CN117045801A - Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors - Google Patents

Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors Download PDF

Info

Publication number
CN117045801A
CN117045801A CN202310516010.XA CN202310516010A CN117045801A CN 117045801 A CN117045801 A CN 117045801A CN 202310516010 A CN202310516010 A CN 202310516010A CN 117045801 A CN117045801 A CN 117045801A
Authority
CN
China
Prior art keywords
cancer
cells
inhibitor
tumor
stm2457
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
CN202310516010.XA
Other languages
Chinese (zh)
Inventor
陈捷凯
孔祥谦
吴凯昕
吴红玲
汤同柯
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.)
Guangzhou Institute of Biomedicine and Health of CAS
Original Assignee
Guangzhou Institute of Biomedicine and Health of CAS
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 Guangzhou Institute of Biomedicine and Health of CAS filed Critical Guangzhou Institute of Biomedicine and Health of CAS
Publication of CN117045801A publication Critical patent/CN117045801A/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene or sparfloxacin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/15Cells of the myeloid line, e.g. granulocytes, basophils, eosinophils, neutrophils, leucocytes, monocytes, macrophages or mast cells; Myeloid precursor cells; Antigen-presenting cells, e.g. dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K35/00Medicinal preparations containing materials or reaction products thereof with undetermined constitution
    • A61K35/12Materials from mammals; Compositions comprising non-specified tissues or cells; Compositions comprising non-embryonic stem cells; Genetically modified cells
    • A61K35/14Blood; Artificial blood
    • A61K35/17Lymphocytes; B-cells; T-cells; Natural killer cells; Interferon-activated or cytokine-activated lymphocytes
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/3955Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against proteinaceous materials, e.g. enzymes, hormones, lymphokines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Immunology (AREA)
  • Hematology (AREA)
  • Organic Chemistry (AREA)
  • Cell Biology (AREA)
  • Engineering & Computer Science (AREA)
  • Biotechnology (AREA)
  • Biomedical Technology (AREA)
  • Virology (AREA)
  • Zoology (AREA)
  • Developmental Biology & Embryology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Oncology (AREA)
  • Endocrinology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The invention belongs to the field of biological medicine, and relates to a m6ARNA methylase inhibitor and immune checkpoint inhibitor combined treatment of tumors. The combined treatment method provided by the invention is to combine a tumor immunotherapeutic agent and STM2457 or derivatives thereof to treat tumors including melanoma and ovarian cancer. The novel anti-tumor drug combination therapy found by the invention has great significance for drug development and clinical treatment of melanoma or ovarian cancer.

Description

Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors
Technical Field
The invention belongs to the field of biological medicine, and relates to a m6A RNA (ribonucleic acid) methylase inhibitor and immune checkpoint inhibitor combined treatment for tumors.
Background
To date, more than 170 post-transcriptional RNA modifications have been found in humans. Among them, N6 methyl adenosine (m 6A) is the most common modification in eukaryotic messenger RNAs (mrnas). m6A modified proteins include METTL3, METTL14, YTHDC1, YTHDF2, and the like. m6A modification plays a key role in regulating RNA processing, splicing, nucleation, translation and stability, which is critical for the development of various human diseases such as cancer. m6A modification is abnormal in various types of cancers and is correlated with patient prognosis. Abnormalities in m6A modification are closely related to the occurrence, development and resistance of malignant tumors. As an oncogene, the m6A modified protein promotes tumor progression by up-regulating oncogenes or down-regulating tumor suppressor genes.
Programmed cell death protein 1 (PD-1) plays an important role in suppressing immune responses and promoting self-tolerance, including modulating T cell activity, activating antigen-specific T cell apoptosis programs and inhibiting apoptosis of conventional T cells. PD-L1 is a transmembrane protein, is considered as a co-inhibitor of immune response, and can bind to PD-1 to reduce proliferation of PD-1 positive cells, inhibit cytokine secretion thereof and induce apoptosis. PD-L1 also plays an important role in the progression of various malignant tumors, inhibiting the recognition and killing of tumor cells by the immune system, and promoting tumor immune escape. Blocking the PD-1/PD-L1 interaction by monoclonal antibodies has had a tremendous impact on cancer treatment.
Melanoma is a malignant tumor that originates from melanocytes. As an invasive skin cancer, the incidence of melanoma is increasing worldwide. Many studies have investigated the relationship of tumor cells to tumor microenvironment. However, more than half of melanoma patients do not respond to immune checkpoint inhibitor treatment, which is closely related to the immunosuppressive microenvironment in which the tumor cells are located. Through the combination with other medicines, the tumor infiltration CD8+ T cell level and the CD8/Treg ratio in the tumor microenvironment are improved, the immune 'cold' tumor is changed into the 'hot' tumor, and the effective rate of tumor immunotherapy is improved, so that the method has important clinical significance.
Disclosure of Invention
In some embodiments, the present invention provides a composition comprising: an anti-tumor immunotherapeutic agent and STM2457 or a derivative thereof.
In some embodiments, the anti-tumor immunotherapeutic agent comprises an immune cell or one or more immune checkpoint inhibitors.
In some embodiments, the immune cell is selected from a T cell, macrophage and/or monocyte or T cell comprising an artificial T cell receptor.
In some embodiments, the T cells are selected from one or more of Natural Killer (NK) cells, cytotoxic T Lymphocytes (CTLs), and regulatory T cells.
In some embodiments, the T cell is selected from a T lymphocyte.
In some embodiments, the T cells are selected from T lymphocytes from cord blood sources.
In some embodiments, the immune checkpoint inhibitor comprises a polypeptide, a monoclonal antibody, or a chemically synthesized small molecule inhibitor.
In some embodiments, the immune checkpoint inhibitor comprises a PD-L1 inhibitor, a PD-1 inhibitor, and/or a CTLA-4 inhibitor.
In some embodiments, the PD-1 inhibitor is an antibody or antigen-binding fragment thereof.
In some embodiments, the PD-1 inhibitor is a PD-1 antibody.
In some embodiments, the anti-tumor immunotherapeutic agent comprises an antibody or antibody fragment that targets a tumor-specific antigen.
In some embodiments, the STM2457 has a structural formula shown in formula (I):
in some embodiments, the composition further comprises a pharmaceutically acceptable carrier, excipient, or stabilizer.
In some embodiments, the composition is an anti-tumor composition.
In some embodiments, the tumor comprises lung cancer (including squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer), gastrointestinal cancer, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma, multiple myeloma, salivary gland cancer, renal cell carcinoma, wilms' cell carcinoma, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, or a human single-cell leukemia cell.
In some embodiments, the inventors have discovered that PD-1 inhibitors (or PD-1 blockers) or T cells have very significant effects in combination with STM2457 in the treatment of melanoma and ovarian cancer, with the combination being far superior to the single administration.
In some embodiments, the combination will even produce a synergistic therapeutic effect beyond the expectations of those skilled in the art.
The combination of anti-CTLA-4 and anti-PD-1 therapies provides the potential for excellent efficacy, possibly due to each drug acting in a complementary or even synergistic manner. But combination therapy may result in greater toxicity. In previously untreated melanoma or recurrent small cell lung cancer patients, the incidence of drug-related grade 3-4 adverse events was 54% to 55%, while the incidence of blockade was 24% to 27%, whereas the incidence of ipp Li Shan antibody alone was 24% to 27%, and the incidence of Nivolumab alone was 15% to 16%. Therefore, the invention finds a new anti-tumor drug combination therapy, and has great significance for drug development and clinical treatment of melanoma.
In some embodiments, the present invention provides a kit comprising a first container, a second container, and a package insert; the first container comprises a medicament of an anti-tumor immunotherapeutic agent, the second container comprises a medicament of STM2457 or a derivative thereof, and the package insert comprises instructions for using the medicament to treat cancer in a subject.
In some embodiments, the anti-tumor immunotherapeutic agent comprises an immune cell or one or more immune checkpoint inhibitors.
In some embodiments, the immune cell is selected from a T cell, macrophage and/or monocyte or T cell comprising an artificial T cell receptor.
In some embodiments, the T cells are selected from one or more of Natural Killer (NK) cells, cytotoxic T Lymphocytes (CTLs), and regulatory T cells.
In some embodiments, the T cell is selected from a T lymphocyte.
In some embodiments, the T cells are selected from T lymphocytes from cord blood sources.
In some embodiments, the immune checkpoint inhibitor comprises a polypeptide, a monoclonal antibody, or a chemically synthesized small molecule inhibitor.
In some embodiments, the immune checkpoint inhibitor comprises a PD-L1 inhibitor, a PD-1 inhibitor, and/or a CTLA-4 inhibitor.
In some embodiments, the PD-1 inhibitor is an antibody or antigen-binding fragment thereof.
In some embodiments, the PD-1 inhibitor is a PD-1 antibody.
In some embodiments, the anti-tumor immunotherapeutic agent comprises an antibody or antibody fragment that targets a tumor-specific antigen.
In some embodiments, the STM2457 has a structural formula shown in formula (I):
in some embodiments, the invention provides the use of any of the compositions in the manufacture of a medicament for treating cancer.
In some embodiments, the cancer comprises lung cancer (including squamous cell carcinoma, small-cell lung cancer, non-small cell lung cancer), gastrointestinal cancer, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma, multiple myeloma, salivary gland cancer, renal cell carcinoma, wilms' cell carcinoma, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, or a human single-cell leukemia cell.
In some embodiments, the invention provides the use of STM2457 or a derivative thereof in the manufacture of a medicament for the treatment of melanoma or ovarian cancer.
In some embodiments, the STM2457 has a structural formula shown in formula (I):
drawings
The volume and mass measurements of the mouse tumors in the four sets of protocols shown in fig. 1 indicate that STM2457 and PD-1 antibodies used in combination effectively inhibit the growth of mouse melanoma. (fig. 1A) STM2457 and PD-1 antibody groups significantly reduced tumor weight relative to NC control groups in the four-group regimen. (fig. 1B) STM2457 and PD-1 antibody groups significantly reduced tumor volume relative to NC control groups in the four-group regimen.
The results of single cell CD45 flow analysis of mouse tumors in the four sets of protocols in fig. 2 indicate that the combination of STM2457 and PD-1 antibodies can effectively improve tumor microenvironment, i.e., increase the proportion of hematopoietic lineage cells in B16F0 tumors.
The result of the single cell sequencing data analysis of the tumor in FIG. 3 shows that the combination of STM2457 and PD-1 antibodies can effectively promote the mitosis of T cell population and the maintenance of the expression of related genes of stem cell population in B16F0 tumor.
FIG. 4 is a flow chart of the administration of STM2457 in combination with PD-1 antibodies.
Figure 5 shows the combination treatment of ovarian cancer with STM2457 and T cells. (FIG. 5A) A1847 cells (labeled red fluorescence) and T cells, and STM 2457. (fig. 5B) tumor cell death rate (%), STM2457 group vs nc+t group P <0.001 calculated for the number of a1847 cells detected by flow cytometry.
FIG. 6 is a comparison of the inhibition effect of the m6A methylase inhibitor STM2457 alone on different tumor cells.
Figure 7 shows the combination treatment of B16 melanoma with STM2457 and T cells. Tumor cell mortality (%) calculated for the number of B16 cells detected by flow cytometry, STM2457 group vs nc+t group P <0.001.
Detailed Description
The technical solution of the present invention is further illustrated by the following specific examples, which do not represent limitations on the scope of the present invention. Some insubstantial modifications and adaptations of the invention based on the inventive concept by others remain within the scope of the invention.
The beta-cyclodextrin solution used in the following examples was purchased from Aladin-e company (https:// www.aladdin-e.com /), under the designation H108813. The PD-1 antibody used was purchased from BioXcell company (https:// bXcell. Com) under the trade designation BE0146. The PBS buffer used was purchased from HyClone, cat# SH30028.02.
"comprising" or "including" is intended to mean that the compositions (e.g., media) and methods include the recited elements, but not exclude other elements. When used to define compositions and methods, "consisting essentially of … …" means excluding other elements that have any significance to the combination for the purpose. Thus, a composition consisting essentially of the elements defined herein does not exclude other materials or steps that do not materially affect the basic and novel characteristics of the claimed invention. "consisting of … …" means the process steps excluding trace elements and essential elements of other components. Embodiments defined by each of these transitional terms are within the scope of this invention.
The components of the "composition" herein may be present in a mixed form or may be packaged separately. The separately packaged components may also contain their respective adjuvants. The adjuvant refers to a means for assisting the curative effect of the medicine in pharmacy. In the case of separate packages for the components of the composition, the individual components of the separate packages may be administered simultaneously or in any order, wherein the patient is treated with one drug and then administered with the other. The patient refers to a mammalian subject, particularly a human.
The tumor immune therapy is a therapeutic method for controlling and eliminating tumor by restarting and maintaining tumor-immune circulation and restoring normal anti-tumor immune response of the body. "tumor immunotherapeutic" refers to a drug for "tumor immunotherapy" and includes monoclonal antibody immune checkpoint inhibitors, therapeutic antibodies, cancer vaccines, cell therapies, small molecule inhibitors, and the like.
Herein, STM refers to "STM2457".
Example 1
1. Drug configuration
(1) The preparation method of the m6A methylase inhibitor working solution comprises the following steps:
1) Dissolving beta-cyclodextrin in sterilized ultrapure water to obtain beta-cyclodextrin solution with the mass fraction of 20% (w/v), and filtering with a filter membrane with the mass fraction of 0.22 μm in a biosafety cabinet to ensure the sterility of the solution;
2) And (3) dissolving the STM2457 medicine in the beta-cyclodextrin solution in the step (1), and fully and uniformly shaking at 42 ℃ to form a homogeneous STM 2457-beta-cyclodextrin solution. STM2457 was found to have a final concentration of 6.25mg/mL.
(2) Immune checkpoint drug configuration method:
PD-1 antibodies were diluted to 2.5mg/mL using PBS buffer.
2. Evaluation of inhibition of growth of murine melanoma B16F0 Using STM2457 in combination with PD-1 antibody
Mouse melanoma B16F0 (or "B16") was inoculated into 6-8 week old C57BL/6J mice, each with 2X 10≡5 cells. After the tumor mass was reached, injections of different drug combinations (four groups, NC, STM2457, PD-1 antibody, STM2457+PD-1 antibody, respectively) were initiated and the mice were periodically examined for changes in body weight and tumor volume. The results showed that the STM2457+PD-1 antibody group had the most significant tumor growth inhibition effect (FIG. 1).
3. Evaluation of melanoma microenvironment changes Using STM2457 in combination with PD-1 antibodies
Taking tumor obtained by dissecting the above mice, cleaning with PBS, discarding necrotic tissue part, taking part of tumor into 1.5mL centrifuge tube, and cutting to 1mm 3 About the size of the pieces, 1.5mL of pre-warmed DMEM medium (containing 2mg/mL collagenase P,20ug/mL DNase I enzyme final concentration) was added. Digestion is carried out for 15min at 37 ℃, vibration is carried out, filtration is carried out by a 70um filter membrane, and supernatant is removed by centrifugation. 700ul of erythrocyte lysate is added into an EP tube, after 4 DEG lysis for 10min, equal volume of serum is added for termination, a single cell sample can be obtained, then FITC-CD45 antibody staining is carried out, and then flow cytometry analysis is carried out. The results show that the three groups of protocols (STM 2457, PD-1 antibody, STM2457+PD-1 antibody) increased the proportion of hematopoietic lineage cells compared to the NC control group, demonstrating that the combination of STM2457 and PD-1 antibodies effectively improves melanoma microenvironment (FIG. 2).
4. Evaluation of the Effect of STM2457 and PD-1 antibody combination on melanoma Global Gene expression
Taking the cell sample obtained in the step 3, and performing single cell transcriptome (scRNA-seq) analysis. Analysis of the data shows that the STM2457+PD-1 antibody set is effective in promoting T cell mitosis and stem cell population maintenance-related gene expression in the immune microenvironment in B16FO tumors relative to the PD-1 antibody set alone, indicating that STM2457 can enhance T cell activity and reduce T cell failure (FIG. 3).
Note that: the four groups in fig. 1-3 are:
(1) NC group (or nc+nc):
(2) Nc+pd1 group (or PD-1 antibody group):
(3) Stm+nc group (STM 2457 group):
(4) Stm+pd1 group (STM 2457& PD-1 antibody group):
STM injection at 50mg/kg, approximately 1.25 mg/mouse per mouse, once daily; the PD-1 antibody injection dose was 250 ug/dose, 2 times a week. The administration time was 2 weeks since the mice were subcutaneously vaccinated with the B16F0 tumor had a clear feeling of touch (after about 5-7 days). The flow chart is shown in fig. 4.
Example 2 evaluation of growth inhibition of ovarian cancer cells Using STM2457 in combination with T cells
At the cellular level, human ovarian cancer cells a1847 were treated with 1 μ M m6A methylase inhibitor STM2457 and then co-incubated with cord blood-derived T cells.
The method comprises the following specific steps: 1000A 1847 cells were seeded in 96-well plates, cultured for 3 days at a concentration of 1uM STM2457, cord blood-derived T cells of different combination schemes (wherein the number of T cells added was 1:1 in the ratio of the number of A1847 to the number of T cells) were added, four groups (NC; STM2457; NC+T; STM 2457+T) were added, and cells were harvested after 24 hours of incubation, and the number of A1847 cells was examined by flow cytometry to calculate the killing of tumor cells.
The results are shown in FIGS. 5A-5B. It can be seen that STM2457 in combination with T cells is effective in inhibiting the growth of ovarian cancer cells. Wherein, the T cell separation method of the umbilical cord blood source comprises the following steps:
separating mononuclear cells from umbilical cord blood by density gradient centrifugation, specifically, (1) diluting umbilical cord blood with PBS for 2 times, and centrifuging 750g in a clean sterile collecting tube at 25deg.C for 30min; (2) Transferring the MNC of the white middle layer into a new 50mL centrifuge tube; (3) centrifugation at 350g per tube +30mLPBS at 4deg.C for 10min; (4) Removing the supernatant, adding 5mL of erythrocyte lysate, incubating for 10min at 25 ℃, filling PBS (phosphate buffered saline) to dilute the erythrocyte lysate, and centrifuging for 10min at 4 ℃ at 300 g; (5) The cell pellet was resuspended in 25mLPBS, centrifuged at 300g for 10min at 4℃and repeated once more; (6) The cells were resuspended in PBS and counted to obtain cells as mononuclear cells. (7) Biotin-conjugated antibodies (CD 14, CD15, CD16, CD19, CD34, CD36, CD56, CD123, CD235 a) were added to the resuspended mononuclear cells for 5 minutes, followed by incubation with anti-biotin beads for 10 minutes, and the magnetically labeled cells were removed by magnetic column, thereby isolating high purity T lymphocytes.
Example 3 inhibition of different tumors by the m6A methylase inhibitor STM2457 alone
At the cellular level, murine melanoma cells B16 were treated with the 5 μ M m6A methylase inhibitor STM2457, respectively; human breast cancer cells MDA-MB-231 and MCF7; human mononuclear leukemia cells Thp1; mouse lung cancer cell LLC; human melanoma cells a375; human lung cancer cells H1299, H23, H460, H522 were treated.
The method comprises the following steps: 1000 cells were seeded in 96-well plates, treated with 5. Mu.M STM2457 for 4 days, incubated in an incubator for 1h after 10ul of CCK8 (Biyun days) was added to each well, and absorbance at 450nm was measured with an ELISA reader.
The results are shown in FIG. 6. THP-1 and MDA-MB-231 cells are sensitive to STM2457, and MCF7, LLC and H460 have about 20% inhibition effect, and the rest cancer cells are not sensitive.
EXAMPLE 4 evaluation of B16 melanoma cell growth inhibition Using STM2457 in combination with T cells
At the cellular level, murine melanoma cells B16 were treated with 1 μ M m6A methylase inhibitor STM2457 and then co-incubated with T cells from the spleen of the mice.
The method comprises the following specific steps: 1000B 16 cells are planted in a 96-well plate, after treatment culture for 3 days at the concentration of 1 mu M STM2457, spleen-derived T cells of mice with different combination schemes (wherein the number of the added T cells is added according to the ratio of the number of the B16 to the number of the T cells of 1:2) are added, four groups (NC, STM2457, NC+T and STM2457+T respectively) are added, the cells are collected after incubation for 24 hours, the number of the B16 cells is detected by using a flow cytometry, and the killing condition of tumor cells is calculated.
The results are shown in FIG. 7. It can be seen that STM2457 in combination with T cells is effective in inhibiting the growth of melanoma cells.
The method for separating the T cells from the spleen of the mouse comprises the following steps:
the spleen of the dissected mice is soaked in PBS, the spleen is reversely ground by a 1mL syringe, 1mL is filtered by a filter screen. Centrifuging 750g for 5min at 25 ℃ in a filtrate collecting pipe; (2) Removing the supernatant, adding 5mL of erythrocyte lysate, incubating for 10min at 25 ℃, filling PBS (phosphate buffered saline) to dilute the erythrocyte lysate, and centrifuging for 10min at 4 ℃ at 300 g; (5) The cell pellet was resuspended in 25mLPBS, centrifuged at 300g for 10min at 4℃and repeated once more; (6) The cells were resuspended in PBS and counted to obtain cells as mononuclear cells. (7) Biotin-conjugated antibodies (CD 11B, CD11c, CD19, CD45R (B220), CD49B (DX 5), CD105, MHC class II and Ter-119) were added to the resuspended mononuclear cells and incubated for 5min, followed by incubation with anti-biotin beads for 10min, and magnetically labeled cells were removed by magnetic column to isolate high purity T lymphocytes.

Claims (10)

1. A composition, wherein the composition comprises: an anti-tumor immunotherapeutic agent and STM2457 or a derivative thereof.
2. The composition of claim 1, wherein the anti-tumor immunotherapeutic agent comprises an immune cell or one or more immune checkpoint inhibitors.
Preferably, the immune cells are selected from T cells, macrophages and/or monocytes or T cells comprising artificial T cell receptors;
preferably, the T cells are selected from one or more of Natural Killer (NK) cells, cytotoxic T Lymphocytes (CTLs), and regulatory T cells;
preferably, the T cells are selected from T lymphocytes.
3. The composition of any one of claims 1-2, wherein the immune checkpoint inhibitor comprises a polypeptide, a monoclonal antibody, or a chemically synthesized small molecule inhibitor;
preferably, the immune checkpoint inhibitor comprises a PD-L1 inhibitor, a PD-1 inhibitor and/or a CTLA-4 inhibitor;
preferably, the PD-1 inhibitor is an antibody or antigen-binding fragment thereof;
preferably, the PD-1 inhibitor is a PD-1 antibody.
4. The composition of any one of claims 1-2, wherein the anti-tumor immunotherapeutic agent comprises an antibody or antibody fragment that targets a tumor-specific antigen.
5. A composition according to any one of claims 1 to 2, wherein the STM2457 has the structural formula shown in formula (I):
6. the composition of any one of claims 1-2, further comprising a pharmaceutically acceptable carrier, excipient, or stabilizer;
preferably, the composition is an anti-tumor composition;
preferably, the tumor comprises lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma, multiple myeloma, salivary gland cancer, renal cell carcinoma, nephroblastoma, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, or a human mononuclear leukemia cell;
preferably, the cancer comprises ovarian cancer or melanoma.
7. A kit comprising a first container, a second container, and a package insert; the first container comprises a medicament of an anti-tumor immunotherapeutic agent, the second container comprises a medicament of STM2457 or a derivative thereof, and the package insert comprises instructions for using the medicament to treat cancer in a subject.
8. The kit of claim 7, wherein the anti-tumor immunotherapeutic agent comprises an immune cell or one or more immune checkpoint inhibitors;
preferably, the immune cells are selected from T cells, macrophages and/or monocytes or T cells comprising artificial T cell receptors;
preferably, the T cells are selected from one or more of Natural Killer (NK) cells, cytotoxic T Lymphocytes (CTLs), and regulatory T cells;
preferably, the T cells are selected from T lymphocytes.
Preferably, the immune checkpoint inhibitor comprises a polypeptide, monoclonal antibody, or a chemically synthesized small molecule inhibitor;
preferably, the immune checkpoint inhibitor comprises a PD-L1 inhibitor, a PD-1 inhibitor and/or a CTLA-4 inhibitor;
preferably, the PD-1 inhibitor is an antibody or antigen-binding fragment thereof;
preferably, the PD-1 inhibitor is a PD-1 antibody.
Preferably, the anti-tumor immunotherapeutic agent comprises an antibody or antibody fragment that targets a tumor-specific antigen;
preferably, the structural formula of STM2457 is shown in formula (I):
9. use of a composition according to any one of claims 1-6 for the manufacture of a medicament for the treatment of cancer;
preferably, the cancer comprises lung cancer, gastrointestinal cancer, pancreatic cancer, glioblastoma, glioma, cervical cancer, ovarian cancer, liver cancer, hepatoma, bladder cancer, breast cancer, colon cancer, colorectal cancer, endometrial cancer, myeloma, multiple myeloma, salivary gland cancer, renal cell carcinoma, nephroblastoma, basal cell carcinoma, melanoma, prostate cancer, vulval cancer, thyroid cancer, testicular cancer, esophageal cancer, or a human mononuclear leukemia cell;
preferably, the cancer comprises ovarian cancer or melanoma.
Use of stm2457 or a derivative thereof in the manufacture of a medicament for the treatment of melanoma or ovarian cancer;
preferably, the structural formula of STM2457 is shown in formula (I):
CN202310516010.XA 2022-05-13 2023-05-09 Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors Pending CN117045801A (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202210524211 2022-05-13
CN202210524211X 2022-05-13

Publications (1)

Publication Number Publication Date
CN117045801A true CN117045801A (en) 2023-11-14

Family

ID=88663339

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310516010.XA Pending CN117045801A (en) 2022-05-13 2023-05-09 Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors

Country Status (2)

Country Link
CN (1) CN117045801A (en)
WO (1) WO2023217109A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN118021819A (en) * 2024-04-15 2024-05-14 中国医学科学院基础医学研究所 Application of XPO1 inhibitor and METTL inhibitor in combination in preparation of medicines for treating gastric cancer

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020201773A1 (en) * 2019-04-05 2020-10-08 Storm Therapeutics Ltd Mettl3 inhibitory compounds
WO2020207550A1 (en) * 2019-04-07 2020-10-15 Chemestmed Ltd. METHOD OF SUPPRESSING CANCER BY RNA m6A DEMETHYLASE AlkBH5 INHIBITORS
EP4045498A4 (en) * 2019-10-14 2023-09-27 The Regents of the University of California Broad spectrum anti-cancer compounds

Also Published As

Publication number Publication date
WO2023217109A1 (en) 2023-11-16

Similar Documents

Publication Publication Date Title
Zhang et al. Specific decrease in B-cell-derived extracellular vesicles enhances post-chemotherapeutic CD8+ T cell responses
Yan et al. FGL2 promotes tumor progression in the CNS by suppressing CD103+ dendritic cell differentiation
Garg et al. Highly activated and expanded natural killer cells for multiple myeloma immunotherapy
Braza et al. Neutrophil derived CSF1 induces macrophage polarization and promotes transplantation tolerance
Luo et al. Extracellular vesicles from Akkermansia muciniphila elicit antitumor immunity against prostate cancer via modulation of CD8+ T cells and macrophages
Shevtsov et al. Ex vivo Hsp70-activated NK cells in combination with PD-1 inhibition significantly increase overall survival in preclinical models of glioblastoma and lung cancer
Kokowski et al. Radiochemotherapy combined with NK cell transfer followed by second-line PD-1 inhibition in a patient with NSCLC stage IIIb inducing long-term tumor control: a case study
Zhu et al. Stroma-derived fibrinogen-like protein 2 activates cancer-associated fibroblasts to promote tumor growth in lung cancer
Alici et al. Anti-myeloma activity of endogenous and adoptively transferred activated natural killer cells in experimental multiple myeloma model
Li et al. BRD4 inhibition by AZD5153 promotes antitumor immunity via depolarizing M2 macrophages
Yuan et al. Dual-function chimeric antigen receptor T cells targeting c-Met and PD-1 exhibit potent anti-tumor efficacy in solid tumors
Anestakis et al. Carboplatin chemoresistance is associated with CD11b+/Ly6C+ myeloid release and upregulation of TIGIT and LAG3/CD160 exhausted T cells
US20080125390A1 (en) Methods for Modulating Immune and Inflammatory Responses
Wu et al. Role of kynurenine in promoting the generation of exhausted CD8+ T cells in colorectal cancer
Park et al. CU06-1004-induced vascular normalization improves immunotherapy by modulating tumor microenvironment via cytotoxic T cells
Cullen et al. Enhanced tumor metastasis in response to blockade of the chemokine receptor CXCR6 is overcome by NKT cell activation
CN117045801A (en) Combination of m6A RNA methylase inhibitors and immune checkpoint inhibitors for the treatment of tumors
Nakazawa et al. Establishment of an efficient ex vivo expansion strategy for human natural killer cells stimulated by defined cytokine cocktail and antibodies against natural killer cell activating receptors
Zhao et al. TIGIT blockade enhances tumor response to radiotherapy via a CD103+ dendritic cell-dependent mechanism
Strecker et al. AAV-mediated gene transfer of a checkpoint inhibitor in combination with HER2-targeted CAR-NK cells as experimental therapy for glioblastoma
Wang et al. TIGIT immune checkpoint blockade enhances immunity of human peripheral blood NK cells against castration-resistant prostate cancer
JP2017520582A (en) Mesenchymal stromal cells for the treatment of rheumatoid arthritis
Celesti et al. Protective anti-tumor vaccination against glioblastoma expressing the MHC class II transactivator CIITA
US20210169937A1 (en) Method for the treatment of a tumor patient with adoptive t cell immunotherapy
Wei et al. Combination therapy of HIFα inhibitors and Treg depletion strengthen the anti‐tumor immunity in mice

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