WO2024092508A1 - Use of mesenchymal stem cell - Google Patents

Use of mesenchymal stem cell Download PDF

Info

Publication number
WO2024092508A1
WO2024092508A1 PCT/CN2022/128989 CN2022128989W WO2024092508A1 WO 2024092508 A1 WO2024092508 A1 WO 2024092508A1 CN 2022128989 W CN2022128989 W CN 2022128989W WO 2024092508 A1 WO2024092508 A1 WO 2024092508A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
immune effector
cell
mesenchymal stem
receptor
Prior art date
Application number
PCT/CN2022/128989
Other languages
French (fr)
Chinese (zh)
Inventor
钟晓松
顾爱琴
Original Assignee
卡瑞济(北京)生命科技有限公司
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 卡瑞济(北京)生命科技有限公司 filed Critical 卡瑞济(北京)生命科技有限公司
Priority to PCT/CN2022/128989 priority Critical patent/WO2024092508A1/en
Publication of WO2024092508A1 publication Critical patent/WO2024092508A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/28Bone marrow; Haematopoietic stem cells; Mesenchymal stem cells of any origin, e.g. adipose-derived stem cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • 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

Definitions

  • the present invention relates to the field of cell therapy, and in particular to the use of mesenchymal stem cells for enhancing immunotherapy and corresponding pharmaceutical compositions.
  • Chimeric antigen receptor is a synthetic molecule that specifically recognizes antigens expressed on the surface of tumor cells to guide immune effector cells (e.g., T cells, NK cells) genetically engineered to express CAR to eliminate tumors (see Sampson JH et al., EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clinical cancer research: an official journal of the American Association for Cancer Research. 2014; 20(4): 972-984).
  • immune effector cells e.g., T cells, NK cells
  • CAR-T chimeric antigen receptor T cells
  • CAR-T directly target surface antigens of tumor cells through chimeric antigen receptor (CAR) molecules on T cells, thereby achieving the purpose of recognizing and killing tumors, wherein the N-terminus of the chimeric antigen receptor contains an extracellular domain that recognizes the antigen.
  • CAR-T cells can recognize and kill these antigen-positive cells.
  • CAR-T cells have obvious advantages in the treatment of malignant tumors. More and more reports have shown that CAR-T cells are very effective in treating refractory hematological tumors (see Sadelain M et al., Therapeutic T cell engineering. Nature.
  • CAR-T therapy does not meet expectations, one of the most important reasons may be that the related CAR-T cells have poor expansion and limited persistence.
  • the in vivo expansion ability and persistence of CAR-T cells are related to the strength of CAR molecular signals, cytokine selection, stimulation domain, and T cell phenotype.
  • CAR-T cells are also affected by the differentiation of CAR-T cells.
  • Infusion of CAR-T cells with naive-like (TN) and central memory (TCM) phenotypes is associated with T cell persistence in vivo and superior antitumor efficacy (see: Wilkie S et al., Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J Biol Chem. 2010 Aug 13; 285(33): 25538-44.; Shum T et al., Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells.
  • MSC Mesenchymal stem cells
  • Mesenchymal stem cells have inherent immunomodulatory properties, nutritional capacity, high self-renewal ability and strong differentiation potential in vitro.
  • Mesenchymal stem cells affect the function of most immune effector cells through direct contact with immune cells and paracrine activity, and can be easily designed to enhance their immunomodulatory function (see: Song N et al., Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential. Trends Pharmacol Sci. 2020 Sep; 41(9): 653-664.). It has been reported that MSCs inhibit the function of T cells by inhibiting the activation and proliferation of T cells (see: Ghannam S et al., Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol.
  • IL7-IL12 engineered mesenchymal stem cells improved CAR-T cell attack against colorectal cancer cells (see: Hombach AA et al., IL7-IL12 Engineered Mesenchymal Stem Cells (MSCs) Improve A CAR T Cell Attack against Colorectal Cancer Cells. Cells. 2020 Apr 3; 9(4): 873.).
  • MSCs Mesenchymal Stem Cells
  • the present invention has discovered for the first time the effect pattern of mesenchymal stem cells on CAR-T function, and for the first time provided the use of mesenchymal stem cells as CAR-T function enhancers. Specifically, the present invention co-cultured mesenchymal stem cells induced and cultured from the placenta of healthy donors and pregnant women with CAR-T cells of different targets, thereby confirming the effect of mesenchymal stem cells on the killing function of CAR-T cells. Furthermore, the present invention has confirmed for the first time that this effect is manifested through the persistence and differentiation of CAR-T cells.
  • the present invention relates to the use of mesenchymal stem cells for enhancing the efficacy of immunotherapy, wherein the surface of the mesenchymal stem cells does not express or substantially does not express the antigens targeted by the immunotherapy.
  • immunotherapy is cell therapy, that is, a therapy that administers therapeutic cells, preferably, therapeutic cells are immune effector cells.
  • therapeutic cells are nonspecific immune effector cells.
  • therapeutic cells are specific immune effector cells, preferably, immune effector cells are T cells; preferably, immune effector cells are modified immune effector cells; more preferably, immune effector cells are CAR-T cells.
  • human T cells are CD8 + T cells.
  • therapeutic cells are TIL, CAR-NK, TCR-T, CAR-DC, DC loaded with antigens, B cells, etc.
  • the disease targeted by the immunotherapy is a tumor.
  • the tumor is a hematological tumor.
  • the tumor is a solid tumor.
  • the immunotherapy is directed against one or more tumor-associated antigens (TAA) expressed by the tumor, wherein the MSC surface does not express or substantially does not express the tumor-associated antigen molecule.
  • TAA tumor-associated antigen
  • the tumor-associated antigen is IL-13R ⁇ 2, EphA2, CD19 and/or EGFRvIII.
  • mesenchymal stem cells are derived from bone marrow, fat, peripheral blood, umbilical cord and/or placenta. In some embodiments, mesenchymal stem cells are derived from umbilical cord and/or placenta. In some embodiments, mesenchymal stem cells are derived from bone marrow, fat and/or placenta. In some embodiments, mesenchymal stem cells are derived from bone marrow and/or placenta. In some embodiments, mesenchymal stem cells are derived from placenta.
  • the mesenchymal stem cells are co-cultured with the immune effector cells in vitro. In some embodiments, the mesenchymal stem cells are co-administered with the immune effector cells into a subject, wherein the mesenchymal stem cells are administered prior to the immune effector cells, or administered simultaneously with the immune effector cells, or administered after the immune effector cells are administered.
  • the ratio of the number of immune effector cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
  • mesenchymal stem cells promote the immune effector cells (e.g., preferably, CAR-T cells) to secrete cytokines to kill tumor cells.
  • mesenchymal stem cells enhance the direct killing effect of immune effector cells (e.g., preferably, CAR-T cells) on target cells.
  • mesenchymal stem cells promote the proliferation and amplification of immune effector cells (e.g., preferably, CAR-T cells), that is, the number increases.
  • mesenchymal stem cells promote the persistence of therapeutic cells (e.g., preferably, CAR-T cells), that is, the time of survival/existence in vivo is prolonged.
  • mesenchymal stem cells enhance the ability of immune effector cells (e.g., preferably, CAR-T cells) to reach the tumor area.
  • the efficacy enhancer promotes immune effector cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more immune effector cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increase the number of immune effector cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier)
  • mesenchymal stem cells regulate the differentiation of immune effector cells (e.g., preferably, CAR-T cells) to maintain a low differentiation state.
  • mesenchymal stem cells promote immune effector cells (e.g., preferably, CAR-T cells) to differentiate into more memory cell subtypes.
  • the enhancement is achieved by increasing the persistence of immune effector cells and/or maintaining a low differentiation state and/or more differentiation into memory effector cell subtypes.
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • cytokines e.g., interferon gamma (IFN- ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), IL-4, IL-6, IL-10, and/or IL-17A
  • IFN- ⁇ interferon gamma
  • TNF- ⁇ tumor necrosis factor alpha
  • IL-4 tumor necrosis factor alpha
  • IL-6 tumor necrosis factor alpha
  • IL-17A IL-17A
  • immune effector cells e.g., CAR-T cells
  • the immune effector cell is modified.
  • the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • immune effector cells e.g., CAR-T cells
  • the immune effector cell is modified.
  • the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • the immune effector cell is modified.
  • the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
  • the present invention provides the use of mesenchymal stem cells for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of single immune effector cells after administration.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention also provides the use of mesenchymal stem cells for regulating the differentiation of immune effector cells in vivo after administration.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention also provides the use of mesenchymal stem cells for promoting the differentiation of immune effector cells into memory cell subtypes in vivo after administration.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention relates to the use of mesenchymal stem cells in the preparation of anti-tumor drugs, wherein the drug also contains other cells.
  • the other cells are immune effector cells, and the surface of the mesenchymal stem cells does not express or substantially does not express the antigen specifically targeted by the CAR-T cells.
  • the tumor is a hematological tumor. In some more specific embodiments, the tumor is a solid tumor.
  • immune effector cells are T cells.
  • immune effector cells are modified, preferably, transduced and express chimeric antigen receptor (CAR) T cells, i.e., CAR-T cells.
  • CAR chimeric antigen receptor
  • the specific immune effector cells are directed against one or more tumor-associated antigens expressed by the tumor.
  • the target TAA directed against by the specific immune effector cells is IL-13R ⁇ 2, EphA2, CD19 or EGFRvIII.
  • the mesenchymal stem cells are of placental origin. In some specific embodiments, the placental-derived mesenchymal stem cells express CD105 and do not express CD45.
  • the mesenchymal stem cells are co-cultured in vitro with therapeutic cells used in cell therapy. In some embodiments, the mesenchymal stem cells are co-administered with therapeutic cells used in cell therapy into a subject, wherein the mesenchymal stem cells are administered prior to, or simultaneously with, or after the therapeutic cells are administered.
  • the number ratio of the therapeutic cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
  • the present invention relates to the use of mesenchymal stem cells in combination with other cells for preparing anti-tumor drugs, wherein the surface of the mesenchymal stem cells does not substantially express antigens specifically targeted by other cells, and the cancer cells express antigens specifically targeted by other cells.
  • the tumor is a hematological tumor. In some more specific embodiments, the tumor is a solid tumor.
  • immune effector cells are T cells.
  • immune effector cells are modified, preferably, transduced and express chimeric antigen receptor (CAR) T cells, i.e., CAR-T cells.
  • CAR chimeric antigen receptor
  • the specific immune effector cells are directed against one or more tumor-associated antigens expressed by the tumor.
  • the target TAA directed against by the specific immune effector cells is IL-13R ⁇ 2, EphA2, CD19 or EGFRvIII.
  • the mesenchymal stem cells are of placental origin. In some specific embodiments, the mesenchymal stem cells express CD105 and do not express CD45.
  • the mesenchymal stem cells are co-cultured in vitro with therapeutic cells used in cell therapy. In some embodiments, the mesenchymal stem cells are co-administered with therapeutic cells used in cell therapy into a subject, wherein the mesenchymal stem cells are administered prior to, or simultaneously with, or after the therapeutic cells are administered.
  • the number ratio of the therapeutic cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
  • the present invention provides a composition comprising mesenchymal stem cells, preferably placental-derived mesenchymal stem cells.
  • the composition is used as an immunotherapy efficacy enhancer.
  • the immunotherapy is directed against hematological tumors.
  • the immunotherapy is directed against solid tumors.
  • the target molecule of the immunotherapy is IL-13R ⁇ 2, EphA2, CD19 or EGFRvIII.
  • the composition promotes therapeutic cells (e.g., preferably, CAR-T cells) to secrete cytokines to kill tumor cells.
  • the composition enhances the direct killing effect of therapeutic cells (e.g., preferably, CAR-T cells) on target cells.
  • the composition promotes the proliferation of therapeutic cells (e.g., preferably, CAR-T cells), that is, the number increases.
  • the composition enhances the ability of therapeutic cells (e.g., preferably, CAR-T cells) to reach the tumor area.
  • the efficacy enhancer promotes therapeutic cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more therapeutic cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increases the number of therapeutic cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier).
  • mesenchymal stem cells regulate the differentiation of immune effector cells (e.g., preferably, CAR-T cells) to maintain a low differentiation state.
  • mesenchymal stem cells promote immune effector cells (e.g., preferably, CAR-T cells) to differentiate more into memory cell subtypes.
  • the enhancement is achieved by increasing the persistence of immune effector cells and/or remaining in a poorly differentiated state and/or more differentiation into memory effector cell subtypes.
  • the present invention provides the use of mesenchymal stem cells for preparing one or more of the following:
  • the immune effector cell is modified.
  • the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
  • the present invention provides the use of mesenchymal stem cells in combination with immune effector cells for preparing a drug for treating cancer, wherein, preferably, the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the aforementioned mesenchymal stem cells (e.g., placental-derived mesenchymal stem cells) or compositions of the present invention, and further comprising a suitable pharmaceutically acceptable carrier.
  • the pharmaceutically acceptable carrier makes the pharmaceutical composition suitable for administration by the intended route, for example, but not limited to, intravenous injection or local injection of tumors, etc.
  • the pharmaceutical composition of the present invention further comprises other cells (e.g., preferably, therapeutic cells, more preferably, CAR-T cells).
  • the present invention provides a method comprising administering immune effector cells to a subject in need thereof, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the subject has a condition described herein, e.g., the subject has cancer, e.g., the subject has a cancer that expresses a target antigen described herein.
  • the subject is a human.
  • the present invention relates to a method for treating a subject with a disease associated with the expression of a cancer-associated antigen as described herein, comprising administering immune effector cells to a subject in need thereof, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention provides a method for treating a subject with a disease associated with the expression of a tumor antigen, comprising administering an immune effector cell to a subject in need thereof, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cell is modified.
  • the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering with mesenchymal stem cells to the body.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vitro/ex vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells to the body.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention also provides a method for regulating the differentiation of immune effector cells in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the present invention also provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • Figures 1-4 Characterization of placental mesenchymal stem cells from healthy natural delivery pregnant women.
  • Figure 1 shows the microscopic morphology of placental-derived mesenchymal stem cells.
  • Figure 2 shows a flow chart for counting the proliferation of mesenchymal stem cells in vitro (top), and the calculation of the proliferation of mesenchymal stem cells according to the population doubling of six passages in vitro (bottom).
  • Figure 3 shows the immunophenotype of the third generation of MSCs amplified by flow cytometry analysis, with the vertical axis reflecting the number of cells and the horizontal axis representing the staining intensity.
  • the left figure shows that the cell population CD45-specific staining (by anti-CD45mAb) is basically negative; the right figure shows the cell population CD105-specific staining (by anti-CD105mAb), the dark curve represents the isotype-matched negative control mAb staining, and the light color represents the mAb-specific stained cells. It can be seen that the vast majority of cells can be clearly stained with CD105 antibodies.
  • Figure 5 Detection of tumor surface antigen expression in MSC by flow cytometry.
  • A Negative control
  • B C, D
  • MSC mesenchymal stem cell.
  • Figures 6-10 Preparation of CAR-T cells and the effect of mesenchymal stem cells on in vitro CAR-T function.
  • Figure 6 shows a schematic diagram of the structures of three CAR molecules that specifically target CD19, IL13R ⁇ 2 and uPAR.
  • Figure 7 shows a flowchart of the preparation of CAR-T cells together with in vitro functional studies by time nodes.
  • Figure 8 shows the effect of MSC on the proliferation of CD19-IL15 CAR-T cells detected by flow cytometry.
  • CAR-T cells were first co-cultured with mesenchymal stem cells at a ratio of 10:1 for 24 hours, and then collected and co-cultured with/without NALM-6 cells at a ratio of 1:1 for 3 days for flow cytometric analysis.
  • FIG. 9 shows the effect of MSC on the activation of CD19-IL15 CAR-T cells detected by flow cytometry. CAR-T cells were co-cultured with MSC in advance and co-cultured with/without NALM-6 for 6 hours.
  • Figure 10 shows the effect of MSC on cytokine release of CD19-IL15 CAR-T cells detected by CBA method.
  • CAR-T cells were co-cultured with MSC for 24 hours, and then co-cultured with NALM-6 (A) and Raji (B) cells overnight, with an E:T ratio of 1:1, and the co-culture supernatant was collected for cytokine detection.
  • Figures 11-13 Effects of MSCs on the anti-tumor function of CD19-IL15 CAR-T cells in vitro.
  • Figure 11 shows the effect of MSCs on CD19-IL15 CAR-T cells against NALM-6-GL cell lines by flow cytometry analysis.
  • CAR-T cells were first co-cultured with MSCs at a ratio of 10:1 and then co-cultured with NALM-6-GL at a ratio of 1:1.
  • Flow cytometry result analysis graphs and statistical bar graphs are provided.
  • Figure 12 shows the effect of MSCs on CD19-IL15 CAR-T cells against Raji-GL cell lines by flow cytometry analysis.
  • CAR-T cells were co-cultured with mesenchymal stem cells at a ratio of 10:1 in advance and then co-cultured with Raji-GL at a ratio of 1:1.
  • Flow cytometry result analysis graphs and statistical bar graphs are provided.
  • Figure 13 shows the effect of MSCs on CD19-IL15 CAR-T cells against NALM-6-GL cells (A) and against Raji-GL cells (B) by detecting luciferase activity.
  • CAR-T cells were first co-cultured with MSCs at a ratio of 10:1, and then collected and co-cultured with NALM-GL or Raji-GL at three ratios of 1:1, 2:1, and 5:1.
  • Figures 14-16 Effect of MSC on the function of IL13R ⁇ 2 CAR-T cells against U87 cells.
  • Figure 14 shows the activation of CD107a analyzed by flow cytometry.
  • CAR-T cells were first co-cultured with mesenchymal stem cells at a ratio of 10:1, and then co-cultured with/without U87-GL cell line for 6 hours.
  • Figure 15 shows the release of cytokines detected by ELISA.
  • CAR-T cells were co-cultured with MSC for 24 hours, then collected and co-cultured with U87-GL cells overnight at an E:T ratio of 1:1, and the supernatant was obtained for cytokine detection.
  • the 5 bars from left to right represent the U87 cell group alone, the IL13R ⁇ 2 CAR-T cell group alone, the IL13R ⁇ 2 CAR-T cell + U87 cell group, the IL13R ⁇ 2 CAR-T cell + U87 cell group co-cultured with MSC, and the IL13R ⁇ 2 CAR-T cell group co-cultured with MSC.
  • Figure 16 shows the anti-tumor effect analyzed by flow cytometry. CAR-T cells were co-cultured with MSC for 24 hours and then collected and co-cultured with U87-GL cells overnight, with an E:T ratio of 1:1. Flow cytometry result analysis chart and statistical bar graph are provided. "*" represents p ⁇ 0.05; "**” represents p ⁇ 0.01; "***” represents p ⁇ 0.001.
  • GL eGFP-Luc
  • Figures 17-21 Effects of MSC on the function of uPAR CAR-T cells against H460-GL cells.
  • Figure 17 shows the activation of CAR-T cell CD107a.
  • Figure 18 shows the detection of cytokine release.
  • Figure 19 shows the in vitro anti-tumor effect of an E:T ratio of 1:1, and provides a flow cytometry result analysis chart and a statistical bar chart.
  • Figure 20 shows the anti-tumor effect of uPAR CAR-T cells at different E:T ratios by detecting luciferase activity.
  • Figure 21 shows that uPAR CAR-T cells exhibit killing function against MSC.
  • Figures 22-25 Effect of MSC on differentiation of CD19-IL15 CAR-T cells.
  • Figure 22 shows flow cytometric analysis of differentiation of TCM phenotype detected by flow cytometry.
  • CAR-T cells were first co-cultured with mesenchymal stem cells for 24 hours, then collected and co-cultured with/without NALM-6 cells.
  • Figure 23 shows a bar graph of statistical results of the same assay.
  • Figure 24 shows the expression of transcription factor TCF-7 detected by qRT-PCR.
  • Figure 25 shows the release of cytokine IL2 detected by ELISA.
  • CAR-T cells were cultured in medium without IL2 for 3 days, co-cultured with MSC for 24 hours, and then cultured with NALM-6 (A) or Raji cells (B) in the same medium without IL2 overnight, and the co-culture supernatant was obtained for IL2 detection.
  • "*" represents p ⁇ 0.05;
  • "**” represents p ⁇ 0.01;
  • "***” represents p ⁇ 0.001.
  • Figures 26-29 Effect of MSC on the anti-tumor effect of CD19-IL15 CAR-T cells in the NALM-6-GL xenograft mouse model.
  • Figure 27 shows the tumor fluorescence imaging of mice
  • Figure 28 shows the relative quantitative results of fluorescence for each mouse, among which the survival period of the tumor-bearing mice in the control group of CAR-T cells was extremely short, and only the relevant data for the first two weeks were obtained, which is also reflected in the survival curve of the mice shown in Figure 29.
  • "*" represents p ⁇ 0.05;
  • ** represents p ⁇ 0.01;
  • *** represents p ⁇ 0.001.
  • meenchymal stem cells also known in the art as “mesenchymal stem cells (MSC)” refers to a group of multipotent stromal cells derived from the mesoderm, with certain differentiation potential and can differentiate into a variety of cell types. They are mainly derived from and exist in the bone marrow, and also include multipotent cells widely derived from other "non-bone marrow" tissues, such as: placenta, umbilical cord blood, adipose tissue, adult muscle, corneal stroma, deciduous tooth pulp, etc.
  • MSC meenchymal stem cells
  • the isolated mesenchymal stem cell population described herein can be produced by the following method: digesting tissues (e.g., but not limited to, bone marrow, placenta, adipose, umbilical cord, peripheral blood, etc.) containing mesenchymal stem cells with tissue-destroying enzymes to obtain a mesenchymal stem cell population containing mesenchymal stem cells, and isolating or substantially isolating a plurality of mesenchymal stem cells from the remainder. All or any portion of the tissue can be digested to obtain the isolated mesenchymal stem cells described herein.
  • tissues e.g., but not limited to, bone marrow, placenta, adipose, umbilical cord, peripheral blood, etc.
  • the mesenchymal stem cells used as described herein adhere to a tissue culture substrate, for example, a tissue culture vessel surface (e.g., a tissue culture plastic).
  • the mesenchymal stem cells in culture present a general fibroblast-like star-shaped appearance, in which a plurality of cytoplasmic processes extend from a central cell body.
  • the mesenchymal stem cells show these processes more than the fibroblast number, the mesenchymal stem cells are distinguishable in morphology from the fibroblasts cultivated under the same conditions. In morphology, the mesenchymal stem cells are also distinguishable from hematopoietic stem cells, which generally present a more rounded or pebble morphology when cultivated.
  • the isolated mesenchymal stem cells (e.g., isolated multipotent mesenchymal stem cells or isolated mesenchymal stem cell populations) useful in the technical solutions disclosed herein are adherent cells (e.g., adherent to tissue culture plastic) that have the characteristics of multipotent cells or stem cells and express a variety of markers that can be used to identify and/or isolate the cells or cell populations containing the stem cells.
  • adherent cells e.g., adherent to tissue culture plastic
  • the separated mesenchymal stem cells are CD45 - and CD105 + , as detected by flow cytometry.
  • the mesenchymal stem cells are CD73 + , CD90 + and CD105 + , and CD14, CD34 and CD45 expression levels are extremely low/not expressed.
  • the separated mesenchymal stem cells have the potential to differentiate into neural phenotype cells, osteogenic phenotype cells and/or chondrogenic phenotype cells.
  • the mesenchymal stem cells express about 95% of CD105 on the surface, substantially do not express CD45, and can differentiate into osteoblasts after being exposed to osteogenic induction medium for 3-4 weeks.
  • the isolated mesenchymal stem cell populations described above may generally contain about, at least, or no more than 1 ⁇ 10 5 , 5 ⁇ 10 5 , 1 ⁇ 10 6 , 5 ⁇ 10 6 , 1 ⁇ 10 7 , 5 ⁇ 10 7 , 1 ⁇ 10 8 , 5 ⁇ 10 8 , 1 ⁇ 10 9 , 5 ⁇ 10 9 , 1 ⁇ 10 10 , 5 ⁇ 10 10 , 1 ⁇ 10 11 or more isolated mesenchymal stem cells.
  • the isolated mesenchymal stem cell populations useful in the methods of treatment described herein contain at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% viable isolated mesenchymal stem cells, e.g., as determined by, e.g., trypan blue exclusion.
  • the cells or mesenchymal stem cell populations are or can include cells that have been passaged for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 generations or more, or expanded for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 population doublings or more.
  • the mesenchymal stem cells of the present invention do not express or substantially do not express the antigen molecules specifically targeted by cell therapy.
  • the mesenchymal stem cells when referred to herein as "not expressing” and “substantially not expressing” a certain antigen, they have the same meaning, meaning that in a certain specified mesenchymal stem cell population, only a very small number of cells, for example, less than 20% of cells, less than 19% of cells, less than 18% of cells, less than 17% of cells, less than 16% of cells, less than 15% of cells, less than 14% of cells, less than 13% of cells, less than 12% of cells, less than 11% of cells, less than 10% of cells, Less than 9% of cells, less than 8% of cells, less than 7% of cells, less than 6% of cells, less than 5% of cells, less than 4% of cells, less than 3% of cells, less than 2% of cells or less than 1% of cells express the antigen to a
  • the separated mesenchymal stem cells do not express or substantially do not express antigens specifically targeted by immune effector cells (e.g., CAR-T cells). In some embodiments, the separated mesenchymal stem cells do not express or substantially do not express CD19.
  • Placental mesenchymal stem cells suitable for the present invention are Placental mesenchymal stem cells suitable for the present invention.
  • the isolated placental stem cell population described herein can be produced by the following method: digesting placental tissue with a tissue-destroying enzyme to obtain a placental cell population containing placental stem cells, and isolating or substantially isolating a plurality of placental stem cells from the remainder of the placental cells. All or any portion of the placenta can be digested to obtain the isolated placental stem cells described herein.
  • the placental tissue can be a whole placenta (e.g., including the umbilical cord), amnion, chorion, a combination of amnion and chorion, or any combination of the above.
  • the tissue-destroying enzyme is trypsin or collagenase.
  • the isolated placental stem cells contained in the cell population obtained from placental digestion are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of the placental cell population.
  • mesenchymal stem cells derived from the placenta can be fetal or maternal in origin (i.e., can have a maternal or fetal genotype).
  • a placental stem cell population or a cell population comprising placental stem cells can include placental stem cells that are only of fetal or maternal origin, or can include a mixed population of placental stem cells of fetal and maternal origin.
  • the isolated mesenchymal stem cells and/or placental cell populations described herein include placental stem cells obtained directly from the placenta or any part thereof (e.g., chorion, placental villous leaves, etc.) and cell populations containing placental cells.
  • Isolated placental cell populations also include (i.e., two or more) isolated placental stem cell populations in culture and cell populations in containers (e.g., bags).
  • immune effector cell refers to cells that have differentiated (i.e., not hematopoietic stem cells) from the human body and are capable of regulating or influencing the form or realization of an immune response, such as B cells, dendritic cells, natural killer cells, and T cells. It is known in the art to collect naturally occurring immune effector cells in vivo, optionally and preferably after a certain procedure of treatment in vitro, to convert them into therapeutic products and apply them to patients, i.e., immune effector cell therapy. These cell therapy products are an important part of immunotherapy, a new pillar of cancer treatment, which effectively uses the patient's own immune system to attack tumors. Immune effector cell therapy can also be designed to promote the immune system to stop attacking itself (in the presence of autoimmune diseases).
  • CAR-T cell therapy is the most studied immune effector cell therapy in recent years.
  • CAR-T or “CAR T” refers to a T lymphocyte transduced with and expressing a chimeric antigen receptor (CAR).
  • the chimeric antigen receptor refers to one or more sets of polypeptides that, when expressed on immune effector cells, provide the cells with specificity for a target antigen and have intracellular signal generation.
  • the target antigen is expressed by a target cell (usually a cancer cell).
  • CAR includes at least one extracellular binding region, a transmembrane region, and an intracellular signal region. Exemplary CAR construction methods and/or CAR-T cell transduction methods are described in, for example, Chinese Patent Publication No.
  • the CAR-T cell is constructed according to the first generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the second generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the third generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the fourth generation CAR-T technology. In some specific embodiments, the CAR molecule comprises an extracellular specific target antigen binding domain, preferably, the target antigen binding domain is a scFv.
  • the target antigen binding domain is a natural interaction molecule of the target antigen, such as, but not limited to, its ligand (when the target antigen is a receptor) or its soluble receptor fragment (when the target antigen is a ligand).
  • its ligand when the target antigen is a receptor
  • its soluble receptor fragment when the target antigen is a ligand
  • the target antigen is selected from one or more of the following: CD19, CD123, CD22, CD30, CD171, CS-1 (also known as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24), C-type lectin-like molecule-1 (CLL-1 or CLECL1), CD33, epidermal growth factor receptor variant III (EGFRvIII), ganglioside G2 (GD2), ganglioside GD3 (aNeu5Ac (2-8) aNeu5Ac (2-3) bDGalp (1-4) bDGlcp (1-1) Cer), TNF receptor family member B cell maturation (BCMA), Tn antigen ((Tn Ag) or (GalNAc ⁇ -Ser/Thr)), prostate specific membrane antigen (PSMA), receptor tyrosine kinase-like orphan receptor 1 (ROR1), Fms-like tyrosine kinase 3 (FLT3), tumor-associated glycoprotein 72 (TA), TNF receptor
  • the tumor antigen bound by the encoded CAR molecule is selected from one or more of the following: TSHR, CD171, CS-1, CLL-1, GD3, Tn Ag, FLT3, CD38, CD44v6, B7H3, KIT, IL-13R ⁇ 2, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR- ⁇ , SSEA-4, MUC1, EGFR, NCAM, CAIX, LMP2, EphA2, fucosyl GM1, SLE, GM3, TGS5, HMWMAA, o-acetyl GD2, folate receptor ⁇ , TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, ETV
  • the tumor antigen bound by the encoded CAR molecule is selected from one or more of the following: TSHR, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, and OR51E2.
  • the target antigen of the CAR-T cell is selected from CD19, IL-13R ⁇ 2, EphA2 and EGFRvIII.
  • the target antigen of the CAR-T cell is CD19.
  • Examples of such CAR-T cells are described in Ying Zhang et al., Co-expression IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy. J Transl Med. 2022 Sep27; 20(1):432.
  • the target antigen of the CAR-T cell is IL-13R ⁇ 2.
  • Examples of such CAR-T cells are described in Chinese Patent Application No. 202210019437.4.
  • the target antigen of the CAR-T cell is EphA2.
  • Examples of such CAR-T cells are described in Chinese patent applications No. 202110919075.X and An Z, etc., Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1.Oncoimmunology.2021 Aug 16;10(1):1960728.
  • the target antigen of the CAR-T cell is EGFRvIII.
  • Examples of such CAR-T cells are described in Chinese Patent Application No. 202211140506.3.
  • the CAR molecule comprises a transmembrane domain selected from the group consisting of: ⁇ , ⁇ , or ⁇ transmembrane domains of a T cell receptor, CD28, CD3 ⁇ , CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, IL2R ⁇ , IL2R ⁇ , IL7R ⁇ , ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD , CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1
  • the extracellular target antigen binding domain of the CAR molecule is connected to the transmembrane domain by a hinge region.
  • the hinge region comprises the amino acid sequence of a CD8 hinge. In another embodiment, the hinge region comprises the amino acid sequence of an IgG4 hinge.
  • the CAR molecule comprises an intracellular segment.
  • the intracellular segment comprises a costimulatory signaling domain, and the costimulatory signaling domain comprises a functional signaling domain of a protein selected from one or more of the following: CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand specifically binding to CD83, CDS, ICA M-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8 ⁇ , CD8 ⁇ , IL2R ⁇ , IL2R ⁇ , IL7R ⁇ , ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE
  • the intracellular segment comprises a stimulatory signaling domain selected from the signaling domains of CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , common FcR ⁇ (FCER1G), FcR ⁇ (Fc ⁇ R1b), CD79a, CD79b, Fc ⁇ RIIa, DAP10 and DAP12, preferably, the stimulatory signaling domain is a CD3 ⁇ signaling domain.
  • the intracellular segment further comprises an IL15 sequence.
  • the CAR-T cell optionally contains at least one other modification in addition to the exogenously introduced CAR molecule, such as other exogenous genes introduced through the same/different vector as the CAR molecule, and/or modification of the original genomic sequence, such as through gene editing technology, etc.
  • the placental mesenchymal stem cells of the present invention enhance the anti-tumor function of CD19-IL15 CAR-T cells and IL13 CAR-T cells, but inhibit the anti-tumor function of uPAR CAR-T cells.
  • mesenchymal stem cells promote the proliferation and activation of CD19-IL15 CAR-T cells and promote the release of cytokines IL2 and IL4.
  • IL-2 is involved in shaping the transcriptional and metabolic processes that determine the fate of T cells. It is an important cytokine for T cell activation and proliferation and is considered a means of treating cancer.
  • IL-4 is generally considered a typical Th2 cytokine, it has been reported that it can promote the conversion of CD4 + T cells in human thymus and umbilical cord blood into CD8 + T cells, and promote the frequency and function of memory CD8+T cells, thereby promoting rather than weakening Th1 cell immune responses.
  • mesenchymal stem cells promote an increase in the proportion of TCM phenotype cells in CD3 + T cells, including CD4 + and CD8 + T cells.
  • mesenchymal stem cells significantly upregulate the expression of TCF-7, a transcription factor associated with T cell differentiation and involved in the differentiation and development of TCM phenotype cells.
  • CAR-T cells with less differentiated phenotypes such as TN and TCM phenotypes
  • TN and TCM phenotypes are associated with increased characteristics of self-renewal, proliferation, and survival. Therefore, the increase in such cells/CAR-T cells tend to differentiate into such cells, which will enhance the in vivo activity of CAR-T cells or cell populations.
  • tumor and cancer are not mutually exclusive and are used interchangeably herein, covering solid tumors and blood tumors, referring to all neoplastic cell growth and proliferation, whether malignant or benign, and all precancerous and cancerous cells and tissues.
  • cancers suitable for treatment by the methods of the present invention include blood cancers, for example, selected from chronic lymphocytic leukemia (CLL), acute leukemia, acute lymphocytic leukemia (ALL), B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), chronic myeloid leukemia (CML), B-cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B-cell Lymphoma, follicular lymphoma, hairy cell leukemia, small cell or large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma such as Burkitt
  • pharmaceutical composition refers to a mixture of mesenchymal stem cells as an active ingredient and a pharmaceutically acceptable carrier (as appropriate).
  • the pharmaceutical composition facilitates administration of the active ingredient to a patient.
  • the term "pharmaceutically acceptable carrier” refers to a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersant, suspending agent, diluent, excipient, thickener, solvent or encapsulating material, which is involved in carrying or transporting the useful compounds of the present invention in or to the patient's body so that it can perform its intended function.
  • a pharmaceutically acceptable material, composition or carrier such as a liquid or solid filler, stabilizer, dispersant, suspending agent, diluent, excipient, thickener, solvent or encapsulating material, which is involved in carrying or transporting the useful compounds of the present invention in or to the patient's body so that it can perform its intended function.
  • a construct can be carried to or transported from one organ or part of the body to another organ or part of the body.
  • Each carrier must be “acceptable” in the sense of being compatible with the other ingredients of the formulation (including the musk extract described in the present invention) and not harmful to
  • materials that can be used as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethylcellulose, ethylcellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerol, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffers, such as magnesium hydroxide and aluminum hydroxide; surfactants; alginic acid; pyrogen-free water; isotonic saline; Ringer
  • pharmaceutically acceptable carriers also include any and all coatings, antibacterial and antifungal agents, and absorption delaying agents that are compatible with the activity of the compounds of the invention and are physiologically acceptable to patients. Supplementary active compounds may also be incorporated into the composition. "Pharmaceutically acceptable carriers” may further include pharmaceutically acceptable salts of the compounds useful in the present invention. Other ingredients that may be included in the pharmaceutical compositions of the present invention are known in the art and are described, for example, in Remington”s Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
  • the pharmaceutical composition is prepared for intravenous injection.
  • the pharmaceutical composition includes mesenchymal stem cells prepared for intravenous injection.
  • the pharmaceutical composition includes mesenchymal stem cells and CAR-T cells prepared for intravenous injection.
  • Pharmaceutical compositions suitable for injection include sterile aqueous solutions (water-soluble) or dispersions and sterile powders for immediate preparation of sterile injectable solutions or dispersions.
  • suitable carriers include physiological saline, antibacterial water, or fluids that are easy to inject. Injectable compositions must be stable under manufacturing and storage conditions and must prevent contamination by microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or a dispersion medium, containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol and liquid polyethylene glycol, etc.), and suitable mixtures thereof.
  • appropriate fluidity can be maintained by using a coating such as lecithin, by maintaining the desired particle size in the case of a dispersion, and by using a surfactant.
  • the action of microorganisms can be prevented by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, etc.
  • isotonic agents for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition.
  • Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
  • the pharmaceutical composition is prepared for co-culturing with the mesenchymal stem cells of the present invention (e.g., preferably, placental stem cells) before the therapeutic cells therein are used for intravenous injection, so that the therapeutic effect is enhanced in vitro, and this state is continued after administration in vivo. Therefore, the pharmaceutical composition contains mesenchymal stem cells, therapeutic cells (e.g., CAR-T cells), and a culture medium suitable for co-culturing the two, and optionally, a culture container suitable for co-culturing the two.
  • the mesenchymal stem cells of the present invention e.g., preferably, placental stem cells
  • therapeutic cells e.g., CAR-T cells
  • the pharmaceutical composition is suitable for modifying cells from the subject itself (e.g., transgenic modification), and after modification, necessary expansion culture and co-culture with the mesenchymal stem cells of the present invention (e.g., preferably, placental stem cells) are performed, so that the therapeutic effect is enhanced in vitro, and this state is continued after administration in vivo. Therefore, the pharmaceutical composition contains mesenchymal stem cells, necessary reagents for modifying cells from the subject itself (e.g., transfection reagents, transgenic constructs, preferably, viral vector constructs), and suitable culture media, and optionally, a culture container suitable for co-culture of the two.
  • mesenchymal stem cells e.g., transfection reagents, transgenic constructs, preferably, viral vector constructs
  • suitable culture media e.g., a culture container suitable for co-culture of the two.
  • treat refers to slowing down, interrupting, blocking, alleviating, stopping, reducing, or reversing the progression or severity of existing symptoms, disorders, conditions, or diseases.
  • the therapeutic effects on cancer/tumors generally include, but are not limited to, for example, reduction in tumor volume, reduction in the number of cancer cells, reduction in the number of metastases, increase in life expectancy, reduction in cancer cell proliferation, reduction in cancer cell survival, or improvement in various physiological symptoms associated with cancerous disorders.
  • prevention includes inhibition of the occurrence or development of a disease or disorder or symptoms of a particular disease or disorder.
  • subjects with a family history of cancer are candidates for preventive regimens.
  • prevention refers to the administration of a drug before the signs or symptoms of cancer occur, particularly in a subject at risk for cancer.
  • “enhancement” or “promotion” of therapy means being able to enhance the therapeutic effect of existing immunotherapy (e.g., cell therapy, preferably, CAR-T cell therapy), wherein existing immunotherapy can refer to any therapy already used or known in the art, preferably a therapy for cancer, more preferably CAR-T cell therapy.
  • existing immunotherapy e.g., cell therapy, preferably, CAR-T cell therapy
  • existing immunotherapy can refer to any therapy already used or known in the art, preferably a therapy for cancer, more preferably CAR-T cell therapy.
  • Enhanced efficacy means that compared to patients treated with existing therapies that have not been enhanced by the mesenchymal stem cells of the present invention, patients who have received existing therapies enhanced by the mesenchymal stem cells of the present invention have slowed down, interrupted, blocked, relieved, stopped, reduced, or reversed the progression or severity of existing symptoms, illnesses, conditions, or diseases, or have fewer side effects, better treatment experience, and higher quality of life, or in short, have achieved or substantially achieved a better treatment process or outcome than patients who have received existing therapies that have not been enhanced by the mesenchymal stem cells of the present invention.
  • mesenchymal stem cells promote the secretion of cytokines by the therapeutic cells (e.g., preferably, CAR-T cells) to kill tumor cells.
  • mesenchymal stem cells enhance the direct killing effect of therapeutic cells (e.g., preferably, CAR-T cells) on target cells.
  • mesenchymal stem cells promote the proliferation and expansion of therapeutic cells (e.g., preferably, CAR-T cells), that is, the number increases.
  • mesenchymal stem cells promote the persistence of therapeutic cells (e.g., preferably, CAR-T cells), that is, the time of survival/existence in the body is prolonged.
  • mesenchymal stem cells enhance the ability of therapeutic cells (e.g., preferably, CAR-T cells) to reach the tumor area.
  • the efficacy enhancer promotes therapeutic cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more therapeutic cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increase the number of therapeutic cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier).
  • the enhancement is achieved by increasing the persistence of immune effector cells and/or maintaining them in a poorly differentiated state and/or more differentiation into memory effector cell subtypes.
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • cytokines e.g., interferon gamma (IFN- ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), IL-4, IL-6, IL-10, and/or IL-17A
  • IFN- ⁇ interferon gamma
  • TNF- ⁇ tumor necrosis factor alpha
  • IL-4 tumor necrosis factor alpha
  • IL-6 tumor necrosis factor alpha
  • IL-17A IL-17A
  • immune effector cells e.g., CAR-T cells
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • immune effector cells e.g., CAR-T cells
  • mesenchymal stem cells enhance cell therapy by at least one or more of the following:
  • the ratio of the number of therapeutic cells to mesenchymal stem cells used in cell therapy is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
  • the present invention relates to the use of mesenchymal stem cells for enhancing the efficacy of immunotherapy.
  • immunotherapy is cell therapy, that is, a therapy that administers therapeutic cells.
  • therapeutic cells are nonspecific immune effector cells.
  • therapeutic cells are specific immune effector cells, preferably, therapeutic cells are T cells, more preferably, therapeutic cells are modified T cells, and most preferably, therapeutic cells are CAR-T cells.
  • human T cells are CD8+T cells.
  • therapeutic cells are CAR-NK, TCR-T, TIL, CAR-DC, etc. cells.
  • the present invention provides a method comprising administering immune effector cells to a subject in need, wherein the immune effector cells are optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body.
  • the immune effector cells are CAR-T cells.
  • the subject has a condition described herein, for example, the subject suffers from cancer, for example, the subject has a cancer expressing a target antigen described herein.
  • the subject is a human.
  • the present invention relates to a method for treating a subject with a disease associated with the expression of a cancer-associated antigen as described herein, comprising administering an immune effector cell to a subject in need thereof, the immune effector cell being optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cell is a CAR-T cell.
  • the present invention provides a method for treating a subject suffering from a disease associated with the expression of a tumor antigen, comprising administering to a subject in need an immune effector cell, the immune effector cell being optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence for the CAR molecule, and the cell having been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cell is a CAR-T cell.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells to the body.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vitro/ex vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells to the body.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the above method increases the persistence of immune effector cells after administration in a subject.
  • the present invention also provides a method for regulating the differentiation of immune effector cells in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the above method keeps the immune effector cells in a low differentiated state after being administered to the subject.
  • the present invention also provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo.
  • the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo.
  • the immune effector cells are modified.
  • the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
  • the above method allows immune effector cells to differentiate into more memory effector cell subtypes after being administered to a subject.
  • the present invention provides a composition comprising immune effector cells and mesenchymal stem cells, for treating a subject with a disease associated with the expression of a tumor antigen such as a disease as described herein, wherein the immune effector cell comprises a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body.
  • the immune effector cell is a CAR-T cell.
  • the mesenchymal stem cells can be delivered in combination with the immune effector cells, administered before the immune effector cells are administered, administered simultaneously with the immune effector cells, and administered after the immune effector cells are administered.
  • the immune effector cells are co-cultured in vitro/ex vivo with the mesenchymal stem cells before being administered to the subject, and after co-culture, the mesenchymal stem cells are delivered in combination with/not with the immune effector cells.
  • mesenchymal stem cells can be administered after an extended period of time after the immune effector cells are administered.
  • mesenchymal stem cells and immune effector cells or cell groups are administered to the subject at the same time (e.g., administered on the same day) or immune effector cells or cell groups are administered (e.g., 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 10 weeks or more) after administration.
  • therapeutic agents/treatment methods known in the art may be further combined, such as, but not limited to, immune checkpoint inhibitors, targeted drugs, chemotherapy drugs, surgical treatment, radiotherapy, other CAR-T enhancers, and the like.
  • therapeutic agents/treatment methods known in the art may be further combined, such as, but not limited to, immune checkpoint inhibitors, targeted drugs, chemotherapy drugs, surgical treatment, radiotherapy, other CAR-T enhancers, and the like.
  • the killing efficacy of the immune effector cells to the target tumor is enhanced relative to the same immune effector cells that are not co-cultured with mesenchymal stem cells and not delivered in combination.
  • the killing efficacy is enhanced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 120%, 140%, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, 9 times, 10 times, 100 times, 500 times, 1000 times or higher, as evaluated by any index known in the art for assessing disease improvement and/or tumor regression.
  • the disease associated with a tumor antigen is selected from a proliferative disease, such as a cancer or a malignant tumor or a precancerous lesion, such as myelodysplasia, myelodysplastic syndrome or preleukemia, or a non-cancer-related indication associated with the expression of a tumor antigen as described herein.
  • a proliferative disease such as a cancer or a malignant tumor or a precancerous lesion, such as myelodysplasia, myelodysplastic syndrome or preleukemia, or a non-cancer-related indication associated with the expression of a tumor antigen as described herein.
  • the disease is a cancer as described herein above.
  • the tumor antigen is a tumor antigen as described herein above.
  • the present invention provides a method comprising administering a modified immune effector cell to a subject in need, the cell comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body.
  • the immune effector cell is a CAR-T cell.
  • the subject has a condition described herein, for example, the subject suffers from cancer, for example, the subject has a cancer expressing a target antigen described herein.
  • the subject is a human.
  • co-culture/co-administration with the mesenchymal stem cells increases the therapeutic efficacy of the immune effector cells.
  • the present invention provides a pharmaceutical composition for enhancing the efficacy of cell therapy, wherein the pharmaceutical composition comprises mesenchymal stem cells.
  • the cell therapy administers therapeutic cells to subjects in need, and the therapeutic cells are nonspecific immune effector cells.
  • the immune effector cells comprise a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and more preferably the immune effector cells are CAR-T cells.
  • the present invention provides a pharmaceutical composition for treating a subject suffering from a disease such as a disease described herein associated with the expression of a tumor antigen, wherein the pharmaceutical composition comprises immune effector cells and mesenchymal stem cells, and the immune effector cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo.
  • the immune effector cells comprise a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and more preferably the immune effector cells are CAR-T cells.
  • the pharmaceutical composition may further comprise one or more pharmaceutically acceptable or physiologically acceptable carriers, diluents or excipients, which make the pharmaceutical composition suitable for administration via the intended route, such as, but not limited to, intravenous injection or local tumor injection, etc.
  • the present invention provides use of mesenchymal stem cells in preparing an agent for enhancing the therapeutic effect of cell therapy.
  • Placental tissue was processed according to the method of Papait A et al. (Mesenchymal Stromal Cells from Fetal and Maternal Placenta Possess Key Similarities and Differences: Potential Implications for Their Applications in Regenerative Medicine. Cells. 2020 Jan 6; 9(1): 127) with slight modifications.
  • the placental tissue was soaked in a preheated phosphate-buffered saline (PBS) solution containing 10% penicillin and streptomycin for 10 minutes, then washed twice with PBS and cut into 1 mm 3 with scissors. Mononuclear cells were obtained through a 100 ⁇ m sterile cell filter, and lymphocytes were separated by density gradient centrifugation.
  • PBS phosphate-buffered saline
  • Cells were cultured at a density of 1 ⁇ 10 6 cells/cm 2 in DMEM medium containing 20% FBS, 2mM L-glutamine and 100U/ml penicillin/streptomycin at 37°C in a humidified atmosphere containing 5% (v/v) CO 2. After about 6 to 8 days, many colonies had formed. Unattached cells were removed and fresh medium was added. After the confluence reached 85%, the adherent cells were digested with 0.25% (w/v) trypsin/EDTA and re-plated at a cell density of 5 ⁇ 10 3 cells/cm 2. Placental cells were cultured continuously under the same conditions and expanded for 2 to 6 passages (Passage, P). All experiments were performed in a sterile environment with cells harvested between passages 3 and 6 (i.e., P3 and P6) and performed in a biosafety cabinet whenever possible.
  • the characterization of pMSCs was performed according to the guidelines proposed by the International Society for Cellular Therapy (ISCT) (see: Viswanathan S et al., Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy. 2019 Oct; 21(10): 1019-1024.).
  • the immunophenotype of cultured pMSCs was evaluated by flow cytometry. A total of 1 ⁇ 10 6 cells at passage 3 were incubated at room temperature in the dark for 15 minutes. Data acquisition and analysis were performed using a FACSCanto-II flow cytometer and flowjo v.10 software (BD Biosciences).
  • the differentiation induction ability of pMSCs was evaluated by culturing in a specific differentiation induction medium for 3-4 weeks.
  • the induction and detection of osteogenic differentiation were evaluated according to the following references: Jaiswal N et al., Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro. J Cell Biochem 1997; 64: 295-312. and Xu X et al., Dysregulated systemic lymphocytes affect the balance of osteogenic/adipogenic differentiation of bone mesenchymal stem cells after local irradiation. Stem Cell Res Ther 2017; 8: 71.
  • mRNA level expression of specific transcription factors related to the induction of osteoblast formation was further quantitatively evaluated.
  • Total mRNA was extracted from differentiated mesenchymal stem cell cultures and controls using Trizol, and quantitative RT-PCR (qRT-PCR) analysis was performed on a Roche Light Cycler 480II using SYBR Green PCR Master Mix (Fisher Scientific SL) and primers. The reactions were performed in triplicate, and the gene expression values were normalized to the expression values of GAPDH.
  • NALM-6 and Raji cell lines engineered to express GFP and firefly luciferase (Luc) by retroviral transduction were named NALM-6-GL and Raji-GL, respectively.
  • FBS fetal bovine serum
  • retroviral producer cell lines were cultured in 1640 medium without penicillin and streptomycin containing 10% FBS.
  • CAR-T cells targeting the blood tumor antigen CD19 are used in embodiments of the present invention, including CAR-T cells targeting the blood tumor antigen CD19, and CAR-T cells targeting two solid tumor antigens uPAR and IL13R ⁇ 2, respectively.
  • CAR is transferred into T cells via a retroviral vector.
  • Examples of CAR-T cells targeting the blood tumor antigen CD19 are described in Ying Zhang et al., Co-expression of IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy. J Transl Med. 2022 Sep 27; 20(1):432.
  • Examples of CAR-T cells targeting two solid tumor antigens uPAR and IL13R ⁇ 2 are described in Amor, C.
  • CAR-T cells can refer to, for example, the previous paper (An Z et al. Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1. Oncoimmunology. 2021 Aug 16; 10(1): 1960728).
  • the structure of the CAR consists of three parts, namely, the extracellular domain containing the anti-CD19 scFV sequence, the transmembrane domain of CD28, and the intracellular domain of CD28, 4-1BB, CD3 ⁇ and IL15 sequences connected by P2A.
  • PBMCs Human peripheral blood mononuclear cells from healthy donors were isolated by gradient centrifugation using lymphocyte separation medium (MP Biomedicals). T cells in PBMCs were stimulated with anti-CD3/CD28 T cell activation Dynabeads (Invitrogen). After 48 h of bead activation, T cells were transduced with retroviral supernatant by centrifugation on Retronectin (Takara)-coated plates. T cells were immunoblotted for CAR expression on day 7. T cells were cultured in X-VIVO-15 medium containing 5% human AB serum (SIGMA), 100 U/ml IL-2, 100 U/mL penicillin, and 100 ⁇ g/mL streptomycin (EallBio Life Sciences).
  • SIGMA human AB serum
  • Flow cytometry was performed using a BD FacsCanto II Plus instrument (BD Biosciences) and analyzed using FlowJo v.10 software (Treestar, Inc. Ashland, OR). T cells were detected using APC-conjugated anti-human CD3 antibodies (BD Biosciences), V450-conjugated anti-human CD4 (BD Biosciences), PE-Cy7-conjugated anti-human CD8 (BD Biosciences), and PE-Cy7-conjugated anti-human CCR7 (BD Biosciences).
  • PE-Cy5-conjugated anti-human CD95 (BD Biosciences), Alexa Fluor 700-conjugated anti-human CD27 (BD Biosciences), and goat anti-mouse IgG (Fab specific) F(ab')2 fragment antibodies (Sigma).
  • CAR was detected by staining with FITC-labeled goat anti-mouse IgG (H+L) antibodies (Sigma).
  • MSC cells were stained with PE-Cy7-conjugated anti-human CD105 (ThermoFisher Scientific) and V450-conjugated anti-human CD45 (BD Biosciences).
  • T cell degranulation was detected by BV421-conjugated anti-human CD107a-APC (ThermoFisher Scientific). After incubation with antibodies, cells were washed with PBS, resuspended in PBS containing 1% FBS, and analyzed on an analyzer.
  • CAR-T cells were co-cultured with MSC cells at a ratio of 10:1 for 24 hours, and then CAR-T cells were collected and co-cultured with NALM-6 cells at a ratio of 1:1 in a 24-well plate, where each well contained 1 ⁇ l of anti-human CD107a antibody (BD Bioscience) and Golgi Stop TM (BD Bioscience). After 6 hours, the cells were harvested and incubated with anti-human CD3 antibody (BD Bioscience), and the CD107a degranulation of CAR-T cells was detected by flow cytometry analysis. CD19-IL15 CAR-T cells were used as an example.
  • the experiment was divided into four groups: (1) CAR-T cells that were not co-cultured with MSCs in advance as a control; (2) CAR-T cells that were co-cultured with MSCs in advance; (3) CAR-T cells that were co-cultured with NALM-6 cells; (4) CAR-T cells that were first co-cultured with MSCs and then collected and co-cultured with NALM-6 cells.
  • CAR-T cells were prepared using cells from three healthy donors, co-cultured with the NALM-6 cell line, and T cell proliferation was analyzed using flow cytometry based on carboxyfluorescein succinimidyl ester (CFSE) dye.
  • CFSE-labeled CAR-T cells were first co-cultured with MSC cells in a round-bottom 24-well plate at a density of 1 ⁇ 10 6 :1 ⁇ 10 5 cells/ml (i.e., a ratio of 10:1) for 24 hours in triplicate. CAR-T cells were then collected and co-cultured with NALM-6 at a ratio of 10:1 for 3 days.
  • T cell proliferation was determined by CFSE dye dilution of CD3-positive cells using a BD FacsCanto II Plus instrument (BD Biosciences). Flow cytometric data were analyzed using Flow Jo v.10 software (Tree star, Inc. Ashland, OR). The groups in this experiment were the same as those in the CD107a degranulation experiment.
  • CAR-T cells Two methods were used to detect the cytotoxic function of CAR-T cells. One was to detect the GFP signal of target cells by flow cytometry, and the other was to detect the chemiluminescence of target cells using a live imaging analysis system. CAR-T cells were first co-cultured with MSC cells at a ratio of 10:1 for 24 hours, then re-collected and co-cultured with NALM-6-GL and Raji-GL cell lines at various E:T ratios (1:1 and 0.5:1) in 24-well plates. After 24 hours, cells were collected and target cells were detected using a BD FacsCanto II Plus instrument (BD Biosciences), and data were analyzed using Flow Jo v.10 software (Treestar, Inc. Ashland, OR).
  • chemiluminescence method cells (96-well plates) were added with substrates, and the signals were collected using a PerkinElmer photochemical imaging system. The intensity of the fluorescence signal was analyzed using a live imaging analysis system to evaluate the cytotoxicity of CAR-T cells.
  • the experiment included three groups: (1) target cells only; (2) CAR-T cells co-cultured with target cells without prior co-culture with MSCs; and (3) CAR-T cells co-cultured with MSCs beforehand and then collected for co-culture with target cells.
  • the expression of human interferon gamma (IFN- ⁇ ), tumor necrosis factor alpha (TNF- ⁇ ), IL-4, IL-6, IL-10, and IL-17A in the supernatant of co-cultured cells was evaluated using a commercial cytometric bead array (CBA) kit (BD Biosciences) according to the manufacturer's procedures. The level of human IFN- ⁇ was also measured by using The ELISA kit (R&D, Minnesota, USA) was used for evaluation according to the manufacturer's instructions.
  • CBA cytometric bead array
  • CAR-T cells were cultured in medium without IL2 for 3 days and then co-cultured with mesenchymal stem cells and target cells in the same medium without IL2. The supernatant was collected and used The release of IL2 was detected by ELISA kit (R&D, Minnesota, USA).
  • CAR-T cells were co-cultured with MSC cells at a ratio of 10:1 for 5 days, then re-collected and co-cultured with NALM-6-GL and Raji-GL cell lines at an E:T ratio of 10:1 in 24-well plates for 24 hours. The co-cultured cells were then collected for flow cytometry analysis of T cell subsets.
  • NOD-SCID mice Female NOD-SCID mice aged six to eight weeks were purchased from Charles River Laboratories and carefully raised under pathogen-free conditions. After one week of adaptive feeding, NOD-SCID mice were injected with 1 ⁇ 10 6 NALM-6-GL cells via the tail vein to construct a tumor xenograft mouse model. A total of 12 mice were randomly divided into three groups.
  • mice were euthanized after reaching the euthanasia criteria.
  • mesenchymal stem cells from the placentas of eight normal donors and determined whether the cells were mesenchymal stem cells by identifying their morphology, immunophenotype, and differentiation potential, referring to the standardized criteria listed by ISCT.
  • Adherent cells were successfully cultured from all samples using standard culture conditions and methods for mesenchymal stem cells. These cells adhered to the plastic and showed a uniform spindle-forming fibroblast morphology ( Figure 1).
  • Figure 1 We calculated the growth dynamics from P1 to P6 by trypan blue staining and found that the proliferation of mesenchymal stem cells from different donor placentas was basically the same (Figure 2).
  • the three CAR molecules described above were constructed according to the method described in Example 1 and transduced into T cells to obtain CAR-T cells, which were verified by flow cytometry.
  • CAR-T cells were also co-cultured with MSCs at a ratio of 10:1 for 24 hours in advance, and then co-cultured with/without NALM-6 at a ratio of 1:1 for 6 hours.
  • CAR-T cells that were not co-cultured with MSCs and co-cultured with/without NALM-6 served as controls.
  • CD19-IL15 CAR-T cells attacked NALM-6 cells
  • the secretion of IL4 and IL10 in CAR-T cells co-cultured with MSCs increased significantly, while the secretion of IFN- ⁇ , IL17A, and TNF- ⁇ did not change significantly.
  • CD19-IL15 CAR-T cells attacked Raji cells
  • the secretion of IL4 and IL17A in CAR-T cells co-cultured with mesenchymal stem cells was significantly increased, while the secretion of IFN- ⁇ , IL17A and IL10 did not change significantly.
  • CAR-T cells were co-cultured with MSCs at a ratio of 10:1 for 24 hours, and then CAR-T cells were collected and co-cultured with NALM-6-GL and Raji-GL cells at various E:T ratios.
  • the survival of tumor cells was analyzed by flow cytometry to detect GFP fluorescence (see Figures 11 and 12, respectively) and luciferase activity by IVIS imaging system (see Figure 13, A and B, respectively) to evaluate cytotoxic function.
  • the killing ability of CD19-IL15 CAR-T cells was significantly enhanced after co-culture with mesenchymal stem cells.
  • Example 5 Mesenchymal stem cells enhance the function of CAR-T cells targeting IL13R ⁇ 2 in vitro
  • Example 6 Mesenchymal stem cells inhibited the function of uPAR-targeted CAR-T cells in vitro
  • MSCs have different functions on different CAR-T cells. MSCs caused a decrease in the killing ability of uPAR-targeted CAR-T cells.
  • MSCs caused a decrease in the killing ability of uPAR-targeted CAR-T cells.
  • MSCs enhanced the activation of CD107a ( Figure 17) and promoted the release of cytokine IFN- ⁇ ( Figure 18), but severely hindered its cytotoxic function ( Figures 19-20).
  • E:T ratio was 2:1, MSCs reduced the killing ability of CAR-T cells by 22%.
  • mesenchymal stem cells regulate the function of CAR-T cells and used CD19-IL15 CAR-T cells as the research object to find the possible mechanism by which mesenchymal stem cells promote CAR-T function.
  • mesenchymal stem cells affected the differentiation of CAR-T cells.
  • CAR-T cells were co-cultured with MSC for one day and collected with or without co-culture with NALM-6 cells.
  • CAR-T cells were then collected to detect T cell subsets.
  • Figures 22-23 compared with CD19-IL15 CAR-T cells, the proportion of TCM phenotype in CD3+ cells was significantly increased in CAR-T cells co-cultured with MSC.
  • the IL2 released by MSC-treated CAR-T cells did not change significantly, but when co-cultured with tumor cells such as NALM-6 and Raji, the IL2 secreted by MSC-treated CAR-T cells increased significantly.
  • Example 8 Mesenchymal stem cells enhance the anti-tumor ability of CD19-IL15 CAR-T in vivo
  • a xenograft mouse model was constructed by tail vein injection of NALM-6-eGFP-Luc (NALM-6-GL) cells. As shown in FIG26 , 1 ⁇ 10 6 NALM-6-GL cells were injected one day in advance, and 1 ⁇ 10 7 CD19-IL15 CAR-T cells were injected through the tail vein on day 0.
  • the CAR-T cells injected into the experimental group mice were co-cultured with mesenchymal stem cells in vitro for 1 day in advance, while the CAR-T cells injected into the control group mice were not co-cultured with MSCs. In addition, a non-treatment control group without CAR-T injection was set up. Fluorescence imaging of mice was performed regularly.
  • the tumor burden in the CAR-T treatment group was significantly lower than that in the control group, but there was no significant difference between the CAR-T group treated with mesenchymal stem cells and the CAR-T group alone.
  • the survival of the mice was observed and counted. The results showed that after co-culture of CAR-T cells with mesenchymal stem cells, the survival time of mice was significantly prolonged ( FIG29 ).
  • placental mesenchymal stem cells enhanced the anti-tumor function of CD19-IL15 CAR-T cells and IL13 CAR-T cells, but inhibited the anti-tumor function of uPAR CAR-T cells.
  • mesenchymal stem cells promoted the proliferation and activation of CD19-IL15 CAR-T cells and promoted the release of cytokines IL2 and IL4.
  • IL-2 is involved in shaping the transcriptional and metabolic processes that determine the fate of T cells. It is an important cytokine for T cell activation and proliferation and is considered a means of treating cancer.
  • IL-4 is generally considered to be a typical Th2 cytokine, but it has been reported that it can promote the conversion of CD4+T cells in human thymus and umbilical cord blood into CD8+T cells and promote the number and function of memory CD8+T cells, thereby promoting rather than weakening Th1 cell immune responses.
  • mesenchymal stem cells promoted an increase in the proportion of TCM phenotype cells in CD3+T cells, including CD4+ and CD8+T cells.
  • TCF-7 a transcriptional activator involved in the differentiation and development of TCM phenotype cells, was significantly increased in expression. It is well known that CAR-T cells with less differentiated phenotypes, such as TN and TCM phenotypes, are associated with increased properties of self-renewal, proliferation, and survival.
  • CAR-T cell therapy is closely related to the activity and persistence of CAR-T cells. Therefore, all the above results well explain why mesenchymal stem cells can enhance the anti-tumor ability of CD19-IL15 CAR-T cells.
  • This paper also evaluated whether mesenchymal stem cells can promote the anti-tumor efficacy of CD19-IL15 CAR-T cells in a xenograft mouse model.
  • the results showed that CAR-T cells co-cultured with mesenchymal stem cells in vitro significantly prolonged the survival time of NALM-6-GL tumor-bearing mice.
  • BM-MSCs were co-cultured with CD19CAR-T and tumor cells, and it was found that MSCs could not protect tumor cells or affect the anti-tumor ability of CAR-T cells (Zanetti SR et al., Bone marrow MSCs from pediatric patients with B-ALL highly immunosuppress T-cell responses but do not compromise CD19-CAR T-cell activity.
  • CD19-IL15 CAR-T cells were first stimulated by MSCs and then collected and co-cultured with tumor cells, and the results showed that their anti-tumor ability was significantly enhanced.
  • CAR-T cells undergo a series of changes under MSC stimulation, such as phenotype, cytokine release, activation and proliferation ability, leading to the enhancement of their anti-tumor function.
  • mesenchymal stem cells inhibited the function of uPAR CAR-T cells.
  • Previous flow cytometry results showed that MSCs expressed uPAR but did not express CD19 and IL13R ⁇ 2. Therefore, it is speculated that CD19-IL15 CAR-T cells and IL13R ⁇ 2 CAR-T cells did not kill mesenchymal stem cells when co-cultured with mesenchymal stem cells (data not shown), but uPAR CAR-T cells can attack mesenchymal stem cells through their target antigen specificity. As a result, uPAR CAR-T cells may be exhausted due to killing MSCs, resulting in a decrease in their anti-tumor ability. This phenomenon shows that whether mesenchymal stem cells have an enhancing effect on the function of CAR-T cells is a key factor in whether mesenchymal stem cells express the corresponding target antigen.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Organic Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Biomedical Technology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Immunology (AREA)
  • Biotechnology (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Hematology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Microbiology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Biochemistry (AREA)
  • Oncology (AREA)
  • Mycology (AREA)
  • Virology (AREA)
  • Medicines Containing Material From Animals Or Micro-Organisms (AREA)

Abstract

Provided are a method of a mesenchymal stem cell, e.g., a placenta-derived mesenchymal stem cell, for enhancing the treatment of a tumor by a CAR-T cell and/or treating a tumor in combination with a CAR-T cell, corresponding use, and a corresponding composition. The mesenchymal stem cell enhances the proliferation ability and persistence of a CAR-T cell in vivo, and regulates and controls the differentiation of the CAR-T cell, so as to allow the CAR-T cell to be in a low-differentiation state and increase the differentiation of the CAR-T cell into a memory cell subtype.

Description

间充质干细胞的用途Uses of Mesenchymal Stem Cells 技术领域Technical Field
本发明涉及细胞治疗领域。具体地,本发明涉及间充质干细胞用于增强免疫疗法的用途和相应的药物组合物。The present invention relates to the field of cell therapy, and in particular to the use of mesenchymal stem cells for enhancing immunotherapy and corresponding pharmaceutical compositions.
背景技术Background technique
嵌合抗原受体(Chimeric antigen receptor,CAR)是一种人工合成的分子,其通过特异性识别肿瘤细胞表面表达的抗原来引导经基因工程化以表达CAR的免疫效应细胞(例如,T细胞、NK细胞)清除肿瘤(参见Sampson JH等,EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebralglioma and generates host immunity against tumor-antigen loss.Clinical cancer research:an official journal of the American Association for Cancer Research.2014;20(4):972-984)。例如,嵌合抗原受体T细胞(CAR-T)是通过T细胞上的嵌合抗原受体(CAR)分子直接靶向肿瘤细胞的表面抗原,从而达到识别和杀伤肿瘤的目的,其中所述嵌合抗原受体的N端包含识别抗原的胞外域。当所述CAR-T细胞针对的抗原阳性细胞存在时,CAR-T细胞能识别并杀伤这些抗原阳性细胞。CAR-T细胞在治疗恶性肿瘤方面具有明显的优势。越来越多的报道证明,CAR-T细胞在治疗难治性血液病肿瘤方面非常有效(参见Sadelain M等,Therapeutic T cell engineering.Nature.2017 May 24;545(7655):423-431.;June CH和Sadelain M.Chimeric Antigen Receptor Therapy.N Engl J Med.2018 Jul 5;379(1):64-73.;Brudno JN和Kochenderfer JN.Chimeric antigen receptor T-cell therapies for lymphoma.Nat Rev Clin Oncol.2018 Jan;15(1):31-46.;Brudno JN等,T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma.J Clin Oncol.2018 Aug 1;36(22):2267-2280.)。虽然完全缓解率很高,但大多数病人仍会复发;其中有些病人是抗原阴性,有些是抗原低下(参见:Sadelain M等,Therapeutic T cell engineering.Nature.2017 May 24;545(7655):423-431.;June CH和Sadelain M.Chimeric Antigen Receptor Therapy.N Engl J Med.2018 Jul 5;379(1):64-73.;Brudno JN等,T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma.J Clin Oncol.2018 Aug 1;36(22):2267-2280.;Orlando EJ等,Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia.Nat Med.2018 Oct;24(10):1504-1506.;Sotillo E等,Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy.Cancer Discov.2015Dec;5(12):1282-95.;Gardner R等,Acquisition of a CD19-negative myeloid phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy.Blood.2016 May 19;127(20):2406-10.;Orlando EJ等,Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia.Nat Med.2018 Oct;24(10):1504-1506.)。特别是在治疗实体瘤方面,疗效甚微(参见:An Z等,Antitumor activity of the third generation EphA2  CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1.Oncoimmunology.2021 Aug 16;10(1):1960728.;Xu C等,IL-13Rα2humanized scFv-based CAR-T cells exhibit therapeutic activity against glioblastoma.Mol Ther Oncolytics.2022 Jan 10;24:443-451.),仍需很长的路要走。Chimeric antigen receptor (CAR) is a synthetic molecule that specifically recognizes antigens expressed on the surface of tumor cells to guide immune effector cells (e.g., T cells, NK cells) genetically engineered to express CAR to eliminate tumors (see Sampson JH et al., EGFRvIII mCAR-modified T-cell therapy cures mice with established intracerebral glioma and generates host immunity against tumor-antigen loss. Clinical cancer research: an official journal of the American Association for Cancer Research. 2014; 20(4): 972-984). For example, chimeric antigen receptor T cells (CAR-T) directly target surface antigens of tumor cells through chimeric antigen receptor (CAR) molecules on T cells, thereby achieving the purpose of recognizing and killing tumors, wherein the N-terminus of the chimeric antigen receptor contains an extracellular domain that recognizes the antigen. When the antigen-positive cells targeted by the CAR-T cells exist, the CAR-T cells can recognize and kill these antigen-positive cells. CAR-T cells have obvious advantages in the treatment of malignant tumors. More and more reports have shown that CAR-T cells are very effective in treating refractory hematological tumors (see Sadelain M et al., Therapeutic T cell engineering. Nature. 2017 May 24; 545(7655): 423-431.; June CH and Sadelain M. Chimeric Antigen Receptor Therapy. N Engl J Med. 2018 Jul 5; 379(1): 64-73.; Brudno JN and Kochenderfer JN. Chimeric antigen receptor T-cell thera pies for lymphoma. Nat Rev Clin Oncol. 2018 Jan; 15(1):31-46.; Brudno JN et al., T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Causes Remissions of Poor-Prognosis Relapsed Multiple Myeloma. J Clin Oncol. 2018 Aug 1; 36(22):2267-2280.). Although the complete remission rate is high, most patients will still relapse; some of these patients are antigen-negative and some are antigen-low (see: Sadelain M et al., Therapeutic T cell engineering. Nature. 2017 May 24; 545(7655): 423-431.; June CH and Sadelain M. Chimeric Antigen Receptor Therapy. N Engl J Med. 2018 Jul 5; 379(1): 64-73.; Brudno JN et al., T Cells Genetically Modified to Express an Anti-B-Cell Maturation Antigen Chimeric Antigen Receptor Cause Remissions of Poor-Prognosis Relapsed Multiple Myeloma.J Clin Oncol.2018 Aug 1;36(22):2267-2280.;Orlando EJ et al., Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphob Lastic leukemia.Nat Med.2018 Oct;24(10):1504-1506.;Sotillo E et al.,Convergence of Acquired Mutations and Alternative Splicing of CD19 Enables Resistance to CART-19 Immunotherapy.Cancer Discov.2015Dec;5(12):1282-95.;Gardner R et al.,Acquisition of a CD19-negative myeloid Phenotype allows immune escape of MLL-rearranged B-ALL from CD19 CAR-T-cell therapy. Blood. 2016 May 19;127(20):2406-10.; Orlando EJ, et al., Genetic mechanisms of target antigen loss in CAR19 therapy of acute lymphoblastic leukemia. Nat Med. 2018 Oct;24(10):1504-1506.). Especially in the treatment of solid tumors, the efficacy is very limited (see: An Z et al., Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1. Oncoimmunology. 2021 Aug 16; 10(1): 1960728.; Xu C et al., IL-13Rα2humanized scFv-based CAR-T cells exhibit therapeutic activity against glioblastoma. Mol Ther Oncolytics. 2022 Jan 10; 24: 443-451.), and there is still a long way to go.
如果CAR-T疗法的结果不达预期,可能有一个最重要的原因是相关CAR-T细胞的扩增性差,且持久性有限。CAR-T细胞的体内扩增能力和持久性与CAR分子信号强度、细胞因子选择、刺激域和T细胞的表型等等都有关。If the results of CAR-T therapy do not meet expectations, one of the most important reasons may be that the related CAR-T cells have poor expansion and limited persistence. The in vivo expansion ability and persistence of CAR-T cells are related to the strength of CAR molecular signals, cytokine selection, stimulation domain, and T cell phenotype.
此外,临床前和临床研究表明,CAR-T细胞的功能也受到CAR-T细胞的分化的影响。灌注具有天真样(TN)和中央记忆(TCM)表型的CAR-T细胞与T细胞在体内的持久性和优越的抗肿瘤效果有关(参见:Wilkie S等,Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4.J Biol Chem.2010 Aug13;285(33):25538-44.;Shum T等,Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells.Cancer Discov.2017 Nov;7(11):1238-1247.;Sukumaran S等,Enhancing the Potency and Specificity of Engineered T Cells for Cancer Treatment.Cancer Discov.2018 Aug;8(8):972-987.;Liu X等,A Chimeric Switch-Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors.Cancer Res.2016 Mar 15;76(6):1578-90.;Torres Chavez A等,Expanding CAR T cells in human platelet lysate renders T cells with in vivo longevity.J Immunother Cancer.2019 Nov 28;7(1):330.)。因此,调控CAR-T细胞的分化,使其保持在低分化状态,将大大增强其抗肿瘤效果。In addition, preclinical and clinical studies have shown that the function of CAR-T cells is also affected by the differentiation of CAR-T cells. Infusion of CAR-T cells with naive-like (TN) and central memory (TCM) phenotypes is associated with T cell persistence in vivo and superior antitumor efficacy (see: Wilkie S et al., Selective expansion of chimeric antigen receptor-targeted T-cells with potent effector function using interleukin-4. J Biol Chem. 2010 Aug 13; 285(33): 25538-44.; Shum T et al., Constitutive Signaling from an Engineered IL7 Receptor Promotes Durable Tumor Elimination by Tumor-Redirected T Cells. Cancer Discov. 2017 Nov; 7(11): 1238-1247.; Sukumaran S et al., Enhancing the Potency and Specificity of Engineered T Cells for Cancer Treatment.Cancer Discov.2018 Aug ;8(8):972-987.;Liu X et al., A Chimeric Switch Receptor Targeting PD1 Augments the Efficacy of Second-Generation CAR T Cells in Advanced Solid Tumors.Cancer Res.2016 Mar 15;76(6):1578-90.;Torres Chavez A et al., Expanding CAR T cells in human platelet lysate renders T cells with in vivo longevity.J Immunother Cancer.2019 Nov 28;7(1):330.). Therefore, regulating the differentiation of CAR-T cells and keeping them in a low-differentiation state will greatly enhance their anti-tumor effect.
间充质干细胞(mesenchymal stem cell,MSC)是一种具有多潜能的基质细胞,在多种组织例如骨髓、脂肪、外周血、脐带、胎盘和其他组织中都有发现。它是临床上最有希望的异体细胞治疗方法。目前,来自骨髓和胎盘的间充质干细胞是本领域最感兴趣的。胎盘中的干细胞相对原始,免疫排斥性低,增殖和分化能力强,易于收集和分离,更重要的是它并不伴随伦理障碍。Mesenchymal stem cells (MSC) are multipotent stromal cells found in a variety of tissues such as bone marrow, fat, peripheral blood, umbilical cord, placenta and other tissues. It is the most promising allogeneic cell therapy in the clinic. Currently, mesenchymal stem cells from bone marrow and placenta are of the greatest interest in this field. Stem cells in the placenta are relatively primitive, have low immune rejection, strong proliferation and differentiation capabilities, are easy to collect and separate, and more importantly, are not accompanied by ethical barriers.
间充质干细胞在体外具有固有的免疫调节特性、营养能力、高自我更新能力和强分化潜力。间充质干细胞通过与免疫细胞的直接接触和旁分泌活动影响大多数免疫效应细胞的功能,并且可以很容易地被设计来增强其免疫调节功能(参见:Song N等,Mesenchymal Stem Cell Immunomodulation:Mechanisms and Therapeutic Potential.Trends Pharmacol Sci.2020 Sep;41(9):653-664.)。有报道称,MSC通过抑制T细胞的激活和增殖来抑制T细胞的功能(参见:Ghannam S等,Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype.J Immunol.2010 Jul 1;185(1):302-12.),并发现MSC抑制naive CD4+T细胞分化为Th17细胞,并抑制IL-17、IL22、IFN-γ和TNF-α的产生(参见:Ghannam S等,Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype.J Immunol.2010 Jul 1;185(1):302-12.),诱导T调节细胞表型或T细胞过敏(参见:Glennie S等,Bone marrow mesenchymal stem  cells induce division arrest anergy of activated T cells.Blood.2005 Apr 1;105(7):2821-7.)。关于间充质干细胞对CAR-T功能的影响的报告很少。有报道称骨髓间充质干细胞抑制T细胞反应,但不影响CD19 CAR-T细胞的活性(参见:Zanetti SR等,Bone marrow MSC from pediatric patients with B-ALL highly immunosuppress T-cell responses but do not compromise CD19-CAR T-cell activity.J Immunother Cancer.2020 Aug;8(2):e001419.),另一项研究发现IL7-IL12工程间充质干细胞改善CAR-T细胞对结直肠癌细胞的攻击(参见:Hombach AA等,IL7-IL12 Engineered Mesenchymal Stem Cells(MSCs)Improve A CAR T Cell Attack Against Colorectal Cancer Cells.Cells.2020 Apr 3;9(4):873.)。目前,MSC对CAR-T功能的影响仍有争议。间充质干细胞是促进CAR-T功能还是抑制CAR-T功能需要更多的研究。Mesenchymal stem cells have inherent immunomodulatory properties, nutritional capacity, high self-renewal ability and strong differentiation potential in vitro. Mesenchymal stem cells affect the function of most immune effector cells through direct contact with immune cells and paracrine activity, and can be easily designed to enhance their immunomodulatory function (see: Song N et al., Mesenchymal Stem Cell Immunomodulation: Mechanisms and Therapeutic Potential. Trends Pharmacol Sci. 2020 Sep; 41(9): 653-664.). It has been reported that MSCs inhibit the function of T cells by inhibiting the activation and proliferation of T cells (see: Ghannam S et al., Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol. 2010 Jul 1; 185(1): 302-12.), and it was found that MSCs inhibited the differentiation of naive CD4+T cells into Th17 cells and inhibited the expression of IL-17 and IL22. , IFN-γ and TNF-α production (see: Ghannam S et al., Mesenchymal stem cells inhibit human Th17 cell differentiation and function and induce a T regulatory cell phenotype. J Immunol. 2010 Jul 1; 185(1): 302-12.), induce T regulatory cell phenotype or T cell allergy (see: Glennie S et al., Bone marrow mesenchymal stem cells induce division arrest anergy of activated T cells. Blood. 2005 Apr 1; 105(7): 2821-7.). There are few reports on the effects of mesenchymal stem cells on CAR-T function. It has been reported that bone marrow mesenchymal stem cells suppress T cell responses but do not affect the activity of CD19 CAR-T cells (see: Zanetti SR et al., Bone marrow MSCs from pediatric patients with B-ALL highly immunosuppress T-cell responses but do not compromise CD19-CAR T-cell activity. J Immunother Cancer. 2020 Aug; 8(2): e001419.). Another study found that IL7-IL12 engineered mesenchymal stem cells improved CAR-T cell attack against colorectal cancer cells (see: Hombach AA et al., IL7-IL12 Engineered Mesenchymal Stem Cells (MSCs) Improve A CAR T Cell Attack Against Colorectal Cancer Cells. Cells. 2020 Apr 3; 9(4): 873.). Currently, the effect of MSC on CAR-T function is still controversial. Whether mesenchymal stem cells promote or inhibit CAR-T function requires more research.
如何提高CAR-T细胞治疗肿瘤的效果是本领域研究人员的首要目标。How to improve the effectiveness of CAR-T cell therapy for tumors is the primary goal of researchers in this field.
发明内容Summary of the invention
本发明首次发现了间充质干细胞对CAR-T功能的影响模式,并且首次提供了间充质干细胞作为CAR-T功能增强剂的用途。具体地,本发明通过将从健康捐赠者和孕妇的胎盘中诱导和培养的间充质干细胞与不同靶标的CAR-T细胞共同培养,从而确认了间充质干细胞对CAR-T细胞杀伤功能的影响,进一步地,本发明首次确认了这种影响通过CAR-T细胞的持久性和分化而体现。The present invention has discovered for the first time the effect pattern of mesenchymal stem cells on CAR-T function, and for the first time provided the use of mesenchymal stem cells as CAR-T function enhancers. Specifically, the present invention co-cultured mesenchymal stem cells induced and cultured from the placenta of healthy donors and pregnant women with CAR-T cells of different targets, thereby confirming the effect of mesenchymal stem cells on the killing function of CAR-T cells. Furthermore, the present invention has confirmed for the first time that this effect is manifested through the persistence and differentiation of CAR-T cells.
在一个方面,本发明涉及间充质干细胞用于增强免疫疗法疗效的用途,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫疗法所针对的抗原。在一些实施方案中,免疫疗法是细胞疗法,即,施用治疗性细胞的疗法,优选地,治疗性细胞是免疫效应细胞。在一些更具体的实施方案中,治疗性细胞是非特异性免疫效应细胞。在一些更具体的实施方案中,治疗性细胞是特异性免疫效应细胞,优选地,免疫效应细胞是T细胞;优选地,免疫效应细胞是经修饰的免疫效应细胞;更优选地,免疫效应细胞是CAR-T细胞。在一些实施方案中,人T细胞是CD8 +T细胞。在另一些具体的实施方案中,治疗性细胞是TIL、CAR-NK,TCR-T、CAR-DC、已负载抗原的DC、B细胞等等细胞。 In one aspect, the present invention relates to the use of mesenchymal stem cells for enhancing the efficacy of immunotherapy, wherein the surface of the mesenchymal stem cells does not express or substantially does not express the antigens targeted by the immunotherapy. In some embodiments, immunotherapy is cell therapy, that is, a therapy that administers therapeutic cells, preferably, therapeutic cells are immune effector cells. In some more specific embodiments, therapeutic cells are nonspecific immune effector cells. In some more specific embodiments, therapeutic cells are specific immune effector cells, preferably, immune effector cells are T cells; preferably, immune effector cells are modified immune effector cells; more preferably, immune effector cells are CAR-T cells. In some embodiments, human T cells are CD8 + T cells. In other specific embodiments, therapeutic cells are TIL, CAR-NK, TCR-T, CAR-DC, DC loaded with antigens, B cells, etc.
在一些实施方案中,所述免疫疗法针对的疾病是肿瘤。在一些更具体的实施方案中,所述肿瘤是血液系统肿瘤。在一些更具体的实施方案中,所述肿瘤是实体瘤。在一些更具体的实施方案中,所述免疫疗法针对肿瘤所表达的一种或多种肿瘤相关抗原(Tumor associated antigen,TAA),其中MSC表面不表达或实质上不表达所述肿瘤相关抗原分子。在一个优选的实施方案中,所述肿瘤相关抗原是IL-13Rα2、EphA2、CD19和/或EGFRvIII。In some embodiments, the disease targeted by the immunotherapy is a tumor. In some more specific embodiments, the tumor is a hematological tumor. In some more specific embodiments, the tumor is a solid tumor. In some more specific embodiments, the immunotherapy is directed against one or more tumor-associated antigens (TAA) expressed by the tumor, wherein the MSC surface does not express or substantially does not express the tumor-associated antigen molecule. In a preferred embodiment, the tumor-associated antigen is IL-13Rα2, EphA2, CD19 and/or EGFRvIII.
在一些实施方案中,间充质干细胞是骨髓、脂肪、外周血、脐带和/或胎盘来源的。在一些实施方案中,间充质干细胞是脐带和/或胎盘来源的。在一些实施方案中,间充质干细胞是骨髓、脂肪和/或胎盘来源的。在一些实施方案中,间充质干细胞是骨髓和/或胎盘来源的。在一些实施方案中,间充质干细胞是胎盘来源的。In some embodiments, mesenchymal stem cells are derived from bone marrow, fat, peripheral blood, umbilical cord and/or placenta. In some embodiments, mesenchymal stem cells are derived from umbilical cord and/or placenta. In some embodiments, mesenchymal stem cells are derived from bone marrow, fat and/or placenta. In some embodiments, mesenchymal stem cells are derived from bone marrow and/or placenta. In some embodiments, mesenchymal stem cells are derived from placenta.
在一些实施方案中,间充质干细胞表达CD105。在一些实施方案中,间充质干细胞不表达CD45。在一些优选的实施方案中,间充质干细胞是CD105 +CD45 -。在一些优选的实施方案中,间充质干细胞具有被诱导分化为成骨细胞的能力。在一些优选的实施方案中, 间充质干细胞具有被诱导分化为成脂细胞的能力。在一些优选的实施方案中,间充质干细胞具有被诱导分化为成软骨细胞的能力。 In some embodiments, the mesenchymal stem cells express CD105. In some embodiments, the mesenchymal stem cells do not express CD45. In some preferred embodiments, the mesenchymal stem cells are CD105 + CD45 - . In some preferred embodiments, the mesenchymal stem cells have the ability to be induced to differentiate into osteoblasts. In some preferred embodiments, the mesenchymal stem cells have the ability to be induced to differentiate into adipocytes. In some preferred embodiments, the mesenchymal stem cells have the ability to be induced to differentiate into chondrocytes.
在一些实施方案中,将所述间充质干细胞与所述免疫效应细胞在体外共培养。在一些实施方案中,将所述间充质干细胞与所述免疫效应细胞共同施用于受试者体内,其中间充质干细胞先于免疫效应细胞施用,或与其同时施用,或在施用免疫效应细胞之后施用。In some embodiments, the mesenchymal stem cells are co-cultured with the immune effector cells in vitro. In some embodiments, the mesenchymal stem cells are co-administered with the immune effector cells into a subject, wherein the mesenchymal stem cells are administered prior to the immune effector cells, or administered simultaneously with the immune effector cells, or administered after the immune effector cells are administered.
在一些实施方案中,所述免疫效应细胞与所述间充质干细胞的数量比例为100:1,或90:1,或80:1,或70:1,或60:1,或50:1,或40:1,或30:1,或20:1,或10:1,或5:1,或4:1,或2:1,或1:1,或1:2,或1:4,或1:5,或1:10,或介于其之间的比例。In some embodiments, the ratio of the number of immune effector cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
在一些具体的实施方案中,间充质干细胞促进所述免疫效应细胞(例如,优选地,CAR-T细胞)分泌细胞因子杀伤肿瘤细胞。在另一些优选的实施方案中,间充质干细胞增强免疫效应细胞(例如,优选地,CAR-T细胞)对靶细胞的直接杀伤作用。在一些具体的实施方案中,间充质干细胞促进免疫效应细胞(例如,优选地,CAR-T细胞)增殖扩增,即,数量增加。在一些具体的实施方案中,间充质干细胞促进治疗性细胞(例如,优选地,CAR-T细胞)持久性增加,即,在体内存活/存在的时间延长。在一些具体的实施方案中,间充质干细胞增强免疫效应细胞(例如,优选地,CAR-T细胞)到达肿瘤区域的能力。在一些具体的实施方案中,所述疗效增强剂促进免疫效应细胞(例如,优选地,CAR-T细胞)透过体内屏障(例如,血脑屏障)而使得更多免疫效应细胞(例如,优选地,CAR-T细胞)到达肿瘤(例如,胶质细胞瘤)区域,即,增加透过体内屏障(例如,血脑屏障)的免疫效应细胞(例如,优选地,CAR-T细胞)的数量。在一些具体的实施方案中,间充质干细胞调节免疫效应细胞(例如,优选地,CAR-T细胞)的分化,使其保持低分化状态。在一些具体的实施方案中,间充质干细胞促进免疫效应细胞(例如,优选地,CAR-T细胞)更多分化为记忆性细胞亚型。在一些优选的实施方案中,所述增强是通过免疫效应细胞的持久性增加和/或保持在低分化状态和/或更多分化为记忆性效应细胞亚型而实现的。In some specific embodiments, mesenchymal stem cells promote the immune effector cells (e.g., preferably, CAR-T cells) to secrete cytokines to kill tumor cells. In other preferred embodiments, mesenchymal stem cells enhance the direct killing effect of immune effector cells (e.g., preferably, CAR-T cells) on target cells. In some specific embodiments, mesenchymal stem cells promote the proliferation and amplification of immune effector cells (e.g., preferably, CAR-T cells), that is, the number increases. In some specific embodiments, mesenchymal stem cells promote the persistence of therapeutic cells (e.g., preferably, CAR-T cells), that is, the time of survival/existence in vivo is prolonged. In some specific embodiments, mesenchymal stem cells enhance the ability of immune effector cells (e.g., preferably, CAR-T cells) to reach the tumor area. In some specific embodiments, the efficacy enhancer promotes immune effector cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more immune effector cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increase the number of immune effector cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier) In some specific embodiments, mesenchymal stem cells regulate the differentiation of immune effector cells (e.g., preferably, CAR-T cells) to maintain a low differentiation state. In some specific embodiments, mesenchymal stem cells promote immune effector cells (e.g., preferably, CAR-T cells) to differentiate into more memory cell subtypes. In some preferred embodiments, the enhancement is achieved by increasing the persistence of immune effector cells and/or maintaining a low differentiation state and/or more differentiation into memory effector cell subtypes.
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力;(1) Enhance the proliferation ability of immune effector cells in vivo;
(2)增加单个免疫效应细胞体内持久性;(2) increase the in vivo persistence of single immune effector cells;
(3)提升由免疫效应细胞分泌的治疗性细胞因子(例如,干扰素γ(IFN-γ)、肿瘤坏死因子α(TNF-α)、IL-4、IL-6、IL-10和/或IL-17A)的量;(3) increasing the amount of therapeutic cytokines (e.g., interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), IL-4, IL-6, IL-10, and/or IL-17A) secreted by immune effector cells;
(4)增强携带靶抗原的细胞的杀伤;(4) Enhanced killing of cells carrying target antigens;
(5)抑制免疫效应细胞(例如,CAR-T细胞)的分化,使其保持低分化状态;(5) Inhibit the differentiation of immune effector cells (e.g., CAR-T cells) to keep them in a low-differentiation state;
(6)增加记忆性T细胞形成;(6) Increase the formation of memory T cells;
(7)增加靶抗原中和/抑制;(7) Increased target antigen neutralization/inhibition;
(8)延缓病灶进展/缩小病灶体积;(8) Delaying lesion progression/reducing lesion size;
(9)逆转接受治疗的受试者体内对免疫疗法的抑制因素/环境/情形;(9) reversing inhibitory factors/environments/conditions against immunotherapy in the treated subjects;
(10)提高接受治疗的受试者生存率和/或延长其生存期(即,寿命延长)。(10) Improving the survival rate and/or prolonging the survival period of the subjects receiving treatment (i.e., extending life span).
优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。Preferably, the immune effector cell is modified. Preferably, the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力;(1) Enhance the proliferation ability of immune effector cells in vivo;
(2)增加单个免疫效应细胞体内持久性;(2) increase the in vivo persistence of single immune effector cells;
(3)增强携带靶抗原的细胞的杀伤;(3) Enhanced killing of cells carrying target antigens;
(4)抑制免疫效应细胞(例如,CAR-T细胞)的分化,使其保持低分化状态;(4) Inhibit the differentiation of immune effector cells (e.g., CAR-T cells) to keep them in a low-differentiation state;
(5)增加记忆性T细胞形成;(5) Increase the formation of memory T cells;
(6)延缓病灶进展/缩小病灶体积;(6) Delaying lesion progression/reducing lesion size;
(7)提高接受治疗的受试者生存率和/或延长其生存期(即,寿命延长)。(7) Improving the survival rate and/or prolonging the survival period of the subjects receiving treatment (i.e., extending life span).
优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。Preferably, the immune effector cell is modified. Preferably, the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力或增加单个免疫效应细胞体内持久性;(1) Enhance the proliferation capacity of immune effector cells in vivo or increase the persistence of single immune effector cells in vivo;
(2)抑制CAR-T细胞的分化,使其保持低分化状态;(2) Inhibit the differentiation of CAR-T cells, keeping them in a low-differentiation state;
(3)增加记忆性T细胞形成;和(3) increase the formation of memory T cells; and
(4)提高接受治疗的受试者生存率和/或延长其生存期。(4) Improve the survival rate and/or prolong the survival of the subjects receiving treatment.
优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。Preferably, the immune effector cell is modified. Preferably, the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
因而,在另一个方面,本发明提供了间充质干细胞用于增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的用途。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。Thus, in another aspect, the present invention provides the use of mesenchymal stem cells for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of single immune effector cells after administration. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
本发明还提供了间充质干细胞用于调节免疫效应细胞在施用后的体内分化的用途。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。The present invention also provides the use of mesenchymal stem cells for regulating the differentiation of immune effector cells in vivo after administration. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
本发明还提供了间充质干细胞用于促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的用途。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。The present invention also provides the use of mesenchymal stem cells for promoting the differentiation of immune effector cells into memory cell subtypes in vivo after administration. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
在一个方面,本发明涉及间充质干细胞在制备抗肿瘤药物中的用途,其中所述药物还含有其他细胞。在一个具体的实施方案中,所述其他细胞是免疫效应细胞,且所述间充质干细胞表面不表达或实质上不表达被所述CAR-T细胞所特异性靶向的抗原。In one aspect, the present invention relates to the use of mesenchymal stem cells in the preparation of anti-tumor drugs, wherein the drug also contains other cells. In a specific embodiment, the other cells are immune effector cells, and the surface of the mesenchymal stem cells does not express or substantially does not express the antigen specifically targeted by the CAR-T cells.
在一些更具体的实施方案中,所述肿瘤是血液系统肿瘤。在一些更具体的实施方案中,所述肿瘤是实体瘤。In some more specific embodiments, the tumor is a hematological tumor.In some more specific embodiments, the tumor is a solid tumor.
在一些具体的实施方案中,免疫效应细胞是T细胞。在一些具体的实施方案中,免疫效应细胞是经修饰的,优选地,转导并表达嵌合抗原受体(CAR)的T细胞,即,CAR-T细胞。In some specific embodiments, immune effector cells are T cells. In some specific embodiments, immune effector cells are modified, preferably, transduced and express chimeric antigen receptor (CAR) T cells, i.e., CAR-T cells.
在一些具体的实施方案中,特异性免疫效应细胞针对肿瘤所表达的一种或多种肿瘤相关抗原。在一个优选的实施方案中,特异性免疫效应细胞针对的靶TAA是IL-13Rα2、EphA2、CD19或EGFRvIII。In some specific embodiments, the specific immune effector cells are directed against one or more tumor-associated antigens expressed by the tumor. In a preferred embodiment, the target TAA directed against by the specific immune effector cells is IL-13Rα2, EphA2, CD19 or EGFRvIII.
在一些实施方案中,间充质干细胞是胎盘来源的。在一些具体的实施方案中,胎盘来源的间充质干细胞表达CD105且不表达CD45。In some embodiments, the mesenchymal stem cells are of placental origin. In some specific embodiments, the placental-derived mesenchymal stem cells express CD105 and do not express CD45.
在一些实施方案中,将所述间充质干细胞与细胞疗法中所用的治疗性细胞在体外共培养。在一些实施方案中,将所述间充质干细胞与细胞疗法中所用的治疗性细胞共同施用于受试者体内,其中间充质干细胞先于治疗性细胞施用,或与其同时施用,或在施用治疗性细胞之后施用。In some embodiments, the mesenchymal stem cells are co-cultured in vitro with therapeutic cells used in cell therapy. In some embodiments, the mesenchymal stem cells are co-administered with therapeutic cells used in cell therapy into a subject, wherein the mesenchymal stem cells are administered prior to, or simultaneously with, or after the therapeutic cells are administered.
在一些实施方案中,所述治疗性细胞与所述间充质干细胞的数量比例为100:1,或90:1,或80:1,或70:1,或60:1,或50:1,或40:1,或30:1,或20:1,或10:1,或5:1,或4:1,或2:1,或1:1,或1:2,或1:4,或1:5,或1:10,或介于其之间的比例。In some embodiments, the number ratio of the therapeutic cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
在一个方面,本发明涉及间充质干细胞与其他细胞联合用于制备抗肿瘤药物的用途,其中所述间充质干细胞表面实质上不表达被其他细胞所特异性靶向的抗原,且所述癌症的细胞表达被其他细胞所特异性靶向的抗原。In one aspect, the present invention relates to the use of mesenchymal stem cells in combination with other cells for preparing anti-tumor drugs, wherein the surface of the mesenchymal stem cells does not substantially express antigens specifically targeted by other cells, and the cancer cells express antigens specifically targeted by other cells.
在一些更具体的实施方案中,所述肿瘤是血液系统肿瘤。在一些更具体的实施方案中,所述肿瘤是实体瘤。In some more specific embodiments, the tumor is a hematological tumor.In some more specific embodiments, the tumor is a solid tumor.
在一些实施方案中,其他细胞是免疫效应细胞。在一些实施方案中,免疫效应细胞是T细胞。在一些具体的实施方案中,免疫效应细胞是经修饰的,优选地,转导并表达嵌合抗原受体(CAR)的T细胞,即,CAR-T细胞。In some embodiments, other cells are immune effector cells. In some embodiments, immune effector cells are T cells. In some specific embodiments, immune effector cells are modified, preferably, transduced and express chimeric antigen receptor (CAR) T cells, i.e., CAR-T cells.
在一些具体的实施方案中,特异性免疫效应细胞针对肿瘤所表达的一种或多种肿瘤相关抗原。在一个优选的实施方案中,特异性免疫效应细胞针对的靶TAA是IL-13Rα2、EphA2、CD19或EGFRvIII。In some specific embodiments, the specific immune effector cells are directed against one or more tumor-associated antigens expressed by the tumor. In a preferred embodiment, the target TAA directed against by the specific immune effector cells is IL-13Rα2, EphA2, CD19 or EGFRvIII.
在一些实施方案中,间充质干细胞是胎盘来源的。在一些具体的实施方案中,间充质干细胞表达CD105且不表达CD45。In some embodiments, the mesenchymal stem cells are of placental origin. In some specific embodiments, the mesenchymal stem cells express CD105 and do not express CD45.
在一些实施方案中,将所述间充质干细胞与细胞疗法中所用的治疗性细胞在体外共培养。在一些实施方案中,将所述间充质干细胞与细胞疗法中所用的治疗性细胞共同施用于受试者体内,其中间充质干细胞先于治疗性细胞施用,或与其同时施用,或在施用治疗性细胞之后施用。In some embodiments, the mesenchymal stem cells are co-cultured in vitro with therapeutic cells used in cell therapy. In some embodiments, the mesenchymal stem cells are co-administered with therapeutic cells used in cell therapy into a subject, wherein the mesenchymal stem cells are administered prior to, or simultaneously with, or after the therapeutic cells are administered.
在一些实施方案中,所述治疗性细胞与所述间充质干细胞的数量比例为100:1,或90:1,或80:1,或70:1,或60:1,或50:1,或40:1,或30:1,或20:1,或10:1,或5:1,或4:1,或2:1,或1:1,或1:2,或1:4,或1:5,或1:10,或介于其之间的比例。In some embodiments, the number ratio of the therapeutic cells to the mesenchymal stem cells is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
在又一个方面,本发明提供了一种组合物,其包含间充质干细胞,优选地,胎盘源 性间充质干细胞。所述组合物用作免疫疗法的疗效增强剂。在一个具体的实施方案中,所述免疫疗法针对血液系统肿瘤。在另一个具体的实施方案中,所述免疫疗法针对实体瘤。在一个优选的实施方案中,所述免疫疗法的靶分子是IL-13Rα2、EphA2、CD19或EGFRvIII。In another aspect, the present invention provides a composition comprising mesenchymal stem cells, preferably placental-derived mesenchymal stem cells. The composition is used as an immunotherapy efficacy enhancer. In a specific embodiment, the immunotherapy is directed against hematological tumors. In another specific embodiment, the immunotherapy is directed against solid tumors. In a preferred embodiment, the target molecule of the immunotherapy is IL-13Rα2, EphA2, CD19 or EGFRvIII.
在一些具体的实施方案中,组合物促进治疗性细胞(例如,优选地,CAR-T细胞)分泌细胞因子杀伤肿瘤细胞。在另一些具体的实施方案中,组合物增强治疗性细胞(例如,优选地,CAR-T细胞)对靶细胞的直接杀伤作用。在一些具体的实施方案中,组合物促进治疗性细胞(例如,优选地,CAR-T细胞)增殖,即,数量增加。在一些优选的实施方案中,组合物增强治疗性细胞(例如,优选地,CAR-T细胞)到达肿瘤区域的能力。在一些具体的实施方案中,所述疗效增强剂促进治疗性细胞(例如,优选地,CAR-T细胞)透过体内屏障(例如,血脑屏障)而使得更多治疗性细胞(例如,优选地,CAR-T细胞)到达肿瘤(例如,胶质细胞瘤)区域,即,增加透过体内屏障(例如,血脑屏障)的治疗性细胞(例如,优选地,CAR-T细胞)的数量。在一些具体的实施方案中,间充质干细胞调节免疫效应细胞(例如,优选地,CAR-T细胞)的分化,使其保持低分化状态。在一些具体的实施方案中,间充质干细胞促进免疫效应细胞(例如,优选地,CAR-T细胞)更多分化为记忆性细胞亚型。在一些优选的实施方案中,所述增强是通过免疫效应细胞的持久性增加和/或保持在低分化状态和/或更多分化为记忆性效应细胞亚型而实现的。In some specific embodiments, the composition promotes therapeutic cells (e.g., preferably, CAR-T cells) to secrete cytokines to kill tumor cells. In other specific embodiments, the composition enhances the direct killing effect of therapeutic cells (e.g., preferably, CAR-T cells) on target cells. In some specific embodiments, the composition promotes the proliferation of therapeutic cells (e.g., preferably, CAR-T cells), that is, the number increases. In some preferred embodiments, the composition enhances the ability of therapeutic cells (e.g., preferably, CAR-T cells) to reach the tumor area. In some specific embodiments, the efficacy enhancer promotes therapeutic cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more therapeutic cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increases the number of therapeutic cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier). In some specific embodiments, mesenchymal stem cells regulate the differentiation of immune effector cells (e.g., preferably, CAR-T cells) to maintain a low differentiation state. In some specific embodiments, mesenchymal stem cells promote immune effector cells (e.g., preferably, CAR-T cells) to differentiate more into memory cell subtypes. In some preferred embodiments, the enhancement is achieved by increasing the persistence of immune effector cells and/or remaining in a poorly differentiated state and/or more differentiation into memory effector cell subtypes.
在又一个方面,本发明提供了间充质干细胞的用途,用于制备以下一种或多种:In yet another aspect, the present invention provides the use of mesenchymal stem cells for preparing one or more of the following:
(1)增强免疫效应细胞体内增殖能力和/或单个免疫效应细胞体内持久性的试剂;(1) Agents that enhance the in vivo proliferation capacity of immune effector cells and/or the in vivo persistence of single immune effector cells;
(2)使得免疫效应细胞保持低分化状态的免疫效应细胞分化抑制剂;(2) immune effector cell differentiation inhibitors that keep immune effector cells in a low-differentiation state;
(3)使得记忆性免疫效应细胞亚型形成增加的试剂;(3) Agents that increase the formation of memory immune effector cell subtypes;
(4)细胞(优选免疫效应细胞)疗法的疗效增强剂;(4) An agent for enhancing the efficacy of cell (preferably immune effector cell) therapy;
其中,优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。Wherein, preferably, the immune effector cell is modified. Preferably, the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
在又一个方面,本发明提供了间充质干细胞与免疫效应细胞联合用于制备治疗癌症的药物的用途,其中,优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides the use of mesenchymal stem cells in combination with immune effector cells for preparing a drug for treating cancer, wherein, preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
在又一个方面,本发明提供了药物组合物,其包含本发明前述的间充质干细胞(例如,胎盘源性间充质干细胞)或组合物,另外还包含适当的可药用载体。所述可药用载体使得所述药物组合物适合于通过预期的途径给药,例如,但不限于,静脉注射或肿瘤局部注射等等。在一些实施方案中,本发明的药物组合物进一步包含其他细胞(例如,优选地,治疗性细胞,更优选地,CAR-T细胞)。In another aspect, the present invention provides a pharmaceutical composition comprising the aforementioned mesenchymal stem cells (e.g., placental-derived mesenchymal stem cells) or compositions of the present invention, and further comprising a suitable pharmaceutically acceptable carrier. The pharmaceutically acceptable carrier makes the pharmaceutical composition suitable for administration by the intended route, for example, but not limited to, intravenous injection or local injection of tumors, etc. In some embodiments, the pharmaceutical composition of the present invention further comprises other cells (e.g., preferably, therapeutic cells, more preferably, CAR-T cells).
在另一个方面,本发明提供了一种方法,包括向有需要的受试者施用免疫效应细胞,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。在一个实施方案中,受试者具有本文所描述的病症,例如,受试者患有癌症,例如,受试者具有表达本文所述的靶抗原的癌症。在一个实施方案 中,受试者是人。In another aspect, the present invention provides a method comprising administering immune effector cells to a subject in need thereof, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells. In one embodiment, the subject has a condition described herein, e.g., the subject has cancer, e.g., the subject has a cancer that expresses a target antigen described herein. In one embodiment, the subject is a human.
在另一个方面,本发明涉及治疗患有与如本文所述的癌症相关抗原的表达相关的疾病的受试者的方法,包括向有需要的受试者施用免疫效应细胞,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention relates to a method for treating a subject with a disease associated with the expression of a cancer-associated antigen as described herein, comprising administering immune effector cells to a subject in need thereof, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
在又一个方面中,本发明提供了治疗患有与肿瘤抗原的表达相关的疾病的受试者的方法,其包括向有需要的受试者施用免疫效应细胞,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a method for treating a subject with a disease associated with the expression of a tumor antigen, comprising administering an immune effector cell to a subject in need thereof, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cell is modified. Preferably, the immune effector cell is a T cell. More preferably, the immune effector cell is a CAR-T cell.
在又一个方面,本发明提供了增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering with mesenchymal stem cells to the body. Therefore, in a specific embodiment, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vitro/ex vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells to the body. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
本发明还提供了调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。The present invention also provides a method for regulating the differentiation of immune effector cells in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
本发明还提供了促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。The present invention also provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells.
附图说明BRIEF DESCRIPTION OF THE DRAWINGS
图1-4:健康自然分娩孕妇的胎盘间充质干细胞的表征。图1所示为胎盘源性间充质干细胞的显微镜下形态。图2所示为间充质干细胞体外增殖计数的流程图(上图),以及按照体外六次传代的群体倍增计算间充质干细胞的增殖(下图)。图3所示为通过流式细胞术分析扩增的MSC第3代的免疫表型,纵轴反映细胞数量,横轴代表染色强度。左图显示细胞群体CD45特异性染色(通过抗CD45mAb)基本为阴性;右图显示细胞群体CD105特异性染色(通过抗CD105mAb),深色曲线代表同型匹配的阴性对照mAb染色,浅色代表mAb特异性染色的细胞,可见绝大多数细胞可明显被CD105抗体所染色。图4所示为通过茜素红染色显示胎盘源性间充质干细胞的成骨分化能力,诱导成骨分化后可见明显团块、结节样钙沉积被染色。MSC,间充质干细胞,N=3。Figures 1-4: Characterization of placental mesenchymal stem cells from healthy natural delivery pregnant women. Figure 1 shows the microscopic morphology of placental-derived mesenchymal stem cells. Figure 2 shows a flow chart for counting the proliferation of mesenchymal stem cells in vitro (top), and the calculation of the proliferation of mesenchymal stem cells according to the population doubling of six passages in vitro (bottom). Figure 3 shows the immunophenotype of the third generation of MSCs amplified by flow cytometry analysis, with the vertical axis reflecting the number of cells and the horizontal axis representing the staining intensity. The left figure shows that the cell population CD45-specific staining (by anti-CD45mAb) is basically negative; the right figure shows the cell population CD105-specific staining (by anti-CD105mAb), the dark curve represents the isotype-matched negative control mAb staining, and the light color represents the mAb-specific stained cells. It can be seen that the vast majority of cells can be clearly stained with CD105 antibodies. Figure 4 shows the osteogenic differentiation ability of placental-derived mesenchymal stem cells by Alizarin red staining. After inducing osteogenic differentiation, obvious clumps and nodular calcium deposits can be stained. MSC, mesenchymal stem cells, N=3.
图5:通过流式细胞术检测MSC中肿瘤表面抗原的表达。(A)阴性对照;(B,C,D)分别检测MSC表面的uPAR、IL13Rα2和CD19的表达。MSC,间充质干细胞。Figure 5: Detection of tumor surface antigen expression in MSC by flow cytometry. (A) Negative control; (B, C, D) Detection of the expression of uPAR, IL13Rα2 and CD19 on the surface of MSC, respectively. MSC, mesenchymal stem cell.
图6-10:CAR-T细胞的制备和间充质干细胞对体外CAR-T功能的影响。图6所示为特异性靶向CD19、IL13Rα2和uPAR的三种CAR分子的结构示意图。图7所示为CAR-T细胞的制备连同体外功能研究按时间节点的流程图。图8所示为流式细胞仪检测MSC对CD19-IL15 CAR-T细胞增殖的影响。CAR-T细胞首先与间充质干细胞以10:1的比例共培养24小时,随后收集并以1:1的比例与/不与NALM-6细胞共培养3天,进行流式细胞术分析。相同条件下(包括与/不与NALM-6细胞共培养)未经与MSC共培养的CAR-T细胞作为对照组。图9所示为流式细胞仪检测MSC对CD19-IL15 CAR-T细胞活化的影响。CAR-T细胞提前与MSC共培养,并与/不与NALM-6共培养6小时。图10所示用CBA方法检测MSC对CD19-IL15 CAR-T细胞的细胞因子释放的影响。CAR-T细胞与MSC共培养24小时,随后与NALM-6(A)和Raji(B)细胞共培养过夜,E:T比例为1:1,收集共培养的上清液进行细胞因子检测。对于每种细胞因子,5个条形从左向右分别代表单独靶肿瘤细胞组、单独CD19-IL15 CAR-T细胞组、CD19-IL15 CAR-T细胞+靶肿瘤细胞组、经过MSC共培养的CD19-IL15 CAR-T细胞+靶肿瘤细胞组和经过MSC共培养的CD19-IL15 CAR-T细胞组。数据显示为平均值±SEM。"*"代表p<0.05;"**"代表p<0.01;"***"代表p<0.001;MSC:间充质干细胞,N=3。CAR-T细胞N=3。Figures 6-10: Preparation of CAR-T cells and the effect of mesenchymal stem cells on in vitro CAR-T function. Figure 6 shows a schematic diagram of the structures of three CAR molecules that specifically target CD19, IL13Rα2 and uPAR. Figure 7 shows a flowchart of the preparation of CAR-T cells together with in vitro functional studies by time nodes. Figure 8 shows the effect of MSC on the proliferation of CD19-IL15 CAR-T cells detected by flow cytometry. CAR-T cells were first co-cultured with mesenchymal stem cells at a ratio of 10:1 for 24 hours, and then collected and co-cultured with/without NALM-6 cells at a ratio of 1:1 for 3 days for flow cytometric analysis. CAR-T cells that were not co-cultured with MSC under the same conditions (including co-culture with/without NALM-6 cells) were used as the control group. Figure 9 shows the effect of MSC on the activation of CD19-IL15 CAR-T cells detected by flow cytometry. CAR-T cells were co-cultured with MSC in advance and co-cultured with/without NALM-6 for 6 hours. Figure 10 shows the effect of MSC on cytokine release of CD19-IL15 CAR-T cells detected by CBA method. CAR-T cells were co-cultured with MSC for 24 hours, and then co-cultured with NALM-6 (A) and Raji (B) cells overnight, with an E:T ratio of 1:1, and the co-culture supernatant was collected for cytokine detection. For each cytokine, the 5 bars from left to right represent the target tumor cell group alone, the CD19-IL15 CAR-T cell group alone, the CD19-IL15 CAR-T cell + target tumor cell group, the CD19-IL15 CAR-T cell + target tumor cell group co-cultured with MSC, and the CD19-IL15 CAR-T cell group co-cultured with MSC. Data are shown as mean ± SEM. "*" represents p < 0.05; "**" represents p < 0.01; "***" represents p < 0.001; MSC: mesenchymal stem cells, N = 3. CAR-T cells N = 3.
图11-13:MSC对体外CD19-IL15 CAR-T细胞抗肿瘤功能的影响。图11所示为通过流式细胞术分析MSC对CD19-IL15 CAR-T细胞抗NALM-6-GL细胞系的影响。CAR-T细胞首先与MSC以10:1的比例共培养,然后与NALM-6-GL共培养,比例为1:1。提供了流式细胞术结果分析图和统计柱状图。图12所示为通过流式细胞术分析MSC对CD19-IL15 CAR-T细胞抗Raji-GL细胞系的影响。CAR-T细胞事先与间充质干细胞以10:1的比例共同培养,然后与Raji-GL以1:1的比例共同培养,提供了流式细胞术结果分析图和统计柱状图。图13所示为通过检测荧光素酶活性,分析MSC对CD19-IL15 CAR-T细胞抗NALM-6-GL细胞(A)和抗Raji-GL细胞(B)的影响。其中,CAR-T细胞首先与MSC以10:1的比例共培养,然后收集,并以1:1,2:1,和5:1三个比例与NALM-GL或Raji-GL共培 养。"*"代表p<0.05;"**"代表p<0.01;"***"代表p<0.001;GL:eGFP-Luc;MSC:间充质干细胞,N=3;E:T比例为效靶比;CAR-T细胞N=3。Figures 11-13: Effects of MSCs on the anti-tumor function of CD19-IL15 CAR-T cells in vitro. Figure 11 shows the effect of MSCs on CD19-IL15 CAR-T cells against NALM-6-GL cell lines by flow cytometry analysis. CAR-T cells were first co-cultured with MSCs at a ratio of 10:1 and then co-cultured with NALM-6-GL at a ratio of 1:1. Flow cytometry result analysis graphs and statistical bar graphs are provided. Figure 12 shows the effect of MSCs on CD19-IL15 CAR-T cells against Raji-GL cell lines by flow cytometry analysis. CAR-T cells were co-cultured with mesenchymal stem cells at a ratio of 10:1 in advance and then co-cultured with Raji-GL at a ratio of 1:1. Flow cytometry result analysis graphs and statistical bar graphs are provided. Figure 13 shows the effect of MSCs on CD19-IL15 CAR-T cells against NALM-6-GL cells (A) and against Raji-GL cells (B) by detecting luciferase activity. Among them, CAR-T cells were first co-cultured with MSCs at a ratio of 10:1, and then collected and co-cultured with NALM-GL or Raji-GL at three ratios of 1:1, 2:1, and 5:1. "*" represents p < 0.05; "**" represents p < 0.01; "***" represents p < 0.001; GL: eGFP-Luc; MSC: mesenchymal stem cells, N = 3; E:T ratio is effector-target ratio; CAR-T cells N = 3.
图14-16:MSC对IL13Rα2 CAR-T细胞抗U87细胞的功能的影响。图14所示为通过流式细胞术分析CD107a的激活情况。CAR-T细胞首先以10:1的比例与间充质干细胞共培养,然后与/不与U87-GL细胞系共培养6小时。图15所示为通过ELISA检测细胞因子的释放。CAR-T细胞与MSC共培养24小时,然后收集并与U87-GL细胞共培养过夜,E:T比例为1:1,获取上清液进行细胞因子检测。对于每种细胞因子,5个条形从左向右分别代表单独U87细胞组、单独IL13Rα2 CAR-T细胞组、IL13Rα2 CAR-T细胞+U87细胞组、经过MSC共培养的IL13Rα2 CAR-T细胞+U87细胞组和经过MSC共培养的IL13Rα2 CAR-T细胞组。图16所示为流式细胞术分析抗肿瘤的效果。CAR-T细胞与MSC共培养24小时,然后收集与U87-GL细胞共培养过夜,E:T比例为1:1。提供了流式细胞术结果分析图和统计柱状图。"*"代表p<0.05;"**"代表p<0.01;"***"代表p<0.001。GL:eGFP-Luc;MSC:间充质干细胞,N=3;CAR-T细胞N=3。Figures 14-16: Effect of MSC on the function of IL13Rα2 CAR-T cells against U87 cells. Figure 14 shows the activation of CD107a analyzed by flow cytometry. CAR-T cells were first co-cultured with mesenchymal stem cells at a ratio of 10:1, and then co-cultured with/without U87-GL cell line for 6 hours. Figure 15 shows the release of cytokines detected by ELISA. CAR-T cells were co-cultured with MSC for 24 hours, then collected and co-cultured with U87-GL cells overnight at an E:T ratio of 1:1, and the supernatant was obtained for cytokine detection. For each cytokine, the 5 bars from left to right represent the U87 cell group alone, the IL13Rα2 CAR-T cell group alone, the IL13Rα2 CAR-T cell + U87 cell group, the IL13Rα2 CAR-T cell + U87 cell group co-cultured with MSC, and the IL13Rα2 CAR-T cell group co-cultured with MSC. Figure 16 shows the anti-tumor effect analyzed by flow cytometry. CAR-T cells were co-cultured with MSC for 24 hours and then collected and co-cultured with U87-GL cells overnight, with an E:T ratio of 1:1. Flow cytometry result analysis chart and statistical bar graph are provided. "*" represents p<0.05; "**" represents p<0.01; "***" represents p<0.001. GL: eGFP-Luc; MSC: mesenchymal stem cells, N=3; CAR-T cells N=3.
图17-21:MSC对uPAR CAR-T细胞抗H460-GL细胞的功能的影响。图17所示为CAR-T细胞CD107a的激活情况。图18所示为细胞因子释放的检测。图19所示为E:T比例为1:1的体外抗肿瘤效果,提供了流式细胞术结果分析图和统计柱状图。图20所示为通过检测荧光素酶的活性,反映在不同的E:T比例下,uPAR CAR-T细胞的抗肿瘤效果。图21所示为uPAR CAR-T细胞对MSC表现出杀伤功能。GL:eGFP-Luc;MSC:间充质干细胞,N=3;E:T比例为效靶比;CAR-T细胞N=3。Figures 17-21: Effects of MSC on the function of uPAR CAR-T cells against H460-GL cells. Figure 17 shows the activation of CAR-T cell CD107a. Figure 18 shows the detection of cytokine release. Figure 19 shows the in vitro anti-tumor effect of an E:T ratio of 1:1, and provides a flow cytometry result analysis chart and a statistical bar chart. Figure 20 shows the anti-tumor effect of uPAR CAR-T cells at different E:T ratios by detecting luciferase activity. Figure 21 shows that uPAR CAR-T cells exhibit killing function against MSC. GL: eGFP-Luc; MSC: mesenchymal stem cells, N = 3; E:T ratio is the effector-target ratio; CAR-T cells N = 3.
图22-25:MSC对CD19-IL15 CAR-T细胞分化的影响。图22所示为流式细胞仪检测TCM表型的分化情况的流式细胞术分析图。CAR-T细胞首先与间充质干细胞共培养24小时,然后收集,并与/不与NALM-6细胞共培养。图23所示为同一测定的统计结果柱状图。图24所示为通过qRT-PCR检测转录因子TCF-7的表达。图25所示为通过ELISA检测细胞因子IL2的释放。CAR-T细胞在不含IL2的培养基中培养3天,与MSC共培养24小时,然后与NALM-6(A)或Raji细胞(B)在不含IL2的相同培养基中培养过夜,获取共培养的上清液进行IL2检测。"*"代表p<0.05;"**"代表p<0.01;"***"代表p<0.001。MSC:间充质干细胞,N=3;CAR-T细胞N=3。Figures 22-25: Effect of MSC on differentiation of CD19-IL15 CAR-T cells. Figure 22 shows flow cytometric analysis of differentiation of TCM phenotype detected by flow cytometry. CAR-T cells were first co-cultured with mesenchymal stem cells for 24 hours, then collected and co-cultured with/without NALM-6 cells. Figure 23 shows a bar graph of statistical results of the same assay. Figure 24 shows the expression of transcription factor TCF-7 detected by qRT-PCR. Figure 25 shows the release of cytokine IL2 detected by ELISA. CAR-T cells were cultured in medium without IL2 for 3 days, co-cultured with MSC for 24 hours, and then cultured with NALM-6 (A) or Raji cells (B) in the same medium without IL2 overnight, and the co-culture supernatant was obtained for IL2 detection. "*" represents p < 0.05; "**" represents p < 0.01; "***" represents p < 0.001. MSC: mesenchymal stem cells, N = 3; CAR-T cells N = 3.
图26-29:MSC对CD19-IL15 CAR-T细胞在NALM-6-GL异种移植小鼠模型中的抗肿瘤效果的影响。图26所示为体内实验的流程图,注明了时间节点。1×10 6个NALM-6-GL细胞经尾静脉注射于NOD-SCID小鼠(N=12)体内,24小时后通过尾静脉注射1×10 7个CAR-T细胞,其中包含与/不与MSC共同培养的CAR-T细胞。小鼠的分组方式为随机的3组,每组4只小鼠。图27所示为小鼠的肿瘤荧光成像,图28所示为每只小鼠的荧光相对定量结果,其中CAR-T细胞的对照组成瘤小鼠生存期极短,仅获取前两周的相关数据,这也反映在图29所示的小鼠的生存曲线中。"*"代表p<0.05;"**"代表p<0.01;"***"代表p<0.001。 Figures 26-29: Effect of MSC on the anti-tumor effect of CD19-IL15 CAR-T cells in the NALM-6-GL xenograft mouse model. Figure 26 shows a flow chart of the in vivo experiment, with time nodes indicated. 1×10 6 NALM-6-GL cells were injected into NOD-SCID mice (N=12) via the tail vein, and 1×10 7 CAR-T cells were injected via the tail vein 24 hours later, including CAR-T cells co-cultured with/without MSC. The mice were randomly divided into 3 groups, with 4 mice in each group. Figure 27 shows the tumor fluorescence imaging of mice, and Figure 28 shows the relative quantitative results of fluorescence for each mouse, among which the survival period of the tumor-bearing mice in the control group of CAR-T cells was extremely short, and only the relevant data for the first two weeks were obtained, which is also reflected in the survival curve of the mice shown in Figure 29. "*" represents p<0.05;"**" represents p<0.01;"***" represents p<0.001.
具体实施方式Detailed ways
如在本文中所用的,单数形式“一个”、“一种”、“该”和“所述”可以包括多于一个或一种的被指代物,除非上下文另外明确规定。如本文所用,“约”应理解为是指处在该所提及数字的-5%至+5%的范围内。此外,本文中的所有数值范围都应理解为包含该范围内的所有整数或分数。本文所公开的组合物可以不含本文未具体公开的任何要素。因此,使用术语“包括”或“包含”的实施方案的公开内容包括“基本上由所指明的组分组成”和“由所指明的组分组成”的实施方案的公开内容。As used herein, the singular forms "a", "an", "the" and "said" may include more than one or one referent unless the context clearly dictates otherwise. As used herein, "about" should be understood to mean within the range of -5% to +5% of the referenced number. In addition, all numerical ranges herein are understood to include all integers or fractions within the range. The compositions disclosed herein may be free of any elements not specifically disclosed herein. Therefore, disclosures of embodiments using the term "comprising" or "including" include disclosures of embodiments "consisting essentially of the specified components" and "consisting of the specified components."
如本文所用,术语“间充质干细胞”,本领域也称为“间质干细胞(mesenchymal stem cell,MSC)”,是指一类源自中胚层的,具有一定分化潜能、可以分化成多种细胞类型的多能基质细胞群。其主要源自和存在于骨髓,此外也包括广泛源自其他“非骨髓”组织的多能细胞,例如:胎盘、脐带血、脂肪组织、成人肌肉、角膜基质、乳牙牙髓等等组织。As used herein, the term "mesenchymal stem cells", also known in the art as "mesenchymal stem cells (MSC)", refers to a group of multipotent stromal cells derived from the mesoderm, with certain differentiation potential and can differentiate into a variety of cell types. They are mainly derived from and exist in the bone marrow, and also include multipotent cells widely derived from other "non-bone marrow" tissues, such as: placenta, umbilical cord blood, adipose tissue, adult muscle, corneal stroma, deciduous tooth pulp, etc.
适合于本发明的间充质干细胞Mesenchymal stem cells suitable for the present invention
可以通过以下方法产生本文所述的分离的间充质干细胞群体:用组织破坏酶消化包含间充质干细胞的组织(例如,但不限于,骨髓、胎盘、脂肪、脐带、外周血等)以获得包含间充质干细胞的间充质干细胞群体,和从剩余物中分离或基本分离多个间充质干细胞。可以将全部或任何部分的组织消化以获得本文所述的分离的间充质干细胞。The isolated mesenchymal stem cell population described herein can be produced by the following method: digesting tissues (e.g., but not limited to, bone marrow, placenta, adipose, umbilical cord, peripheral blood, etc.) containing mesenchymal stem cells with tissue-destroying enzymes to obtain a mesenchymal stem cell population containing mesenchymal stem cells, and isolating or substantially isolating a plurality of mesenchymal stem cells from the remainder. All or any portion of the tissue can be digested to obtain the isolated mesenchymal stem cells described herein.
当在原代培养或在细胞培养中培养时,如本文所述使用的间充质干细胞贴壁到组织培养基底上,例如,组织培养容器表面(例如,组织培养塑料制品)。培养中的间充质干细胞呈现出一般成纤维样星形外观,其中多个胞质突从中央细胞体中伸出。然而,由于间充质干细胞显示出比成纤维细胞数目更多的这些过程,因此间充质干细胞在形态上与相同条件下培养的成纤维细胞是可区分的。在形态上,间充质干细胞还与造血干细胞是可区分的,造血干细胞在培养时一般呈现出更圆的或卵石形态。When cultivated in primary culture or in cell culture, the mesenchymal stem cells used as described herein adhere to a tissue culture substrate, for example, a tissue culture vessel surface (e.g., a tissue culture plastic). The mesenchymal stem cells in culture present a general fibroblast-like star-shaped appearance, in which a plurality of cytoplasmic processes extend from a central cell body. However, since the mesenchymal stem cells show these processes more than the fibroblast number, the mesenchymal stem cells are distinguishable in morphology from the fibroblasts cultivated under the same conditions. In morphology, the mesenchymal stem cells are also distinguishable from hematopoietic stem cells, which generally present a more rounded or pebble morphology when cultivated.
在本文所公开的技术方案中有用的分离的间充质干细胞(例如,分离的多能间充质干细胞或分离的间充质干细胞群体)是贴壁细胞(例如在组织培养塑料上贴壁),其具有多能细胞或干细胞的特征并且表达可用于鉴别和/或分离所述细胞或包含所述干细胞的细胞群体的多种标志物。The isolated mesenchymal stem cells (e.g., isolated multipotent mesenchymal stem cells or isolated mesenchymal stem cell populations) useful in the technical solutions disclosed herein are adherent cells (e.g., adherent to tissue culture plastic) that have the characteristics of multipotent cells or stem cells and express a variety of markers that can be used to identify and/or isolate the cells or cell populations containing the stem cells.
在一些实施方案中,所述分离的间充质干细胞是CD45 -和CD105 +,如通过流式细胞术所检测的。在一些实施方案中,所述间充质干细胞是CD73 +、CD90 +和CD105 +,并且CD14、CD34和CD45表达水平极低/不表达。在某些实施方案中,分离的间充质干细胞具有分化为神经表型细胞、成骨表型细胞和/或软骨形成表型细胞的潜能。在一个具体的实施方案中,所述间充质干细胞表面表达约95%的CD105,基本上不表达CD45,并在暴露于成骨诱导培养基3-4周后可分化为成骨细胞。 In some embodiments, the separated mesenchymal stem cells are CD45 - and CD105 + , as detected by flow cytometry. In some embodiments, the mesenchymal stem cells are CD73 + , CD90 + and CD105 + , and CD14, CD34 and CD45 expression levels are extremely low/not expressed. In certain embodiments, the separated mesenchymal stem cells have the potential to differentiate into neural phenotype cells, osteogenic phenotype cells and/or chondrogenic phenotype cells. In a specific embodiment, the mesenchymal stem cells express about 95% of CD105 on the surface, substantially do not express CD45, and can differentiate into osteoblasts after being exposed to osteogenic induction medium for 3-4 weeks.
如上所述的分离的间充质干细胞群体一般可以包含约、至少或不超过1×10 5、5×10 5、1×10 6、5×10 6、1×10 7、5×10 7、1×10 8、5×10 8、1×10 9、5×10 9、1×10 10、5×10 10、1×10 11或更多个分离的间充质干细胞。在本文所述的治疗方法中有用的分离的间充质干细胞群体包含至少50%、55%、60%、65%、70%、75%、80%、85%、90%、95%、98% 或99%的活的分离的间充质干细胞,例如,如通过(例如)台盼蓝拒染法所确定的。 The isolated mesenchymal stem cell populations described above may generally contain about, at least, or no more than 1×10 5 , 5×10 5 , 1×10 6 , 5×10 6 , 1×10 7 , 5×10 7 , 1×10 8 , 5×10 8 , 1×10 9 , 5×10 9 , 1×10 10 , 5×10 10 , 1×10 11 or more isolated mesenchymal stem cells. The isolated mesenchymal stem cell populations useful in the methods of treatment described herein contain at least 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90%, 95%, 98% or 99% viable isolated mesenchymal stem cells, e.g., as determined by, e.g., trypan blue exclusion.
对于任何上述间充质干细胞或间充质干细胞群体,所述细胞或间充质干细胞群体是或者可以包含已传代了至少1、2、3、4、5、6、7、8、9、10、12、14、16、18或20代或更多代,或者扩增了1、2、3、4、5、6、7、8、9、10、12、14、16、18、20、22、24、26、28、30、32、34、36、38或40次群体倍增或更多次群体倍增的细胞。For any of the above mesenchymal stem cells or mesenchymal stem cell populations, the cells or mesenchymal stem cell populations are or can include cells that have been passaged for at least 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18 or 20 generations or more, or expanded for 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 12, 14, 16, 18, 20, 22, 24, 26, 28, 30, 32, 34, 36, 38 or 40 population doublings or more.
在优选的实施方案中,本发明的间充质干细胞不表达或实质上不表达被细胞疗法所特异性针对的抗原分子。如无另外说明,当本文提及间充质干细胞“不表达”和“实质上不表达”某种抗原时,其具有相同的含义,意指在某个指定的间充质干细胞群体中,只有极少数的细胞,例如,少于20%的细胞,少于19%的细胞,少于18%的细胞,少于17%的细胞,少于16%的细胞,少于15%的细胞,少于14%的细胞,少于13%的细胞,少于12%的细胞,少于11%的细胞,少于10%的细胞,少于9%的细胞,少于8%的细胞,少于7%的细胞,少于6%的细胞,少于5%的细胞,少于4%的细胞,少于3%的细胞,少于2%的细胞或少于1%的细胞表达该抗原至可被检测到的程度;或者,虽然该抗原在多于少数的细胞上表达,但表达量极低,以至于与被细胞正常表达的分子相比,或者与正常表达该抗原分子的细胞上的表达量相比,该表达量可忽略不计,甚至低于常规方法的检测下限。例如,在某些实施方案中,分离的间充质干细胞不表达或实质上不表达被免疫效应细胞(例如,CAR-T细胞)所特异性靶向的抗原。在一些实施方案中,分离的间充质干细胞不表达或实质上不表达CD19。In a preferred embodiment, the mesenchymal stem cells of the present invention do not express or substantially do not express the antigen molecules specifically targeted by cell therapy. Unless otherwise specified, when the mesenchymal stem cells are referred to herein as "not expressing" and "substantially not expressing" a certain antigen, they have the same meaning, meaning that in a certain specified mesenchymal stem cell population, only a very small number of cells, for example, less than 20% of cells, less than 19% of cells, less than 18% of cells, less than 17% of cells, less than 16% of cells, less than 15% of cells, less than 14% of cells, less than 13% of cells, less than 12% of cells, less than 11% of cells, less than 10% of cells, Less than 9% of cells, less than 8% of cells, less than 7% of cells, less than 6% of cells, less than 5% of cells, less than 4% of cells, less than 3% of cells, less than 2% of cells or less than 1% of cells express the antigen to a detectable degree; or, although the antigen is expressed on more than a few cells, the expression is extremely low, so that compared with the molecules normally expressed by the cells, or compared with the expression on cells that normally express the antigen molecule, the expression is negligible, or even below the detection limit of conventional methods. For example, in certain embodiments, the separated mesenchymal stem cells do not express or substantially do not express antigens specifically targeted by immune effector cells (e.g., CAR-T cells). In some embodiments, the separated mesenchymal stem cells do not express or substantially do not express CD19.
适合于本发明的胎盘间充质干细胞Placental mesenchymal stem cells suitable for the present invention
可以通过以下方法产生本文所述的分离的胎盘干细胞群体:用组织破坏酶消化胎盘组织以获得包含胎盘干细胞的胎盘细胞群体,和从所述胎盘细胞的剩余物中分离或基本分离多个胎盘干细胞。可以将全部或任何部分的胎盘消化以获得本文所述的分离的胎盘干细胞。在具体的实施方式中,例如,所述胎盘组织可以是整个胎盘(例如,包括脐带)、羊膜、绒毛膜、羊膜和绒毛膜的组合,或任何上述的组合。在其他具体的实施方式中,所述组织破坏酶是胰蛋白酶或胶原酶。在多种实施方式中,包含在得自胎盘消化的细胞群体内的所述分离的胎盘干细胞是所述胎盘细胞群体的至少50%、60%、70%、80%、90%、95%、99%或至少99.5%。The isolated placental stem cell population described herein can be produced by the following method: digesting placental tissue with a tissue-destroying enzyme to obtain a placental cell population containing placental stem cells, and isolating or substantially isolating a plurality of placental stem cells from the remainder of the placental cells. All or any portion of the placenta can be digested to obtain the isolated placental stem cells described herein. In specific embodiments, for example, the placental tissue can be a whole placenta (e.g., including the umbilical cord), amnion, chorion, a combination of amnion and chorion, or any combination of the above. In other specific embodiments, the tissue-destroying enzyme is trypsin or collagenase. In various embodiments, the isolated placental stem cells contained in the cell population obtained from placental digestion are at least 50%, 60%, 70%, 80%, 90%, 95%, 99% or at least 99.5% of the placental cell population.
其中,源自胎盘的间充质干细胞(也可称为胎盘源性间充质干细胞、胎盘衍生间充质干细胞、胎盘间充质干细胞、胎盘干细胞,上述术语在本文可互换地使用)在来源上可以是胎儿的或母体的(即,可以具有母体或胎儿的基因型)。胎盘干细胞群体或包含胎盘干细胞的细胞群体可以包含仅是胎儿或母体来源的胎盘干细胞,或者可以包含胎儿和母体来源的胎盘干细胞的混合群体。Among them, mesenchymal stem cells derived from the placenta (also referred to as placental-derived mesenchymal stem cells, placental-derived mesenchymal stem cells, placental mesenchymal stem cells, and the above terms are used interchangeably herein) can be fetal or maternal in origin (i.e., can have a maternal or fetal genotype). A placental stem cell population or a cell population comprising placental stem cells can include placental stem cells that are only of fetal or maternal origin, or can include a mixed population of placental stem cells of fetal and maternal origin.
本文所述的分离的间充质干细胞和/或胎盘细胞群体(例如,两种或更多种分离的胎盘干细胞)包括直接获自胎盘或其任何部分(例如,绒毛膜、胎盘绒毛叶等)的胎盘干细胞和含有胎盘细胞的细胞群体。分离的胎盘细胞群体还包括培养物中的(即两种或更多种)分离的胎盘干细胞群体和容器(例如,袋)中的细胞群体。The isolated mesenchymal stem cells and/or placental cell populations described herein (e.g., two or more isolated placental stem cells) include placental stem cells obtained directly from the placenta or any part thereof (e.g., chorion, placental villous leaves, etc.) and cell populations containing placental cells. Isolated placental cell populations also include (i.e., two or more) isolated placental stem cell populations in culture and cell populations in containers (e.g., bags).
适合于本发明的免疫效应细胞Immune effector cells suitable for the present invention
术语“免疫效应细胞”是来自人体的已分化成(即,不是造血干细胞)能够调节或影响免疫反应的形式或实现的细胞,例如B细胞、树突细胞、自然杀伤细胞和T细胞。本领域已知收集体内天然存在的免疫效应细胞,任选地并且是优选地在体外经过一定程序的处理之后,转化为治疗产品并施用于患者的治疗方法,即免疫效应细胞疗法。这些细胞疗法产品是癌症治疗新支柱——免疫疗法的重要部分,有效地利用患者自身的免疫系统来攻击肿瘤。免疫效应细胞疗法也可以被设计为促进免疫系统停止攻击自身(在存在自身免疫性疾病的情况下)。The term "immune effector cell" refers to cells that have differentiated (i.e., not hematopoietic stem cells) from the human body and are capable of regulating or influencing the form or realization of an immune response, such as B cells, dendritic cells, natural killer cells, and T cells. It is known in the art to collect naturally occurring immune effector cells in vivo, optionally and preferably after a certain procedure of treatment in vitro, to convert them into therapeutic products and apply them to patients, i.e., immune effector cell therapy. These cell therapy products are an important part of immunotherapy, a new pillar of cancer treatment, which effectively uses the patient's own immune system to attack tumors. Immune effector cell therapy can also be designed to promote the immune system to stop attacking itself (in the presence of autoimmune diseases).
CAR-T细胞疗法是近年来被研究最多的免疫效应细胞疗法。术语“CAR-T”或“CAR T”是指转导并表达了嵌合抗原受体(CAR)的T淋巴细胞。所述嵌合抗原受体是指一组或多组多肽,当其在免疫效应细胞上表达时,给所述的细胞提供针对靶抗原的特异性,并且具有细胞内信号产生。所述靶抗原被靶细胞(通常是癌细胞)表达。通常,CAR包括至少一个胞外结合区、跨膜区和胞内信号区。示例性的CAR构建方法和/或CAR-T细胞的转导方法记载于例如中国专利公开号CN114014941A之中。在一些具体的实施方案中,所述CAR-T细胞是根据第一代CAR-T技术构建的。在一些具体的实施方案中,所述CAR-T细胞是根据第二代CAR-T技术构建的。在一些具体的实施方案中,所述CAR-T细胞是根据第三代CAR-T技术构建的。在一些具体的实施方案中,所述CAR-T细胞是根据第四代CAR-T技术构建的。在一些具体的实施方案中,所述CAR分子包含胞外特异性靶抗原结合域,优选地,所述靶抗原结合域是scFv。优选地,所述靶抗原结合域是靶抗原的天然相互作用分子,例如但不限于,其配体(当靶抗原是受体时)或其可溶性受体片段(当靶抗原是配体时)。在一些实施方案中,靶抗原选自以下一种或多种:CD19、CD123、CD22、CD30、CD171、CS-1(也被称为CD2子集1、CRACC、SLAMF7、CD319、和19A24)、C型凝集素样分子-1(CLL-1或CLECL1)、CD33、表皮生长因子受体变体III(EGFRvIII)、神经节苷脂G2(GD2)、神经节苷脂GD3(aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、TNF受体家族成员B细胞成熟(BCMA)、Tn抗原((Tn Ag)或(GalNAcα-Ser/Thr))、前列腺特异性膜抗原(PSMA)、受体酪氨酸激酶样孤儿受体1(ROR1)、Fms样酪氨酸激酶3(FLT3)、肿瘤相关的糖蛋白72(TAG72)、CD38、CD44v6、癌胚抗原(CEA)、上皮细胞粘附分子(EPCAM)、B7H3(CD276)、KIT(CD117)、白介素13受体亚基α-2(IL-13Rα2或CD213A2)、间皮素、白介素11受体α(IL-11Ra)、前列腺干细胞抗原(PSCA)、蛋白酶丝氨酸21(Testisin或PRSS21)、血管内皮生长因子受体2(VEGFR2)、路易斯(Y)抗原、CD24、血小板衍生生长因子受体β(PDGFR-β)、阶段特异性胚胎抗原-4(SSEA-4)、CD20、叶酸受体α、受体酪氨酸蛋白激酶ERBB2(Her2/neu)、细胞表面相关的粘蛋白1(MUC1)、表皮生长因子受体(EGFR)、神经细胞粘附分子(NCAM)、Prostase、前列腺酸性磷酸酶(PAP)、突变的延伸因子2(ELF2M)、肝配蛋白B2、成纤维细胞活化蛋白α(FAP)、胰岛素样生长因子1受体(IGF-I受体)、碳酸酐酶IX(CAIX)、蛋白酶体(Prosome、Macropain)亚基、β型、9(LMP2)、糖蛋白100(gp100)、由断点簇区(BCR)和Alelson鼠白血病病毒癌基因同源物1(AB1)组成的癌基因融合蛋白(bcr- abl)、酪氨酸酶、肝配蛋白A型受体2(EphA2)、岩藻糖基GM1、唾液酸基路易斯粘附分子(sLe)、神经节苷脂GM3(aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、转谷氨酰胺酶5(TGS5)、高分子量黑素瘤相关抗原(HMWMAA)、邻乙酰基GD2神经节苷脂(OAcGD2)、叶酸受体β、肿瘤血管内皮标记1(TEM1/CD248)、肿瘤血管内皮标记7相关的(TEM7R)、Claudin6(CLDN6)、促甲状腺激素受体(TSHR)、G蛋白偶联受体C类5组、成员D(GPRC5D)、X染色体开放阅读框61(CXORF61)、CD97、CD179a、间变性淋巴瘤激酶(ALK)、聚唾液酸、胎盘特异性1(PLAC1)、globoH glycoceramide的己糖部分(GloboH)、乳腺分化抗原(NY-BR-1)、uroplakin 2(UPK2)、甲型肝炎病毒细胞受体1(HAVCR1)、肾上腺素受体β3(ADRB3)、pannexin 3(PANX3)、G蛋白偶联受体20(GPR20)、淋巴细胞抗原6复合物、基因座K9(LY6K)、嗅觉受体51E2(OR51E2)、TCRγ交替阅读框蛋白(TARP)、肾母细胞瘤蛋白(WT1)、癌/睾丸抗原1(NY-ESO-1)、癌症/睾丸抗原2(LAGE-1A)、黑素瘤相关抗原1(MAGE-A1)、ETS易位变异基因6,位于染色体12p(ETV6-AML)、精子蛋白17(SPA17)、X抗原家族,成员1A(XAGE1)、血管生成素结合细胞表面受体2(Tie2)、黑素瘤癌睾丸抗原-1(MAD-CT-1)、黑素瘤癌睾丸抗原-2(MAD-CT-2)、FOS相关抗原1、肿瘤蛋白质p53(p53)、p53突变体、prostein、存活蛋白、端粒酶、前列腺癌肿瘤抗原-1(PCTA-1或半乳凝素8)、由T细胞识别的黑素瘤抗原1(MelanA或MART1)、大鼠肉瘤(Ras)突变体、人端粒酶逆转录酶(hTERT)、肉瘤易位断点、细胞凋亡的黑素瘤抑制剂(ML-IAP)、ERG(跨膜蛋白酶,丝氨酸2(TMPRSS2)ETS融合基因)、N-乙酰葡糖胺基转移酶V(NA17)、配对盒蛋白Pax-3(PAX3)、雄激素受体、细胞周期蛋白B1、V-myc鸟髓细胞瘤病病毒癌基因神经母细胞瘤衍生的同源物(MYCN)、Ras同源物家族成员C(RhoC)、酪氨酸酶相关蛋白2(TRP-2)、细胞色素P450 1B1(CYP1B1)、CCCTC结合因子(锌指蛋白)样(BORIS或印记位点的调节物的兄弟)、由T细胞识别的鳞状细胞癌抗原3(SART3)、配对盒蛋白Pax-5(PAX5)、proacrosin结合蛋白sp32(OY-TES1)、淋巴细胞特异性蛋白酪氨酸激酶(LCK)、A激酶锚定蛋白4(AKAP-4)、滑膜肉瘤,X断点2(SSX2)、高级糖化终产物受体(RAGE-1)、肾uibiguitous 1(RU1)、肾uibiguitous 2(RU2)、legumain、人类乳头瘤病毒E6(HPV E6)、人类乳头瘤病毒E7(HPV E7)、肠羧基酯酶、突变的热休克蛋白70-2(mut hosp 70-2)、CD79a、CD79b、CD72、白细胞相关免疫球蛋白样受体1(LAIR1)、IgA受体的Fc片段(FCAR或CD89)、白细胞免疫球蛋白样受体亚家族A成员2(LILRA2)、CD300分子样家族成员f(CD300LF)、C型凝集素结构域家族12成员A(CLEC12A)、骨髓基质细胞抗原2(BST2)、含有EGF样模块粘蛋白样激素受体样2(EMR2)、淋巴细胞抗原75(LY75)、磷脂酰肌醇蛋白聚糖-3(GPC3)、Fc受体样5(FCRL5)、与免疫球蛋白λ样多肽1(IGLL1)。CAR-T cell therapy is the most studied immune effector cell therapy in recent years. The term "CAR-T" or "CAR T" refers to a T lymphocyte transduced with and expressing a chimeric antigen receptor (CAR). The chimeric antigen receptor refers to one or more sets of polypeptides that, when expressed on immune effector cells, provide the cells with specificity for a target antigen and have intracellular signal generation. The target antigen is expressed by a target cell (usually a cancer cell). Typically, CAR includes at least one extracellular binding region, a transmembrane region, and an intracellular signal region. Exemplary CAR construction methods and/or CAR-T cell transduction methods are described in, for example, Chinese Patent Publication No. CN114014941A. In some specific embodiments, the CAR-T cell is constructed according to the first generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the second generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the third generation CAR-T technology. In some specific embodiments, the CAR-T cell is constructed according to the fourth generation CAR-T technology. In some specific embodiments, the CAR molecule comprises an extracellular specific target antigen binding domain, preferably, the target antigen binding domain is a scFv. Preferably, the target antigen binding domain is a natural interaction molecule of the target antigen, such as, but not limited to, its ligand (when the target antigen is a receptor) or its soluble receptor fragment (when the target antigen is a ligand). In some embodiments, the target antigen is selected from one or more of the following: CD19, CD123, CD22, CD30, CD171, CS-1 (also known as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24), C-type lectin-like molecule-1 (CLL-1 or CLECL1), CD33, epidermal growth factor receptor variant III (EGFRvIII), ganglioside G2 (GD2), ganglioside GD3 (aNeu5Ac (2-8) aNeu5Ac (2-3) bDGalp (1-4) bDGlcp (1-1) Cer), TNF receptor family member B cell maturation (BCMA), Tn antigen ((Tn Ag) or (GalNAcα-Ser/Thr)), prostate specific membrane antigen (PSMA), receptor tyrosine kinase-like orphan receptor 1 (ROR1), Fms-like tyrosine kinase 3 (FLT3), tumor-associated glycoprotein 72 (TAG72), CD38, CD44v6, carcinoembryonic antigen (CEA), epithelial cell adhesion molecule (EPCAM), B7H3 (CD276), KIT (CD117), interleukin 13 receptor subunit α-2 (IL-13Rα2 or CD 213A2), mesothelin, interleukin 11 receptor alpha (IL-11Ra), prostate stem cell antigen (PSCA), testisin or PRSS21, vascular endothelial growth factor receptor 2 (VEGFR2), Lewis (Y) antigen, CD24, platelet-derived growth factor receptor beta (PDGFR-β), stage-specific embryonic antigen-4 (SSEA-4), CD20, folate receptor alpha, receptor tyrosine protein kinase ERBB2 (Her2/neu), cell surface-associated mucin 1 (MUC1), epidermal growth factor receptor (EGFR) , neural cell adhesion molecule (NCAM), prostase, prostatic acid phosphatase (PAP), mutated elongation factor 2 (ELF2M), ephrin B2, fibroblast activation protein alpha (FAP), insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX), proteasome (Prosome, Macropain) subunit, beta type, 9 (LMP2), glycoprotein 100 (gp100), oncogene fusion protein composed of breakpoint cluster region (BCR) and Alelson murine leukemia virus oncogene homolog 1 (AB1) (bcr- abl), tyrosinase, ephrin type A receptor 2 (EphA2), fucosyl GM1, sialyl Lewis adhesion molecule (sLe), ganglioside GM3 (aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer), transglutaminase 5 (TGS5), high molecular weight melanoma-associated antigen (HMWMAA), o-acetyl GD2 ganglioside (OAcGD2), folate receptor β, tumor endothelial marker 1 (TEM1/CD248), tumor endothelial marker 7-related (T7-related EM7R), Claudin6 (CLDN6), thyroid stimulating hormone receptor (TSHR), G protein-coupled receptor class C group 5, member D (GPRC5D), X chromosome open reading frame 61 (CXORF61), CD97, CD179a, anaplastic lymphoma kinase (ALK), polysialic acid, placenta-specific 1 (PLAC1), hexose portion of globoH glycoceramide (GloboH), breast differentiation antigen (NY-BR-1), uroplakin 2 (UPK2), hepatitis A HAVCR1, adrenergic receptor β3 (ADRB3), pannexin 3 (PANX3), G protein-coupled receptor 20 (GPR20), lymphocyte antigen 6 complex, locus K9 (LY6K), olfactory receptor 51E2 (OR51E2), TCRγ alternate reading frame protein (TARP), nephroblastoma protein (WT1), cancer/testis antigen 1 (NY-ESO-1), cancer/testis antigen 2 (LAGE-1A), melanoma associated antigen 1 (MAGE-A1), ETS-prone ETV6-AML, sperm protein 17 (SPA17), X-antigen family, member 1A (XAGE1), angiopoietin binding cell surface receptor 2 (Tie2), melanoma cancer testis antigen-1 (MAD-CT-1), melanoma cancer testis antigen-2 (MAD-CT-2), FOS-related antigen 1, tumor protein p53 (p53), p53 mutant, prostein, survivin, telomerase, prostate cancer tumor antigen-1 (PCTA-1 or galectin 8), T cell Recognized melanoma antigen 1 (MelanA or MART1), rat sarcoma (Ras) mutant, human telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, melanoma inhibitor of apoptosis (ML-IAP), ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene), N-acetylglucosaminyltransferase V (NA17), paired box protein Pax-3 (PAX3), androgen receptor, cyclin B1, V-myc avian myelocytic virus oncogene neuroblastoma-derived homolog (MYCN) , Ras homolog family member C (RhoC), tyrosinase-related protein 2 (TRP-2), cytochrome P450 1B1 (CYP1B1), CCCTC binding factor (zinc finger protein)-like (brother of regulator of BORIS or imprinting sites), squamous cell carcinoma antigen recognized by T cells 3 (SART3), paired box protein Pax-5 (PAX5), proacrosin binding protein sp32 (OY-TES1), lymphocyte-specific protein tyrosine kinase (LCK), A kinase anchoring protein 4 (AKAP-4), Synovial sarcoma, SSX2, RAGE-1, RU1, RU2, legumain, HPV E6, HPV E7, intestinal carboxylesterase, mutated heat shock protein 70-2, CD79a, CD79b, CD72, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), IgA receptor Fc fragment (FCAR or CD89), leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2), CD300 molecule-like family member f (CD300LF), C-type lectin domain family 12 member A (CLEC12A), bone marrow stromal cell antigen 2 (BST2), mucin-like hormone receptor-like 2 (EMR2) containing an EGF-like module, lymphocyte antigen 75 (LY75), phosphatidylinositol proteoglycan-3 (GPC3), Fc receptor-like 5 (FCRL5), and immunoglobulin lambda-like polypeptide 1 (IGLL1).
在一些实施方案中,被编码的CAR分子结合的肿瘤抗原选自以下一种或多种:TSHR、CD171、CS-1、CLL-1、GD3、Tn Ag、FLT3、CD38、CD44v6、B7H3、KIT、IL-13Rα2、IL-11Ra、PSCA、PRSS21、VEGFR2、LewisY、CD24、PDGFR-β,SSEA-4、MUC1、EGFR、NCAM、CAIX、LMP2、EphA2、岩藻糖基GM1,SLE、GM3、TGS5、HMWMAA、邻乙酰基GD2、叶酸受体β、TEM1/CD248、TEM7R、CLDN6、GPRC5D、CXORF61、CD97、 CD179a、ALK、聚唾液酸,PLAC1、GloboH、NY-BR-1、UPK2、HAVCR1、ADRB3、PANX3、GPR20、LY6K、OR51E2、TARP、WT1、ETV6-AML、精子蛋白17,XAGE1、Tie 2、MAD-CT-1、MAD-CT-2、Fos相关抗原1、p53突变体、hTERT、肉瘤易位断点、ML-IAP,ERG(TMPRSS2ETS融合基因)、NA17,PAX3,雄激素受体,细胞周期蛋白B1,MYCN、RhoC、CYP1B1、BORIS、SART3、PAX5、OY-TES1、LCK、AKAP-4、SSX2、CD79a、CD79b、CD72、LAIR1、FCAR、LILRA2、CD300LF、CLEC12A、BST2、EMR2、LY75、GPC3、FCRL5、和IGLL1。In some embodiments, the tumor antigen bound by the encoded CAR molecule is selected from one or more of the following: TSHR, CD171, CS-1, CLL-1, GD3, Tn Ag, FLT3, CD38, CD44v6, B7H3, KIT, IL-13Rα2, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-β, SSEA-4, MUC1, EGFR, NCAM, CAIX, LMP2, EphA2, fucosyl GM1, SLE, GM3, TGS5, HMWMAA, o-acetyl GD2, folate receptor β, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53 mutant, hTERT, sarcoma translocation breakpoint, ML-IAP, ERG (TMPR SS2ETS fusion gene), NA17, PAX3, androgen receptor, cyclin B1, MYCN, RhoC, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
在某些实施方案中,被编码的CAR分子结合的肿瘤抗原选自以下一种或多种:TSHR、CLDN6、GPRC5D、CXORF61、CD97、CD179a、ALK、聚唾液酸、PLAC1、GloboH、NY-BR-1、UPK2、HAVCR1、ADRB3、PANX3、GPR20、LY6K、和OR51E2。In certain embodiments, the tumor antigen bound by the encoded CAR molecule is selected from one or more of the following: TSHR, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADRB3, PANX3, GPR20, LY6K, and OR51E2.
在一些具体的实施方案中,所述CAR-T细胞的靶抗原选自CD19、IL-13Rα2、EphA2和EGFRvIII。In some specific embodiments, the target antigen of the CAR-T cell is selected from CD19, IL-13Rα2, EphA2 and EGFRvIII.
在一个具体的实施方案中,所述CAR-T细胞的靶抗原是CD19,此类CAR-T细胞的实例如Ying Zhang等,Co-expression IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy.J Transl Med.2022 Sep27;20(1):432.中所描述。In a specific embodiment, the target antigen of the CAR-T cell is CD19. Examples of such CAR-T cells are described in Ying Zhang et al., Co-expression IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy. J Transl Med. 2022 Sep27; 20(1):432.
在一个具体的实施方案中,所述CAR-T细胞的靶抗原是IL-13Rα2,此类CAR-T细胞的实例如中国专利申请号202210019437.4中所描述。In a specific embodiment, the target antigen of the CAR-T cell is IL-13Rα2. Examples of such CAR-T cells are described in Chinese Patent Application No. 202210019437.4.
在一个具体的实施方案中,所述CAR-T细胞的靶抗原是EphA2,此类CAR-T细胞的实例如中国专利申请号202110919075.X和An Z等,Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1.Oncoimmunology.2021 Aug 16;10(1):1960728.中所描述。In a specific embodiment, the target antigen of the CAR-T cell is EphA2. Examples of such CAR-T cells are described in Chinese patent applications No. 202110919075.X and An Z, etc., Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1.Oncoimmunology.2021 Aug 16;10(1):1960728.
在一个具体的实施方案中,所述CAR-T细胞的靶抗原是EGFRvIII,此类CAR-T细胞的实例如中国专利申请号202211140506.3中所描述。In a specific embodiment, the target antigen of the CAR-T cell is EGFRvIII. Examples of such CAR-T cells are described in Chinese Patent Application No. 202211140506.3.
在一些具体的实施方案中,所述CAR分子包含选自以下的跨膜结构域:T细胞受体的α、β、或ζ的跨膜结构域、CD28、CD3ε、CD45、CD4、CD5、CD8、CD9、CD16、CD22、CD33、CD37、CD64、CD80、CD86、CD134、CD137、CD154、KIRDS2、OX40、CD2、CD27、LFA-1(CD11a、CD18)、ICOS(CD278)、4-1BB(CD137)、GITR、CD40、BAFFR、HVEM(LIGHTR)、SLAMF7、NKp80(KLRF1)、CD160、CD19、IL2Rβ、IL2Rγ、IL7Rα、ITGA1、VLA1、CD49a、ITGA4、IA4、CD49D、ITGA6、VLA-6、CD49f、ITGAD、CD11d、ITGAE、CD103、ITGAL、CD11a、LFA-1、ITGAM、CD11b、ITGAX、CD11c、ITGB1、CD29、ITGB2、CD18、LFA-1、ITGB7、TNFR2、DNAM1(CD226)、SLAMF4(CD244、2B4)、CD84、CD96(Tactile)、CEACAM1、CRTAM、Ly9(CD229)、CD160(BY55)、PSGL1、CD100(SEMA4D)、SLAMF6(NTB-A、Ly108)、SLAM(SLAMF1、CD150、IPO-3)、BLAME(SLAMF8)、SELPLG(CD162)、LTBR、PAG/Cbp、NKp44、NKp30、NKp46、NKG2D、和/或NKG2C的跨膜结构域。优选地,所述跨膜结构域是CD28跨膜结构域或CD8跨膜结构 域或CD4跨膜结构域。In some specific embodiments, the CAR molecule comprises a transmembrane domain selected from the group consisting of: α, β, or ζ transmembrane domains of a T cell receptor, CD28, CD3ε, CD45, CD4, CD5, CD8, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD154, KIRDS2, OX40, CD2, CD27, LFA-1 (CD11a, CD18), ICOS (CD278), 4-1BB (CD137), GITR, CD40, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, IL2Rβ, IL2Rγ, IL7Rα, ITGA1, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD , CD11d, ITGAE, CD103, ITGAL, CD11a, LFA-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 (SEMA4D), SLAMF6 (NTB-A, Ly108), SLAM (SLAMF1, CD150, IPO-3), BLAME (SLAMF8), SELPLG (CD162), LTBR, PAG/Cbp, NKp44, NKp30, NKp46, NKG2D, and/or NKG2C transmembrane domain. Preferably, the transmembrane domain is a CD28 transmembrane domain or a CD8 transmembrane domain or a CD4 transmembrane domain.
在一些实施方案中,CAR分子的所述胞外靶抗原结合域由铰链区连接到所述跨膜结构域。在一个实施方案中,铰链区包含CD8铰链的氨基酸序列。在另一个实施方案中,铰链区包含IgG4铰链的氨基酸序列。In some embodiments, the extracellular target antigen binding domain of the CAR molecule is connected to the transmembrane domain by a hinge region. In one embodiment, the hinge region comprises the amino acid sequence of a CD8 hinge. In another embodiment, the hinge region comprises the amino acid sequence of an IgG4 hinge.
在一些具体的实施方案中,所述CAR分子包含胞内段。在一些具体的实施方案中,所述胞内段包含共刺激信号结构域,所述共刺激信号传导结构域包含选自如下一种或多种的蛋白质的功能信号传导结构域:CD27、CD28、4-1BB(CD137)、OX40、CD30、CD40、PD-1、ICOS,淋巴细胞功能相关抗原-1(LFA-1)、CD2、CD7、LIGHT、NKG2C、B7-H3、特异性结合CD83的配体、CDS、ICAM-1、GITR、BAFFR、HVEM(LIGHTR)、SLAMF7、NKp80(KLRF1)、CD160、CD19、CD4、CD8α、CD8β、IL2Rβ、IL2Rγ、IL7Rα、ITGA4、VLA1、CD49a、ITGA4、IA4、CD49D、ITGA6、VLA-6、CD49f、ITGAD、CD11d、ITGAE、CD103、ITGAL、CD11a、LFA-1、ITGAM、CD11b、ITGAX、CD11c、ITGB1、CD29、ITGB2、CD18、LFA-1、ITGB7、TNFR2、TRANCE/RANKL、DNAM1(CD226)、SLAMF4(CD244、2B4)、CD84、CD96(Tactile)、CEACAM1、CRTAM、Ly9(CD229)、CD160(BY55)、PSGL1、CD100(SEMA4D)、CD69、SLAMF6(NTB-A、Ly108)、SLAM(SLAMF1、CD150、IPO-3)、BLAME(SLAMF8)、SELPLG(CD162)、LTBR、LAT、GADS、SLP-76、PAG/Cbp、NKp44、NKp30、NKp46、或NKG2D,优选地,所述共刺激信号结构域是CD28共刺激结构域和/或4-1BB共刺激结构域。In some specific embodiments, the CAR molecule comprises an intracellular segment. In some specific embodiments, the intracellular segment comprises a costimulatory signaling domain, and the costimulatory signaling domain comprises a functional signaling domain of a protein selected from one or more of the following: CD27, CD28, 4-1BB (CD137), OX40, CD30, CD40, PD-1, ICOS, lymphocyte function-associated antigen-1 (LFA-1), CD2, CD7, LIGHT, NKG2C, B7-H3, a ligand specifically binding to CD83, CDS, ICA M-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), CD160, CD19, CD4, CD8α, CD8β, IL2Rβ, IL2Rγ, IL7Rα, ITGA4, VLA1, CD49a, ITGA4, IA4, CD49D, ITGA6, VLA-6, CD49f, ITGAD, CD11d, ITGAE, CD103, ITGAL, CD11a, LF A-1, ITGAM, CD11b, ITGAX, CD11c, ITGB1, CD29, ITGB2, CD18, LFA-1, ITGB7, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD229), CD160 (BY55), PSGL1, CD100 ( Preferably, the co-stimulatory signaling domain is a CD28 co-stimulatory domain and/or a 4-1BB co-stimulatory domain.
在一些具体的实施方案中,所述胞内段包含选自CD3ζ、CD3γ、CD3δ、CD3ε、常见FcRγ(FCER1G)、FcRβ(FcεR1b)、CD79a、CD79b、FcγRIIa、DAP10和DAP12的信号传导结构域的刺激信号结构域,优选地,所述刺激信号结构域是CD3ζ信号传导结构域。在一些具体的实施方案中,所述胞内段进一步包含IL15序列。In some specific embodiments, the intracellular segment comprises a stimulatory signaling domain selected from the signaling domains of CD3ζ, CD3γ, CD3δ, CD3ε, common FcRγ (FCER1G), FcRβ (FcεR1b), CD79a, CD79b, FcγRIIa, DAP10 and DAP12, preferably, the stimulatory signaling domain is a CD3ζ signaling domain. In some specific embodiments, the intracellular segment further comprises an IL15 sequence.
在一些实施方案中,所述CAR-T细胞除了外源引入的CAR分子之外,还任选地包含至少另一种修饰,例如通过与CAR分子相同/不同载体引入的其他外源基因,和/或对基因组原有序列的修饰,例如通过基因编辑技术等。In some embodiments, the CAR-T cell optionally contains at least one other modification in addition to the exogenously introduced CAR molecule, such as other exogenous genes introduced through the same/different vector as the CAR molecule, and/or modification of the original genomic sequence, such as through gene editing technology, etc.
在一些实施方案中,本发明的胎盘间充质干细胞增强了CD19-IL15 CAR-T细胞和IL13 CAR-T细胞的抗肿瘤功能,但抑制了uPAR CAR-T细胞的抗肿瘤功能。在一些实施方案中,间充质干细胞促进CD19-IL15 CAR-T细胞的增殖和激活,并促进细胞因子IL2和IL4的释放。IL-2参与塑造决定T细胞命运的转录和代谢过程。它是T细胞激活和增殖的重要细胞因子,被认为是治疗癌症的一种手段。而IL-4通常被认为是一种典型的Th2细胞因子,但有报道称它能促进人类胸腺和脐带血中的CD4 +T细胞转化为CD8 +T细胞,并促进记忆性CD8+T细胞的频率和功能,从而促进而不是削弱Th1细胞免疫反应。在一些实施方案中,间充质干细胞促进了CD3 +T细胞中TCM表型细胞的比例增加,包括CD4 +和CD8 +T细胞。在一个实施方案中,间充质干细胞显著地上调了与T细胞分化有关、参与TCM表型细胞分化和发展的转录因子TCF-7的表达。本领域公知的是,具有较少分化表型的CAR-T细胞,如TN和TCM表型,与自我更新、增殖和存活的特性增加有关。因此此类细胞增多 /CAR-T细胞倾向于分化为此类细胞,将使得CAR-T细胞或细胞群的体内活性增强。 In some embodiments, the placental mesenchymal stem cells of the present invention enhance the anti-tumor function of CD19-IL15 CAR-T cells and IL13 CAR-T cells, but inhibit the anti-tumor function of uPAR CAR-T cells. In some embodiments, mesenchymal stem cells promote the proliferation and activation of CD19-IL15 CAR-T cells and promote the release of cytokines IL2 and IL4. IL-2 is involved in shaping the transcriptional and metabolic processes that determine the fate of T cells. It is an important cytokine for T cell activation and proliferation and is considered a means of treating cancer. While IL-4 is generally considered a typical Th2 cytokine, it has been reported that it can promote the conversion of CD4 + T cells in human thymus and umbilical cord blood into CD8 + T cells, and promote the frequency and function of memory CD8+T cells, thereby promoting rather than weakening Th1 cell immune responses. In some embodiments, mesenchymal stem cells promote an increase in the proportion of TCM phenotype cells in CD3 + T cells, including CD4 + and CD8 + T cells. In one embodiment, mesenchymal stem cells significantly upregulate the expression of TCF-7, a transcription factor associated with T cell differentiation and involved in the differentiation and development of TCM phenotype cells. It is well known in the art that CAR-T cells with less differentiated phenotypes, such as TN and TCM phenotypes, are associated with increased characteristics of self-renewal, proliferation, and survival. Therefore, the increase in such cells/CAR-T cells tend to differentiate into such cells, which will enhance the in vivo activity of CAR-T cells or cell populations.
术语“肿瘤”和“癌症”在本文中并不互相排斥且可互换地使用,涵盖实体瘤和血液肿瘤,指所有赘生性(neoplastic)细胞生长和增殖,无论是恶性的还是良性的,及所有癌前(pre-cancerous)和癌性细胞和组织。在某些实施方案中,适合于通过本发明的方法来治疗的癌症包括血液癌症,例如,选自慢性淋巴细胞白血病(CLL),急性白血病,急性淋巴性白血病(ALL),B细胞急性淋巴性白血病(B-ALL),T-细胞急性淋巴性白血病(T-ALL),慢性骨髓性白血病(CML),B细胞幼淋巴细胞白血病,母细胞性浆细胞树突状细胞瘤,伯基特淋巴瘤,弥散性大B细胞淋巴瘤,滤泡型淋巴瘤,多毛细胞白血病,小细胞或大细胞-滤泡型淋巴瘤,恶性淋巴组织增生状况,MALT淋巴瘤,套细胞淋巴瘤,边缘区淋巴瘤,多发性骨髓瘤,脊髓发育不良和骨髓增生异常综合征,非霍奇金淋巴瘤如Burkitt淋巴瘤等,霍奇金淋巴瘤,浆母细胞性淋巴瘤,浆细胞样树突细胞瘤,瓦尔登斯特伦巨球蛋白血症,或白血病前期的一种或多种的癌症。癌症也可以包括实体瘤,例如选自结肠癌,直肠癌,肾细胞癌,肝癌,肺的非小细胞癌,小肠癌,食道癌,黑素瘤,骨癌,胰腺癌,皮肤癌,头或颈癌,皮肤或眼内恶性黑素瘤,子宫癌,卵巢癌,直肠癌,肛区癌,胃癌,睾丸癌,子宫癌,输卵管癌,子宫内膜癌,宫颈癌,阴道癌,霍奇金淋巴瘤,非霍奇金淋巴瘤,内分泌系统癌,甲状腺癌,甲状旁腺癌,肾上腺癌,软组织肉瘤,尿道癌,阴茎癌,儿童实体瘤,膀胱癌,肾或输尿管癌,肾盂癌,中枢神经系统瘤(CNS),原发性CNS淋巴瘤,肿瘤血管发生,脊椎肿瘤,脑干神经胶质瘤,垂体腺瘤,卡波西肉瘤,表皮样癌,鳞状细胞癌,T细胞淋巴瘤,环境诱发的癌症,所述癌症的组合和所述癌症的转移性形式。The terms "tumor" and "cancer" are not mutually exclusive and are used interchangeably herein, covering solid tumors and blood tumors, referring to all neoplastic cell growth and proliferation, whether malignant or benign, and all precancerous and cancerous cells and tissues. In certain embodiments, cancers suitable for treatment by the methods of the present invention include blood cancers, for example, selected from chronic lymphocytic leukemia (CLL), acute leukemia, acute lymphocytic leukemia (ALL), B-cell acute lymphocytic leukemia (B-ALL), T-cell acute lymphocytic leukemia (T-ALL), chronic myeloid leukemia (CML), B-cell prolymphocytic leukemia, blastic plasmacytoid dendritic cell neoplasm, Burkitt's lymphoma, diffuse large B-cell Lymphoma, follicular lymphoma, hairy cell leukemia, small cell or large cell-follicular lymphoma, malignant lymphoproliferative conditions, MALT lymphoma, mantle cell lymphoma, marginal zone lymphoma, multiple myeloma, myelodysplasia and myelodysplastic syndrome, non-Hodgkin lymphoma such as Burkitt lymphoma, Hodgkin lymphoma, plasmablastic lymphoma, plasmacytoid dendritic cell neoplasm, Waldenstrom's macroglobulinemia, or one or more of the following: Cancer can also include solid tumors, for example selected from the group consisting of colon cancer, rectal cancer, renal cell carcinoma, liver cancer, non-small cell carcinoma of the lung, small intestine cancer, esophageal cancer, melanoma, bone cancer, pancreatic cancer, skin cancer, cancer of the head or neck, cutaneous or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal region, stomach cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer, cervical cancer, vaginal cancer, Hodgkin lymphoma, non-Hodgkin lymphoma, cancer of the endocrine system, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, solid tumors in children, bladder cancer, cancer of the kidney or ureter, renal pelvis cancer, central nervous system tumors (CNS), primary CNS lymphoma, tumor angiogenesis, spinal tumors, brain stem gliomas, pituitary adenomas, Kaposi's sarcoma, epidermoid carcinoma, squamous cell carcinoma, T-cell lymphoma, environmentally induced cancers, combinations of said cancers and metastatic forms of said cancers.
术语“药物组合物”是指,本发明中的间充质干细胞作为有效成分与可药用载体(根据情况而定)的混合物。药物组合物有助于有效成分向患者的施用。The term "pharmaceutical composition" refers to a mixture of mesenchymal stem cells as an active ingredient and a pharmaceutically acceptable carrier (as appropriate). The pharmaceutical composition facilitates administration of the active ingredient to a patient.
如本文所用,术语“可药用载体”是指药学上可接受的材料、组合物或载体,例如液体或固体填充剂,稳定剂,分散剂,悬浮剂,稀释剂,赋形剂,增稠剂,溶剂或包封材料,涉及在患者体内或向患者体内携带或运输本发明有用的化合物,使其可以发挥其预期的功能。通常,这种构建物可以被携带至、或从一个器官或身体的一部分运输至另一个器官或身体的一部分。在与制剂的其他成分(包括本发明所述的麝香提取物)相容且对患者无害的意义上,每种载体必须是“可接受的”。可用作药学上可接受的载体的材料的一些实例包括:糖,例如乳糖,葡萄糖和蔗糖;淀粉,如玉米淀粉和马铃薯淀粉;纤维素及其衍生物,如羧甲基纤维素钠,乙基纤维素和醋酸纤维素;粉末黄蓍胶;麦芽;明胶;滑石;赋形剂,如可可脂和栓剂蜡;油,如花生油,棉籽油,红花油,芝麻油,橄榄油,玉米油和豆油;二醇,如丙二醇;多元醇,如甘油,山梨糖醇,甘露糖醇和聚乙二醇;酯类,如油酸乙酯和月桂酸乙酯;琼脂;缓冲剂,如氢氧化镁和氢氧化铝;表面活性剂;海藻酸;无热原水;等渗盐水;林格氏溶液;乙醇;磷酸盐缓冲液;和药物制剂中使用的其他无毒相容物质。如本文所用,“可药用载体”还包括与本发明化合物的活性相容的且对于患者是生理学上可接受的任何和所有包衣、抗细菌和抗真菌剂、和吸收延迟剂等。增补性活性化合物也可以掺入组合物中。“可药用载体”可以进一步包括可用于本发明的化合物的药学上可接受盐。可以包 括在本发明的药物组合物中的其他成分是本领域已知的并且描述于例如Remington”s Pharmaceutical Sciences(Genaro,Ed。,Mack Publishing Co.,1985,Easton,PA),其通过引用并入本文。As used herein, the term "pharmaceutically acceptable carrier" refers to a pharmaceutically acceptable material, composition or carrier, such as a liquid or solid filler, stabilizer, dispersant, suspending agent, diluent, excipient, thickener, solvent or encapsulating material, which is involved in carrying or transporting the useful compounds of the present invention in or to the patient's body so that it can perform its intended function. Typically, such a construct can be carried to or transported from one organ or part of the body to another organ or part of the body. Each carrier must be "acceptable" in the sense of being compatible with the other ingredients of the formulation (including the musk extract described in the present invention) and not harmful to the patient. Some examples of materials that can be used as pharmaceutically acceptable carriers include: sugars, such as lactose, glucose and sucrose; starches, such as corn starch and potato starch; cellulose and its derivatives, such as sodium carboxymethylcellulose, ethylcellulose and cellulose acetate; powdered tragacanth; malt; gelatin; talc; excipients, such as cocoa butter and suppository waxes; oils, such as peanut oil, cottonseed oil, safflower oil, sesame oil, olive oil, corn oil and soybean oil; glycols, such as propylene glycol; polyols, such as glycerol, sorbitol, mannitol and polyethylene glycol; esters, such as ethyl oleate and ethyl laurate; agar; buffers, such as magnesium hydroxide and aluminum hydroxide; surfactants; alginic acid; pyrogen-free water; isotonic saline; Ringer's solution; ethanol; phosphate buffer; and other nontoxic compatible substances used in pharmaceutical formulations. As used herein, "pharmaceutically acceptable carriers" also include any and all coatings, antibacterial and antifungal agents, and absorption delaying agents that are compatible with the activity of the compounds of the invention and are physiologically acceptable to patients. Supplementary active compounds may also be incorporated into the composition. "Pharmaceutically acceptable carriers" may further include pharmaceutically acceptable salts of the compounds useful in the present invention. Other ingredients that may be included in the pharmaceutical compositions of the present invention are known in the art and are described, for example, in Remington"s Pharmaceutical Sciences (Genaro, Ed., Mack Publishing Co., 1985, Easton, PA), which is incorporated herein by reference.
在一些实施方案中,配制药物组合物用于静脉内注射。在一些实施方案中,药物组合物包含配制用于静脉内注射的间充质干细胞。在一些实施方案中,药物组合物包含配制用于静脉内注射的间充质干细胞和CAR-T细胞。适于注射使用的药物组合物包括无菌水溶液(水溶性的)或分散体、和用于即时制备无菌可注射溶液或分散体的无菌粉末。对于静脉内施用,合适的载体包括生理盐水,抑菌水,或达到易于注射程度的流体。可注射组合物在制造和储存条件下必须是稳定的,并且必须防止微生物如细菌和真菌的污染。载体可以是溶剂或分散介质,含有例如水,乙醇,多元醇(例如,甘油,丙二醇和液体聚乙二醇等),以及它们的合适混合物。例如,可以通过使用诸如卵磷脂的涂层、在分散体的情况下通过维持所需的颗粒尺寸、和通过使用表面活性剂,来保持适当的流动性。通过各种抗细菌剂和抗真菌剂,例如对羟基苯甲酸酯,氯丁醇,苯酚,抗坏血酸,硫柳汞等,可以防止微生物的作用。在许多情况下,优选在组合物中包含等渗剂,例如糖,多元醇如甘露醇,山梨糖醇,氯化钠。通过在组合物中包括延迟吸收的试剂,例如单硬脂酸铝和明胶,可以实现可注射组合物的延长吸收。In some embodiments, the pharmaceutical composition is prepared for intravenous injection. In some embodiments, the pharmaceutical composition includes mesenchymal stem cells prepared for intravenous injection. In some embodiments, the pharmaceutical composition includes mesenchymal stem cells and CAR-T cells prepared for intravenous injection. Pharmaceutical compositions suitable for injection include sterile aqueous solutions (water-soluble) or dispersions and sterile powders for immediate preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers include physiological saline, antibacterial water, or fluids that are easy to inject. Injectable compositions must be stable under manufacturing and storage conditions and must prevent contamination by microorganisms such as bacteria and fungi. The carrier can be a solvent or a dispersion medium, containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol and liquid polyethylene glycol, etc.), and suitable mixtures thereof. For example, appropriate fluidity can be maintained by using a coating such as lecithin, by maintaining the desired particle size in the case of a dispersion, and by using a surfactant. The action of microorganisms can be prevented by various antibacterial and antifungal agents, such as parabens, chlorobutanol, phenol, ascorbic acid, thimerosal, etc. In many cases, it will be preferable to include isotonic agents, for example, sugars, polyalcohols such as mannitol, sorbitol, sodium chloride in the composition. Prolonged absorption of the injectable compositions can be brought about by including in the composition an agent which delays absorption, for example, aluminum monostearate and gelatin.
在一些实施方案中,配制药物组合物适用于在将其中的治疗性细胞用于静脉内注射之前,与本发明的间充质干细胞(例如,优选,胎盘干细胞)共培养,使其治疗效应在体外即得到增强,并将此状态延续至施用于体内后。因此,所述药物组合物中包含间充质干细胞,治疗性细胞(例如,CAR-T细胞),以及适用于两者共培养的培养基,以及可选地,适用于两者共培养的培养容器。In some embodiments, the pharmaceutical composition is prepared for co-culturing with the mesenchymal stem cells of the present invention (e.g., preferably, placental stem cells) before the therapeutic cells therein are used for intravenous injection, so that the therapeutic effect is enhanced in vitro, and this state is continued after administration in vivo. Therefore, the pharmaceutical composition contains mesenchymal stem cells, therapeutic cells (e.g., CAR-T cells), and a culture medium suitable for co-culturing the two, and optionally, a culture container suitable for co-culturing the two.
在一些实施方案中,配制药物组合物适用于对来自受试者自身的细胞进行修饰(例如,转基因修饰),并在修饰之后,进行必要的扩增培养和与本发明的间充质干细胞(例如,优选,胎盘干细胞)的共培养,使其治疗效应在体外即得到增强,并将此状态延续至施用于体内后。因此,所述药物组合物中包含间充质干细胞,对来自受试者自身的细胞进行修饰的必需的试剂(例如,转染试剂,转基因构建体,优选地,病毒载体构建体),以及合适的培养基,以及可选地,适用于两者共培养的培养容器。In some embodiments, the pharmaceutical composition is suitable for modifying cells from the subject itself (e.g., transgenic modification), and after modification, necessary expansion culture and co-culture with the mesenchymal stem cells of the present invention (e.g., preferably, placental stem cells) are performed, so that the therapeutic effect is enhanced in vitro, and this state is continued after administration in vivo. Therefore, the pharmaceutical composition contains mesenchymal stem cells, necessary reagents for modifying cells from the subject itself (e.g., transfection reagents, transgenic constructs, preferably, viral vector constructs), and suitable culture media, and optionally, a culture container suitable for co-culture of the two.
用于本文时,“治疗”指减缓、中断、阻滞、缓解、停止、降低、或逆转已存在的症状、病症、病况或疾病的进展或严重性。对癌症/肿瘤的治疗效果通常包括但不限于例如肿瘤体积减小、癌细胞数量减少、转移数量减少、预期寿命增加、癌细胞增殖减少、癌细胞存活率降低或与癌性病症有关的各种生理学症状的改善。As used herein, "treat" refers to slowing down, interrupting, blocking, alleviating, stopping, reducing, or reversing the progression or severity of existing symptoms, disorders, conditions, or diseases. The therapeutic effects on cancer/tumors generally include, but are not limited to, for example, reduction in tumor volume, reduction in the number of cancer cells, reduction in the number of metastases, increase in life expectancy, reduction in cancer cell proliferation, reduction in cancer cell survival, or improvement in various physiological symptoms associated with cancerous disorders.
用于本文时,“预防”包括对疾病或病症或特定疾病或病症的症状的发生或发展的抑制。在一些实施方案中,具有癌症家族病史的受试者是预防性方案的候选。通常,在癌症的背景中,术语“预防”是指在癌症的病征或症状发生前,特别是在具有癌症风险的受试者中发生前的药物施用。As used herein, "prevention" includes inhibition of the occurrence or development of a disease or disorder or symptoms of a particular disease or disorder. In some embodiments, subjects with a family history of cancer are candidates for preventive regimens. Generally, in the context of cancer, the term "prevention" refers to the administration of a drug before the signs or symptoms of cancer occur, particularly in a subject at risk for cancer.
用于本文时,疗法的“增强”或“促进”意指能够提升已有免疫疗法(例如,细胞 疗法,优选地,CAR-T细胞疗法)的治疗效果,其中已有免疫疗法可以指任何本领域已经使用或知晓的疗法,优选针对癌症的疗法,更优选CAR-T细胞疗法。疗效得到增强,是指相比于没有被本发明的间充质干细胞所增强的已有疗法治疗的患者,在接受了被本发明的间充质干细胞所增强的已有疗法的患者中更加减缓、更加中断、更加阻滞、更加缓解、更加停止、更加降低、或更加逆转已存在的症状、病症、病况或疾病的进展或严重性,或者副作用更少、治疗体验更好、生存质量更高,或者简而言之,取得了或者基本上取得了比接受没有被本发明的间充质干细胞所增强的已有疗法治疗的患者更好的治疗过程或结局。As used herein, "enhancement" or "promotion" of therapy means being able to enhance the therapeutic effect of existing immunotherapy (e.g., cell therapy, preferably, CAR-T cell therapy), wherein existing immunotherapy can refer to any therapy already used or known in the art, preferably a therapy for cancer, more preferably CAR-T cell therapy. Enhanced efficacy means that compared to patients treated with existing therapies that have not been enhanced by the mesenchymal stem cells of the present invention, patients who have received existing therapies enhanced by the mesenchymal stem cells of the present invention have slowed down, interrupted, blocked, relieved, stopped, reduced, or reversed the progression or severity of existing symptoms, illnesses, conditions, or diseases, or have fewer side effects, better treatment experience, and higher quality of life, or in short, have achieved or substantially achieved a better treatment process or outcome than patients who have received existing therapies that have not been enhanced by the mesenchymal stem cells of the present invention.
在一些具体的实施方案中,间充质干细胞促进所述治疗性细胞(例如,优选地,CAR-T细胞)分泌细胞因子杀伤肿瘤细胞。在另一些具体的实施方案中,间充质干细胞增强治疗性细胞(例如,优选地,CAR-T细胞)对靶细胞的直接杀伤作用。在一些具体的实施方案中,间充质干细胞促进治疗性细胞(例如,优选地,CAR-T细胞)增殖扩增,即,数量增加。在一些具体的实施方案中,间充质干细胞促进治疗性细胞(例如,优选地,CAR-T细胞)持久性增加,即,在体内存活/存在的时间延长。在一些具体的实施方案中,间充质干细胞增强治疗性细胞(例如,优选地,CAR-T细胞)到达肿瘤区域的能力。在一些具体的实施方案中,所述疗效增强剂促进治疗性细胞(例如,优选地,CAR-T细胞)透过体内屏障(例如,血脑屏障)而使得更多治疗性细胞(例如,优选地,CAR-T细胞)到达肿瘤(例如,胶质细胞瘤)区域,即,增加透过体内屏障(例如,血脑屏障)的治疗性细胞(例如,优选地,CAR-T细胞)的数量。在一些优选的实施方案中,所述增强是通过免疫效应细胞的持久性增加和/或保持在低分化状态和/或更多分化为记忆性效应细胞亚型而实现的。In some specific embodiments, mesenchymal stem cells promote the secretion of cytokines by the therapeutic cells (e.g., preferably, CAR-T cells) to kill tumor cells. In other specific embodiments, mesenchymal stem cells enhance the direct killing effect of therapeutic cells (e.g., preferably, CAR-T cells) on target cells. In some specific embodiments, mesenchymal stem cells promote the proliferation and expansion of therapeutic cells (e.g., preferably, CAR-T cells), that is, the number increases. In some specific embodiments, mesenchymal stem cells promote the persistence of therapeutic cells (e.g., preferably, CAR-T cells), that is, the time of survival/existence in the body is prolonged. In some specific embodiments, mesenchymal stem cells enhance the ability of therapeutic cells (e.g., preferably, CAR-T cells) to reach the tumor area. In some specific embodiments, the efficacy enhancer promotes therapeutic cells (e.g., preferably, CAR-T cells) to penetrate the body barrier (e.g., blood-brain barrier) so that more therapeutic cells (e.g., preferably, CAR-T cells) reach the tumor (e.g., glioma) area, that is, increase the number of therapeutic cells (e.g., preferably, CAR-T cells) that penetrate the body barrier (e.g., blood-brain barrier). In some preferred embodiments, the enhancement is achieved by increasing the persistence of immune effector cells and/or maintaining them in a poorly differentiated state and/or more differentiation into memory effector cell subtypes.
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力;(1) Enhance the proliferation ability of immune effector cells in vivo;
(2)增加单个免疫效应细胞体内持久性;(2) increase the in vivo persistence of single immune effector cells;
(3)提升由免疫效应细胞分泌的治疗性细胞因子(例如,干扰素γ(IFN-γ)、肿瘤坏死因子α(TNF-α)、IL-4、IL-6、IL-10和/或IL-17A)的量;(3) increasing the amount of therapeutic cytokines (e.g., interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), IL-4, IL-6, IL-10, and/or IL-17A) secreted by immune effector cells;
(4)增强携带靶抗原的细胞的杀伤;(4) Enhanced killing of cells carrying target antigens;
(5)抑制免疫效应细胞(例如,CAR-T细胞)的分化,使其保持低分化状态;(5) Inhibit the differentiation of immune effector cells (e.g., CAR-T cells) to keep them in a low-differentiation state;
(6)增加记忆性T细胞形成;(6) Increase the formation of memory T cells;
(7)增加靶抗原中和/抑制;(7) Increased target antigen neutralization/inhibition;
(8)延缓病灶进展/缩小病灶体积;(8) Delaying lesion progression/reducing lesion size;
(9)逆转接受治疗的受试者体内对免疫疗法的抑制因素/环境/情形;(9) reversing inhibitory factors/environments/conditions against immunotherapy in the treated subjects;
(10)提高接受治疗的受试者生存率和/或延长其生存期(即,寿命延长)。(10) Improving the survival rate and/or prolonging the survival period of the subjects receiving treatment (i.e., extending life span).
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力;(1) Enhance the proliferation ability of immune effector cells in vivo;
(2)增加单个免疫效应细胞体内持久性;(2) increase the in vivo persistence of single immune effector cells;
(3)增强携带靶抗原的细胞的杀伤;(3) Enhanced killing of cells carrying target antigens;
(4)抑制免疫效应细胞(例如,CAR-T细胞)的分化,使其保持低分化状态;(4) Inhibit the differentiation of immune effector cells (e.g., CAR-T cells) to keep them in a low-differentiation state;
(5)增加记忆性T细胞形成;(5) Increase the formation of memory T cells;
(6)延缓病灶进展/缩小病灶体积;(6) Delaying lesion progression/reducing lesion size;
(7)提高接受治疗的受试者生存率和/或延长其生存期(即,寿命延长)。(7) Improving the survival rate and/or prolonging the survival period of the subjects receiving treatment (i.e., extending life span).
在一些具体的实施方案中,间充质干细胞增强细胞疗法至少体现在如以下所述的一项或多项:In some specific embodiments, mesenchymal stem cells enhance cell therapy by at least one or more of the following:
(1)增强免疫效应细胞体内增殖能力或增加单个免疫效应细胞体内持久性;(1) Enhance the proliferation capacity of immune effector cells in vivo or increase the persistence of single immune effector cells in vivo;
(2)抑制CAR-T细胞的分化,使其保持低分化状态;(2) Inhibit the differentiation of CAR-T cells, keeping them in a low-differentiation state;
(3)增加记忆性T细胞形成;和(3) increase the formation of memory T cells; and
(4)提高接受治疗的受试者生存率和/或延长其生存期。(4) Improve the survival rate and/or prolong the survival of the subjects receiving treatment.
在一些实施方案中,细胞疗法所用的治疗性细胞与间充质干细胞的数量比例为100:1,或90:1,或80:1,或70:1,或60:1,或50:1,或40:1,或30:1,或20:1,或10:1,或5:1,或4:1,或2:1,或1:1,或1:2,或1:4,或1:5,或1:10,或介于其之间的比例。In some embodiments, the ratio of the number of therapeutic cells to mesenchymal stem cells used in cell therapy is 100:1, or 90:1, or 80:1, or 70:1, or 60:1, or 50:1, or 40:1, or 30:1, or 20:1, or 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or a ratio therebetween.
治疗方法/组合疗法Treatment methods/combination therapies
在一个方面,本发明涉及间充质干细胞用于增强免疫疗法疗效的用途。在一些实施方案中,免疫疗法是细胞疗法,即,施用治疗性细胞的疗法。在一些更具体的实施方案中,治疗性细胞是非特异性免疫效应细胞。在一些更具体的实施方案中,治疗性细胞是特异性免疫效应细胞,优选地,治疗性细胞是T细胞,更优选地,治疗性细胞是经修饰的T细胞,最优选地,治疗性细胞是CAR-T细胞。在一些实施方案中,人T细胞是CD8+T细胞。在另一些具体的实施方案中,治疗性细胞是CAR-NK,TCR-T、TIL、CAR-DC等等细胞。In one aspect, the present invention relates to the use of mesenchymal stem cells for enhancing the efficacy of immunotherapy. In some embodiments, immunotherapy is cell therapy, that is, a therapy that administers therapeutic cells. In some more specific embodiments, therapeutic cells are nonspecific immune effector cells. In some more specific embodiments, therapeutic cells are specific immune effector cells, preferably, therapeutic cells are T cells, more preferably, therapeutic cells are modified T cells, and most preferably, therapeutic cells are CAR-T cells. In some embodiments, human T cells are CD8+T cells. In other specific embodiments, therapeutic cells are CAR-NK, TCR-T, TIL, CAR-DC, etc. cells.
在另一个方面,本发明提供了一种方法,包括向有需要的受试者施用免疫效应细胞,所述免疫效应细胞任选地是经修饰的,优选地包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是CAR-T细胞。在一个实施方案中,受试者具有本文所描述的病症,例如,受试者患有癌症,例如,受试者具有表达本文所述的靶抗原的癌症。在一个实施方案中,受试者是人。In another aspect, the present invention provides a method comprising administering immune effector cells to a subject in need, wherein the immune effector cells are optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body. Preferably, the immune effector cells are CAR-T cells. In one embodiment, the subject has a condition described herein, for example, the subject suffers from cancer, for example, the subject has a cancer expressing a target antigen described herein. In one embodiment, the subject is a human.
在另一个方面,本发明涉及治疗患有与如本文所述的癌症相关抗原的表达相关的疾病的受试者的方法,包括向有需要的受试者施用免疫效应细胞,所述免疫效应细胞任选地是经修饰的,优选地包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention relates to a method for treating a subject with a disease associated with the expression of a cancer-associated antigen as described herein, comprising administering an immune effector cell to a subject in need thereof, the immune effector cell being optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cell is a CAR-T cell.
在又一个方面中,本发明提供了治疗患有与肿瘤抗原的表达相关的疾病的受试者的方法,其包括向有需要的受试者施用免疫效应细胞,所述免疫效应细胞任选地是经修饰的,优选地包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免 疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a method for treating a subject suffering from a disease associated with the expression of a tumor antigen, comprising administering to a subject in need an immune effector cell, the immune effector cell being optionally modified, preferably comprising a CAR molecule and/or a vector molecule comprising a coding sequence for the CAR molecule, and the cell having been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cell is a CAR-T cell.
在又一个方面,本发明提供了增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。优选地,上述方法使得免疫效应细胞在施用于受试者体内后持久性增加。In another aspect, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells to the body. Therefore, in a specific embodiment, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vitro/ex vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for enhancing the in vivo proliferation ability of immune effector cells after administration in vivo and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-administering the immune effector cells with mesenchymal stem cells to the body. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells. Preferably, the above method increases the persistence of immune effector cells after administration in a subject.
本发明还提供了调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内调节免疫效应细胞在施用后的体内分化的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。优选地,上述方法使得免疫效应细胞在施用于受试者体内后保持在低分化状态。The present invention also provides a method for regulating the differentiation of immune effector cells in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for regulating the differentiation of immune effector cells in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells. Preferably, the above method keeps the immune effector cells in a low differentiated state after being administered to the subject.
本发明还提供了促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。因此,在一个具体的实施方案中,本发明提供了体外/离体促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞在体外/离体与间充质干细胞共培养。在另一个具体的实施方案中,本发明提供了体内促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是经修饰的。优选地,所述免疫效应细胞是T细胞。更优选地,所述免疫效应细胞是CAR-T细胞。优选地,上述方法使得免疫效应细胞在施用于受试者体内后更多分化为记忆性效应细胞亚型。The present invention also provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering them with mesenchymal stem cells in vivo. Therefore, in a specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vitro/ex vivo, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo. In another specific embodiment, the present invention provides a method for promoting the differentiation of immune effector cells to memory cell subtypes in vivo after administration in vivo, the method comprising co-administering the immune effector cells with mesenchymal stem cells in vivo. Preferably, the immune effector cells are modified. Preferably, the immune effector cells are T cells. More preferably, the immune effector cells are CAR-T cells. Preferably, the above method allows immune effector cells to differentiate into more memory effector cell subtypes after being administered to a subject.
在另一个方面,本发明提供了包含免疫效应细胞和间充质干细胞的组合物,用于治疗患有与肿瘤抗原的表达相关的疾病如本文所述的病症的受试者,其中所述免疫效应细胞包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a composition comprising immune effector cells and mesenchymal stem cells, for treating a subject with a disease associated with the expression of a tumor antigen such as a disease as described herein, wherein the immune effector cell comprises a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body. Preferably, the immune effector cell is a CAR-T cell.
在任何上述方法或用途的某些实施方案中,所述间充质干细胞可与所述免疫效应细 胞组合递送,在所述免疫效应细胞施用之前施用,与所述免疫效应细胞同时施用,在所述免疫效应细胞施用之后施用。备选地,免疫效应细胞在施用于受试者之前,与所述间充质干细胞在体外/离体共培养,并且在共培养后,所述间充质干细胞与/不与所述免疫效应细胞组合递送。备选地,间充质干细胞可以在免疫效应细胞施用之后延长的时间后,施用。在一个实施方案中,间充质干细胞与免疫效应细胞或细胞群被同时(例如,在同一天施用)施用于受试者或免疫效应细胞或细胞群施用后(例如,施用后1天,2天,3天,4天,5天,6天,7天,2周,3周,4周,6周,8周,10周或以上)施用于受试者。In certain embodiments of any of the above methods or uses, the mesenchymal stem cells can be delivered in combination with the immune effector cells, administered before the immune effector cells are administered, administered simultaneously with the immune effector cells, and administered after the immune effector cells are administered. Alternatively, the immune effector cells are co-cultured in vitro/ex vivo with the mesenchymal stem cells before being administered to the subject, and after co-culture, the mesenchymal stem cells are delivered in combination with/not with the immune effector cells. Alternatively, mesenchymal stem cells can be administered after an extended period of time after the immune effector cells are administered. In one embodiment, mesenchymal stem cells and immune effector cells or cell groups are administered to the subject at the same time (e.g., administered on the same day) or immune effector cells or cell groups are administered (e.g., 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 2 weeks, 3 weeks, 4 weeks, 6 weeks, 8 weeks, 10 weeks or more) after administration.
在任何上述方法或用途的实施方案中,还可以进一步组合其他本领域已知的治疗剂/治疗方法,例如,但不限于,免疫检查点抑制剂、靶向药、化疗药物、手术治疗、放疗、其他CAR-T增效剂,等等。In any of the above-mentioned methods or embodiments of the use, other therapeutic agents/treatment methods known in the art may be further combined, such as, but not limited to, immune checkpoint inhibitors, targeted drugs, chemotherapy drugs, surgical treatment, radiotherapy, other CAR-T enhancers, and the like.
在任何上述方法或用途的实施方案中,还可以进一步组合其他本领域已知的治疗剂/治疗方法,例如,但不限于,免疫检查点抑制剂、靶向药、化疗药物、手术治疗、放疗、其他CAR-T增效剂,等等。In any of the above methods or embodiments of the use, other therapeutic agents/treatment methods known in the art may be further combined, such as, but not limited to, immune checkpoint inhibitors, targeted drugs, chemotherapy drugs, surgical treatment, radiotherapy, other CAR-T enhancers, and the like.
在任何上述方法或用途的实施方案中,在所述共培养/组合递送时,治疗性细胞与间充质干细胞以约选自以下的比例存在:间充质干细胞:治疗性细胞=5:1、4:1、3:1、2:1、1:1、1:2、1:3、1:4、1:5、1:6、1:7、1:8、1:9、1:10、1:11、1:12、1:13、1:14、1:15、1:16、1:17、1:18、1:19、1:20、1:30、1:40、1:50、1:60、1:70、1:80、1:90、1:100,或之间的任何比例。In any embodiment of the above method or use, during the co-culture/combined delivery, the therapeutic cells and mesenchymal stem cells are present in a ratio approximately selected from the following: mesenchymal stem cells: therapeutic cells = 5: 1, 4: 1, 3: 1, 2: 1, 1: 1, 1: 2, 1: 3, 1: 4, 1: 5, 1: 6, 1: 7, 1: 8, 1: 9, 1: 10, 1: 11, 1: 12, 1: 13, 1: 14, 1: 15, 1: 16, 1: 17, 1: 18, 1: 19, 1: 20, 1: 30, 1: 40, 1: 50, 1: 60, 1: 70, 1: 80, 1: 90, 1: 100, or any ratio therebetween.
在任何上述方法或用途的实施方案中,经共培养/组合递送,所述免疫效应细胞对目的肿瘤的杀伤效力,相对于未与间充质干细胞共培养也未组合递送的相同免疫效应细胞,得到增强。在优选的实施方案中,杀伤效力增强了10%、20%、30%、40%、50%、60%、70%、80%、90%、100%、120%、140%、2倍、3倍、4倍、5倍、6倍、7倍、8倍、9倍、10倍、100倍、500倍、1000倍或更高,如利用本领域已知的任何评估疾病改善和/或肿瘤消退的指标所评价的。In any of the above methods or embodiments of purposes, through co-culture/combined delivery, the killing efficacy of the immune effector cells to the target tumor is enhanced relative to the same immune effector cells that are not co-cultured with mesenchymal stem cells and not delivered in combination. In a preferred embodiment, the killing efficacy is enhanced by 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 120%, 140%, 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, 9 times, 10 times, 100 times, 500 times, 1000 times or higher, as evaluated by any index known in the art for assessing disease improvement and/or tumor regression.
在任何上述方法或用途的某些实施方案中,与肿瘤抗原,例如,本文所述的肿瘤抗原相关的疾病选自增生性疾病,诸如癌症或恶性肿瘤或癌前病变,如脊髓发育不良,骨髓增生异常综合征或白血病前期,或与本文所述的肿瘤抗原的表达相关联的非癌症相关的适应证。在一个实施方案中,所述疾病是本文如前所述的癌症。在一些实施方案中,所述肿瘤抗原是本文如前所述的肿瘤抗原。In certain embodiments of any of the above methods or uses, the disease associated with a tumor antigen, e.g., a tumor antigen as described herein, is selected from a proliferative disease, such as a cancer or a malignant tumor or a precancerous lesion, such as myelodysplasia, myelodysplastic syndrome or preleukemia, or a non-cancer-related indication associated with the expression of a tumor antigen as described herein. In one embodiment, the disease is a cancer as described herein above. In some embodiments, the tumor antigen is a tumor antigen as described herein above.
在另一个方面,本发明提供了一种方法,包括向有需要的受试者施用经修饰的免疫效应细胞,所述细胞包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞是CAR-T细胞。在一个实施方案中,受试者具有本文所描述的病症,例如,受试者患有癌症,例如,受试者具有表达本文所述的靶抗原的癌症。在一个实施方案中,受试者是人。In another aspect, the present invention provides a method comprising administering a modified immune effector cell to a subject in need, the cell comprising a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and the cell has been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells to the body. Preferably, the immune effector cell is a CAR-T cell. In one embodiment, the subject has a condition described herein, for example, the subject suffers from cancer, for example, the subject has a cancer expressing a target antigen described herein. In one embodiment, the subject is a human.
在一些优选的实施方案中,与所述间充质干细胞的共同培养/共同施用增加了所述免疫效应细胞的疗效。In some preferred embodiments, co-culture/co-administration with the mesenchymal stem cells increases the therapeutic efficacy of the immune effector cells.
制药用途/药物组合物Pharmaceutical use/pharmaceutical composition
在另一个方面,本发明提供了药物组合物,所述药物组合物用于增强细胞疗法的疗效,其中所述药物组合物包含间充质干细胞。所述细胞疗法向有需要的受试者施用治疗性细胞,所述治疗性细胞是非特异性免疫效应细胞。优选地,所述免疫效应细胞包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,更优选地所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a pharmaceutical composition for enhancing the efficacy of cell therapy, wherein the pharmaceutical composition comprises mesenchymal stem cells. The cell therapy administers therapeutic cells to subjects in need, and the therapeutic cells are nonspecific immune effector cells. Preferably, the immune effector cells comprise a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and more preferably the immune effector cells are CAR-T cells.
在另一个方面,本发明提供了药物组合物,用于治疗患有与肿瘤抗原的表达相关的疾病如本文所述的病症的受试者,其中所述药物组合物包含免疫效应细胞和间充质干细胞,且所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内。优选地,所述免疫效应细胞包含CAR分子和/或包含所述CAR分子的编码序列的载体分子,更优选地所述免疫效应细胞是CAR-T细胞。In another aspect, the present invention provides a pharmaceutical composition for treating a subject suffering from a disease such as a disease described herein associated with the expression of a tumor antigen, wherein the pharmaceutical composition comprises immune effector cells and mesenchymal stem cells, and the immune effector cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo. Preferably, the immune effector cells comprise a CAR molecule and/or a vector molecule comprising a coding sequence of the CAR molecule, and more preferably the immune effector cells are CAR-T cells.
在任何上述实施方案中,药物组合物可以进一步包含一种或多种药学上可接受的或生理上可接受的载体、稀释剂或赋形剂,其使得所述药物组合物适合于通过预期的途径给药,例如,但不限于,静脉注射或肿瘤局部注射等等。In any of the above embodiments, the pharmaceutical composition may further comprise one or more pharmaceutically acceptable or physiologically acceptable carriers, diluents or excipients, which make the pharmaceutical composition suitable for administration via the intended route, such as, but not limited to, intravenous injection or local tumor injection, etc.
因此,在另一个方面,本发明提供了间充质干细胞在制备细胞疗法的疗效增强剂中的用途。Therefore, in another aspect, the present invention provides use of mesenchymal stem cells in preparing an agent for enhancing the therapeutic effect of cell therapy.
本文引用或提及的所有专利,专利申请,出版物,技术文献和/或学术文章以及其他参考文献在法律允许的范围内通过引用的方式,以其全部内容并入本文。All patents, patent applications, publications, technical documents and/or scholarly articles and other references cited or referred to herein are incorporated by reference in their entirety to the extent permitted by law.
实施例Example
现在参考以下实施例描述本发明。提供这些实施例仅用于举例说明的目的,本发明不局限于这些实施例,而是涵盖基于本文提供的教导而显而易见的所有变化。The present invention will now be described with reference to the following examples. These examples are provided for illustrative purposes only, and the present invention is not limited to these examples, but encompasses all variations that are obvious based on the teaching provided herein.
实施例1.材料和方法Example 1. Materials and methods
本研究(包括其每个部分,例如其引用或参考的部分)得到了北京世纪坛医院机构审查委员会的批准和所有参与者的知情同意。胎盘来自首都医科大学附属北京世纪坛医院的健康捐赠者。所有处理人类胎盘的方案、间充质干细胞的分离和培养、体外的细胞实验和体内的异种移植小鼠实验都得到了北京世纪坛医院伦理委员会的批准。This study (including each of its parts, such as those cited or referenced therein) was approved by the Institutional Review Board of Beijing Shijitan Hospital and informed consent was obtained from all participants. Placentas were obtained from healthy donors of Beijing Shijitan Hospital affiliated to Capital Medical University. All protocols for processing human placentas, isolation and culture of mesenchymal stem cells, in vitro cell experiments, and in vivo xenograft mouse experiments were approved by the Ethics Committee of Beijing Shijitan Hospital.
胎盘间充质干细胞(pMSC)的分离和培养Isolation and culture of placental mesenchymal stem cells (pMSC)
胎盘组织的处理方式按照Papait A等的方法(Mesenchymal Stromal Cells from Fetal and Maternal Placenta Possess Key Similarities and Differences:Potential Implications for Their Applications in Regenerative Medicine.Cells.2020 Jan 6;9(1):127)的基础上稍作修改。首先将胎盘组织浸泡在含有10%青霉素和链霉素的预热磷酸盐缓冲盐水(PBS)溶液中10分钟,然后再用PBS清洗两次,并用剪刀剪成1mm 3。通过100μm无菌细胞过滤器获得单个核细胞,并由密度梯度离心分离淋巴细胞。细胞在含有20%FBS、2mM L-谷氨酰胺和100U/ml青/链霉素的DMEM培养基中,以1×10 6细胞/cm 2的密度,在37℃下含5%(v/v)CO 2的 湿润空气中维持培养。大约6至8天后,许多集落已经形成。去除未贴附的细胞,补充新鲜培养基。在汇合度达到85%后,用0.25%(w/v)胰酶/EDTA将粘附的细胞消化,并以5×10 3细胞/cm 2的细胞密度重新铺种。胎盘细胞在相同条件下连续培养,扩增2至6代(Passage,P)。所有的实验都是用第3代至第6代(即P3和P6)之间收获的细胞在无菌环境中进行的,并尽可能在生物安全柜中进行。 Placental tissue was processed according to the method of Papait A et al. (Mesenchymal Stromal Cells from Fetal and Maternal Placenta Possess Key Similarities and Differences: Potential Implications for Their Applications in Regenerative Medicine. Cells. 2020 Jan 6; 9(1): 127) with slight modifications. First, the placental tissue was soaked in a preheated phosphate-buffered saline (PBS) solution containing 10% penicillin and streptomycin for 10 minutes, then washed twice with PBS and cut into 1 mm 3 with scissors. Mononuclear cells were obtained through a 100 μm sterile cell filter, and lymphocytes were separated by density gradient centrifugation. Cells were cultured at a density of 1×10 6 cells/cm 2 in DMEM medium containing 20% FBS, 2mM L-glutamine and 100U/ml penicillin/streptomycin at 37°C in a humidified atmosphere containing 5% (v/v) CO 2. After about 6 to 8 days, many colonies had formed. Unattached cells were removed and fresh medium was added. After the confluence reached 85%, the adherent cells were digested with 0.25% (w/v) trypsin/EDTA and re-plated at a cell density of 5×10 3 cells/cm 2. Placental cells were cultured continuously under the same conditions and expanded for 2 to 6 passages (Passage, P). All experiments were performed in a sterile environment with cells harvested between passages 3 and 6 (i.e., P3 and P6) and performed in a biosafety cabinet whenever possible.
胎盘间充质干细胞(pMSC)的扩增和表征Expansion and Characterization of Placental Mesenchymal Stem Cells (pMSC)
按照Lechanteur等人(Clinical-scale expansion of mesenchymal stromal cells:a large banking experience.J Transl Med.2016 May 20;14(1):145.)的方法,通过累积的每代群体倍增数来评估pMSC的扩增情况。将第1代的间充质干细胞以5×10 3cells/cm 2的细胞密度铺种在六孔板中。然后每5天收获细胞,用台盼蓝染色计数,并以相同的细胞密度5×10 3细胞/cm 2重新铺种。重复该过程,直到第6代。 According to the method of Lechanteur et al. (Clinical-scale expansion of mesenchymal stromal cells: a large banking experience. J Transl Med. 2016 May 20; 14(1): 145.), the expansion of pMSCs was evaluated by the cumulative population doublings per generation. The first generation of mesenchymal stem cells was plated in a six-well plate at a cell density of 5×10 3 cells/cm 2. Then the cells were harvested every 5 days, counted by trypan blue staining, and re-plated at the same cell density of 5×10 3 cells/cm 2. The process was repeated until the 6th generation.
pMSC的表征是根据国际细胞治疗学会(ISCT)提出的指南进行的(参见:Viswanathan S等,Mesenchymal stem versus stromal cells:International Society for Cell & Gene Therapy
Figure PCTCN2022128989-appb-000001
Mesenchymal Stromal Cell committee position statement on nomenclature.Cytotherapy.2019Oct;21(10):1019-1024.)。培养的pMSC的免疫表型通过流式细胞仪评估。总共1×10 6个第3代细胞,在室温下避光孵育15分钟。使用FACSCanto-II流式细胞仪和flowjo v.10软件(BD Biosciences)进行数据采集和分析。通过在特定的分化诱导培养基中培养3-4周来评估pMSC的分化诱导能力。对于成骨分化的诱导和检测评估参照文献进行:Jaiswal N等,Osteogenic differentiation of purified,culture-expanded human mesenchymal stem cells in vitro.J Cell Biochem 1997;64:295-312.和Xu X等,Dysregulated systemic lymphocytes affect the balance of osteogenic/adipogenic differentiation of bone mesenchymal stem cells after local irradiation.Stem Cell Res Ther 2017;8:71。间充质干细胞用茜素红染色以检测钙质沉积,并通过检测与诱导成骨细胞形成有关的特定转录因子的mRNA水平表达,以进一步定量评估。使用Trizol从分化的间充质干细胞培养物和对照中提取总mRNA,并使用SYBR Green PCR Master Mix(Fisher Scientific SL)和引物,在罗氏Light Cycler 480II上实施定量RT-PCR(qRT-PCR)分析。反应以一式三份进行,基因表达值归一化至GAPDH的表达值。
The characterization of pMSCs was performed according to the guidelines proposed by the International Society for Cellular Therapy (ISCT) (see: Viswanathan S et al., Mesenchymal stem versus stromal cells: International Society for Cell & Gene Therapy
Figure PCTCN2022128989-appb-000001
Mesenchymal Stromal Cell committee position statement on nomenclature. Cytotherapy. 2019 Oct; 21(10): 1019-1024.). The immunophenotype of cultured pMSCs was evaluated by flow cytometry. A total of 1×10 6 cells at passage 3 were incubated at room temperature in the dark for 15 minutes. Data acquisition and analysis were performed using a FACSCanto-II flow cytometer and flowjo v.10 software (BD Biosciences). The differentiation induction ability of pMSCs was evaluated by culturing in a specific differentiation induction medium for 3-4 weeks. The induction and detection of osteogenic differentiation were evaluated according to the following references: Jaiswal N et al., Osteogenic differentiation of purified, culture-expanded human mesenchymal stem cells in vitro. J Cell Biochem 1997; 64: 295-312. and Xu X et al., Dysregulated systemic lymphocytes affect the balance of osteogenic/adipogenic differentiation of bone mesenchymal stem cells after local irradiation. Stem Cell Res Ther 2017; 8: 71. Mesenchymal stem cells were stained with Alizarin Red to detect calcium deposition, and the mRNA level expression of specific transcription factors related to the induction of osteoblast formation was further quantitatively evaluated. Total mRNA was extracted from differentiated mesenchymal stem cell cultures and controls using Trizol, and quantitative RT-PCR (qRT-PCR) analysis was performed on a Roche Light Cycler 480II using SYBR Green PCR Master Mix (Fisher Scientific SL) and primers. The reactions were performed in triplicate, and the gene expression values were normalized to the expression values of GAPDH.
细胞系Cell lines
人B型急性淋巴细胞白血病细胞系NALM-6和人Burkitt淋巴瘤细胞Raji以及逆转录病毒包装细胞系PG13和Phoenix ECO均购自ATCC。通过逆转录病毒转导而被工程化为表达GFP和萤火虫荧光素酶(Luc)的NALM-6和Raji细胞系分别命名为NALM-6-GL和Raji-GL。这些细胞系在含有10%胎牛血清(FBS,Biosera)、100U/mL青霉素和100μg/mL链霉素(EallBio Life Sciences)的1640培养基(Lonza)中进行良好培养。逆转录病毒生产者细胞系在含有10%FBS的不加青链霉素的1640培养基中培养。Human B-type acute lymphoblastic leukemia cell line NALM-6 and human Burkitt lymphoma cell Raji as well as retroviral packaging cell lines PG13 and Phoenix ECO were purchased from ATCC. NALM-6 and Raji cell lines engineered to express GFP and firefly luciferase (Luc) by retroviral transduction were named NALM-6-GL and Raji-GL, respectively. These cell lines were well cultured in 1640 medium (Lonza) containing 10% fetal bovine serum (FBS, Biosera), 100 U/mL penicillin, and 100 μg/mL streptomycin (EallBio Life Sciences). Retroviral producer cell lines were cultured in 1640 medium without penicillin and streptomycin containing 10% FBS.
嵌合抗原受体-T细胞的产生Generation of chimeric antigen receptor-T cells
三种特异性靶向CAR-T细胞用于本发明的实施例,包括靶向血液肿瘤抗原CD19的CAR-T细胞,和分别靶向两种实体瘤抗原uPAR和IL13Rα2的CAR-T细胞。CAR通过逆转录 病毒载体转移到T细胞中。靶向血液肿瘤抗原CD19的CAR-T细胞的实例如Ying Zhang等,Co-expression IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy.J Transl Med.2022 Sep 27;20(1):432中所描述。靶向两种实体瘤抗原uPAR和IL13Rα2的CAR-T细胞的实例分别如Amor,C.等,Senolytic CAR T cells reverse senescence-associated pathologies.Nature 583,127–132(2020).和中国专利申请号202210019437.4中所描述。CAR-T细胞的普适性制备方法可以参照例如此前的论文(An Z等.Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1.Oncoimmunology.2021 Aug 16;10(1):1960728)。Three specific targeted CAR-T cells are used in embodiments of the present invention, including CAR-T cells targeting the blood tumor antigen CD19, and CAR-T cells targeting two solid tumor antigens uPAR and IL13Rα2, respectively. CAR is transferred into T cells via a retroviral vector. Examples of CAR-T cells targeting the blood tumor antigen CD19 are described in Ying Zhang et al., Co-expression of IL-15 receptor alpha with IL-15 reduces toxicity via limiting IL-15 systemic exposure during CAR-T immunotherapy. J Transl Med. 2022 Sep 27; 20(1):432. Examples of CAR-T cells targeting two solid tumor antigens uPAR and IL13Rα2 are described in Amor, C. et al., Senolytic CAR T cells reverse senescence-associated pathologies. Nature 583, 127–132 (2020). and Chinese patent application No. 202210019437.4. The universal preparation method of CAR-T cells can refer to, for example, the previous paper (An Z et al. Antitumor activity of the third generation EphA2 CAR-T cells against glioblastoma is associated with interferon gamma induced PD-L1. Oncoimmunology. 2021 Aug 16; 10(1): 1960728).
具体地,CAR的详细构造和CAR-T细胞的制备过程见图6和图7。所述CAR的结构由三部分组成,即含有anti-CD19 scFV序列的胞外结构域、CD28的跨膜结构域和用P2A连接的CD28、4-1BB、CD3ζ和IL15序列的胞内结构域。我们以前的研究发现,含有CD28、4-1BB和/或CD3ζ的原代人CD8+T细胞的胞内域在促进细胞因子释放方面比只含有一个或两个结构域的要好(参见:Zhong XS等,Chimeric antigen receptors combining 4-1BB and CD28signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+T cell-mediated tumor eradication.Mol Ther.2010 Feb;18(2):413-20.)。Specifically, the detailed structure of CAR and the preparation process of CAR-T cells are shown in Figures 6 and 7. The structure of the CAR consists of three parts, namely, the extracellular domain containing the anti-CD19 scFV sequence, the transmembrane domain of CD28, and the intracellular domain of CD28, 4-1BB, CD3ζ and IL15 sequences connected by P2A. Our previous studies have found that the intracellular domain of primary human CD8+T cells containing CD28, 4-1BB and/or CD3ζ is better than that containing only one or two domains in promoting cytokine release (see: Zhong XS et al., Chimeric antigen receptors combining 4-1BB and CD28 signaling domains augment PI3kinase/AKT/Bcl-XL activation and CD8+T cell-mediated tumor eradication. Mol Ther. 2010 Feb; 18(2): 413-20.).
使用淋巴细胞分离培养基(MP Biomedicals)通过梯度离心分离来自健康捐赠者的人外周血单个核细胞(PBMC)。用抗CD3/CD28T细胞活化Dynabeads(Invitrogen)刺激PBMC中的T细胞。珠子激活48小时后,在Retronectin(Takara)-包被板上通过离心法用逆转录病毒上清液转导T细胞。在第7天对T细胞进行免疫印迹检测CAR的表达。T细胞在X-VIVO-15培养基中培养,该培养基含有5%人AB血清(SIGMA)、100U/ml IL-2、100U/mL青霉素和100μg/mL链霉素(EallBio Life Sciences)。Human peripheral blood mononuclear cells (PBMCs) from healthy donors were isolated by gradient centrifugation using lymphocyte separation medium (MP Biomedicals). T cells in PBMCs were stimulated with anti-CD3/CD28 T cell activation Dynabeads (Invitrogen). After 48 h of bead activation, T cells were transduced with retroviral supernatant by centrifugation on Retronectin (Takara)-coated plates. T cells were immunoblotted for CAR expression on day 7. T cells were cultured in X-VIVO-15 medium containing 5% human AB serum (SIGMA), 100 U/ml IL-2, 100 U/mL penicillin, and 100 μg/mL streptomycin (EallBio Life Sciences).
流式细胞术分析Flow cytometric analysis
使用BD FacsCanto II Plus仪器(BD Biosciences)进行流式细胞测定,并使用FlowJo v.10软件(Tree star,Inc.Ashland,OR)进行分析。使用APC偶联的抗人CD3抗体(BD Biosciences)、V450偶联的抗人CD4(BD Biosciences)、PE-Cy7偶联的抗人CD8(BD Biosciences)、PE-Cy7偶联的抗人CCR7(BD Biosciences)检测T细胞。PE-Cy5偶联的抗人CD95(BD Biosciences),Alexa Fluor 700偶联的抗人CD27(BD Biosciences)和山羊抗小鼠IgG(Fab特异性)F(ab')2片段抗体(Sigma)。用FITC标记的山羊抗小鼠IgG(H+L)抗体(Sigma)染色检测CAR。MSC细胞用PE-Cy7偶联的抗人CD105(ThermoFisher Scientific)和V450偶联的抗人CD45(BD Biosciences)染色。T细胞的脱颗粒现象通过BV421偶联的抗人CD107a-APC(ThermoFisher Scientific)检测。与抗体孵育后,用PBS清洗细胞,然后用含有1%FBS的PBS重悬,上机分析。Flow cytometry was performed using a BD FacsCanto II Plus instrument (BD Biosciences) and analyzed using FlowJo v.10 software (Treestar, Inc. Ashland, OR). T cells were detected using APC-conjugated anti-human CD3 antibodies (BD Biosciences), V450-conjugated anti-human CD4 (BD Biosciences), PE-Cy7-conjugated anti-human CD8 (BD Biosciences), and PE-Cy7-conjugated anti-human CCR7 (BD Biosciences). PE-Cy5-conjugated anti-human CD95 (BD Biosciences), Alexa Fluor 700-conjugated anti-human CD27 (BD Biosciences), and goat anti-mouse IgG (Fab specific) F(ab')2 fragment antibodies (Sigma). CAR was detected by staining with FITC-labeled goat anti-mouse IgG (H+L) antibodies (Sigma). MSC cells were stained with PE-Cy7-conjugated anti-human CD105 (ThermoFisher Scientific) and V450-conjugated anti-human CD45 (BD Biosciences). T cell degranulation was detected by BV421-conjugated anti-human CD107a-APC (ThermoFisher Scientific). After incubation with antibodies, cells were washed with PBS, resuspended in PBS containing 1% FBS, and analyzed on an analyzer.
检测CAR-T细胞的CD107a脱颗粒Detection of CD107a degranulation of CAR-T cells
将CAR-T细胞与MSC细胞以10:1的比例共培养24小时,然后收集CAR-T并与NALM-6细胞以1:1的比例在24孔板中共培养,其中每孔都含有1μl抗人CD107a抗体(BD  Bioscience)和Golgi Stop TM(BD Bioscience)。6小时后,收获细胞并与抗人CD3抗体(BD Bioscience)孵化,通过流式细胞术分析检测CAR-T细胞的CD107a脱颗粒情况。以CD19-IL15 CAR-T细胞为例。实验分为四组:(1)事先不与MSC共培养的CAR-T细胞作为对照;(2)事先与MSC共培养的CAR-T细胞;(3)与NALM-6细胞共培养的CAR-T细胞;(4)先与MSC共培养,再收集并与NALM-6细胞共培养的Car-T细胞。 CAR-T cells were co-cultured with MSC cells at a ratio of 10:1 for 24 hours, and then CAR-T cells were collected and co-cultured with NALM-6 cells at a ratio of 1:1 in a 24-well plate, where each well contained 1 μl of anti-human CD107a antibody (BD Bioscience) and Golgi Stop TM (BD Bioscience). After 6 hours, the cells were harvested and incubated with anti-human CD3 antibody (BD Bioscience), and the CD107a degranulation of CAR-T cells was detected by flow cytometry analysis. CD19-IL15 CAR-T cells were used as an example. The experiment was divided into four groups: (1) CAR-T cells that were not co-cultured with MSCs in advance as a control; (2) CAR-T cells that were co-cultured with MSCs in advance; (3) CAR-T cells that were co-cultured with NALM-6 cells; (4) CAR-T cells that were first co-cultured with MSCs and then collected and co-cultured with NALM-6 cells.
羧基荧光素琥珀酰亚胺酯稀释试验Carboxyfluorescein succinimidyl ester dilution test
利用来自三个健康捐赠者的细胞制备了CAR-T细胞,与NALM-6细胞系进行共培养,并使用基于羧基荧光素琥珀酰亚胺酯(CFSE)染料的流式细胞术分析T细胞增殖。首先将CFSE标记的CAR-T细胞与MSC细胞在圆底24孔板中以1×10 6:1×10 5细胞/ml的密度(即10:1的比例)共培养24小时,一式三份。然后收集CAR-T细胞并与NALM-6以10:1的配比共培养3天。使用BD FacsCanto II Plus仪器(BD Biosciences)通过CD3阳性细胞的CFSE染料稀释测定T细胞增殖。使用Flow Jo v.10软件(Tree star,Inc.Ashland,OR)分析流式细胞仪数据。本实验的组别与CD107a脱颗粒实验的组别相同。 CAR-T cells were prepared using cells from three healthy donors, co-cultured with the NALM-6 cell line, and T cell proliferation was analyzed using flow cytometry based on carboxyfluorescein succinimidyl ester (CFSE) dye. CFSE-labeled CAR-T cells were first co-cultured with MSC cells in a round-bottom 24-well plate at a density of 1×10 6 :1×10 5 cells/ml (i.e., a ratio of 10:1) for 24 hours in triplicate. CAR-T cells were then collected and co-cultured with NALM-6 at a ratio of 10:1 for 3 days. T cell proliferation was determined by CFSE dye dilution of CD3-positive cells using a BD FacsCanto II Plus instrument (BD Biosciences). Flow cytometric data were analyzed using Flow Jo v.10 software (Tree star, Inc. Ashland, OR). The groups in this experiment were the same as those in the CD107a degranulation experiment.
细胞毒性试验Cytotoxicity assay
用两种方法来检测CAR-T细胞的细胞毒性功能。一种是通过流式细胞术检测靶细胞的GFP信号,另一种是用活体成像分析系统检测靶细胞的化学发光。CAR-T细胞首先与MSC细胞以10:1的比例共培养24小时,然后重新收集,并在24孔板中以多种E:T比例(1:1和0.5:1)与NALM-6-GL和Raji-GL细胞系共培养。24小时后,收集细胞,用BD FacsCanto II Plus仪器(BD Biosciences)检测靶细胞,数据使用Flow Jo v.10软件(Tree star,Inc.Ashland,OR)分析。对于化学发光法,将细胞(96孔板)加入底物,然后使用PerkinElmer光化学成像系统收集信号。用活体成像分析系统分析荧光信号的强度来评价CAR-T细胞的细胞毒性。实验包括三组:(1)仅有目标细胞;(2)事先未与MSC共培养的CAR-T细胞与目标细胞共培养;(3)事先与MSC共培养的CAR-T细胞,然后收集与目标细胞共培养。Two methods were used to detect the cytotoxic function of CAR-T cells. One was to detect the GFP signal of target cells by flow cytometry, and the other was to detect the chemiluminescence of target cells using a live imaging analysis system. CAR-T cells were first co-cultured with MSC cells at a ratio of 10:1 for 24 hours, then re-collected and co-cultured with NALM-6-GL and Raji-GL cell lines at various E:T ratios (1:1 and 0.5:1) in 24-well plates. After 24 hours, cells were collected and target cells were detected using a BD FacsCanto II Plus instrument (BD Biosciences), and data were analyzed using Flow Jo v.10 software (Treestar, Inc. Ashland, OR). For the chemiluminescence method, cells (96-well plates) were added with substrates, and the signals were collected using a PerkinElmer photochemical imaging system. The intensity of the fluorescence signal was analyzed using a live imaging analysis system to evaluate the cytotoxicity of CAR-T cells. The experiment included three groups: (1) target cells only; (2) CAR-T cells co-cultured with target cells without prior co-culture with MSCs; and (3) CAR-T cells co-cultured with MSCs beforehand and then collected for co-culture with target cells.
细胞因子产生测定Cytokine production assay
CAR-T细胞首先与MSC细胞以10:1的比例共培养24小时,然后重新收集并与NALM-6-GL和Raji-GL细胞系以E:T=1:1的比例在24孔板上共培养。共培养24小时后,收集上清液进行细胞因子检测。根据制造商的程序,使用商业细胞计数珠阵列(CBA)试剂盒(BD Biosciences)评估共培养细胞上清液中人干扰素γ(IFN-γ)、肿瘤坏死因子α(TNF-α)、IL-4、IL-6、IL-10和IL-17A的表达。人IFN-γ的水平还通过使用
Figure PCTCN2022128989-appb-000002
ELISA试剂盒(R&D,Minnesota,USA)按照制造商的说明进行评估。
CAR-T cells were first co-cultured with MSC cells at a ratio of 10:1 for 24 hours, and then re-collected and co-cultured with NALM-6-GL and Raji-GL cell lines at a ratio of E:T = 1:1 on a 24-well plate. After 24 hours of co-culture, the supernatant was collected for cytokine detection. The expression of human interferon gamma (IFN-γ), tumor necrosis factor alpha (TNF-α), IL-4, IL-6, IL-10, and IL-17A in the supernatant of co-cultured cells was evaluated using a commercial cytometric bead array (CBA) kit (BD Biosciences) according to the manufacturer's procedures. The level of human IFN-γ was also measured by using
Figure PCTCN2022128989-appb-000002
The ELISA kit (R&D, Minnesota, USA) was used for evaluation according to the manufacturer's instructions.
为了检测细胞因子IL2,CAR-T细胞在不含IL2的培养基中培养3天,然后与间充质干细胞和靶细胞在不含IL2的相同培养基中共同培养。收集上清液,使用
Figure PCTCN2022128989-appb-000003
ELISA试剂盒(R&D,Minnesota,USA)检测IL2的释放。
To detect the cytokine IL2, CAR-T cells were cultured in medium without IL2 for 3 days and then co-cultured with mesenchymal stem cells and target cells in the same medium without IL2. The supernatant was collected and used
Figure PCTCN2022128989-appb-000003
The release of IL2 was detected by ELISA kit (R&D, Minnesota, USA).
细胞亚群分析Cell Subpopulation Analysis
CAR-T细胞与MSC细胞以10:1的比例共培养5天,然后重新收集并在24孔板中以10:1的E:T比例与NALM-6-GL和Raji-GL细胞系共培养24小时,然后收集共培养后的细胞进 行流式细胞术分析T细胞亚群。CAR-T cells were co-cultured with MSC cells at a ratio of 10:1 for 5 days, then re-collected and co-cultured with NALM-6-GL and Raji-GL cell lines at an E:T ratio of 10:1 in 24-well plates for 24 hours. The co-cultured cells were then collected for flow cytometry analysis of T cell subsets.
脑内注射NALM-GL细胞的异种移植小鼠模型Xenograft mouse model with intracerebral injection of NALM-GL cells
六至八周大的雌性NOD-SCID小鼠购自Charles River实验室,并在无病原体的条件下精心饲养。适应性喂养一周后,经尾静脉给NOD-SCID小鼠注射1×10 6个NALM-6-GL细胞,构建肿瘤异种移植小鼠模型。共12只小鼠被随机分为3组。对照组是只注射了NALM-6-GL的模型组(N=4)。实验组为CAR-T细胞治疗组,包括CAR-T细胞与间充质干细胞共培养(N=4)和不与间充质干细胞共培养的CAR-T细胞(N=4)。实验组在模型构建24小时后同样经尾静脉注射1×10 7/天CAR-T细胞,连续三天。使用活体成像分析系统(IVIS,Xenogen,Alameda,CA,USA),每周通过生物发光成像监测肿瘤的进展。根据首都医科大学附属北京世纪坛医院的实验动物管理规定,小鼠达到安乐死标准后被安乐死。 Female NOD-SCID mice aged six to eight weeks were purchased from Charles River Laboratories and carefully raised under pathogen-free conditions. After one week of adaptive feeding, NOD-SCID mice were injected with 1×10 6 NALM-6-GL cells via the tail vein to construct a tumor xenograft mouse model. A total of 12 mice were randomly divided into three groups. The control group was a model group injected with NALM-6-GL only (N=4). The experimental group was a CAR-T cell treatment group, including CAR-T cells co-cultured with mesenchymal stem cells (N=4) and CAR-T cells not co-cultured with mesenchymal stem cells (N=4). The experimental group was also injected with 1×10 7 /day CAR-T cells via the tail vein 24 hours after model construction for three consecutive days. The progression of the tumor was monitored weekly by bioluminescence imaging using an in vivo imaging analysis system (IVIS, Xenogen, Alameda, CA, USA). According to the experimental animal management regulations of Beijing Shijitan Hospital affiliated to Capital Medical University, mice were euthanized after reaching the euthanasia criteria.
统计分析Statistical Analysis
所有数据至少包含三个生物重复,用GraphPad Prism 7软件(GraphPad Software,San Diego,CA)进行分析。数据以平均值±SEM表示,使用非配对t检验来评估差异。异种移植小鼠的总体存活率用Kaplan-Meier方法测量,用Cox比例风险回归分析进行分组比较。如果P<0.05认为差异具有统计学显著性。All data included at least three biological replicates and were analyzed using GraphPad Prism 7 software (GraphPad Software, San Diego, CA). Data are presented as mean ± SEM, and differences were assessed using unpaired t-tests. Overall survival of xenografted mice was measured using the Kaplan-Meier method, and group comparisons were performed using Cox proportional hazards regression analysis. Differences were considered statistically significant if P < 0.05.
实施例2.胎盘间充质干细胞的扩增和特征分析Example 2. Expansion and Characterization of Placental Mesenchymal Stem Cells
我们从8位正常捐赠者的胎盘中分离和培养了间充质干细胞,并参照ISCT列出的标准化标准,通过识别其形态、免疫表型和分化潜能来确定细胞是否是间充质干细胞。使用间充质干细胞标准培养条件和方法,从所有样本中都成功培养出贴壁细胞。这些细胞粘附在塑料上,显示出均匀的纺锤形成纤维细胞的形态(图1)。我们通过台盼蓝染色计算从P1到P6的生长动态,发现来自不同供体胎盘的间充质干细胞的增殖情况基本相同(图2)。我们通过流式细胞术检测了P3的间充质干细胞的免疫表型。结果显示,CD105(MSC的标志之一)呈阳性(>95%),而CD45(造血细胞标记)为阴性(图3)。接下来,我们在体外评估了MSC的诱导分化潜能。来自P3的间充质干细胞被暴露在成骨诱导培养基中,并进行了3-4周的培养。然后用茜素红染色检测钙的沉积。结果显示,在成骨细胞诱导的细胞中检测到明显的钙沉积(图4)。We isolated and cultured mesenchymal stem cells from the placentas of eight normal donors and determined whether the cells were mesenchymal stem cells by identifying their morphology, immunophenotype, and differentiation potential, referring to the standardized criteria listed by ISCT. Adherent cells were successfully cultured from all samples using standard culture conditions and methods for mesenchymal stem cells. These cells adhered to the plastic and showed a uniform spindle-forming fibroblast morphology (Figure 1). We calculated the growth dynamics from P1 to P6 by trypan blue staining and found that the proliferation of mesenchymal stem cells from different donor placentas was basically the same (Figure 2). We detected the immunophenotype of mesenchymal stem cells from P3 by flow cytometry. The results showed that CD105 (one of the markers of MSC) was positive (>95%), while CD45 (a hematopoietic cell marker) was negative (Figure 3). Next, we evaluated the induction differentiation potential of MSC in vitro. Mesenchymal stem cells from P3 were exposed to osteogenic induction medium and cultured for 3-4 weeks. Calcium deposition was then detected by alizarin red staining. The results showed that significant calcium deposition was detected in osteoblast-induced cells ( FIG. 4 ).
在探索MSC对CAR-T细胞功能的影响之前,我们首先检测了MSC表面是否表达常见的肿瘤靶抗原。流式细胞仪的结果显示,uPAR在间充质干细胞的表面表达,但CD19和IL13Rα2没有表达(图5)。Before exploring the effect of MSC on CAR-T cell function, we first detected whether common tumor target antigens were expressed on the surface of MSC. The results of flow cytometry showed that uPAR was expressed on the surface of mesenchymal stem cells, but CD19 and IL13Rα2 were not expressed (Figure 5).
实施例3.CAR-T细胞的产生Example 3. Generation of CAR-T cells
按照如实施例1中所述的方法构建如前所述的三种CAR分子并转导到T细胞中获得CAR-T细胞,通过流式细胞术验证。The three CAR molecules described above were constructed according to the method described in Example 1 and transduced into T cells to obtain CAR-T cells, which were verified by flow cytometry.
实施例4.pMSC在体外增强了CD19-IL15靶向CAR-T细胞的功能Example 4. pMSCs enhance the function of CD19-IL15 targeted CAR-T cells in vitro
我们从四个方面评估了MSC对CD19-IL15 CAR-T功能的影响,包括CAR-T增殖、CD107a激活、细胞因子释放和细胞毒性。We evaluated the effects of MSCs on CD19-IL15 CAR-T function from four aspects, including CAR-T proliferation, CD107a activation, cytokine release, and cytotoxicity.
在探索MSC对CD19-IL15 CAR-T细胞增殖影响的实验中,首先将CAR-T细胞与 MSC以E:T=10:1的比例共培养24小时,然后收集CAR-T细胞,用CFSE标记,并与NALM-6细胞以E:T的比例共同培养。T的比例为1:1。三天后,收获细胞并与适量的抗人类CD3抗体孵化,进行流式细胞术分析。以CFSE标记为对照,在相同条件下将未与MSC共培养的CAR-T细胞与/不与NALM-6细胞共培养。结果显示MSC促进了CAR-T细胞的增殖(图8)。In the experiment to explore the effect of MSC on the proliferation of CD19-IL15 CAR-T cells, CAR-T cells were first co-cultured with MSC at an E:T = 10:1 ratio for 24 hours, and then the CAR-T cells were collected, labeled with CFSE, and co-cultured with NALM-6 cells at an E:T ratio of 1:1. Three days later, the cells were harvested and incubated with an appropriate amount of anti-human CD3 antibody for flow cytometry analysis. Using CFSE labeling as a control, CAR-T cells that were not co-cultured with MSC were co-cultured with/without NALM-6 cells under the same conditions. The results showed that MSC promoted the proliferation of CAR-T cells (Figure 8).
接下来,我们探讨了MSC对CAR-T细胞CD107a激活的影响。CAR-T细胞也提前与MSC以10:1的比例共同培养24小时,然后与/不与NALM-6以1:1的比例共同培养6小时。与/不与NALM-6共培养的未经MSC共培养的CAR-T细胞作为对照。与对照组相比,当CAR-T细胞事先与MSC共培养,然后再与NALM-6共培养,则CD107a阳性细胞比例呈现出明显增加(图9,22.7±1.36 vs 30.47±1.26,P=0.0138)。Next, we explored the effect of MSC on CD107a activation of CAR-T cells. CAR-T cells were also co-cultured with MSCs at a ratio of 10:1 for 24 hours in advance, and then co-cultured with/without NALM-6 at a ratio of 1:1 for 6 hours. CAR-T cells that were not co-cultured with MSCs and co-cultured with/without NALM-6 served as controls. Compared with the control group, when CAR-T cells were co-cultured with MSCs in advance and then co-cultured with NALM-6, the proportion of CD107a-positive cells showed a significant increase (Figure 9, 22.7±1.36 vs 30.47±1.26, P=0.0138).
然后,我们收集共培养的细胞的上清液,通过细胞计数珠阵列试剂盒和ELISA检测细胞因子的释放。CAR-T细胞与MSC共培养24小时,然后重新收集并与/不与NALM-6或Raji细胞共培养过夜。如图10的A(涉及NALM-6的结果)和B(涉及Raji的结果)所示,无论是否与MSC共培养,CD19-IL15 CAR-T细胞在抗肿瘤过程中,细胞因子IL10、IL4和IL6、IFN-γ、TNF-α和IL17的释放没有明显变化。当CD19-IL15 CAR-T细胞攻击NALM-6细胞时,发现与MSC共培养的CAR-T细胞中IL4和IL10的分泌量明显增加,而IFN-γ、IL17A和TNF-α的分泌量没有明显变化。当CD19-IL15 CAR-T细胞攻击Raji细胞时,发现与间充质干细胞共培养的CAR-T细胞中IL4和IL17A的分泌量明显增加,而IFN-γ、IL17A和IL10的分泌量没有明显变化。We then collected the supernatants of the co-cultured cells and assayed the release of cytokines by cytometry bead array kit and ELISA. CAR-T cells were co-cultured with MSCs for 24 h and then re-collected and co-cultured with/without NALM-6 or Raji cells overnight. As shown in Figure 10A (results involving NALM-6) and B (results involving Raji), the release of cytokines IL10, IL4 and IL6, IFN-γ, TNF-α, and IL17 by CD19-IL15 CAR-T cells during the anti-tumor process did not change significantly, regardless of whether they were co-cultured with MSCs. When CD19-IL15 CAR-T cells attacked NALM-6 cells, it was found that the secretion of IL4 and IL10 in CAR-T cells co-cultured with MSCs increased significantly, while the secretion of IFN-γ, IL17A, and TNF-α did not change significantly. When CD19-IL15 CAR-T cells attacked Raji cells, it was found that the secretion of IL4 and IL17A in CAR-T cells co-cultured with mesenchymal stem cells was significantly increased, while the secretion of IFN-γ, IL17A and IL10 did not change significantly.
最后,我们评估了MSC对CD19-IL15 CAR-T细胞毒性的影响。将CAR-T细胞与MSC以10:1的比例共培养24小时,然后收集CAR-T细胞,与NALM-6-GL和Raji-GL细胞以多种E:T比例共同培养。通过流式细胞仪检测GFP荧光(分别见图11和12)和IVIS成像系统检测荧光素酶活性(分别见图13的A和B)来分析肿瘤细胞的存活情况,从而评估细胞毒功能。如上述附图所表明的,CD19-IL15 CAR-T细胞与间充质干细胞共同培养后,其杀伤能力被显著增强。Finally, we evaluated the effect of MSC on the cytotoxicity of CD19-IL15 CAR-T cells. CAR-T cells were co-cultured with MSCs at a ratio of 10:1 for 24 hours, and then CAR-T cells were collected and co-cultured with NALM-6-GL and Raji-GL cells at various E:T ratios. The survival of tumor cells was analyzed by flow cytometry to detect GFP fluorescence (see Figures 11 and 12, respectively) and luciferase activity by IVIS imaging system (see Figure 13, A and B, respectively) to evaluate cytotoxic function. As shown in the above figures, the killing ability of CD19-IL15 CAR-T cells was significantly enhanced after co-culture with mesenchymal stem cells.
实施例5.间充质干细胞在体外增强靶向IL13Rα2的CAR-T细胞功能Example 5. Mesenchymal stem cells enhance the function of CAR-T cells targeting IL13Rα2 in vitro
我们还以IL13Rα2靶向的CAR-T细胞为例,探讨了MSC对CAR-T细胞靶向胶质瘤功能的影响。我们从CD107a活性、细胞因子释放和杀伤能力三个方面评估了间充质干细胞对CAR-T细胞靶向IL13Rα2胶质瘤功能的影响。详细的实验方法与前一实施例,即评估MSC对CD19-IL15 CAR-T细胞功能影响的研究方法一致。如图14-16所示,在1:1的E:T的比例下,MSC没有引起IL13Rα2 CAR-T细胞的CD107a活化增加,也不影响IFN-γ和IL6的释放(图14、15),但提高了CAR-T细胞的杀伤能力(图16)。We also took IL13Rα2-targeted CAR-T cells as an example to explore the effect of MSC on the function of CAR-T cells targeting glioma. We evaluated the effect of mesenchymal stem cells on the function of CAR-T cells targeting IL13Rα2 glioma from three aspects: CD107a activity, cytokine release, and killing ability. The detailed experimental method is consistent with the previous embodiment, that is, the research method for evaluating the effect of MSC on the function of CD19-IL15 CAR-T cells. As shown in Figures 14-16, at an E:T ratio of 1:1, MSC did not cause an increase in CD107a activation of IL13Rα2 CAR-T cells, nor did it affect the release of IFN-γ and IL6 (Figures 14, 15), but it increased the killing ability of CAR-T cells (Figure 16).
实施例6.间充质干细胞在体外抑制了uPAR靶向CAR-T细胞的功能Example 6. Mesenchymal stem cells inhibited the function of uPAR-targeted CAR-T cells in vitro
我们发现,MSC对不同的CAR-T细胞有不同的功能。MSC导致了uPAR靶向CAR-T细胞杀伤能力的下降。我们探讨了MSC对uPAR靶向CAR-T细胞功能的影响,发现MSC增强了CD107a的激活(图17),促进了细胞因子IFN-γ的释放(图18),但严重阻碍了其细胞 毒性功能(图19-20)。当E:T比例为2:1时,MSC使CAR-T细胞的杀伤能力减少22%。当E:T比例为2:1时,间充质干细胞使CAR-T细胞的杀伤能力下降22%,并严重破坏杀伤能力;在E:T比例为1:1时,杀伤力严重受损,导致杀伤力下降高达92%。进一步的研究显示,当E:T比率为1:1时,uPAR CAR-T细胞杀伤高达77%的间充质干细胞(图21)。We found that MSCs have different functions on different CAR-T cells. MSCs caused a decrease in the killing ability of uPAR-targeted CAR-T cells. We explored the effect of MSCs on the function of uPAR-targeted CAR-T cells and found that MSCs enhanced the activation of CD107a (Figure 17) and promoted the release of cytokine IFN-γ (Figure 18), but severely hindered its cytotoxic function (Figures 19-20). When the E:T ratio was 2:1, MSCs reduced the killing ability of CAR-T cells by 22%. When the E:T ratio was 2:1, mesenchymal stem cells reduced the killing ability of CAR-T cells by 22% and severely damaged the killing ability; at an E:T ratio of 1:1, the killing ability was severely impaired, resulting in a decrease in killing ability of up to 92%. Further studies showed that when the E:T ratio was 1:1, uPAR CAR-T cells killed up to 77% of mesenchymal stem cells (Figure 21).
实施例7.MSC调节T细胞分化Example 7. MSC regulates T cell differentiation
探讨了间充质干细胞如何调节CAR-T细胞的功能,并以CD19-IL15 CAR-T细胞为研究对象,寻找间充质干细胞促进CAR-T功能的可能机制。我们发现,间充质干细胞影响了CAR-T细胞的分化。将CAR-T细胞与MSC共培养一天,并收集与/不与NALM-6细胞共培养。然后收集CAR-T细胞来检测T细胞亚群。如图22-23所示,与CD19-IL15 CAR-T细胞相比,CD3+细胞中TCM表型的比例在与MSC共培养的CAR-T细胞中明显增加。进一步分析发现,其在CD4+细胞和CD8+细胞中的比例也明显增加。qRT-PCR的结果显示,与T细胞分化有关的转录因子TCF-7的表达明显上调(图24)。同时,我们检测了T细胞增殖和分化的必要细胞因子IL2的分泌。事先将CAR-T细胞与MSC共培养24小时,然后与NALM-6和Raji细胞在不含IL2的X-VIVO-15培养基中共培养过夜。收集上清液,用ELISA检测IL2的含量。如图25所示,与CD19-IL15 CAR-T细胞组相比,MSC处理的CAR-T细胞释放的IL2没有明显变化,但当与NALM-6和Raji等肿瘤细胞共培养时,MSC处理的CAR-T细胞分泌的IL2明显增加。We explored how mesenchymal stem cells regulate the function of CAR-T cells, and used CD19-IL15 CAR-T cells as the research object to find the possible mechanism by which mesenchymal stem cells promote CAR-T function. We found that mesenchymal stem cells affected the differentiation of CAR-T cells. CAR-T cells were co-cultured with MSC for one day and collected with or without co-culture with NALM-6 cells. CAR-T cells were then collected to detect T cell subsets. As shown in Figures 22-23, compared with CD19-IL15 CAR-T cells, the proportion of TCM phenotype in CD3+ cells was significantly increased in CAR-T cells co-cultured with MSC. Further analysis found that its proportion in CD4+ cells and CD8+ cells was also significantly increased. The results of qRT-PCR showed that the expression of TCF-7, a transcription factor related to T cell differentiation, was significantly upregulated (Figure 24). At the same time, we detected the secretion of IL2, a necessary cytokine for T cell proliferation and differentiation. CAR-T cells were co-cultured with MSCs for 24 hours in advance and then co-cultured with NALM-6 and Raji cells in X-VIVO-15 medium without IL2 overnight. The supernatant was collected and the IL2 content was detected by ELISA. As shown in Figure 25, compared with the CD19-IL15 CAR-T cell group, the IL2 released by MSC-treated CAR-T cells did not change significantly, but when co-cultured with tumor cells such as NALM-6 and Raji, the IL2 secreted by MSC-treated CAR-T cells increased significantly.
实施例8.间充质干细胞增强体内CD19-IL15 CAR-T的抗肿瘤能力Example 8. Mesenchymal stem cells enhance the anti-tumor ability of CD19-IL15 CAR-T in vivo
通过尾静脉注射NALM-6-eGFP-Luc(NALM-6-GL)细胞构建异种移植小鼠模型,如图26所示,提前一天注射1×10 6NALM-6-GL细胞,在第0天通过尾静脉注射1×10 7个CD19-IL15 CAR-T细胞。实验组小鼠注射的CAR-T细胞事先与间充质干细胞体外共培养1天,而对照组小鼠注射的CAR-T细胞未与MSC共培养。另外还设置未注射CAR-T的非治疗对照组。定期对小鼠进行荧光成像。如图27和图28所示,CAR-T治疗组的肿瘤负荷明显低于对照组,但与间充质干细胞处理的CAR-T组与只用CAR-T组之间没有明显差异。观察并统计了小鼠的生存情况。结果发现,CAR-T细胞与间充质干细胞共培养后,小鼠的生存时间明显延长(图29)。 A xenograft mouse model was constructed by tail vein injection of NALM-6-eGFP-Luc (NALM-6-GL) cells. As shown in FIG26 , 1×10 6 NALM-6-GL cells were injected one day in advance, and 1×10 7 CD19-IL15 CAR-T cells were injected through the tail vein on day 0. The CAR-T cells injected into the experimental group mice were co-cultured with mesenchymal stem cells in vitro for 1 day in advance, while the CAR-T cells injected into the control group mice were not co-cultured with MSCs. In addition, a non-treatment control group without CAR-T injection was set up. Fluorescence imaging of mice was performed regularly. As shown in FIG27 and FIG28 , the tumor burden in the CAR-T treatment group was significantly lower than that in the control group, but there was no significant difference between the CAR-T group treated with mesenchymal stem cells and the CAR-T group alone. The survival of the mice was observed and counted. The results showed that after co-culture of CAR-T cells with mesenchymal stem cells, the survival time of mice was significantly prolonged ( FIG29 ).
讨论discuss
在上述实施例中,胎盘间充质干细胞增强了CD19-IL15 CAR-T细胞和IL13 CAR-T细胞的抗肿瘤功能,但抑制了uPAR CAR-T细胞的抗肿瘤功能。具体地,结果显示,间充质干细胞促进CD19-IL15 CAR-T细胞的增殖和激活,并促进细胞因子IL2和IL4的释放。IL-2参与塑造决定T细胞命运的转录和代谢过程。它是T细胞激活和增殖的重要细胞因子,并被认为是治疗癌症的一种手段。IL-4通常被认为是一种典型的Th2细胞因子,但有报道称它能促进人类胸腺和脐带血中的CD4+T细胞转化为CD8+T细胞,并促进记忆性CD8+T细胞的数量和功能,从而促进而非削弱Th1细胞免疫反应。我们还发现,间充质干细胞促进了CD3+T细胞中TCM表型细胞的比例增加,包括CD4+和CD8+T细胞。TCF-7,一种 参与TCM表型细胞分化和发展的转录激活剂,表达量显著增加。众所周知,具有较少分化表型的CAR-T细胞,如TN和TCM表型,与自我更新、增殖和存活的特性增加有关。In the above examples, placental mesenchymal stem cells enhanced the anti-tumor function of CD19-IL15 CAR-T cells and IL13 CAR-T cells, but inhibited the anti-tumor function of uPAR CAR-T cells. Specifically, the results showed that mesenchymal stem cells promoted the proliferation and activation of CD19-IL15 CAR-T cells and promoted the release of cytokines IL2 and IL4. IL-2 is involved in shaping the transcriptional and metabolic processes that determine the fate of T cells. It is an important cytokine for T cell activation and proliferation and is considered a means of treating cancer. IL-4 is generally considered to be a typical Th2 cytokine, but it has been reported that it can promote the conversion of CD4+T cells in human thymus and umbilical cord blood into CD8+T cells and promote the number and function of memory CD8+T cells, thereby promoting rather than weakening Th1 cell immune responses. We also found that mesenchymal stem cells promoted an increase in the proportion of TCM phenotype cells in CD3+T cells, including CD4+ and CD8+T cells. TCF-7, a transcriptional activator involved in the differentiation and development of TCM phenotype cells, was significantly increased in expression. It is well known that CAR-T cells with less differentiated phenotypes, such as TN and TCM phenotypes, are associated with increased properties of self-renewal, proliferation, and survival.
CAR-T细胞疗法的疗效与CAR-T细胞的活性和持久性密切相关,因此,上述所有结果很好地解释了为什么间充质干细胞可以增强CD19-IL15 CAR-T细胞的抗肿瘤能力。The efficacy of CAR-T cell therapy is closely related to the activity and persistence of CAR-T cells. Therefore, all the above results well explain why mesenchymal stem cells can enhance the anti-tumor ability of CD19-IL15 CAR-T cells.
本文还评估了间充质干细胞是否能促进CD19-IL15 CAR-T细胞在异种移植小鼠模型中的抗肿瘤疗效。结果显示,体外与间充质干细胞共培养的CAR-T细胞明显延长了NALM-6-GL成瘤小鼠的生存时间。在以往报道中,BM-MSC与CD19CAR-T和肿瘤细胞共同培养,发现MSC并不能保护肿瘤细胞,也不影响CAR-T细胞的抗肿瘤能力(Zanetti SR等人,Bone marrow MSC from pediatric patients with B-ALL highly immunosuppress T-cell responses but do not compromise CD19-CAR T-cell activity.J Immunother Cancer 2020;8:e001419.)。然而,在本文中,CD19-IL15 CAR-T细胞首先被MSC刺激,然后收集起来与肿瘤细胞共培养,结果显示其抗肿瘤能力被明显增强。CAR-T细胞在MSC刺激下发生一系列的变化,如表型、细胞因子释放、活化和增殖能力等,导致其抗肿瘤功能的增强。This paper also evaluated whether mesenchymal stem cells can promote the anti-tumor efficacy of CD19-IL15 CAR-T cells in a xenograft mouse model. The results showed that CAR-T cells co-cultured with mesenchymal stem cells in vitro significantly prolonged the survival time of NALM-6-GL tumor-bearing mice. In previous reports, BM-MSCs were co-cultured with CD19CAR-T and tumor cells, and it was found that MSCs could not protect tumor cells or affect the anti-tumor ability of CAR-T cells (Zanetti SR et al., Bone marrow MSCs from pediatric patients with B-ALL highly immunosuppress T-cell responses but do not compromise CD19-CAR T-cell activity. J Immunother Cancer 2020; 8: e001419.). However, in this paper, CD19-IL15 CAR-T cells were first stimulated by MSCs and then collected and co-cultured with tumor cells, and the results showed that their anti-tumor ability was significantly enhanced. CAR-T cells undergo a series of changes under MSC stimulation, such as phenotype, cytokine release, activation and proliferation ability, leading to the enhancement of their anti-tumor function.
在这个实验中,我们还发现间充质干细胞抑制了uPAR CAR-T细胞的功能。之前的流式分析结果表明MSC表达uPAR而不表达CD19和IL13Rα2。因此,推测CD19-IL15 CAR-T细胞和IL13Rα2 CAR-T细胞与间充质干细胞共培养时没有杀伤间充质干细胞(数据未显示),但uPAR CAR-T细胞可以经由其靶抗原特异性而攻击间充质干细胞。从而,uPAR CAR-T细胞可能因为杀伤MSC而衰竭,并导致其抗肿瘤能力下降。这一现象表明,间充质干细胞是否对CAR-T细胞的功能有增强作用,一个关键因素在于间充质干细胞是否表达相应的靶抗原。In this experiment, we also found that mesenchymal stem cells inhibited the function of uPAR CAR-T cells. Previous flow cytometry results showed that MSCs expressed uPAR but did not express CD19 and IL13Rα2. Therefore, it is speculated that CD19-IL15 CAR-T cells and IL13Rα2 CAR-T cells did not kill mesenchymal stem cells when co-cultured with mesenchymal stem cells (data not shown), but uPAR CAR-T cells can attack mesenchymal stem cells through their target antigen specificity. As a result, uPAR CAR-T cells may be exhausted due to killing MSCs, resulting in a decrease in their anti-tumor ability. This phenomenon shows that whether mesenchymal stem cells have an enhancing effect on the function of CAR-T cells is a key factor in whether mesenchymal stem cells express the corresponding target antigen.

Claims (47)

  1. 间充质干细胞用于增强针对疾病的免疫疗法的用途,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫疗法所针对的抗原。Use of mesenchymal stem cells for enhancing immunotherapy against a disease, wherein the mesenchymal stem cells do not express or substantially do not express the antigen targeted by the immunotherapy on their surface.
  2. 权利要求1的用途,其中免疫疗法是细胞疗法。The use of claim 1, wherein the immunotherapy is cell therapy.
  3. 权利要求2的用途,其中所述细胞疗法中所用治疗性细胞是免疫效应细胞,任选地,经修饰的免疫效应细胞。The use of claim 2, wherein the therapeutic cells used in the cell therapy are immune effector cells, optionally modified immune effector cells.
  4. 权利要求3的用途,其中免疫效应细胞是T细胞。The use of claim 3, wherein the immune effector cells are T cells.
  5. 权利要求3的用途,其中免疫效应细胞是CAR-T细胞。The use of claim 3, wherein the immune effector cells are CAR-T cells.
  6. 权利要求3的用途,其中免疫效应细胞选自TIL、CAR-NK,TCR-T、CAR-DC、已负载抗原的DC、B细胞中的一种或多种。The use of claim 3, wherein the immune effector cells are selected from one or more of TIL, CAR-NK, TCR-T, CAR-DC, DC loaded with antigen, and B cells.
  7. 权利要求1-6任一项的用途,其中所述疾病是肿瘤或自身免疫病。The use according to any one of claims 1 to 6, wherein the disease is a tumor or an autoimmune disease.
  8. 权利要求7的用途,其中所述疾病是肿瘤。The use according to claim 7, wherein the disease is a tumor.
  9. 权利要求8的用途,其中所述免疫疗法针对肿瘤所表达的一种或多种肿瘤相关抗原。The use of claim 8, wherein the immunotherapy is directed against one or more tumor-associated antigens expressed by the tumor.
  10. 权利要求9的用途,其中所述一种或多种肿瘤相关抗原选自CD19、CD123、CD22、CD30、CD171、CS-1(也被称为CD2子集1、CRACC、SLAMF7、CD319、和19A24)、C型凝集素样分子-1(CLL-1或CLECL1)、CD33、表皮生长因子受体变体III(EGFRvIII)、神经节苷脂G2(GD2)、神经节苷脂GD3(aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、TNF受体家族成员B细胞成熟(BCMA)、Tn抗原((Tn Ag)或(GalNAcα-Ser/Thr))、前列腺特异性膜抗原(PSMA)、受体酪氨酸激酶样孤儿受体1(ROR1)、Fms样酪氨酸激酶3(FLT3)、肿瘤相关的糖蛋白72(TAG72)、CD38、CD44v6、癌胚抗原(CEA)、上皮细胞粘附分子(EPCAM)、B7H3(CD276)、KIT(CD117)、白介素13受体亚基α-2(IL-13Rα2或CD213A2)、间皮素、白介素11受体α(IL-11Ra)、前列腺干细胞抗原(PSCA)、蛋白酶丝氨酸21(Testisin或PRSS21)、血管内皮生长因子受体2(VEGFR2)、路易斯(Y)抗原、CD24、血小板衍生生长因子受体β(PDGFR-β)、阶段特异性胚胎抗原-4(SSEA-4)、CD20、叶酸受体α、受体酪氨酸蛋白激酶ERBB2(Her2/neu)、细胞表面相关的粘蛋白1(MUC1)、表皮生长因子受体(EGFR)、神经细胞粘附分子(NCAM)、Prostase、前列腺酸性磷酸酶(PAP)、突变的延伸因子2(ELF2M)、肝配蛋白B2、成纤维细胞活化蛋白α(FAP)、胰岛素样生长因子1受体(IGF-I受体)、碳酸酐酶IX(CAIX)、蛋白酶体(Prosome、Macropain)亚基、β型、9(LMP2)、糖蛋白100(gp100)、由断点簇区(BCR)和Alelson鼠白血病病毒癌基因同源物1(AB1)组成的癌基因融合蛋白(bcr-abl)、酪氨酸酶、肝配蛋白A型受体2(EphA2)、岩藻糖基GM1、唾液酸基路易斯粘附分子(sLe)、神经节苷脂GM3(aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、转谷氨酰胺酶5(TGS5)、高分子量黑素瘤相关抗原(HMWMAA)、邻乙酰基GD2神经节苷脂(OAcGD2)、叶酸受体β、肿瘤血管内皮标记1(TEM1/CD248)、肿瘤血管内皮标记7相关的(TEM7R)、Claudin 6(CLDN6)、促甲状腺激素受体(TSHR)、G蛋白偶联受体C类5组、成员D(GPRC5D)、X染色体开放阅读框61(CXORF61)、CD97、CD179a、间变性淋巴瘤激酶(ALK)、聚唾液酸、胎盘特异性1(PLAC1)、globoH  glycoceramide的己糖部分(GloboH)、乳腺分化抗原(NY-BR-1)、uroplakin 2(UPK2)、甲型肝炎病毒细胞受体1(HAVCR1)、肾上腺素受体β3(ADRB3)、pannexin 3(PANX3)、G蛋白偶联受体20(GPR20)、淋巴细胞抗原6复合物、基因座K9(LY6K)、嗅觉受体51E2(OR51E2)、TCRγ交替阅读框蛋白(TARP)、肾母细胞瘤蛋白(WT1)、癌/睾丸抗原1(NY-ESO-1)、癌症/睾丸抗原2(LAGE-1A)、黑素瘤相关抗原1(MAGE-A1)、ETS易位变异基因6,位于染色体12p(ETV6-AML)、精子蛋白17(SPA17)、X抗原家族,成员1A(XAGE1)、血管生成素结合细胞表面受体2(Tie2)、黑素瘤癌睾丸抗原-1(MAD-CT-1)、黑素瘤癌睾丸抗原-2(MAD-CT-2)、FOS相关抗原1、肿瘤蛋白质p53(p53)、p53突变体、prostein、存活蛋白、端粒酶、前列腺癌肿瘤抗原-1(PCTA-1或半乳凝素8)、由T细胞识别的黑素瘤抗原1(MelanA或MART1)、大鼠肉瘤(Ras)突变体、人端粒酶逆转录酶(hTERT)、肉瘤易位断点、细胞凋亡的黑素瘤抑制剂(ML-IAP)、ERG(跨膜蛋白酶,丝氨酸2(TMPRSS2)ETS融合基因)、N-乙酰葡糖胺基转移酶V(NA17)、配对盒蛋白Pax-3(PAX3)、雄激素受体、细胞周期蛋白B1、V-myc鸟髓细胞瘤病病毒癌基因神经母细胞瘤衍生的同源物(MYCN)、Ras同源物家族成员C(RhoC)、酪氨酸酶相关蛋白2(TRP-2)、细胞色素P450 1B1(CYP1B1)、CCCTC结合因子(锌指蛋白)样(BORIS或印记位点的调节物的兄弟)、由T细胞识别的鳞状细胞癌抗原3(SART3)、配对盒蛋白Pax-5(PAX5)、proacrosin结合蛋白sp32(OY-TES1)、淋巴细胞特异性蛋白酪氨酸激酶(LCK)、A激酶锚定蛋白4(AKAP-4)、滑膜肉瘤,X断点2(SSX2)、高级糖化终产物受体(RAGE-1)、肾uibiguitous 1(RU1)、肾uibiguitous 2(RU2)、legumain、人类乳头瘤病毒E6(HPV E6)、人类乳头瘤病毒E7(HPV E7)、肠羧基酯酶、突变的热休克蛋白70-2(mut hosp 70-2)、CD79a、CD79b、CD72、白细胞相关免疫球蛋白样受体1(LAIR1)、IgA受体的Fc片段(FCAR或CD89)、白细胞免疫球蛋白样受体亚家族A成员2(LILRA2)、CD300分子样家族成员f(CD300LF)、C型凝集素结构域家族12成员A(CLEC12A)、骨髓基质细胞抗原2(BST2)、含有EGF样模块粘蛋白样激素受体样2(EMR2)、淋巴细胞抗原75(LY75)、磷脂酰肌醇蛋白聚糖-3(GPC3)、Fc受体样5(FCRL5)、与免疫球蛋白λ样多肽1(IGLL1)中的一种或多种。The use of claim 9, wherein the one or more tumor-associated antigens are selected from CD19, CD123, CD22, CD30, CD171, CS-1 (also known as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24), C-type lectin-like molecule-1 (CLL-1 or CLECL1), CD33, epidermal growth factor receptor variant III (EGFRvIII), ganglioside G2 (GD2), ganglioside GD3 (aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer), TNF receptor family member B cell maturation (BCMA), Tn antigen ((Tn Ag) or (GalNAcα-Ser/Thr)), prostate specific membrane antigen (PSMA), receptor tyrosine kinase ROS-like orphan receptor 1 (ROR1), Fms-like tyrosine kinase 3 (FLT3), tumor-associated glycoprotein 72 (TAG72), CD38, CD44v6, carcinoembryonic antigen (CEA), epithelial cell adhesion molecule (EPCAM), B7H3 (CD276), KIT (CD117), interleukin 13 receptor subunit alpha-2 (IL-13Rα2 or CD213A2), mesothelin, interleukin 11 receptor alpha (IL-11Ra), prostate stem cell antigen (PSCA), testisin serine 21 (testisin or PRSS21), vascular endothelial growth factor receptor 2 (VEGFR2), Lewis (Y) antigen, CD24, platelet-derived growth factor receptor beta (PDGFR-β), stage-specific embryonic antigen-4 (SSEA-4), CD20, folate receptor alpha, receptor tyrosine protein Kinase ERBB2 (Her2/neu), cell surface associated mucin 1 (MUC1), epidermal growth factor receptor (EGFR), neural cell adhesion molecule (NCAM), prostase, prostatic acid phosphatase (PAP), mutated elongation factor 2 (ELF2M), ephrin B2, fibroblast activation protein alpha (FAP), insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX), proteasome (Prosome, Macropain) subunit, beta type, 9 (LMP2), glycoprotein 100 (gp100), oncogene fusion protein composed of breakpoint cluster region (BCR) and Alelson murine leukemia virus oncogene homolog 1 (AB1) (bcr-abl), tyrosinase, ephrin type A receptor 2 (EphA2), fucosyl GM1 , sialyl Lewis adhesion molecule (sLe), ganglioside GM3 (aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer), transglutaminase 5 (TGS5), high molecular weight melanoma associated antigen (HMWMAA), o-acetyl GD2 ganglioside (OAcGD2), folate receptor β, tumor endothelial marker 1 (TEM1/CD248), tumor endothelial marker 7 related (TEM7R), Claudin 6 (CLDN6), thyroid stimulating hormone receptor (TSHR), G protein-coupled receptor class 5 group C, member D (GPRC5D), X chromosome open reading frame 61 (CXORF61), CD97, CD179a, anaplastic lymphoma kinase (ALK), polysialic acid, placenta-specific 1 (PLAC1), globoH hexose moiety of glycoceramide (GloboH), breast differentiation antigen (NY-BR-1), uroplakin 2 (UPK2), hepatitis A virus cell receptor 1 (HAVCR1), adrenergic receptor β3 (ADRB3), pannexin 3 (PANX3), G protein-coupled receptor 20 (GPR20), lymphocyte antigen 6 complex, locus K9 (LY6K), olfactory receptor 51E2 (OR51E2), TCRγ alternate reading frame protein (TARP), Wilms tumor protein (WT1), cancer/testis antigen 1 (NY-ESO-1), cancer/testis antigen 2 (LAGE-1A), melanoma-associated antigen 1 (MAGE-A1), ETS translocation variant gene 6, located on chromosome 12p (ETV6-AML), sperm protein 17 (SPA17), X antigen family, member 1A (XA GE1), angiopoietin binding cell surface receptor 2 (Tie2), melanoma cancer testis antigen-1 (MAD-CT-1), melanoma cancer testis antigen-2 (MAD-CT-2), FOS-related antigen 1, tumor protein p53 (p53), p53 mutant, prostein, survivin, telomerase, prostate cancer tumor antigen-1 (PCTA-1 or galectin 8), melanoma antigen recognized by T cells 1 (MelanA or MART1), rat sarcoma (Ras) mutant, human telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, melanoma inhibitor of apoptosis (ML-IAP), ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene), N-acetylglucosaminyltransferase V (NA17), paired box protein Pax-3 (PAX3), androgen receptor, cyclin B1, V-myc avian Myelocytic neoplastic disease viral oncogene neuroblastoma-derived homolog (MYCN), Ras homolog family member C (RhoC), tyrosinase-related protein 2 (TRP-2), cytochrome P450 1B1 (CYP1B1), CCCTC binding factor (zinc finger protein)-like (brother of BORIS or regulator of imprinting sites), squamous cell carcinoma antigen recognized by T cells 3 (SART3), paired box protein Pax-5 (PAX5), proacrosin binding protein sp32 (OY-TES1), lymphocyte-specific protein tyrosine kinase (LCK), A kinase anchoring protein 4 (AKAP-4), synovial sarcoma, X breakpoint 2 (SSX2), receptor for advanced glycation end products (RAGE-1), renal uibiguitous 1 (RU1), renal uibiguitous 2 (RU2), legumain, human papilloma HPV E6 (HPV E6), human papillomavirus E7 (HPV E7), intestinal carboxylesterase, mutant heat shock protein 70-2 (mut hosp 70-2), CD79a, CD79b, CD72, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), Fc fragment of IgA receptor (FCAR or CD89), leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2), CD300 molecule-like family member f (CD300LF), C-type lectin domain family 12 member A (CLEC12A), bone marrow stromal cell antigen 2 (BST2), EGF-like module-containing mucin-like hormone receptor-like 2 (EMR2), lymphocyte antigen 75 (LY75), phosphatidylinositol proteoglycan-3 (GPC3), Fc receptor-like 5 (FCRL5), and immunoglobulin lambda-like polypeptide 1 (IGLL1) One or more.
  11. 权利要求9的用途,其中所述一种或多种肿瘤相关抗原选自TSHR、CD171、CS-1、CLL-1、GD3、Tn Ag、FLT3、CD38、CD44v6、B7H3、KIT、IL-13Rα2、IL-11Ra、PSCA、PRSS21、VEGFR2、LewisY、CD24、PDGFR-β,SSEA-4、MUC1、EGFR、NCAM、CAIX、LMP2、EphA2、岩藻糖基GM1,SLE、GM3、TGS5、HMWMAA、邻乙酰基GD2、叶酸受体β、TEM1/CD248、TEM7R、CLDN6、GPRC5D、CXORF61、CD97、CD179a、ALK、聚唾液酸,PLAC1、GloboH、NY-BR-1、UPK2、HAVCR1、ADRB3、PANX3、GPR20、LY6K、OR51E2、TARP、WT1、ETV6-AML、精子蛋白17,XAGE1、Tie 2、MAD-CT-1、MAD-CT-2、Fos相关抗原1、p53突变体、hTERT、肉瘤易位断点、ML-IAP,ERG(TMPRSS2ETS融合基因)、NA17,PAX3,雄激素受体,细胞周期蛋白B1,MYCN、RhoC、CYP1B1、BORIS、SART3、PAX5、OY-TES1、LCK、AKAP-4、SSX2、CD79a、CD79b、CD72、LAIR1、FCAR、LILRA2、CD300LF、CLEC12A、BST2、EMR2、LY75、GPC3、FCRL5、和IGLL1中的一种或多种。The use of claim 9, wherein the one or more tumor-associated antigens are selected from TSHR, CD171, CS-1, CLL-1, GD3, Tn Ag, FLT3, CD38, CD44v6, B7H3, KIT, IL-13Rα2, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-β, SSEA-4, MUC1, EGFR, NCAM, CAIX, LMP2, EphA2, fucosyl GM1, SLE, GM3, TGS5, HMWMAA, o-acetyl GD2, folate receptor β, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, A One or more of DRB3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53 mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2ETS fusion gene), NA17, PAX3, androgen receptor, cyclin B1, MYCN, RhoC, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
  12. 权利要求9的用途,其中所述一种或多种肿瘤相关抗原选自CD19、IL-13Rα2、EphA2和 EGFRvIII中的一种或多种。The use of claim 9, wherein the one or more tumor-associated antigens are selected from one or more of CD19, IL-13Rα2, EphA2 and EGFRvIII.
  13. 间充质干细胞用于增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的用途,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原。Use of mesenchymal stem cells for enhancing the in vivo proliferation capacity of immune effector cells after administration and/or increasing the in vivo persistence of single immune effector cells after administration, wherein the mesenchymal stem cells do not express or substantially do not express antigens targeted by the immune effector cells on their surface.
  14. 权利要求13的用途,其中免疫效应细胞是经修饰的。The use of claim 13, wherein the immune effector cells are modified.
  15. 权利要求13的用途,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The use of claim 13, wherein the immune effector cells are T cells, optionally CAR-T cells.
  16. 间充质干细胞用于调节免疫效应细胞在施用后的体内分化和/或促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的用途,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原。Use of mesenchymal stem cells for regulating the in vivo differentiation of immune effector cells after administration and/or promoting the in vivo differentiation of immune effector cells into memory cell subtypes after administration, wherein the surface of the mesenchymal stem cells does not express or substantially does not express the antigens targeted by the immune effector cells.
  17. 权利要求16的用途,其中免疫效应细胞是经修饰的。The use of claim 16, wherein the immune effector cells are modified.
  18. 权利要求16的用途,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The use of claim 16, wherein the immune effector cells are T cells, optionally CAR-T cells.
  19. 间充质干细胞在制备抗肿瘤药物中的用途,其中所述药物还含有免疫效应细胞,并且所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原。Use of mesenchymal stem cells in the preparation of anti-tumor drugs, wherein the drugs also contain immune effector cells, and the surface of the mesenchymal stem cells does not express or substantially does not express antigens targeted by the immune effector cells.
  20. 权利要求19的用途,其中免疫效应细胞是经修饰的。The use of claim 19, wherein the immune effector cells are modified.
  21. 权利要求19的用途,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The use of claim 19, wherein the immune effector cells are T cells, optionally CAR-T cells.
  22. 间充质干细胞的用途,用于制备以下一种或多种:Use of mesenchymal stem cells for preparing one or more of the following:
    (1)增强免疫效应细胞体内增殖能力和/或单个免疫效应细胞体内持久性的试剂;(1) Agents that enhance the in vivo proliferation capacity of immune effector cells and/or the in vivo persistence of single immune effector cells;
    (2)使得免疫效应细胞保持低分化状态的免疫效应细胞分化抑制剂;(2) immune effector cell differentiation inhibitors that keep immune effector cells in a low-differentiation state;
    (3)使得记忆性免疫效应细胞亚型形成增加的试剂;(3) Agents that increase the formation of memory immune effector cell subtypes;
    (4)细胞(优选免疫效应细胞)疗法的疗效增强剂;(4) An agent for enhancing the efficacy of cell (preferably immune effector cell) therapy;
    其中所述间充质干细胞表面不表达或实质上不表达被所述细胞所针对的抗原。The mesenchymal stem cells do not express or substantially do not express the antigen targeted by the cells on their surface.
  23. 权利要求22的用途,其中免疫效应细胞是经修饰的。The use of claim 22, wherein the immune effector cells are modified.
  24. 权利要求22的用途,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The use of claim 22, wherein the immune effector cells are T cells, optionally CAR-T cells.
  25. 间充质干细胞与免疫效应细胞联合用于制备治疗癌症的药物的用途,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原。The invention relates to the use of mesenchymal stem cells in combination with immune effector cells for preparing a drug for treating cancer, wherein the surface of the mesenchymal stem cells does not express or substantially does not express the antigen targeted by the immune effector cells.
  26. 权利要求25的用途,其中免疫效应细胞是经修饰的。The use of claim 25, wherein the immune effector cells are modified.
  27. 权利要求25的用途,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The use of claim 25, wherein the immune effector cells are T cells, optionally CAR-T cells.
  28. 药物组合物,其包含间充质干细胞和免疫效应细胞,另外还任选地包含适当的可药用载体,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原,所述药物用于治疗肿瘤或自身免疫病。A pharmaceutical composition comprising mesenchymal stem cells and immune effector cells, and optionally further comprising a suitable pharmaceutically acceptable carrier, wherein the surface of the mesenchymal stem cells does not express or substantially does not express antigens targeted by the immune effector cells, and the drug is used to treat tumors or autoimmune diseases.
  29. 权利要求28的药物组合物,其中免疫效应细胞是经修饰的。The pharmaceutical composition of claim 28, wherein the immune effector cells are modified.
  30. 权利要求28的药物组合物,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The pharmaceutical composition of claim 28, wherein the immune effector cells are T cells, optionally CAR-T cells.
  31. 治疗肿瘤的方法,所述方法包括向患有所述肿瘤的受试者施用免疫效应细胞,且所述细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内,其中其中所述免疫效应细胞所针对的所述肿瘤的肿瘤相关抗原,并且所述间充质干细胞表面不表达或实质上不表达被所述免疫效应细胞所针对的抗原。A method for treating a tumor, the method comprising administering immune effector cells to a subject having the tumor, wherein the cells have been co-cultured with mesenchymal stem cells in vitro/ex vivo, and/or co-administered with mesenchymal stem cells in vivo, wherein the immune effector cells are directed against a tumor-associated antigen of the tumor, and the mesenchymal stem cells do not express or substantially do not express the antigen directed against by the immune effector cells on their surface.
  32. 权利要求31的方法,其中免疫效应细胞是经修饰的。The method of claim 31, wherein the immune effector cells are modified.
  33. 权利要求31的方法,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The method of claim 31, wherein the immune effector cell is a T cell, optionally a CAR-T cell.
  34. 权利要求31的方法,其中所述肿瘤相关抗原选自CD19、CD123、CD22、CD30、CD171、CS-1(也被称为CD2子集1、CRACC、SLAMF7、CD319、和19A24)、C型凝集素样分子-1(CLL-1或CLECL1)、CD33、表皮生长因子受体变体III(EGFRvIII)、神经节苷脂G2(GD2)、神经节苷脂GD3(aNeu5Ac(2-8)aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、TNF受体家族成员B细胞成熟(BCMA)、Tn抗原((Tn Ag)或(GalNAcα-Ser/Thr))、前列腺特异性膜抗原(PSMA)、受体酪氨酸激酶样孤儿受体1(ROR1)、Fms样酪氨酸激酶3(FLT3)、肿瘤相关的糖蛋白72(TAG72)、CD38、CD44v6、癌胚抗原(CEA)、上皮细胞粘附分子(EPCAM)、B7H3(CD276)、KIT(CD117)、白介素13受体亚基α-2(IL-13Rα2或CD213A2)、间皮素、白介素11受体α(IL-11Ra)、前列腺干细胞抗原(PSCA)、蛋白酶丝氨酸21(Testisin或PRSS21)、血管内皮生长因子受体2(VEGFR2)、路易斯(Y)抗原、CD24、血小板衍生生长因子受体β(PDGFR-β)、阶段特异性胚胎抗原-4(SSEA-4)、CD20、叶酸受体α、受体酪氨酸蛋白激酶ERBB2(Her2/neu)、细胞表面相关的粘蛋白1(MUC1)、表皮生长因子受体(EGFR)、神经细胞粘附分子(NCAM)、Prostase、前列腺酸性磷酸酶(PAP)、突变的延伸因子2(ELF2M)、肝配蛋白B2、成纤维细胞活化蛋白α(FAP)、胰岛素样生长因子1受体(IGF-I受体)、碳酸酐酶IX(CAIX)、蛋白酶体(Prosome、Macropain)亚基、β型、9(LMP2)、糖蛋白100(gp100)、由断点簇区(BCR)和Alelson鼠白血病病毒癌基因同源物1(AB1)组成的癌基因融合蛋白(bcr-abl)、酪氨酸酶、肝配蛋白A型受体2(EphA2)、岩藻糖基GM1、唾液酸基路易斯粘附分子(sLe)、神经节苷脂GM3(aNeu5Ac(2-3)bDGalp(1-4)bDGlcp(1-1)Cer)、转谷氨酰胺酶5(TGS5)、高分子量黑素瘤相关抗原(HMWMAA)、邻乙酰基GD2神经节苷脂(OAcGD2)、叶酸受体β、肿瘤血管内皮标记1(TEM1/CD248)、肿瘤血管内皮标记7相关的(TEM7R)、Claudin 6(CLDN6)、促甲状腺激素受体(TSHR)、G蛋白偶联受体C类5组、成员D(GPRC5D)、X染色体开放阅读框61(CXORF61)、CD97、CD179a、间变性淋巴瘤激酶(ALK)、聚唾液酸、胎盘特异性1(PLAC1)、globoH glycoceramide的己糖部分(GloboH)、乳腺分化抗原(NY-BR-1)、uroplakin 2(UPK2)、甲型肝炎病毒细胞受体1(HAVCR1)、肾上腺素受体β3(ADRB3)、pannexin 3(PANX3)、G蛋白偶联受体20(GPR20)、淋巴细胞抗原6复合物、基因座K9(LY6K)、嗅觉受体51E2(OR51E2)、TCRγ交替阅读框蛋白(TARP)、肾母细胞瘤蛋白(WT1)、癌/睾丸抗原1(NY-ESO-1)、癌症/睾丸抗原2(LAGE-1A)、黑素瘤相关抗原1(MAGE-A1)、ETS易位变异基因6,位于染色体12p(ETV6-AML)、精子蛋白17(SPA17)、X抗原家族,成员1A(XAGE1)、血管生成素结合细胞表面受体2(Tie2)、黑素瘤癌睾丸抗原-1(MAD-CT-1)、黑素瘤癌睾丸抗原-2(MAD-CT-2)、FOS相关抗原1、肿瘤蛋白质p53(p53)、p53突变体、prostein、存活蛋白、端粒酶、前列腺癌肿瘤抗原-1(PCTA-1或半乳凝素8)、由T细胞识别的黑素瘤抗原1(MelanA或MART1)、大鼠肉瘤(Ras)突变体、人端粒酶逆转录酶(hTERT)、肉瘤易位断点、细胞凋亡的黑素瘤抑制剂(ML-IAP)、ERG(跨膜蛋白酶,丝氨酸2(TMPRSS2)ETS融合基因)、N-乙酰葡糖胺基转移酶V(NA17)、配对盒蛋白Pax-3(PAX3)、雄激素受体、细胞周期蛋白B1、V-myc鸟髓细胞瘤病病毒癌基因神经母细胞瘤衍生的同源物(MYCN)、Ras同源物家 族成员C(RhoC)、酪氨酸酶相关蛋白2(TRP-2)、细胞色素P450 1B1(CYP1B1)、CCCTC结合因子(锌指蛋白)样(BORIS或印记位点的调节物的兄弟)、由T细胞识别的鳞状细胞癌抗原3(SART3)、配对盒蛋白Pax-5(PAX5)、proacrosin结合蛋白sp32(OY-TES1)、淋巴细胞特异性蛋白酪氨酸激酶(LCK)、A激酶锚定蛋白4(AKAP-4)、滑膜肉瘤,X断点2(SSX2)、高级糖化终产物受体(RAGE-1)、肾uibiguitous 1(RU1)、肾uibiguitous 2(RU2)、legumain、人类乳头瘤病毒E6(HPV E6)、人类乳头瘤病毒E7(HPV E7)、肠羧基酯酶、突变的热休克蛋白70-2(mut hosp 70-2)、CD79a、CD79b、CD72、白细胞相关免疫球蛋白样受体1(LAIR1)、IgA受体的Fc片段(FCAR或CD89)、白细胞免疫球蛋白样受体亚家族A成员2(LILRA2)、CD300分子样家族成员f(CD300LF)、C型凝集素结构域家族12成员A(CLEC12A)、骨髓基质细胞抗原2(BST2)、含有EGF样模块粘蛋白样激素受体样2(EMR2)、淋巴细胞抗原75(LY75)、磷脂酰肌醇蛋白聚糖-3(GPC3)、Fc受体样5(FCRL5)、与免疫球蛋白λ样多肽1(IGLL1)中的一种或多种。The method of claim 31, wherein the tumor-associated antigen is selected from CD19, CD123, CD22, CD30, CD171, CS-1 (also known as CD2 subset 1, CRACC, SLAMF7, CD319, and 19A24), C-type lectin-like molecule-1 (CLL-1 or CLECL1), CD33, epidermal growth factor receptor variant III (EGFRvIII), ganglioside G2 (GD2), ganglioside GD3 (aNeu5Ac (2-8) aNeu5Ac (2-3) bDGalp (1-4) bDGlcp (1-1) Cer), TNF receptor family member B cells Maturation (BCMA), Tn antigen ((Tn Ag) or (GalNAcα-Ser/Thr)), prostate-specific membrane antigen (PSMA), receptor tyrosine kinase-like orphan receptor 1 (ROR1), Fms-like tyrosine kinase 3 (FLT3), tumor-associated glycoprotein 72 (TAG72), CD38, CD44v6, carcinoembryonic antigen (CEA), epithelial cell adhesion molecule (EPCAM), B7H3 (CD276), KIT (CD117), interleukin 13 receptor subunit α-2 (IL-13Rα2 or CD213A2), mesothelin, interleukin 11 receptor α (IL-11Ra), prostate stem cells Antigen (PSCA), testisin or PRSS21, vascular endothelial growth factor receptor 2 (VEGFR2), Lewis (Y) antigen, CD24, platelet-derived growth factor receptor beta (PDGFR-β), stage-specific embryonic antigen-4 (SSEA-4), CD20, folate receptor alpha, receptor tyrosine protein kinase ERBB2 (Her2/neu), cell surface-associated mucin 1 (MUC1), epidermal growth factor receptor (EGFR), neural cell adhesion molecule (NCAM), prostase, prostatic acid phosphatase (PAP), mutated elongation factor 2 (ELF2 M), ephrin B2, fibroblast activation protein alpha (FAP), insulin-like growth factor 1 receptor (IGF-I receptor), carbonic anhydrase IX (CAIX), proteasome (Prosome, Macropain) subunit, beta type, 9 (LMP2), glycoprotein 100 (gp100), oncogene fusion protein composed of breakpoint cluster region (BCR) and Alelson murine leukemia virus oncogene homolog 1 (AB1) (bcr-abl), tyrosinase, ephrin type A receptor 2 (EphA2), fucosyl GM1, sialyl Lewis adhesion molecule (sLe), ganglioside GM3 (aNeu5Ac(2 -3)bDGalp(1-4)bDGlcp(1-1)Cer), transglutaminase 5 (TGS5), high molecular weight melanoma associated antigen (HMWMAA), o-acetyl GD2 ganglioside (OAcGD2), folate receptor β, tumor endothelial marker 1 (TEM1/CD248), tumor endothelial marker 7 related (TEM7R), Claudin 6 (CLDN6), thyroid stimulating hormone receptor (TSHR), G protein coupled receptor class C 5 group, member D (GPRC5D), X chromosome open reading frame 61 (CXORF61), CD97, CD179a, anaplastic lymphoma kinase ( ALK), polysialic acid, placenta-specific 1 (PLAC1), hexose moiety of globoH glycoceramide (GloboH), breast differentiation antigen (NY-BR-1), uroplakin 2 (UPK2), hepatitis A virus cell receptor 1 (HAVCR1), adrenergic receptor β3 (ADRB3), pannexin 3 (PANX3), G protein-coupled receptor 20 (GPR20), lymphocyte antigen 6 complex, locus K9 (LY6K), olfactory receptor 51E2 (OR51E2), TCRγ alternate reading frame protein (TARP), nephroblastoma protein (WT1 ), cancer/testis antigen 1 (NY-ESO-1), cancer/testis antigen 2 (LAGE-1A), melanoma-associated antigen 1 (MAGE-A1), ETS translocation variant gene 6, located on chromosome 12p (ETV6-AML), sperm protein 17 (SPA17), X antigen family, member 1A (XAGE1), angiopoietin binding cell surface receptor 2 (Tie2), melanoma cancer testis antigen-1 (MAD-CT-1), melanoma cancer testis antigen-2 (MAD-CT-2), FOS-associated antigen 1, tumor protein p53 (p53), p53 mutant, prostein, survivin, telomerase, prostate adenocarcinoma tumor antigen-1 (PCTA-1 or galectin 8), melanoma antigen recognized by T cells 1 (MelanA or MART1), rat sarcoma (Ras) mutant, human telomerase reverse transcriptase (hTERT), sarcoma translocation breakpoints, melanoma inhibitor of apoptosis (ML-IAP), ERG (transmembrane protease, serine 2 (TMPRSS2) ETS fusion gene), N-acetylglucosaminyltransferase V (NA17), paired box protein Pax-3 (PAX3), androgen receptor, cyclin B1, V-myc avian myelocytic virus oncogene neuroblastoma-derived homolog (MYCN), Ras homolog family family member C (RhoC), tyrosinase-related protein 2 (TRP-2), cytochrome P450 1B1 (CYP1B1), CCCTC binding factor (zinc finger protein)-like (brother of regulator of BORIS or imprinting sites), squamous cell carcinoma antigen recognized by T cells 3 (SART3), paired box protein Pax-5 (PAX5), proacrosin binding protein sp32 (OY-TES1), lymphocyte-specific protein tyrosine kinase (LCK), A kinase anchoring protein 4 (AKAP-4), synovial sarcoma, X breakpoint 2 (SSX2), receptor for advanced glycation end products (RAGE-1), renal uibiguitous 1 (RU1), renal uibiguitous 2 (RU2), legumain, human papillomavirus E6 (HPV E6), human papilloma HPV E7 (HPV E7), intestinal carboxylesterase, mutated heat shock protein 70-2 (mut hosp 70-2), CD79a, CD79b, CD72, leukocyte-associated immunoglobulin-like receptor 1 (LAIR1), Fc fragment of IgA receptor (FCAR or CD89), leukocyte immunoglobulin-like receptor subfamily A member 2 (LILRA2), CD300 molecule-like family member f (CD300LF), C-type lectin domain family 12 member A (CLEC12A), bone marrow stromal cell antigen 2 (BST2), EGF-like module-containing mucin-like hormone receptor-like 2 (EMR2), lymphocyte antigen 75 (LY75), phosphatidylinositol proteoglycan-3 (GPC3), Fc receptor-like 5 (FCRL5), and immunoglobulin lambda-like polypeptide 1 (IGLL1) One or more.
  35. 权利要求31的方法,其中所述肿瘤相关抗原选自TSHR、CD171、CS-1、CLL-1、GD3、Tn Ag、FLT3、CD38、CD44v6、B7H3、KIT、IL-13Rα2、IL-11Ra、PSCA、PRSS21、VEGFR2、LewisY、CD24、PDGFR-β,SSEA-4、MUC1、EGFR、NCAM、CAIX、LMP2、EphA2、岩藻糖基GM1,SLE、GM3、TGS5、HMWMAA、邻乙酰基GD2、叶酸受体β、TEM1/CD248、TEM7R、CLDN6、GPRC5D、CXORF61、CD97、CD179a、ALK、聚唾液酸,PLAC1、GloboH、NY-BR-1、UPK2、HAVCR1、ADRB3、PANX3、GPR20、LY6K、OR51E2、TARP、WT1、ETV6-AML、精子蛋白17,XAGE1、Tie 2、MAD-CT-1、MAD-CT-2、Fos相关抗原1、p53突变体、hTERT、肉瘤易位断点、ML-IAP,ERG(TMPRSS2ETS融合基因)、NA17,PAX3,雄激素受体,细胞周期蛋白B1,MYCN、RhoC、CYP1B1、BORIS、SART3、PAX5、OY-TES1、LCK、AKAP-4、SSX2、CD79a、CD79b、CD72、LAIR1、FCAR、LILRA2、CD300LF、CLEC12A、BST2、EMR2、LY75、GPC3、FCRL5、和IGLL1中的一种或多种。The method of claim 31, wherein the tumor-associated antigen is selected from TSHR, CD171, CS-1, CLL-1, GD3, Tn Ag, FLT3, CD38, CD44v6, B7H3, KIT, IL-13Rα2, IL-11Ra, PSCA, PRSS21, VEGFR2, LewisY, CD24, PDGFR-β, SSEA-4, MUC1, EGFR, NCAM, CAIX, LMP2, EphA2, fucosyl GM1, SLE, GM3, TGS5, HMWMAA, o-acetyl GD2, folate receptor β, TEM1/CD248, TEM7R, CLDN6, GPRC5D, CXORF61, CD97, CD179a, ALK, polysialic acid, PLAC1, GloboH, NY-BR-1, UPK2, HAVCR1, ADR One or more of B3, PANX3, GPR20, LY6K, OR51E2, TARP, WT1, ETV6-AML, sperm protein 17, XAGE1, Tie 2, MAD-CT-1, MAD-CT-2, Fos-related antigen 1, p53 mutant, hTERT, sarcoma translocation breakpoints, ML-IAP, ERG (TMPRSS2ETS fusion gene), NA17, PAX3, androgen receptor, cyclin B1, MYCN, RhoC, CYP1B1, BORIS, SART3, PAX5, OY-TES1, LCK, AKAP-4, SSX2, CD79a, CD79b, CD72, LAIR1, FCAR, LILRA2, CD300LF, CLEC12A, BST2, EMR2, LY75, GPC3, FCRL5, and IGLL1.
  36. 权利要求31的方法,其中所述肿瘤相关抗原选自CD19、IL-13Rα2、EphA2和EGFRvIII中的一种或多种。The method of claim 31, wherein the tumor-associated antigen is selected from one or more of CD19, IL-13Rα2, EphA2 and EGFRvIII.
  37. 增强免疫效应细胞在施用后的体内增殖能力和/或增加单个免疫效应细胞在施用后的体内持久性的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫疗法所针对的抗原,任选地所述方法是体内/体外/离体的。A method for enhancing the in vivo proliferation capacity of immune effector cells after administration and/or increasing the in vivo persistence of a single immune effector cell after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering the immune effector cells with mesenchymal stem cells into the body, wherein the mesenchymal stem cells do not express or substantially do not express the antigen targeted by the immunotherapy on their surface, and optionally the method is in vivo/in vitro/ex vivo.
  38. 权利要求37的方法,其中免疫效应细胞是经修饰的。The method of claim 37, wherein the immune effector cells are modified.
  39. 权利要求37的方法,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The method of claim 37, wherein the immune effector cell is a T cell, optionally a CAR-T cell.
  40. 调节免疫效应细胞在施用后的体内分化和/或促进免疫效应细胞在施用后的体内向记忆性细胞亚型分化的方法,所述方法包括将所述免疫效应细胞已经在体外/离体与间充质干细胞共培养,和/或与间充质干细胞共同施用至体内,其中所述间充质干细胞表面不表达或实质上不表达被所述免疫疗法所针对的抗原,任选地所述方法是体内/体外/离体的。A method for regulating the in vivo differentiation of immune effector cells after administration and/or promoting the differentiation of immune effector cells into memory cell subtypes in vivo after administration, the method comprising co-culturing the immune effector cells with mesenchymal stem cells in vitro/ex vivo, and/or co-administering the immune effector cells with mesenchymal stem cells into the body, wherein the surface of the mesenchymal stem cells does not express or substantially does not express the antigen targeted by the immunotherapy, and optionally the method is in vivo/in vitro/ex vivo.
  41. 权利要求40的方法,其中免疫效应细胞是经修饰的。The method of claim 40, wherein the immune effector cells are modified.
  42. 权利要求40的方法,其中免疫效应细胞是T细胞,任选地CAR-T细胞。The method of claim 40, wherein the immune effector cell is a T cell, optionally a CAR-T cell.
  43. 前述权利要求任一项的用途或方法或药物组合物,其中间充质干细胞是胎盘来源的。The use or method or pharmaceutical composition of any preceding claim, wherein the mesenchymal stem cells are of placental origin.
  44. 前述权利要求任一项的用途或方法或药物组合物,其中间充质干细胞表达CD105和/或不表达CD45。The use or method or pharmaceutical composition of any preceding claim, wherein the mesenchymal stem cells express CD105 and/or do not express CD45.
  45. 前述权利要求任一项的用途或方法或药物组合物,其中将所述间充质干细胞与细胞疗法中所用的治疗性细胞在体外共培养。The use or method or pharmaceutical composition of any preceding claim, wherein the mesenchymal stem cells are co-cultured in vitro with therapeutic cells used in cell therapy.
  46. 前述权利要求任一项的用途或方法或药物组合物,其中将所述间充质干细胞与细胞疗法中所用的治疗性细胞共同施用于受试者体内,其中间充质干细胞先于治疗性细胞施用,或与其同时施用,或在施用治疗性细胞之后施用。The use or method or pharmaceutical composition of any of the preceding claims, wherein the mesenchymal stem cells are co-administered into a subject with therapeutic cells used in cell therapy, wherein the mesenchymal stem cells are administered before, simultaneously with, or after the therapeutic cells.
  47. 前述权利要求任一项的用途或方法或药物组合物,其中所述间充质干细胞与细胞疗法中所用的治疗性细胞的数量比例为10:1,或5:1,或4:1,或2:1,或1:1、或1:2,或1:4,或1:5,或1:10,或1:20。The use or method or pharmaceutical composition of any preceding claim, wherein the ratio of the number of mesenchymal stem cells to the therapeutic cells used in cell therapy is 10:1, or 5:1, or 4:1, or 2:1, or 1:1, or 1:2, or 1:4, or 1:5, or 1:10, or 1:20.
PCT/CN2022/128989 2022-11-01 2022-11-01 Use of mesenchymal stem cell WO2024092508A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/128989 WO2024092508A1 (en) 2022-11-01 2022-11-01 Use of mesenchymal stem cell

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2022/128989 WO2024092508A1 (en) 2022-11-01 2022-11-01 Use of mesenchymal stem cell

Publications (1)

Publication Number Publication Date
WO2024092508A1 true WO2024092508A1 (en) 2024-05-10

Family

ID=90929331

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/128989 WO2024092508A1 (en) 2022-11-01 2022-11-01 Use of mesenchymal stem cell

Country Status (1)

Country Link
WO (1) WO2024092508A1 (en)

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107184603A (en) * 2017-06-01 2017-09-22 刘未斌 A kind of drug regimen for treating tumour
CN110331134A (en) * 2019-07-19 2019-10-15 上海交通大学医学院附属瑞金医院 A kind of universal cell therapy product and its preparation method and application for expressing antigen recognition region

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107184603A (en) * 2017-06-01 2017-09-22 刘未斌 A kind of drug regimen for treating tumour
CN110331134A (en) * 2019-07-19 2019-10-15 上海交通大学医学院附属瑞金医院 A kind of universal cell therapy product and its preparation method and application for expressing antigen recognition region

Non-Patent Citations (4)

* Cited by examiner, † Cited by third party
Title
LER YIE CHAN: "CAR-T Cells/-NK Cells in Cancer Immunotherapy and the Potential of MSC to Enhance Its Efficacy: A Review", BIOMEDICINES, MDPI, BASEL, vol. 10, no. 4, 1 April 2022 (2022-04-01), Basel , pages 804, XP093168245, ISSN: 2227-9059, DOI: 10.3390/biomedicines10040804 *
MCKENNA MARY K., ENGLISCH ALEXANDER, BRENNER BENJAMIN, SMITH TYLER, HOYOS VALENTINA, SUZUKI MASATAKA, BRENNER MALCOLM K.: "Mesenchymal stromal cell delivery of oncolytic immunotherapy improves CAR-T cell antitumor activity", MOLECULAR THERAPY, ELSEVIER INC., US, vol. 29, no. 5, 1 May 2021 (2021-05-01), US , pages 1808 - 1820, XP093136509, ISSN: 1525-0016, DOI: 10.1016/j.ymthe.2021.02.004 *
SHUIZHONG CEN: "Autophagy enhances mesenchymal stem cell-mediated CD4+ T cell migration and differentiation through CXCL8 and TGF-β1", STEM CELL RESEARCH & THERAPY, BIOMED CENTRAL LTD, LONDON, UK, vol. 10, no. 1, 1 December 2019 (2019-12-01), London, UK , pages 265, XP093168246, ISSN: 1757-6512, DOI: 10.1186/s13287-019-1380-0 *
WEI JIANG: "Immune modulation by mesenchymal stem cells", CELL PROLIFERATION, OXFORD., GB, vol. 53, no. 1, 1 January 2020 (2020-01-01), GB , pages e12712, XP093168247, ISSN: 0960-7722, DOI: 10.1111/cpr.12712 *

Similar Documents

Publication Publication Date Title
US20200407458A1 (en) Anti cd30 chimeric antigen receptor and its use
US11564945B2 (en) Chimeric antigen receptor and use thereof
JP2023038386A (en) Methods and compositions for cellular immunotherapy
WO2019052562A1 (en) Fusion protein of il-4r and use thereof
Bose et al. Tumor-derived vascular pericytes anergize Th cells
US20210213061A1 (en) Genetically engineered cell and application thereof
JP7450892B2 (en) Artificial HLA-positive feeder cell line for NK cells and its use
WO2021136263A1 (en) Modified immune effector cell and preparation method therefor
WO2019024933A1 (en) Gpc3-targeted car nk cell
US11401317B2 (en) Human-CD123-targeting chimeric receptor ligand and application thereof
WO2018056736A1 (en) Novel natural killer cell line and use thereof
WO2020057641A1 (en) Chemokine expressing cell and use thereof
WO2021057906A1 (en) Immune effector cell expressing il-15
US20230036481A1 (en) A novel cd16+ natural killer cell and a method of culturing cd16+ natural killer cell
WO2019210863A1 (en) Immune effector cell and use thereof
CN113677353A (en) Amplification of modified cells and uses thereof
WO2022033483A1 (en) Multifunctional immune effector cell and use thereof
KR20210061361A (en) Methods for expanding antigen-specific CAR-T cells, compositions and uses related thereto
WO2023235440A2 (en) Compositions and methods comprising chimeric adaptor polypeptides
WO2024092508A1 (en) Use of mesenchymal stem cell
CN115463155B (en) Use of mesenchymal stem cells
WO2022037562A1 (en) Engineered immunoresponsive cells and uses thereof
WO2021244654A1 (en) Activation induced cytokine production in immune cells
WO2020052645A1 (en) Cell containing immunomodulator on surface and use thereof
WO2023284875A1 (en) Chimeric antigen receptor