WO2019114751A1 - Utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs - Google Patents

Utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs Download PDF

Info

Publication number
WO2019114751A1
WO2019114751A1 PCT/CN2018/120593 CN2018120593W WO2019114751A1 WO 2019114751 A1 WO2019114751 A1 WO 2019114751A1 CN 2018120593 W CN2018120593 W CN 2018120593W WO 2019114751 A1 WO2019114751 A1 WO 2019114751A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
tumor
cells
antigen
receptor
Prior art date
Application number
PCT/CN2018/120593
Other languages
English (en)
Chinese (zh)
Inventor
李宗海
周敏
Original Assignee
科济生物医药(上海)有限公司
上海市肿瘤研究所
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 科济生物医药(上海)有限公司, 上海市肿瘤研究所 filed Critical 科济生物医药(上海)有限公司
Publication of WO2019114751A1 publication Critical patent/WO2019114751A1/fr

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464474Proteoglycans, e.g. glypican, brevican or CSPG4
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4611T-cells, e.g. tumor infiltrating lymphocytes [TIL], lymphokine-activated killer cells [LAK] or regulatory T cells [Treg]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/463Cellular immunotherapy characterised by recombinant expression
    • A61K39/4631Chimeric Antigen Receptors [CAR]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/464Cellular immunotherapy characterised by the antigen targeted or presented
    • A61K39/4643Vertebrate antigens
    • A61K39/4644Cancer antigens
    • A61K39/464402Receptors, cell surface antigens or cell surface determinants
    • A61K39/464403Receptors for growth factors
    • A61K39/464404Epidermal growth factor receptors [EGFR]
    • 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
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/31Indexing codes associated with cellular immunotherapy of group A61K39/46 characterized by the route of administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/38Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the dose, timing or administration schedule
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K2239/00Indexing codes associated with cellular immunotherapy of group A61K39/46
    • A61K2239/46Indexing codes associated with cellular immunotherapy of group A61K39/46 characterised by the cancer treated
    • A61K2239/50Colon

Definitions

  • the invention belongs to the field of immunotherapy, and particularly relates to an immune effector cell having a receptor for recognizing a tumor antigen and triggering cell activation, and a local tumor radiation therapy combined for tumor treatment.
  • CAR-T cells chimeric antigen receptor-modified T cells
  • CAR-NK chimeric antigen receptor-modified NK cells
  • TCR-T TCR-modified T cells
  • the treatment regimen used is clearing pretreatment with clearing drugs such as fludarabine, cyclophosphamide, etc., and then administering CAR-T cells to patients.
  • One of the objects of the present invention is to provide a method for treating a tumor which improves the therapeutic effect of genetically engineered immune cells on solid tumors, the method comprising administering to a subject having a tumor a combination of immune effector cells and local tumor radiation, and The individual undergoes lymphocyte pre-clearing, the immune effector cell comprising a tumor antigen that recognizes the tumor and eliciting the immune effector cell to activate the receptor.
  • the lymphocytes are first pre-cleared, and then the immune effector cells and the local radiation therapy of the tumor are administered and the time of administration is in no particular order; the local radiation therapy of the tumor can be administered first and then the immune effector cells are administered; It can also be administered at the same time; it can also be administered to the immune effector cells first and then to the local radiation therapy of the tumor.
  • the individual is treated with immune effector cells, and the subject is then treated with localized radiation therapy.
  • the receptor is selected from the group consisting of a Chimeric Antigen Receptor (CAR), a T cell receptor (TCR), a T cell fusion protein (TFP), and a T cell.
  • CAR Chimeric Antigen Receptor
  • TCR T cell receptor
  • TFP T cell fusion protein
  • TAC T cell antigen coupler
  • the local radiation therapy is to irradiate the tumor with a radiation therapy device.
  • the radiation therapy device irradiates the tumor by generating any of the following rays: X-ray , ⁇ rays, ⁇ rays, ⁇ rays, neutrons.
  • the radiation therapy apparatus generates X-rays that perform at least one radiation on the tumor or multiple doses of radiation.
  • the radiation dose of the radiation treatment is between 1 and 100 Gy, preferably between 2 and 80 Gy, and more preferably not higher than 70 Gy.
  • the energy source of the radiation therapy is located in the body or outside of the individual.
  • the chimeric antigen receptor comprises:
  • the tumor antigen is selected from the group consisting of: thyroid stimulating hormone receptor (TSHR); CD171; CS-1; C-type lectin-like molecule-1; ganglioside GD3; Tn antigen; CD19; CD20; ; CD 30; CD70; CD123; CD138; CD33; CD44; CD44v7/8; CD38; CD44v6; B7H3 (CD276), B7H6; KIT (CD117); interleukin 13 receptor subunit alpha (IL-13R ⁇ ); interleukin 11 ⁇ (IL-11R ⁇ ); prostate stem cell antigen (PSCA); prostate specific membrane antigen (PSMA); carcinoembryonic antigen (CEA); NY-ESO-1; HIV-1 Gag; MART-1; gp100; tyrosine Enzyme; mesothelin; EpCAM; protease serine 21 (PRSS21); vascular endothelial growth factor receptor, vascular endothelial growth factor receptor 2 (VEG
  • the immune effector cells are selected from CAR-T cells which specifically recognize EGFR, EGFRvIII, GPC3, Claudin 18.2.
  • the antibody which specifically recognizes a tumor antigen has the amino acid sequence shown in SEQ ID NO: 2.
  • the chimeric antigen receptor has SEQ ID NO: 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25 or 26 Amino acid sequence.
  • the radiation treatment is performed on the day after the administration of the immune effector cells, or after 1 day, 2 days, or 3 days.
  • the method of the present invention further comprising administering to the individual an immune checkpoint inhibitor; preferably, the immune checkpoint inhibitor is a biological therapeutic agent or a small molecule; more preferably, the The immunological checkpoint inhibitor is selected from the group consisting of a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein, or a combination thereof.
  • the immunological checkpoint targeted by the immunological checkpoint inhibitor is selected from the following immunological checkpoint proteins: CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3 , VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR and B-7 family ligands or combinations thereof; preferably, the immunological checkpoint inhibitor is a PD-1 or PDL-1 inhibitor .
  • the immunological checkpoint inhibitor interacts with a ligand selected from the following immunological checkpoint proteins: CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR and B-7 family ligands or combinations thereof.
  • a ligand selected from the following immunological checkpoint proteins: CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR and B-7 family ligands or combinations thereof.
  • the immune effector cells are selected from CAR-T cells which specifically recognize EGFR, EGFRvIII, GPC3, Claudin 18.2, and the immunological checkpoint inhibitor is a monoclonal antibody that specifically recognizes PD-1 or PD-L1.
  • the immune effector cells and the immunological checkpoint inhibitor are administered simultaneously.
  • the therapeutic effect of the method is predicted by the presence of immune effector cells, or the presence of a genetic marker indicative of T cell inflammation, or a combination thereof, preferably by detecting a change in IFN-[gamma] levels.
  • the tumor includes: breast cancer, blood cancer, colon cancer, rectal cancer, renal cell carcinoma, liver cancer, non-small cell cancer of the lung, small bowel cancer, esophageal cancer, melanoma, bone.
  • Cancer pancreatic cancer, skin cancer, glioma, head and neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, anal cancer, stomach cancer, testicular cancer, uterine cancer, fallopian tube cancer, uterus Endometrial cancer, cervical cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, non-Hodgkin's lymphoma, endocrine system cancer, thyroid cancer, parathyroid carcinoma, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, Child solid tumor, bladder cancer, kidney or ureteral cancer, renal pelvic cancer, central nervous system (CNS) tumor, primary CNS lymphoma, tumor angiogenesis, spinal tumor, brain
  • the immune effector cells include: T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells or bone marrow-derived phagocytic cells or a combination thereof; preferably, said The immune effector cells are selected from autologous T cells, allogeneic T cells or allogeneic NK cells, and more preferably, the T cells are autologous T cells.
  • Another object of the present invention is to provide a use of immunotherapeutic cells in combination with a radiation source for the treatment of cancer in the context of clear pretreatment.
  • Another object of the present invention is to provide a combined treatment system for tumors, characterized in that the combination therapy system is a combined treatment system consisting of an apparatus for administering immune effector cells and local radiation therapy to a tumor-bearing individual. And pre-clearing lymphocytes to said individual, said immune effector cells comprising a receptor that recognizes a tumor antigen of said tumor and initiates activation of said immune effector cells.
  • the radiation source includes an alpha radiation source, a beta radiation source, a gamma radiation source, a neutron source, and the like.
  • the radiation source is a radiation device, and the radiation device is a linear accelerator; preferably, the linear accelerator generates X-rays and electron lines.
  • Figure 1A is a plasmid map of the recombinant vector MSCV-EGFRvIII-mCD28Z;
  • Figure 1B is a test for establishing a mouse serum shower model.
  • Figure 2 shows the anti-tumor therapeutic effect of EGFRvIII-m28Z CAR-T cells combined with local tumor radiation therapy on colon cancer subcutaneous xenografts.
  • Figure 3 shows in vivo tumor suppression assays of EGFRvIII-m28Z CAR-T cells in combination with local tumor radiation therapy.
  • Figure 4 shows the survival assay of EGFRvIII-m28Z CAR-T cells in combination with local tumor radiation therapy for colon cancer in mice.
  • Figure 5 shows a plasma IFN- ⁇ concentration test for EGFRvIII-m28Z CAR-T cells in combination with local tumor radiation therapy for mouse colon cancer.
  • Figure 6 shows the detection of tumor local EGFRvIII-m28Z CAR-T cell expansion in a mouse colon cancer model in combination with local tumor radiation therapy with EGFRvIII-m28Z CAR-T cells.
  • the present invention relates to an immune effector cell having a receptor that recognizes a tumor antigen and triggers cell activation, and a combination of local radiation therapy for treating a tumor, it being understood that the present invention is not limited to the methods and experimental conditions described, because such a method and Conditions can vary.
  • the present disclosure derives, at least in part, from the technological cognition of a combination therapy comprising one or more cycles and/or doses of tumor local radiation therapy and immune effector cell therapy, either continuously, in any order, or substantially simultaneously.
  • the regimen can be more effective in treating cancer in some subjects, and/or can initiate, effect, increase, enhance or prolong the activity and/or number of immune cells, or a medically beneficial response through the tumor.
  • immune effector cell refers to a cell that participates in an immune response, for example, to promote an immune effect.
  • immune effector cells include T cells, for example, ⁇ / ⁇ T cells and ⁇ / ⁇ T cells, B cells, natural killer (NK) cells, natural killer T (NKT) cells, mast cells, and bone marrow-derived phagocytic cells.
  • the T cells comprise autologous T cells, xenografted T cells, allogeneic T cells or allogeneic NK cells.
  • immune effector function or immune effector response refers to an immune effector cell, such as a function or response that enhances or promotes an immune attack by a target cell.
  • an immune effector function or response refers to a property of a T cell or NK cell that promotes killing of a target cell or inhibits growth or proliferation.
  • Local radiation therapy or “local radiation treatment” or “local radiation therapy” has the same meaning in the present invention, and is intended to mean local radiation therapy when a radiation illuminates a certain part of the body, causing a reaction of local cells. These include, for example, fractional radiation therapy, non-fractionated radiation therapy, and super-fractionated radiation therapy.
  • Localized radiation therapy further includes irradiating the tumor with a radiation therapy device that, in a particular embodiment, irradiates the tumor by generating any of the following rays: X-rays, alpha rays, beta rays , gamma rays, neutrons.
  • anti-tumor treatments by ionizing radiation that can be performed according to all available techniques: a non-limiting list of them includes: fractionated radiation therapy, accelerated radiation, intensity modulated radiation therapy, image guided radiation therapy, external beam radiation Treatment, sealed source radiotherapy or brachytherapy, unsealed source radiotherapy, three-dimensional conformal radiotherapy, proton therapy, etc.
  • Localized radiation therapy can also be treated with radiation using an energy source located within the body of the individual.
  • the source of radiation can be external or internal to the subject.
  • the treatment is called external beam radiation therapy (EBRT), also known as external irradiation, often using gamma rays, neutrons, X-rays, etc. Radiation, the biological effect of external irradiation is strong.
  • EBRT external beam radiation therapy
  • BT brachytherapy
  • Its action mainly occurs in the passage of radioactive materials and the tissues and organs at the deposition site, but its effect can be Spread all over the body.
  • the effect of internal irradiation is mainly due to the short range and strong ionization of ⁇ and ⁇ rays.
  • the dose of radiation depends on a number of factors, which are well known in the art. Such factors include the organ to be treated, the healthy organ located in the radiation channel that may be adversely affected inadvertently, the patient's tolerance to radiation therapy, and the area of the body in need of treatment.
  • the dose is typically not higher than 100 Gy, preferably, not higher than 80 Gy, and more preferably not higher than 70 Gy.
  • the dose used can be administered once, or it can be divided into small doses and administered to the patient multiple times. It should be emphasized that the invention is not limited to any particular dosage. The dosage will be determined by the treating physician based on the particular factors in a given situation, including the factors described above.
  • the distance between the external source and the point of entry into the patient can be any distance that achieves an acceptable balance between killing the target cells and minimizing side effects. Typically, the distance between the external source and the point of entry into the patient is between 70 and 100 centimeters.
  • Brachytherapy is usually performed by placing a sealed radioactive source in the patient. Typically, the source is placed approximately 0-5 cm from the tissue to be treated.
  • Known methods include interstitial brachytherapy, intraluminal brachytherapy, and surface interstitial brachytherapy.
  • the source of radiation can be permanently or temporarily implanted. Some of the typical radioactive atoms that have been used in permanent implants include iodine-125 and strontium. Some typical radioactive atoms that have been used in temporary implants include technetium-137 and technetium-192.
  • the radiation dose of brachytherapy may be the same as that used in external beam radiation therapy described above.
  • the characteristics of the radioactive atoms used are also taken into account in determining the brachytherapy dose.
  • Immune effector cell administration is administered before, during, and after radiation therapy, and may also be administered in combination, i.e., before, during, and after, during, and after, or before, during, and after radiation therapy.
  • the immune effector cell treatment is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours prior to radiation treatment.
  • the immune effector cell therapy is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 after radiation therapy administration.
  • the term "radiation therapy device” is a device that emits radiation. According to the degree to which the radiation device may cause harm to human health and the environment, the radiation device is classified into Class I, Class II, and Class III from high to low.
  • the medical ray device and the non-medical ray device are classified according to the use.
  • the ray device includes 1).
  • the device for accelerating the charged particles includes: a cyclotron, an electromagnetic induction accelerator, and various acceleration devices; 2) the device for emitting X-rays includes: an X-ray generating device, an X-ray diffractometer, and an X-ray fluorescence analysis. Instruments, etc.; 3). Devices containing radioactive materials.
  • Common medical ray devices include: medical accelerators, radiation therapy, X-ray electron beam accelerators, heavy ion therapy accelerators, proton therapy devices, accelerators for the preparation of radioactive drugs for positron emission computed tomography (PET), and other medical accelerators.
  • medical accelerators radiation therapy, X-ray electron beam accelerators, heavy ion therapy accelerators, proton therapy devices, accelerators for the preparation of radioactive drugs for positron emission computed tomography (PET), and other medical accelerators.
  • PET positron emission computed tomography
  • X-ray deep therapy machine digital subtraction angiography device
  • medical X-ray CT machine general X-ray machine for radiology diagnosis
  • X-ray imaging device dental X-ray machine
  • mammography machine mammography machine
  • radiation therapy simulation positioning machine others X-ray machines above the exemption level.
  • terapéuticaally effective amount refers to a compound that effectively achieves a particular biological result as described herein.
  • the amount, formulation, substance or composition for example, but not limited to, an amount or dose sufficient to promote a T cell response.
  • therapeutically effective amount refers to a compound that effectively achieves a particular biological result as described herein.
  • the amount, formulation, substance or composition for example, but not limited to, an amount or dose sufficient to promote a T cell response.
  • An effective amount of localized radiation therapy means, but is not limited to, an increase, increase or prolongation of anti-tumor activity of an immune effector cell when combined with an immune effector cell; an increase in the number of anti-tumor immune effector cells or activated immune effector cells; and promotion of IFN- ⁇ secretion Radiation dose or radiation source for tumor regression, tumor shrinkage, tumor necrosis.
  • An effective amount of immune effector cells means, but is not limited to, an increase, increase or prolongation of anti-tumor activity of the immune effector cells when combined with local radiation; an increase in the number of anti-tumor immune effector cells or activated immune effector cells; promotion of IFN- ⁇ secretion; tumor The number of immune effector cells that resolve, tumor shrinkage, and tumor necrosis.
  • lymphocyte clearance is the removal of lymphocytes from a subject in the body. This includes administering a lymphocyte scavenger, systemic radiation therapy, or a combination thereof.
  • a lymphocyte scavenger e.g, a SELvIII-binding CAR molecule
  • the subject may be administered alone or in combination with one or more agents capable of substantially clearing the lymphocytes of the subject, systemic radiation therapy, or a combination thereof.
  • the clearing treatment can be administered under conditions sufficient to achieve a subject's lymphocyte clearance of 50%-100%.
  • the number of lymphocytes in the subject is reduced by at least 50%, 51%, 52%, 53%, 54%, 55%, 56%, 57%, 58%, 59%, 60%, 61%, 62%, 63 %, 64%, 65%, 66%, 67%, 68%, 69%, 70%, 71%, 72%, 73%, 74%, 75%, 76%, 77%, 78%, 79%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96% , 97%, 98%, 99% or 100%, the subject's lymphocytes were cleared.
  • Lymphocyte scavengers are anti-tumor chemotherapeutic agents.
  • lymphocyte scavengers include, but are not limited to, fludarabine, cyclophosphamide, or combinations thereof.
  • the treating physician can select a specific lymphocyte scavenger and a suitable dose according to the subject to be treated, such as CAMPATH, anti-CD3 antibody, cyclosporin, FK506, rapamycin, mycophenolic acid, steroid, FR901228, Melphalan, cyclophosphamide, fludarabine, and whole body radiation therapy.
  • Immune effector cell administration is administered before, during, and after the clearing treatment, or in combination, i.e., before, during, and after, during, and after, or before, during, and after the clearing treatment.
  • the clearing treatment is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 before the immune effector cell treatment Hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, Administration is performed on days 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 1 or any combination thereof.
  • the clearing treatment is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, after administration of the immune effector cell therapy, 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days , 17 days, 18 days, 19 days, 20 days, 21 days, 22 days, 23 days, 24 days, 25 days, 26 days, 27 days, 28 days, 29 days, 1 month, or any combination thereof.
  • chimeric antigen receptor refers to a group of polypeptides that, when administered in an immune effector cell, provide said cells with specificity for a target cell, typically a cancer cell, and have Intracellular signal production.
  • CAR typically includes at least one extracellular antigen binding domain, a transmembrane domain, and a cytoplasmic signaling domain (also referred to herein as an "intracellular signaling domain”), including stimulatory molecules derived from the definitions below and / Or a functional signaling domain of a co-stimulatory molecule.
  • the polypeptide groups are contiguous with each other.
  • a polypeptide group includes a dimerization switch that can couple the polypeptides to each other in the presence of a dimerization molecule, for example, an antigen binding domain can be coupled to an intracellular signaling domain.
  • the stimulatory molecule is an ⁇ chain that binds to a T cell receptor complex.
  • the cytoplasmic signaling domain further comprises one or more functional signaling domains derived from at least one costimulatory molecule as defined below.
  • the costimulatory molecule is selected from a costimulatory molecule described herein, such as 4-1BB (ie, CD137), CD27, and/or CD28.
  • a CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain, and an intracellular signaling domain comprising a functional signaling domain derived from a stimulatory molecule.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain, and a functional signaling domain comprising a costimulatory molecule and a functionality derived from a stimulatory molecule The intracellular signaling domain of the signaling domain.
  • the CAR comprises a chimeric fusion protein comprising an extracellular antigen binding domain, a transmembrane domain, and two functional signaling comprising one or more costimulatory molecules.
  • EGFRvIII was selected as a target of CAR-T cells, and in order to more accurately verify the anti-tumor effect in mice, the selected signal peptide, transmembrane region, intracellular region and the like were mouse-derived.
  • the method of preparation is operated according to conventional CAR-T cell preparation methods in the art.
  • the above embodiment selects a CAR of a murine source, but its signal peptide, hinge region, transmembrane region, and the like may be selected from other species depending on the purpose. These include, but are not limited to, human signal peptides, hinge regions, transmembrane regions, intracellular regions.
  • Antibodies can also be selected for murine anti- or humanized antibodies or whole human antibodies against different targets for different purposes.
  • stimulation refers to the binding of a stimulatory molecule (eg, a TCR/CD3 complex or CAR) to its cognate ligand (or a tumor antigen in the case of a CAR), thereby mediating signal transduction events (eg, However, it is not limited to the initial response induced via signal transduction of the TCR/CD3 complex or via signal transduction of a suitable NK receptor or CAR signaling domain. Stimulation can mediate altered expression of certain molecules.
  • a stimulatory molecule eg, a TCR/CD3 complex or CAR
  • the term "irritating molecule” refers to a molecule that provides a cytoplasmic signaling sequence expressed by immune cells (eg, T cells, NK cells, B cells) that modulate the signaling pathway for immune cells in an irritating manner. At least some aspects of activation of immune cells.
  • the signal is a primary signal initiated by binding of, for example, a TCR/CD3 complex to a peptide-loaded MHC molecule, and which results in a T cell response, including, but not limited to, proliferation, activation, differentiation, and the like.
  • a primary cytoplasmic signaling sequence that functions in a stimulatory manner can contain a signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM.
  • ITAM-containing cytoplasmic signaling sequences specifically for use in the present invention include, but are not limited to, those derived from CD3 ⁇ , common FcR ⁇ (FCER1G), Fc ⁇ RIIa, FcR ⁇ (FcEpsilon R1b), CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, DAP10 and DAP12.
  • the intracellular signaling domain in any one or more of the CARs of the invention comprises an intracellular signaling sequence, such as a CD3- ⁇ primary signaling sequence.
  • the primary signaling sequence of CD3- ⁇ is an equivalent residue derived from a human or non-human species such as mouse, rodent, monkey, donkey, and the like.
  • co-stimulatory molecule refers to a homologous binding partner on a T cell that specifically binds to a costimulatory ligand, thereby mediating a costimulatory response of a T cell, such as, but not limited to, proliferation.
  • a costimulatory molecule is a cell surface molecule other than an antigen receptor or its ligand that promotes an effective immune response.
  • Costimulatory molecules include, but are not limited to, MHC class I molecules, BTLA and Toll ligand receptors, and OX40, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278) and 4- 1BB (CD137).
  • costimulatory molecules include CDS, ICAM-1, GITR, BAFFR, HVEM (LIGHTR), SLAMF7, NKp80 (KLRF1), NKp44, NKp30, NKp46, CD160, CD19, CD4, CD8 ⁇ , CD8 ⁇ , IL2R ⁇ , IL2R ⁇ , IL7R ⁇ , 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, NKG2D, NKG2C, TNFR2, TRANCE/RANKL, DNAM1 (CD226), SLAMF4 (CD244, 2B4), CD84, CD96 (Tactile), CEACAM1, CRTAM, Ly9 (CD22), CD19
  • the costimulatory intracellular signaling domain can be an intracellular portion of a costimulatory molecule.
  • Costimulatory molecules can be represented by the following protein families: TNF receptor proteins, immunoglobulin-like proteins, cytokine receptors, integrins, signaling lymphocyte activating molecules (SLAM proteins), and NK cell receptors.
  • Examples of such molecules include CD27, CD28, 4-1BB (CD137), OX40, GITR, CD30, CD40, ICOS, BAFFR, HVEM, ICAM-1, antigen-related antigen-1 (LFA-1), CD2, CDS, CD7, CD287, LIGHT, NKG2C, NKG2D, SLAMF7, NKp80, NKp30, NKp44, NKp46, CD160, B7-H3, and ligands that specifically bind to CD83.
  • the intracellular signaling domain may comprise part or all of the native intracellular signaling domain, or a functional fragment or derivative thereof, of all cells within the molecule.
  • 4-1BB refers to a member of the TNFR superfamily having an amino acid sequence as provided by GenBank Accession No. AAA62478.2, or an equivalent residue from a non-human species such as a mouse, rodent, monkey, donkey, and the like;
  • the "4-1BB costimulatory domain” is defined as amino acid residues 214-255 of GenBank Accession No. AAA62478.2, or equivalent residues from non-human species such as mice, rodents, monkeys, ticks, and the like.
  • the "4-1BB costimulatory domain” is an equivalent residue from a human or from a non-human species such as a mouse, rodent, monkey, donkey, and the like.
  • intracellular signaling domain refers to an intracellular portion of a molecule.
  • the intracellular signaling domain produces a signal that promotes the immune effector function of cells containing CAR, such as CART cells.
  • immune effector functions in, for example, CART cells include cell lytic activity and helper activity, including secretion of cytokines.
  • the intracellular signaling domain can comprise a first order intracellular signaling domain.
  • Exemplary primary intracellular signaling domains include those derived from molecules responsible for primary stimulation or antigen dependent stimulation.
  • the intracellular signaling domain can comprise a costimulatory intracellular domain.
  • the first-level intracellular signaling domain can include a signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM.
  • ITAM immunoreceptor tyrosine-based activation motif
  • Examples of primary cytoplasmic signaling sequences containing ITAM include, but are not limited to, those derived from: CD3 ⁇ , common FcR ⁇ (FCER1G), Fc ⁇ RIIa, FcR ⁇ (FcEpsilon R1b), CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD79a, CD79b, DAP10 And DAP12.
  • T cell receptor a characteristic marker on the surface of all T cells, binds to CD3 with a non-covalent bond to form a TCR-CD3 complex.
  • the TCR is responsible for identifying antigens that bind to major histocompatibility complex molecules.
  • TCR is a heterodimer composed of two different peptide chains, consisting of two peptide chains, ⁇ and ⁇ . Each peptide chain can be further divided into variable region (V region), constant region (C region), transmembrane. The region and the cytoplasmic region are several parts; it is characterized by a short cytoplasmic region.
  • the TCR molecule belongs to the immunoglobulin superfamily, and its antigen specificity exists in the V region; the V region (V ⁇ , V ⁇ ) has three hypervariable regions CDR1, CDR2, and CDR3, among which the CDR3 mutation is the largest, which directly determines the TCR antigen. Binding specificity. When the TCR recognizes the MHC-antigen peptide complex, CDR1, CDR2 recognizes and binds to the side wall of the MHC molecule antigen binding groove, and CDR3 binds directly to the antigen peptide.
  • TCR is divided into two categories: TCR1 and TCR2; TCR1 consists of two chains of ⁇ and ⁇ , and TCR2 consists of two chains of ⁇ and ⁇ .
  • T cell fusion protein includes various polypeptide-derived recombinant polypeptides constituting a TCR, which are capable of binding to a surface antigen on a target cell, and to other polypeptides of the intact TCR complex. The effect is usually located on the surface of the T cell.
  • TFP consists of an antigen binding domain consisting of a TCR subunit and a human or humanized antibody domain, wherein the TCR subunit comprises at least a portion of the TCR extracellular domain, the transmembrane domain, and the TCR intracellular domain.
  • the TCR subunit is operably linked to the antibody domain, wherein the extracellular, transmembrane, and intracellular signal domains of the TCR subunit are derived from CD3 epsilon or CD3 gamma, and the TFP is integrated TCR expressed on T cells.
  • T cell antigen coupler includes three functional domains: 1. Tumor targeting domain, including single-chain antibody, designed ankyrin repeat protein (DARPin). Or other targeting group; 2.
  • the extracellular domain domain is a single-chain antibody that binds to CD3, thereby bringing the TAC receptor closer to the TCR receptor; 3.
  • the transmembrane region and the intracellular region of the CD4 co-receptor Among them, the intracellular domain is linked to protein kinase LCK, which catalyzes the phosphorylation of immunoreceptor tyrosine activation motifs (ITAMs) of TCR complexes as an initial step in T cell activation.
  • ITAMs immunoreceptor tyrosine activation motifs
  • antibody refers to a protein or polypeptide sequence derived from an immunoglobulin molecule that specifically binds an antigen.
  • Antibodies can be polyclonal or monoclonal, multi-stranded or single-stranded, or intact immunoglobulins, and can be derived from natural or recombinant sources.
  • the antibody can be a tetramer of immunoglobulin molecules.
  • antibody fragment refers to at least a portion of an antibody that retains the ability to specifically interact with an epitope of an antigen (eg, by binding, steric hindrance, stabilization/destabilization, spatial distribution).
  • antibody fragments include, but are not limited to, Fab, Fab', F(ab')2, Fv fragments, scFv antibody fragments, disulfide-linked Fvs (sdFv), Fd fragments consisting of VH and CH1 domains, Linear antibodies, single domain antibodies such as sdAb (VL or VH), camelid VHH domain, multispecific antibodies formed by antibody fragments (eg, bivalent fragments comprising two Fab fragments joined by a disulfide bond in the hinge region) and An isolated CDR or other epitope binding fragment of an antibody.
  • Antigen-binding fragments can also be incorporated into single domain antibodies, maximal antibodies, minibodies, Nanobodies, intrabodies, diabodies, triabodies, tetrabodies, v-NARs, and bis-scFvs (see, for example, Hollinger and Hudson, Nature Biotechnology (23): 1126-1136, 2005).
  • scFv refers to a fusion protein comprising at least one variable region antibody fragment comprising a light chain and at least one antibody fragment comprising a variable region of a heavy chain, wherein said light and heavy chain variable regions are contiguous (for example, via a synthetic linker such as a short flexible polypeptide linker), and can be expressed as a single-chain polypeptide, and wherein the scFv retains the specificity of the intact antibody from which it is derived.
  • a synthetic linker such as a short flexible polypeptide linker
  • an scFv can have the VL and VH variable regions in any order (eg, relative to the N-terminus and C-terminus of the polypeptide), and the scFv can include a VL-linker-VH or A VH-linker-VL can be included.
  • antibody heavy chain refers to the larger of the two polypeptide chains that are present in the antibody molecule in their naturally occurring configuration and which typically determine the type to which the antibody belongs.
  • antibody light chain refers to the smaller of the two polypeptide chains present in the antibody molecule in their naturally occurring configuration.
  • the ⁇ (k) and ⁇ (l) light chains refer to the isoforms of the two major antibody light chains.
  • recombinant antibody refers to an antibody produced using recombinant DNA techniques, such as, for example, an antibody expressed by a bacteriophage or yeast expression system.
  • the term should also be interpreted to mean an antibody that has been produced by synthesizing a DNA molecule encoding an antibody (and wherein the DNA molecule expresses the antibody protein) or an amino acid sequence of a specified antibody, wherein the DNA or amino acid sequence has been obtained using recombinant DNA or is available in the art. And well known amino acid sequence techniques are available.
  • antigen refers to a molecule that elicits an immune response.
  • the immune response can involve activation of the antibody-producing or cells with specific immunity or both.
  • any macromolecule comprising virtually all proteins or peptides can serve as an antigen.
  • the antigen can be derived from recombinant or genomic DNA.
  • any DNA comprising a nucleotide sequence or a partial nucleotide sequence encoding a protein that causes an immune response, thus encoding an "antigen.”
  • the antigen need not be encoded only by the full length nucleotide sequence of the gene.
  • the invention includes, but is not limited to, the use of partial nucleotide sequences of more than one gene, and these nucleotide sequences are arranged in different combinations to encode a polypeptide that elicits a desired immune response.
  • the antigen does not need to be encoded by a "gene” at all.
  • the antigen may be produced synthetically, or may be derived from a biological sample, or may be a macromolecule other than a polypeptide.
  • biological samples can include, but are not limited to, tissue samples, tumor samples, cells or liquids with other biological components.
  • Tumor antigen refers to a common antigen of a particular hyperproliferative disease.
  • the hyperproliferative disorder antigen of the invention is derived from cancer.
  • the tumor antigen of the present invention includes, but is not limited to, thyroid stimulating hormone receptor (TSHR); CD171; CS-1; C-type lectin-like molecule-1; ganglioside GD3; Tn antigen; CD19; CD20; CD 22; CD 30; CD 70; CD 123; CD 138; CD33; CD44; CD44v7/8; CD38; CD44v6; B7H3 (CD276), B7H6; KIT (CD117); interleukin 13 receptor subunit ⁇ (IL-13R ⁇ ); 11 receptor alpha (IL-11R ⁇ ); prostate stem cell antigen (PSCA); prostate specific membrane antigen (PSMA); carcinoembryonic antigen (CEA); NY-ESO-1; HIV-1 Gag; MART-1; gp100; Lysin; mesothelin
  • cancer refers to a broad class of disorders characterized by hyperproliferative cell growth in vitro (eg, transformed cells) or in vivo.
  • Conditions which may be treated or prevented by the methods of the invention include, for example, various neoplasms, including benign or malignant tumors, various hyperplasias and the like.
  • the methods of the invention may achieve inhibition and/or reversal of undesirable hyperproliferative cell growth involved in such conditions.
  • cancer examples include, but are not limited to, breast cancer, blood cancer, colon cancer, rectal cancer, renal cell carcinoma, liver cancer, non-small cell cancer of the lung, small intestine cancer, esophageal cancer, melanoma, bone cancer, pancreatic cancer, Skin cancer, glioma, head and neck cancer, skin or intraocular malignant melanoma, uterine cancer, ovarian cancer, rectal cancer, anal cancer, gastric cancer, testicular cancer, uterine cancer, fallopian tube cancer, endometrial cancer, cervix Cancer, vaginal cancer, vulvar cancer, Hodgkin's disease, non-Hodgkin's lymphoma, endocrine system cancer, thyroid cancer, parathyroid carcinoma, adrenal cancer, soft tissue sarcoma, urethral cancer, penile cancer, solid tumor of child, bladder Cancer, renal or ureteral cancer, renal pelvic cancer, central nervous system (CNS) tumor, primary CNS lymphoma, tumor angiogenesis
  • transfected or “transformed” or “transduced” refers to the process by which an exogenous nucleic acid is transferred or introduced into a host cell.
  • a “transfected” or “transformed” or “transduced” cell is one that has been transfected, transformed or transduced with an exogenous nucleic acid.
  • the cells include primary subject cells and their progeny.
  • the term "specifically binds” refers to an antibody or ligand that recognizes and binds to a binding partner (eg, tumor antigen) protein present in a sample, but the antibody or ligand does not substantially recognize or bind other molecules in the sample. .
  • biologically equivalent refers to an agent different from a reference compound, an immune effector cell, or an irradiation agent required to produce an effect equivalent to a reference compound or a reference amount of a reference compound, an immune effector cell, or an irradiation-producing effect. the amount.
  • refractory refers to a disease, such as cancer, which does not respond to treatment.
  • the refractory cancer can be resistant to treatment prior to or at the onset of treatment.
  • the refractory cancer can be resistant during treatment.
  • Refractory cancer is also known as resistant cancer.
  • refractory cancers include, but are not limited to, cancers that are insensitive to radiotherapy, relapse after radiotherapy, insensitive to chemotherapy, relapse after chemotherapy, insensitive to CAR-T therapy, or relapse after treatment.
  • the treatment regimens described herein can be used for refractory or recurrent malignancies.
  • Relapsed refers to the return of a disease (eg, cancer) or signs and symptoms of a disease, such as cancer, over a period of improvement, for example, after a previous treatment of a therapy, such as a cancer therapy.
  • a disease eg, cancer
  • signs and symptoms of a disease such as cancer
  • a therapy such as a cancer therapy
  • therapeutic agent refers to any medical product that produces a therapeutic response in a subject. These include, but are not limited to, immunostimulatory agents, T cell growth factors, interleukins, antibodies and vaccines, chemotherapeutic agents, or combinations thereof.
  • An immunostimulatory agent is a substance (drug and nutrient) that stimulates the immune system by inducing activation of any one of the components of the immune system or increasing the activity of any of its components.
  • Immunostimulants include bacterial vaccines, colony stimulating factors, interferons, interleukins, other immunostimulants, therapeutic vaccines, vaccine combinations, and viral vaccines.
  • T cell growth factor is a protein that stimulates T cell proliferation.
  • T cell growth factors include Il-2, IL-7, IL-15, IL-17, IL21, and IL-33.
  • Interleukins are a group of cytokines that are first thought to be expressed by white blood cells. The function of the immune system depends to a large extent on interleukins, and many of the rare defects of interleukins have been described, all characterized by autoimmune diseases or immunodeficiencies. Most of the interleukins are synthesized by helper CD4 T lymphocytes, as well as by monocytes, macrophages, and endothelial cells. They promote the development and differentiation of T and B lymphocytes as well as hematopoietic cells.
  • interleukins examples include IL-1, IL-2, IL-3, IL-4, IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, IL- 12. IL-13, IL-14, IL-15 and IL-17.
  • Chemotherapeutic agents include alkylating agents such as thiotepa and cyclophosphamide (CYTOXAN); alkyl sulfonates such as busulfan, acetaminophen and piperazine; aziridines such as benzotropipine ( Benzodopa), carbofuran, meturedopa and uredopa; ethyleneimine and methylamelamines, including hexamethylene melamine, triethylene melamine, three Ethylenephosphoramide, triethyl thiophosphoramide and trimethylolomelamine; nitrogen mustards such as chlorambucil, naphthyl mustard, cholophosphamide, estramustine, ifosfamide, nitrogen Mustard, oxychloride mustard, melphalan, neo-nitrogen mustard, fentanyl sterol, pine benzene mustard, tromethamine, uracil mustard; nitroure
  • an antihormonal agent for regulating or inhibiting the action of a hormone on a tumor
  • an antiestrogenic agent including, for example, tamoxifen, raloxifene, aromatase inhibitory 4 (5) -imidazole, 4-hydroxytamoxifen, trovaxifene, keoxifene, LY117018, ol's ketone and faremis (Fareston); and antiandrogens such as flutamide, nis Rummet, bicalutamide, leuprolide, and goserelin; and a pharmaceutically acceptable salt, acid or derivative of any of the above.
  • cancer therapeutics include sorafenib and other protein kinase inhibitors such as afatinib, axitinib, bevacizumab, cetuximab, crizotinib, dasatinib, erg Lotitinib, fotininib, gefitinib, imatinib, lapatinib, levabinib, moritinib, nilotinib, panitumumab, pazopanib, pega Tani, ranibizumab, rosobinib, trastuzumab, vandetanib, vemurafenib, and sunitinib; sirolimus (rapamycin), ivimo Division and other mTOR inhibitors.
  • protein kinase inhibitors such as afatinib, axitinib, bevacizumab, cetuximab, crizotinib, dasatinib,
  • chemotherapeutic agents include topoisomerase I inhibitors (eg, irinotecan, topotecan, camptothecin and its analogs or metabolites, and doxorubicin); topoisomerase II inhibition Agents (eg etoposide, teniposide and daunorubicin); alkylating agents (eg melphalan, chlorambucil, busulfan, thiotepa, ifosfamide, carmustine) , lomustine, semustine, streptozotocin, azomethamine, methotrexate, mitomycin C, and cyclophosphamide); DNA intercalators (eg, cisplatin, oxaliplatin, and Carboplatin); DNA intercalators and free radical generators such as bleomycin; and nucleoside mimics (eg 5-fluorouracil, capecitabine, gemcitabine, fludarabine, cytarabine,
  • chemotherapeutic agents that disrupt cell replication include: paclitaxel, docetaxel, and related analogs; vincristine, vinblastine, and related analogs; thalidomide, lenalidomide, and related analogs (eg, CC-5013 and CC-4047); protein tyrosine kinase inhibitors (eg imatinib mesylate and gefitinib); proteasome inhibitors (eg bortezomib); NF- ⁇ B inhibitors including I ⁇ B Inhibitors of kinases; antibodies that bind to proteins overexpressed in cancer, and other inhibitors of proteins or enzymes known to be upregulated, overexpressed or activated in cancer, which inhibit downregulation of cells.
  • Local radiation and immune effector cell therapy combined with checkpoint inhibitors and therapeutic agents may be more effective in treating cancer in some subjects, and/or may initiate, achieve, increase, enhance or prolong immune cells (Including the activity and/or number of T cells, B cells, NK cells and/or others, or promoting IFN- ⁇ secretion, or communicating a medically beneficial response (including regression, necrosis or elimination thereof) through the tumor.
  • the immune effector cells are administered concurrently with the checkpoint inhibitor, or the immune effector cell therapy is administered chronologically before or after the checkpoint inhibitor.
  • the immune effector cell treatment is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours prior to administration of the checkpoint inhibitor.
  • 12 hours 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 Administration on days, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 1 or any combination thereof.
  • the immune effector cell treatment is 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours after administration of the checkpoint inhibitor. , 12 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, 13 days, 14 days, 15 days, 16 Administration on days, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 1 or any combination thereof.
  • the therapeutic agent is administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 before the administration of the immune effector cells.
  • the therapeutic agent is administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 days, 8 days, 9 hours, 10 hours, 11 days, 12 days, 13 days, 14 days, 15 days, 16 days, Administration is performed on days 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 1 or any combination thereof.
  • the therapeutic agent is administered 1 hour, 2 hours, 3 hours, 4 hours, 5 hours, 6 hours, 7 hours, 8 hours, 9 hours, 10 hours, 11 hours, 12 hours after administration of the immune effector cells.
  • biological therapeutic agent refers to any medical product that is manufactured in a biological source or extracted from a biological source.
  • Biopharmaceuticals are different from chemically synthesized pharmaceutical products.
  • biological agents include vaccines, blood or blood components, allergens, somatic cells, gene therapy, tissues, recombinant therapeutic proteins, including antibody therapeutics and fusion proteins, and living cells.
  • Biological products can be composed of sugars, proteins or nucleic acids or complex combinations of these substances, or can be living entities such as cells and tissues.
  • Biological products are isolated from a variety of natural sources (human, animal or microbial) and can be produced by biotechnological methods and other techniques.
  • Specific examples of biological therapeutic agents include, but are not limited to, immunostimulatory agents, T cell growth factors, interleukins, antibodies, fusion proteins, and vaccines such as cancer vaccines.
  • treating refers to slowing or ameliorating the progression, severity and/or duration of a proliferative disorder, or amelioration of hyperplasia, due to administration of one or more therapies (eg, one or more therapeutic agents, such as the CAR of the invention).
  • One or more symptoms of a sexual condition preferably, one or more discernible symptoms.
  • the term “treating” refers to ameliorating at least one measurable physical parameter of a proliferative disorder, such as tumor growth, that is not necessarily discernible by a patient.
  • the term “treating” refers to inhibiting the progression of a proliferative disorder physically, by, for example, stabilizing physical parameters, physiologically, or both, by, for example, stabilizing the discernible symptoms.
  • the term “treating” refers to reducing or stabilizing tumor size, counting cancer cells, or prolonging the survival of an individual.
  • improving survival refers to an increase in the lifespan or quality of life of a subject having cancer or a proliferative disease. For example, improving survival also includes promoting cancer remission, preventing tumor invasion, preventing tumor recurrence, slowing tumor growth, preventing tumor growth, reducing tumor size, and reducing total cancer cell count.
  • treating cancer is not meant to be an absolute term.
  • the methods of the invention seek to reduce tumor size or number of cancer cells, promote cancer into remission, or prevent the growth of cancer cells in size or number of cells. In some cases, treatment results in an improved prognosis.
  • autologous refers to any substance derived from an individual that is subsequently introduced to the same individual.
  • allogeneic refers to any substance derived from a different animal of the same species as the individual into which the substance will be introduced. When the genes of one or more loci are not identical, two or more individuals are considered to be allogeneic to each other. In certain aspects, allogeneic materials from individuals of the same species may be genetically sufficiently different to interact antigenically.
  • tumor response refers to a cellular response including, but not limited to, triggering programmed cell death.
  • anti-tumor response refers to an immune system response including, but not limited to, activating T cells to challenge an antigen or antigen presenting cells.
  • small molecule refers to a low molecular weight ( ⁇ 900 Daltons) organic compound that can help regulate biological processes with a size on the order of 10-9 m. Most drugs are small molecules.
  • the immune checkpoint regulates T cell function in the immune system.
  • T cells play a key role in cell-mediated immunity.
  • the checkpoint protein interacts with a specific ligand that signals the T cells and substantially shuts down or inhibits T cell function.
  • Cancer cells utilize this system by driving high levels of checkpoint protein expression on their surface, which results in the control of T cells expressing checkpoint proteins on the surface of T cells entering the tumor microenvironment, thus suppressing the anti-cancer immune response . As such, inhibition of checkpoint proteins will result in restoration of T cell function and immune response against cancer cells.
  • checkpoint proteins include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4 (of the CD2 family of molecules, and All NK, ⁇ and memory CD8+ ( ⁇ ) T cells are expressed), CD160 (also known as BY55), CGEN-15049, CHK 1 and CHK2 kinase, A2aR and various B-7 family ligands.
  • PD-1 antibody refers to an antibody that antagonizes the activity and/or proliferation of lymphocytes by agonizing PD-1.
  • antagonistic activity refers to a decrease (or decrease) in at least about 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90% or more of lymphocyte proliferation or activity.
  • antagonistagonism can be used interchangeably with the terms “inhibiting” and “inhibiting”.
  • PD-1 Programmed cell death protein 1
  • PD-1 has two ligands, PD-L1 and PD-L2, which are members of the B7 family.
  • PD-L1 protein is up-regulated in macrophages and dendritic cells (DC) in response to LPS and GM-CSF treatment, and up-regulated on T and B cells after TCR and B cell receptor signaling, In resting mice, PDL1 mRNA can be detected in the heart, lung, thymus, spleen and kidney. PD-1 negatively regulates T cell responses.
  • CTLA4 cytotoxic T lymphocyte-associated protein
  • CTLA4 is a protein receptor that down-regulates the immune system.
  • CTLA4 is found on the surface of T cells, which results in cellular immune attack on antigen.
  • T cell challenge can be initiated by stimulating the CD28 receptor on T cells.
  • T cell challenge can be turned off by stimulating the CTLA4 receptor.
  • the first class of immunotherapy Yervoy (a monoclonal antibody that targets CTLA-4 on the surface of T cells), is approved for the treatment of melanoma.
  • the checkpoint inhibitor is a biological therapeutic agent or a small molecule.
  • the checkpoint inhibitor is a monoclonal antibody, a humanized antibody, a fully human antibody, a fusion protein, or a combination thereof.
  • the checkpoint inhibitor inhibits checkpoint proteins, which can be CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4 , CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or combinations thereof.
  • the checkpoint inhibitor interacts with a ligand that can be a checkpoint protein: CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160, CGEN-15049, CHK 1, CHK2, A2aR, B-7 family ligands or combinations thereof.
  • the therapeutic agent is an immunostimulatory agent, a T cell growth factor, an interleukin, an antibody, a vaccine, a chemotherapeutic drug, or a combination thereof.
  • the immunological checkpoint inhibitor is administered prior to, concurrently with, or subsequent to the topical radiation therapy, or the immunological checkpoint inhibitor is administered prior to, concurrently with, or subsequent to the immunotherapeutic cell therapy.
  • Checkpoint inhibitors include any agent that blocks or inhibits the inhibitory pathway of the immune system in a statistically significant manner. Such inhibitors may include small molecule inhibitors, or may include antibodies that bind to and block or inhibit the immunological checkpoint receptor or antigen-binding fragments thereof, or antibodies that bind to and block or inhibit the immunological checkpoint receptor ligand.
  • Exemplary checkpoint molecules that can be targeted for blockade or inhibition include, but are not limited to, CTLA-4, PDL1, PDL2, PD1, B7-H3, B7-H4, BTLA, HVEM, GAL9, LAG3, TIM3, VISTA , KIR, 2B4 (belonging to the CD2 family of molecules, and expressed on all NK, ⁇ and memory CD8+ ( ⁇ ) T cells), CD160 (also known as BY55), CGEN-15049, CHK 1 and CHK2 kinase, A2aR and various B-7 family ligand.
  • B7 family ligands include, but are not limited to, B7-1, B7-2, B7-DC, B7-H1, B7-H2, B7-H3, B7-H4, B7-H5, B7-H6, and B7-H7.
  • Checkpoint inhibitors include antibodies or antigen-binding fragments thereof, other binding proteins, biological therapeutics or small molecules that bind to and block or inhibit the activity of one or more of the following: CTLA-4, PDL1, PDL2 , PD1, BTLA, HVEM, TIM3, GAL9, LAG3, VISTA, KIR, 2B4, CD160 and CGEN-15049.
  • Illustrative immunological checkpoint inhibitors include tromezumab (CTLA-4 blocking antibody), anti-OX40, PD-L1 monoclonal antibody (anti-B7-H1; MEDI4736), MK-3475 (PD-1 blocking) , Nivolumab (anti-PD1 antibody), CT-011 (anti-PD1 antibody), BY55 monoclonal antibody, AMP224 (anti-PDL1 antibody), BMS-936559 (anti-PDL1 antibody), MPLDL3280A (anti-PDL1 antibody), MSB0010718C (antibiotic) PDL1 antibody) and Yervoy/Ipilimumab (anti-CTLA-4 checkpoint inhibitor).
  • Checkpoint protein ligands include, but are not limited to, PD-L1, PD-L2, B7-H3, B7-H4, CD28, CD86, and TIM-3.
  • Suitable anti-CTLA-4 antagonists for use in the methods of the invention include, but are not limited to, anti-CTLA4 antibodies, human anti-CTLA4 antibodies, mouse anti-CTLA4 antibodies, mammalian anti-CTLA4 antibodies, humanized anti-CTLA4 antibodies, monoclonal antibodies CTLA4 antibody, polyclonal anti-CTLA4 antibody, chimeric anti-CTLA4 antibody, MDX-010 (Ipilizumab), Trumeimumab, anti-CD28 antibody, anti-CTLA4adnectin, anti-CTLA4 domain antibody, single-stranded anti-CTLA4 fragment , heavy chain anti-CTLA4 fragment, light chain anti-CTLA4 fragment, inhibitor of CTLA4 agonizing the co-stimulatory pathway, antibody disclosed in PCT Publication No.
  • CTLA-4 antibodies are described in U.S. Patent Nos. 5,811,097, 5,855,887, 6,051,227 and 6,984,720; PCT Publication No. WO 01/14424 and WO 00/37504; and U.S. Publication Nos. 2002/0039581 and 2002/086014.
  • Other anti-CTLA-4 antibodies that can be used in the methods of the invention include, for example, those disclosed in WO 98/42752; U.S. Patent Nos.
  • Additional anti-CTLA4 antagonists include, but are not limited to, the ability to disrupt the ability of the CD28 antigen to bind to its cognate ligand, inhibit the ability of CTLA4 to bind its cognate ligand, enhance T cell responses via a costimulatory pathway, and disrupt B7 binding to CD28. And/or the ability of CTLA4 to disrupt B7's ability to activate costimulatory pathways, disrupt CD80's ability to bind to CD28 and/or CTLA4, disrupt CD80's ability to activate costimulatory pathways, disrupt CD86 binding to CD28 and/or CTLA4, and disrupt CD86 activation The ability to co-stimulate pathways, as well as any inhibitor that disrupts the activation of the costimulatory pathway in general.
  • Such requisites include small molecule inhibitors of CD28, CD80, CD86, CTLA4, and other members of the costimulatory pathway; antibodies to CD28, CD80, CD86, CTLA4, and other members of the costimulatory pathway; CD28 for the costimulatory pathway, Antisense molecules of CD80, CD86, CTLA4 and other members; CDnectin, CD80, CD86, CTLA4 and other member of the adnectin for co-stimulatory pathways, CDi, CD80, CD86, CTLA4 and other members of the RNAi inhibitors of the co-stimulatory pathway ( Both single-stranded and double-stranded, as well as other anti-CTLA4 antagonists.
  • the treatment is by clinical outcome; the anti-tumor activity of the T cell is increased, enhanced or prolonged; the increase in the number of anti-tumor T cells or activated T cells, promotes IFN- ⁇ secretion, or Combination decision.
  • the clinical outcome is tumor regression; tumor shrinkage; tumor necrosis; anti-tumor response through the immune system; tumor enlargement, recurrence or spread, or a combination thereof.
  • the therapeutic effect is predicted by the presence of T cells, the presence of a genetic marker indicative of T cell inflammation, promotion of IFN-[gamma] secretion, or a combination thereof.
  • the methods described herein are used to treat cancer.
  • the methods described herein can be used to reduce the size of a solid tumor or to reduce the number of cancer cells in a cancer.
  • the methods described herein can be used to slow the rate of cancer cell growth.
  • the methods described herein can be used to stop the rate of cancer cell growth.
  • An immune effector cell, therapeutic agent, checkpoint inhibitor, biological therapeutic, or pharmaceutical composition as disclosed herein can be administered to an individual by a variety of routes including, for example, orally or parenterally, such as intravenously, intramuscularly, subcutaneously, Intraorbital, intracapsular, intraperitoneal, intrarectal, intracisternal, intratumoral, intravasally, intradermal or by passive or accelerated absorption through the skin, for example, by skin patch or transdermal iontophoresis.
  • the therapeutic agent, checkpoint inhibitor, biological therapeutic, or pharmaceutical composition can also be administered to a site of a pathological condition, such as intravenously or intraarterially into a blood vessel that supplies the tumor.
  • the total amount of agent to be administered in practicing the methods of the invention may be administered as a single dose by bolus or by infusion over a relatively short period of time, or may be administered using a fractionated treatment regimen, wherein over extended periods of time Multiple doses are administered in segments.
  • One skilled in the art will recognize that the amount of composition that treats a pathological condition in a subject depends on a number of factors, including the age and general health of the subject, as well as the route of administration and the number of treatments to be administered. With these factors in mind, the technician will adjust the specific dose as needed. In general, initially, phase I and phase II clinical trials are used to determine the radiation dose, mode, and dose of immune effector cells, as well as the route and frequency of administration.
  • a range such as 95-99% identity includes a range having 95%, 96%, 97%, 98%, or 99% identity, and includes subranges such as 96-99%, 96-98%, 96-97%, 97-99%, 97-98%, and 98-99% identity. This does not apply regardless of the width of the range.
  • Exemplary antigen receptors of the present invention including CAR, and methods for engineering and introducing a receptor into a cell, are described, for example, in Chinese Patent Application Publication No. CN107058354A, CN107460201A, CN105194661A, CN105315375A, CN105713881A, CN106146666A, CN106519037A, CN106554414A. , CN105331585A, CN106397593A, CN106467573A, CN104140974A, International Patent Application Publication No. WO2017186121A1, WO2018006882A1, WO2015172339A8, WO2018018958A1.
  • this example employs a second generation CAR that targets EGFRvIII, which is used to construct a transmembrane domain and an intracellular domain of CAR using the gene sequence of the mouse for the needs of later animal experiments.
  • a coding sequence of a mouse CD8 ⁇ signal peptide (SEQ ID NO: 1), a coding sequence for recognizing human activated EGFR, a monoclonal antibody that also recognizes human EGFRvIII (SEQ ID NO: 2), a murine CD8 alpha hinge region, and
  • the coding sequence of the transmembrane region (SEQ ID NO: 3), the coding sequence of the murine CD28 intracellular domain (SEQ ID NO: 4), and the coding sequence of the murine CD3 sputum intracellular domain (SEQ ID NO: 5) were sequentially ligated in vitro.
  • 293T cells were infected with the recombinant vector MSCV-EGFRvIII-mCD28Z to obtain a packaged retrovirus. Infection methods are routine infection methods in the preparation of T cells expressing chimeric antigen receptors in the art.
  • mice purchased from Shanghai Xipuer-Beikai Experimental Animal Co., Ltd.
  • the spleen T lymphocytes of Balb/c mice were taken, and the purified mouse CD3+ T lymphocytes were added to the Dynabeads Mouse T-activator CD3 in a volume ratio of 1:1. /CD28 (Thermo Fisher), washed once with PBS, activated, and cultured in an incubator.
  • the medium was RPMI 1640 complete medium.
  • Mouse spleen T lymphocytes activated for 24 h were inoculated into a retronectin (Takara T100A)-coated 12-well plate, and retrovirus infection was added overnight to obtain mouse EGFRvIII-m28Z CAR-T cells.
  • Balb/c mice are mice with normal immune systems. Balb/c mice were given 5Gy ⁇ -rays for whole body irradiation. On the 14th day after irradiation, 50 ⁇ l of mouse mandibular blood was collected into anticoagulation tubes, and PerCP Cy5.5-labeled anti-mouse CD3 antibody was added and incubated for 1 hour at room temperature. 450 ⁇ l of red blood cell lysate was added, and after ten minutes, the BD flow analyzer was used to detect the CD3 positive rate.
  • CT26 cell model (CT26-EGFRvIII) of mouse EGFR expressing the amino acid epitopes of chimeric human EGFR 287-302 was removed by conventional molecular molecular means to remove mouse EGFR exon 2-7 exon.
  • CT26 cells were purchased from the American Type Culture Collection (ATCC CRL-2638).
  • the tumor volume was 150-250 mm 3 , and the tumor-bearing mice were divided into 5 groups (6 in each group), which were:
  • UT (untreated) cell group (UT) Day13 tail vein infusion of 5 ⁇ 10 6 untreated mouse T cells;
  • tumor local radiation treatment group (X-ray): Day13 tumor locally received 10Gy X-ray irradiation; (2) EGFRvIII-m28Z CAR-T cell group (EGFRvIII-m28Z): Day13 tail vein 5 ⁇ 10 6 EGFRvIII-m28Z CAR-T cells were infused.
  • X-ray tumor local radiation treatment group
  • EGFRvIII-m28Z CAR-T cell group
  • EGFRvIII-m28Z CAR-T+ tumor local radiation treatment group (EGFRvIII-m28Z+X-ray): Day13 injection of 5 ⁇ 10 6 EGFRvIII-m28Z CAR-T cells, Day15 tumor localization at 1 day interval 10Gy X-ray irradiation;
  • Tumor local radiation therapy + EGFRvIII-m28Z CAR-T group (X-ray+EGFRvIII-m28Z): Day13 tumor locally received 10Gy X-ray irradiation, and Day 15 injection 5 ⁇ interval 1 day interval 10 6 EGFRvIII-m28Z CAR-T cells.
  • Figure 2 shows the results of tumor local radiation therapy combined with EGFRvIII-m28Z CAR-T cells to inhibit tumor growth in vivo.
  • the CAR-T treatment group EGFRvIII-m28Z group
  • the combination group EGFRvIII-m28Z+X-ray group, X-ray+EGFRvIII-m28Z group
  • Narrowing P ⁇ 0.05, Two way ANOVA
  • the tumor growth rate of the combined group was not significantly different from that of the CAR-T treatment group alone, indicating that the local radiation of the tumor could not be improved with CAR-T after the clearing pretreatment.
  • the role of cell combination in inhibiting tumor growth.
  • the tumor inhibition rate in the local radiation treatment group was about 27.27% ⁇ 18.61%, and the tumor suppression rate in the EGFRvIII-m28Z group.
  • the tumor inhibition rates of about 82.48% ⁇ 5.86%, EGFRvIII-m28Z+X-ray group and X-ray+EGFRvIII-m28Z group were 90.98% ⁇ 2.26% and 84.02% ⁇ 3.61%, respectively.
  • Example 3 EGFRvIII-m28Z CAR-T cells combined with local radiation of tumors for detection of IFN- ⁇ concentration in mouse colon cancer model
  • Example 4 EGFRvIII-m28Z CAR-T combined with local tumor radiation therapy for cell copy number detection of tumor local EGFRvIII-m28Z CAR-T in mouse colon cancer model
  • the tumor tissue DNA of Day27 to Day 40 days after the treatment in Example 2 was Day 27 to Day 40, and the EGFRvIII-m28Z CAR-T copy number was detected by real-time quantitative PCR.
  • Tumor tissue CAR T copy number detection DNA was extracted from each group of mouse tumor tissues, and the retroviral vector fragment sequence was detected by Taqman probe method to determine the DNA copy number of 806-m28z-CAR;
  • the primer sequence of the Taq probe method is as follows:
  • Probe primer sequence 5'-(FAM)-ATGGCCGCGA GACGGCACCT-(BHQ1)-3'
  • a CAR-T prepared by targeting mouse anti-EGFRvIII (SEQ ID NO: 2), murine transmembrane domain and intracellular domain, etc.
  • the coding sequence of the human CD8 ⁇ signal peptide (SEQ ID NO: 6), the coding sequence of the human CD8 ⁇ hinge region and the transmembrane region (SEQ ID NO: 7), and the coding sequence of the human CD28 transmembrane domain can be selected ( SEQ ID NO: 10), the coding sequence of the human CD28 intracellular domain (SEQ ID NO: 8), and the coding sequence of the human CD3 intracellular domain (SEQ ID NO: 9) were prepared.
  • the amino acid sequence of the chimeric antigen receptor targeting EFGRvIII employed is set forth in any one of SEQ ID NOs: 20, 21, 22.
  • the CAR used may be targeted to other antigens, such as GPC3 (exemplary, the amino acid sequence of the chimeric antigen receptor targeting GPC3 is SEQ ID NO: 11, 12, 13, 14, 15 CLD18A2 (exemplary, the amino acid sequence of the chimeric antigen receptor targeting CLD18A2 is as shown in any of SEQ ID NOs: 16, 17, 18, 19); mesothelin (exemplary, targeted) The amino acid sequence of the chimeric antigen receptor of mesothelin is as shown in any of SEQ ID NOs: 23, 24, 25, and 26).

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Cell Biology (AREA)
  • Microbiology (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Dermatology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

La présente invention concerne une utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs. L'invention se rapporte plus précisément à un procédé d'administration d'une utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie tumorale locale à un individu souffrant d'une tumeur pour effectuer un traitement, et la réalisation d'une lymphodéplétion sur l'individu ; les cellules effectrices immunitaires comprennent un antigène tumoral qui reconnaît une tumeur et un récepteur qui déclenche l'activation des cellules effectrices immunitaires. La présente invention a été confirmée au moyen de tests : le procédé de traitement de tumeurs selon la présente invention améliore l'effet anticancéreux pour des patients atteints de cancer qui ne sont pas sensibles au traitement CAR-T ou qui ont subi une récidive après le traitement, et améliore également les effets thérapeutiques anticancéreux pour des patients atteints de cancer, tels que l'élimination de tumeurs et la prolongation de la durée de vie.
PCT/CN2018/120593 2017-12-12 2018-12-12 Utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs WO2019114751A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201711318760 2017-12-12
CN201711318760.7 2017-12-12

Publications (1)

Publication Number Publication Date
WO2019114751A1 true WO2019114751A1 (fr) 2019-06-20

Family

ID=66819541

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2018/120593 WO2019114751A1 (fr) 2017-12-12 2018-12-12 Utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs

Country Status (1)

Country Link
WO (1) WO2019114751A1 (fr)

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111166878A (zh) * 2020-01-06 2020-05-19 上海鑫湾生物科技有限公司 靶向肿瘤抗原的抗体与iNKT细胞的组合的制备方法与用途
CN113896801A (zh) * 2021-10-08 2022-01-07 南京凯地医疗技术有限公司 靶向人Claudin18.2和NKG2DL的嵌合抗原受体细胞及其制备方法和应用
WO2022028623A1 (fr) 2020-08-07 2022-02-10 佧珐药业有限公司 Cellules modifiées et procédé de modification de cellules
WO2023274303A1 (fr) 2021-06-29 2023-01-05 科济生物医药(上海)有限公司 Polypeptide chimérique pour la régulation de l'activité physiologique cellulaire

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN106519037A (zh) * 2015-09-11 2017-03-22 科济生物医药(上海)有限公司 可活化的嵌合受体
CN107074973A (zh) * 2014-07-29 2017-08-18 辉瑞大药厂 用于癌症免疫疗法的EGFRvIII特异性嵌合抗原受体
CN107446051A (zh) * 2016-05-31 2017-12-08 上海恒润达生生物科技有限公司 靶向cd19的嵌合抗原受体及其用途

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107074973A (zh) * 2014-07-29 2017-08-18 辉瑞大药厂 用于癌症免疫疗法的EGFRvIII特异性嵌合抗原受体
CN106519037A (zh) * 2015-09-11 2017-03-22 科济生物医药(上海)有限公司 可活化的嵌合受体
CN107446051A (zh) * 2016-05-31 2017-12-08 上海恒润达生生物科技有限公司 靶向cd19的嵌合抗原受体及其用途

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN111166878A (zh) * 2020-01-06 2020-05-19 上海鑫湾生物科技有限公司 靶向肿瘤抗原的抗体与iNKT细胞的组合的制备方法与用途
CN111166878B (zh) * 2020-01-06 2024-01-02 上海鑫湾生物科技有限公司 靶向肿瘤抗原的抗体与iNKT细胞的组合的制备方法与用途
WO2022028623A1 (fr) 2020-08-07 2022-02-10 佧珐药业有限公司 Cellules modifiées et procédé de modification de cellules
WO2023274303A1 (fr) 2021-06-29 2023-01-05 科济生物医药(上海)有限公司 Polypeptide chimérique pour la régulation de l'activité physiologique cellulaire
CN113896801A (zh) * 2021-10-08 2022-01-07 南京凯地医疗技术有限公司 靶向人Claudin18.2和NKG2DL的嵌合抗原受体细胞及其制备方法和应用
CN113896801B (zh) * 2021-10-08 2022-05-17 南京凯地医疗技术有限公司 靶向人Claudin18.2和NKG2DL的嵌合抗原受体细胞及其制备方法和应用

Similar Documents

Publication Publication Date Title
WO2019114762A1 (fr) Utilisation combinée de cellules immunes effectrices et de rayonnement pour le traitement de tumeurs
KR102584300B1 (ko) 암을 표적하는 키메라 항원 수용체
US20240075070A1 (en) Chimeric antigen receptor t cells targeting the tumor microenvironment
Kohrt et al. Targeting CD137 enhances the efficacy of cetuximab
WO2019114751A1 (fr) Utilisation combinée de cellules effectrices immunitaires et d'une radiothérapie pour le traitement de tumeurs
CN111727373A (zh) 靶向bcma的嵌合抗原受体及其用途
KR20190101979A (ko) 합성 면역 수용체 및 이의 사용 방법
CN111787938A (zh) 靶向bcma的嵌合抗原受体、靶向cd19的嵌合抗原受体及组合疗法
CN113286811A (zh) 改善过继性细胞疗法的效力和安全性
CN110225756A (zh) 与用于穿透实体瘤的细胞系统相关的组合物和方法
CN112218886A (zh) 嵌合吞噬受体的表达载体、基因修饰的宿主细胞及其用途
US20190358262A1 (en) Methods for modulation of car-t cells
KR20180012851A (ko) 고 친화성 및 고 특이성을 가진 tcr-유사 항체 결합 도메인을 포함하는 친화성 엔티티 및 이의 용도
AU2019271819A1 (en) Genetically engineered cell and application thereof
CN113490689A (zh) Tcr融合蛋白及表达tcr融合蛋白的细胞
JP2021501801A (ja) 癌の処置に用いるための免疫刺激アゴニスト抗体
CN113164408A (zh) 肿瘤联合免疫治疗
EP3805366A1 (fr) Méthode de thérapie par lymphocytes t améliorée
CN114616337A (zh) 嵌合CD3融合蛋白与基于anti-CD3的双特异性T细胞激活元件的联合表达
AU2020373899A1 (en) Drug for treating cancer, combination drug, drug composition, immune responsive cell, nucleic acid delivery vehicle, and product
CA3134439A1 (fr) Variants d'interleukine-2 a activite biologique modifiee
EP4321533A1 (fr) Utilisation d'immunothérapie cellulaire
WO2023044039A1 (fr) Compositions et méthodes pour traiter le cancer
US20230295564A1 (en) Galectin-9 Specific Binding Agents for Use in Treating Cancer
Murad Developing Improved Preclinical Models for Effective Chimeric Antigen Receptor (CAR) T Cell Therapy for Solid Tumors

Legal Events

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

Ref document number: 18887338

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 18887338

Country of ref document: EP

Kind code of ref document: A1