WO2022052982A1 - USE OF PPARα (PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR α) LIGAND IN PREPARATION OF MEDICINE - Google Patents

USE OF PPARα (PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR α) LIGAND IN PREPARATION OF MEDICINE Download PDF

Info

Publication number
WO2022052982A1
WO2022052982A1 PCT/CN2021/117407 CN2021117407W WO2022052982A1 WO 2022052982 A1 WO2022052982 A1 WO 2022052982A1 CN 2021117407 W CN2021117407 W CN 2021117407W WO 2022052982 A1 WO2022052982 A1 WO 2022052982A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
tumor
pparα
mice
inhibitor
Prior art date
Application number
PCT/CN2021/117407
Other languages
French (fr)
Chinese (zh)
Inventor
曾文峰
张富尧
Original Assignee
昭明泽康(北京)生物医药科技有限公司
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by 昭明泽康(北京)生物医药科技有限公司 filed Critical 昭明泽康(北京)生物医药科技有限公司
Priority to CN202180062556.1A priority Critical patent/CN116367861A/en
Publication of WO2022052982A1 publication Critical patent/WO2022052982A1/en

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
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/42Oxazoles
    • A61K31/4211,3-Oxazoles, e.g. pemoline, trimethadione
    • 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
    • A61K39/46Cellular immunotherapy
    • A61K39/461Cellular immunotherapy characterised by the cell type used
    • A61K39/4615Dendritic cells
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/46Cellular immunotherapy
    • A61K39/462Cellular immunotherapy characterized by the effect or the function of the cells
    • A61K39/4622Antigen presenting cells
    • 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
    • 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 technical field of medicine and biology, and in particular relates to the application of PPAR ⁇ (peroxisome proliferator-activated receptor ⁇ ) ligands in the preparation of medicines.
  • PPAR ⁇ peroxisome proliferator-activated receptor ⁇
  • tumor cells use various immune evasion strategies to evade T cell immune surveillance, such as: resisting T cell killing of tumor cells, high expression of death receptors to induce T cell death, immune checkpoint signaling or immunosuppression Sexual factors (TGF- ⁇ , PGE2, etc.) inhibit T cell activation and induce a large number of suppressive immune cell subsets (regulatory T cells or myeloid-derived suppressor cells).
  • TGF- ⁇ , PGE2, etc. immunosuppression Sexual factors
  • suppressive immune cell subsets regulatory T cells or myeloid-derived suppressor cells.
  • the main problem of tumor immune escape is the inability of infiltrating T cells in tumor tissue to be fully activated and the inability to generate durable memory T cells. Therefore, enhancing or improving the activation and survival of T cells in the tumor microenvironment and generating memory T cells to maintain long-term anti-tumor activity will be the core task of developing tumor immunotherapy.
  • T cell activation initiates a series of intracellular signaling cascades, which ultimately lead to different biological events such as T cell proliferation, immune function, or death, depending on the strength and duration of the activation signal.
  • the full activation of T cells requires two independent signals: the first signal is an antigen-specific signal, which is bound by the T cell receptor (TCR) on the surface of the T cell membrane and the antigen-MHC complex on the surface of the antigen presenting cell , causing activation of downstream tyrosine kinases; the second signal is mediated by cytokines or co-stimulatory molecules on the surface of antigen-presenting cells, such as B7.1 (CD80) and B7.2 (CD86).
  • TCR T cell receptor
  • cytokines or co-stimulatory molecules on the surface of antigen-presenting cells, such as B7.1 (CD80) and B7.2 (CD86).
  • TAK1 TGF- ⁇ -activated kinase 1
  • TGF- ⁇ -activated kinase 1 can integrate signals that cause T cell development, activation, survival and other functions, and is at the core of T cell function regulation 3 .
  • PPAR ⁇ knockout mice As well as in PPAR ⁇ knockout mice, it was found that the NF- ⁇ B and JNK pathways downstream of TAK1 in T cells were selectively activated, resulting in increased production of cytokines such as IFN- ⁇ , TNF- ⁇ and IL-2 6 .
  • cytokines such as IFN- ⁇ , TNF- ⁇ and IL-2 6 .
  • PPAR ⁇ can inhibit the NF- ⁇ B cell pathway in the presence of activators, indicating that PPAR ⁇ is an important negative regulator in T cells.
  • PPAR ⁇ inhibitors can have the same T cell regulation effect.
  • PPAR ⁇ as an intracellular activation signal checkpoint molecule of T cells, binds with PPAR ⁇ ligand molecules to release the TAK1 kinase hidden by PPAR ⁇ and increase the activation of downstream NF- ⁇ B and JNK kinases. , improve the sensitivity of T cells, strengthen T cell activation, and promote T cells to secrete more cytokines (such as IL-2 and IFN- ⁇ ) to promote their own proliferation and anti-tumor activity ( Figure 13). And in combination with immunotherapy (including immune checkpoint inhibitors and anti-tumor therapeutic vaccines), it can enhance the control of tumor growth by immunotherapy, and can also promote the generation of memory T cells to prevent tumor recurrence.
  • immunotherapy including immune checkpoint inhibitors and anti-tumor therapeutic vaccines
  • DCs such as the presentation of tumor-associated antigens and the provision of costimulatory molecules and other signals. Therefore, the function of DCs is indispensable for a successful antitumor response.
  • immune cells are disadvantaged in the process of highly competing with tumor cells for nutrients such as glucose, fatty acids, and amino acids. Therefore, immune cells, such as DCs, are usually in a state of extreme nutritional starvation and cannot reach a fully activated or mature state. Therefore, the metabolic state of DC can reflect its activation state and function.
  • Current studies have shown that events such as increased mitochondrial activity and enhanced fatty acid oxidation in DCs can cause immune dysfunction in DCs, preventing them from fully activating T cells.
  • PPAR ⁇ can be activated by both endogenous and exogenous lipid molecular ligands 12 .
  • These lipid molecules include saturated fatty acids (palm, stearic acid), unsaturated fatty acids (oleic acid, linoleic acid, arachidonic acid), 2-oleoyl-1-palmitin glycerol-3-phosphocholine ( 16:0/18:0 GPC), hexadecylamide ethanol (PEA), etc. 13 .
  • PPAR ⁇ is sensitive to fatty acid sensing and can be rapidly activated to initiate transcription and regulate the expression of fatty acid ⁇ -oxidation and lipid metabolism-related genes 14,15 . Regulation of the metabolic level of DC cells by PPAR ⁇ ligands can improve the immunosuppression of DCs caused by the immune microenvironment, thereby improving the anti-tumor activity of T cells.
  • the present invention first relates to the application of PPAR ⁇ (peroxisome proliferator-activated receptor ⁇ ) ligands in the preparation of medicines, and the functions of the medicines include any one or more of the following functions,
  • the activation signal refers to an increase in the phosphorylation level of JNK, and the T cell is preferably a CD8+ T cell;
  • the activation of T cells refers to making T cells secrete more IFN- ⁇ and IL-2 and at the same time increase the survival time of T cells, the T cells are preferably CD8+ T cells, more preferably , for tumor-infiltrating CD8+ T cells;
  • the differentiation refers to promoting the differentiation of T cells into memory T cells
  • the regulating metabolism refers to: down-regulating the fatty acid oxidation/oxidative phosphorylation level of DC cells, and up-regulating the level of glycolysis;
  • the DC are bone marrow-derived DCs, more preferably, DCs infiltrating the tumor microenvironment;
  • Restoring the regulatory function of DC on T cells refers to: down-regulating the ratio of CD4+ T cells to regulatory T cells, increasing the ratio of tumor-killing CD8+ T cells, and activating tumor-killing The function of CD8+ T cells;
  • the activation of tumor-killing CD8+ T cells refers to: making tumor-killing CD8+ T cells secrete more IFN- ⁇ and IL-2 while increasing tumor-killing CD8+ T cells survival time;
  • the immune checkpoint inhibitor is preferably a PD-L1/PD-1 antibody, and preferably, the tumor is a tumor that is effectively treated by an immune checkpoint inhibitor ;
  • the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigen through PEG2000-DSPE micelles.
  • the PPAR ⁇ ligand is preferably an inhibitor of PPAR ⁇ , more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
  • the present invention also relates to a group of PPAR ⁇ inhibitors, the inhibitors are ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
  • the present invention also relates to a method for treating tumors, which comprises administering a therapeutically effective amount of an immune checkpoint inhibitor and a PPAR ⁇ ligand at the same time;
  • the tumor is a tumor that is effectively treated by an immune checkpoint inhibitor, and the immune checkpoint inhibitor is a PD-L1/PD-1 antibody;
  • the PPAR ⁇ ligand is preferably an inhibitor of PPAR ⁇ , more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
  • the present invention also relates to a method for treating tumors, which comprises administering a therapeutically effective amount of an anti-tumor vaccine and a PPAR ⁇ ligand at the same time;
  • the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigens through PEG2000-DSPE micelles, and the preparation method of the micelles can refer to the records of CN201410570624;
  • the PPAR ⁇ ligand is preferably an inhibitor of PPAR ⁇ , more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
  • the present invention also relates to an anti-tumor combined preparation comprising:
  • the immune checkpoint inhibitor is PD-L1/PD-1 antibody
  • the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigens through PEG2000-DSPE micelles;
  • the PPAR ⁇ ligand is preferably an inhibitor of PPAR ⁇ , more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
  • PPAR ⁇ inhibitor GW6471 enhances the anti-tumor effect of PD-1 antibody (PD-1 mAb) on MC38
  • PPAR ⁇ inhibitor GW6471 increases the proportion of memory CD8+ T cells induced by PD-L1 antibody.
  • the PPAR ⁇ inhibitor GW6471 enhances the activation of CD8+ T cells by tumor-infiltrating DCs.
  • FIG. 14 The PPAR ⁇ inhibitor GW6471 inhibits the induction of regulatory T cells by tumor exosomes [in vitro experiment].
  • Figure 18 Diagram of the mechanism of interaction between PPAR ⁇ and TAK1 in T cells.
  • CD11c-DTR mouse model which is characterized by the presence of a diphtheria toxin response element (DTR, DT: diphtheria toxin, R: response Original), in the case of intraperitoneal injection of this toxin, CD11c-positive DC cells derived from mouse bone marrow will be eliminated.
  • DTR diphtheria toxin response element
  • R response Original
  • the mouse CD11c-DTR/PPAR ⁇ -/- chimeric bone marrow transfer model that is, on the basis of the first model, the PPAR ⁇ in the mouse genome is further knocked out, which is characterized by the simultaneous transfer of the same amount of CD11c-DTR bone marrow Cells and PPAR ⁇ -/- myeloid cells, when mice were treated with diphtheria toxin, only PPAR ⁇ -/- DC cells remained in the body, but no wild-type PPAR ⁇ function DC cells.
  • the role of the PPAR ⁇ signaling pathway in DC cells on tumor growth and in anti-tumor immunity can be investigated without the need for antibody or small molecule drug treatment, and only by observing tumor growth.
  • Example 1 The PPAR ⁇ inhibitor GW6471 enhances the phosphorylation of T cell activation-related kinases in T cells
  • CD8+ T cells were treated with different concentrations of Fenofibrate (20 ⁇ M) or GW6471 (10 ⁇ M) for 20 minutes, 2.5ug/ml anti-CD3 antibody and 0.5ug/ml anti-CD28 antibody were used to activate T cells for 20 minutes.
  • CD8+ T cells were collected, and the cells were lysed with Lysis Buffer on ice, and the intracellular proteins were extracted.
  • Western Blot experiments were performed to investigate the changes of PPAR ⁇ , JNK phosphorylation and I-kB degradation.
  • Example 2 PPAR ⁇ inhibitor GW6471 and agonist Fenofibrate enhance the survival and function of CD8+ T cells
  • CD8+ T cells were treated with different concentrations of Fenofibrate (20 ⁇ M) or GW6471 (10 ⁇ M) for 24, 48, and 72 hours, the cells were harvested, and 0.5ug/ml anti-CD3 antibody and 0.1ug/ml anti-CD3 antibody were used again. 72 hours after CD28 antibody activation of CD8+ T cells, the cell culture supernatant was collected, and the levels of IFN- ⁇ and IL-2 in the supernatant were detected by ELISA.
  • the results in Figure 2 show that after pretreatment with GW6471, the state of CD8+ T cells was adjusted, and after restimulation, more IFN- ⁇ and IL-2 could be secreted.
  • the CD8+ T cells in the 24-hour pretreatment group could secrete 3 times the IFN- ⁇ produced by the CD8+ T cells in the unpretreated and restimulated group, while the CD8+ T cells in the 72-hour pretreatment group could be stimulated by Activates and produces more IL-2 that maintains T cell survival.
  • CD8+ T cells pretreated with Fenofibrate for 24, 48 and 72 hours were able to produce more IFN- ⁇ , but not IL-2.
  • PPAR ⁇ ligand name/number IC50 EC 200 GW6471 34.61uM 0.0022uM Fenofibrate >40uM 0.0076uM ZM0282 31.49uM 0.0058uM ZM0283 0.3537uM 0.0087uM ZM0284 7.495uM 0.0258uM ZM0285 0.1303uM 0.7892uM ZM0286 12.71uM Unable to reach within lethal dose ZM0325 30.9uM Unable to reach within lethal dose ZM0326 2.417uM Unable to reach within lethal dose ZM0329 40.96uM Unable to reach within lethal dose ZM0330 8.423uM Unable to reach within lethal dose ZM0332 >40uM Unable to reach within lethal dose
  • IC 50 median lethal dose.
  • mice The typical PPAR ⁇ inhibitor GW6471 enhances the anti-tumor effect of PD-1/PD-L1 antibody on MC38-OT I, activates the function of tumor-infiltrating T lymphocytes, increases the proportion of memory T cells, and prolongs the survival of mice. Survival period
  • mice-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is easy to detect and recognize tumor antigen-specific T lymphocytes) was used to subcutaneously inoculate C57 mice, which can be measured in about 5 days to tumor formation (average tumor volume was approximately 50 mm 3 ).
  • CTR tumor-bearing mice control group
  • mice A total of 4 groups of tumor-bearing mice were tested, with 6 mice in each group.
  • mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 11th day after inoculation, and GW6471 was given once every other day, for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 0.1 ml).
  • mice-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is easy to detect and recognize tumor antigen-specific T lymphocytes) was used to subcutaneously inoculate C57 mice, which can be measured in about 5 days to tumor formation (average tumor volume was approximately 50 mm 3 ).
  • CTR tumor-bearing mice control group
  • mice There were 3 groups of tumor-bearing mice in the experiment, with 7 mice in each group.
  • mice were treated with PD-1 antibody on the 8th and 11th day after inoculation, 50ug per mouse, intraperitoneal injection (PD-1 mAb was diluted in PBS, the concentration of 0.5mg/ml, each mouse was intraperitoneally injected with 0.1ml).
  • PD-1 mAb was diluted in PBS, the concentration of 0.5mg/ml, each mouse was intraperitoneally injected with 0.1ml).
  • the mice were treated with GW6471, 30 mg/kg, orally administered daily on the 5th day after inoculation.
  • GW6471 was ground with a mortar, diluted and suspended in 0.3% CMC-Na solution to reach a concentration of 3 mg/ml and stored at 4°C. The drug was taken out before administration every day, and it was administered by gavage after equilibrating to room temperature.
  • mice were divided into 4 groups with 6 mice in each group, and were treated with PD-L1 antibody and GW6471 respectively. This method is completely the same as the experimental scheme (1).
  • GW6471 can significantly improve the ability of tumor-infiltrating CD8+ T lymphocytes to secrete perforin, Granzyme B and IFN ⁇ , that is, the function of killing tumor cells.
  • Activity of T cells Ki67
  • mice were divided into four groups as above, with 6 mice in each group, and were treated with PD-L1 antibody and GW6471 respectively. This method is completely the same as the experimental scheme (1).
  • the MC38-OT I tumor-bearing model was constructed as above.
  • CTR tumor-bearing mice control group
  • mice A total of 4 groups of tumor-bearing mice were tested, with 6 mice in each group.
  • mice were treated with PD-L1 antibody on days 7, 10 and 17 after inoculation, 200ug per mouse, intraperitoneal injection (PD-L1 mAb diluted in PBS) , the concentration is 2mg/ml, each mouse is intraperitoneally injected with 0.1ml).
  • mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 11th day after inoculation, and GW6471 was given once every other day, for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 0.1 ml).
  • Example 4 The anti-tumor effect of typical PPAR ⁇ inhibitor GW6471 combined with PD-L1 antibody mainly depends on DC cells, especially PPAR ⁇ -/-DC
  • CD11c-DTR and CD11c-DTR/PPAR ⁇ -/- chimeric mouse models First, wild-type C57 mice (female) were irradiated with 10 Gy whole body for 10 minutes to kill bone marrow cells. On the second day, the bone marrow cells of CD11c-DTR mice were taken and injected into the irradiated mice by tail vein according to the number of cells per mouse at 5 ⁇ 10 6 . To construct CD11c-DTR/PPAR ⁇ -/- chimeric mice, CD11c-DTR mice and PPAR ⁇ -/- mouse bone marrow cells were mixed 1:1 and injected into the irradiated mice via tail vein. After two weeks, the antibiotic ampicillin was given to observe their living conditions, and the experiment could be carried out after two months.
  • Inoculation of tumor cells Use a 1 ml sterile syringe to subcutaneously inoculate the model mice constructed above, 0.1 ml per mouse (ie, 50 ⁇ 10 4 MC38-OT I cells per mouse). Take care to mix the cells well before each pipetting of the cell suspension.
  • mice The mouse-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is convenient for the detection of specific T lymphocytes) was used to subcutaneously inoculate the constructed CD11c-DTR mice. Tumor formation of about 50 mm3 was seen.
  • the experiment was divided into 3 groups, which were
  • CTR tumor-bearing mice control group
  • mice A total of 4 groups of tumor-bearing mice were tested, with 5 mice in each group.
  • mice were treated with GW6471, 10 mg/kg, intraperitoneal injection, and GW6471 was administered every other day (GW6471 stock solution was 25 mg/ml dissolved in DMSO, before administration, Dilute in pre-warmed PBS at 55°C to complete dissolution, the concentration is 2mg/ml, and each mouse is injected intraperitoneally with 1ml).
  • Wild-type C57 mice were inoculated with MC38-OT I cells. After the model was constructed, they were divided into 3 groups with 9 mice in each group, namely the tumor-bearing control group (CTR) and the wild-type DC treatment group (WT). DC) and PPAR ⁇ knockout DC treatment group (PPAR ⁇ -/- DC).
  • CTR tumor-bearing control group
  • WT wild-type DC treatment group
  • DC PPAR ⁇ knockout DC treatment group
  • PPAR ⁇ -/- DC PPAR ⁇ knockout DC treatment group
  • Tumor growth statistics During the tumor growth period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length ⁇ width ⁇ width/2, and the tumor growth curve was drawn.
  • mice were divided into two groups: tumor-bearing control group and GW6471 administration group.
  • the GW6471 administration group received intraperitoneal injection of 10 mg/kg GW6471 from the 7th day after tumor receiving, once every two days, for a total of 6 times.
  • the mice were sacrificed, and the tumor tissue was surgically removed, ground and digested to obtain tumor tissue cells.
  • tumor-infiltrating DCs (CD45+MHCII+CD11c+F4/80-) were sorted by flow cytometry, incubated with 2 mg/ml OVA antigen for 48 hours, and then washed away with antigen.
  • the OTCD8+ T cells obtained by magnetic bead sorting from the spleen of OTI transgenic mice were co-incubated at a ratio of 1:8. After 3 days, the proliferation status and secretion of IFN- gamma level.
  • Flow cytometry was used to detect the dilution ratio of CFSE to determine the division state of OTI CD8+ T cells. Generally speaking, cell proliferation is about obvious, and the higher the dilution of CFSE, the lower the average fluorescence intensity. The level of IFN- ⁇ secreted by OTI CD8+ T cells can also be detected by ELISA kit to indicate the activation state of OTI CD8+ T cells.
  • Wild-type and PPAR ⁇ knockout mice were inoculated with MC38-OTI tumor cells, respectively. On the 20th day after receiving the tumor, the mice were sacrificed, and the tumor tissue was surgically removed, ground and digested to obtain tumor tissue cells.
  • In vitro treatment pre-incubate anti-mouse IFN- ⁇ antibody in an ELISPOT plate one day before the isolation of mouse intratumoral T cells at a concentration of 5 ⁇ g/ml, incubate overnight at 4°C, discard the antibody the next day, and add blocking solution ( RPMI 1640 medium containing 10% fetal bovine serum, 1% penicillin-streptomycin and ⁇ -mercaptoethanol) was blocked at room temperature for 2 h. Then, the isolated mouse tumor T cells were added to the plate at 3 ⁇ 105 per well, and incubated with 15 ⁇ g/ml OVA257-264 antigen peptide for 48 h in a 37°C incubator.
  • blocking solution RPMI 1640 medium containing 10% fetal bovine serum, 1% penicillin-streptomycin and ⁇ -mercaptoethanol
  • Cell staining Fluorescent staining of the above cell suspension, the color scheme is APC-CD45, PE/Cy7-CD3, APC/Cy7-CD8, PE-Tetramer (Tet), after mixing the antibody with the cells, stain at 4°C in the dark 30min. The staining was terminated by washing once with PBS containing 2% FBS.
  • Tumor cells were cultured in a medium containing 10% exosome-depleted fetal bovine serum (100,000 g, 2 h) for 48 h, and the supernatant was collected. First, cell debris in the supernatant was removed by centrifugation at 4000 rpm for 2 h. Then, the fractions larger than 100 kDa in the supernatant were collected by using a 100 kDa ultrafiltration tube at a speed of 4000 rpm, 30 min/time.
  • TDE Tumor-derived Exosomes
  • the collected components were added to the exosome extraction reagent (EXOTC10A-1, SBI) at a ratio of 5:1 (v:v), mixed well and placed at 4°C for at least 12 hours. After 12 hours, the precipitate appeared, centrifuged at 3000 rpm for 30 min, and the precipitate was separated, which was exosomes. After resuspending in PBS according to a certain volume, the protein content was determined by BCA to standardize the content of exosomes, and the packaged and stored at -80°C for future use.
  • EXOTC10A-1, SBI exosome extraction reagent
  • Processing of tumor exosomes can alter the metabolic state and function of bone marrow-derived DCs, mimicking the effects of the tumor microenvironment.
  • Seahorse XF24 cell culture plate by Seahorse uses Corning TM Cell-Tak Cell and Tissue Adhesive Reagent (to promote the adhesion of suspended cells to the bottom of the cell culture plate) 3.5 ⁇ g/cm 2
  • Corning TM Cell-Tak Cell and Tissue Adhesive Reagent to promote the adhesion of suspended cells to the bottom of the cell culture plate
  • bone marrow-derived DCs pretreated with exosomes or GW6471 for 48h were seeded in culture plates at 300,000 per well, with five replicate wells per group, 0.5ml system, and cells adhered for 4h.
  • Seahorse hippocampal bioenergetics assay levels of overall cellular metabolism: Probe plates were hydrated overnight. Before assay, cells were replaced with pre-warmed 525 ⁇ l XF Base medium (Seahorse Bioscience) supplemented with 10 mM glucose, 2 mM L-glutamine and 1 mM sodium pyruvate, and adjusted to pH 7.4 with NaOH Available after filtering.
  • Oligomycin oligomycin
  • FCCP trifluoromethoxyphenylhydrazone carbonyl cyanide
  • Rotenone rotenone
  • Antimycin A Antimycin A
  • Seahorse hippocampal bioenergetics assay level of fatty acid oxidation: Probe plate hydrated overnight. 45 min before the test, the cells were replaced with pre-warmed 450 ⁇ l FAO assay medium (fatty acid oxidation assay medium, containing 111 mM NaCl, 4.7 mM KCl, 2 mM MgSO 4 , 1.2 mM Na 2 HPO 4 , 2.5 mM glucose, 0.5 mM meat Alkaline and 5 mM HEPES sulfonic acid buffered saline).
  • FAO assay medium fatty acid oxidation assay medium, containing 111 mM NaCl, 4.7 mM KCl, 2 mM MgSO 4 , 1.2 mM Na 2 HPO 4 , 2.5 mM glucose, 0.5 mM meat Alkaline and 5 mM HEPES sulfonic acid buffered saline.
  • ETO an inhibitor of carnitine palmitoyl transporter 1
  • DCs derived from bone marrow were seeded in 24-well cell culture plates at 300,000 per well, and the supernatant was collected after pretreatment with exosomes or GW6471 for 48 hours.
  • Lactate Colorimetric Assay Kit (Biovision) was used to detect the lactic acid content in the supernatant, and the absorbance at 570 nm was read for quantitative analysis.
  • TDE tumor exosomes
  • OCR DC cell oxygen consumption
  • Fig. 13a marker of cellular oxidative phosphorylation levels
  • Fig. 13b fatty acid oxidation levels
  • Secretion ( Figure 13c) Annotation of cellular glycolysis levels. Therefore, tumor exosomes can mimic the regulation of the metabolic state of DCs by the tumor microenvironment in vitro.
  • GW6471 could significantly reduce the level of oxidative phosphorylation and fatty acid oxidation of DC, and restore the level of glycolysis (Fig. 13), and the metabolic state change caused by GW6471 contributed to the recovery of DC function.
  • Foxp3-GFP transgenic mice are characterized by gene editing to place the gene expressing green fluorescent protein (GFP) behind the promoter of the transcription factor Foxp3 responsible for the development of CD4+ regulatory T cells. Therefore, we can easily detect the transformation of CD4+ T cells into regulatory T cells in vivo or in vitro by the method of fluorescence detection (GFP positive means Foxp3 positive), rather than the complex intracellular protein staining method in the past. condition.
  • GFP green fluorescent protein
  • OTI (C57BL/6-Tg(TcraTcrb) 1100Mjb/J) transgenic mice are characterized by gene editing, so that the T cell receptors of the CD8+ T cells of the mice can specifically recognize the 257-264 of the OVA protein.
  • the structure of the polypeptide is activated and proliferated.
  • the transgenic mice are commonly used in studies investigating antigen-specific immune responses.
  • BMDCs were first pretreated with exosomes and 15uM GW6471 for 48h.
  • splenocytes of Foxp3-GFP transgenic mice were taken, and CD4+ cells were sorted by magnetic beads, and counted after centrifugation.
  • the above pretreated DCs (2 ⁇ 10 4 ) were combined with CFSE (carboxyfluorescein diacetate succinimidyl ester, commonly used to label and monitor the proliferation state of cells) labeled CD8+ from the spleen of OTI transgenic mice.
  • T cells (2 x 105 ) were co-cultured at a ratio of 1:10 in U-bottom 96-well plates. On the fourth day, 100U/well of recombinant mouse IL-2 cytokine was added to promote the survival of T cells.
  • BMDCs were first incubated with exosomes, 2mg/ml OVA antigen, and 15uM GW6471 for 48h.
  • spleen cells of OT I transgenic mice were taken, and CD8+ cells were sorted by magnetic beads and counted after centrifugation.
  • the above pretreated DCs (5 ⁇ 10 4 ) were co-cultured with OTI CD8+ T cells (4 ⁇ 10 5 ) at a ratio of 1:8 in a U-bottom 96-well plate.
  • Inoculation of tumor cells Use a 1 ml sterile syringe to subcutaneously inoculate female wild-type C57 mice with 0.1 ml per mouse (ie, inoculate 5 ⁇ 10 4 TC-1 cells per mouse). Take care to mix the cells well before each pipetting of the cell suspension. Tumor formation of about 50 mm3 was seen in about 7 days.
  • CTR tumor-bearing mice control group
  • mice There were 4 groups of tumor-bearing mice, 10 mice in each group.
  • mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 8th day after inoculation, and GW6471 was given once every other day for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 1 ml).

Abstract

Provided is use of a PPARα (peroxisome proliferator-activated receptor α) ligand in the preparation of a medicine. The medicine alone or in any combination has the following functions: (1) activating an activation signal in T cells; (2) activating T cells; (3) promoting the differentiation of T cells; (4) regulating the metabolic state of DCs; (5) restoring the regulatory function of DCs on T cells; (6) improving the antitumor effect of immune checkpoint inhibitors; and (7) improving the therapeutic effect of antitumor vaccines.

Description

PPARα(过氧化物酶体增殖物激活受体α)配体在制备药物中的应用Application of PPARα (peroxisome proliferator-activated receptor α) ligand in the preparation of medicine 技术领域technical field
本发明属于医药生物技术领域,具体的,涉及PPARα(过氧化物酶体增殖剂激活受体α)配体在制备药物中的应用。The invention belongs to the technical field of medicine and biology, and in particular relates to the application of PPARα (peroxisome proliferator-activated receptor α) ligands in the preparation of medicines.
背景技术Background technique
肿瘤生长过程中,肿瘤细胞会采用多种免疫逃逸策略规避T细胞的免疫监视,例如:抵抗T细胞对肿瘤细胞的杀伤、高表达死亡受体诱导T细胞死亡、通过免疫检查点信号或免疫抑制性因子(TGF-β、PGE2等)抑制T细胞活化以及诱导产生大量抑制性免疫细胞亚群(调节性T细胞或髓样来源的抑制性细胞)。简而言之,肿瘤免疫逃逸的主要问题是肿瘤组织中浸润的T细胞无法被充分激活以及无法产生持久的记忆性T细胞。因此,提高或改善肿瘤微环境中T细胞的活化与存活,并产生记忆性T细胞以维持长期的抗肿瘤活性将是开发肿瘤免疫疗法的核心任务。During tumor growth, tumor cells use various immune evasion strategies to evade T cell immune surveillance, such as: resisting T cell killing of tumor cells, high expression of death receptors to induce T cell death, immune checkpoint signaling or immunosuppression Sexual factors (TGF-β, PGE2, etc.) inhibit T cell activation and induce a large number of suppressive immune cell subsets (regulatory T cells or myeloid-derived suppressor cells). In short, the main problem of tumor immune escape is the inability of infiltrating T cells in tumor tissue to be fully activated and the inability to generate durable memory T cells. Therefore, enhancing or improving the activation and survival of T cells in the tumor microenvironment and generating memory T cells to maintain long-term anti-tumor activity will be the core task of developing tumor immunotherapy.
T细胞活化会启动一系列细胞内信号级联反应,并根据激活信号的强度和持续时间,最终引起T细胞增殖、执行免疫功能或死亡等不同的生物学事件。T细胞的完全激活需要两个互相独立的信号:第一信号是抗原特异性信号,由T细胞膜表面的T细胞受体(T cell receptor,TCR)与抗原呈递细胞表面的抗原-MHC复合物结合,引起下游酪氨酸激酶的活化;第二信号是通过细胞因子或抗原呈递细胞表面的共刺激分子,诸如B7.1(CD80)和B7.2(CD86)介导产生。如果只接受第一信号会导致T细胞失能,而单独激活第二信号也不会引起T细胞的活化。因此两个信号之间的协同作用对T细胞发挥免疫功能具有重要的意义。第一信号和第二信号的充分激活,将引起NF-κB和MAPK(JNK,p38和ERK)等重要的激酶磷酸化,并激活下游的转录因子,促进T细胞活化。而T细胞活化后,在细胞因子,例如IL-2、IL-7和IL-15的作用下,上述激酶进一步被活化,则会启动T细胞的增殖以及向记忆性T细胞分化 1,2 。有研究表明TAK1(TGF-β激活激酶1)能够整合引起T细胞发育、活化、存活等功能的信号,处于T细胞功能调控的核心位置 3 T cell activation initiates a series of intracellular signaling cascades, which ultimately lead to different biological events such as T cell proliferation, immune function, or death, depending on the strength and duration of the activation signal. The full activation of T cells requires two independent signals: the first signal is an antigen-specific signal, which is bound by the T cell receptor (TCR) on the surface of the T cell membrane and the antigen-MHC complex on the surface of the antigen presenting cell , causing activation of downstream tyrosine kinases; the second signal is mediated by cytokines or co-stimulatory molecules on the surface of antigen-presenting cells, such as B7.1 (CD80) and B7.2 (CD86). Receiving only the first signal would result in T cell inactivation, while activation of the second signal alone would not cause T cell activation. Therefore, the synergistic effect between the two signals is of great significance to the immune function of T cells. The sufficient activation of the first signal and the second signal will cause the phosphorylation of important kinases such as NF-κB and MAPK (JNK, p38 and ERK), and activate the downstream transcription factors to promote T cell activation. After activation of T cells, the above-mentioned kinases are further activated under the action of cytokines, such as IL-2, IL-7 and IL-15, to initiate T cell proliferation and differentiation into memory T cells 1,2 . Studies have shown that TAK1 (TGF-β-activated kinase 1) can integrate signals that cause T cell development, activation, survival and other functions, and is at the core of T cell function regulation 3 .
PPARα与TAK1之间存在相互作用,例如小鼠的肝细胞中敲除TAK1后发现,不仅PPARα的mRNA水平降低,相应的PPARα下游控制脂代谢和脂肪酸氧化相关的基因表达量也降低,同时遏制了肝细胞的脂肪酸氧化能力,导致肝细胞中脂类沉积 5 ,说明TAK1的活化可以上调PPARα的胞内表达量。以及PPARα敲除小鼠中发现T细胞中的TAK1下游的NF-κB和JNK通路被选择性活化,并导致更多的IFN-γ,TNF-α和IL-2等细胞因子的产生 6 。此外PPARα在激活剂存在的情况下能够抑制NF-κB细胞通路,说明PPARα在T细胞中是一个重要的负调控因子。但是,基于现有的理论和固有思路之限制,对于PPARα抑制剂是否能够起到同样的T细胞调节效果,目前未见报道。 There is an interaction between PPARα and TAK1. For example, after knocking out TAK1 in mouse hepatocytes, it was found that not only the mRNA level of PPARα was reduced, but also the expression of genes related to the control of lipid metabolism and fatty acid oxidation downstream of PPARα. The fatty acid oxidation capacity of hepatocytes leads to lipid deposition in hepatocytes 5 , indicating that activation of TAK1 can upregulate the intracellular expression of PPARα. As well as in PPARα knockout mice, it was found that the NF-κB and JNK pathways downstream of TAK1 in T cells were selectively activated, resulting in increased production of cytokines such as IFN-γ, TNF-α and IL-2 6 . In addition, PPARα can inhibit the NF-κB cell pathway in the presence of activators, indicating that PPARα is an important negative regulator in T cells. However, based on the limitations of existing theories and inherent thinking, there is no report on whether PPARα inhibitors can have the same T cell regulation effect.
尽管原有观点认为PPARα的激活剂具有抗炎作用,可以通过抑制NF-κB的转录活性抑制T细胞的活化。但是最近有研究显示,使用PPARα选择性激活剂Fenofibrate预处理过的CD8+T细胞在过继性T细胞治疗中显示出更优异的抗肿瘤效果 8 。而且泛PPAR激动剂Bezafibrate配合免疫检查点抑制剂的联用策略使肿瘤得到很好的控制,荷瘤小鼠生存期显著延长 9 。上述研究均支持我们的理论推断,但是它们的分析认为该效应得益于PPARα对T细胞代谢状态的调节而非PPARα直接影响了T细胞活化相关的信号通路。 Although the original view is that activators of PPARα have anti-inflammatory effects, they can inhibit the activation of T cells by inhibiting the transcriptional activity of NF-κB. However, recent studies have shown that CD8+ T cells pretreated with the PPARα-selective activator Fenofibrate show superior antitumor effects in adoptive T cell therapy 8 . In addition, the combination strategy of pan-PPAR agonist Bezafibrate and immune checkpoint inhibitors can control the tumor well, and the survival time of tumor-bearing mice is significantly prolonged 9 . The above studies all support our theoretical inference, but their analysis suggests that the effect is due to the regulation of PPARα on T cell metabolic state rather than PPARα directly affecting T cell activation-related signaling pathways.
通过系统的研究,我们前期发现,PPARα作为T细胞的胞内激活信号检查点分子,通过PPARα的配体分子与其结合,来释放被PPARα掩藏的TAK1激酶,增加下游NF-κB和JNK激酶的活化,提高T细胞的敏感性,强化T细胞活化,促进T细胞分泌更多的细胞因子(如IL-2和IFN-γ)促进自身的增殖与抗肿瘤活性(图13)。而且在与免疫治疗(包括免疫检查点抑制剂和抗肿瘤治疗性疫苗)联用中,能够增强免疫治疗对肿瘤生长的控制,更能够促进记忆性T细胞的产生,防止肿瘤的复发。Through systematic research, we previously found that PPARα, as an intracellular activation signal checkpoint molecule of T cells, binds with PPARα ligand molecules to release the TAK1 kinase hidden by PPARα and increase the activation of downstream NF-κB and JNK kinases. , improve the sensitivity of T cells, strengthen T cell activation, and promote T cells to secrete more cytokines (such as IL-2 and IFN-γ) to promote their own proliferation and anti-tumor activity (Figure 13). And in combination with immunotherapy (including immune checkpoint inhibitors and anti-tumor therapeutic vaccines), it can enhance the control of tumor growth by immunotherapy, and can also promote the generation of memory T cells to prevent tumor recurrence.
此外,由于T细胞发挥抗肿瘤活性往往需要DC的辅助,例如呈递肿瘤相关抗原、提供共刺激分子等 信号。因此,成功的抗肿瘤反应中DC的功能是不可或缺的。但是,肿瘤微环境中,免疫细胞在与肿瘤细胞高度竞争营养物质,例如葡萄糖、脂肪酸和氨基酸的过程中出与劣势。所以免疫细胞,例如DC,通常处于营养极度匮乏状态,无法达到完全活化或成熟状态。因此,DC的代谢状态能够反映其活化状态与功能。目前的研究表明DC胞内线粒体活性升高、脂肪酸氧化增强等事件会引起DC的免疫功能紊乱,使其无法充分激活T细胞,而抑制脂肪酸氧化可以重塑DC的胞内代谢途径,恢复其对T细胞的激活能力 10,11 。PPARα作为II型核激素受体超家族成员,能够接受内源性和外源性脂类分子配体的激活 12 。这些脂类分子包括饱和脂肪酸(棕榈树、硬脂酸)、不饱和脂肪酸(油酸、亚油酸、花生四烯酸)、2-油酰-1-棕榈锡甘油-3-磷酸胆碱(16:0/18:0GPC)、十六酰胺乙醇(PEA)等 13 。PPARα对脂肪酸的感应很敏感,可以被迅速激活而启动转录,调控脂肪酸β氧化和脂代谢相关基因的表达 14,15 。通过PPARα的配体对DC细胞的代谢水平进行调控可以改善免疫微环境对DC造成的免疫抑制,进而改善T细胞的抗肿瘤活性。 In addition, the anti-tumor activity of T cells often requires the help of DCs, such as the presentation of tumor-associated antigens and the provision of costimulatory molecules and other signals. Therefore, the function of DCs is indispensable for a successful antitumor response. However, in the tumor microenvironment, immune cells are disadvantaged in the process of highly competing with tumor cells for nutrients such as glucose, fatty acids, and amino acids. Therefore, immune cells, such as DCs, are usually in a state of extreme nutritional starvation and cannot reach a fully activated or mature state. Therefore, the metabolic state of DC can reflect its activation state and function. Current studies have shown that events such as increased mitochondrial activity and enhanced fatty acid oxidation in DCs can cause immune dysfunction in DCs, preventing them from fully activating T cells. Activation capacity of T cells 10,11 . As a member of the type II nuclear hormone receptor superfamily, PPARα can be activated by both endogenous and exogenous lipid molecular ligands 12 . These lipid molecules include saturated fatty acids (palm, stearic acid), unsaturated fatty acids (oleic acid, linoleic acid, arachidonic acid), 2-oleoyl-1-palmitin glycerol-3-phosphocholine ( 16:0/18:0 GPC), hexadecylamide ethanol (PEA), etc. 13 . PPARα is sensitive to fatty acid sensing and can be rapidly activated to initiate transcription and regulate the expression of fatty acid β-oxidation and lipid metabolism-related genes 14,15 . Regulation of the metabolic level of DC cells by PPARα ligands can improve the immunosuppression of DCs caused by the immune microenvironment, thereby improving the anti-tumor activity of T cells.
我们的研究表明,PPARα的配体可以降低DC的脂肪酸氧化/氧化磷酸化水平,提高糖酵解水平,恢复DC对CD8+T细胞,特别是抗原特异性CD8+T细胞的功能,同时减少CD4+调节性T细胞的产生,极大的缓解了肿瘤微环境的免疫抑制,从而达到更好的抗肿瘤活性。Our study shows that ligands of PPARα can reduce the level of fatty acid oxidation/oxidative phosphorylation in DC, increase the level of glycolysis, and restore the function of DC on CD8+ T cells, especially antigen-specific CD8+ T cells, while reducing CD4+ The generation of regulatory T cells greatly relieves the immunosuppression of the tumor microenvironment, thereby achieving better anti-tumor activity.
参考文献:references:
1 Paul,S.&Schaefer,B.C.A new look at T cell receptor signaling to nuclear factor-κB.Trends in immunology 34,269-281,doi:https://doi.org/10.1016/j.it.2013.02.002(2013).1 Paul, S. & Schaefer, BCA new look at T cell receptor signaling to nuclear factor-κB. Trends in immunology 34, 269-281, doi: https://doi.org/10.1016/j.it.2013.02.002(2013) .
2 Berard,M.,Brandt,K.,Paus,S.B.&Tough,D.F.IL-15 Promotes the Survival of Naive and Memory Phenotype CD8<sup>+</sup>T Cells.The Journal of Immunology 170,5018-5026,doi:10.4049/jimmunol.170.10.5018(2003).2 Berard, M., Brandt, K., Paus, SB&Tough, DFIL-15 Promotes the Survival of Naive and Memory Phenotype CD8<sup>+</sup>T Cells. The Journal of Immunology 170,5018-5026, doi: 10.4049/jimmunol.170.10.5018 (2003).
3 Wan,Y.Y.,Chi,H.,Xie,M.,Schneider,M.D.&Flavell,R.A.The kinase TAK1 integrates antigen and cytokine receptor signaling for T cell development,survival and function.Nature Immunology 7,851,doi:10.1038/ni1355 https://www.nature.com/articles/ni1355#supplementary-information(2006).3 Wan, YY, Chi, H., Xie, M., Schneider, MD & Flavell, RA //www.nature.com/articles/ni1355#supplementary-information(2006).
4 Ajibade,A.A.,Wang,H.Y.&Wang,R.-F.Cell type-specific function of TAK1 in innate immune signaling.Trends in immunology 34,307-316,doi:https://doi.org/10.1016/j.it.2013.03.007(2013).4 Ajibade,AA,Wang,HY&Wang,R.-F.Cell type-specific function of TAK1 in innate immune signaling.Trends in immunology 34,307-316,doi:https://doi.org/10.1016/j.it. 2013.03.007 (2013).
5 Inokuchi-Shimizu,S.et al.TAK1-mediated autophagy and fatty acid oxidation prevent hepatosteatosis and tumorigenesis.The Journal of clinical investigation 124,3566-3578,doi:10.1172/JCI74068(2014).5 Inokuchi-Shimizu, S. et al. TAK1-mediated autophagy and fatty acid oxidation prevent hepatosteatosis and tumorigenesis. The Journal of clinical investigation 124, 3566-3578, doi:10.1172/JCI74068(2014).
6 Dunn,S.E.et al.Peroxisome proliferator–activated receptor(PPAR)αexpression in T cells mediates gender differences in development of T cell–mediated autoimmunity.The Journal of experimental medicine 204,321-330,doi:10.1084/jem.20061839(2007).6 Dunn, SE et al. Peroxisome proliferator–activated receptor(PPAR)αexpression in T cells mediates gender differences in development of T cell–mediated autoimmunity. The Journal of experimental medicine 204,321-330,doi:10.1084/jem.20061839(2007) .
7 Bansal,T.et al.Arjunolic acid,a peroxisome proliferator-activated receptor alpha agonist,regresses cardiac fibrosis by inhibiting non-canonical TGF-beta signaling.The Journal of biological chemistry 292,16440-16462,doi:10.1074/jbc.M117.788299(2017).7 Bansal, T. et al. Arjunolic acid, a peroxisome proliferator-activated receptor alpha agonist, regresses cardiac fibrosis by inhibiting non-canonical TGF-beta signaling. The Journal of biological chemistry 292, 16440-16462, doi:10.1074/jbc. M117.788299 (2017).
8 Zhang,Y.et al.Enhancing CD8+T Cell Fatty Acid Catabolism within a Metabolically Challenging Tumor Microenvironment Increases the Efficacy of Melanoma Immunotherapy.Cancer cell 32,377-391.e379,doi:https://doi.org/10.1016/j.ccell.2017.08.004(2017).8 Zhang, Y. et al. Enhancing CD8+T Cell Fatty Acid Catabolism within a Metabolically Challenging Tumor Microenvironment Increases the Efficacy of Melanoma Immunotherapy. Cancer cell 32,377-391.e379,doi:https://doi.org/10.1016/j .ccell.2017.08.004(2017).
9 Chamoto,K.et al.Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity.Proceedings of the National Academy of Sciences 114,E761-E770,doi:10.1073/pnas.1620433114(2017).9 Chamoto, K. et al. Mitochondrial activation chemicals synergize with surface receptor PD-1 blockade for T cell-dependent antitumor activity. Proceedings of the National Academy of Sciences 114, E761-E770, doi:10.1073/pnas.1620433114(2017) .
10 Zhao,F.et al.Paracrine Wnt5a-β-Catenin Signaling Triggers a Metabolic Program that Drives Dendritic Cell Tolerization.Immunity 48,147-160.e147(2018).doi.org/10.1016/j.immuni.2017.12.00410 Zhao, F. et al. Paracrine Wnt5a-β-Catenin Signaling Triggers a Metabolic Program that Drives Dendritic Cell Tolerization. Immunity 48, 147-160.e147(2018).doi.org/10.1016/j.immuni.2017.12.004
11 Malinarich,F.et al.High mitochondrial respiration and glycolytic capacity represent a metabolic phenotype of human tolerogenic dendritic cells.J Immunol 194,5174-5186(2015).DOI:https://doi.org/10.4049/jimmunol.130331611 Malinarich, F. et al. High mitochondrial respiration and glycolytic capacity represent a metabolic phenotype of human tolerogenic dendritic cells. J Immunol 194, 5174-5186 (2015). DOI: https://doi.org/10.4049/jimmunol.1303316
12 Xu,H.E.et al.Structural determinants of ligand binding selectivity between the peroxisome proliferator-activated receptors.Proceedings Of the National Academy Of Sciences Of the United States Of America 98,13919-13924(2001).doi.org/10.1073/pnas.24141019812 Xu, HE et al. Structural determinants of ligand binding selectivity between the peroxisome proliferator-activated receptors. Proceedings Of the National Academy Of Sciences Of the United States Of America 98, 13919-13924(2001).doi.org/10.1073/pnas .241410198
13 Forman,B.M.et al.Hypolipidemic drugs,polyunsaturated fatty acids,and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta.Proceedings Of the National Academy Of Sciences Of the United States Of America 94,4312-4317(1997).doi.org/10.1073/pnas.94.9.431213 Forman, BM et al. Hypolipidemic drugs, polyunsaturatedfatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proceedings Of the National Academy Of Sciences Of the United States Of America 94,4312-4317(1997). doi.org/10.1073/pnas.94.9.4312
14 Yamazaki,K.et al.Microarray analysis of gene expression changes in mouse liver induced by peroxisome proliferator activated receptor alpha agonists.Biochemical And Biophysical Research Communications 290,1114-1122(2002).doi.org/10.1006/bbrc.2001.631914 Yamazaki, K. et al. Microarray analysis of gene expression changes in mouse liver induced by peroxisome proliferator activated receptor alpha agonists. Biochemical And Biophysical Research Communications 290, 1114-1122 (2002).doi.org/10.1006/bbrc.2001.6319
15 Issemann,I.et al.Activation Of a Member Of the Steroid-Hormone Receptor Superfamily by Peroxisome Proliferators.Nature 347,645-650(1990).doi:10.1038/347645a0.15 Issemann, I. et al. Activation Of a Member Of the Steroid-Hormone Receptor Superfamily by Peroxisome Proliferators. Nature 347, 645-650 (1990). doi: 10.1038/347645a0.
发明内容SUMMARY OF THE INVENTION
本发明首先涉及PPARα(过氧化物酶体增殖物激活受体α)配体在制备药物中的应用,所述药物的功能包括如下功能的任意一种或多种,The present invention first relates to the application of PPARα (peroxisome proliferator-activated receptor α) ligands in the preparation of medicines, and the functions of the medicines include any one or more of the following functions,
(1)激活T细胞内的活化信号,所述的活化信号是指JNK磷酸化水平上升,所述的T细胞优选为CD8+T细胞;(1) Activating the activation signal in the T cell, the activation signal refers to an increase in the phosphorylation level of JNK, and the T cell is preferably a CD8+ T cell;
(2)激活T细胞,所述的激活T细胞指使T细胞分泌更多的IFN-γ和IL-2且同时增加T细胞的存活时间,所述的T细胞优选为CD8+T细胞,更优选的,为肿瘤浸润性CD8+T细胞;(2) Activating T cells, the activation of T cells refers to making T cells secrete more IFN-γ and IL-2 and at the same time increase the survival time of T cells, the T cells are preferably CD8+ T cells, more preferably , for tumor-infiltrating CD8+ T cells;
(3)促进T细胞分化,所述的分化指促进T细胞分化为记忆性T细胞;(3) promoting the differentiation of T cells, the differentiation refers to promoting the differentiation of T cells into memory T cells;
(4)调节树突状细胞(Dendritic Cell,DC细胞)的代谢状态,所述的调节代谢是指:下调DC细胞的脂肪酸氧化/氧化磷酸化水平,并上调糖酵解水平;所述的DC为骨髓来源的DC,更优选的,为浸润在肿瘤微环境中的DC;(4) Regulating the metabolic state of dendritic cells (Dendritic Cell, DC cells), the regulating metabolism refers to: down-regulating the fatty acid oxidation/oxidative phosphorylation level of DC cells, and up-regulating the level of glycolysis; the DC are bone marrow-derived DCs, more preferably, DCs infiltrating the tumor microenvironment;
(5)恢复DC对T细胞的调控功能,所述的恢复DC的调控功能指:下调CD4+T细胞转变为调节性T细胞的比例、增加肿瘤杀伤性CD8+T细胞的比例、激活肿瘤杀伤性CD8+T细胞的功能;所述的激活肿瘤杀伤性CD8+T细胞指:使肿瘤杀伤性CD8+T细胞分泌更多的IFN-γ和IL-2且同时增加肿瘤杀伤性CD8+T细胞的存活时间;(5) Restoring the regulatory function of DC on T cells, the said restoring the regulatory function of DC refers to: down-regulating the ratio of CD4+ T cells to regulatory T cells, increasing the ratio of tumor-killing CD8+ T cells, and activating tumor-killing The function of CD8+ T cells; the activation of tumor-killing CD8+ T cells refers to: making tumor-killing CD8+ T cells secrete more IFN-γ and IL-2 while increasing tumor-killing CD8+ T cells survival time;
(6)提高免疫检查点抑制剂的抗肿瘤效果,所述的免疫检查点抑制剂优选为PD-L1/PD-1抗体,优选的,所述的肿瘤为免疫检查点抑制剂治疗有效的肿瘤;(6) Improve the anti-tumor effect of an immune checkpoint inhibitor, the immune checkpoint inhibitor is preferably a PD-L1/PD-1 antibody, and preferably, the tumor is a tumor that is effectively treated by an immune checkpoint inhibitor ;
(7)提高抗肿瘤疫苗的治疗效果,所述的抗肿瘤疫苗优选为通过PEG2000-DSPE胶束装载多肽/蛋白肿瘤抗原的纳米疫苗。(7) To improve the therapeutic effect of anti-tumor vaccine, the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigen through PEG2000-DSPE micelles.
所述的PPARα配体优选为PPARα的抑制剂,更优选的,所述的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。The PPARα ligand is preferably an inhibitor of PPARα, more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
本发明还涉及一组PPARα的抑制剂,所述的抑制剂为ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。The present invention also relates to a group of PPARα inhibitors, the inhibitors are ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
本发明还涉及一种治疗肿瘤的方法,所述的方法为,同时给予治疗有效量的免疫检查点抑制剂和PPARα配体;The present invention also relates to a method for treating tumors, which comprises administering a therapeutically effective amount of an immune checkpoint inhibitor and a PPARα ligand at the same time;
优选的,所述的肿瘤为免疫检查点抑制剂治疗有效的肿瘤,所述的免疫检查点抑制剂为PD-L1/PD-1抗体;Preferably, the tumor is a tumor that is effectively treated by an immune checkpoint inhibitor, and the immune checkpoint inhibitor is a PD-L1/PD-1 antibody;
优选的,所述的PPARα配体优选为PPARα的抑制剂,更优选的,所述的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。Preferably, the PPARα ligand is preferably an inhibitor of PPARα, more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
本发明还涉及一种治疗肿瘤的方法,所述的方法为,同时给予治疗有效量的抗肿瘤疫苗和PPARα配体;The present invention also relates to a method for treating tumors, which comprises administering a therapeutically effective amount of an anti-tumor vaccine and a PPARα ligand at the same time;
优选的,所述的抗肿瘤疫苗优选为通过PEG2000-DSPE胶束装载多肽/蛋白肿瘤抗原的纳米疫苗,所述的胶束的制备方法可参照CN201410570624的记载;Preferably, the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigens through PEG2000-DSPE micelles, and the preparation method of the micelles can refer to the records of CN201410570624;
优选的,所述的PPARα配体优选为PPARα的抑制剂,更优选的,所述的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。Preferably, the PPARα ligand is preferably an inhibitor of PPARα, more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
本发明还涉及一种抗肿瘤联用制剂,所述的制剂包含:The present invention also relates to an anti-tumor combined preparation comprising:
(1)治疗有效量PPARα配体;(1) A therapeutically effective amount of PPARα ligand;
(2)治疗有效量的免疫检查点抑制剂或治疗有效量的抗肿瘤疫苗;(2) A therapeutically effective amount of an immune checkpoint inhibitor or a therapeutically effective amount of an antitumor vaccine;
(3)必要的药用辅料。(3) Necessary pharmaceutical excipients.
优选的,所述的免疫检查点抑制剂为PD-L1/PD-1抗体;Preferably, the immune checkpoint inhibitor is PD-L1/PD-1 antibody;
优选的,所述的抗肿瘤疫苗优选为通过PEG2000-DSPE胶束装载多肽/蛋白肿瘤抗原的纳米疫苗;Preferably, the anti-tumor vaccine is preferably a nano-vaccine loaded with polypeptide/protein tumor antigens through PEG2000-DSPE micelles;
优选的,所述的PPARα配体优选为PPARα的抑制剂,更优选的,所述的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。Preferably, the PPARα ligand is preferably an inhibitor of PPARα, more preferably, the inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330 , ZM0332, ZM0333.
附图说明Description of drawings
图1、T细胞中PPARα抑制剂GW6471增强T细胞活化相关激酶的磷酸化【体外实验】。Figure 1. The PPARα inhibitor GW6471 in T cells enhances the phosphorylation of T cell activation-related kinases [in vitro experiments].
图2、PPARα抑制剂GW6471增强CD8+T细胞的存活及功能【体外实验】。Figure 2. The PPARα inhibitor GW6471 enhances the survival and function of CD8+ T cells [in vitro experiment].
图3、PPARα抑制剂GW6471增强PD-L1抗体对MC38-OT I的抗肿瘤效果。Figure 3. PPARα inhibitor GW6471 enhances the anti-tumor effect of PD-L1 antibody on MC38-OT I.
图4、PPARα抑制剂GW6471延长PD-L1抗体治疗的MC38-OT I荷瘤小鼠生存期。Figure 4. The PPARα inhibitor GW6471 prolongs the survival of PD-L1 antibody-treated MC38-OT I tumor-bearing mice.
图5、PPARα抑制剂GW6471增强PD-1抗体(PD-1 mAb)对MC38的抗肿瘤效果Figure 5. PPARα inhibitor GW6471 enhances the anti-tumor effect of PD-1 antibody (PD-1 mAb) on MC38
图6、PPARα抑制剂GW6471对肿瘤浸润性T淋巴细胞功能及活性的影响。Figure 6. Effect of PPARα inhibitor GW6471 on the function and activity of tumor-infiltrating T lymphocytes.
图7、PPARα抑制剂GW6471增加PD-L1抗体诱导产生的记忆性CD8+T细胞比例。Figure 7. PPARα inhibitor GW6471 increases the proportion of memory CD8+ T cells induced by PD-L1 antibody.
图8、PPRAα抑制剂GW6471的抗肿瘤作用主要依赖于CD8+T细胞。Figure 8. The antitumor effect of PPRAα inhibitor GW6471 mainly depends on CD8+ T cells.
图9、PPARα抑制剂GW6471联合PD-L1抗体的抗肿瘤作用主要依赖于DC细胞。Figure 9. The anti-tumor effect of PPARα inhibitor GW6471 combined with PD-L1 antibody mainly depends on DC cells.
图10、PPARα-/-DC的抗肿瘤作用。Figure 10. Antitumor effect of PPARα-/-DC.
图11、PPARα抑制剂GW6471增强肿瘤浸润性DC对CD8+T细胞的活化。Figure 11. The PPARα inhibitor GW6471 enhances the activation of CD8+ T cells by tumor-infiltrating DCs.
图12、PPARα-/-的肿瘤浸润性DC增强肿瘤抗原特异性CD8+T细胞的比例和活性。Figure 12. Tumor-infiltrating DCs of PPARα-/- enhance the proportion and activity of tumor antigen-specific CD8+ T cells.
图13、PPARα抑制剂GW6471对DC代谢状态的调控【体外实验】。Figure 13. Regulation of DC metabolic state by PPARα inhibitor GW6471 [in vitro experiment].
图14、PPARα抑制剂GW6471抑制肿瘤外泌体对调节性T细胞的诱导【体外实验】。Figure 14. The PPARα inhibitor GW6471 inhibits the induction of regulatory T cells by tumor exosomes [in vitro experiment].
图15、PPARα抑制剂GW6471恢复DC对CD8+T细胞的活化能力【体外实验】。Figure 15. The PPARα inhibitor GW6471 restores the ability of DC to activate CD8+ T cells [in vitro experiment].
图16、PPARα抑制剂GW6471增强E7疫苗对TC-1的抗肿瘤效果。Figure 16. PPARα inhibitor GW6471 enhances the anti-tumor effect of E7 vaccine on TC-1.
图17、PPARα抑制剂GW6471延长E7疫苗治疗的TC-1荷瘤小鼠生存期。Figure 17. The PPARα inhibitor GW6471 prolongs the survival of E7 vaccine-treated TC-1 tumor-bearing mice.
图18、T细胞中PPARα与TAK1相互作用机制图。Figure 18. Diagram of the mechanism of interaction between PPARα and TAK1 in T cells.
图19、qRT-PCR检测GW6471及其结构类似物对PPARα控制的下游靶基因Cpt1a表达的影响。Figure 19. The effects of GW6471 and its structural analogs on the expression of the downstream target gene Cpt1a controlled by PPARα were detected by qRT-PCR.
具体实施方式detailed description
小鼠模型:mouse model:
1、CD11c-DTR小鼠模型,该模型的特点是该小鼠的CD11c基因(该基因为DC细胞的表面标志物)前面嵌入了一个白喉毒素响应原件(DTR,DT:白喉毒素,R:响应原件),在腹腔注射该毒素的情况下,小鼠骨髓来源的CD11c阳性DC细胞将会被清除。构建该模型是因为DC的删除实验中,使用抗体的方法效率很低,故采取转基因小鼠的方式。1. CD11c-DTR mouse model, which is characterized by the presence of a diphtheria toxin response element (DTR, DT: diphtheria toxin, R: response Original), in the case of intraperitoneal injection of this toxin, CD11c-positive DC cells derived from mouse bone marrow will be eliminated. This model was constructed because the method of using antibodies in the DC deletion experiment was very inefficient, so the method of transgenic mice was adopted.
2、小鼠CD11c-DTR/PPARα-/-嵌合骨髓转移模型,即在第一个模型的基础上进一步敲除小鼠基因组中的PPARα,其特点是,同时转移等量的CD11c-DTR骨髓细胞和PPARα-/-骨髓细胞之后,小鼠经白喉毒素处理时,体内将只剩下PPARα-/-的DC细胞,而没有野生型PPARα功能的DC细胞。在这种情况下,无需抗体或小分子药物治疗,仅观察肿瘤的生长便可以考察DC细胞中PPARα信号通路对肿瘤生长、以及在抗肿瘤免疫中起到的作用。2. The mouse CD11c-DTR/PPARα-/- chimeric bone marrow transfer model, that is, on the basis of the first model, the PPARα in the mouse genome is further knocked out, which is characterized by the simultaneous transfer of the same amount of CD11c-DTR bone marrow Cells and PPARα-/- myeloid cells, when mice were treated with diphtheria toxin, only PPARα-/- DC cells remained in the body, but no wild-type PPARα function DC cells. In this case, the role of the PPARα signaling pathway in DC cells on tumor growth and in anti-tumor immunity can be investigated without the need for antibody or small molecule drug treatment, and only by observing tumor growth.
实施例一:T细胞中PPARα抑制剂GW6471增强T细胞活化相关激酶的磷酸化Example 1: The PPARα inhibitor GW6471 enhances the phosphorylation of T cell activation-related kinases in T cells
细胞培养及处理方法:Cell culture and processing methods:
(1)野生型C57BL6小鼠处死后,取脾脏细胞,裂解红细胞,之后用CD8磁珠(StemCell公司)分离CD8+T细胞。(1) After the wild-type C57BL6 mice were sacrificed, spleen cells were taken, red blood cells were lysed, and CD8+ T cells were separated by CD8 magnetic beads (StemCell).
(2)使用不同浓度的Fenofibrate(20μM)或GW6471(10μM)处理CD8+T细胞20分钟后,再使用2.5ug/ml anti-CD3抗体和0.5ug/ml anti-CD28抗体激活T细胞20分钟。收集CD8+T细胞,并用Lysis Buffer冰上裂解细胞,提取胞内蛋白,进行WesternBlot实验,考察PPARα、JNK磷酸化和I-kB降解的变化。(2) After CD8+ T cells were treated with different concentrations of Fenofibrate (20μM) or GW6471 (10μM) for 20 minutes, 2.5ug/ml anti-CD3 antibody and 0.5ug/ml anti-CD28 antibody were used to activate T cells for 20 minutes. CD8+ T cells were collected, and the cells were lysed with Lysis Buffer on ice, and the intracellular proteins were extracted. Western Blot experiments were performed to investigate the changes of PPARα, JNK phosphorylation and I-kB degradation.
结果如图1所示,与单独anti-CD3和anti-CD28抗体激活组相比,GW6471(结构如下式1所示)和Fenofibrate预处理组的T细胞胞内呈现出更强的JNK磷酸化水平,而I-kB的降解并未变的更强烈。同时胞浆中PPARα的含量显著降低,说明经GW6471和Fenofibrate预处理后,PPARα很有可能发生构象改变,脱离与TAK1的物理结合,转而与共刺激因子/共抑制因子形成转录调控复合物进入细胞核中,并行使其它的功能。该结果很好的证实了我们的推测,即PPARα的配体处理能够解除PPARα与TAK1的物理结合,进而释放TAK1,激活其下游激酶的功能,促进T细胞活化、存活甚至向记忆性T细胞分化。The results are shown in Figure 1. Compared with the anti-CD3 and anti-CD28 antibody activation groups alone, the T cells in the GW6471 (structure shown in Formula 1 below) and Fenofibrate pretreatment groups showed stronger JNK phosphorylation levels in the cells. , while the degradation of I-kB did not become more intense. At the same time, the content of PPARα in the cytoplasm was significantly reduced, indicating that after pretreatment with GW6471 and Fenofibrate, PPARα is likely to undergo a conformational change, break away from the physical association with TAK1, and instead form a transcriptional regulatory complex with co-stimulators/co-repressors and enter the nucleus. , and perform other functions in parallel. This result well confirms our speculation that ligand treatment of PPARα can release the physical binding of PPARα and TAK1, thereby releasing TAK1, activating its downstream kinase function, and promoting T cell activation, survival and even differentiation into memory T cells. .
Figure PCTCN2021117407-appb-000001
Figure PCTCN2021117407-appb-000001
实施例二:PPARα抑制剂GW6471和激动剂Fenofibrate增强CD8+T细胞的存活和功能Example 2: PPARα inhibitor GW6471 and agonist Fenofibrate enhance the survival and function of CD8+ T cells
细胞培养及处理方法:Cell culture and processing methods:
(1)野生型C57BL6小鼠处死后,取脾脏细胞,裂解红细胞,之后用CD8a磁珠(StemCell公司)分离CD8+T细胞。(1) After the wild-type C57BL6 mice were sacrificed, spleen cells were taken, red blood cells were lysed, and then CD8+ T cells were separated with CD8a magnetic beads (StemCell).
(2)使用不同浓度的Fenofibrate(20μM)或GW6471(10μM)处理CD8+T细胞24、48、72小时后,收取细胞,并再使用0.5ug/ml anti-CD3抗体和0.1ug/ml anti-CD28抗体激活CD8+T细胞72小时后,收取细胞培养上清,ELISA检测上清中IFN-γ和IL-2的水平。(2) After CD8+ T cells were treated with different concentrations of Fenofibrate (20μM) or GW6471 (10μM) for 24, 48, and 72 hours, the cells were harvested, and 0.5ug/ml anti-CD3 antibody and 0.1ug/ml anti-CD3 antibody were used again. 72 hours after CD28 antibody activation of CD8+ T cells, the cell culture supernatant was collected, and the levels of IFN-γ and IL-2 in the supernatant were detected by ELISA.
(3)使用不同浓度的Fenofibrate、GW6471、类似物处理CD8+T细胞24小时后,收取细胞,并使用Trizol法抽提mRNA,反转录之后,进行qRT-PCR检测Cpt1a的表达量。(3) After treating CD8+ T cells with different concentrations of Fenofibrate, GW6471, and analogs for 24 hours, the cells were harvested, and mRNA was extracted by Trizol method. After reverse transcription, qRT-PCR was performed to detect the expression of Cpt1a.
如图2的结果显示,通过预处理GW6471之后,CD8+T细胞的状态得到调整,经过再刺激后,能够分泌更多的IFN-γ和IL-2。特别是预处理24小时组的CD8+T细胞,能分泌3倍于未经预处理然后再刺激组CD8+T细胞所产生的IFN-γ,而预处理72小时组的CD8+T细胞能够被激活并产生更多维持T细胞存活的IL-2。而预处理Fenofibrate 24、48和72小时之后的CD8+T细胞能够产生更多的IFN-γ,但是没有更多的IL-2产生。The results in Figure 2 show that after pretreatment with GW6471, the state of CD8+ T cells was adjusted, and after restimulation, more IFN-γ and IL-2 could be secreted. In particular, the CD8+ T cells in the 24-hour pretreatment group could secrete 3 times the IFN-γ produced by the CD8+ T cells in the unpretreated and restimulated group, while the CD8+ T cells in the 72-hour pretreatment group could be stimulated by Activates and produces more IL-2 that maintains T cell survival. In contrast, CD8+ T cells pretreated with Fenofibrate for 24, 48 and 72 hours were able to produce more IFN-γ, but not IL-2.
进一步的,我们使用了一组GW6471化合物的结构类似物(表1)对CD8+T细胞重复上述刺激试验。结果显示,GW6471及其部分类似物的处理既能够增强CD8+T细胞的功能,还能够促进其长时间存活,而Fenofibrate处理能够增强CD8+T细胞的功能,但不影响CD8+T细胞的增殖和存活(表2)。此外,为了验证GW6471的结构类似物(表1)是否具有与GW6471类似的功能,我们使用qRT-PCR的方法检测了这些结构类似物(表1)如何影响PPARα控制的下游靶基因Cpt1a的表达,其中Fenofibrate为PPARα激动剂对照,GW6471为PPARα抑制剂对照。如图19所示,GW6471的结构类似物(表1)均能够抑制Cpt1a的表达,显示它们能够通过与GW6471类似的机制抑制PPARα的活性及改变PPARα与TAK1物理结合的能力。Further, we repeated the above stimulation experiments on CD8+ T cells using a panel of structural analogs of the GW6471 compound (Table 1). The results showed that the treatment of GW6471 and some of its analogs could not only enhance the function of CD8+ T cells, but also promote their long-term survival, while Fenofibrate treatment could enhance the function of CD8+ T cells, but did not affect the proliferation of CD8+ T cells. and survival (Table 2). In addition, to verify whether the structural analogs of GW6471 (Table 1) have similar functions to GW6471, we used qRT-PCR to examine how these structural analogs (Table 1) affected the expression of the PPARα-controlled downstream target gene Cpt1a, Among them, Fenofibrate is a PPARα agonist control, and GW6471 is a PPARα inhibitor control. As shown in Figure 19, the structural analogs of GW6471 (Table 1) were all able to inhibit the expression of Cpt1a, indicating that they could inhibit the activity of PPARα and change the ability of PPARα to physically bind to TAK1 through a mechanism similar to that of GW6471.
表1、GW6471结构类似物的结构示意图Table 1. Schematic diagram of the structural analogs of GW6471
Figure PCTCN2021117407-appb-000002
Figure PCTCN2021117407-appb-000002
Figure PCTCN2021117407-appb-000003
Figure PCTCN2021117407-appb-000003
Figure PCTCN2021117407-appb-000004
Figure PCTCN2021117407-appb-000004
Figure PCTCN2021117407-appb-000005
Figure PCTCN2021117407-appb-000005
Figure PCTCN2021117407-appb-000006
Figure PCTCN2021117407-appb-000006
表2、PPARα配体对CD8+T细胞的影响Table 2. Effects of PPARα ligands on CD8+ T cells
PPARα配体名称/编号PPARα ligand name/number IC 50 IC50 EC 200 EC 200
GW6471GW6471 34.61uM34.61uM 0.0022uM0.0022uM
FenofibrateFenofibrate >40uM>40uM 0.0076uM0.0076uM
ZM0282ZM0282 31.49uM31.49uM 0.0058uM0.0058uM
ZM0283ZM0283 0.3537uM0.3537uM 0.0087uM0.0087uM
ZM0284ZM0284 7.495uM7.495uM 0.0258uM0.0258uM
ZM0285ZM0285 0.1303uM0.1303uM 0.7892uM0.7892uM
ZM0286ZM0286 12.71uM12.71uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0325ZM0325 30.9uM30.9uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0326ZM0326 2.417uM2.417uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0329ZM0329 40.96uM40.96uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0330ZM0330 8.423uM8.423uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0332ZM0332 >40uM>40uM 致死剂量以内无法达到Unable to reach within lethal dose
ZM0333ZM0333 >40uM>40uM 致死剂量以内无法达到Unable to reach within lethal dose
EC 200:与不处理组相比,使IFN-γ产量加倍的最小剂量 EC200 : Minimum dose to double IFN-γ production compared to no treatment group
IC 50:半数致死剂量。 IC 50 : median lethal dose.
实验例三:典型的PPARα抑制剂GW6471增强PD-1/PD-L1抗体对MC38-OT I的抗肿瘤效果、激活肿瘤浸润性T淋巴细胞的功能,增加记忆性T细胞比例并延长小鼠的存活期Experimental example 3: The typical PPARα inhibitor GW6471 enhances the anti-tumor effect of PD-1/PD-L1 antibody on MC38-OT I, activates the function of tumor-infiltrating T lymphocytes, increases the proportion of memory T cells, and prolongs the survival of mice. Survival period
细胞培养及动物模型建模方法:Cell culture and animal model modeling methods:
(1)MC38-OT I细胞(自带抗原ovalbumin257-264,便于检测能够识别肿瘤抗原特异性T淋巴细胞)的复苏及培养:使用DMEM培养基(含10%胎牛血清)。将MC38-OT I肿瘤细胞冻存管从液氮保存罐中取出,立即放入37℃水浴中快速溶解,然后将细胞悬液移入含10ml培养基的离心桶中,900rpm离心5分钟后去除上清,用新鲜培养基重悬后,将细胞转移到细胞培养瓶中,加入10-15ml培养基混悬沉淀细胞,调整细胞浓度后,置于37℃、体积分数5%CO 2饱和湿度培养箱中培养。在维持培养过程中,每天观察细胞状态并及时更换新鲜培养基。当细胞贴壁生长至90%汇合度时,用0.05%胰酶消化,按照1:3的比例进行传代培养。实验当天,将生长状态良好,汇合度达到90%的细胞胰酶消化后,用新鲜培养基中和胰酶,900rpm离心,弃上清后加入无菌PBS重悬,计数,将细胞悬液的密度调整为5×10 6/ml待用。 (1) Recovery and culture of MC38-OT I cells (with their own antigen ovalbumin257-264, which is easy to detect and recognize tumor antigen-specific T lymphocytes): use DMEM medium (containing 10% fetal bovine serum). The MC38-OT I tumor cell cryopreservation tube was taken out from the liquid nitrogen storage tank, immediately placed in a 37°C water bath to dissolve rapidly, and then the cell suspension was transferred into a centrifuge bucket containing 10ml of culture medium, centrifuged at 900rpm for 5 minutes, and then removed. After resuspending with fresh medium, transfer the cells to a cell culture flask, add 10-15ml medium to suspend the pelleted cells, adjust the cell concentration, and place it in a 37°C, volume fraction 5% CO 2 saturated humidity incubator cultivated in. During the maintenance culture, the cell status was observed every day and fresh medium was replaced in time. When the cells adhered to 90% confluence, they were digested with 0.05% trypsin and subcultured at a ratio of 1:3. On the day of the experiment, trypsinize the cells that grow well and reach 90% confluence, neutralize the trypsin with fresh medium, centrifuge at 900 rpm, discard the supernatant, add sterile PBS to resuspend, count, and count the cells in the suspension. The density was adjusted to 5×10 6 /ml for use.
(2)接种肿瘤细胞:皮下接种雌性野生型C57小鼠,每只小鼠0.1ml(每只小鼠接种50×10 4个MC38-OTI细胞)。 (2) Inoculation of tumor cells: female wild-type C57 mice were subcutaneously inoculated with 0.1 ml per mouse (50×10 4 MC38-OTI cells were inoculated per mouse).
实验方案(一):PPARα抑制剂GW6471增强PD-L1抗体(PD-L1 mAb)对MC38-OT I的抗肿瘤效果及延长了小鼠的存活期Experimental protocol (1): PPARα inhibitor GW6471 enhances the anti-tumor effect of PD-L1 antibody (PD-L1 mAb) on MC38-OT I and prolongs the survival of mice
(1)采用鼠源结肠癌细胞系MC38-OT I(自带抗原ovalbumin257-264,便于检测能够识别肿瘤抗原特异性T淋巴细胞),对C57小鼠进行皮下接种,约在5天左右可测量到肿瘤形成(平均肿瘤体积约为50mm 3)。 (1) The mouse-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is easy to detect and recognize tumor antigen-specific T lymphocytes) was used to subcutaneously inoculate C57 mice, which can be measured in about 5 days to tumor formation (average tumor volume was approximately 50 mm 3 ).
(2)荷瘤小鼠组别设计,试验共分4组,分别为(2) Group design of tumor-bearing mice, the experiment was divided into 4 groups, respectively
a)荷瘤小鼠对照组(CTR);a) tumor-bearing mice control group (CTR);
b)荷瘤小鼠抗体治疗组(PD-L1 mAb);b) Antibody treatment group of tumor-bearing mice (PD-L1 mAb);
c)荷瘤小鼠给药组(GW6471);c) Tumor-bearing mice administration group (GW6471);
d)荷瘤小鼠联合治疗组(GW6471+PD-L1 mAb);d) Tumor-bearing mice combined treatment group (GW6471+PD-L1 mAb);
试验荷瘤小鼠共4组,每组6只。A total of 4 groups of tumor-bearing mice were tested, with 6 mice in each group.
(3)治疗方案:如图3a所示,按接种为0天计算,在接种后第7,10和17天对小鼠进行PD-L1抗体治疗,200ug每只小鼠,腹腔注射(PD-L1 mAb稀释于PBS中,浓度为2mg/ml,每只小鼠腹腔注射0.1ml)。与此同时,在接种后第11天开始对小鼠给与GW6471治疗,10mg/kg,腹腔注射,隔一天给与一次GW6471,共10次(GW6471储液为25mg/ml溶解于DMSO中,待给药前,稀释于55℃预热的PBS中至完全溶解,浓度为2mg/ml,每只小鼠腹腔注射0.1ml)。(3) Treatment scheme: As shown in Figure 3a, according to the day of inoculation, the mice were treated with PD-L1 antibody on the 7th, 10th and 17th days after inoculation, 200ug per mouse, intraperitoneal injection (PD-L1 antibody) L1 mAb was diluted in PBS at a concentration of 2 mg/ml, and each mouse was injected intraperitoneally with 0.1 ml). At the same time, the mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 11th day after inoculation, and GW6471 was given once every other day, for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 0.1 ml).
(4)肿瘤生长及生存期统计:在给药期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线,记录小鼠死亡情况。(4) Statistics of tumor growth and survival period: During the administration period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn, and the small size was recorded. Mouse death.
结果如图3b所示,相比于对照组,GW6471治疗组小鼠肿瘤生长趋势有轻微的减缓,但是效果并不显著。PD-L1抗体组小鼠肿瘤体积虽然也得到缓解,但在此治疗的基础上,联合GW6471可显著减缓MC38-OT I肿瘤的生长。与此同时如图4所示,联合GW6471和PD-L1抗体对小鼠生存期的增长也有显著的改善。The results are shown in Figure 3b. Compared with the control group, the tumor growth trend of the mice in the GW6471 treatment group was slightly slowed down, but the effect was not significant. Although the tumor volume of the mice in the PD-L1 antibody group was also relieved, on the basis of this treatment, the combination of GW6471 could significantly slow down the growth of MC38-OT I tumors. At the same time, as shown in Figure 4, the combination of GW6471 and PD-L1 antibody also significantly improved the survival of mice.
实验方案(二):PPARα抑制剂GW6471增强PD-1抗体(PD-1 mAb)对MC38-OTI的抗肿瘤效果Experimental protocol (2): PPARα inhibitor GW6471 enhances the anti-tumor effect of PD-1 antibody (PD-1 mAb) on MC38-OTI
(1)采用鼠源结肠癌细胞系MC38-OT I(自带抗原ovalbumin257-264,便于检测能够识别肿瘤抗原特异性T淋巴细胞),对C57小鼠进行皮下接种,约在5天左右可测量到肿瘤形成(平均肿瘤体积约为50mm 3)。 (1) The mouse-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is easy to detect and recognize tumor antigen-specific T lymphocytes) was used to subcutaneously inoculate C57 mice, which can be measured in about 5 days to tumor formation (average tumor volume was approximately 50 mm 3 ).
(2)荷瘤小鼠组别设计,试验共分3组,分别为(2) Group design of tumor-bearing mice, the experiment is divided into 3 groups, respectively
a)荷瘤小鼠对照组(CTR);a) tumor-bearing mice control group (CTR);
b)荷瘤小鼠抗体治疗组(PD-1 mAb);b) Antibody treatment group of tumor-bearing mice (PD-1 mAb);
c)荷瘤小鼠联合治疗组(GW6471+PD-1 mAb);c) Tumor-bearing mice combined treatment group (GW6471+PD-1 mAb);
试验荷瘤小鼠共3组,每组7只。There were 3 groups of tumor-bearing mice in the experiment, with 7 mice in each group.
(3)治疗方案:按接种为0天计算,在接种后第8和11天对小鼠进行PD-1抗体治疗,50ug每只小鼠,腹腔注射(PD-1 mAb稀释于PBS中,浓度为0.5mg/ml,每只小鼠腹腔注射0.1ml)。与此同时,在接种后第5天开始对小鼠给与GW6471治疗,30mg/kg,每天灌胃给药。GW6471使用研钵研碎,稀释并混悬于0.3%CMC-Na溶液中,达到3mg/ml浓度并置于4℃保存,每天给药前将药物取出,平衡到室温后进行灌胃给药。(3) Treatment plan: Calculated as day 0 of inoculation, mice were treated with PD-1 antibody on the 8th and 11th day after inoculation, 50ug per mouse, intraperitoneal injection (PD-1 mAb was diluted in PBS, the concentration of 0.5mg/ml, each mouse was intraperitoneally injected with 0.1ml). At the same time, the mice were treated with GW6471, 30 mg/kg, orally administered daily on the 5th day after inoculation. GW6471 was ground with a mortar, diluted and suspended in 0.3% CMC-Na solution to reach a concentration of 3 mg/ml and stored at 4°C. The drug was taken out before administration every day, and it was administered by gavage after equilibrating to room temperature.
(4)肿瘤生长及生存期统计:在给药期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线。到接瘤后第28天时,统计肿瘤体积的变化趋势,即将每只小鼠第28天的肿瘤体积与第8天的肿瘤体积对比,计算出变化倍数。如果变化倍数为0,说明小鼠肿瘤已经消除;如果变化倍数大于0小于1,说明小鼠肿瘤得到控制,体积并未进一步增长;变化倍数大于1,说明小鼠肿瘤没有得到控制,继续进展。(4) Statistics of tumor growth and survival period: During the administration period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn. On the 28th day after receiving the tumor, the change trend of the tumor volume was calculated, that is, the tumor volume on the 28th day of each mouse was compared with the tumor volume on the 8th day, and the change fold was calculated. If the change fold is 0, it means that the mouse tumor has been eliminated; if the change fold is greater than 0 and less than 1, it means that the mouse tumor is controlled and the volume has not further increased; if the change fold is greater than 1, the mouse tumor is not controlled and continues to progress.
结果如图5a所示,相比于对照组,PD-1抗体组和PD-1抗体联合GW6471组的小鼠肿瘤生长趋势显著减缓。而PD-1抗体组小鼠平均肿瘤体积在接瘤后第28天显示出一定的复发趋势,而联用组对肿瘤体积的控制依然很好,体现出显著的疗效优势。可以从图5b中观察到类似的趋势,即PD-1抗体联合GW6471可显著减缓MC38-OT I肿瘤的生长。The results are shown in Figure 5a. Compared with the control group, the tumor growth trend of the mice in the PD-1 antibody group and the PD-1 antibody combined with GW6471 group was significantly slowed down. The average tumor volume of the mice in the PD-1 antibody group showed a certain recurrence trend on the 28th day after receiving the tumor, while the combination group still controlled the tumor volume well, showing a significant curative effect advantage. A similar trend can be observed from Figure 5b, that the combination of PD-1 antibody with GW6471 significantly slowed the growth of MC38-OT I tumors.
实验方案(三):PPARα抑制剂GW6471增强肿瘤浸润性T淋巴细胞功能及活性Experimental protocol (3): PPARα inhibitor GW6471 enhances the function and activity of tumor-infiltrating T lymphocytes
(1)构建MC38-OT I荷瘤小鼠模型,待模型构建完毕,分为4组,每组6只小鼠,并分别给与PD-L1抗体和GW6471治疗。此方法完全同实验方案(一)。(1) The MC38-OT I tumor-bearing mouse model was constructed. After the model was constructed, the mice were divided into 4 groups with 6 mice in each group, and were treated with PD-L1 antibody and GW6471 respectively. This method is completely the same as the experimental scheme (1).
(2)在接种后第21天,取各组小鼠肿瘤组织。利用手术剪将组织剪碎后置于2mg/ml的胶原酶Ⅳ中37℃,220rpm消化1h。然后在滤网上对组织进行研磨过滤,离心弃上清后,用PBS重悬,制备成单细胞悬液。(2) On the 21st day after inoculation, the tumor tissue of each group of mice was collected. The tissue was minced with surgical scissors and placed in 2 mg/ml collagenase IV for 1 h at 37°C and 220 rpm. The tissue was then ground and filtered on a filter screen, and the supernatant was discarded by centrifugation, and then resuspended in PBS to prepare a single-cell suspension.
(3)细胞染色:将上述细胞悬液进行荧光染色,配色方案为APC-CD45,PE/Cy7-CD3,APC/Cy7-CD8,PE-Perforin,FITC-Granzyme B,Percp/Cy5.5-IFNγ,BV421-Ki67。其中CD45,CD3和CD8为针对表面抗原的抗体,直接在4℃避光染色30min。其余为针对细胞内抗原的抗体,需将细胞固定过夜后,透膜后进行染色。待染色完成后,清洗细胞后重悬进行流式细胞检测。(3) Cell staining: The above cell suspension was fluorescently stained, and the color scheme was APC-CD45, PE/Cy7-CD3, APC/Cy7-CD8, PE-Perforin, FITC-Granzyme B, Percp/Cy5.5-IFNγ , BV421-Ki67. Among them, CD45, CD3 and CD8 are antibodies against surface antigens, which were directly stained at 4°C for 30 min in the dark. The rest are antibodies against intracellular antigens, which need to be fixed overnight and stained after permeabilization. After staining, the cells were washed and resuspended for flow cytometry.
结果如图6所示,在PD-L1抗体治疗的基础上, GW6471可以显著提高肿瘤浸润性CD8+T淋巴细胞分泌 perforin、Granzyme B和IFNγ的能力即杀伤肿瘤细胞的功能,同时增强了CD8+T细胞的活性(Ki67)The results are shown in Figure 6. On the basis of PD-L1 antibody treatment, GW6471 can significantly improve the ability of tumor-infiltrating CD8+ T lymphocytes to secrete perforin, Granzyme B and IFNγ, that is, the function of killing tumor cells. Activity of T cells (Ki67) .
实验方案(四):PPRAα抑制剂GW6471增加PD-L1抗体诱导产生的记忆性CD8+T细胞比例Experimental protocol (4): PPRAα inhibitor GW6471 increases the proportion of memory CD8+ T cells induced by PD-L1 antibody
(1)构建MC38-OT I荷瘤小鼠模型,待模型构建完毕,同上分为四组,每组6只小鼠,并分别给与PD-L1抗体和GW6471治疗。此方法完全同实验方案(一)。(1) The MC38-OT I tumor-bearing mouse model was constructed. After the model was constructed, the mice were divided into four groups as above, with 6 mice in each group, and were treated with PD-L1 antibody and GW6471 respectively. This method is completely the same as the experimental scheme (1).
(2)在接种后第15天,取各组小鼠肿瘤组织。利用手术剪将组织剪碎后置于2mg/ml的胶原酶Ⅳ中37℃,220rpm消化1h。然后在滤网上对组织进行研磨过滤,离心弃上清后,用PBS重悬,制备成单细胞悬液。(2) On the 15th day after inoculation, the tumor tissue of each group of mice was collected. The tissue was minced with surgical scissors and placed in 2 mg/ml collagenase IV for 1 h at 37°C and 220 rpm. The tissue was then ground and filtered on a filter screen, and the supernatant was discarded by centrifugation, and then resuspended in PBS to prepare a single-cell suspension.
(3)细胞染色:将上述细胞悬液进行荧光染色,配色方案为BV605-CD45,APC/Cy7-CD8,PE-CD62L,FITC-CD44,Percp/Cy5.5-CD3,APC-KLGR1。上述所有抗体均为针对表面抗原的抗体,直接在4℃避光染色30min。待染色完成后,清洗细胞后重悬进行流式细胞检测,并将CD45+CD3+CD8+CD44+KLRG1-CD62L+的细胞视为记忆性CD8+T细胞类群。(3) Cell staining: The above cell suspension was fluorescently stained, and the color scheme was BV605-CD45, APC/Cy7-CD8, PE-CD62L, FITC-CD44, Percp/Cy5.5-CD3, APC-KLGR1. All the above antibodies were antibodies against surface antigens, and were directly stained at 4°C for 30 min in the dark. After staining, the cells were washed and resuspended for flow cytometry, and CD45+CD3+CD8+CD44+KLRG1-CD62L+ cells were regarded as a memory CD8+ T cell population.
结果如图7所示,与对照组相比,单独使用GW6471不能提高肿瘤组织中记忆性CD8+T细胞比例,而PD-L1抗体和GW6471联用PD-L1抗体组均能够显著调高该比例。更重要得是, 在PD-L1抗体治疗的基础上, GW6471可以进一步显著提高肿瘤组织中记忆性CD8+T细胞比例,使荷瘤小鼠长期获益,同时支持了图4的结果,即GW6471与PD-L1抗体联用显著延长荷瘤小鼠的生存期。 The results are shown in Figure 7. Compared with the control group, the use of GW6471 alone cannot increase the proportion of memory CD8+ T cells in the tumor tissue, while the PD-L1 antibody and GW6471 combined with the PD-L1 antibody group can significantly increase the proportion. . More importantly, on the basis of PD-L1 antibody treatment, GW6471 can further significantly increase the proportion of memory CD8+ T cells in tumor tissue , which brings long-term benefits to tumor-bearing mice, and supports the results in Figure 4, that is, GW6471 Combination with PD-L1 antibody significantly prolonged the survival of tumor-bearing mice.
实验方案(五):PPRAα抑制剂GW6471的抗肿瘤作用主要依赖于CD8+T细胞Experimental scheme (5): The anti-tumor effect of PPRAα inhibitor GW6471 mainly depends on CD8+ T cells
(1)同上构建MC38-OT I荷瘤模型。(1) The MC38-OT I tumor-bearing model was constructed as above.
(2)荷瘤小鼠组别设计,试验共分4组,分别为(2) Group design of tumor-bearing mice, the experiment was divided into 4 groups, respectively
a)荷瘤小鼠对照组(CTR);a) tumor-bearing mice control group (CTR);
b)荷瘤小鼠联合治疗组(GW6471+PD-L1 mAb);b) tumor-bearing mice combined treatment group (GW6471+PD-L1 mAb);
c)荷瘤小鼠删除CD4组(GW6471+PD-L1 mAb+α-CD4 Ab)c) CD4 deletion group in tumor-bearing mice (GW6471+PD-L1 mAb+α-CD4 Ab)
d)荷瘤小鼠删除CD8组(GW6471+PD-L1 mAb+α-CD8 Ab)d) CD8 deletion group in tumor-bearing mice (GW6471+PD-L1 mAb+α-CD8 Ab)
试验荷瘤小鼠共4组,每组6只。A total of 4 groups of tumor-bearing mice were tested, with 6 mice in each group.
(3)治疗方案:按接种为0天计算,在接种后第7,10和17天对小鼠进行PD-L1抗体治疗,200ug每只小鼠,腹腔注射(PD-L1 mAb稀释于PBS中,浓度为2mg/ml,每只小鼠腹腔注射0.1ml)。与此同时,在接种后第11天开始对小鼠给与GW6471治疗,10mg/kg,腹腔注射,隔一天给与一次GW6471,共10次(GW6471储液为25mg/ml溶解于DMSO中,待给药前,稀释于55℃预热的PBS中至完全溶解,浓度为2mg/ml,每只小鼠腹腔注射0.1ml)。(3) Treatment plan: Calculated as day 0 of inoculation, mice were treated with PD-L1 antibody on days 7, 10 and 17 after inoculation, 200ug per mouse, intraperitoneal injection (PD-L1 mAb diluted in PBS) , the concentration is 2mg/ml, each mouse is intraperitoneally injected with 0.1ml). At the same time, the mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 11th day after inoculation, and GW6471 was given once every other day, for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 0.1 ml).
(4)肿瘤生长统计:在给药期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线。(4) Statistics of tumor growth: During the administration period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn.
结果如图8所示, 删除CD8+T细胞后肿瘤生长和对照组相同,显著抵消了PD-L1抗体和GW6471的联 合治疗效果,而相比之下删除CD4+T细胞效果并没有那么明显。以上则说明GW6471同PD-L1抗体的联合治疗效果主要依赖于CD8+T细胞。 The results are shown in Figure 8. The tumor growth after deletion of CD8+ T cells was the same as that of the control group, which significantly offset the combined treatment effect of PD-L1 antibody and GW6471, while the effect of deletion of CD4+ T cells was not so obvious. . The above shows that the combined therapeutic effect of GW6471 and PD-L1 antibody mainly depends on CD8+ T cells.
实施例四:典型PPARα抑制剂GW6471联合PD-L1抗体的抗肿瘤作用主要依赖于DC细胞,特别是PPARα-/-DCExample 4: The anti-tumor effect of typical PPARα inhibitor GW6471 combined with PD-L1 antibody mainly depends on DC cells, especially PPARα-/-DC
动物模型建模和细胞培养方法:Animal model modeling and cell culture methods:
(1)CD11c-DTR和CD11c-DTR/PPARα-/-嵌合小鼠模型构建:首先将野生型C57小鼠(雌)进行10Gy全身辐照10分钟杀死骨髓细胞。第二天,取CD11c-DTR小鼠骨髓细胞,按照5×10 6每只小鼠的细胞数量尾静脉注射到辐照小鼠体内。若构建CD11c-DTR/PPARα-/-嵌合小鼠需将CD11c-DTR小鼠和PPARα-/-小鼠骨髓细胞1:1混合后尾静脉注射到辐照小鼠体内。之后给与抗生素氨苄两周,观察其生活情况,两个月后方可进行实验。 (1) Construction of CD11c-DTR and CD11c-DTR/PPARα-/- chimeric mouse models: First, wild-type C57 mice (female) were irradiated with 10 Gy whole body for 10 minutes to kill bone marrow cells. On the second day, the bone marrow cells of CD11c-DTR mice were taken and injected into the irradiated mice by tail vein according to the number of cells per mouse at 5×10 6 . To construct CD11c-DTR/PPARα-/- chimeric mice, CD11c-DTR mice and PPARα-/- mouse bone marrow cells were mixed 1:1 and injected into the irradiated mice via tail vein. After two weeks, the antibiotic ampicillin was given to observe their living conditions, and the experiment could be carried out after two months.
(2)MC38-OT I细胞的复苏及培养:复苏细胞前预先将细胞培养室内超净台进行紫外照射30min,DMEM培养基(含10%胎牛血清)放置室温备用。待准备工作结束后,将肿瘤细胞冻存管从液氮保存罐中取出,立即放入37℃水浴中快速溶解,然后将细胞悬液移入含10ml培养基的离心桶中,900rpm离心5分钟后去除上清,用新鲜培养基重悬后移入细胞培养瓶中,加入10-15ml培养基混悬沉淀细胞,调整细胞浓度后,置于37℃、体积分数5%CO 2饱和湿度培养箱中培养。在维持培养过程中,每天观察细胞状态并及时更换新鲜培养基。当细胞贴壁生长至90%汇合度时,用0.05%胰酶消化,按照1:3的比例进行传代培养。实验当天,将生长状态良好,汇合度达到90%的细胞胰酶消化后,用新鲜培养基中和胰酶,900rpm离心,弃上清后加入无菌PBS重悬,计数,将细胞悬液的密度调整为5×10 6/ml待用。 (2) Recovery and culture of MC38-OT I cells: Before recovering the cells, the ultra-clean bench in the cell culture room was irradiated with ultraviolet light for 30 minutes, and the DMEM medium (containing 10% fetal bovine serum) was placed at room temperature for use. After the preparation is over, take the tumor cell cryopreservation tube out of the liquid nitrogen storage tank, immediately put it into a 37°C water bath to dissolve quickly, and then transfer the cell suspension into a centrifuge bucket containing 10 ml of culture medium, and centrifuge at 900 rpm for 5 minutes. Remove the supernatant, resuspend it with fresh medium and transfer it to a cell culture flask, add 10-15ml medium to suspend the precipitated cells, adjust the cell concentration, and place it in a 37°C, volume fraction 5% CO 2 saturated humidity incubator for cultivation . During the maintenance culture, the cell status was observed every day and fresh medium was replaced in time. When the cells adhered to 90% confluence, they were digested with 0.05% trypsin and subcultured at a ratio of 1:3. On the day of the experiment, trypsinize the cells that grow well and reach 90% confluence, neutralize the trypsin with fresh medium, centrifuge at 900 rpm, discard the supernatant, add sterile PBS to resuspend, count, and count the cells in the suspension. The density was adjusted to 5×10 6 /ml for use.
(3)接种肿瘤细胞:利用1ml无菌注射器皮下接种上述构建的模型小鼠,每只小鼠0.1ml(即每只小鼠接种50×10 4个MC38-OT I细胞)。注意每次吸取细胞悬液前将细胞混合均匀。 (3) Inoculation of tumor cells: Use a 1 ml sterile syringe to subcutaneously inoculate the model mice constructed above, 0.1 ml per mouse (ie, 50×10 4 MC38-OT I cells per mouse). Take care to mix the cells well before each pipetting of the cell suspension.
实验方案(一):PPARα抑制剂GW6471联合PD-L1抗体的抗肿瘤效果主要依赖于DC细胞Experimental scheme (1): The anti-tumor effect of PPARα inhibitor GW6471 combined with PD-L1 antibody mainly depends on DC cells
(1)采用鼠源性结肠癌细胞系MC38-OT I(自带抗原ovalbumin257-264,便于检测特异性T淋巴细胞),对构建的CD11c-DTR小鼠进行皮下接种,约在5天左右可看到约50mm 3的肿瘤形成。 (1) The mouse-derived colon cancer cell line MC38-OT I (with its own antigen ovalbumin257-264, which is convenient for the detection of specific T lymphocytes) was used to subcutaneously inoculate the constructed CD11c-DTR mice. Tumor formation of about 50 mm3 was seen.
(2)荷瘤小鼠组别设计(2) Group design of tumor-bearing mice
试验共分3组,分别为The experiment was divided into 3 groups, which were
a)荷瘤小鼠对照组(CTR);a) tumor-bearing mice control group (CTR);
b)荷瘤小鼠注射DT组(CTR(DT));b) Tumor-bearing mice injected with DT (CTR(DT));
c)荷瘤小鼠抗体治疗组(GW6471+PD-L1 mAb);c) Antibody treatment group of tumor-bearing mice (GW6471+PD-L1 mAb);
d)荷瘤小鼠联合治疗DT组(GW6471+PD-L1 mAb(DT));d) Tumor-bearing mice combined with DT group (GW6471+PD-L1 mAb(DT));
试验荷瘤小鼠共4组,每组5只。A total of 4 groups of tumor-bearing mice were tested, with 5 mice in each group.
(3)治疗方案:按接种日为第0天计算,在接种后第6天,每只b)组和d)组的小鼠腹腔注射500ng DT,隔一天给一次以便删除DC。第7,10和17天对小鼠进行PD-L1抗体治疗,200ug每只小鼠,腹腔注射(PD-L1 mAb稀释于PBS中,浓度为2mg/ml,每只小鼠腹腔注射0.1ml)。与此同时,在接种后第8天开始对小鼠给与GW6471治疗,10mg/kg,腹腔注射,隔一天给与一次GW6471(GW6471储液为25mg/ml溶解于DMSO中,待给药前,稀释于55℃预热的PBS中至完全溶解,浓度为2mg/ml,每只小鼠腹腔注射1ml)。(3) Treatment plan: Calculated according to the day of inoculation as the 0th day, on the 6th day after inoculation, each mouse in group b) and group d) was intraperitoneally injected with 500ng DT, once every other day to delete DC. Mice were treated with PD-L1 antibody on days 7, 10 and 17, 200ug per mouse, i.p. (PD-L1 mAb diluted in PBS at a concentration of 2mg/ml, 0.1ml per mouse i.p.) . At the same time, on the 8th day after inoculation, mice were treated with GW6471, 10 mg/kg, intraperitoneal injection, and GW6471 was administered every other day (GW6471 stock solution was 25 mg/ml dissolved in DMSO, before administration, Dilute in pre-warmed PBS at 55°C to complete dissolution, the concentration is 2mg/ml, and each mouse is injected intraperitoneally with 1ml).
(4)肿瘤生长及生存期统计:在给药期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线,记录小鼠死亡情况。(4) Statistics of tumor growth and survival period: During the administration period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn, and the small size was recorded. Mouse death.
结果如图9所示,相比于GW6471+PD-L1 mAb治疗组, 给与DT(即删除DC)后小鼠肿瘤体积失去控 制,同对照组一样,说明该联合治疗方案依赖于DC细胞的参与The results are shown in Figure 9. Compared with the GW6471+PD-L1 mAb treatment group, the tumor volume of the mice lost control after DT (that is, the deletion of DC) , which was the same as the control group, indicating that the combined treatment regimen depends on DC cells. participation .
实验方案(二):PPARα -/-DC在抗肿瘤作用中占据重要地位 Experimental scheme (2): PPARα -/- DCs play an important role in the anti-tumor effect
(1)在野生型C57小鼠上接种MC38-OT I细胞,待模型构建完毕,分为3组,每组9只小鼠,即荷瘤对照组(CTR),野生型DC治疗组(WT DC)和PPARα敲除DC治疗组(PPARα -/-DC)。 (1) Wild-type C57 mice were inoculated with MC38-OT I cells. After the model was constructed, they were divided into 3 groups with 9 mice in each group, namely the tumor-bearing control group (CTR) and the wild-type DC treatment group (WT). DC) and PPARα knockout DC treatment group (PPARα -/- DC).
(2)治疗方案:按接种为0天计算,在接种后第6、13、20天对小鼠进行DC注射治疗,每只小鼠靠近接瘤处的皮下注射5×10 6个DC细胞。其中野生型DC治疗组和PPARα敲除DC治疗组所接受的DC分别来自野生型C57小鼠和PPARα敲除小鼠。取出的骨髓细胞经体外分化成为成熟的DC后,经过2mg/ml OVA抗原的刺激过夜,再收集DC,计数,进行注射治疗。 (2) Treatment plan: Calculated as day 0 of inoculation, DC injection was performed on the mice on the 6th, 13th and 20th days after inoculation, and 5×10 6 DC cells were injected subcutaneously near the tumor site of each mouse. The DCs received in the wild-type DC treatment group and the PPARα knockout DC treatment group were obtained from wild-type C57 mice and PPARα knockout mice, respectively. After the extracted bone marrow cells were differentiated into mature DCs in vitro, they were stimulated with 2 mg/ml OVA antigen overnight, and then the DCs were collected, counted, and injected for treatment.
(3)肿瘤生长统计:肿瘤生长期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线。(3) Tumor growth statistics: During the tumor growth period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn.
结果如图10所示,与对照组相比,野生型DC治疗组能够有效控制肿瘤生长。而PPARα敲除DC治疗组的治疗效果显著优于野生型DC治疗组,从而进一步体现了PPARα -/-DC在抗肿瘤中的重要地位。 The results are shown in Figure 10. Compared with the control group, the wild-type DC treatment group could effectively control the tumor growth. The treatment effect of PPARα knockout DC treatment group was significantly better than that of wild-type DC treatment group, which further reflected the important role of PPARα -/- DC in anti-tumor.
实验方案(三):GW6471和PPARα敲除对DC激活T细胞功能的影响Experimental Protocol (3): Effects of GW6471 and PPARα Knockout on the Function of DC-Activated T Cells
(1)野生型小鼠接种MC38-OTI肿瘤细胞后,小鼠分为两组:荷瘤对照组和GW6471给药组。GW6471给药组从接瘤后第7天开始腹腔注射10mg/kg GW6471,每两天给药一次,共给药6次。在接瘤后第22天,处死小鼠,手术取出肿瘤组织,研磨、消化后获得肿瘤组织细胞。(1) After wild-type mice were inoculated with MC38-OTI tumor cells, the mice were divided into two groups: tumor-bearing control group and GW6471 administration group. The GW6471 administration group received intraperitoneal injection of 10 mg/kg GW6471 from the 7th day after tumor receiving, once every two days, for a total of 6 times. On the 22nd day after receiving the tumor, the mice were sacrificed, and the tumor tissue was surgically removed, ground and digested to obtain tumor tissue cells.
(2)体外处理:通过流式细胞分选的方法分选出肿瘤浸润性DC(CD45+MHCII+CD11c+F4/80-),与2mg/ml OVA抗原孵育48小时,之后洗去抗原,与从OTI转基因小鼠脾脏中通过磁珠分选的方法获得的OTICD8+T细胞(CFSE预染色)按照1:8的比例进行共孵育,3天后检测OTI CD8+T细胞的增殖状况和分泌IFN-γ的水平。(2) In vitro treatment: tumor-infiltrating DCs (CD45+MHCII+CD11c+F4/80-) were sorted by flow cytometry, incubated with 2 mg/ml OVA antigen for 48 hours, and then washed away with antigen. The OTCD8+ T cells (pre-stained with CFSE) obtained by magnetic bead sorting from the spleen of OTI transgenic mice were co-incubated at a ratio of 1:8. After 3 days, the proliferation status and secretion of IFN- gamma level.
(3)检测方法:流式细胞仪检测CFSE稀释倍数,以此确定OTI CD8+T细胞的分裂状态。通常来说细胞增殖约明显,CFSE稀释倍数越多,平均荧光强度越低。OTI CD8+T细胞分泌的IFN-γ的水平也可以通过ELISA试剂盒来检测,来说明OTI CD8+T细胞的激活状态。(3) Detection method: Flow cytometry was used to detect the dilution ratio of CFSE to determine the division state of OTI CD8+ T cells. Generally speaking, cell proliferation is about obvious, and the higher the dilution of CFSE, the lower the average fluorescence intensity. The level of IFN-γ secreted by OTI CD8+ T cells can also be detected by ELISA kit to indicate the activation state of OTI CD8+ T cells.
结果显示,相对于接瘤对照组,GW6471给药组的小鼠肿瘤浸润性DC能够更有效的激活OTI CD8+T细胞——增殖更强烈(图11a),且分泌更多的IFN-γ(图11b)The results showed that compared with the tumor receiving control group, tumor-infiltrating DCs in the GW6471-administered group could more effectively activate OTI CD8+ T cells—proliferated more strongly (Fig. 11a) and secreted more IFN-γ (Fig. 11a). Figure 11b)
实验方案(四):PPARα敲除对DC激活T细胞功能的影响Experimental protocol (4): The effect of PPARα knockout on the function of DC-activated T cells
(1)野生型和PPARα敲除小鼠分别接种MC38-OTI肿瘤细胞。接瘤后第20天,处死小鼠,手术取出肿瘤组织,研磨、消化后获得肿瘤组织细胞。(1) Wild-type and PPARα knockout mice were inoculated with MC38-OTI tumor cells, respectively. On the 20th day after receiving the tumor, the mice were sacrificed, and the tumor tissue was surgically removed, ground and digested to obtain tumor tissue cells.
体外处理:在分离小鼠肿瘤内T细胞的前一天在ELISPOT板中预孵育anti-mouse IFN-γ抗体,作用浓度为5μg/ml,4℃孵育过夜,次日弃去抗体,加入封闭液(含10%胎牛血清、1%青霉素链霉素和β-巯基乙醇的RPMI 1640培养基)室温封闭2h。随后将分离得到的小鼠肿瘤T细胞按照3×105每孔加入板中,并孵育15μg/ml OVA257-264抗原肽,37℃培养箱培养48h。48h后弃培养液,加入去离子水裂解细胞,5min/次,共2次。用PBST(含0.05%Tween-20的1×PBS)溶液洗3遍,随后加入生物素标记的anti-mouse IFN-γ抗体,室温孵育2h。PBST溶液洗4遍,随后加入链亲和素标记的辣根过氧化物酶(streptavidin-HRP),室温孵育1 h。最后1×PBS洗3遍后加入AEC底物显色5-60min,加入去离子水终止反应,继续冲洗干净后将ELISPOT板子吹干。In vitro treatment: pre-incubate anti-mouse IFN-γ antibody in an ELISPOT plate one day before the isolation of mouse intratumoral T cells at a concentration of 5 μg/ml, incubate overnight at 4°C, discard the antibody the next day, and add blocking solution ( RPMI 1640 medium containing 10% fetal bovine serum, 1% penicillin-streptomycin and β-mercaptoethanol) was blocked at room temperature for 2 h. Then, the isolated mouse tumor T cells were added to the plate at 3 × 105 per well, and incubated with 15 μg/ml OVA257-264 antigen peptide for 48 h in a 37°C incubator. After 48 h, the culture medium was discarded, and deionized water was added to lyse the cells, 5 min/time, 2 times in total. Wash with PBST (1×PBS containing 0.05% Tween-20) solution for 3 times, then add biotin-labeled anti-mouse IFN-γ antibody, and incubate at room temperature for 2 h. After washing with PBST solution for 4 times, streptavidin-labeled horseradish peroxidase (streptavidin-HRP) was added and incubated for 1 h at room temperature. Finally, after washing with 1×PBS for 3 times, AEC substrate was added for 5-60 min, and deionized water was added to stop the reaction. After washing, the ELISPOT plate was dried.
细胞染色:将上述细胞悬液进行荧光染色,配色方案为APC-CD45,PE/Cy7-CD3,APC/Cy7-CD8,PE-Tetramer(Tet),抗体与细胞混合后,在4℃避光染色30min。之后用含有2%FBS的PBS溶液清洗一次,终止染色。Cell staining: Fluorescent staining of the above cell suspension, the color scheme is APC-CD45, PE/Cy7-CD3, APC/Cy7-CD8, PE-Tetramer (Tet), after mixing the antibody with the cells, stain at 4°C in the dark 30min. The staining was terminated by washing once with PBS containing 2% FBS.
(2)检测方法:Ellispot结果通过荧光酶斑点分析仪CTL analyzer LLC进行扫描和计数分析。另外一部分肿瘤组织单细胞悬液直接进行细胞染色,由于我们使用的PE-Tetramer(Tet)抗体能够直接识别T细胞表面能够与肿瘤相关抗原配对的T细胞受体,因此这种分析方法可以直接检测得到肿瘤抗原特异性Tet+CD8+T细胞的比例。(2) Detection method: Ellispot results were scanned and counted by luciferase spot analyzer CTL analyzer LLC. Another part of the tumor tissue single cell suspension was directly stained. Since the PE-Tetramer (Tet) antibody we used can directly recognize T cell receptors on the surface of T cells that can pair with tumor-associated antigens, this analysis method can directly detect The ratio of tumor antigen-specific Tet+CD8+ T cells was obtained.
结果显示,与野生型小鼠相比,PPARα敲除小鼠的肿瘤组织中含有更高比例的抗原特异性Tet+CD8+T细胞比例(图12a),而且这些细胞能够产生强的IFN-γ信号(图12b)。这些结果说明PPARα敲除小鼠的肿瘤浸润性DC具有更强的抗原呈递和激活CD8+T细胞的能力。The results showed that compared with wild-type mice, the tumor tissues of PPARα knockout mice contained a higher proportion of antigen-specific Tet+CD8+ T cells (Fig. 12a), and these cells were able to produce strong IFN-γ signal (Fig. 12b). These results suggest that tumor-infiltrating DCs from PPARα knockout mice have stronger ability to present antigen and activate CD8+ T cells.
实验例五:PPARα抑制剂GW6471对DC代谢状态的调控,进而调控DC的功能Experimental example 5: PPARα inhibitor GW6471 regulates the metabolic state of DC, thereby regulating the function of DC
细胞培养及处理方法:Cell culture and processing methods:
骨髓来源DC的获取:取6-8周龄C57BL/6小鼠,解剖获取长骨和胫骨,将骨髓吹出,红细胞裂解液2ml处理1min裂解红细胞,无血清培养基中和后离心,重悬获得小鼠骨髓细胞悬液,以3×10 6细胞/皿种于10cm培养皿中。完全培养基(含10%FBS、50μM巯基乙醇的RPMI 1640)加入20ng/ml mGM-CSF(重组小鼠粒-巨噬细胞集落刺激因子)培养按第0天计算,第3天补加10ml完全培养基(含20ng/ml mGM-CSF)。培养至第6天,收集未贴壁细胞为未成熟的BMDCs。经流式鉴定CD11c+的DCs纯度达90%以上,可用于后续实验。 Acquisition of bone marrow-derived DC: Take 6-8 week old C57BL/6 mice, dissect the long bones and tibias, blow out the bone marrow, lyse the red blood cells with 2 ml of erythrocyte lysate for 1 min, neutralize them in serum-free medium, and then resuspend them to obtain small cells. The mouse bone marrow cell suspension was seeded in a 10cm petri dish at 3×10 6 cells/dish. Complete medium (RPMI 1640 containing 10% FBS, 50 μM mercaptoethanol) was added with 20ng/ml mGM-CSF (recombinant mouse granulocyte-macrophage colony stimulating factor). Medium (containing 20ng/ml mGM-CSF). On day 6, unadherent cells were collected as immature BMDCs. The purity of CD11c+ DCs identified by flow cytometry was more than 90% and could be used in subsequent experiments.
肿瘤外泌体(Tumor-derived Exosomes,TDE)的获取:肿瘤细胞培养在含有10%已去除外泌体的胎牛血清(100,000g,2h)的培养基中48h后,收集上清。首先,4000rpm离心2h去除上清中的细胞碎片。然后,利用100kDa的超滤管以4000rpm、30min/次的速度收集上清中大于100kDa的组份。收集的组份以5:1(v:v)加入外泌体提取试剂(EXOTC10A-1,SBI),充分混合后4℃放置至少12h。待12h后沉淀出现,3000rpm离心30min,分离出沉淀物,即为外泌体。PBS按照一定的体积重悬后,BCA测定蛋白含量以标定外泌体的含量,分装后置于-80℃保存,备用。Acquisition of Tumor-derived Exosomes (TDE): Tumor cells were cultured in a medium containing 10% exosome-depleted fetal bovine serum (100,000 g, 2 h) for 48 h, and the supernatant was collected. First, cell debris in the supernatant was removed by centrifugation at 4000 rpm for 2 h. Then, the fractions larger than 100 kDa in the supernatant were collected by using a 100 kDa ultrafiltration tube at a speed of 4000 rpm, 30 min/time. The collected components were added to the exosome extraction reagent (EXOTC10A-1, SBI) at a ratio of 5:1 (v:v), mixed well and placed at 4°C for at least 12 hours. After 12 hours, the precipitate appeared, centrifuged at 3000 rpm for 30 min, and the precipitate was separated, which was exosomes. After resuspending in PBS according to a certain volume, the protein content was determined by BCA to standardize the content of exosomes, and the packaged and stored at -80°C for future use.
肿瘤外泌体的处理可以改变骨髓来源DC的代谢状态和功能,模拟肿瘤微环境的影响。Processing of tumor exosomes can alter the metabolic state and function of bone marrow-derived DCs, mimicking the effects of the tumor microenvironment.
实验方案(一):GW6471对DC代谢状态的调控Experimental Protocol (1): Regulation of DC Metabolic Status by GW6471
(1)考察DC细胞的氧化磷酸化水平:(1) Investigate the oxidative phosphorylation level of DC cells:
细胞处理:Seahorse(总体细胞代谢水平检测试剂盒,购自安捷伦)的Seahorse XF24细胞培养板利用Corning TM Cell-Tak Cell and Tissue Adhesive试剂(促进悬浮细胞黏附到细胞培养板底面)3.5μg/cm 2进行贴壁预处理后,经过外泌体或GW6471预处理48h的骨髓来源的DCs按照30万每孔接种于培养板中,每组五个复孔,0.5ml体系,细胞贴壁4h。 Cell processing: Seahorse XF24 cell culture plate by Seahorse (Total Cell Metabolism Level Detection Kit, purchased from Agilent) uses Corning Cell-Tak Cell and Tissue Adhesive Reagent (to promote the adhesion of suspended cells to the bottom of the cell culture plate) 3.5 μg/cm 2 After adherence pretreatment, bone marrow-derived DCs pretreated with exosomes or GW6471 for 48h were seeded in culture plates at 300,000 per well, with five replicate wells per group, 0.5ml system, and cells adhered for 4h.
Seahorse海马生物能量测定(总体细胞代谢水平):探针板水化过夜。在检测之前,细胞更换为预热的525μl XF Base培养基(Seahorse Bioscience)培养,该基础培养基中另加入10mM葡萄糖,2mM L-谷氨酰胺和1mM丙酮酸钠,并用NaOH调至PH为7.4过滤后可用。Oligomycin(寡霉素),FCCP(三氟甲氧基苯腙羰基氰化物),Rotenone(魚藤酮)和Antimycin A(抗霉素A)分别用XF基础培养基稀释为1mM,1.5mM,100nM和1mM,体积为75μl加入到探针板A,B,C孔中(Rotenone和antimycin-A混合配置一起加入C孔)。待准备完毕即可上机检测细胞氧化磷酸化水平。Seahorse hippocampal bioenergetics assay (levels of overall cellular metabolism): Probe plates were hydrated overnight. Before assay, cells were replaced with pre-warmed 525 μl XF Base medium (Seahorse Bioscience) supplemented with 10 mM glucose, 2 mM L-glutamine and 1 mM sodium pyruvate, and adjusted to pH 7.4 with NaOH Available after filtering. Oligomycin (oligomycin), FCCP (trifluoromethoxyphenylhydrazone carbonyl cyanide), Rotenone (rotenone) and Antimycin A (antimycin A) were diluted in XF basal medium to 1 mM, 1.5 mM, 100 nM and 1 mM, respectively , a volume of 75 μl was added to wells A, B, and C of the probe plate (Rotenone and antimycin-A were mixed together in well C). After the preparation is completed, the oxidative phosphorylation level of the cells can be detected on the machine.
(2)考察DC细胞的脂肪酸氧化水平:(2) To investigate the fatty acid oxidation level of DC cells:
细胞处理:Seahorse XF24细胞培养板利用Corning TM Cell-Tak Cell and Tissue Adhesive试剂3.5μg/cm 2进行贴壁预处理后,经过外泌体或GW6471预处理48h的骨髓来源的DCs按照30万每孔接种于培养板中,每组五个复孔,0.5ml体系,细胞在底物限制性培养基(仅含有0.5mM葡萄糖,1.0mM L-谷氨酰胺,0.5mM肉碱and 1%FBS的DMEM)中培养过夜。 Cell treatment: Seahorse XF24 cell culture plates were pretreated with Corning TM Cell-Tak Cell and Tissue Adhesive reagent 3.5μg/cm 2 for adherent pretreatment, and bone marrow-derived DCs pretreated with exosomes or GW6471 for 48h were prepared at a rate of 300,000 per well. Inoculated in culture plates, five replicate wells per group, 0.5 ml system, and cells were plated in substrate-limiting medium (DMEM containing only 0.5 mM glucose, 1.0 mM L-glutamine, 0.5 mM carnitine and 1% FBS). ) overnight.
Seahorse海马生物能量测定(脂肪酸氧化水平):探针板水化过夜。在上机检测前45min,细胞更换为预热的450μl FAO assay medium(脂肪酸氧化检测培养基,含111mM NaCl,4.7mM KCl,2mM MgSO 4,1.2mM Na 2HPO 4,2.5mM葡萄糖,0.5mM肉碱and 5mM HEPES磺酸缓冲盐溶液)中培养。并利用该培养基配置100μM的Etomoxir(简称ETO,肉碱棕榈酰基转运蛋白1的抑制剂)75μl,加入探针板A孔中。待准备完毕即可上机检测脂肪酸氧化程度。 Seahorse hippocampal bioenergetics assay (level of fatty acid oxidation): Probe plate hydrated overnight. 45 min before the test, the cells were replaced with pre-warmed 450 μl FAO assay medium (fatty acid oxidation assay medium, containing 111 mM NaCl, 4.7 mM KCl, 2 mM MgSO 4 , 1.2 mM Na 2 HPO 4 , 2.5 mM glucose, 0.5 mM meat Alkaline and 5 mM HEPES sulfonic acid buffered saline). 75 μl of 100 μM Etomoxir (abbreviated as ETO, an inhibitor of carnitine palmitoyl transporter 1) was prepared by using the medium, and added to well A of the probe plate. After the preparation is complete, you can go on the machine to detect the degree of fatty acid oxidation.
(3)考察DC细胞的乳酸分泌水平:(3) Investigate the lactate secretion level of DC cells:
细胞处理:骨髓来源的DC按照30万每孔接种于24孔细胞培养板中,经过外泌体或GW6471预处理48h后,收集上清。Cell treatment: DCs derived from bone marrow were seeded in 24-well cell culture plates at 300,000 per well, and the supernatant was collected after pretreatment with exosomes or GW6471 for 48 hours.
检测方式:使用比色法乳酸检测试剂盒(Lactate Colorimetric Assay Kit,Biovision)检测上清中的乳酸含量,并读取570nm处吸光值进行定量分析。Detection method: Lactate Colorimetric Assay Kit (Biovision) was used to detect the lactic acid content in the supernatant, and the absorbance at 570 nm was read for quantitative analysis.
结果显示,肿瘤外泌体(TDE)能够显著增加DC细胞的耗氧量(OCR)——细胞氧化磷酸化水平的标志(图13a),以及脂肪酸氧化水平(图13b),同时降低了乳酸的分泌量(图13c)——细胞糖酵解水平的标注。因此,肿瘤外泌体在体外能够模拟肿瘤微环境对DC代谢状态的调控。在此基础上,GW6471能够显著降低DC的氧化磷酸化水平、脂肪酸氧化水平,并恢复糖酵解水平(图13),而且GW6471引起的代谢状态改变有助于DC功能的恢复。The results showed that tumor exosomes (TDE) were able to significantly increase DC cell oxygen consumption (OCR), a marker of cellular oxidative phosphorylation levels (Fig. 13a), and fatty acid oxidation levels (Fig. 13b), while reducing lactate levels. Secretion (Figure 13c) - Annotation of cellular glycolysis levels. Therefore, tumor exosomes can mimic the regulation of the metabolic state of DCs by the tumor microenvironment in vitro. On this basis, GW6471 could significantly reduce the level of oxidative phosphorylation and fatty acid oxidation of DC, and restore the level of glycolysis (Fig. 13), and the metabolic state change caused by GW6471 contributed to the recovery of DC function.
实验方案(二):GW6471对DC诱导产生调节性T细胞的影响Experimental protocol (2): The effect of GW6471 on DC induction of regulatory T cells
小鼠模型介绍:Introduction to the mouse model:
Foxp3-GFP转基因小鼠的特点是通过基因编辑手段,将表达绿色荧光蛋白(GFP)的基因置于负责CD4+调节性T细胞发育的转录因子Foxp3的启动子之后。因此,我们可以方便的通过荧光检测的方法(GFP阳性即为Foxp3阳性),而非以往比较复杂的胞内蛋白染色的方法,来考察小鼠体内或体外CD4+T细胞转变成调节性T细胞的状况。Foxp3-GFP transgenic mice are characterized by gene editing to place the gene expressing green fluorescent protein (GFP) behind the promoter of the transcription factor Foxp3 responsible for the development of CD4+ regulatory T cells. Therefore, we can easily detect the transformation of CD4+ T cells into regulatory T cells in vivo or in vitro by the method of fluorescence detection (GFP positive means Foxp3 positive), rather than the complex intracellular protein staining method in the past. condition.
OTI(C57BL/6-Tg(TcraTcrb)1100Mjb/J)转基因小鼠的特点是通过基因编辑手段,使该小鼠的CD8+T细胞的T细胞受体能够特异性识别OVA蛋白的第257-264位多肽的结构,并被激活、增殖。该转基因小鼠常用于考察抗原特异性免疫反应的研究中。OTI (C57BL/6-Tg(TcraTcrb) 1100Mjb/J) transgenic mice are characterized by gene editing, so that the T cell receptors of the CD8+ T cells of the mice can specifically recognize the 257-264 of the OVA protein. The structure of the polypeptide is activated and proliferated. The transgenic mice are commonly used in studies investigating antigen-specific immune responses.
(1)细胞处理:首先将BMDCs用外泌体以及15uM GW6471预处理48h。另外,取Foxp3-GFP转基因小鼠的脾细胞,通过磁珠分选出CD4+细胞,离心后计数。然后,将以上预处理的DCs(2×10 4)与CFSE(羧基荧光素二醋酸琥珀酰亚胺酯,常用来标记并监控细胞的增殖状态)标记的取自OTI转基因小鼠脾脏的CD8+T细胞(2×10 5)以1:10的比例共培养与U型底96孔板中。第四天时加入100U/孔重组小鼠IL-2细胞因子促进T细胞存活。 (1) Cell treatment: BMDCs were first pretreated with exosomes and 15uM GW6471 for 48h. In addition, splenocytes of Foxp3-GFP transgenic mice were taken, and CD4+ cells were sorted by magnetic beads, and counted after centrifugation. Then, the above pretreated DCs (2×10 4 ) were combined with CFSE (carboxyfluorescein diacetate succinimidyl ester, commonly used to label and monitor the proliferation state of cells) labeled CD8+ from the spleen of OTI transgenic mice. T cells (2 x 105 ) were co-cultured at a ratio of 1:10 in U-bottom 96-well plates. On the fourth day, 100U/well of recombinant mouse IL-2 cytokine was added to promote the survival of T cells.
(2)检测方式:共培养7天后,细胞通过流式分析CD4+Foxp3+细胞的比例。(2) Detection method: After 7 days of co-culture, the proportion of CD4+Foxp3+ cells was analyzed by flow cytometry.
结果显示,肿瘤外泌体预处理的DC能够诱导更多的调节性T细胞(CD4+Foxp+)产生(图14),而GW6471可以显著降低调节性T细胞,使之恢复到未经处理的对照组水平(图14)。The results showed that DCs pretreated with tumor exosomes were able to induce more regulatory T cells (CD4+Foxp+) production (Fig. 14), while GW6471 could significantly reduce regulatory T cells and restore them to untreated controls group level (Figure 14).
实验方案(三):GW6471恢复DC对CD8+T细胞的活化作用Experimental protocol (3): GW6471 restores the activation effect of DC on CD8+ T cells
(1)细胞处理:首先将BMDCs用外泌体和2mg/ml OVA抗原,以及15uM GW6471共同孵育48h。另外,取OT I转基因小鼠的脾细胞,通过磁珠分选出CD8+细胞,离心后计数。然后,将以上预处理的DCs(5×10 4)与OTI CD8+T细胞(4×10 5)以1:8的比例共培养与U型底96孔板中。 (1) Cell treatment: BMDCs were first incubated with exosomes, 2mg/ml OVA antigen, and 15uM GW6471 for 48h. In addition, spleen cells of OT I transgenic mice were taken, and CD8+ cells were sorted by magnetic beads and counted after centrifugation. Then, the above pretreated DCs (5×10 4 ) were co-cultured with OTI CD8+ T cells (4×10 5 ) at a ratio of 1:8 in a U-bottom 96-well plate.
(2)检测方式:共培养3天后,收集上清,通过ELISA的方法检测OTI CD8+T细胞分泌的IFN-γ。(2) Detection method: After 3 days of co-culture, the supernatant was collected, and the IFN-γ secreted by OTI CD8+ T cells was detected by ELISA.
结果显示,肿瘤外泌体预处理的DC能够抑制CD8+T细胞的活化,而GW6471显著提高了OTI CD8+T细胞分泌的IFN-γ水平(图15),充分说明GW6471恢复了DC对CD8+T的活化。The results showed that DCs pretreated with tumor exosomes could inhibit the activation of CD8+ T cells, while GW6471 significantly increased the level of IFN-γ secreted by OTI CD8+ T cells (Fig. 15), which fully demonstrated that GW6471 restored the effect of DC on CD8+ T cells. activation of T.
实验例六:PPARα抑制剂GW6471增强E7疫苗对TC-1的抗肿瘤效果并延长小鼠的生存期Experimental Example 6: PPARα inhibitor GW6471 enhances the anti-tumor effect of E7 vaccine on TC-1 and prolongs the survival of mice
(一)、细胞培养及动物模型建模方法:(1) Cell culture and animal model modeling methods:
(1)TC-1细胞的复苏及培养:复苏细胞前预先将细胞培养室内超净台进行紫外照射30min,RPMI1640培养基(含10%胎牛血清)放置室温备用。待准备工作结束后,将肿瘤细胞冻存管从液氮保存罐中取出,立即放入37℃水浴中快速溶解,然后将细胞悬液移入含10ml培养基的离心桶中,900rpm离心5分钟后去除上清,用新鲜培养基重悬后,转移入细胞培养瓶中,加入10-15ml培养基混悬沉淀细胞,调整细胞浓度后,置于37℃、体积分数5%CO 2饱和湿度培养箱中培养。在维持培养过程中,每天观察细胞状态并及时更换新鲜培养基。当细胞贴壁生长至90%汇合度时,用0.05%胰酶消化,按照1:3的比例进行传代培养。实验当天,将生长状态良好,汇合度达到90%的细胞胰酶消化后,用新鲜培养基中和胰酶,900rpm离心,弃上清后加入无菌PBS重悬,计数,将细胞悬液的密度调整为5×10 5/ml待用。 (1) Recovery and culture of TC-1 cells: Before recovering the cells, the ultra-clean bench in the cell culture room was irradiated with UV light for 30 minutes, and the RPMI1640 medium (containing 10% fetal bovine serum) was placed at room temperature for use. After the preparations are over, take the tumor cell cryopreservation tube out of the liquid nitrogen storage tank, immediately put it into a 37°C water bath to dissolve quickly, and then transfer the cell suspension into a centrifuge bucket containing 10 ml of medium, and centrifuge at 900 rpm for 5 minutes. Remove the supernatant, resuspend it with fresh medium, transfer it into a cell culture flask, add 10-15ml medium to suspend the pelleted cells, adjust the cell concentration, and place it in a 37°C, volume fraction 5% CO 2 saturated humidity incubator cultivated in. During the maintenance culture, the cell status was observed every day and fresh medium was replaced in time. When the cells adhered to 90% confluence, they were digested with 0.05% trypsin and subcultured at a ratio of 1:3. On the day of the experiment, trypsinize the cells that grow well and reach 90% confluence, neutralize the trypsin with fresh medium, centrifuge at 900 rpm, discard the supernatant, add sterile PBS to resuspend, count, and count the cells in the suspension. The density was adjusted to 5×10 5 /ml until use.
(2)接种肿瘤细胞:利用1ml无菌注射器皮下接种雌性野生型C57小鼠,每只小鼠0.1ml(即每只小鼠接种5×10 4个TC-1细胞)。注意每次吸取细胞悬液前将细胞混合均匀。约在7天左右可看到约50mm 3的肿瘤形成。 (2) Inoculation of tumor cells: Use a 1 ml sterile syringe to subcutaneously inoculate female wild-type C57 mice with 0.1 ml per mouse (ie, inoculate 5×10 4 TC-1 cells per mouse). Take care to mix the cells well before each pipetting of the cell suspension. Tumor formation of about 50 mm3 was seen in about 7 days.
(二)、治疗方案:(2) Treatment plan:
(3)荷瘤小鼠组别设计(3) Group design of tumor-bearing mice
试验共分4组,分别为The experiment was divided into 4 groups, which were
d)荷瘤小鼠对照组(CTR);d) tumor-bearing mice control group (CTR);
e)荷瘤小鼠疫苗治疗组(E7 vaccine)(制备方法可参见CN201410570624);e) tumor-bearing mice vaccine treatment group (E7 vaccine) (for the preparation method, please refer to CN201410570624);
f)荷瘤小鼠给药组(GW6471);f) Tumor-bearing mice administration group (GW6471);
g)荷瘤小鼠联合治疗组(GW6471+E7 vaccine);g) tumor-bearing mice combined treatment group (GW6471+E7 vaccine);
试验荷瘤小鼠共4组,每组10只。There were 4 groups of tumor-bearing mice, 10 mice in each group.
(3)治疗方案:按接种为0天计算,在接种后第7,14和21天对小鼠进行E7疫苗治疗(疫苗的制备参见本实验室在先工作:CN201410570624.7),E7(20)/MPLA/PEG-PE(20/10/1000w/w/w)/每只小鼠,皮下注射。与此同时,在接种后第8天开始对小鼠给与GW6471治疗,10mg/kg,腹腔注射,隔一天给与一次GW6471,共10次(GW6471储液为25mg/ml溶解于DMSO中,待给药前,稀释于55℃预热的PBS中至完全溶解,浓度为2mg/ml,每只小鼠腹腔注射1ml)。(3) Treatment plan: Calculated as day 0 of inoculation, the mice were treated with E7 vaccine on the 7th, 14th and 21st days after inoculation (for the preparation of vaccine, please refer to the previous work of our laboratory: CN201410570624.7), E7 (20 )/MPLA/PEG-PE(20/10/1000w/w/w)/each mouse, subcutaneous injection. At the same time, the mice were treated with GW6471, 10 mg/kg, intraperitoneally, on the 8th day after inoculation, and GW6471 was given once every other day for a total of 10 times (GW6471 stock solution was 25 mg/ml dissolved in DMSO, waiting for Before administration, it was diluted in pre-warmed PBS at 55°C until completely dissolved, the concentration was 2 mg/ml, and each mouse was injected intraperitoneally with 1 ml).
(4)肿瘤生长及生存期统计:在给药期间,每周进行两次测量各组小鼠肿瘤生长情况,按照长×宽×宽/2计算出肿瘤体积,并绘制肿瘤生长曲线,记录小鼠死亡情况。(4) Statistics of tumor growth and survival period: During the administration period, the tumor growth of mice in each group was measured twice a week, the tumor volume was calculated according to length×width×width/2, and the tumor growth curve was drawn, and the small size was recorded. Mouse death.
结果如图16所示,相比于E7疫苗治疗组,联合GW6471可进一步显著减缓TC-1的肿瘤生长。与此同时如图17所示,联合GW6471和E7疫苗对小鼠生存期的增长也有显著的改善。The results are shown in Figure 16. Compared with the E7 vaccine treatment group, the combination of GW6471 can further significantly slow down the tumor growth of TC-1. At the same time, as shown in Figure 17, the combination of GW6471 and E7 vaccine also significantly improved the survival of mice.
最后需要说明的是,以上实施例仅用作帮助本领域技术人员理解本发明的实质,不用做对保护范围的限定。Finally, it should be noted that the above embodiments are only used to help those skilled in the art to understand the essence of the present invention, and do not limit the protection scope.

Claims (7)

  1. PPARα配体在制备药物中的应用,所述的药物的功能包括如下功能的任意一种或多种:The application of PPARα ligand in the preparation of medicine, the function of the medicine includes any one or more of the following functions:
    (1)激活T细胞的活化信号,所述的活化信号是使JNK磷酸化水平上升,所述的T细胞优选为CD8+T细胞;(1) an activation signal for activating T cells, the activation signal is to increase the phosphorylation level of JNK, and the T cells are preferably CD8+ T cells;
    (2)激活T细胞,(2) activate T cells,
    所述的激活T细胞指使T细胞分泌更多的IFN-γ和IL-2且同时增加T细胞的存活时间,The activation of T cells refers to making T cells secrete more IFN-γ and IL-2 and at the same time increase the survival time of T cells,
    所述的T细胞优选为CD8+T细胞,更优选的,为肿瘤浸润性CD8+T细胞;The T cells are preferably CD8+ T cells, more preferably, tumor-infiltrating CD8+ T cells;
    (3)促进T细胞分化,所述的分化指促进T细胞分化为记忆性T细胞;(3) promoting the differentiation of T cells, the differentiation refers to promoting the differentiation of T cells into memory T cells;
    (4)调节树突状细胞(Dendritic Cell,DC细胞)的代谢状态,(4) Regulate the metabolic state of dendritic cells (DC cells),
    所述的调节代谢是指:下调DC细胞的脂肪酸氧化/氧化磷酸化水平,并上调糖酵解水平;The regulating metabolism refers to: down-regulating the fatty acid oxidation/oxidative phosphorylation level of DC cells, and up-regulating the level of glycolysis;
    所述的DC为骨髓来源的DC,更优选的,为浸润在肿瘤微环境中的DC;The DCs are bone marrow-derived DCs, more preferably, DCs infiltrated in the tumor microenvironment;
    (5)恢复DC对T细胞的调控功能,(5) Restore the regulatory function of DC on T cells,
    所述的恢复DC的调控功能指:下调CD4+T细胞转变为调节性T细胞的比例、增加肿瘤杀伤性CD8+T细胞的比例、激活肿瘤杀伤性CD8+T细胞的功能;The restoration of the regulatory function of DC refers to: down-regulating the ratio of CD4+ T cells transformed into regulatory T cells, increasing the ratio of tumor-killing CD8+ T cells, and activating the function of tumor-killing CD8+ T cells;
    所述的激活肿瘤杀伤性CD8+T细胞指:使肿瘤杀伤性CD8+T细胞分泌更多的IFN-和IL-2且同时增加肿瘤杀伤性CD8+T细胞的存活时间;The activation of tumor-killing CD8+ T cells refers to: making tumor-killing CD8+ T cells secrete more IFN- and IL-2 while increasing the survival time of tumor-killing CD8+ T cells;
    (6)提高免疫检查点抑制剂的抗肿瘤效果,所述的免疫检查点抑制剂优选为PD-L1/PD-1抗体,优选的,所述的肿瘤为免疫检查点抑制剂治疗有效的肿瘤;(6) Improve the anti-tumor effect of an immune checkpoint inhibitor, the immune checkpoint inhibitor is preferably a PD-L1/PD-1 antibody, and preferably, the tumor is a tumor that is effectively treated by an immune checkpoint inhibitor ;
    (7)提高抗肿瘤疫苗的治疗效果,所述的抗肿瘤疫苗优选为以PEG2000-DSPE胶束装载多肽/蛋白肿瘤抗原的纳米疫苗。(7) Improve the therapeutic effect of the anti-tumor vaccine, the anti-tumor vaccine is preferably a nano-vaccine with PEG2000-DSPE micelles loaded with polypeptide/protein tumor antigens.
  2. 根据权利要求1所述的应用,其特征在于,所述的PPARα配体为PPARα的抑制剂,优选的,所述的PPARα的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。The application according to claim 1, wherein the PPARα ligand is a PPARα inhibitor, preferably, the PPARα inhibitor is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284, ZM0285 , ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
  3. 一组PPARα的抑制剂,所述的抑制剂为ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。A group of PPARα inhibitors, the inhibitors are ZM0282, ZM0283, ZM0284, ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
  4. 一种抗肿瘤联用制剂,所述的制剂包含:An anti-tumor combined preparation comprising:
    (1)治疗有效量PPARα配体;(1) A therapeutically effective amount of PPARα ligand;
    (2)治疗有效量的免疫检查点抑制剂或治疗有效量的抗肿瘤疫苗;(2) A therapeutically effective amount of an immune checkpoint inhibitor or a therapeutically effective amount of an antitumor vaccine;
    (3)必要的药用辅料。(3) Necessary pharmaceutical excipients.
  5. 根据权利要求4所述的联用制剂,其特征在于,所述的PPARα配体为PPARα的抑制剂,优选的,所述的PPARα的抑制剂为GW6471分子及其结构类似物ZM0282、ZM0283、ZM0284、ZM0285、ZM0286、ZM0325、ZM0326、ZM0329、ZM0330、ZM0332、ZM0333。The combined preparation according to claim 4, wherein the PPARα ligand is an inhibitor of PPARα, preferably, the inhibitor of PPARα is GW6471 molecule and its structural analogs ZM0282, ZM0283, ZM0284 , ZM0285, ZM0286, ZM0325, ZM0326, ZM0329, ZM0330, ZM0332, ZM0333.
  6. 根据权利要求4或5所述的联用制剂,其特征在于,所述的免疫检查点抑制剂为PD-L1/PD-1抗体。The combined preparation according to claim 4 or 5, wherein the immune checkpoint inhibitor is a PD-L1/PD-1 antibody.
  7. 根据权利要求4或5所述的联用制剂,其特征在于,所述的抗肿瘤疫苗为以PEG2000-DSPE胶束装载多肽/蛋白肿瘤抗原的疫苗。The combined preparation according to claim 4 or 5, wherein the anti-tumor vaccine is a vaccine loaded with polypeptide/protein tumor antigens in PEG2000-DSPE micelles.
PCT/CN2021/117407 2020-09-14 2021-09-09 USE OF PPARα (PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR α) LIGAND IN PREPARATION OF MEDICINE WO2022052982A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202180062556.1A CN116367861A (en) 2020-09-14 2021-09-09 Application of PPARα (peroxisome proliferator-activated receptor α) ligand in preparation of medicines

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010961007 2020-09-14
CN202010961007.5 2020-09-14

Publications (1)

Publication Number Publication Date
WO2022052982A1 true WO2022052982A1 (en) 2022-03-17

Family

ID=80632616

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/117407 WO2022052982A1 (en) 2020-09-14 2021-09-09 USE OF PPARα (PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR α) LIGAND IN PREPARATION OF MEDICINE

Country Status (2)

Country Link
CN (1) CN116367861A (en)
WO (1) WO2022052982A1 (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1321152A (en) * 1998-08-07 2001-11-07 葛兰素集团有限公司 Substd. oxazoles and thiazoles derivs. as hPPAR gamma and hPPAR alpha activators
CN104288758A (en) * 2014-10-22 2015-01-21 中国科学院生物物理研究所 Micellar polypeptide vaccine by taking polyethylene glycol phospholipid as carrier
CN104918922A (en) * 2012-12-20 2015-09-16 因森普深2公司 Triazolone compounds and uses thereof
US20160193190A1 (en) * 2012-02-08 2016-07-07 University Of Cincinnati Targeting Metabolic Adaptive Responses to Chemotherapy
CN106822901A (en) * 2016-11-01 2017-06-13 中山大学 Peroxisome proliferator-activated receptor α is to the direct regulation and control effect of carnitine palmityl transferase 1C and antitumor application thereof

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
SG11201807022XA (en) * 2016-02-25 2018-09-27 Memorial Sloan Kettering Cancer Center Recombinant mva or mvadele3l expressing human flt3l and use thereof as immuno-therapeutic agents against solid tumors
CN108338994A (en) * 2017-01-25 2018-07-31 杭州康万达医药科技有限公司 Application of the oncolytic virus as the immunostimulant for treating tumour and/or cancer

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN1321152A (en) * 1998-08-07 2001-11-07 葛兰素集团有限公司 Substd. oxazoles and thiazoles derivs. as hPPAR gamma and hPPAR alpha activators
US20160193190A1 (en) * 2012-02-08 2016-07-07 University Of Cincinnati Targeting Metabolic Adaptive Responses to Chemotherapy
CN104918922A (en) * 2012-12-20 2015-09-16 因森普深2公司 Triazolone compounds and uses thereof
CN104288758A (en) * 2014-10-22 2015-01-21 中国科学院生物物理研究所 Micellar polypeptide vaccine by taking polyethylene glycol phospholipid as carrier
CN106822901A (en) * 2016-11-01 2017-06-13 中山大学 Peroxisome proliferator-activated receptor α is to the direct regulation and control effect of carnitine palmityl transferase 1C and antitumor application thereof

Non-Patent Citations (5)

* Cited by examiner, † Cited by third party
Title
BRAVO YALDA; BACCEI CHRISTOPHER S.; BROADHEAD ALEX; BUNDEY RICHARD; CHEN AUSTIN; CLARK RYAN; CORREA LUCIA; JACINTHO JASON D.; LORR: "Identification of the first potent, selective and bioavailable PPARα antago", BIOORGANIC & MEDICINAL CHEMISTRY LETTERS, vol. 24, no. 10, 4 April 2014 (2014-04-04), AMSTERDAM, NL , pages 2267 - 2272, XP028646763, ISSN: 0960-894X, DOI: 10.1016/j.bmcl.2014.03.090 *
FANG, LIANG: "Pharmaceutical Polymer Materials", 31 August 2015, CHINA MEDICAL SCIENCE AND TECHNOLOGY PRESS , CN , ISBN: 978-7-5067-7419-2, article FANG, LIANG: "(8) For Surface Modification of NanoFormulations", pages: 166 - 167, XP009535729 *
GAO JIAMING, YUAN SHUPING, JIN JIANHUA, SHI JUANJUAN, HOU YONGZHONG: "PPARα regulates tumor progression, foe or friend?", EUROPEAN JOURNAL OF PHARMACOLOGY, vol. 765, 15 October 2015 (2015-10-15), NL , pages 560 - 564, XP055910935, ISSN: 0014-2999, DOI: 10.1016/j.ejphar.2015.09.027 *
XIAOZHE YIN; WENFENG ZENG; BOWEN WU; ZIHAO WANG; HONGJIAN TIAN; LUYAO WANG; LUOYANG WANG; XIULI WEI; YAN QIN; FAYUN ZHANG; CHUNLIN: "Abstract C109: Tumor derived exosomal lipids-induced dendritic cell immunosuppression by PPARa", MOLECULAR CANCER THERAPEUTICS, vol. 18, no. 12, Suppl., 1 December 2019 (2019-12-01), pages C109, XP009535490, DOI: 10.1158/1535-7163.TARG-19-C109 *
YIN XIAOZHE, ZENG WENFENG, WU BOWEN, WANG LUOYANG, WANG ZIHAO, TIAN HONGJIAN, WANG LUYAO, JIANG YUNHAN, CLAY RYAN, WEI XIULI, QIN : "PPARα Inhibition Overcomes Tumor-Derived Exosomal Lipid-Induced Dendritic Cell Dysfunction", CELL REPORTS, vol. 33, no. 3, 1 October 2020 (2020-10-01), US , pages 1 - 20, XP055910925, ISSN: 2211-1247, DOI: 10.1016/j.celrep.2020.108278 *

Also Published As

Publication number Publication date
CN116367861A (en) 2023-06-30

Similar Documents

Publication Publication Date Title
Huang et al. Antigen-loaded monocyte administration induces potent therapeutic antitumor T cell responses
Radhakrishnan et al. Reprogrammed FoxP3+ T regulatory cells become IL-17+ antigen-specific autoimmune effectors in vitro and in vivo
Silk et al. Utilizing the adjuvant properties of CD1d-dependent NK T cells in T cell–mediated immunotherapy
Xiang et al. Kinsenoside attenuates liver fibro-inflammation by suppressing dendritic cells via the PI3K-AKT-FoxO1 pathway
CN110846281B (en) Anti-tumor vaccine based on exosomes
KR20090051598A (en) Vaccine comprising monocyte or immature myeloid cells(imc) which was loaded with the ligand of natural killer t cell and antigen
Su et al. Anti-tumor efficacy of a hepatocellular carcinoma vaccine based on dendritic cells combined with tumor-derived autophagosomes in murine models
EP3460052B1 (en) Improved allogeneic dendritic cells for use in cancer treatment
US10137153B2 (en) Methods for treating an infectious or neoplastic disease
Xu et al. High-avidity antitumor T-cell generation by toll receptor 8–primed, myeloid-derived dendritic cells is mediated by IL-12 production
WO2022052982A1 (en) USE OF PPARα (PEROXISOME PROLIFERATOR-ACTIVATED RECEPTOR α) LIGAND IN PREPARATION OF MEDICINE
Russo et al. Lymphocytes genetically modified to express tumor antigens target DCs in vivo and induce antitumor immunity
Zhang et al. In Situ biomimetic Nanoformulation for metastatic cancer immunotherapy
KR101055666B1 (en) Vaccine comprising Monocyte or immature myeloid cells(IMC) which was loaded with the ligand of natural killer T cell and antigen
WO2020145222A1 (en) Novel neoantigens and cancer immunotherapy using same
CN109642214A (en) Technology for effective activation NKT cell
US20170274069A1 (en) Method for the cancer treatment and prevention of metastatic disease
EA009327B1 (en) Method for producing antineoplastic vaccine based on surface antigens of endothelial cells
US11850279B2 (en) Platforms and methods for optimizing host antigen presentation and host antitumor and antipathogen immunity
Zou et al. Treatment effect of tuftsin and antigen peptide combined with immune cells on colorectal cancer
Dong et al. Vaccination with FasL-/TCL plus MHSP65 induces improved anti-lung cancer immunity in mice
양수빈 A Hepatitis B Virus-derived Peptide Exerts an Anticancer Effect via TNF/iNOS-producing Dendritic Cells in Tumor-bearing Mouse Model
Jæhger Preclinical Evaluation of Novel Drug Delivery Platforms for the Improvement of Adoptive T cell Therapy
Zhang et al. LKB1 Positively Regulates Dendritic Cell-induced T Cell Immunity and Suppresses Tumor Development
Garcia-Robledo et al. Dendritic cells and glioblastoma

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: 21866034

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: 21866034

Country of ref document: EP

Kind code of ref document: A1