方法,所述一种或多种T细胞激活剂包含CD28激动剂。
  82. 根据权利要求69-81中任一项所述的方法,所述一种或多种T细胞激活剂包含抗CD28的抗体和/或其抗原结合片段。
  83. 根据权利要求69-82中任一项所述的方法,所述使TIL与所述一种或多种T细胞激活剂接触包含选自以下组的一种或多种方式:(1)将所述一种或多种T细胞激活剂添加至所述TIL的细胞培养基中;(2)将表达所述一种或多种T细胞激活剂的工程化细胞添加至所述TIL的细胞培养基中;(3)将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
  84. 根据权利要求83所述的方法,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为至少约30ng/mL。
  85. 根据权利要求83-84中任一项所述的方法,每一种所述T细胞激活剂在所述TIL的细胞培养基中的初始浓度各自独立地为约30ng/mL-约300ng/mL。
  86. 根据权利要求83-85中任一项所述的方法,所述固相介质的直径为约500纳米至约10微米。
  87. 根据权利要求83-86中任一项所述的方法,所述固相介质的直径为约1纳米至约500纳米。
  88. 根据权利要求83-87中任一项所述的方法,所述固相介质的直径通过透射电子显微镜测量。
  89. 根据权利要求83-88中任一项所述的方法,所述固相介质包含聚合物。
  90. 根据权利要求83-89中任一项所述的方法,每mg所述固相介质中包含的每一种所述T细胞激活剂的量各自独立地至少为约25μg。
  91. 根据权利要求83-90中任一项所述的方法,以约2:1-约1:2的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
  92. 根据权利要求83-91中任一项所述的方法,以约1:100-约1:2000的所述固相介质与所述TIL的比例,将包含所述一种或多种T细胞激活剂的固相介质添加至所述TIL的细胞培养基中。
  93. 根据权利要求69-92中任一项所述的方法,所述免疫检查点抑制剂包含抑制PD-1与PD-L1和/或PD-L2相互作用的物质。
  94. 根据权利要求69-93中任一项所述的方法,所述免疫检查点抑制剂包含PD-1抑制剂。
  95. 根据权利要求69-94中任一项所述的方法,所述免疫检查点抑制剂包含PD-1的抗体和/或其抗原结合片段。
  96. 根据权利要求69-95中任一项所述的方法,所述免疫检查点抑制剂包含具有以约100pM 或更低的K D值结合PD-1的能力的免疫检查点抑制剂。
  97. 根据权利要求69-96中任一项所述的方法,所述免疫检查点抑制剂包含具有以约100pM或更低的EC 50值结合PD-1的能力的免疫检查点抑制剂。
  98. 根据权利要求69-97中任一项所述的方法,所述免疫检查点抑制剂包含具有以约1nM或更低的IC 50值抑制PD-1与PD-L1和/或PD-L2结合的能力的免疫检查点抑制剂。
  99. 根据权利要求69-98中任一项所述的方法,所述免疫检查点抑制剂包含HCDR3,且所述HCDR3包含SEQ ID NO:3和17中任一项所示的氨基酸序列。
  100. 根据权利要求69-99中任一项所述的方法,所述免疫检查点抑制剂包含HCDR2,且所述HCDR2包含SEQ ID NO:2和16中任一项所示的氨基酸序列。
  101. 根据权利要求69-100中任一项所述的方法,所述免疫检查点抑制剂包含HCDR1,且所述HCDR1包含SEQ ID NO:1和15中任一项所示的氨基酸序列。
  102. 根据权利要求69-101中任一项所述的方法,所述免疫检查点抑制剂包含LCDR3,且所述LCDR3包含SEQ ID NO:6和20中任一项所示的氨基酸序列。
  103. 根据权利要求69-102中任一项所述的方法,所述免疫检查点抑制剂包含LCDR2,且所述LCDR2包含SEQ ID NO:5和19中任一项所示的氨基酸序列。
  104. 根据权利要求69-103中任一项所述的方法,所述免疫检查点抑制剂包含LCDR1,且所述LCDR1包含SEQ ID NO:4和18中任一项所示的氨基酸序列。
  105. 根据权利要求69-104中任一项所述的方法,所述免疫检查点抑制剂包含VH,且所述VH包含SEQ ID NO:7、11和21中任一项所示的氨基酸序列。
  106. 根据权利要求69-105中任一项所述的方法,所述免疫检查点抑制剂包含VL,且所述VL包含SEQ ID NO:8、12和22中任一项所示的氨基酸序列。
  107. 根据权利要求69-106中任一项所述的方法,所述免疫检查点抑制剂选自以下组:(1)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:1和15中任一项所示的HCDR1,氨基酸序列如SEQ ID NO:2和16中任一项所示的HCDR2,以及氨基酸序列如SEQ ID NO:3和17中任一项所示的HCDR3,且所述轻链包含氨基酸序列如SEQ ID NO:4和18中任一项所示的LCDR1,氨基酸序列如SEQ ID NO:5和19中任一项所示的LCDR2,以及氨基酸序列如SEQ ID NO:6和20中任一项所示的LCDR3;(2)包含下述重链和轻链的抗体或其抗原结合片段,所述重链包含氨基酸序列如SEQ ID NO:7、11和21中任一项所示的VH,且所述轻链包含氨基酸序列如SEQ ID NO:8、12和22中任一项所示的VL。
  108. 根据权利要求95-107中任一项所述的方法,其中所述抗体选自:嵌合抗体,人源化抗体和全人源抗体。
  109. 根据权利要求95-108中任一项所述的方法,其中所述抗原结合片段选自:Fab,Fab′,Fv片段,F(ab') 2,F(ab) 2,scFv,di-scFv,VHH和dAb。
  110. 根据权利要求69-109中任一项所述的方法,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地至少为约0.1μg/mL。
  111. 根据权利要求69-110中任一项所述的方法,每一种所述免疫检查点抑制剂在所述TIL的细胞培养基中的初始浓度各自独立地为约0.1μg/mL-约20μg/mL。
  112. 根据权利要求69-111中任一项所述的方法,其中,在步骤(B)中,使所述TIL基本上同时与所述一种或多种T细胞激活剂以及所述一种或多种T细胞生长因子接触。
  113. 根据权利要求69-112中任一项所述的方法,所述一种或多种T细胞生长因子选自以下组的一种或多种:IL-2、IL-7、IL-12、IL-15、IL-21、γ干扰素、以及它们的功能活性片段。
  114. 根据权利要求69-113中任一项所述的方法,所述一种或多种T细胞生长因子包含IL-2和/或其功能活性片段。
  115. 根据权利要求69-114中任一项所述的方法,所述TIL与所述一种或多种T细胞生长因子接触包含将所述T细胞生长因子添加至所述TIL的细胞培养基中。
  116. 根据权利要求69-115中任一项所述的方法,每一种所述T细胞生长因子在所述TIL的细胞培养基中的初始浓度各自独立地为至少约300IU/mL。
  117. 根据权利要求69-116中任一项所述的方法,其还包含:在步骤(A)、步骤(B)和/或步骤(C)中,使所述TIL与饲养细胞共培养。
  118. 根据权利要求117所述的方法,其中,在步骤(B)中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触且与所述饲养细胞共培养。
  119. 根据权利要求117-118中任一项所述的方法,其中,在步骤(B)中,使所述TIL与所述一种或多种T细胞激活剂和/或所述一种或多种T细胞生长因子接触一定时间之后,再与所述饲养细胞共培养。
  120. 根据权利要求119所述的方法,所述一定时间为至少约2小时。
  121. 根据权利要求119-120中任一项所述的方法,所述一定时间为约6小时至约72小时。
  122. 根据权利要求119-121中任一项所述的方法,所述一定时间为约12小时至约48小时。
  123. 根据权利要求119-121中任一项所述的方法,所述一定时间为约6小时、约12小时、约24小时、约48小时、或约72小时。
  124. 根据权利要求119-123中任一项所述的方法,所述饲养细胞包含抗原呈递细胞。
  125. 根据权利要求119-124中任一项所述的方法,所述饲养细胞包含选自以下组的一种或多种:外周单个核细胞,树突状细胞,和人工抗原呈递细胞。
  126. 根据权利要求119-125中任一项所述的方法,所述饲养细胞为外周单个核细胞。
  127. 根据权利要求119-126中任一项所述的方法,所述饲养细胞为经过辐照的饲养细胞。
  128. 根据权利要求119-127中任一项所述的方法,所述TIL与所述饲养细胞共培养包含使所述饲养细胞的表面与所述TIL的表面相接触。
  129. 根据权利要求119-128中任一项所述的方法,所述TIL与所述饲养细胞共培养包含将所述饲养细胞添加至所述TIL的细胞培养基中。
  130. 根据权利要求119-129中任一项所述的方法,以约40:1-约400:1的所述饲养细胞与所述TIL的比例,将所述饲养细胞添加至所述TIL的细胞培养基中。
  131. 根据权利要求69-130中任一项所述的方法,所述源自肿瘤组织且未经体外扩增的TIL为源自所述肿瘤组织的碎片的TIL。
  132. 根据权利要求131所述的方法,所述碎片的体积为约1立方毫米至约27立方毫米。
  133. 一种肿瘤浸润淋巴细胞(TIL),所述TIL经过权利要求1-132中任一项所述的方法获得。
  134. 一种组合物,其包含权利要求133所述的TIL。
  135. 一种药物组合物,其包含权利要求133所述的TIL和/或权利要求134所述的组合物,以及任选地药学上可接受的载体。
  136. 一种影响肿瘤细胞生长的方法,包含向受试者施用权利要求133所述的TIL、权利要求134所述的组合物和/或权利要求135所述的药物组合物。
  137. 权利要求133所述的TIL、权利要求134所述的组合物和/或权利要求135所述的药物组合物在制备药物中的应用,所述药物用于预防和/或***。
  138. 根据权利要求137所述的应用,其中,所述肿瘤为实体瘤。
  139. 根据权利要求137-138中任一项所述的应用,其中,所述肿瘤选自以下组的一种或多种:黑色素瘤、卵巢癌、***、肺癌、膀胱癌、乳腺癌、头颈癌、胰腺癌、肝癌、胃癌、结直肠癌、和肾癌。
PCT/CN2022/075341 2021-02-08 2022-02-07 肿瘤浸润淋巴细胞的制备方法及其用途 WO2022166947A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280003156.8A CN115315509B (zh) 2021-02-08 2022-02-07 肿瘤浸润淋巴细胞的制备方法及其用途

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110180195.2 2021-02-08
CN202110180195 2021-02-08

Publications (1)

Publication Number Publication Date
WO2022166947A1 true WO2022166947A1 (zh) 2022-08-11

Family

ID=82740850

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/075341 WO2022166947A1 (zh) 2021-02-08 2022-02-07 肿瘤浸润淋巴细胞的制备方法及其用途

Country Status (2)

Country Link
CN (1) CN115315509B (zh)
WO (1) WO2022166947A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023138598A1 (zh) * 2022-01-19 2023-07-27 苏州沙砾生物科技有限公司 肿瘤浸润淋巴细胞在疾病治疗中的用途

Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103429264A (zh) * 2011-03-31 2013-12-04 默沙东公司 针对人程序性死亡受体pd-1的抗体的稳定制剂和有关的治疗
CN105713878A (zh) * 2015-12-21 2016-06-29 杭州特马赛生物技术有限公司 一种体外扩增cd8+t细胞的方法
CN106119194A (zh) * 2016-08-04 2016-11-16 英普乐孚生物技术(上海)有限公司 一种恶性胸水来源的til细胞的分离培养方法
CN106244538A (zh) * 2016-08-04 2016-12-21 英普乐孚生物技术(上海)有限公司 一种恶性腹水来源的til细胞的分离培养方法
CN110199016A (zh) * 2016-11-17 2019-09-03 艾欧凡斯生物治疗公司 残余肿瘤浸润淋巴细胞及其制备和使用方法
CN110462027A (zh) * 2017-01-06 2019-11-15 艾欧凡斯生物治疗公司 用肿瘤坏死因子受体超家族(tnfrsf)激动剂扩增肿瘤浸润淋巴细胞(til)及til和tnfrsf激动剂的治疗组合
CN110938594A (zh) * 2019-09-12 2020-03-31 夏建川 一种功能增强型til细胞的培养方法
CN111228472A (zh) * 2020-01-16 2020-06-05 卡替(上海)细胞生物技术有限公司 通过til免疫细胞改善胃癌的方法
CN111801415A (zh) * 2017-11-06 2020-10-20 路德维格癌症研究院 扩增淋巴细胞的方法
CN111836887A (zh) * 2018-01-08 2020-10-27 艾欧凡斯生物治疗公司 产生富含肿瘤抗原特异性t细胞的til产品的方法

Patent Citations (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN103429264A (zh) * 2011-03-31 2013-12-04 默沙东公司 针对人程序性死亡受体pd-1的抗体的稳定制剂和有关的治疗
CN105713878A (zh) * 2015-12-21 2016-06-29 杭州特马赛生物技术有限公司 一种体外扩增cd8+t细胞的方法
CN106119194A (zh) * 2016-08-04 2016-11-16 英普乐孚生物技术(上海)有限公司 一种恶性胸水来源的til细胞的分离培养方法
CN106244538A (zh) * 2016-08-04 2016-12-21 英普乐孚生物技术(上海)有限公司 一种恶性腹水来源的til细胞的分离培养方法
CN110199016A (zh) * 2016-11-17 2019-09-03 艾欧凡斯生物治疗公司 残余肿瘤浸润淋巴细胞及其制备和使用方法
CN110462027A (zh) * 2017-01-06 2019-11-15 艾欧凡斯生物治疗公司 用肿瘤坏死因子受体超家族(tnfrsf)激动剂扩增肿瘤浸润淋巴细胞(til)及til和tnfrsf激动剂的治疗组合
CN111801415A (zh) * 2017-11-06 2020-10-20 路德维格癌症研究院 扩增淋巴细胞的方法
CN111836887A (zh) * 2018-01-08 2020-10-27 艾欧凡斯生物治疗公司 产生富含肿瘤抗原特异性t细胞的til产品的方法
CN110938594A (zh) * 2019-09-12 2020-03-31 夏建川 一种功能增强型til细胞的培养方法
CN111228472A (zh) * 2020-01-16 2020-06-05 卡替(上海)细胞生物技术有限公司 通过til免疫细胞改善胃癌的方法

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
ANONYMOUS: "Light Chain of Nivolumab in PDB entry 5wt9 ‹ PDBe ‹ EMBL-EBI", PROTEIN DATA BANK IN EUROPE, 1 December 2020 (2020-12-01), pages 1 - 2, XP055957237, Retrieved from the Internet <URL:https://www.ebi.ac.uk/pdbe/entry/pdb/5wt9/protein/2> [retrieved on 20220902] *
GEUKES FOPPEN M.H.; DONIA M.; SVANE I.M.; HAANEN J.B.A.G.: "Tumor-infiltrating lymphocytes for the treatment of metastatic cancer", MOLECULAR ONCOLOGY, ELSEVIER, vol. 9, no. 10, 1 January 1900 (1900-01-01), pages 1918 - 1935, XP029341866, ISSN: 1574-7891, DOI: 10.1016/j.molonc.2015.10.018 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023138598A1 (zh) * 2022-01-19 2023-07-27 苏州沙砾生物科技有限公司 肿瘤浸润淋巴细胞在疾病治疗中的用途

Also Published As

Publication number Publication date
CN115315509A (zh) 2022-11-08
CN115315509B (zh) 2024-05-07

Similar Documents

Publication Publication Date Title
JP7451627B2 (ja) キメラ受容体及びその使用方法
JP7034934B2 (ja) キメラ抗原及びt細胞受容体、並びに使用方法
KR102660362B1 (ko) 조작된 감마 델타 t 세포
TW202045547A (zh) 靶向dll3的嵌合抗原受體和結合劑
JP2021513839A (ja) 改変された多能性幹細胞並びに製造方法及び使用方法
WO2022135525A1 (zh) 肿瘤浸润淋巴细胞的制备方法及其用途
KR20210038922A (ko) 키메라 항원 수용체 요법 t 세포 확장 동역학 및 그의 용도
WO2022105816A1 (zh) 肿瘤浸润淋巴细胞的培养方法及其用途
WO2022166947A1 (zh) 肿瘤浸润淋巴细胞的制备方法及其用途
WO2022223013A1 (zh) 一种修饰的肿瘤浸润淋巴细胞及其用途
JP2023553634A (ja) 治療用のcar t細胞を特徴付けるための方法および試薬
US20200339944A1 (en) Methods of manufacturing allogeneic car t cells
KR20210144679A (ko) TCRαβ+ 세포 고갈 효율을 향상시키는 방법
WO2023284721A1 (zh) 一种免疫细胞的培养方法及其用途
WO2023011433A1 (zh) 一种修饰的肿瘤浸润淋巴细胞及其用途
WO2023011434A1 (zh) 一种修饰的免疫细胞及其用途
CN114908050B (zh) 肿瘤浸润淋巴细胞的制备方法及其用途
WO2023125772A1 (zh) 一种修饰的肿瘤浸润淋巴细胞及其用途
WO2022228492A1 (zh) 一种修饰的肿瘤浸润淋巴细胞及其用途
WO2023138598A1 (zh) 肿瘤浸润淋巴细胞在疾病治疗中的用途
JP2024504397A (ja) Car t細胞医薬品のための製剤および方法
JP2024510162A (ja) 免疫細胞機能の改善

Legal Events

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

Ref document number: 22749226

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22749226

Country of ref document: EP

Kind code of ref document: A1