CN117653733A - Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment - Google Patents

Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment Download PDF

Info

Publication number
CN117653733A
CN117653733A CN202311680205.4A CN202311680205A CN117653733A CN 117653733 A CN117653733 A CN 117653733A CN 202311680205 A CN202311680205 A CN 202311680205A CN 117653733 A CN117653733 A CN 117653733A
Authority
CN
China
Prior art keywords
cancer
piezo1
cells
inhibitor
technology
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202311680205.4A
Other languages
Chinese (zh)
Inventor
刘玉英
梁俊波
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Institute of Basic Medical Sciences of CAMS
Original Assignee
Institute of Basic Medical Sciences of CAMS
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 Institute of Basic Medical Sciences of CAMS filed Critical Institute of Basic Medical Sciences of CAMS
Priority to CN202311680205.4A priority Critical patent/CN117653733A/en
Publication of CN117653733A publication Critical patent/CN117653733A/en
Pending legal-status Critical Current

Links

Landscapes

  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the fields of biology, medicine and clinic, and particularly relates to application of a novel immune check point PIEZO1 pathway in tumor T cell treatment. Specifically, the invention provides application of PIEZO1 inhibitor in treating cancer, improving T cell killing ability and improving treatment effect of immune checkpoint inhibitor. The novel immune checkpoint PIEZO1 and the downstream path thereof open up an innovative strategy of tumor immunotherapy.

Description

Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment
Technical Field
The invention belongs to the fields of biology, medicine and clinic, and particularly relates to application of a novel immune check point PIEZO1 pathway in tumor T cell treatment.
Background
Cancer immunotherapy is defined as a method of combating cancer by generating or enhancing immune responses to cancer cells. In the last decade, two types of immunotherapy using immune checkpoint inhibitors to enhance natural anti-tumor activity and to administer specific anti-tumor immune cells by adoptive cell therapy have been very effective in cancer treatment.
Currently, the most widespread immunotherapeutic types are monoclonal antibodies directed against regulatory immune checkpoint molecules that inhibit T cell activation, in particular cytotoxic T lymphocyte-associated protein-4 (CTLA-4), programmed cell death-1 (PD-1) and programmed death ligand 1 (PD-L1). Immune checkpoint blockade (Immune checkpoint blockade, ICB) has enjoyed great success in treating a variety of cancer types, such as metastatic melanoma, non-small cell lung cancer, and renal cancer. Although this approach to treatment has been successfully applied to many solid tumors, the significant response to current ICB therapies is limited to a few cancer patients, as its primary mechanism relies on the enhancement of existing potentially tumor-reactive T cell populations in the patient's body, and most of the responding patients then relapse. Thus, immune checkpoint therapy alone may fail in a less immunogenic cancer type.
This highlights the urgent need to explore more efficient strategies by determining new immune checkpoints. To date, considerable effort has been expended to find T cell immune checkpoints based on biochemical signals, and we need to explore new T cell immune checkpoints in addition to biochemical signals and as targets in future clinical tumor immunotherapy.
Disclosure of Invention
The present invention discovers that PIEZO1 is a potential mechanical immune checkpoint in cytotoxic T cells that can be manipulated to enhance T cell traction and subsequently cytotoxicity to cancer cells, thereby killing tumor cells, thus providing a new treatment for cancer.
Specifically, the invention provides the following technical scheme:
application of PIEZO1
In a first aspect, the present invention provides the use of a PIEZO1 inhibitor in the preparation of:
1) A product for the treatment of cancer, which comprises,
2) A product for improving the killing capacity of T cells,
3) A product that enhances the therapeutic effect of immune checkpoint inhibitors (immune checkpoint inhibitors, ICIs).
The product comprises medicines, medicine compositions, medicine combinations and the like.
More preferably, the immune checkpoint is PD1 and the immune checkpoint inhibitor is an antibody targeting PD 1.
In particular, the killing ability is manifested in traction, enhanced cytotoxicity, and degree of tumor infiltration.
Pharmaceutical combination composition
In another aspect, the invention provides a pharmaceutical combination composition for treating cancer comprising a PIEZO1 inhibitor and an immune checkpoint inhibitor (immune checkpoint inhibitors, ICIs).
In a specific embodiment, the dosage forms of the PIEZO1 inhibitor and the immune checkpoint inhibitor in the pharmaceutical combination composition are the same or different.
In a specific embodiment, the PIEZO1 inhibitor and immune checkpoint inhibitor in the pharmaceutical combination composition are administered simultaneously or sequentially, in particular, may be administered at intervals of 0, 1, 2, 3, 4, 5, 6, 7 or more days.
The dosage form and the mode of administration of the pharmaceutical combination composition of the present invention are not particularly limited. Representative modes of administration include, but are not limited to: oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous) injection, and topical administration. Representative dosage forms include tablets, pills, powders, granules, capsules, lozenges, syrups, liquids, emulsions, suspensions, controlled release formulations, aerosols, films, injections, intravenous drip formulations, transdermal absorption formulations, ointments, lotions, adhesive formulations, suppositories, pellets, nasal formulations, pulmonary formulations, eye drops and the like.
In a specific embodiment, the dosage ratio of the PIEZO1 inhibitor and immune checkpoint inhibitor in the pharmaceutical combination composition is 1:0.1-2; specifically included are 3:1, 2:1, 1:1, 1:2, 1:3.
More preferably, the immune checkpoint is PD1 and the immune checkpoint inhibitor is an antibody targeting PD 1.
Preferably, the pharmaceutical combination composition further comprises pharmaceutically acceptable excipients, including any one or more of diluents, excipients, fillers, binders, wetting agents, disintegrants, emulsifiers, co-solvents, solubilizers, osmotic pressure regulators, surfactants, coating materials, colorants, pH regulators, antioxidants, bacteriostats or buffers.
T cell preparation method and product application
In another aspect, the present invention provides a method for preparing T cells with high killing ability, the method comprising the step of pre-treating T cells with a PIEZO1 inhibitor, or the method comprising the step of obtaining T cells with reduced expression of PIEZO1 after knocking down the PIEZO 1.
Preferably, the pretreatment refers to a step of pretreatment (cultivation) in a medium containing a PIEZO1 inhibitor.
More specifically, the PIEZO1 inhibitor may be contained in the medium in an amount of 0.01 to 10. Mu.M or more, for example, 1. Mu.M, 2. Mu.M, 3. Mu.M, 4. Mu.M, 5. Mu.M, 6. Mu.M, 7. Mu.M, 8. Mu.M, 9. Mu.M, 10. Mu.M.
Preferably, the method of knocking down the PIEZO1 includes RNA interference technology (RNAi), antisense oligonucleotide (ASO) technology, CRISPR technology, TALEN technology, ZFN technology, cre-loxP gene recombination technology.
Preferably, the reduced amount of PIEZO1 expression may comprise a reduction in the amount of PIEZO1 expression of at least 1% -100%, e.g. 1%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100% relative to a control. The control includes the median expression level of the biomarker in an individual without the disease or disorder, or an internal control (e.g., housekeeping gene, housekeeping gene), or a sample from one patient group/population.
Preferably, the method is performed in vitro.
Preferably, the method is non-therapeutic.
In particular, the T cells with high killing capacity have the advantages of enhanced traction force, enhanced cytotoxicity and enhanced tumor infiltration degree relative to control T cells, and have better treatment effect in treating cancers.
Meanwhile, the invention provides the T cell prepared by the method.
Preferably, the T cells are soluble in a suitable solution to maintain cell activity.
Meanwhile, the invention also provides application of the T cells prepared by the method in the treatment of cancer and the preparation of cancer drugs.
Therapeutic method
In another aspect, the invention provides a method of treating cancer comprising administering a PIEZO1 inhibitor or T cells prepared by the foregoing method to a patient.
Preferably, the methods of treatment may also be used in combination with other methods of treatment of cancer.
Preferably, the treatment methods of the other cancers include surgical therapy, chemotherapy, radiation therapy, gene therapy, immunotherapy, and the like.
Specifically, the chemotherapy is used to administer to a patient a chemical (chemo drug) such as: thiotepa, cyclophosphamide (cyclophosphamide), difluoromethylornithine (DMFO), retinoic acid (retinoic acid), piposulfan (piposulfan), dolastatin (dolastatin), busulfan (busulfan), imperosulfan (imposulfan), chlorophosphonate (clodronate), camptothecin (camptothecin), bryostatin (bryostatin), capecitabine (capecitabine), carboplatin, procarbazine, plicomycin (plicomycin), gemcitabine (gemcitabine), novelt (novelbine), pharmaceutically acceptable salts, acids or derivatives of any of the foregoing.
Specifically, the immunotherapy (immunotherapy) is a therapeutic method for artificially enhancing or inhibiting the immune function of the body to treat a disease by indicating a lowered or enhanced immune state of the body. Immunotherapy of tumors aims at activating the human immune system, killing cancer cells and tumor tissues by means of autoimmune functions. Generally rely on the use of immune effector cells and molecules to target and destroy cancer cells.
In particular, the gene therapy is directed to the administration of therapeutic polynucleotides to a patient. Viral vectors for expressing polynucleotides are well known in the art and include eukaryotic expression systems such as adenovirus, adeno-associated virus, retrovirus, herpes virus, lentivirus, poxvirus (including vaccinia virus) and papillomavirus (including SV 40). Alternatively, the polynucleotide may be administered using a lipid-based carrier, such as a liposome.
In particular, the radiation therapy involves ionizing radiation of the tumor, effecting extensive damage to DNA, DNA precursors, replication and repair of DNA, assembly and maintenance of chromosomes. The ionizing radiation includes x-ray radiation, ultraviolet radiation, infrared radiation, gamma ray radiation, microwave radiation, or the like.
In particular, the surgical procedure includes resection, which refers to the removal, excision and/or destruction of all or a portion of cancerous tissue. Surgical treatments also include laser surgery, cryosurgery, electrosurgery, microscope-controlled surgery, and the like.
General concepts
The immune checkpoint (immune checkpoint) of the invention refers to a receptor for programmed death and a ligand thereof. Immune checkpoint blocking therapies based on apoptosis receptors and their ligands increase the aggressiveness of the host immune system to tumor cells by inhibiting the binding of apoptosis receptors and their ligands.
Preferably, the immune checkpoint (immune checkpoint) comprises PD1, CTLA4, VISTA, IDO, CD137, TIGIT, TIM3, BTLA, CD27L, CD40, LAG-3, CD270, GITR, SIRPalpha, or the like.
The immune checkpoint inhibitor (immune checkpoint inhibitors, ICIs) is an inhibitor targeting the immune checkpoint.
The term "inhibitor" as used herein refers to a substance that targets, reduces or inhibits at least one activity of a particular gene or protein of interest. In particular, the present invention is directed to active agents that target, reduce or inhibit PIEZO1 and immune checkpoints.
Preferably, the inhibitor comprises synthetic or naturally occurring.
Preferably, the inhibitor comprises an agent that reduces expression of a gene or protein of interest by: RNA interference technology (RNAi), antisense oligonucleotide (ASO) technology, CRISPR technology, TALEN technology, ZFN technology, cre-loxP gene recombination technology.
Preferably, the inhibitor further comprises a compound, an antibody, which specifically targets a gene or protein of interest.
Preferably, the PIEZO1 inhibitor comprises an agent that reduces expression of PIEZO1 by the following technique: RNA interference technology (RNAi), antisense oligonucleotide (ASO) technology, CRISPR technology, TALEN technology, ZFN technology, cre-loxP gene recombination technology.
Preferably, the PIEZO1 inhibitor further comprises a compound and an antibody which specifically target PIEZO 1.
Preferably, the PIEZO1 inhibitor comprises GsMTx4, D-GsMTx4, gsMTx4 TFA.
Most preferably, the PIEZO1 inhibitor is GsMTx4.
Preferably, the PIEZO1 inhibitor is a specific PIEZO1 targeting agent used in RNA interference technology; more specifically, shRNA as used in the embodiments of the present invention.
Preferably, the PD1 inhibitor is an antibody that targets PD 1.
The antibodies of the invention include those polyclonal or monoclonal antibodies which are intact or shortened antibodies (e.g., F (ab ') 2 (Fab', fab or Fv fragments) antibodies, chimeric, humanized or fully humanized antibodies.
Cancers of the invention include, but are not limited to, cervical cancer, seminoma, testicular lymphoma, prostate cancer, ovarian cancer, lung cancer (e.g., small cell lung cancer SCLC, non-small cell lung cancer NSCLC, lung adenocarcinoma), rectal cancer, breast cancer, cutaneous squamous cell carcinoma, colon cancer, liver cancer, pancreatic cancer, esophageal cancer, thyroid cancer, transitional bladder epithelial cancer, leukemia (e.g., acute lymphoblastic leukemia ALL, acute myelogenous leukemia AML, chronic myelogenous leukemia CML, chronic lymphocytic leukemia CLL), brain tumor, stomach cancer, peritoneal cancer, head and neck cancer, endometrial cancer, kidney cancer, female genital tract cancer, carcinoma in situ, neurofibromas, bone cancer, skin cancer, gastrointestinal stromal tumors, mast cell tumors, multiple myeloma, melanoma, glioma, mesothelioma, neuroendocrine tumors (e.g., pancreatic neuroendocrine tumors or carcinomas, gastric neuroendocrine tumors or carcinomas, intestinal neuroendocrine tumors or carcinomas).
More specifically, the cancers of the present invention include cancers of the head, neck, eye, mouth, throat, esophagus, trachea, throat, pharynx, chest, bone, lung, colon, rectum, stomach, prostate, bladder, uterus, cervix, breast, ovary, testes, skin, thyroid, blood, lymph node, kidney, liver, pancreas, brain or central nervous system, pleura, peritoneum, neuroendocrine system.
Preferably, the cancer is melanoma, colon cancer, gastric cancer, liver cancer.
Most preferably, the specific embodiment of the invention is verified for melanoma and colon cancer, and proves that the T cells treated by the PIEZO1 inhibitor have higher treatment effect on the melanoma and the colon cancer, and the treatment effect can be further improved when the PD1 inhibitor is used in combination.
The term "subject" as used herein refers to any animal (e.g., mammal), including but not limited to humans, non-human primates, rodents, etc., that will become the recipient of a particular treatment. In general, the terms "subject" and "patient" are used interchangeably herein when referring to a human subject.
In certain embodiments, the subject (preferably a human) has, or is suspected of having, a cancer or an autoimmune disease.
By "treating" as used herein is meant alleviating or alleviating at least one symptom associated with such a condition, or slowing or reversing the progression of such a condition. Within the meaning of the present invention, the term "treatment" also means inhibiting, delaying the onset of the disorder (i.e. the period prior to the clinical manifestation of the disease) and/or reducing the risk of developing or worsening the disease. For example, the term "treatment" in connection with cancer may refer to eliminating or reducing tumor burden in a patient, or preventing, delaying or inhibiting metastasis, etc.
The T cells of the invention include autologous T cells or allogeneic T cells. Preferably, the autologous T cells are derived from human umbilical cord blood or peripheral blood. Preferably, the T cells are also mature commercial cell line products. In particular, the T cells may also be genetically engineered to express exogenous proteins, such as TCR-T cells or CAR-T cells.
Preferably, the T cells comprise CD8 + T、CD4 + T, TCR-T or CAR-T cells; more preferably, the T cell is CD8 + T cells.
Preferably, the T cells are prepared for administration as an injection, including any site injection, e.g., intradermal, subcutaneous, intramuscular, intravenous.
The cytotoxic T lymphocytes (CTL cells) of the invention are commonly referred to as CD8 + T cells, a key component of the adaptive immune system, play an important role in the immune system against pathogens such as viruses, bacteria and tumors.
In summary, the scheme of the application has the following effects:
1. the novel immune check point mediated by PIEZO1 of the application clarifies a novel mechanism of participation of T cells in tumor immune killing, reasonably explains novel types of T cell immune check points except biochemical signals, and has good clinical application prospect.
2. When PIEZO1 inhibitor GsMT×4 is added into T cells or PIEZO1 is knocked down, cytotoxicity of the T cells can be enhanced, and tumor immune killing infiltration function is enhanced, so that tumor growth is inhibited.
3. The novel immune checkpoint PIEZO1 and the downstream path thereof open up an innovative strategy of tumor immunotherapy, target the PIEZO1 mechanical channel path, increase the traction of T cells by inhibiting the PIEZO1 mechanical channel path, and enhance the immune killing capability; meanwhile, the GsMT multiplied 4 and alpha PD-1 combined drug can greatly improve the T cell tumor recognition and killing function, and develop ideas for the clinical targeted drug development.
Drawings
FIGS. 1A-B are graphs showing the results of cytotoxicity of T cells against B16F10 cells and MC38 cells after treatment of T cells with the PIEZO1 inhibitor GsMT X4 and knocking down of PIEZO1, A being B16F10 cells and B being MC38 cells.
FIG. 2 is a graph showing the results of cell traction measurements after T cells were treated with the PIEZO1 inhibitor GsMT×4.
FIG. 3 is a graph showing the results of detection of T cell tumor infiltration levels in mice treated with the PIEZO1 inhibitor GsMT×4 after adoptive treatment of B16F10 tumor mice.
FIGS. 4A-B are graphs showing the results of detection of tumor volume and survival of mice after adoptive treatment of mice with T cells after PIEZO1 inhibition using GsMT X4.
Fig. 5 is a graph of the results of detection of tumor volume in mice after adoptive treatment of mice with T cells knocked down with PIEZO 1.
FIGS. 6A-B are graphs of results of detection of tumor volume and survival of mice after treatment of mice with GsMT x4 and/or αPD-1 treated T cells.
Detailed Description
The present invention is further described in terms of the following examples, which are given by way of illustration only, and not by way of limitation, of the present invention, and any person skilled in the art may make any modifications to the equivalent examples using the teachings disclosed above. Any simple modification or equivalent variation of the following embodiments according to the technical substance of the present invention falls within the scope of the present invention.
Various cell lines, drugs and experimental animals used in the following examples:
B16F10 murine melanoma cell line and MC38 murine colon cancer cell line were purchased from ATCC;
GxMT x4 is available from Selleck corporation;
c57BL/6J mice were purchased from the medical laboratory animal center of the national academy of medical science;
CD45.1 + OT-1TCR mice were derived from OT-1 transgenic mice and CD45.1 + Breeding mice;
the OVA-B16 tumor mice are molded by a laboratory;
alpha PD-1 was purchased from Bio X Cell company.
Example one, inhibition and knock-down of PIEZO1 followed by enhanced T cytotoxicity
1. Experimental procedure
We will pre-treat pmel 1TCR transgenic CD8 with GsMT×4 and Yoda1 (GsMT×4-1. Mu.M and Yoda 1-2. Mu.M, inhibiting or activating PIEZO 1) for 24 hours, respectively + T-cell (CD 8) + T cells were cultured in IMDM medium containing 10% fetal bovine serum, 50. Mu.M beta. -mercaptoethanol and 50U/ml IL-2) and B16F10 cells or MC38 cells were co-cultured at a ratio of 20:1 for 4 hours.
Meanwhile, aiming at target points of shRNAs shown in SEQ ID NO.1 and SEQ ID NO.4, sense and anti-sense sequences are synthesized, a Piezo1-shRNAs lentiviral vector is constructed, T cells are transduced to obtain Piezo1-shRNAs Pmel-1T cells, and Piezo1-shRNAs Pmel-1T cells and B16F10 cells are co-cultured, and apoptosis of CD 45-tumor cells is detected by using a flow cytometry.
2. Experimental results
Inhibition or knockdown of the level of PIEZO1 in the B16F10 melanoma cell line enhanced T cell cytotoxicity, thereby promoting tumor cell apoptosis (fig. 1A), with the same results obtained in MC38 murine colon cancer cells (fig. 1B).
Example two T cell traction enhancement after PIEZO1 inhibition
1. Experimental procedure
The glass bottom of the confocal dish was pretreated with APTES (Sigma-Aldrich), after polymerization, the gel surface was covered with fluorescent bead solution for 15 min for immobilization, the gel was activated with sulfanilamide-sanpah, polylysine (PLL) was coated overnight at 4℃and then cells were incubated on PAA gel with 5. Mu.g/mL of anti-CD 3. Epsilon. Antibody for 60min at 37℃to ensure activation. Taking fluorescent images of cell phase contrast and fluorescent beads, and measuring displacement fields generated in the T cell activation process.
2. Experimental results
PIEZO1 inhibitor GsMT×4 pretreated mouse CD8 prepared in example 1 + T cells significantly enhanced traction in either resting or activated states (fig. 2).
Example three, gsMT×4 increases T cell tumor infiltration
1. The experimental steps are as follows:
from CD45.1 + Obtaining activated CD8 in OT-1TCR mice + T cells (methods of acquisition see Nat immunol 2021Mar;22 (3): 358-369.) were pretreated with GsMT×4 (1. Mu.M) for 24 hours and adoptively transferred by tail vein injection into C57BJ/6L mice carrying OVA-B16 tumors. CD45.1 + T cell flow assay for tumor infiltration.
2. Experimental results:
on receiving GsMT x4 pretreatment CD45.1 + Obtaining activated CD8 in OT-1TCR mice + Flow analysis of tumor CD45.1 in T cell treated C57BJ/6L mice + T cell infiltration was significantly increased (figure 3).
Example IV GsMT×4 inhibits T cells after PIEZO1, inhibits tumor growth in mice and prolongs survival of mice
1. The experimental steps are as follows:
OT-1CD8 pretreated with GsMT X4 (1. Mu.M) for 24 hours + Adoptive transfer of T cells into OVA-B16 tumor mice, recording and observing tumor growth conditions of mice, and recording miceLife time.
2. Experimental results:
gsmt×4 significantly delayed the growth of the mouse B16F10 tumor (fig. 4A) and prolonged the survival of the mice (fig. 4B).
Example five, T cells after PIEZO1 knockdown have an inhibitory effect on mouse tumor growth
1. The experimental steps are as follows:
at OT-1CD8 + And constructing a PIEZO1-shRNA stable transfer cell strain in the T cells, transferring the PIEZO1-shRNA stable transfer cell strain into an OVA-B16 tumor mouse body in a adoptive manner, and recording and observing the tumor growth condition of the mouse.
2. Experimental results:
OT-1CD8 knocked down PIEZO1 + T cells had a significant inhibitory effect on the growth of the mouse B16F10 tumor (FIG. 5).
The combination of GsMT multiplied by 4 and alpha PD-1 can better promote the inhibition function of T cells on tumor growth.
1. The experimental steps are as follows:
for OT-1CD8 + After 24 hours of stimulation with or without GsMT x4 (1. Mu.M), these T cells were injected into mice via the tail vein, and some mice were also intraperitoneally administered with anti-PD-1 antibodies (200. Mu.g per mouse, once every three days, three total administrations), and tumor growth was noted.
2. Experimental results:
OT-1CD8 treated with GsMT X4 in combination with alpha PD-1 compared to the control group + T cells had significant inhibition of OVA-B16 tumors in mice (FIG. 6A), and survival of mice treated with the combination was significantly prolonged (FIG. 6B).

Claims (10)

  1. Use of a piezo1 inhibitor for the preparation of:
    1) A product for the treatment of cancer, which comprises,
    2) A product for improving the killing capacity of T cells,
    3) A product that enhances the therapeutic effect of an immune checkpoint inhibitor;
    preferably, the immune checkpoint comprises PD1, CTLA4, VISTA, IDO, CD137, TIGIT, TIM3, BTLA, CD27L, CD, LAG-3, CD270, GITR, sirpa;
    preferably, the immune checkpoint is PD1;
    preferably, the inhibitor of PD1 is an antibody that targets PD1;
    preferably, the antibodies include monoclonal antibodies, single chain antibodies, chimeric antibodies, multispecific antibodies, humanized antibodies, and fully human antibodies;
    preferably, the T cells comprise CD8 + T、CD4 + T, TCR-T or CAR-T cells;
    preferably, the T cell is CD8 + T。
  2. 2. The use of claim 1, the PIEZO1 inhibitor comprising an agent that reduces expression of PIEZO1 by: RNA interference technology, antisense oligonucleotide technology, CRISPR technology, TALEN technology, ZFN technology, cre-loxP gene recombination technology;
    preferably, the PIEZO1 inhibitor is an agent used in RNA interference technology;
    preferably, the PIEZO1 inhibitor further comprises a compound, antibody that specifically targets PIEZO 1;
    preferably, the PIEZO1 inhibitor comprises GsMTx4, D-GsMTx4, gsMTx4 TFA;
    most preferably, the PIEZO1 inhibitor is GsMTx4;
    preferably, the antibodies include monoclonal antibodies, single chain antibodies, chimeric antibodies, multispecific antibodies, humanized antibodies.
  3. 3. The use of claim 1, wherein the cancer comprises cancer of the head, neck, eye, mouth, larynx, esophagus, trachea, larynx, pharynx, chest, bone, lung, colon, rectum, stomach, prostate, bladder, uterus, cervix, breast, ovary, testis, skin, thyroid, blood, lymph node, kidney, liver, pancreas, brain or central nervous system, pleura, peritoneum, neuroendocrine system.
  4. 4. The use of claim 1, wherein the cancer comprises melanoma, myeloma, thymoma, sarcoma, lung cancer, liver cancer, non-hodgkin's lymphoma, skin cancer, uterine cancer, breast cancer, pancreatic cancer, colon cancer, anal cancer, renal cancer, bladder cancer, prostate cancer, ovarian cancer, brain cancer, vascular endothelial tumor, head and neck cancer, thyroid cancer, testicular cancer, gastrointestinal cancer, pleural mesothelioma, peritoneal mesothelioma, neuroendocrine tumor or cancer;
    preferably, the cancer is melanoma, colon cancer, gastric cancer, liver cancer.
  5. 5. A pharmaceutical combination composition for treating cancer, the pharmaceutical combination composition comprising a PIEZO1 inhibitor and an immune checkpoint inhibitor;
    preferably, the dosage forms of the PIEZO1 inhibitor and the immune checkpoint inhibitor in the pharmaceutical combination composition are the same or different;
    preferably, the PIEZO1 inhibitor and immune checkpoint inhibitor in the pharmaceutical combination composition are administered simultaneously or sequentially;
    preferably, the mode of administration includes oral, spray inhalation, rectal, nasal, buccal, subcutaneous, intravenous, intramuscular, intraperitoneal, intrathecal, endocardial or intrasternal administration;
    preferably, the dosage ratio of the PIEZO1 inhibitor and the immune checkpoint inhibitor in the pharmaceutical combination composition is 1:0.1-2;
    preferably, the immune checkpoint is PD1;
    preferably, the immune checkpoint inhibitor is an antibody targeting PD 1.
  6. 6. The pharmaceutical combination composition of claim 5, further comprising a pharmaceutically acceptable carrier, diluent or excipient;
    preferably, the pharmaceutically acceptable carrier, diluent or excipient comprises a glidant, sweetener, diluent, preservative, dye, colorant, flavoring agent, surfactant, wetting agent, dispersing agent, suspending agent, stabilizer, isotonic agent, surfactant or emulsifier.
  7. 7. A method for preparing T cells with high killing ability, the method comprising a step of pretreating T cells with a PIEZO1 inhibitor, or the method comprising a step of obtaining T cells with reduced expression of PIEZO1 after knocking down PIEZO 1;
    preferably, the pretreatment refers to a step of pretreatment in a medium containing a PIEZO1 inhibitor;
    more specifically, the content of the PIEZO1 inhibitor in the medium may be 0.01 to 10. Mu.M or more;
    preferably, the method for knocking down the PIEZO1 comprises RNA interference technology, antisense oligonucleotide technology, CRISPR technology, TALEN technology, ZFN technology and Cre-loxP gene recombination technology;
    preferably, the PIEZO1 inhibitor comprises an agent that reduces expression of PIEZO1 by the following technique: RNA interference technology, antisense oligonucleotide technology, CRISPR technology, TALEN technology, ZFN technology, cre-loxP gene recombination technology;
    preferably, the PIEZO1 inhibitor is an agent used in RNA interference technology;
    preferably, the PIEZO1 inhibitor further comprises a compound, antibody that specifically targets PIEZO 1;
    preferably, the PIEZO1 inhibitor comprises GsMTx4, D-GsMTx4, gsMTx4 TFA;
    most preferably, the pretreatment is treatment of T cells with 1 μm GsMTx4;
    preferably, the antibody comprises a monoclonal antibody, a single chain antibody, a chimeric antibody, a multispecific antibody, a humanized antibody;
    preferably, the method is performed in vitro;
    preferably, the method is non-therapeutic.
  8. 8. The method of claim 9, wherein the T cells comprise autologous T cells or allogeneic T cells;
    preferably, the autologous T cells are derived from human umbilical cord blood or peripheral blood;
    preferably, the T cells may also be mature commercial cell line products;
    preferably, the T cells comprise CD8 + T、CD4 + T, TCR T or CAR T cells;
    preferably, the T cell is CD8 + T cells.
  9. 9. A cell prepared by the method of claim 7;
    preferably, the T cells are soluble in a suitable solution to maintain cellular activity;
    specifically, the T cells have any one or more characteristics of traction enhancement, cytotoxicity enhancement and tumor infiltration degree enhancement relative to common T cells;
    preferably, the T cells comprise CD8 + T、CD4 + T, TCR T or CAR T cells;
    preferably, the T cell is CD8 + T cells.
  10. 10. Use of the cell of claim 9 for the preparation of a medicament for cancer;
    preferably, the cancer comprises a cancer of the head, neck, eye, mouth, throat, esophagus, trachea, larynx, pharynx, chest, bone, lung, colon, rectum, stomach, prostate, bladder, uterus, cervix, breast, ovary, testis, skin, thyroid, blood, lymph node, kidney, liver, pancreas, brain or central nervous system, pleura, peritoneum, neuroendocrine system;
    preferably, the cancer comprises melanoma, myeloma, thymoma, sarcoma, lung cancer, liver cancer, non-hodgkin lymphoma, skin cancer, uterine cancer, breast cancer, pancreatic cancer, colon cancer, anal cancer, kidney cancer, bladder cancer, prostate cancer, ovarian cancer, brain cancer, vascular endothelial tumor, head and neck cancer, thyroid cancer, testicular cancer, gastrointestinal cancer, pleural mesothelioma, peritoneal mesothelioma, neuroendocrine tumor or cancer;
    preferably, the cancer is melanoma, colon cancer, stomach cancer, liver cancer;
    preferably, the T cells comprise CD8 + T、CD4 + T, TCR T or CAR T cells;
    preferably, the T cell is CD8 + T cells.
CN202311680205.4A 2023-12-08 2023-12-08 Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment Pending CN117653733A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202311680205.4A CN117653733A (en) 2023-12-08 2023-12-08 Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202311680205.4A CN117653733A (en) 2023-12-08 2023-12-08 Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment

Publications (1)

Publication Number Publication Date
CN117653733A true CN117653733A (en) 2024-03-08

Family

ID=90065816

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202311680205.4A Pending CN117653733A (en) 2023-12-08 2023-12-08 Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment

Country Status (1)

Country Link
CN (1) CN117653733A (en)

Similar Documents

Publication Publication Date Title
JP2021176863A (en) Methods for enhancing potency of immune cells
US11331345B2 (en) PD-1 CAR NK-92 cell and preparation method and use thereof
CN106573981A (en) Combination therapies with anti CD40 antibodies
Wu et al. HER2-specific chimeric antigen receptor-engineered natural killer cells combined with apatinib for the treatment of gastric cancer
CN113663076B (en) Cancer combination therapy composition
Tabana et al. Tackling immune targets for breast cancer: Beyond PD-1/PD-L1 Axis
JP2024041920A (en) New uses of phenothiazines or compounds with similar structures in pharmaceuticals
CN109937051A (en) Treat the raised method of TIM-3
US20190160099A1 (en) Pharmaceutical composition and use thereof
CN117653733A (en) Application of novel immune checkpoint PIEZO1 pathway in tumor T cell treatment
CN112190718B (en) Gene transfer vector and preparation method thereof, and anti-tumor drug and preparation method thereof
CN111419832B (en) Pharmaceutical composition and application thereof in preparation of tumor treatment drugs
CN107858352A (en) A kind of shRNA cluster sequences and its expression vector and purposes
US20220378879A1 (en) Application of peg interferon and protooncogene product targeting inhibitor in synergistic treatment of renal carcinoma
Esmaeilzadeh et al. Chimeric Antigen Receptor (CAR) T cell immunotherapy for solid tumors
CN116063556B (en) IPS-derived CAR-NK cells and their use for the treatment of cancer
Mangal et al. Engineering Metabolism of Chimeric Antigen Receptor (CAR) Cells for Developing Efficient Immunotherapies. Cancers 2021, 13, 1123
CN115925974B (en) Preparation method of universal IPS-derived CAR-NK cells for solid tumors
CN117143254B (en) Chimeric Antigen Receptor (CAR) and application thereof in anticancer
CN115944650B (en) Application of tumor infiltration cells in preparation of antitumor drugs and model construction method
CN113117088B (en) Use of inhibitors of calcium activated chloride channels in tumor immunotherapy
WO2022268162A1 (en) Method and composition for treating tumors
US20220305100A1 (en) Methods of vaccination and use of cd47 blockade
Dhar et al. Neoangiogenesis and immune-regulation: Two armour of VEGF in the tumor microenvironment
US20230102924A1 (en) Methods of Treating Cancer Using Checkpoint Inhibitors in Combination with Purine Cleaving Enzymes

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination