[go: up one dir, main page]

WO2025185599A1 - Mrna tumor vaccine encoding membrane-bound il-12 cytokine adjuvant, and use thereof - Google Patents

Mrna tumor vaccine encoding membrane-bound il-12 cytokine adjuvant, and use thereof

Info

Publication number
WO2025185599A1
WO2025185599A1 PCT/CN2025/080420 CN2025080420W WO2025185599A1 WO 2025185599 A1 WO2025185599 A1 WO 2025185599A1 CN 2025080420 W CN2025080420 W CN 2025080420W WO 2025185599 A1 WO2025185599 A1 WO 2025185599A1
Authority
WO
WIPO (PCT)
Prior art keywords
mrna
tumor
cancer
ova
domain
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/CN2025/080420
Other languages
French (fr)
Chinese (zh)
Other versions
WO2025185599A8 (en
Inventor
傅阳心
梁永
彭琨
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tsinghua University
Original Assignee
Tsinghua University
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 Tsinghua University filed Critical Tsinghua University
Publication of WO2025185599A1 publication Critical patent/WO2025185599A1/en
Publication of WO2025185599A8 publication Critical patent/WO2025185599A8/en
Pending legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/39Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/04Immunostimulants

Definitions

  • the present invention belongs to the field of biomedicine technology, and specifically relates to an mRNA tumor vaccine encoding a membrane-bound IL-12 cytokine adjuvant and uses thereof.
  • Cancer is a major threat to human health. Abnormal cell proliferation or differentiation leads to dysfunction, unregulated cell growth, and local tissue invasion and metastasis. Cancer cells, due to genetic mutations, express specific neoantigens that can be recognized and killed by immune T lymphocytes, forming the cornerstone of tumor immunotherapy.
  • CTLs cytotoxic T lymphocytes
  • Traditional vaccines such as protein-based vaccines combined with alum, often fail to generate robust CTL responses against viral infections or tumors; while stronger adjuvants can enhance CTL responses, they are often accompanied by severe toxicity (Awate et al., 2013; Coffman et al., 2010). Antigen generation and adjuvant selection are crucial for vaccine success.
  • mRNA vaccine platforms offer a promising strategy for generating more potent antigens and effective adjuvants for the prevention of viral infections and the treatment of cancer (Barbier et al., 2022; Beck et al., 2021; Chaudhary et al., 2021).
  • Most clinical mRNA vaccines use methylated bases to prevent innate immune induction and reduce toxicity, although this may also limit the adjuvant effect of mRNA (Andries et al., 2015; Kariko et al., 2005).
  • Lipid nanoparticles (LNPs) for mRNA delivery can act as built-in adjuvants, inducing the production of proinflammatory cytokines such as IL-1 ⁇ and IL-6, thereby promoting antigen-specific CD4+ follicular helper T cell (Tfh) and B cell responses (Alameh et al., 2021; Tahtinen et al., 2022).
  • cytokines such as IL-6 may inhibit the differentiation of naive T cells into effector cells, which are crucial in anti-tumor immune responses (Huseni et al., 2023). Therefore, selecting the right adjuvant for mRNA cancer vaccines is crucial to ensure their efficacy and safety.
  • IL-12 itself is a highly toxic cytokine. Directly injecting IL-12 protein as an adjuvant with RNA vaccine or expressing free IL-12 with mRNA will lead to serious treatment-related side effects. Therefore, how to create an immune microenvironment that is conducive to the differentiation of cytotoxic T cells for the T cell activation process of mRNA vaccine and reduce the side effects of the immune process on the body is crucial to improving the efficacy of mRNA tumor vaccines.
  • the purpose of the present invention is to provide an mRNA tumor vaccine encoding a membrane-bound IL-12 cytokine adjuvant, using RNA technology to simultaneously express IL-12 and tumor antigens on antigen-presenting cells in peripheral lymphoid tissues, and use membrane-bound IL-12 as an adjuvant to improve the therapeutic effect of conventional mRNA tumor vaccines; compared with directly expressing free IL-12 protein, the cell membrane-anchored form of IL-12 protein can limit IL-12 to the surface of antigen-presenting cells in peripheral lymphoid tissues, while not affecting its adjuvant effect and avoiding the release of IL-12 into peripheral tissues to cause peripheral toxicity.
  • the present invention provides an mRNA tumor vaccine encoding a membrane-type IL-12 cytokine adjuvant, wherein the mRNA comprises three parts: a sequence encoding IL-12 (IL12), a sequence encoding a membrane domain (MD), and a sequence encoding a tumor antigen (Ag).
  • IL12 a sequence encoding IL-12
  • MD membrane domain
  • Ag tumor antigen
  • the sequence encoding IL-12 (IL12) and the sequence encoding the membrane domain (MD) are located on one mRNA, and the sequence encoding the tumor antigen (Ag) is located on another mRNA; or the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD) and the sequence encoding the tumor antigen (Ag) are located on the same mRNA, and the arrangement order of the three parts is IL12-L1-MD-L2-Ag, or Ag-L2-IL12-L1-MD; wherein L1 is a glycine/serine (Glycine/Serine, Gly/Ser) polypeptide linker, L2 is an IRES sequence or a Glycine/Serine or a 2A polypeptide linker, and L2 is preferably a 2A polypeptide linker.
  • L1 is a glycine/serine (Glycine/Serine, Gly/Ser) polypeptide linker
  • the tumor antigen includes a tumor neoantigen derived from a mutation, a tumor-specific antigen derived from a virus, or a tumor-related antigen that is highly expressed.
  • the mutation-derived tumor neoantigens include KARS G12C, KRASG12D, EGFRvIII or BRAF V600E ;
  • the viral-derived tumor-specific antigens include HPV E6/E7, endogenous retroviral antigen hERT, LMP1 or LMP2;
  • the tumor-related antigens highly expressed include WT1, MAGE-A3, GP100, NY-ESO-I, HER2/Neu, Claudin18.2, Mesothelin or MUC.
  • the sequence encoding the IL-12 cytokine comprises an ORF encoding an IL-12B polypeptide and an IL-12A polypeptide, wherein IL-12B and IL-12A are connected by a polypeptide linker, the linker comprises a Gly/Ser linker, and the Gly/Ser linker comprises (GnS)m, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 and m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20; the linker is preferably a (G4S) 3 linker.
  • the membrane domain may include a transmembrane domain, preferably a transmembrane domain plus an intracellular domain
  • the transmembrane domain includes the CD80 transmembrane domain, the CD86 transmembrane domain, the CD4 transmembrane domain, the CD8 transmembrane domain, the VEGFR transmembrane domain, and the PDGF-RB transmembrane domain, preferably the CD80 transmembrane domain
  • the intracellular domain includes the CD80 intracellular domain, the CD86 intracellular domain, the CD4 intracellular domain, the CD8 intracellular domain, the VEGFR intracellular domain, the PDGF-RB intracellular domain, and a truncation of the PDGF-RB intracellular domain
  • the intracellular domain and the transmembrane domain may come from the same gene combination or different gene combinations; preferably, a combination of the CD80 transmembrane domain plus the CD80 intracellular domain.
  • the mRNA tumor vaccine further comprises a nucleic acid vector loaded with mRNA, and the nucleic acid vector comprises liposomes, LPP or LPX, preferably LNP nanoliposomes.
  • the present invention further provides a pharmaceutical composition
  • a pharmaceutical composition comprising the mRNA tumor vaccine of the present invention and a second anticancer agent, wherein the second anticancer agent comprises an immune checkpoint inhibitor and/or a bispecific T cell engager (BiTE).
  • the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.
  • the present invention further provides the use of the mRNA tumor vaccine or pharmaceutical composition of the present invention in the preparation of a medicament for treating or preventing tumors. In yet another aspect, the present invention further provides the use of the mRNA tumor vaccine or pharmaceutical composition of the present invention in treating or preventing tumors. In yet another aspect, the present invention further provides a method for treating or preventing tumors in a subject, the method comprising administering the mRNA tumor vaccine or pharmaceutical composition of the present invention to the subject.
  • the tumor is selected from B-cell lymphoma, bronchial cancer, prostate cancer, bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, head and neck cancer, cervical cancer, uterine or endometrial cancer, oral cancer, laryngeal cancer, salivary gland cancer, thymic cancer, adrenal cancer, osteosarcoma, chondrosarcoma, adipose cancer, testicular cancer, malignant fibrous histiocytoma, colorectal cancer, melanoma, gastric cancer, pancreatic cancer, lung cancer, liver cancer, kidney cancer, bile duct cancer, small intestine cancer or appendix cancer, squamous cell carcinoma, breast cancer and ovarian cancer.
  • the present invention we have successfully improved the therapeutic effect of existing mRNA tumor vaccines by adding a cell membrane-anchored IL-12 sequence to mRNA.
  • the cell membrane-anchored form of IL-12 protein can limit IL-12 to the surface of antigen-presenting cells in peripheral lymphoid tissue, greatly reducing the release of IL-12 protein into the peripheral blood without affecting the adjuvant effect of IL-12, and significantly reducing the systemic toxicity produced by IL-12 protein.
  • the addition of membrane-anchored IL-12 sequences can significantly improve the therapeutic effect of mRNA vaccines. Therefore, we believe that mRNA tumor vaccines based on membrane-anchored IL-12 have the potential to become a new generation of tumor therapeutic vaccines and achieve better therapeutic effects in a variety of clinical tumors.
  • Figure 1 shows that IL-12 can act as an adjuvant to enhance the antitumor efficacy of mRNA vaccines.
  • Six hours after vaccination 2.5 ⁇ g of IL2-Fc (C), 5 ⁇ g of IL15-Fc (D), or 0.25 ⁇ g of IL12-Fc protein (E) mixed with sodium alginate was injected intramuscularly into the same site.
  • C-E Tumor growth curves for each treatment are shown. Data are presented as mean ⁇ standard error of variance (SEM) and are from two independent experiments. P values were determined by two-way analysis of variance with Bonfferoni's multiple comparison test (C-E). **P ⁇ 0.01, ***P ⁇ 0.001, ****P ⁇ 0.0001.
  • FIG. 2 shows the construction of RNA tumor vaccines adjuvanted with free or membrane-anchored IL12.
  • A Schematic diagram of the structures of the free sIL12-OVA and membrane-anchored mtIL12-OVA mRNA vaccines.
  • B DC2.4 cells were transfected with CTRL mRNA (top), mtEGFP-OVA RNA (center), or mtIL12-OVA mRNA (bottom). 24 hours after transfection, cells were harvested and stained with antibodies against IL12p40 and OVA peptide-MHC complexes, and the expression of membrane IL12 or OVA-pMHC complexes was analyzed by flow cytometry.
  • C DC2.4 cells were transfected with mtIL12-OVA mRNA or sIL12-OVA mRNA. 24 hours after transfection, cell culture fluid was harvested to measure the concentration of secreted free IL12.
  • Figure 3 shows that RNA tumor vaccines adjuvanted with membrane-anchored IL12 exhibit similar antitumor efficacy but lower peripheral toxicity compared to vaccines adjuvanted with free IL12.
  • Figure 4 shows that the mtIL12 mRNA vaccine specifically activates antigen-specific T cells but does not affect bystander T cells or NK cells.
  • C57BL/6J mice received 5 ⁇ g of mtIL12 mRNA or 5 ⁇ g of sIL12 mRNA by intramuscular injection. 36 hours later, peripheral blood was collected and blocked with BFA solution for 6 hours, and the proportion of IFN ⁇ -positive cell subsets was analyzed by flow cytometry.
  • Figure 5 shows that the mtIL12-OVA mRNA vaccine has a superior anti-tumor effect compared to OVA mRNA or mtIL12 mRNA alone.
  • Tumor-bearing mice were injected intramuscularly with 2.5 ⁇ g of mRNA encoding mtEGFP-OVA, mtIL12-OVA, or mtIL12 on days 8 and 12, and tumor growth curves were recorded.
  • Figure 6 shows that the antitumor effect of an RNA tumor vaccine containing a membrane-anchored IL12 adjuvant is dependent on CD8 + T cells.
  • Anti-NK1.1 (200 ⁇ g), anti-CD8 (200 ⁇ g), anti-CD4 (200 ⁇ g), and anti-CSF1R (500 ⁇ g) antibodies were administered one day before treatment and continued twice weekly for the next two weeks.
  • Tumor growth curves (A) and survival curves (B) were recorded.
  • C C57BL/6J mice received 2.5 ⁇ g of mtEGFP-OVA mRNA or 2.5 ⁇ g of mtIL12-OVA mRNA via intramuscular injection on days 0 and 5.
  • OVA-specific T cells were measured in the draining lymph nodes on day 8 after the first vaccination and in the spleen on day 13 after the first vaccination.
  • Figure 7 shows that the mtIL12-based vaccine produces stronger effector T cells in the OVA antigen model.
  • B6 mice were immunized with 2.5 ⁇ g mtEGFP-OVA or mtIL12-OVA RNA intramuscularly on days 0 and 7, and blood was collected on day 14 for flow cytometry analysis.
  • B Flow cytometry analysis showed that IL12 immunization promoted the differentiation of OVA-specific T cells from a precursor phenotype (Ly108+CD69-) to an effector phenotype (Ly108-CD69-).
  • C Peripheral blood cells were incubated with the OT1 peptide (SIINFEKL) for 3 hours and then analyzed by flow cytometry.
  • OVA-specific T cells from mice immunized with the IL12 vaccine expressed higher levels of effector molecules (GZMB, IFN-gamma), higher expression of the effector T cell-specific transcription factor T-bet, and lower expression of the immunosuppressive molecule PD-1.
  • GZMB effector molecules
  • IFN-gamma effector molecules
  • T-bet T cell-specific transcription factor
  • PD-1 immunosuppressive molecule
  • FIG. 8 Demonstrates that cis-delivery of IL12 and antigen can generate more antigen-specific T cells.
  • A B6 mice were intravenously injected with 2 ⁇ g mtEGFP-OVA, 2 ⁇ g mtEGFP-OVA + 1 ⁇ g mtIL12, and 1 ⁇ g mtEGFP-OVA + 1 ⁇ g mtIL12-OVA RNA-LNP on days 0 and 7, and blood was collected on day 14 for flow cytometric analysis.
  • mice treated with the cis-delivery of IL12 and antigen vaccine (1 ⁇ g mtEGFP-OVA + 1 ⁇ g mtIL12-OVA) generated higher numbers of antigen-specific T cells in peripheral blood.
  • Figure 9 shows that membrane-bound IL12 adjuvanted mRNA vaccines overcome resistance to immune checkpoint blockade.
  • Anti-CTLA4 (200 ⁇ g) and anti-PD1 (200 ⁇ g) were given on day 6 after the first vaccination and continued twice a week for two weeks. Tumor growth curves and mouse survival were recorded.
  • FIG. 10 shows that the mtIL12-E7 vaccine inhibits the growth of HPV-associated TC-1 tumors.
  • Tumor-bearing mice were treated with an intramuscular injection of 1 ⁇ g of mRNA encoding mtEGFP-E7, mtIL12-E7, or mtIL12 on day 14 and boosted with 2 ⁇ g of the same mRNA vaccine on day 20.
  • Tumor growth curves and mouse survival (A) and body weight changes (B) were recorded. It can be seen that neither the control E7 vaccine nor mtIL12 alone failed to control tumor growth in mice bearing advanced TC-1 tumors.
  • mice treated with the mtIL12-E7 mRNA vaccine did not experience significant weight loss compared to the untreated group (B).
  • FIG. 11 shows that the mtIL12-based vaccine generates enhanced effector T cells in the E7 antigen model.
  • A TC-1 tumor-bearing mice were intramuscularly injected with 2.5 ⁇ g mtEGFP-E7 on day 12 after tumor implantation. On day 18, they were immunized with either 2.5 ⁇ g mtEGFP-E7 or 2.5 ⁇ g mtIL12-E7. Tumors were harvested and analyzed by flow cytometry on day 22.
  • B Flow cytometry analysis shows the gates and expression of selected surface markers for antigen-specific T cells.
  • C Flow cytometry analysis shows that IL12 immunization promotes the intratumoral enrichment of E7-specific T cells.
  • T cells immunized with mtIL12-E7 are more likely to remain in the effector state (LY108-CD69-) rather than the exhausted state (LY108-CD69+). They also express higher levels of the effector molecule GZMB and lower levels of the inhibitory marker PD1.
  • FIG 12 shows that mtIL12 adjuvant can be applied to the design of mRNA vaccines for endogenous retroviral-derived tumor neoantigens.
  • P15E protein derived from gp70 retroviral gene expression, has been detected in many cancers, including the MC38 colon adenocarcinoma cell line (Ye et al., 2020). 4 ⁇ 10 5 MC38 tumor cells were subcutaneously injected into C57BL/6J mice on day 0. After tumor formation, mice were injected intramuscularly with 2.5 ⁇ g of mtIL12-p15E or control mtEGFP-p15E mRNA-LNP on days 12 and 16, and tumor growth curves were monitored twice a week. As shown in Figure 7, control P15E mRNA vaccine treatment barely inhibited tumor growth, while mtIL12-P15E treatment induced significant tumor regression and complete eradication (Figure 7).
  • FIG. 13 shows that the mtIL12-based vaccine generates enhanced effector T cells in the p15E antigen model.
  • A MC38 tumor-bearing mice were intramuscularly injected with 2.5 ⁇ g mtEGFP-p15E mRNA on day 12 after tumor inoculation. They were then immunized again with either 2.5 ⁇ g mtEGFP-p15E or 2.5 ⁇ g mtIL12-p15E mRNA on day 18. Peripheral blood was collected for flow cytometry analysis on day 21.
  • B Flow cytometry analysis showed that mtIL12-p15E immunization generated more p15E-specific T cells. Furthermore, compared to T cells immunized with mtEGFP-p15E, these T cells expressed higher expression of the effector molecules GZMB and IFN-gamma, and lower expression of the inhibitory marker PD1.
  • the present invention provides an mRNA tumor vaccine encoding a membrane-type IL-12 cytokine adjuvant, wherein the mRNA comprises three parts: a sequence encoding IL-12 (IL12), a sequence encoding a membrane domain (MD), and a sequence encoding a tumor antigen (Ag).
  • IL12 a sequence encoding IL-12
  • MD membrane domain
  • Ag tumor antigen
  • the sequence encoding IL-12 (IL12) and the sequence encoding the membrane domain (MD) are located on one mRNA, and the sequence encoding the tumor antigen (Ag) is located on another mRNA; or the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD) and the sequence encoding the tumor antigen (Ag) are located on the same mRNA, and the arrangement order of the three parts is IL12-L1-MD-L2-Ag, or Ag-L2-IL12-L1-MD; wherein L1 is a Glycine/Serine polypeptide linker, and L2 is an IRES sequence or a Glycine/Serine or 2A polypeptide linker.
  • the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD), and the sequence encoding the tumor antigen (Ag) are located on the same mRNA.
  • IL12 IL-12
  • MD membrane domain
  • Ag tumor antigen
  • the same mRNA can ensure that IL12, MD, and Ag are expressed simultaneously in the same cell, avoiding the situation where some cells only express IL-12-MD or Ag due to differences in the transfection efficiency of the two mRNAs. It can more accurately achieve the synchronous expression of the three, ensure their coordination in quantity and time, and facilitate the synergistic effect between them.
  • the synthesis, purification, and quality control costs of a single mRNA are significantly lower than the parallel production of two independent mRNAs.
  • the tumor antigens described in the present invention may include tumor neoantigens of mutation origin such as KARS G12C, KRAS G12D, EGFRvIII and BRAF V600E ; tumor-specific antigens of viral origin such as HPV E6/E7, endogenous retrovirus antigens (hERT), LMP1 and LMP2; tumor-related antigens highly expressed such as WT1, MAGE-A3, GP100, NY-ESO-I, HER2/Neu, Claudin18.2, Mesothelin and MUC, etc.
  • the type of antigen can be changed according to the tumor-specific antigen.
  • Ovalbumin is a protein antigen that is widely used to induce cellular and humoral immune responses in cancer immunotherapy and is commonly used as a tumor model antigen in the art.
  • OVA is used as an example of a tumor-specific antigen.
  • Interleukin-12 is a multipotent cytokine encoded by two independent genes, IL-12A (p35) and IL-12B (p40), which exist as an active heterodimer (p70) or a homodimer of p40 (p80).
  • IL-12B and IL-12A are linked by a polypeptide linker comprising a Gly/Ser linker comprising (GnS)m, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 and m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20.
  • the two subunits of the IL-12 cytokine are linked by a (G4S) 3 linker.
  • the membrane domain of the present invention can be a transmembrane domain plus an intracellular domain, and the transmembrane domain includes the CD80 transmembrane domain, the CD86 transmembrane domain, the CD4 transmembrane domain, the CD8 transmembrane domain, the VEGFR transmembrane domain, and the PDGF-RB transmembrane domain, preferably the CD80 transmembrane domain; the intracellular domain includes the CD80 intracellular domain, the CD86 intracellular domain, the CD4 intracellular domain, the CD8 intracellular domain, the VEGFR intracellular domain, the PDGF-RB intracellular domain, and a truncated PDGF-RB intracellular domain.
  • the intracellular domain and the transmembrane domain can be derived from the same gene combination or different gene combinations. For example, the following transmembrane domain sequence and intracellular domain sequence can be added together, but the present invention is not limited thereto:
  • VVISAILALVVLTIISLIILI Human PDGF-RB transmembrane domain, VVISAILALVVLTIISLIILI (SEQ ID NO: 3)
  • IL-2 is essential for T cell activation and IL-15 is highly effective in expanding T cell numbers (Guo et al., 2021; Zou et al., 2024), we observed limited efficacy of these cytokines in enhancing antitumor activity in the context of mRNA vaccines.
  • IL-12 can induce a Th1-type immune microenvironment and promote the differentiation of CD8+ T cells with tumor-killing potential (Hewitt et al., 2020; Tucker et al., 2020; Tugues et al., 2015; Zou et al., 2024).
  • IL-12 was the most potent cytokine adjuvant for enhancing the antitumor effects of mRNA cancer vaccines.
  • mRNA vaccine expressing a tumor antigen and a membrane-bound IL-12 adjuvant.
  • IL-12 induces and expands an understudied subset of pre-effector T cells.
  • mtIL-12 could provide a robust antitumor response while minimizing toxicity.
  • mice C57BL/6J, BALB/c, and C57BL/6J-Tg(TcraTcrb)1100Mjb/J (OT1 TCR transgenic) mice, 6–8 weeks old, were purchased from Vital River or Jackson Laboratory. Mice were housed in a SPF environment, and all animal experiments were conducted in accordance with the Regulations for Laboratory Animal Care of Tsinghua University.
  • MC38, B16F10, TC-1, and DC2.4 cell lines were purchased from the American Type Culture Collection (ATCC). Freestyle 293-F (R79007) was purchased from Invitrogen.
  • the B16F10-OVA cell line was derived from a single-cell clone after lentiviral expression of OVA. All cell lines were routinely tested for mycoplasma contamination.
  • MC38, B16F10, and TC-1 cells were cultured in Dulbecco's modified Eagle's medium (DMEA) supplemented with 10% heat-inactivated fetal bovine serum, 100 U/mL penicillin, and 100 U/mL streptomycin at 37°C and 5% CO2 .
  • DMEA Dulbecco's modified Eagle's medium
  • DC2.4 cells were cultured in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum, 2 mmol/L L-glutamine, 0.1 mmol/L MEM non-essential amino acids, 100 U/mL penicillin, and 100 U/mL streptomycin at 37°C in 5% CO2.
  • 293-F cells were cultured in SMM 293-TI medium (M293TI; Sino Biological).
  • Anti-CD4 (clone GK1.5), anti-CD8 (clone 53-5.8), anti-mCSF1R (clone AFS98), anti-NK1.1 (clone PK136), anti-PD-L1 antibodies (10F.9G2), anti-mCTLA4 (clone 4F10), and anti-mIL12p75 (clone JR2-9A5) were purchased from Bio X Cell.
  • IL2-Fc, IL15-Fc, and IL12-Fc were produced in-house. Cytokine coding sequences were fused to the human IgG1 Fc fragment via a GGGGS linker and cloned into the pEE12.4 vector. Protein-encoding plasmids were transfected into 293F cells, and the supernatant was purified by Protein-A affinity chromatography (GE Healthcare) according to established protocols.
  • mice MC38 cells (4 ⁇ 105 ), TC-1 cells (3 ⁇ 105 ), and B16F10-OVA cells (2-5 ⁇ 105 ) were inoculated subcutaneously on the right dorsum of mice. After tumor establishment, tumor length (a) and width (b) were measured three times weekly, and tumor volume was calculated as a*b*b/2. Mice were randomly grouped according to tumor size. At specific time points, mice were injected with 1-5 ⁇ g of mRNA-nanoliposome complexes encoding different coding regions, or with empty liposomes as a control.
  • mice were intraperitoneally injected with 200 ⁇ g of anti-CD8 antibody, 200 ⁇ g of anti-CD4 antibody, 200 ⁇ g of anti-NK1.1 antibody, or 500 ⁇ g of anti-CSFIR antibody every three days, starting one day before mRNA vaccination.
  • 2 ⁇ g of IL2-Fc, 3.5 ⁇ g of IL15-Fc, and 0.25 ⁇ g of IL12-Fc were mixed with sodium alginate and injected intramuscularly.
  • 200 ⁇ g of anti-PD-L1 antibody and 200 ⁇ g of anti-CTLA4 antibody were injected intraperitoneally every three days.
  • mice were sacrificed when the length, width or height of the tumor exceeded 2 cm, or the tumor size exceeded 1500 mm 3 , or the body weight of tumor-bearing mice decreased by more than 20%.
  • CBA Cytometric Bead Array
  • the CBAMouse Th1/Th2/Th17 Kit (BD Biosciences) was used according to the manufacturer's instructions to measure cytokine levels in mouse serum and tumor tissue homogenates.
  • ELISA was performed using a 96-well microplate (Corning Costar). 2 ⁇ g/mL (100 ⁇ L/well) of capture antibody was added to the plate and adsorbed overnight at 4°C. The plate was washed with PBS and then blocked with blocking buffer (PBS containing 0.05% TWEEN-20 and 5% skim milk). Serum or tumor homogenate diluted in blocking buffer was then added and incubated at 37°C for 1.5 hours.
  • blocking buffer PBS containing 0.05% TWEEN-20 and 5% skim milk
  • splenocytes from mice were stimulated with 1 mM SIINFEKL peptide (New England Peptide) at a concentration of 108 cells/mL. After a 2-hour peptide pulse, the peptide was washed away, and splenocytes were cultured at a concentration of 106 cells/mL in T cell culture medium supplemented with 5 ng/mL mouse recombinant IL-2 (mrIL-2) and 10 ng/mL mrIL-12 (R&D), or 5 ng/mL mrIL-2 alone, and supplemented daily. On day 4, 2 ⁇ 106 Ficoll-separated, viable OT-1 cells were adoptively transferred into B16-OVA tumor-bearing mice via intravenous injection.
  • SIINFEKL peptide New England Peptide
  • mice 10 5 naive OT-1 cells were transferred into naive mice, and 24 hours after cell transfer, the mice were immunized with 10 ⁇ g of mtEGFP-OVA or mtIL12-OVA mRNA vaccine. Seven days after immunization, activated OT-1 cells were isolated from the spleen of the mice, purified, and 5 ⁇ 10 5 OT-1 cells were transferred intravenously into B16-OVA tumor-bearing mice.
  • DNA plasmid containing the complete sequence of mRNA and carrying poly-A sequence was digested to prepare in vitro transcription template.
  • Co-transcription capping was performed using the T7 Co-transcription RNA Synthesis Kit (C3111).
  • mRNA produced by in vitro transcription was decapped using the cellulose method and stored at -80°C for subsequent experiments.
  • mRNA was dissolved in 100 mM sodium citrate buffer (pH 4.0).
  • DSPC, cholesterol, DMG-PEG2000, and SM102 were dissolved in anhydrous ethanol at a molar ratio of 38.5:10:1.5:50.
  • the LNP:RNA mass ratio was adjusted between 30:1 and 50:1.
  • the RNA in sodium citrate and the LNP in anhydrous ethanol were then mixed at a volume ratio of 3:1 using a microfluidic system to prepare mRNA-LNP nanoliposomes.
  • the mRNA nanoliposome solution was dialyzed against PBS buffer. The dialyzed mRNA nanoliposomes were suitable for in vitro and in vivo animal experiments.
  • mice were randomly assigned to different groups based on tumor volume. Data were analyzed using GraphPad Prism statistical software and presented as mean ⁇ SEM. P values were calculated using two-way ANOVA for tumor curves and log-rank tests for mouse survival curves. All other data were analyzed using unpaired two-tailed t-tests. A p value ⁇ 0.05 indicated significant differences.
  • Example 1 IL-12 enhances the efficacy of mRNA tumor vaccines to achieve superior tumor control
  • T lymphocyte-stimulating cytokines including IL2, IL15, and IL12, for tumor therapy. These cytokines are known to promote T cell proliferation, activation, or effector differentiation through different downstream transcription factors (Propper and Balkwill, 2022).
  • IL2 T lymphocyte-stimulating cytokines
  • IL15 T cell proliferation, activation, or effector differentiation through different downstream transcription factors
  • IL12 T cell proliferation, activation, or effector differentiation through different downstream transcription factors
  • Membrane-bound IL-12-based mRNA vaccine achieves superior anti-tumor efficacy with limited toxicity
  • IL-12 as an adjuvant for mRNA vaccines, we constructed an IL12-OVA fusion mRNA vaccine for treatment of an OVA-expressing B16F10-OVA tumor model.
  • the secreted form of IL-12 was linked to the OT1 epitope-encoding sequence via a 2A linker (sIL12-OVA) ( Figure 2A, upper panel, sequence 19, i.e., SEQ ID NO: 67).
  • the IL-12 coding sequence was replaced with the less immunogenic EGFP (Skelton et al., 2001).
  • IL-12 is known to activate CD4, CD8, and NK cells and induce potent IFN- ⁇ secretion (Guo et al., 2012).
  • mtIL12 membrane-bound IL12
  • mtIL12-OVA membrane-bound IL12 adjuvanted mRNA vaccine
  • IL-12 was fused to the transmembrane domain of the CD80 molecule and then linked to the tumor antigen via a 2A linker (Figure 2A, lower panel, sequence 10, i.e., SEQ ID NO: 58).
  • Figure 2A lower panel, sequence 10, i.e., SEQ ID NO: 58.
  • Figure 2B sequence 10, i.e., SEQ ID NO: 58.
  • sIL12-OVA vaccine significantly improved therapeutic efficacy and prolonged tumor control compared with the control mRNA vaccine ( Figure 3A, sIL12-OVA sequence 19 (SEQ ID NO: 67), control sequence S1 (SEQ ID NO: 69)).
  • Figure 3B sIL12-OVA mRNA vaccine caused a significant decrease in body weight in vaccinated mice ( Figure 3B), indicating the presence of systemic toxicity.
  • mtIL12-OVA showed similar tumor growth inhibition and survival-prolonging effects as sIL12-OVA ( Figure 3A, mtIL12-OVA sequence 10 (SEQ ID NO: 58), sIL12-OVA sequence 19 (SEQ ID NO: 67)).
  • mtIL12-OVA administration did not exacerbate the toxicity associated with soluble IL12.
  • the body weight decreased briefly but recovered rapidly ( Figure 3B).
  • mtIL12-OVA-induced serum IL-12, IFN- ⁇ , and MCP-1 levels were significantly reduced (Figure 2C).
  • IL-12 was only detected on the surface of monocytes, dendritic cells, and macrophages within dLNs (Figure 4A). These results indicate that membrane-bound IL12 effectively restricts IL-12 to the surface of APCs, preventing its release into peripheral tissues.
  • Example 3 mtIL12-based mRNA vaccines rely on CD8 T cells to achieve tumor control and induce unique pre-effector CD8 T cell subsets
  • IL-12-based therapies can act on multiple immune cell types, including CD4+ T cells, CD8+ T cells, NK cells, and macrophages, to optimize tumor control (Xue et al., 2022).
  • CD4+ T cells CD8+ T cells
  • NK cells NK cells
  • macrophages to selectively delete specific immune cell subsets.
  • Deletion of NK cells, CD4+ T cells, or macrophages did not affect the anti-tumor effect of the vaccine ( Figure 6A and B, sequence 19 (SEQ ID NO:67)).
  • deletion of CD8+ T cells greatly impaired vaccine-mediated tumor suppression and mouse survival benefits, indicating that CD8+ T cell responses are critical for the anti-tumor effect of the vaccine.
  • mice immunized with mtIL12-OVA or control OVA mRNA vaccines stimulated them with different dilutions of SIINFEKL peptide, and analyzed IFN- ⁇ secretion by ELISA.
  • Baseline IFN- ⁇ secretion was low in both groups, but after peptide stimulation, splenocytes from both groups secreted IFN- ⁇ in a dose-dependent manner, and the amount detected in the mtIL12-OVA group was significantly higher (Figure 7D).
  • peripheral antigen-specific T cells induced by the mtIL12-OVA vaccine are highly potent pre-effector T cells that exhibit higher sensitivity and stronger effector responses when they encounter antigen again, thereby achieving enhanced tumor suppression compared to traditional OVA mRNA vaccines.
  • mice were intravenously injected with 2 ⁇ g mtEGFP-OVA, 2 ⁇ g mtEGFP-OVA + 1 ⁇ g mtIL12, or 1 ⁇ g mtEGFP-OVA + 1 ⁇ g mtIL12-OVA RNA-LNP on days 0 and 7. Blood was collected on day 14 for flow cytometry analysis (Figure 8A).
  • mice delivered with IL12 and antigen in cis (OVA+mtIL12-OVA) generated a higher proportion and number of antigen-specific T cells in peripheral blood ( Figure 8B, Sequence S1 (SEQ ID NO: 69), Sequence 19 (SEQ ID NO: 67), Sequence S15 (SEQ ID NO: 83)).
  • Immune checkpoint blockade (ICB) therapy has shown promising results in the clinical setting (Hargadon et al., 2018); however, only a minority of patients achieve effective responses (Sharma et al., 2017).
  • TILs tumor-infiltrating lymphocytes
  • mtIL12-adjuvanted mRNA cancer vaccines could overcome resistance to ICB therapy by providing sufficient antigen-specific TILs to immune-cold tumors.
  • Example 5 Vaccine design using membrane-bound IL12 adjuvant for E6/E7 antigens in HPV-related tumors
  • mice bearing advanced TC-1 tumors neither the control E7 vaccine nor mtIL12 treatment alone failed to control tumor growth (Figure 10A, Sequence S15 (SEQ ID NO:83), Sequence S4 (SEQ ID NO:72), Sequence 18 (SEQ ID NO:66)).
  • Figure 10A Sequence S15 (SEQ ID NO:83), Sequence S4 (SEQ ID NO:72), Sequence 18 (SEQ ID NO:66)
  • TC-1 tumors continued to grow to approximately 500 mm 3 after two doses of mtIL12-E7 vaccination, they ultimately regressed, with approximately 80% of tumors successfully eradicated ( FIG10A ).
  • mice treated with the mtIL12-E7 mRNA vaccine did not experience significant weight loss compared to the untreated group ( FIG10B ).
  • TC-1 tumor-bearing mice were immunized with 2.5 ⁇ g mtEGFP-E7 on day 12 of tumor implantation and with either 2.5 ⁇ g mtEGFP-E7 or 2.5 ⁇ g mtIL12-E7 on day 18.
  • Tumor tissue was harvested and analyzed by flow cytometry on day 22 ( Figure 11A). The results showed that IL12 immunization promoted the intratumoral enrichment of E7-specific T cells.
  • T cells immunized with mtGFP-E7 were more likely to remain in the effector state (LY108-CD69-) rather than the exhausted state (LY108-CD69+), and expressed higher levels of the effector molecule GZMB and lower levels of the inhibitory marker PD-1 ( Figure 11C).
  • Membrane-bound IL12 adjuvant is suitable for vaccine design against retroviral-derived tumor neoantigens
  • Endogenous retroviruses integrate into the host genome and are typically silent in healthy tissues, but their expression is reactivated under pathological conditions such as cancer (Cherkasova et al., 2013; Smith et al., 2018). These reactivated ERV proteins provide viable targets for cancer vaccine development.
  • the p15E protein derived from the gp70 retroviral gene, has been detected in many cancers, including the MC38 colon adenocarcinoma cell line (Ye et al., 2020). C57BL/6 mice were subcutaneously inoculated with 4 ⁇ 10 5 MC38 tumors.
  • mice were injected intramuscularly with 2.5 ⁇ g of mtIL12-p15E or control mtEGFP-p15E mRNA-LNP on days 12 and 16 after tumor establishment, and tumor growth curves were monitored twice weekly. The results are shown in Figure 12.
  • the control P15E mRNA vaccine treatment had minimal effect on tumor growth, while mtIL12-P15E treatment induced significant tumor regression and complete eradication (Figure 12, Sequence S5 (SEQ ID NO: 73), Sequence S9 (SEQ ID NO: 77)).
  • mice MC38 tumor-bearing mice were immunized with 2.5 ⁇ g of mtEGFP-p15E intramuscularly on day 12 after tumor implantation.
  • 2.5 ⁇ g of mtEGFP-p15E or 2.5 ⁇ g of mtIL12-p15E was injected intramuscularly.
  • Peripheral blood was collected on day 21 for flow cytometry analysis ( Figure 13A). The results showed that compared to the mtEGFP-p15E mRNA vaccine, mtIL12-p15E immunization resulted in more p15E-specific T cells.
  • T cells immunized with mtGFP-p15E expressed higher levels of the effector molecule IFN-gamma and lower levels of the immunosuppressive molecule PD-1.
  • Cytokines act as a tertiary signal during T cell activation.
  • the self-adjuvant effect of mRNA-LNPs primarily induces the production of proinflammatory cytokines, including IL-1 ⁇ , IL-6, and type I interferons (Li et al., 2022; Tahtinen et al., 2022), which may not optimally support antigen-specific CD8+ T cell responses.
  • IL-12 was the most effective cytokine in enhancing the antitumor effect of mRNA vaccines.
  • T cell dysfunction is characterized by established mechanisms by which tumor cells evade T cell-mediated cytotoxicity (Zebley et al., 2024). Studies have shown that naive tumor-specific T cells rapidly acquire a dysfunctional phenotype within hours of entering the TME (Philip et al., 2017; Rudloff et al., 2023). In our studies, conventional mRNA vaccines expanded a large number of progenitor-like antigen-specific T cells, but these cells exhibited limited antitumor activity. We speculate that similar mechanisms may hinder the differentiation of progenitor T cells generated by conventional mRNA vaccines into functional effector cells.
  • Introducing IL-12 signaling during the priming phase of mRNA vaccines facilitates the differentiation of antigen-specific T cells into a pre-effector phenotype.
  • these pre-effector CD8+ T cells secrete few effector cytokines in circulation but exhibit increased sensitivity and stronger effector responses upon re-encountering tumor antigens, which may help explain their limited peripheral toxicity but enhanced antitumor efficacy.
  • These cells also express lower levels of inhibitory PD-1 and display resistance to the suppressive TME, enhancing their antitumor capacity.
  • membrane-bound IL-12 as an adjuvant significantly enhanced the therapeutic efficacy of mRNA tumor vaccines while minimizing toxicity.
  • This strategy is applicable to a variety of clinically relevant tumor antigens and has considerable potential for clinical translation.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Immunology (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Epidemiology (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The present invention relates to an mRNA tumor vaccine encoding a membrane-bound IL-12 cytokine adjuvant, and the use thereof. By means of the mRNA tumor vaccine, the toxicity of IL-12 is reduced and the therapeutic effect of the mRNA tumor vaccine is improved. The mRNA contains three parts: a sequence (IL12) encoding IL-12, a sequence (MD) encoding a membrane domain, and a sequence (Ag) encoding a tumor antigen.

Description

编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗及其用途mRNA tumor vaccine encoding membrane-bound IL-12 cytokine adjuvant and its use

相关申请交叉引用Cross-reference to related applications

本申请要求于2024年3月4日提交的中国国家申请号202410245327.9的优先权权益,所述申请通过引用整体并入本文。This application claims the benefit of priority to Chinese National Application No. 202410245327.9, filed on March 4, 2024, which is incorporated herein by reference in its entirety.

技术领域Technical Field

本发明属于生物医药技术领域,具体的,涉及一种编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗及其用途。The present invention belongs to the field of biomedicine technology, and specifically relates to an mRNA tumor vaccine encoding a membrane-bound IL-12 cytokine adjuvant and uses thereof.

背景技术Background Art

癌症是人类健康的重大威胁疾病,由于细胞增殖或分化异常,导致功能异常、细胞生长不受调控,局部组织侵袭和转移。癌症细胞由于基因突变表达特异的肿瘤新生抗原,可以被免疫T淋巴细胞识别和杀伤,是肿瘤免疫治疗的基础。Cancer is a major threat to human health. Abnormal cell proliferation or differentiation leads to dysfunction, unregulated cell growth, and local tissue invasion and metastasis. Cancer cells, due to genetic mutations, express specific neoantigens that can be recognized and killed by immune T lymphocytes, forming the cornerstone of tumor immunotherapy.

针对免疫原性肿瘤的细胞毒性T淋巴细胞(CTLs)数量不足是免疫治疗面临的一个重大挑战(Kalbasi和Ribas,2020)。传统的疫苗,例如基于蛋白质的疫苗与明矾结合,通常无法产生针对病毒感染或肿瘤的强烈CTL反应;而更强的佐剂虽然可以增强CTL反应,但往往伴随着严重的毒性(Awate等人,2013;Coffman等人,2010)。抗原的产生和佐剂的选择对于疫苗的成功至关重要。近年来开发的mRNA疫苗平台为产生更强大的抗原和有效的佐剂提供了一个有前景的策略,用于预防病毒感染和治疗癌症(Barbier等人,2022;Beck等人,2021;Chaudhary等人,2021)。为了增加抗原表达,大多数临床mRNA疫苗使用甲基化修饰的碱基以防止先天免疫感应并减少毒性,尽管这可能也会限制mRNA的佐剂效应(Andries等人,2015;Kariko等人,2005)。用于mRNA递送的脂质纳米颗粒(LNPs)可以作为内置佐剂,诱导促炎症细胞因子如IL-1β和IL-6的产生,从而促进抗原特异性CD4+滤泡辅助T细胞(Tfh)和B细胞反应(Alameh等人,2021;Tahtinen等人,2022)。然而,细胞因子如IL-6可能会抑制幼稚T细胞分化为效应细胞,而效应T细胞在抗肿瘤免疫反应中是至关重要的(Huseni等人,2023)。因此,选择合适的mRNA癌症疫苗佐剂对于确保其疗效和安全性至关重要。对于诱导肿瘤特异性杀伤性T细胞而言,营造一个以IL-12和TNF-α为主导的Th1型免疫微环境非常重要。然而,IL-12本身是一个毒性很大的细胞因子,直接将IL-12蛋白作为佐剂和RNA疫苗共注射或者用mRNA表达游离IL-12都会导致严重的治疗相关副作用;所以如何为mRNA疫苗的T细胞激活过程营造一个有利于杀伤性T细胞分化的免疫微环境,并且降低免疫过程对机体产生的副作用对于提升mRNA肿瘤疫苗的疗效至关重要。Insufficient numbers of cytotoxic T lymphocytes (CTLs) targeting immunogenic tumors are a major challenge for immunotherapy (Kalbasi and Ribas, 2020). Traditional vaccines, such as protein-based vaccines combined with alum, often fail to generate robust CTL responses against viral infections or tumors; while stronger adjuvants can enhance CTL responses, they are often accompanied by severe toxicity (Awate et al., 2013; Coffman et al., 2010). Antigen generation and adjuvant selection are crucial for vaccine success. Recently developed mRNA vaccine platforms offer a promising strategy for generating more potent antigens and effective adjuvants for the prevention of viral infections and the treatment of cancer (Barbier et al., 2022; Beck et al., 2021; Chaudhary et al., 2021). To increase antigen expression, most clinical mRNA vaccines use methylated bases to prevent innate immune induction and reduce toxicity, although this may also limit the adjuvant effect of mRNA (Andries et al., 2015; Kariko et al., 2005). Lipid nanoparticles (LNPs) for mRNA delivery can act as built-in adjuvants, inducing the production of proinflammatory cytokines such as IL-1β and IL-6, thereby promoting antigen-specific CD4+ follicular helper T cell (Tfh) and B cell responses (Alameh et al., 2021; Tahtinen et al., 2022). However, cytokines such as IL-6 may inhibit the differentiation of naive T cells into effector cells, which are crucial in anti-tumor immune responses (Huseni et al., 2023). Therefore, selecting the right adjuvant for mRNA cancer vaccines is crucial to ensure their efficacy and safety. For the induction of tumor-specific killer T cells, it is important to create a Th1 immune microenvironment dominated by IL-12 and TNF-α. However, IL-12 itself is a highly toxic cytokine. Directly injecting IL-12 protein as an adjuvant with RNA vaccine or expressing free IL-12 with mRNA will lead to serious treatment-related side effects. Therefore, how to create an immune microenvironment that is conducive to the differentiation of cytotoxic T cells for the T cell activation process of mRNA vaccine and reduce the side effects of the immune process on the body is crucial to improving the efficacy of mRNA tumor vaccines.

最新研究发现肌肉注射的mRNA-LNP主要引流到引流淋巴结和脾脏,被巨噬细胞和树突状细胞捕获并表达蛋白(Kimberly,J.H.等Mol Ther Nucleic Acids.2023年11月24日;35(1):102083.)。引流淋巴结和脾脏是肿瘤疫苗触发适应性免疫反应的重要器官,因此将IL-12特异递送到引流淋巴结和脾脏,避免IL-12释放到外周组织,可能在提高肿瘤mRNA疫苗免疫效果的同时降低IL-12引起的外周毒副作用。Recent studies have found that intramuscularly injected mRNA-LNPs are primarily distributed to the draining lymph nodes and spleen, where they are captured by macrophages and dendritic cells and express proteins (Kimberly, J.H. et al. Mol Ther Nucleic Acids. 2023 Nov 24;35(1):102083.). Draining lymph nodes and spleen are important organs for tumor vaccines to trigger adaptive immune responses. Therefore, specifically delivering IL-12 to the draining lymph nodes and spleen to prevent IL-12 from being released into peripheral tissues may improve the immune effect of tumor mRNA vaccines while reducing the peripheral toxic side effects caused by IL-12.

发明内容Summary of the Invention

有鉴于此,本发明的目的在于提供编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗,利用RNA技术将IL-12和肿瘤抗原同时表达在外周淋巴组织中的抗原递呈细胞上,膜型IL-12作为佐剂提高常规mRNA肿瘤疫苗的治疗效果;与直接表达游离的IL-12蛋白相比,细胞膜锚定形式的IL-12蛋白可以将IL-12限定在外周淋巴组织的抗原递呈细胞表面,在不影响其佐剂效应的同时避免IL-12释放到外周组织引起外周毒性。In view of this, the purpose of the present invention is to provide an mRNA tumor vaccine encoding a membrane-bound IL-12 cytokine adjuvant, using RNA technology to simultaneously express IL-12 and tumor antigens on antigen-presenting cells in peripheral lymphoid tissues, and use membrane-bound IL-12 as an adjuvant to improve the therapeutic effect of conventional mRNA tumor vaccines; compared with directly expressing free IL-12 protein, the cell membrane-anchored form of IL-12 protein can limit IL-12 to the surface of antigen-presenting cells in peripheral lymphoid tissues, while not affecting its adjuvant effect and avoiding the release of IL-12 into peripheral tissues to cause peripheral toxicity.

为了实现上述发明目的,本发明提供了以下技术方案:In order to achieve the above-mentioned object of the invention, the present invention provides the following technical solutions:

在一个方面,本发明提供了一种编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗,其中,所述mRNA包含编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)三部分。In one aspect, the present invention provides an mRNA tumor vaccine encoding a membrane-type IL-12 cytokine adjuvant, wherein the mRNA comprises three parts: a sequence encoding IL-12 (IL12), a sequence encoding a membrane domain (MD), and a sequence encoding a tumor antigen (Ag).

根据本发明的具体实施方式,所述编码IL-12的序列(IL12)和所述编码膜结构域的序列(MD)位于一条mRNA上,所述编码肿瘤抗原的序列(Ag)位于另一条mRNA上;或者所述编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)位于同一条mRNA上,且所述三部分的排列顺序为IL12-L1-MD-L2-Ag,或Ag-L2-IL12-L1-MD;其中L1为甘氨酸/丝氨酸(Glycine/Serine,Gly/Ser)多肽连接子,L2为IRES序列或Glycine/Serine或2A多肽连接子,L2优选2A多肽连接子。According to a specific embodiment of the present invention, the sequence encoding IL-12 (IL12) and the sequence encoding the membrane domain (MD) are located on one mRNA, and the sequence encoding the tumor antigen (Ag) is located on another mRNA; or the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD) and the sequence encoding the tumor antigen (Ag) are located on the same mRNA, and the arrangement order of the three parts is IL12-L1-MD-L2-Ag, or Ag-L2-IL12-L1-MD; wherein L1 is a glycine/serine (Glycine/Serine, Gly/Ser) polypeptide linker, L2 is an IRES sequence or a Glycine/Serine or a 2A polypeptide linker, and L2 is preferably a 2A polypeptide linker.

根据本发明的具体实施方式,所述肿瘤抗原包括突变来源的肿瘤新抗原、病毒来源的肿瘤特异性抗原或肿瘤高表达的相关抗原。According to a specific embodiment of the present invention, the tumor antigen includes a tumor neoantigen derived from a mutation, a tumor-specific antigen derived from a virus, or a tumor-related antigen that is highly expressed.

根据本发明的具体实施方式,所述突变来源的肿瘤新抗原包括KARS G12C、KRASG12D、EGFRvIII或BRAFV600E;所述病毒来源的肿瘤特异性抗原包括HPV E6/E7、内源性逆转录病毒抗原hERT、LMP1或LMP2;所述肿瘤高表达的相关抗原包括WT1、MAGE-A3、GP100、NY-ESO-I、HER2/Neu、Claudin18.2、Mesothelin或MUC。According to a specific embodiment of the present invention, the mutation-derived tumor neoantigens include KARS G12C, KRASG12D, EGFRvIII or BRAF V600E ; the viral-derived tumor-specific antigens include HPV E6/E7, endogenous retroviral antigen hERT, LMP1 or LMP2; the tumor-related antigens highly expressed include WT1, MAGE-A3, GP100, NY-ESO-I, HER2/Neu, Claudin18.2, Mesothelin or MUC.

根据本发明的具体实施方式,所述编码IL-12细胞因子的序列包含编码IL-12B多肽和IL-12A多肽的ORF,其中IL-12B和IL-12A通过多肽接头(linker)连接,所述linker包括Gly/Ser linker,所述Gly/Ser linker包含(GnS)m,其中n为1、2、3、4、5、6、7、8、9、10、15或20且m为1、2、3、4、5、6、7、8、9、10、15或20;所述linker优选为(G4S)3linker。According to a specific embodiment of the present invention, the sequence encoding the IL-12 cytokine comprises an ORF encoding an IL-12B polypeptide and an IL-12A polypeptide, wherein IL-12B and IL-12A are connected by a polypeptide linker, the linker comprises a Gly/Ser linker, and the Gly/Ser linker comprises (GnS)m, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 and m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20; the linker is preferably a (G4S) 3 linker.

根据本发明的具体实施方式,所述膜结构域可以包含跨膜结构域,优选包含跨膜结构域加胞内域,跨膜结构域包括CD80跨膜域、CD86跨膜域、CD4跨膜域、CD8跨膜域、VEGFR跨膜域、PDGF-RB跨膜域,优选为CD80跨膜域;胞内域包括CD80胞内域、CD86胞内域、CD4胞内域、CD8胞内域、VEGFR胞内域、PDGF-RB胞内域、PDGF-RB胞内域截短体,胞内域和跨膜域可以来自同一个基因组合或不同基因组合;优选为CD80跨膜域加CD80胞内域的组合。According to a specific embodiment of the present invention, the membrane domain may include a transmembrane domain, preferably a transmembrane domain plus an intracellular domain, the transmembrane domain includes the CD80 transmembrane domain, the CD86 transmembrane domain, the CD4 transmembrane domain, the CD8 transmembrane domain, the VEGFR transmembrane domain, and the PDGF-RB transmembrane domain, preferably the CD80 transmembrane domain; the intracellular domain includes the CD80 intracellular domain, the CD86 intracellular domain, the CD4 intracellular domain, the CD8 intracellular domain, the VEGFR intracellular domain, the PDGF-RB intracellular domain, and a truncation of the PDGF-RB intracellular domain, and the intracellular domain and the transmembrane domain may come from the same gene combination or different gene combinations; preferably, a combination of the CD80 transmembrane domain plus the CD80 intracellular domain.

根据本发明的具体实施方式,所述mRNA肿瘤疫苗还包含负载mRNA的核酸载体,所述核酸载体包括脂质体、LPP或LPX,优选为LNP纳米脂质体。According to a specific embodiment of the present invention, the mRNA tumor vaccine further comprises a nucleic acid vector loaded with mRNA, and the nucleic acid vector comprises liposomes, LPP or LPX, preferably LNP nanoliposomes.

在另一方面,本发明还提供了一种药物组合物,其包含本发明的mRNA肿瘤疫苗和第二抗癌剂,所述第二抗癌剂包括免疫检查点抑制剂和/或双特异性T细胞衔接器(BiTE)。优选地,所述药物组合物还包含药学上可接受的辅料。In another aspect, the present invention further provides a pharmaceutical composition comprising the mRNA tumor vaccine of the present invention and a second anticancer agent, wherein the second anticancer agent comprises an immune checkpoint inhibitor and/or a bispecific T cell engager (BiTE). Preferably, the pharmaceutical composition further comprises a pharmaceutically acceptable excipient.

在又一方面,本发明还提供了本发明的mRNA肿瘤疫苗或药物组合物在制备用于治疗或预防肿瘤的药物中的应用。在又一方面,本发明还提供了本发明的mRNA肿瘤疫苗或药物组合物在用于治疗或预防肿瘤中的应用。在又一方面,本发明还提供了一种治疗或预防受试者的肿瘤的方法,所述方法包括向所述受试者施用本发明的mRNA肿瘤疫苗或药物组合物。In yet another aspect, the present invention further provides the use of the mRNA tumor vaccine or pharmaceutical composition of the present invention in the preparation of a medicament for treating or preventing tumors. In yet another aspect, the present invention further provides the use of the mRNA tumor vaccine or pharmaceutical composition of the present invention in treating or preventing tumors. In yet another aspect, the present invention further provides a method for treating or preventing tumors in a subject, the method comprising administering the mRNA tumor vaccine or pharmaceutical composition of the present invention to the subject.

根据本发明的具体实施方式,其中,所述肿瘤选自B细胞淋巴瘤、支气管癌、前列腺癌、膀胱癌、脑或中枢神经系统癌症、外周神经系统癌症、食道癌、头颈癌、宫颈癌、子宫或子宫内膜癌、口腔癌、喉癌、唾液腺癌、胸腺癌、肾上腺癌、骨肉瘤、软骨肉瘤、脂肪癌、睾丸癌、恶性纤维组织细胞瘤、结肠直肠癌、黑色素瘤、胃癌、胰腺癌、肺癌、肝癌、肾癌、胆管癌、小肠癌或阑尾癌、鳞状细胞癌、乳腺癌和卵巢癌。According to a specific embodiment of the present invention, the tumor is selected from B-cell lymphoma, bronchial cancer, prostate cancer, bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, head and neck cancer, cervical cancer, uterine or endometrial cancer, oral cancer, laryngeal cancer, salivary gland cancer, thymic cancer, adrenal cancer, osteosarcoma, chondrosarcoma, adipose cancer, testicular cancer, malignant fibrous histiocytoma, colorectal cancer, melanoma, gastric cancer, pancreatic cancer, lung cancer, liver cancer, kidney cancer, bile duct cancer, small intestine cancer or appendix cancer, squamous cell carcinoma, breast cancer and ovarian cancer.

有益效果Beneficial effects

在本发明中,我们通过在mRNA中加入一段细胞膜锚定的IL-12序列,成功地提升了现有mRNA肿瘤疫苗的治疗效果。与直接表达游离的IL-12蛋白相比,细胞膜锚定形式的IL-12蛋白可以将IL-12限定在外周淋巴组织的抗原递呈细胞表面,在不影响IL-12佐剂效果的同时大大降低了IL-12蛋白释放到外周血中,显著降低了IL-12蛋白产生的系统性毒性。在多种类型的肿瘤疫苗中,膜锚定的IL-12序列的加入都可以显著的提升mRNA疫苗的治疗效果。因此,我们认为,以膜锚定IL-12为基础的mRNA肿瘤疫苗有潜力成为新一代的肿瘤治疗性疫苗,并在多种临床肿瘤中实现更好的治疗效果。In the present invention, we have successfully improved the therapeutic effect of existing mRNA tumor vaccines by adding a cell membrane-anchored IL-12 sequence to mRNA. Compared with directly expressing free IL-12 protein, the cell membrane-anchored form of IL-12 protein can limit IL-12 to the surface of antigen-presenting cells in peripheral lymphoid tissue, greatly reducing the release of IL-12 protein into the peripheral blood without affecting the adjuvant effect of IL-12, and significantly reducing the systemic toxicity produced by IL-12 protein. In various types of tumor vaccines, the addition of membrane-anchored IL-12 sequences can significantly improve the therapeutic effect of mRNA vaccines. Therefore, we believe that mRNA tumor vaccines based on membrane-anchored IL-12 have the potential to become a new generation of tumor therapeutic vaccines and achieve better therapeutic effects in a variety of clinical tumors.

附图说明BRIEF DESCRIPTION OF THE DRAWINGS

为了更清楚地说明本申请实施例中的技术方案,下面将对实施例中所需要使用的附图作简单地介绍。In order to more clearly illustrate the technical solutions in the embodiments of the present application, the following briefly introduces the drawings required for use in the embodiments.

图1显示IL-12可以作为佐剂促进mRNA疫苗的抗肿瘤效果。(A-E)C57BL/6J小鼠(n=4-6)皮下接种4×105B16F10-OVA细胞,在肿瘤接种后第8天和第12天通过肌肉注射接受5μg编码OVA的mRNA。在接种疫苗后的6小时,将2.5μg IL2-Fc(C)、5μgIL15-Fc(D)或0.25μg IL12-Fc蛋白(E)与海藻酸钠混合后肌肉注射到同一部位。(B)在第一次接种疫苗后的第9天,采集外周血,通过流式细胞术分析计算OVA特异性T细胞的数量。(C-E)每种治疗的肿瘤生长曲线如图所示。数据以均值±标准误(SEM)表示,来自两次独立实验。P值通过双向方差分析结合Bonfferoni多重比较检验确定(C-E)。**P<0.01,***P<0.001,****P<0.0001。Figure 1 shows that IL-12 can act as an adjuvant to enhance the antitumor efficacy of mRNA vaccines. (A-E) C57BL/6J mice (n = 4-6) were subcutaneously inoculated with 4× 10⁵ B16F10-OVA cells and received 5 μg of OVA-encoding mRNA via intramuscular injection on days 8 and 12 after tumor inoculation. Six hours after vaccination, 2.5 μg of IL2-Fc (C), 5 μg of IL15-Fc (D), or 0.25 μg of IL12-Fc protein (E) mixed with sodium alginate was injected intramuscularly into the same site. (B) Peripheral blood was collected on day 9 after the first vaccination, and the number of OVA-specific T cells was enumerated by flow cytometry. (C-E) Tumor growth curves for each treatment are shown. Data are presented as mean ± standard error of variance (SEM) and are from two independent experiments. P values were determined by two-way analysis of variance with Bonfferoni's multiple comparison test (C-E). **P<0.01, ***P<0.001, ****P<0.0001.

图2显示构建含游离或膜锚定IL12佐剂的RNA肿瘤疫苗。(A)游离形式sIL12-OVA和膜锚定形式mtIL12-OVAmRNA疫苗的结构示意图。(B)DC2.4细胞转染了CTRL mRNA(上图)、mtEGFP-OVARNA(中图)或mtIL12-OVAmRNA(下图)。转染后24小时,收集细胞并用IL12p40和抗OVA肽-MHC复合物抗体染色,通过流式细胞术分析膜IL12或OVA-pMHC复合物的表达。(C)DC2.4细胞转染了mtIL12-OVAmRNA或sIL12-OVAmRNA,转染后24小时,收集细胞培养液以测量分泌的游离IL12浓度。Figure 2 shows the construction of RNA tumor vaccines adjuvanted with free or membrane-anchored IL12. (A) Schematic diagram of the structures of the free sIL12-OVA and membrane-anchored mtIL12-OVA mRNA vaccines. (B) DC2.4 cells were transfected with CTRL mRNA (top), mtEGFP-OVA RNA (center), or mtIL12-OVA mRNA (bottom). 24 hours after transfection, cells were harvested and stained with antibodies against IL12p40 and OVA peptide-MHC complexes, and the expression of membrane IL12 or OVA-pMHC complexes was analyzed by flow cytometry. (C) DC2.4 cells were transfected with mtIL12-OVA mRNA or sIL12-OVA mRNA. 24 hours after transfection, cell culture fluid was harvested to measure the concentration of secreted free IL12.

图3显示含膜锚定IL12佐剂的RNA肿瘤疫苗相较含游离性IL12佐剂的疫苗具有相似的抗肿瘤效果,但外周毒性更低。(A,B)C57BL/6J小鼠(n=6)接种了4×105B16F10-OVA细胞,在接种后第8天和第12天通过肌肉注射2.5μg编码mtEGFP-OVA、mtIL12-OVA或sIL12-OVA的mRNA-LNP疫苗。记录肿瘤生长曲线和小鼠生存曲线(A)和体重变化(B)。(C)C57BL/6J小鼠(n=4)通过肌肉注射2.5μg编码mtIL12-OVA或sIL12-OVA的mRNA,在注射后24小时和48小时收集血清,测量促炎细胞因子浓度。Figure 3 shows that RNA tumor vaccines adjuvanted with membrane-anchored IL12 exhibit similar antitumor efficacy but lower peripheral toxicity compared to vaccines adjuvanted with free IL12. (A, B) C57BL/6J mice (n=6) were inoculated with 4×10 5 B16F10-OVA cells and injected intramuscularly with 2.5 μg of mRNA-LNP vaccines encoding mtEGFP-OVA, mtIL12-OVA, or sIL12-OVA on days 8 and 12 post-inoculation. Tumor growth curves, mouse survival curves (A), and body weight changes (B) were recorded. (C) C57BL/6J mice (n=4) were injected intramuscularly with 2.5 μg of mRNA encoding mtIL12-OVA or sIL12-OVA. Serum was collected 24 and 48 hours after injection for measurement of proinflammatory cytokine concentrations.

图4显示mtIL12 mRNA疫苗特异性激活抗原特异性T细胞,但不影响旁观者T细胞或NK细胞。(A)C57/B6J小鼠(n=4)通过肌肉注射2.5μg编码mtIL12-OVA或sIL12-OVA的mRNA,在接种疫苗后24小时,解剖dLNs,通过流式细胞术检测表面表达IL12p40的细胞亚群。(B)C57BL/6J小鼠(n=4-5)过继转移5×105OT1 T细胞,并在1天后接种了5μg编码mtEGFP-OVA、mtIL12-OVA或sIL12-OVA的mRNA疫苗。在免疫后42小时,通过腹腔注射50μL BFA。在免疫后48小时解剖并消化引流淋巴结(dLN),通过流式细胞术检测IFN-γ+细胞。(C)C57BL/6J小鼠通过肌肉注射接受了5μg mtIL12mRNA或5μg sIL12 mRNA。36小时后,收集外周血,加入BFA溶液阻断6小时,然后流式细胞术分析IFNγ阳性的细胞亚群比例。Figure 4 shows that the mtIL12 mRNA vaccine specifically activates antigen-specific T cells but does not affect bystander T cells or NK cells. (A) C57/B6J mice (n = 4) were intramuscularly injected with 2.5 μg of mRNA encoding mtIL12-OVA or sIL12-OVA. 24 hours after vaccination, dLNs were dissected, and cell subsets expressing IL12p40 on their surface were analyzed by flow cytometry. (B) C57BL/6J mice (n = 4-5) were adoptively transferred with 5 × 10 5 OT1 T cells and vaccinated one day later with 5 μg of mRNA vaccine encoding mtEGFP-OVA, mtIL12-OVA, or sIL12-OVA. 42 hours after immunization, 50 μL of BFA were injected intraperitoneally. 48 hours after immunization, draining lymph nodes (dLNs) were dissected and digested, and IFN-γ+ cells were analyzed by flow cytometry. (C) C57BL/6J mice received 5 μg of mtIL12 mRNA or 5 μg of sIL12 mRNA by intramuscular injection. 36 hours later, peripheral blood was collected and blocked with BFA solution for 6 hours, and the proportion of IFNγ-positive cell subsets was analyzed by flow cytometry.

图5显示mtIL12-OVAmRNA疫苗相较单独的OVAmRNA或者mtIL12 mRNA具有更好的抗肿瘤效果。C57BL/6J小鼠(n=6)接种4×105B16F10-OVA细胞,携带肿瘤的小鼠在第8天和第12天通过肌肉注射2.5μg编码mtEGFP-OVA、mtIL12-OVA或mtIL12的mRNA,记录了肿瘤生长曲线。Figure 5 shows that the mtIL12-OVA mRNA vaccine has a superior anti-tumor effect compared to OVA mRNA or mtIL12 mRNA alone. C57BL/6J mice (n=6) were inoculated with 4×10 5 B16F10-OVA cells. Tumor-bearing mice were injected intramuscularly with 2.5 μg of mRNA encoding mtEGFP-OVA, mtIL12-OVA, or mtIL12 on days 8 and 12, and tumor growth curves were recorded.

图6显示含膜锚定IL12佐剂的RNA肿瘤疫苗抗肿瘤效果依赖于CD8+T细胞。C57BL/6J小鼠(n=6)接种了4×105B16F10-OVA细胞,在接种后第8天和第12天肌肉注射2.5μg编码mtIL12-OVA的mRNA治疗。在治疗开始前一天,给予抗NK1.1(200μg)、抗CD8(200μg)、抗CD4(200μg)和抗CSF1R(500μg)抗体,并在接下来的两周内每周两次继续给药。记录肿瘤生长曲线(A)和生存曲线(B)。(C)C57BL/6J小鼠在第0天和第5天通过肌肉注射接受了2.5μg mtEGFP-OVAmRNA或2.5μg mtIL12-OVAmRNA。在第一次接种疫苗后的第8天测量引流淋巴结中OVA特异性T细胞,在第一次接种疫苗后的第13天测量脾脏中OVA特异性T细胞。Figure 6 shows that the antitumor effect of an RNA tumor vaccine containing a membrane-anchored IL12 adjuvant is dependent on CD8 + T cells. C57BL/6J mice (n=6) were inoculated with 4× 105 B16F10-OVA cells and treated intramuscularly with 2.5 μg of mtIL12-OVA mRNA on days 8 and 12 after inoculation. Anti-NK1.1 (200 μg), anti-CD8 (200 μg), anti-CD4 (200 μg), and anti-CSF1R (500 μg) antibodies were administered one day before treatment and continued twice weekly for the next two weeks. Tumor growth curves (A) and survival curves (B) were recorded. (C) C57BL/6J mice received 2.5 μg of mtEGFP-OVA mRNA or 2.5 μg of mtIL12-OVA mRNA via intramuscular injection on days 0 and 5. OVA-specific T cells were measured in the draining lymph nodes on day 8 after the first vaccination and in the spleen on day 13 after the first vaccination.

图7显示基于mtIL12的疫苗在OVA抗原模型中产生了更强的效应性T细胞。(A)B6小鼠在第0天和第七天肌肉注射2.5ug mtEGFP-OVA或者mtIL12-OVARNA免疫,第14天取血进行流式分析。(B)流式分析显示IL12免疫促进OVA特异性T细胞从前体表型(Ly108+CD69-)到效应表型(Ly108-CD69-)分化。(C)外周血细胞与OT1 peptide(SIINFEKL)共孵育3小时后进行流式染色分析。结果发现IL12疫苗免疫产生的小鼠OVA特异性T细胞表达更高水平的效应分子(GZMB,IFN-gamma),高表达效应T细胞特异性转录因子T-bet,低表达免疫抑制性分子PD-1。(D)mtEGFP-OVA或者mtIL12-OVA免疫完的小鼠中分选同等数量的OVA特异性T细胞,并与不同浓度的OT1peptide(SIINFEKL)共孵育,4小时后测量培养上清中IFN-gamma的浓度。Figure 7 shows that the mtIL12-based vaccine produces stronger effector T cells in the OVA antigen model. (A) B6 mice were immunized with 2.5 μg mtEGFP-OVA or mtIL12-OVA RNA intramuscularly on days 0 and 7, and blood was collected on day 14 for flow cytometry analysis. (B) Flow cytometry analysis showed that IL12 immunization promoted the differentiation of OVA-specific T cells from a precursor phenotype (Ly108+CD69-) to an effector phenotype (Ly108-CD69-). (C) Peripheral blood cells were incubated with the OT1 peptide (SIINFEKL) for 3 hours and then analyzed by flow cytometry. The results showed that OVA-specific T cells from mice immunized with the IL12 vaccine expressed higher levels of effector molecules (GZMB, IFN-gamma), higher expression of the effector T cell-specific transcription factor T-bet, and lower expression of the immunosuppressive molecule PD-1. (D) Equal numbers of OVA-specific T cells were isolated from mice immunized with mtEGFP-OVA or mtIL12-OVA and incubated with different concentrations of OT1 peptide (SIINFEKL). IFN-gamma concentrations in the culture supernatant were measured 4 hours later.

图8.显示顺式递送IL12和抗原可以产生更多的抗原特异性T细胞。(A)B6小鼠在第0天和第7天静脉注射2ug mtEGFP-OVA,2ug mtEGFP-OVA+1ug mtIL12和1ug mtEGFP-OVA+1ug mtIL12-OVARNA-LNP,第14天取血进行流式分析。(B)流式分析显示顺式递送IL12和抗原疫苗治疗的小鼠(1ug mtEGFP-OVA+1ug mtIL12-OVA)产生了更高数目的外周血抗原特异性T细胞。Figure 8. Demonstrates that cis-delivery of IL12 and antigen can generate more antigen-specific T cells. (A) B6 mice were intravenously injected with 2 μg mtEGFP-OVA, 2 μg mtEGFP-OVA + 1 μg mtIL12, and 1 μg mtEGFP-OVA + 1 μg mtIL12-OVA RNA-LNP on days 0 and 7, and blood was collected on day 14 for flow cytometric analysis. (B) Flow cytometric analysis shows that mice treated with the cis-delivery of IL12 and antigen vaccine (1 μg mtEGFP-OVA + 1 μg mtIL12-OVA) generated higher numbers of antigen-specific T cells in peripheral blood.

图9显示膜结合型IL12佐剂mRNA疫苗克服免疫检查点阻断耐药性。(A)C57BL/6J小鼠(n=6)皮下接种4×105B16F10-OVA细胞,在肿瘤接种后第11天和第15天接受了2.5μg编码mtIL12-OVA的mRNA治疗。在第一次接种疫苗后的第6天,给予抗CTLA4(200μg)和抗PD1(200μg),并持续两周,每周两次。记录了肿瘤生长曲线和小鼠存活率。Figure 9 shows that membrane-bound IL12 adjuvanted mRNA vaccines overcome resistance to immune checkpoint blockade. (A) C57BL/6J mice (n=6) were subcutaneously inoculated with 4×105B16F10-OVA cells and received 2.5μg of mRNA encoding mtIL12-OVA on days 11 and 15 after tumor inoculation. Anti-CTLA4 (200μg) and anti-PD1 (200μg) were given on day 6 after the first vaccination and continued twice a week for two weeks. Tumor growth curves and mouse survival were recorded.

图10显示mtIL12-E7疫苗抑制HPV相关的TC-1肿瘤生长。C57BL/6J小鼠(n=6)接种了4×105TC-1细胞,携带肿瘤的小鼠在第14天肌肉注射1μg编码mtEGFP-E7、mtIL12-E7或mtIL12的mRNA治疗,并在第20天用2μg相同mRNA疫苗进行加强免疫。记录了肿瘤生长曲线和小鼠存活率(A),以及体重变化(B)。可以看到在晚期TC-1肿瘤携带小鼠中,对照E7疫苗或mtIL12单独治疗均未能控制肿瘤生长。然而,尽管TC-1肿瘤在两次mtIL12-E7疫苗接种后继续生长至约500mm3,但肿瘤最终消退,约80%的肿瘤成功根除(A)。重要的是,与未治疗组相比,接受mtIL12-E7 mRNA疫苗治疗的小鼠没有显著的体重下降(B)。Figure 10 shows that the mtIL12-E7 vaccine inhibits the growth of HPV-associated TC-1 tumors. C57BL/6J mice (n=6) were vaccinated with 4×105 TC-1 cells. Tumor-bearing mice were treated with an intramuscular injection of 1 μg of mRNA encoding mtEGFP-E7, mtIL12-E7, or mtIL12 on day 14 and boosted with 2 μg of the same mRNA vaccine on day 20. Tumor growth curves and mouse survival (A) and body weight changes (B) were recorded. It can be seen that neither the control E7 vaccine nor mtIL12 alone failed to control tumor growth in mice bearing advanced TC-1 tumors. However, although TC-1 tumors continued to grow to approximately 500 mm3 after two mtIL12-E7 vaccinations, the tumors ultimately regressed, with approximately 80% of tumors successfully eradicated (A). Importantly, mice treated with the mtIL12-E7 mRNA vaccine did not experience significant weight loss compared to the untreated group (B).

图11显示基于mtIL12的疫苗在E7抗原模型中产生了更强的效应性T细胞。(A).TC-1荷瘤小鼠在肿瘤种植第12天肌肉注射2.5ug mtEGFP-E7,第18天肌肉注射2.5ug mtEGFP-E7或者2.5ug mtIL12-E7免疫,第22天取肿瘤进行流式分析。(B).流式分析展示抗原特异性T细胞的圈门和部分表面标志表达。(C).流式分析显示IL12免疫促进E7特异性T细胞的肿瘤内富集,并且相对于mtGFP-E7免疫产生的T细胞,mtIL12-E7免疫产生的T细胞更多停留在效应状态(LY108-CD69-)而不是耗竭状态(LY108-CD69+),并且表达更高的效应分子GZMB,和更低的抑制性标志PD1。Figure 11 shows that the mtIL12-based vaccine generates enhanced effector T cells in the E7 antigen model. (A) TC-1 tumor-bearing mice were intramuscularly injected with 2.5 μg mtEGFP-E7 on day 12 after tumor implantation. On day 18, they were immunized with either 2.5 μg mtEGFP-E7 or 2.5 μg mtIL12-E7. Tumors were harvested and analyzed by flow cytometry on day 22. (B) Flow cytometry analysis shows the gates and expression of selected surface markers for antigen-specific T cells. (C) Flow cytometry analysis shows that IL12 immunization promotes the intratumoral enrichment of E7-specific T cells. Compared to T cells immunized with mtGFP-E7, T cells immunized with mtIL12-E7 are more likely to remain in the effector state (LY108-CD69-) rather than the exhausted state (LY108-CD69+). They also express higher levels of the effector molecule GZMB and lower levels of the inhibitory marker PD1.

图12显示mtIL12佐剂可以应用到内源性逆转录病毒来源的肿瘤新抗原的mRNA疫苗设计。P15E蛋白,源自gp70逆转录病毒基因表达,在许多癌症中被检测到,包括MC38结肠腺癌细胞系(Ye等人,2020)。第0天将4X105的MC38肿瘤细胞皮下注射到C57BL/6J小鼠中。肿瘤形成后,在第12和第16天小鼠肌肉注射2.5μg的mtIL12-p15E或者对照mtEGFP-p15E mRNA-LNP,每周监测两次肿瘤生长曲线。结果如图7所示对照P15E mRNA疫苗治疗几乎不能抑制肿瘤生长,而mtIL12-P15E治疗诱导了显著的肿瘤消退和完全根除(图7)。Figure 12 shows that mtIL12 adjuvant can be applied to the design of mRNA vaccines for endogenous retroviral-derived tumor neoantigens. P15E protein, derived from gp70 retroviral gene expression, has been detected in many cancers, including the MC38 colon adenocarcinoma cell line (Ye et al., 2020). 4×10 5 MC38 tumor cells were subcutaneously injected into C57BL/6J mice on day 0. After tumor formation, mice were injected intramuscularly with 2.5 μg of mtIL12-p15E or control mtEGFP-p15E mRNA-LNP on days 12 and 16, and tumor growth curves were monitored twice a week. As shown in Figure 7, control P15E mRNA vaccine treatment barely inhibited tumor growth, while mtIL12-P15E treatment induced significant tumor regression and complete eradication (Figure 7).

图13显示基于mtIL12的疫苗在p15E抗原模型中产生了更强的效应性T细胞。(A).MC38荷瘤小鼠在肿瘤接种后第12天肌肉注射2.5ug mtEGFP-p15E mRNA,第18天进行2.5ug mtEGFP-p15E或者2.5ug mtIL12-p15E mRNA疫苗再次免疫,第21天取外周血进行流式分析。(B).流式分析显示mtIL12-p15E免疫产生了更多的p15E抗原特异性T细胞,并且相对于mtEGFP-p15E免疫产生的T细胞,mtIL12-p15E免疫产生的T细胞表达更高的效应分子GZMB和IFN-gamma,和更低的抑制性标志PD1。Figure 13 shows that the mtIL12-based vaccine generates enhanced effector T cells in the p15E antigen model. (A) MC38 tumor-bearing mice were intramuscularly injected with 2.5 μg mtEGFP-p15E mRNA on day 12 after tumor inoculation. They were then immunized again with either 2.5 μg mtEGFP-p15E or 2.5 μg mtIL12-p15E mRNA on day 18. Peripheral blood was collected for flow cytometry analysis on day 21. (B) Flow cytometry analysis showed that mtIL12-p15E immunization generated more p15E-specific T cells. Furthermore, compared to T cells immunized with mtEGFP-p15E, these T cells expressed higher expression of the effector molecules GZMB and IFN-gamma, and lower expression of the inhibitory marker PD1.

具体实施方式DETAILED DESCRIPTION

本发明提供了一种编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗,其中,所述mRNA包含编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)三部分。The present invention provides an mRNA tumor vaccine encoding a membrane-type IL-12 cytokine adjuvant, wherein the mRNA comprises three parts: a sequence encoding IL-12 (IL12), a sequence encoding a membrane domain (MD), and a sequence encoding a tumor antigen (Ag).

根据本发明的具体实施方式,所述编码IL-12的序列(IL12)和所述编码膜结构域的序列(MD)位于一条mRNA上,所述编码肿瘤抗原的序列(Ag)位于另一条mRNA上;或者所述编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)位于同一条mRNA上,所述三部分的排列顺序为IL12-L1-MD-L2-Ag,或Ag-L2-IL12-L1-MD;其中L1为Glycine/Serine多肽连接子,L2为IRES序列或Glycine/Serine或2A多肽连接子。According to a specific embodiment of the present invention, the sequence encoding IL-12 (IL12) and the sequence encoding the membrane domain (MD) are located on one mRNA, and the sequence encoding the tumor antigen (Ag) is located on another mRNA; or the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD) and the sequence encoding the tumor antigen (Ag) are located on the same mRNA, and the arrangement order of the three parts is IL12-L1-MD-L2-Ag, or Ag-L2-IL12-L1-MD; wherein L1 is a Glycine/Serine polypeptide linker, and L2 is an IRES sequence or a Glycine/Serine or 2A polypeptide linker.

优选的是,所述编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)位于同一条mRNA上。这是因为同一mRNA可保证IL12、MD和Ag在同一细胞内同时表达,避免因两条mRNA转染效率差异导致部分细胞仅表达IL-12-MD或Ag,能更精准地实现三者的同步表达,保证它们在量和时间上的协调性,有利于发挥它们之间的协同作用。另外,能够优化生产流程和降低生产成本,单条mRNA的合成、纯化及质控成本显著低于两条独立mRNA的并行生产,在基因治疗等实际应用中,只需要递送一条mRNA进入细胞,相比于递送两条mRNA,操作更简单,递送效率更高,也降低了因递送多条mRNA带来的载体容量限制、细胞摄取差异等问题,提高了治疗的可行性和可操作性。单一分子结构也更容易保证稳定性(如避免两条mRNA的降解速率差异),且批次间一致性更高。Preferably, the sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD), and the sequence encoding the tumor antigen (Ag) are located on the same mRNA. This is because the same mRNA can ensure that IL12, MD, and Ag are expressed simultaneously in the same cell, avoiding the situation where some cells only express IL-12-MD or Ag due to differences in the transfection efficiency of the two mRNAs. It can more accurately achieve the synchronous expression of the three, ensure their coordination in quantity and time, and facilitate the synergistic effect between them. In addition, it is possible to optimize the production process and reduce production costs. The synthesis, purification, and quality control costs of a single mRNA are significantly lower than the parallel production of two independent mRNAs. In practical applications such as gene therapy, only one mRNA needs to be delivered into the cell. Compared with delivering two mRNAs, the operation is simpler and the delivery efficiency is higher. It also reduces the problems of carrier capacity limitations and cell uptake differences caused by delivering multiple mRNAs, thereby improving the feasibility and operability of the treatment. A single molecular structure is also easier to ensure stability (such as avoiding differences in the degradation rates of the two mRNAs) and has higher consistency between batches.

本发明中描述的肿瘤抗原可以包括突变来源的肿瘤新抗原如KARS G12C、KRAS G12D、EGFRvIII和BRAFV600E;病毒来源的肿瘤特异性抗原如HPV E6/E7、内源性逆转录病毒抗原(hERT)、LMP1和LMP2;肿瘤高表达的相关抗原如WT1、MAGE-A3、GP100、NY-ESO-I、HER2/Neu、Claudin18.2、Mesothelin和MUC等。可以根据肿瘤特异性抗原而改变抗原种类。卵清蛋白(OVA)是一种蛋白抗原,广泛用于在癌症免疫治疗中引发细胞和体液免疫反应,在本领域中常用作肿瘤模式抗原。在本发明的实施例中,以OVA作为肿瘤特异性抗原的实例。The tumor antigens described in the present invention may include tumor neoantigens of mutation origin such as KARS G12C, KRAS G12D, EGFRvIII and BRAF V600E ; tumor-specific antigens of viral origin such as HPV E6/E7, endogenous retrovirus antigens (hERT), LMP1 and LMP2; tumor-related antigens highly expressed such as WT1, MAGE-A3, GP100, NY-ESO-I, HER2/Neu, Claudin18.2, Mesothelin and MUC, etc. The type of antigen can be changed according to the tumor-specific antigen. Ovalbumin (OVA) is a protein antigen that is widely used to induce cellular and humoral immune responses in cancer immunotherapy and is commonly used as a tumor model antigen in the art. In the embodiments of the present invention, OVA is used as an example of a tumor-specific antigen.

白细胞介素-12(IL-12)是由两个独立基因IL-12A(p35)和IL-12B(p40)编码的一种多效能细胞因子,它们以一种活性异源二聚体(p70)或p40的同型二聚体(p80)形式存在。在本发明中,IL-12B和IL-12A通过多肽接头(linker)连接,所述linker包括Gly/Ser linker,所述Gly/Ser linker包含(GnS)m,其中n为1、2、3、4、5、6、7、8、9、10、15或20且m为1、2、3、4、5、6、7、8、9、10、15或20;在本发明的实施例中,IL-12细胞因子的两个亚基通过(G4S)3linker连接。Interleukin-12 (IL-12) is a multipotent cytokine encoded by two independent genes, IL-12A (p35) and IL-12B (p40), which exist as an active heterodimer (p70) or a homodimer of p40 (p80). In the present invention, IL-12B and IL-12A are linked by a polypeptide linker comprising a Gly/Ser linker comprising (GnS)m, wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20 and m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, or 20. In embodiments of the present invention, the two subunits of the IL-12 cytokine are linked by a (G4S) 3 linker.

本发明的膜结构域可以为跨膜结构域加胞内域,跨膜结构域包括CD80跨膜域、CD86跨膜域、CD4跨膜域、CD8跨膜域、VEGFR跨膜域、PDGF-RB跨膜域,优选为CD80跨膜域;胞内域包括CD80胞内域、CD86胞内域、CD4胞内域、CD8胞内域、VEGFR胞内域、PDGF-RB胞内域、PDGF-RB胞内域截短体,胞内域和跨膜域可以来自同一个基因组合或不同基因组合。例如,可以使用下述跨膜域序列和胞内域序列相加,但是本发明不限于此:The membrane domain of the present invention can be a transmembrane domain plus an intracellular domain, and the transmembrane domain includes the CD80 transmembrane domain, the CD86 transmembrane domain, the CD4 transmembrane domain, the CD8 transmembrane domain, the VEGFR transmembrane domain, and the PDGF-RB transmembrane domain, preferably the CD80 transmembrane domain; the intracellular domain includes the CD80 intracellular domain, the CD86 intracellular domain, the CD4 intracellular domain, the CD8 intracellular domain, the VEGFR intracellular domain, the PDGF-RB intracellular domain, and a truncated PDGF-RB intracellular domain. The intracellular domain and the transmembrane domain can be derived from the same gene combination or different gene combinations. For example, the following transmembrane domain sequence and intracellular domain sequence can be added together, but the present invention is not limited thereto:

1.人CD8A跨膜域,IYIWAPLAGTCGVLLLSLVITLYCY(SEQ ID NO:85)1. Human CD8A transmembrane domain, IYIWAPLAGTCGVLLLSLVITLYCY (SEQ ID NO: 85)

2.人PDGF-RB跨膜域,VVISAILALVVLTIISLIILI(SEQ ID NO:3)2. Human PDGF-RB transmembrane domain, VVISAILALVVLTIISLIILI (SEQ ID NO: 3)

3.人CD80跨膜域,LLPSWAITLISVNGIFVICCL(SEQ ID NO:5)3. Human CD80 transmembrane domain, LLPSWAITLISVNGIFVICCL (SEQ ID NO: 5)

4.人CD86跨膜域,WITAVLPTVIICVMVFCLILW(SEQ ID NO:7)4. Human CD86 transmembrane domain, WITAVLPTVIICVMVFCLILW (SEQ ID NO: 7)

5.鼠CD80跨膜域,TLVLFGAGAVITVVVIVVII(SEQ ID NO:9)5. Mouse CD80 transmembrane domain, TLVLFGAGAVITVVVIVVII (SEQ ID NO: 9)

6.人CD8A胞内域(ICD)氨基酸序列,LYCNHRNRRRVCKCPRPVVKSGDKPSLSARYV(SEQ ID NO:11)6. Human CD8A intracellular domain (ICD) amino acid sequence, LYCNHRNRRRVCKCPRPVVKSGDKPSLSARYV (SEQ ID NO: 11)

7.人PDGF-RB胞内域(ICD)氨基酸序列,
7. Human PDGF-RB intracellular domain (ICD) amino acid sequence,

8.截短的人PDGF-RB胞内域(ICD)氨基酸序列
8. Truncated human PDGF-RB intracellular domain (ICD) amino acid sequence

9.人CD80胞内域(ICD)氨基酸序列
9. Human CD80 intracellular domain (ICD) amino acid sequence

10.人CD86胞内域(ICD)氨基酸序列
10. Human CD86 intracellular domain (ICD) amino acid sequence

11.鼠CD80胞内域(ICD)氨基酸序列
11. Mouse CD80 intracellular domain (ICD) amino acid sequence

下面结合具体实施例,对本发明作进一步详细的阐述,下述实施例不用于限制本发明,仅用于说明本发明。以下实施例中所使用的实验方法如无特殊说明,实施例中未注明具体条件的实验方法,通常按照常规条件,下述实施例中所使用的材料、试剂等,如无特殊说明,均可从商业途径得到。The present invention will be further described in detail below with reference to specific examples. The following examples are not intended to limit the present invention but are merely intended to illustrate the present invention. The experimental methods used in the following examples are generally based on conventional conditions unless otherwise specified. The materials and reagents used in the following examples are all commercially available unless otherwise specified.

实施例Example

我们想知道T细胞刺激细胞因子是否能够快速扩增抗原启动的CTLs,并有效增强抗肿瘤免疫反应。尽管IL-2对T细胞激活至关重要,IL-15能够高效扩增T细胞数量(Guo等人,2021;Zou等人,2024),但在mRNA疫苗的背景下,我们观察到这些细胞因子对增强抗肿瘤活性的作用有限。IL-12能够诱导Th1型免疫微环境,促进具有肿瘤杀伤能力的CD8+T细胞分化(Hewitt等人,2020;Tucker等人,2020;Tugues等人,2015;Zou等人,2024)。值得注意的是,我们观察到IL-12是增强mRNA癌症疫苗抗肿瘤效果的最有效细胞因子佐剂。为了优化IL-12的刺激效应并减少外周毒性,我们设计了一种表达肿瘤抗原和膜结合型IL-12佐剂的mRNA疫苗。我们的研究结果揭示了IL-12诱导并扩增了一种未被充分研究的预效应T细胞亚群。我们进一步探讨了mtIL-12如何提供强大的抗肿瘤反应,同时将毒性降至最低。We sought to determine whether T cell-stimulating cytokines could rapidly expand antigen-primed CTLs and effectively enhance antitumor immune responses. Although IL-2 is essential for T cell activation and IL-15 is highly effective in expanding T cell numbers (Guo et al., 2021; Zou et al., 2024), we observed limited efficacy of these cytokines in enhancing antitumor activity in the context of mRNA vaccines. IL-12 can induce a Th1-type immune microenvironment and promote the differentiation of CD8+ T cells with tumor-killing potential (Hewitt et al., 2020; Tucker et al., 2020; Tugues et al., 2015; Zou et al., 2024). Notably, we observed that IL-12 was the most potent cytokine adjuvant for enhancing the antitumor effects of mRNA cancer vaccines. To optimize the stimulatory effects of IL-12 and minimize peripheral toxicity, we designed an mRNA vaccine expressing a tumor antigen and a membrane-bound IL-12 adjuvant. Our results revealed that IL-12 induces and expands an understudied subset of pre-effector T cells. We further explored how mtIL-12 could provide a robust antitumor response while minimizing toxicity.

材料和方法Materials and methods

小鼠和细胞系:Mice and cell lines:

C57BL/6J,BALB/c,C57BL/6J-Tg(TcraTcrb)1100Mjb/J(OT1 TCR transgenic)小鼠,6-8周龄,购自维通利华或者Jackson Laboratory。小鼠饲养在SPF环境,所有动物实验符合清华大学实验动物管理实施细则。C57BL/6J, BALB/c, and C57BL/6J-Tg(TcraTcrb)1100Mjb/J (OT1 TCR transgenic) mice, 6–8 weeks old, were purchased from Vital River or Jackson Laboratory. Mice were housed in a SPF environment, and all animal experiments were conducted in accordance with the Regulations for Laboratory Animal Care of Tsinghua University.

MC38、B16F10、TC-1和DC2.4细胞系购自美国典型培养物保藏中心(ATCC)。Freestyle 293-F(R79007)购自Invitrogen。B16F10-OVA细胞系是通过慢病毒表达OVA后从单细胞克隆中筛选得到的。所有细胞系均常规检测支原体污染。MC38、B16F10和TC-1细胞在含有10%热灭活胎牛血清、100U/mL青霉素和100U/mL链霉素的Dulbecco改良Eagle培养基中培养,条件为37℃、5% CO2。DC2.4细胞在含有10%热灭活胎牛血清、2mmol/L L-谷氨酰胺、0.1mmol/LMEM非必需氨基酸、100U/mL青霉素和100U/mL链霉素的RPMI 1640培养基中培养,条件为37℃、5% CO2。293-F细胞在SMM 293-TI培养基(M293TI;Sino Biological)中培养。MC38, B16F10, TC-1, and DC2.4 cell lines were purchased from the American Type Culture Collection (ATCC). Freestyle 293-F (R79007) was purchased from Invitrogen. The B16F10-OVA cell line was derived from a single-cell clone after lentiviral expression of OVA. All cell lines were routinely tested for mycoplasma contamination. MC38, B16F10, and TC-1 cells were cultured in Dulbecco's modified Eagle's medium (DMEA) supplemented with 10% heat-inactivated fetal bovine serum, 100 U/mL penicillin, and 100 U/mL streptomycin at 37°C and 5% CO₂ . DC2.4 cells were cultured in RPMI 1640 medium containing 10% heat-inactivated fetal bovine serum, 2 mmol/L L-glutamine, 0.1 mmol/L MEM non-essential amino acids, 100 U/mL penicillin, and 100 U/mL streptomycin at 37°C in 5% CO2. 293-F cells were cultured in SMM 293-TI medium (M293TI; Sino Biological).

试剂:Reagents:

抗CD4(克隆GK1.5)、抗CD8(克隆53-5.8)、抗mCSF1R(克隆AFS98)、抗NK1.1(克隆PK136)、抗PD-L1抗体(10F.9G2)、抗mCTLA4(克隆4F10)和抗mIL12p75(克隆JR2-9A5)购自Bio X Cell。IL2-Fc、IL15-Fc和IL12-Fc为本实验室自行生产。具体方法为:将细胞因子编码序列与人类IgG1 Fc片段通过GGGGS连接子融合,并克隆到pEE12.4载体中。将编码蛋白的质粒转染到293F细胞中,上清液通过Protein-A亲和层析(GE Healthcare)纯化,按照既定协议进行操作。Anti-CD4 (clone GK1.5), anti-CD8 (clone 53-5.8), anti-mCSF1R (clone AFS98), anti-NK1.1 (clone PK136), anti-PD-L1 antibodies (10F.9G2), anti-mCTLA4 (clone 4F10), and anti-mIL12p75 (clone JR2-9A5) were purchased from Bio X Cell. IL2-Fc, IL15-Fc, and IL12-Fc were produced in-house. Cytokine coding sequences were fused to the human IgG1 Fc fragment via a GGGGS linker and cloned into the pEE12.4 vector. Protein-encoding plasmids were transfected into 293F cells, and the supernatant was purified by Protein-A affinity chromatography (GE Healthcare) according to established protocols.

小鼠肿瘤模型建立和治疗:Mouse tumor model establishment and treatment:

MC38细胞(4×105)、TC-1细胞(3×105)和B16F10-OVA细胞(2-5×105)细胞接种到小鼠的右侧背部皮下,肿瘤建立后,每周三次测量肿瘤的长(a),宽(b),计算肿瘤体积=a*b*b/2。小鼠按肿瘤大小随机分组。小鼠在特定的时间点注射1-5μg不同编码区的mRNA-纳米脂质体复合物,或者脂质体空载对照。删除不同细胞亚群实验,从mRNA疫苗接种前一天开始,每3天腹腔注射200μg抗CD8抗体、200μg抗CD4抗体、200μg抗NK1.1抗体、或500μg抗CSFIR抗体。在mRNA疫苗接种后6小时,将2μg IL2-Fc、3.5μg IL15-Fc和0.25μg IL12-Fc与海藻酸钠混合后肌肉注射。从首次mRNA疫苗接种后第6天开始,每3天腹腔注射200μg抗PD-L1抗体和200μg抗CTLA4抗体。MC38 cells (4× 10⁵ ), TC-1 cells (3× 10⁵ ), and B16F10-OVA cells (2-5× 10⁵ ) were inoculated subcutaneously on the right dorsum of mice. After tumor establishment, tumor length (a) and width (b) were measured three times weekly, and tumor volume was calculated as a*b*b/2. Mice were randomly grouped according to tumor size. At specific time points, mice were injected with 1-5 μg of mRNA-nanoliposome complexes encoding different coding regions, or with empty liposomes as a control. To exclude experiments targeting different cell subsets, mice were intraperitoneally injected with 200 μg of anti-CD8 antibody, 200 μg of anti-CD4 antibody, 200 μg of anti-NK1.1 antibody, or 500 μg of anti-CSFIR antibody every three days, starting one day before mRNA vaccination. Six hours after mRNA vaccination, 2 μg of IL2-Fc, 3.5 μg of IL15-Fc, and 0.25 μg of IL12-Fc were mixed with sodium alginate and injected intramuscularly. Starting on day 6 after the first mRNA vaccination, 200 μg of anti-PD-L1 antibody and 200 μg of anti-CTLA4 antibody were injected intraperitoneally every three days.

当肿瘤的长、宽或高超过2cm,或者肿瘤大小超过1500mm3,或者荷瘤小鼠体重下降超过20%时,处死小鼠。Mice were sacrificed when the length, width or height of the tumor exceeded 2 cm, or the tumor size exceeded 1500 mm 3 , or the body weight of tumor-bearing mice decreased by more than 20%.

流式细胞术分析Flow cytometric analysis

细胞消化离心后制成细胞悬液,先加入anti-FcgRIII/II(克隆2.4G2)抗体冰上放置15min阻断非特异结合,然后加入相应的荧光偶联抗体4℃避光放置30min染色。Fixable viability Dye eFluorTM 506或者Dye eFluorTM 780染色用来排除死细胞。染色后PBS洗去游离抗体,进行流式细胞上机实验,数据使用FlowJo软件(Treestar)进行分析。After digestion and centrifugation, cells were prepared into a cell suspension. Anti-FcgRIII/II (clone 2.4G2) was added to block nonspecific binding and incubated on ice for 15 minutes. The corresponding fluorescently conjugated antibodies were then added and incubated at 4°C in the dark for 30 minutes for staining. Fixable viability Dye eFluor™ 506 or Dye eFluor™ 780 was used to exclude dead cells. After staining, free antibodies were washed with PBS to remove free antibodies. Flow cytometry was performed, and data were analyzed using FlowJo software (Treestar).

流式微球阵列(Cytometric Bead Array,CBA)和ELISA分析血清和组织样本Cytometric Bead Array (CBA) and ELISA analysis of serum and tissue samples

CBAMouse Th1/Th2/Th17 Kit(BD Biosciences)依据说明书操作测量小鼠血清和肿瘤组织匀浆中的细胞因子水平。ELISA采用96孔微孔板(Corning Costar),2μg/mL(100μL/孔)捕获抗体加入到板中4℃过夜吸附。PBS洗板,然后加入封闭液(PBS含0.05%TWEEN-20和5%脱脂牛奶)封闭后,加入用封闭液稀释的血清或肿瘤匀浆液,37℃放置1.5小时。PBST洗板后加入碱性磷酸酶(Alkaline Phosphatase)标记的Goat Anti-Human IgG二抗,37℃放置50min。PBST洗板后加入100μL p-Nitrophenyl Phosphate显色后用SPECTROstar Nano(BMG LABTECH),在405nm波长读板。The CBAMouse Th1/Th2/Th17 Kit (BD Biosciences) was used according to the manufacturer's instructions to measure cytokine levels in mouse serum and tumor tissue homogenates. ELISA was performed using a 96-well microplate (Corning Costar). 2 μg/mL (100 μL/well) of capture antibody was added to the plate and adsorbed overnight at 4°C. The plate was washed with PBS and then blocked with blocking buffer (PBS containing 0.05% TWEEN-20 and 5% skim milk). Serum or tumor homogenate diluted in blocking buffer was then added and incubated at 37°C for 1.5 hours. After washing with PBST, alkaline phosphatase-labeled Goat Anti-Human IgG secondary antibody was added and incubated at 37°C for 50 minutes. After washing with PBST, 100 μL of p-Nitrophenyl Phosphate was added for color development and the plate was read at 405 nm using a SPECTROstar Nano (BMG LABTECH).

T细胞体外培养和过继细胞转移:T cell culture in vitro and adoptive cell transfer:

为了在体外生成激活的OT-I细胞毒性T细胞(CTLs),从小鼠脾细胞中用1mM SIINFEKL肽(New England Peptide)以108细胞/mL的浓度刺激。肽脉冲2小时后,洗去肽,将脾细胞以106细胞/mL的浓度在T细胞培养基中培养,添加5ng/mL小鼠重组IL-2(mrIL-2)和10ng/mL mrIL-12(R&D),或仅添加5ng/mL mrIL-2,并每日补充。在第4天,将2×106Ficoll分离的活OT-1细胞通过静脉注射过继转移到B16-OVA肿瘤小鼠中。To generate activated OT-1 cytotoxic T cells (CTLs) in vitro, splenocytes from mice were stimulated with 1 mM SIINFEKL peptide (New England Peptide) at a concentration of 108 cells/mL. After a 2-hour peptide pulse, the peptide was washed away, and splenocytes were cultured at a concentration of 106 cells/mL in T cell culture medium supplemented with 5 ng/mL mouse recombinant IL-2 (mrIL-2) and 10 ng/mL mrIL-12 (R&D), or 5 ng/mL mrIL-2 alone, and supplemented daily. On day 4, 2 × 106 Ficoll-separated, viable OT-1 cells were adoptively transferred into B16-OVA tumor-bearing mice via intravenous injection.

对于体内细胞转移实验,将105个幼稚OT1细胞转移到幼稚小鼠中,并在细胞转移后24小时用10μg mtEGFP-OVA或mtIL12-OVAmRNA疫苗进行免疫。免疫后第7天,从小鼠脾脏中分离出激活的OT-1细胞,纯化后将5×105个OT-1细胞通过静脉注射转移到B16-OVA肿瘤小鼠中。For in vivo cell transfer experiments, 10 5 naive OT-1 cells were transferred into naive mice, and 24 hours after cell transfer, the mice were immunized with 10 μg of mtEGFP-OVA or mtIL12-OVA mRNA vaccine. Seven days after immunization, activated OT-1 cells were isolated from the spleen of the mice, purified, and 5 × 10 5 OT-1 cells were transferred intravenously into B16-OVA tumor-bearing mice.

mRNA生产mRNA production

包含mRNA完整序列并且携带poly-A序列的DNA质粒通过酶切法制备体外转录模板,模板通过T7Co-transcription RNASynthesis Kit(C3111)进行共转录加帽。体外转录生产的mRNA通过纤维素法去除双链RNA,并储存于-80冰箱以待后续实验。DNA plasmid containing the complete sequence of mRNA and carrying poly-A sequence was digested to prepare in vitro transcription template. Co-transcription capping was performed using the T7 Co-transcription RNA Synthesis Kit (C3111). mRNA produced by in vitro transcription was decapped using the cellulose method and stored at -80°C for subsequent experiments.

mRNA纳米脂质体包装:mRNA nanoliposome packaging:

mRNA被溶解于pH=4.0的100毫摩柠檬酸钠缓冲液中。DSPC,胆固醇,DMG-PEG2000,SM102以38.5:10:1.5:50的摩尔比溶解在无水乙醇中。调整LNP和RNA质量比为30:1-50:1,然后将溶于柠檬酸钠溶液的RNA和溶于无水乙醇的LNP混合物以3:1体积比用微流控系统混合,制备为mRNA-LNP纳米脂质体。将mRNA纳米脂质体溶液放入PBS缓冲液中透析,透析完成的mRNA纳米脂质体可以用于体内外动物实验。mRNA was dissolved in 100 mM sodium citrate buffer (pH 4.0). DSPC, cholesterol, DMG-PEG2000, and SM102 were dissolved in anhydrous ethanol at a molar ratio of 38.5:10:1.5:50. The LNP:RNA mass ratio was adjusted between 30:1 and 50:1. The RNA in sodium citrate and the LNP in anhydrous ethanol were then mixed at a volume ratio of 3:1 using a microfluidic system to prepare mRNA-LNP nanoliposomes. The mRNA nanoliposome solution was dialyzed against PBS buffer. The dialyzed mRNA nanoliposomes were suitable for in vitro and in vivo animal experiments.

数据分析Data Analysis

荷瘤小鼠治疗前,根据肿瘤体积随机分配到不同组。数据使用GraphPad Prism统计软件分析,展示为平均值±SEM。肿瘤曲线用two-way ANOVA分析方法计算P值,小鼠存活曲线用Log-rank test分析计算P值,其他数据用unpaired two-tailed t-tests检验分析。p<0.05代表有显著性差异。Before treatment, tumor-bearing mice were randomly assigned to different groups based on tumor volume. Data were analyzed using GraphPad Prism statistical software and presented as mean ± SEM. P values were calculated using two-way ANOVA for tumor curves and log-rank tests for mouse survival curves. All other data were analyzed using unpaired two-tailed t-tests. A p value < 0.05 indicated significant differences.

结果示于以下实施例中。The results are shown in the following examples.

实施例1.IL-12增强mRNA肿瘤疫苗的疗效以实现优越的肿瘤控制Example 1. IL-12 enhances the efficacy of mRNA tumor vaccines to achieve superior tumor control

有效诱导、激活和分化CD8+T细胞需要除了TCR-pMHC识别和共刺激信号之外的第三信号(Tugues等人,2015)。尽管目前的mRNA-LNP疫苗能够扩增大量肿瘤特异性CD8+T细胞(图1的A-B,序列S1(SEQ ID NO:69)),但它们仅部分抑制肿瘤生长(图1的C-E)。已知这种mRNA疫苗能够诱导高水平的IL-1β、IL-6和I型干扰素,从而促进免疫激活(Li等人,2022)。然而,这些细胞因子可能无法有效激活CD8+T细胞以实现最佳的肿瘤控制。为了确定能够与mRNA疫苗协同作用的关键细胞因子信号,我们分别筛选了几种T淋巴细胞刺激细胞因子,包括IL2、IL15和IL12,以用于肿瘤治疗。这些细胞因子已知可以通过不同的下游转录因子促进T细胞增殖、激活或效应分化(Propper和Balkwill,2022)。为了将这些细胞因子递送至引流淋巴结(dLN)以协助T细胞启动,我们将特定的Fc融合细胞因子与mRNA疫苗共同肌肉注射到肿瘤携带小鼠的同一部位(图1的A)。尽管IL-2和IL-15未能进一步增强mRNA疫苗的抗肿瘤效果(图1的C-D),但IL-12显著增强了疫苗的疗效,导致明显的肿瘤抑制(图1的E)。这些结果促使我们将IL-12作为佐剂纳入mRNA疫苗中,以更好地激活抗原特异性T细胞并改善肿瘤控制。Effective induction, activation, and differentiation of CD8+ T cells requires a third signal in addition to TCR-pMHC recognition and co-stimulatory signals (Tugues et al., 2015). Although current mRNA-LNP vaccines can expand a large number of tumor-specific CD8+ T cells (Figure 1A-B, Sequence S1 (SEQ ID NO:69)), they only partially inhibit tumor growth (Figure 1C-E). Such mRNA vaccines are known to induce high levels of IL-1β, IL-6, and type I interferon, thereby promoting immune activation (Li et al., 2022). However, these cytokines may not effectively activate CD8+ T cells for optimal tumor control. To identify key cytokine signals that can synergize with mRNA vaccines, we screened several T lymphocyte-stimulating cytokines, including IL2, IL15, and IL12, for tumor therapy. These cytokines are known to promote T cell proliferation, activation, or effector differentiation through different downstream transcription factors (Propper and Balkwill, 2022). To deliver these cytokines to the draining lymph nodes (dLN) to assist T cell priming, we co-injected specific Fc-fused cytokines with the mRNA vaccine intramuscularly into the same site of tumor-bearing mice (Figure 1A). Although IL-2 and IL-15 failed to further enhance the anti-tumor effect of the mRNA vaccine (Figure 1C-D), IL-12 significantly enhanced the efficacy of the vaccine, resulting in significant tumor inhibition (Figure 1E). These results prompted us to include IL-12 as an adjuvant in the mRNA vaccine to better activate antigen-specific T cells and improve tumor control.

实施例2.膜结合型IL-12基mRNA疫苗实现优越的抗肿瘤疗效且毒性有限Example 2. Membrane-bound IL-12-based mRNA vaccine achieves superior anti-tumor efficacy with limited toxicity

为了研究IL-12作为mRNA疫苗佐剂的潜力,我们构建了一种IL12-OVA融合mRNA疫苗,用于治疗表达OVA的B16F10-OVA肿瘤模型。在此构建中,IL-12的分泌型与OT1表位编码序列通过2A连接子连接(sIL12-OVA)(图2的A,上图,序列19,即SEQ ID NO:67)。对于对照mRNA疫苗,IL-12编码序列被替换为免疫原性较低的EGFP(Skelton等人,2001)。已知IL12能够激活CD4、CD8和NK细胞,并诱导强效的IFN-γ分泌(Guo等人,2012)。我们假设sIL12-OVAmRNA疫苗后翻译的IL-12蛋白的系统性扩散可能导致观察到的毒性(Hewitt等人,2020)。为解决这一问题,我们进一步开发了一种膜结合型IL12(mtIL12)佐剂mRNA疫苗(mtIL12-OVA),旨在将IL-12限制在dLN或脾脏中的抗原递呈细胞表面。在此构建中,IL-12与CD80分子的跨膜结构域融合,然后通过2A连接子与肿瘤抗原连接(图2的A,下图,序列10,即SEQ ID NO:58)。体外转染DC2.4细胞证实mtIL12-OVAmRNA转染后可以在细胞表面同时呈递OT-1多肽/MHC-I复合物和IL-12细胞因子(图2的B,序列10,即SEQ ID NO:58)。体外转染sIL12-OVAmRNA能够诱导IL-12分泌到培养上清中(图2的C,序列19,即SEQ ID NO:67);而mtIL12-OVAmRNA转染证实IL-12锚定在细胞表面(图2的B,序列10,即SEQ ID NO:58),几乎没有渗漏到上清液中(图2的B-C)。To investigate the potential of IL-12 as an adjuvant for mRNA vaccines, we constructed an IL12-OVA fusion mRNA vaccine for treatment of an OVA-expressing B16F10-OVA tumor model. In this construct, the secreted form of IL-12 was linked to the OT1 epitope-encoding sequence via a 2A linker (sIL12-OVA) (Figure 2A, upper panel, sequence 19, i.e., SEQ ID NO: 67). For the control mRNA vaccine, the IL-12 coding sequence was replaced with the less immunogenic EGFP (Skelton et al., 2001). IL-12 is known to activate CD4, CD8, and NK cells and induce potent IFN-γ secretion (Guo et al., 2012). We hypothesized that systemic spread of the translated IL-12 protein following sIL12-OVA mRNA vaccination might contribute to the observed toxicity (Hewitt et al., 2020). To address this issue, we further developed a membrane-bound IL12 (mtIL12) adjuvanted mRNA vaccine (mtIL12-OVA) designed to restrict IL-12 to the surface of antigen-presenting cells in dLN or spleen. In this construct, IL-12 was fused to the transmembrane domain of the CD80 molecule and then linked to the tumor antigen via a 2A linker (Figure 2A, lower panel, sequence 10, i.e., SEQ ID NO: 58). In vitro transfection of DC2.4 cells confirmed that mtIL12-OVA mRNA transfection could simultaneously present the OT-1 peptide/MHC-I complex and the IL-12 cytokine on the cell surface (Figure 2B, sequence 10, i.e., SEQ ID NO: 58). In vitro transfection of sIL12-OVA mRNA induced IL-12 secretion into the culture supernatant (Figure 2C, sequence 19, i.e., SEQ ID NO: 67); while mtIL12-OVA mRNA transfection confirmed that IL-12 was anchored on the cell surface (Figure 2B, sequence 10, i.e., SEQ ID NO: 58), with almost no leakage into the supernatant (Figure 2B-C).

在B16-OVA肿瘤携带小鼠中,sIL12-OVA疫苗显著改善了治疗效果并延长了肿瘤控制时间,与对照mRNA疫苗相比(图3的A,sIL12-OVA序列19(SEQ ID NO:67),对照序列S1(SEQ ID NO:69))。然而,sIL12-OVAmRNA疫苗导致接种小鼠体重显著下降(图3的B),表明存在系统性毒性。在体内,mtIL12-OVA显示出与sIL12-OVA相似的肿瘤生长抑制和延长生存的效果(图3的A,mtIL12-OVA序列10(SEQ ID NO:58),sIL12-OVA序列19(SEQ ID NO:67)),值得注意的是,mtIL12-OVA给药并未加剧与可溶性IL12相关的毒性,与对照OVAmRNA治疗组小鼠一样,体重短暂下降后快速恢复(图3的B)。与sIL12-OVA相比,mtIL12-OVA诱导的血清IL-12、IFN-γ和MCP-1水平显著降低(图2的C)。且IL-12仅在dLN内的单核细胞、树突状细胞和巨噬细胞表面检测到(图4的A)。这些结果表明,膜结合型IL12有效地将IL-12限制在APCs表面,防止其释放到外周组织。In B16-OVA tumor-bearing mice, sIL12-OVA vaccine significantly improved therapeutic efficacy and prolonged tumor control compared with the control mRNA vaccine (Figure 3A, sIL12-OVA sequence 19 (SEQ ID NO: 67), control sequence S1 (SEQ ID NO: 69)). However, sIL12-OVA mRNA vaccine caused a significant decrease in body weight in vaccinated mice (Figure 3B), indicating the presence of systemic toxicity. In vivo, mtIL12-OVA showed similar tumor growth inhibition and survival-prolonging effects as sIL12-OVA (Figure 3A, mtIL12-OVA sequence 10 (SEQ ID NO: 58), sIL12-OVA sequence 19 (SEQ ID NO: 67)). Notably, mtIL12-OVA administration did not exacerbate the toxicity associated with soluble IL12. As with the control OVA mRNA-treated mice, the body weight decreased briefly but recovered rapidly (Figure 3B). Compared with sIL12-OVA, mtIL12-OVA-induced serum IL-12, IFN-γ, and MCP-1 levels were significantly reduced (Figure 2C). IL-12 was only detected on the surface of monocytes, dendritic cells, and macrophages within dLNs (Figure 4A). These results indicate that membrane-bound IL12 effectively restricts IL-12 to the surface of APCs, preventing its release into peripheral tissues.

为了进一步阐明为什么mtIL12佐剂疫苗能够有效分离毒性副作用与增强的抗肿瘤效果,我们进行了实验,将1×106OT-1TCR转基因CD8 T细胞分选并转移到野生型(WT)小鼠中,随后用对照OVA、sIL12-OVA或mtIL12-OVAmRNA疫苗进行免疫(图4的B和4C,序列S1(SEQ ID NO:69),10(SEQ ID NO:58),19(SEQ ID NO:67))。与对照OVA疫苗相比,sIL12-OVA和mtIL12-OVA疫苗在dLN中诱导了相当数量的抗原特异性IFN-γ阳性的细胞(图4的B)。然而,sIL12-OVA疫苗还在dLN(图4的B)和外周血(图4的C)中诱导了更高数量的IFN-γ阳性的旁观者CD8+T细胞、CD4+T细胞和NK细胞,而mtIL12-OVA组中未观察到这种现象。这种差异可能解释了sIL12-OVA组中更高的血清IFN-γ水平和外周毒性,而mtIL12-OVA组中未观察到这些现象。综上所述,这些结果表明,膜结合型IL-12细胞因子佐剂的mRNA疫苗能够显著增强抗肿瘤效果,同时最小化不良反应。To further elucidate why mtIL12-adjuvanted vaccines can effectively separate toxic side effects from enhanced antitumor effects, we conducted experiments in which 1×106 OT-1 TCR transgenic CD8 T cells were sorted and transferred into wild-type (WT) mice, which were then immunized with control OVA, sIL12-OVA, or mtIL12-OVA mRNA vaccines (Figure 4B and 4C, sequences S1 (SEQ ID NO: 69), 10 (SEQ ID NO: 58), 19 (SEQ ID NO: 67)). Compared with the control OVA vaccine, sIL12-OVA and mtIL12-OVA vaccines induced a considerable number of antigen-specific IFN-γ-positive cells in the dLN (Figure 4B). However, the sIL12-OVA vaccine also induced higher numbers of IFN-γ-positive bystander CD8+ T cells, CD4+ T cells, and NK cells in dLN (Figure 4B) and peripheral blood (Figure 4C), which was not observed in the mtIL12-OVA group. This difference may explain the higher serum IFN-γ levels and peripheral toxicity in the sIL12-OVA group, which were not observed in the mtIL12-OVA group. Taken together, these results indicate that mRNA vaccines with membrane-bound IL-12 cytokine adjuvants can significantly enhance antitumor effects while minimizing adverse reactions.

在体内,mtIL12-OVAmRNA疫苗治疗效果优于单独使用OVA-mRNA或着单独的mtIL12 mRNA治疗(图5,序列S1(SEQ ID NO:69),序列19(SEQ ID NO:67),序列S15(SEQ ID NO:83)),表明IL-12细胞因子和抗原的同时呈递协同增强了抗肿瘤效果。In vivo, the therapeutic effect of mtIL12-OVA mRNA vaccine was superior to that of OVA-mRNA or mtIL12 mRNA alone (Figure 5, sequence S1 (SEQ ID NO: 69), sequence 19 (SEQ ID NO: 67), sequence S15 (SEQ ID NO: 83)), indicating that the simultaneous presentation of IL-12 cytokine and antigen synergistically enhanced the anti-tumor effect.

实施例3.基于mtIL12的mRNA疫苗依赖CD8 T细胞实现肿瘤控制并诱导独特的预效应CD8 T细胞亚群Example 3. mtIL12-based mRNA vaccines rely on CD8 T cells to achieve tumor control and induce unique pre-effector CD8 T cell subsets

基于IL-12的疗法可以作用于多种免疫细胞类型,包括CD4+T细胞、CD8+T细胞、NK细胞和巨噬细胞,以优化肿瘤控制(Xue等人,2022)。为了确定哪些细胞亚群对于mtIL12-OVAmRNA疫苗介导的肿瘤控制是必需的,我们使用抗体选择性地删除特定免疫细胞亚群。删除NK细胞、CD4+T细胞或巨噬细胞并未影响疫苗的抗肿瘤效果(图6的A和B,序列19(SEQ ID NO:67))。相比之下,CD8+T细胞的删除极大地削弱了疫苗介导的肿瘤抑制和小鼠生存获益,表明CD8+T细胞反应对于疫苗的抗肿瘤效果至关重要。IL-12-based therapies can act on multiple immune cell types, including CD4+ T cells, CD8+ T cells, NK cells, and macrophages, to optimize tumor control (Xue et al., 2022). To determine which cell subsets are essential for tumor control mediated by the mtIL12-OVA mRNA vaccine, we used antibodies to selectively delete specific immune cell subsets. Deletion of NK cells, CD4+ T cells, or macrophages did not affect the anti-tumor effect of the vaccine (Figure 6A and B, sequence 19 (SEQ ID NO:67)). In contrast, deletion of CD8+ T cells greatly impaired vaccine-mediated tumor suppression and mouse survival benefits, indicating that CD8+ T cell responses are critical for the anti-tumor effect of the vaccine.

我们进一步探索了mtIL12-OVA疫苗增强抗肿瘤效果的细胞和分子机制。与对照OVAmRNA疫苗相比,mtIL12-OVA疫苗免疫后在引流淋巴结和脾脏中诱导了更高比例的抗原特异性CD8+T细胞(图6的C)。We further explored the cellular and molecular mechanisms by which the mtIL12-OVA vaccine enhanced its anti-tumor effect. Compared with the control OVA mRNA vaccine, the mtIL12-OVA vaccine induced a higher proportion of antigen-specific CD8+ T cells in the draining lymph nodes and spleen after immunization (Figure 6C).

进一步我们通过流式细胞术分析T细胞分化阶段,我们采用了一种基于Ly108和CD69的分群策略,该策略可以区分慢性感染或肿瘤模型中抗原特异性T细胞的分化轨迹(Beltra等人,2020)。C57BL/6J小鼠在第0天和第7天肌肉注射2.5ug mtEGFP-OVA或者mtIL12-OVARNA,第14天取血进行流式分析(图7的A)。对照OVAmRNA主要诱导Ly108+CD69-T细胞亚群(前体状态),而mtIL12-OVA疫苗主要在外周血中诱导Ly108-CD69-T细胞亚群(效应状态)(图7的B)。与对照OVAmRNA组相比,mtIL12-OVA疫苗组的PBMCs中granzyme B阳性或IFN-γ阳性OVA特异性CD8+T细胞的比例显著更高(图7的C)。此外,mtIL12-OVA组中OVA特异性CD8+T细胞表达效应T细胞转录因子T-bet的比例更高,而抑制性分子PD-1的水平更低(图7的C)。为了进一步验证这些细胞的功能能力,我们从mtIL12-OVA或对照OVAmRNA疫苗免疫过的小鼠中分离脾细胞,用不同稀释度的SIINFEKL肽刺激,并通过ELISA分析IFN-γ分泌。两组的基线IFN-γ分泌量均较低,但在肽刺激后,两组的脾细胞均以剂量依赖性方式分泌IFN-γ,且mtIL12-OVA组检测到的量显著更高(图7的D)。这些结果表明,mtIL12-OVA疫苗诱导的外周抗原特异性T细胞是具有高度效力的预效应T细胞,它们在重新遇到抗原时表现出更高的敏感性和更强的效应反应,从而与传统OVAmRNA疫苗相比实现了增强的肿瘤抑制。We further analyzed T cell differentiation stages by flow cytometry. We employed a Ly108- and CD69-based clustering strategy that can distinguish the differentiation trajectories of antigen-specific T cells in chronic infection or tumor models (Beltra et al., 2020). C57BL/6J mice were intramuscularly injected with 2.5 μg mtEGFP-OVA or mtIL12-OVA RNA on days 0 and 7, and blood was collected on day 14 for flow cytometry analysis (Figure 7A). The control OVA mRNA predominantly induced the Ly108+CD69- T cell subset (precursor state), while the mtIL12-OVA vaccine predominantly induced the Ly108-CD69- T cell subset (effector state) in peripheral blood (Figure 7B). Compared with the control OVA mRNA group, the proportion of granzyme B-positive or IFN-γ-positive OVA-specific CD8+ T cells in PBMCs was significantly higher in the mtIL12-OVA vaccine group (Figure 7C). In addition, the proportion of OVA-specific CD8+ T cells expressing the effector T cell transcription factor T-bet was higher in the mtIL12-OVA group, while the level of the inhibitory molecule PD-1 was lower (Figure 7C). To further verify the functional capacity of these cells, we isolated splenocytes from mice immunized with mtIL12-OVA or control OVA mRNA vaccines, stimulated them with different dilutions of SIINFEKL peptide, and analyzed IFN-γ secretion by ELISA. Baseline IFN-γ secretion was low in both groups, but after peptide stimulation, splenocytes from both groups secreted IFN-γ in a dose-dependent manner, and the amount detected in the mtIL12-OVA group was significantly higher (Figure 7D). These results indicate that peripheral antigen-specific T cells induced by the mtIL12-OVA vaccine are highly potent pre-effector T cells that exhibit higher sensitivity and stronger effector responses when they encounter antigen again, thereby achieving enhanced tumor suppression compared to traditional OVA mRNA vaccines.

接下来我们分析mtIL12和OVA抗原串联在一起的设计(mtIL12-OVA)是否比两者的简单混合应用(OVA+mtIL12)免疫效果更好。B6小鼠在第0天和第7天静脉注射2ug mtEGFP-OVA,2ug mtEGFP-OVA+1ug mtIL12或1ug mtEGFP-OVA+1ug mtIL12-OVA RNA-LNP,第14天取血进行流式分析(图8的A)。流式分析显示顺式递送IL12和抗原的小鼠(OVA+mtIL12-OVA)产生了更高比例和数目的外周血抗原特异性T细胞(图8的B,序列S1(SEQ ID NO:69),序列19(SEQ ID NO:67),序列S15(SEQ ID NO:83))。Next, we analyzed whether a design in which mtIL12 and OVA antigens were delivered in tandem (mtIL12-OVA) had a better immune effect than a simple mixture of the two (OVA+mtIL12). B6 mice were intravenously injected with 2 μg mtEGFP-OVA, 2 μg mtEGFP-OVA + 1 μg mtIL12, or 1 μg mtEGFP-OVA + 1 μg mtIL12-OVA RNA-LNP on days 0 and 7. Blood was collected on day 14 for flow cytometry analysis (Figure 8A). Flow cytometry analysis showed that mice delivered with IL12 and antigen in cis (OVA+mtIL12-OVA) generated a higher proportion and number of antigen-specific T cells in peripheral blood (Figure 8B, Sequence S1 (SEQ ID NO: 69), Sequence 19 (SEQ ID NO: 67), Sequence S15 (SEQ ID NO: 83)).

实施例4.膜结合型IL12佐剂mRNA疫苗克服免疫检查点阻断耐药性Example 4. Membrane-bound IL12 adjuvanted mRNA vaccine overcomes immune checkpoint blockade resistance

免疫检查点阻断(ICB)疗法在临床环境中显示出有希望的治疗结果(Hargadon等人,2018),然而,只有少数患者能够有效响应(Sharma等人,2017)。耐药性的一个机制是“免疫冷”肿瘤中缺乏足够的肿瘤浸润性淋巴细胞(TILs)。我们认为,mtIL12佐剂mRNA癌症疫苗可以通过向免疫冷肿瘤中提供足够的抗原特异性TIL来克服ICB疗法的耐药性。为了验证,我们在B16-OVA肿瘤携带小鼠中用mtIL12-OVA(图9的A,序列19,SEQ ID NO:67)或ICB抗体(抗PD-L1加抗CTLA4)进行治疗,这些肿瘤已经充分建立。尽管ICB疗法单独对肿瘤生长和生存几乎没有影响,但mtIL12-OVA疫苗显著抑制了肿瘤生长。此外,将mtIL12-OVA与ICB疗法联合使用进一步改善了肿瘤抑制并延长了生存时间(图9的A)。Immune checkpoint blockade (ICB) therapy has shown promising results in the clinical setting (Hargadon et al., 2018); however, only a minority of patients achieve effective responses (Sharma et al., 2017). One mechanism of resistance is the lack of sufficient tumor-infiltrating lymphocytes (TILs) in “immunologically cold” tumors. We hypothesized that mtIL12-adjuvanted mRNA cancer vaccines could overcome resistance to ICB therapy by providing sufficient antigen-specific TILs to immune-cold tumors. To validate this, we treated B16-OVA tumor-bearing mice with either mtIL12-OVA (Figure 9A, sequence 19, SEQ ID NO:67) or ICB antibodies (anti-PD-L1 plus anti-CTLA4). Although ICB therapy alone had little effect on tumor growth and survival, the mtIL12-OVA vaccine significantly inhibited tumor growth. Furthermore, combining mtIL12-OVA with ICB therapy further improved tumor suppression and prolonged survival (Figure 9A).

实施例5.膜结合型IL12佐剂适用于HPV相关肿瘤的E6/E7抗原的疫苗设计Example 5. Vaccine design using membrane-bound IL12 adjuvant for E6/E7 antigens in HPV-related tumors

我们进一步研究了膜结合型IL12佐剂mRNA疫苗的设计是否可以扩展到临床上相关的肿瘤特异性抗原。E6和E7癌蛋白直接参与了HPV诱导的致癌过程,无论是在临床前还是临床环境中。尽管对E6/E7靶向治疗性疫苗的研究广泛,但进展有限(Grunwitz等人,2019;Peng等人,2021)。我们在模拟HPV相关癌症的TC-1肿瘤小鼠模型中比较了对照E7 mRNA疫苗与mtIL12-E7疫苗的治疗效果(Lin等人,1996)。在晚期晚期TC-1肿瘤携带小鼠中,对照E7疫苗或mtIL12单独治疗均未能控制肿瘤生长(图10的A,序列S15(SEQ ID NO:83),序列S4(SEQ ID NO:72),序列18(SEQ ID NO:66))。然而,尽管TC-1肿瘤在两次mtIL12-E7疫苗接种后继续生长至约500mm3,但肿瘤最终消退,约80%的肿瘤成功根除(图10的A)。重要的是,与未治疗组相比,接受mtIL12-E7 mRNA疫苗治疗的小鼠没有显著的体重下降(图10的B)。We further investigated whether the design of a membrane-bound IL12-adjuvanted mRNA vaccine could be extended to target clinically relevant tumor-specific antigens. The E6 and E7 oncoproteins are directly involved in HPV-induced carcinogenesis, both in preclinical and clinical settings. Despite extensive research into E6/E7-targeted therapeutic vaccines, progress has been limited (Grunwitz et al., 2019; Peng et al., 2021). We compared the therapeutic efficacy of a control E7 mRNA vaccine with that of an mtIL12-E7 vaccine in a TC-1 tumor mouse model that mimics HPV-associated cancer (Lin et al., 1996). In mice bearing advanced TC-1 tumors, neither the control E7 vaccine nor mtIL12 treatment alone failed to control tumor growth (Figure 10A, Sequence S15 (SEQ ID NO:83), Sequence S4 (SEQ ID NO:72), Sequence 18 (SEQ ID NO:66)). However, although TC-1 tumors continued to grow to approximately 500 mm 3 after two doses of mtIL12-E7 vaccination, they ultimately regressed, with approximately 80% of tumors successfully eradicated ( FIG10A ). Importantly, mice treated with the mtIL12-E7 mRNA vaccine did not experience significant weight loss compared to the untreated group ( FIG10B ).

TC-1荷瘤小鼠在肿瘤种植第12天被2.5ug mtEGFP-E7,第18天被2.5ug mtEGFP-E7或者2.5ug mtIL12-E7免疫,第22天取肿瘤组织进行流式分析(图11的A)。结果显示IL12免疫促进E7特异性T细胞的肿瘤内富集,并且相对于mtGFP-E7免疫产生的T细胞,mtIL12-E7免疫产生的T细胞更多停留在效应状态(LY108-CD69-)而不是耗竭状态(LY108-CD69+),并且表达更高的效应分子GZMB,和更低的抑制性标志PD-1(图11的C).TC-1 tumor-bearing mice were immunized with 2.5 μg mtEGFP-E7 on day 12 of tumor implantation and with either 2.5 μg mtEGFP-E7 or 2.5 μg mtIL12-E7 on day 18. Tumor tissue was harvested and analyzed by flow cytometry on day 22 (Figure 11A). The results showed that IL12 immunization promoted the intratumoral enrichment of E7-specific T cells. Compared with T cells immunized with mtGFP-E7, T cells immunized with mtIL12-E7 were more likely to remain in the effector state (LY108-CD69-) rather than the exhausted state (LY108-CD69+), and expressed higher levels of the effector molecule GZMB and lower levels of the inhibitory marker PD-1 (Figure 11C).

实施例6.膜结合型IL12佐剂适用于逆转录病毒来源的肿瘤新抗原的疫苗设计Example 6. Membrane-bound IL12 adjuvant is suitable for vaccine design against retroviral-derived tumor neoantigens

内源性逆转录病毒(ERVs)基因整合到宿主基因组中,并在健康组织中通常保持沉默,但在癌症等病理条件下其表达被重新激活(Cherkasova等人,2013;Smith等人,2018)。这些重新激活的ERV蛋白为癌症疫苗开发提供了可行的靶点。P15E蛋白,源自gp70逆转录病毒基因表达,在许多癌症中被检测到,包括MC38结肠腺癌细胞系(Ye等人,2020)。C57BL/6小鼠皮下接种4X105的MC38肿瘤,小鼠在肿瘤形成后第12和第16天肌肉注射2.5μg的mtIL12-p15E或者对照mtEGFP-p15E mRNA-LNP,每周监测两次肿瘤生长曲线。结果如图12所示对照P15E mRNA疫苗治疗对肿瘤生长的影响最小,而mtIL12-P15E治疗诱导了显著的肿瘤消退和完全根除(图12,序列S5(SEQ ID NO:73),序列S9(SEQ ID NO:77))。Endogenous retroviruses (ERVs) integrate into the host genome and are typically silent in healthy tissues, but their expression is reactivated under pathological conditions such as cancer (Cherkasova et al., 2013; Smith et al., 2018). These reactivated ERV proteins provide viable targets for cancer vaccine development. The p15E protein, derived from the gp70 retroviral gene, has been detected in many cancers, including the MC38 colon adenocarcinoma cell line (Ye et al., 2020). C57BL/6 mice were subcutaneously inoculated with 4 × 10 5 MC38 tumors. Mice were injected intramuscularly with 2.5 μg of mtIL12-p15E or control mtEGFP-p15E mRNA-LNP on days 12 and 16 after tumor establishment, and tumor growth curves were monitored twice weekly. The results are shown in Figure 12. The control P15E mRNA vaccine treatment had minimal effect on tumor growth, while mtIL12-P15E treatment induced significant tumor regression and complete eradication (Figure 12, Sequence S5 (SEQ ID NO: 73), Sequence S9 (SEQ ID NO: 77)).

MC38荷瘤小鼠在肿瘤种植第12天肌肉注射2.5ug mtEGFP-p15E,第18天肌肉注射2.5ug mtEGFP-p15E或者2.5ug mtIL12-p15E免疫,第21天取外周血进行流式分析(图13的A)。结果显示相较于mtEGFP-P15E mRNA疫苗,mtIL12-P15E免疫产生了更多的p15E抗原特异性T细胞,并且相对于mtGFP-p15E免疫产生的T细胞,mtIL12-p15E免疫产生的T细胞表达更高的效应分子IFN-gamma,和更低的免疫抑制性分子PD-1.MC38 tumor-bearing mice were immunized with 2.5 μg of mtEGFP-p15E intramuscularly on day 12 after tumor implantation. On day 18, either 2.5 μg of mtEGFP-p15E or 2.5 μg of mtIL12-p15E was injected intramuscularly. Peripheral blood was collected on day 21 for flow cytometry analysis (Figure 13A). The results showed that compared to the mtEGFP-p15E mRNA vaccine, mtIL12-p15E immunization resulted in more p15E-specific T cells. Furthermore, compared to T cells immunized with mtGFP-p15E, T cells immunized with mtIL12-p15E expressed higher levels of the effector molecule IFN-gamma and lower levels of the immunosuppressive molecule PD-1.

总之,这些实验表明,基于mtIL12的疫苗策略适用于针对多种临床上相关的抗原表位进行癌症治疗。Together, these experiments demonstrate that mtIL12-based vaccine strategies are suitable for cancer therapy targeting a variety of clinically relevant epitopes.

讨论:discuss:

细胞因子在T细胞激活过程中作为第三信号发挥作用。mRNA-LNP的自佐剂效应主要诱导产生促炎症细胞因子,包括IL-1β、IL-6和I型干扰素(Li等人,2022;Tahtinen等人,2022),这些细胞因子可能无法最佳支持抗原特异性CD8+T细胞反应。在筛选了多种T细胞刺激细胞因子后,我们发现IL-12是增强mRNA疫苗抗肿瘤效果的最有效细胞因子。Cytokines act as a tertiary signal during T cell activation. The self-adjuvant effect of mRNA-LNPs primarily induces the production of proinflammatory cytokines, including IL-1β, IL-6, and type I interferons (Li et al., 2022; Tahtinen et al., 2022), which may not optimally support antigen-specific CD8+ T cell responses. After screening multiple T cell-stimulating cytokines, we found that IL-12 was the most effective cytokine in enhancing the antitumor effect of mRNA vaccines.

许多研究都集中在将IL-12局部递送至肿瘤微环境(TME)中,以增强抗肿瘤免疫反应,同时减轻系统性毒性(Nguyen等人,2020)。目前的临床方法包括通过瘤内转染编码IL-12的质粒或mRNA来实现(Hewitt等人,2020;Telli等人,2021)。然而,这些局部递送方法面临重大挑战,尤其是对于深部实体瘤,且仍存在IL-12扩散至循环系统的风险,导致毒性反应和治疗窗口受限。我们的研究表明,局部注射编码可溶性IL-12的mRNA会导致IL-12大量渗漏至血清中,从而引发随后的IFN-γ反应,而IFN-γ是与IL-12相关毒性密切相关的关键促炎症细胞因子。通过将IL-12通过跨膜结构域锚定在转染细胞表面,我们成功地将IL-12的治疗益处与系统性毒性分离。Many studies have focused on local delivery of IL-12 to the tumor microenvironment (TME) to enhance antitumor immune responses while mitigating systemic toxicity (Nguyen et al., 2020). Current clinical approaches involve intratumoral transfection of plasmids or mRNA encoding IL-12 (Hewitt et al., 2020; Telli et al., 2021). However, these local delivery methods face significant challenges, especially for deep-seated solid tumors, and there is still the risk of IL-12 spreading into the circulation, leading to toxic reactions and a limited therapeutic window. Our studies show that local injection of mRNA encoding soluble IL-12 leads to a substantial leakage of IL-12 into the serum, which subsequently triggers an IFN-γ response, a key proinflammatory cytokine closely associated with IL-12-related toxicity. By anchoring IL-12 to the surface of transfected cells via its transmembrane domain, we successfully decoupled the therapeutic benefits of IL-12 from its systemic toxicity.

T细胞功能障碍的特征是肿瘤细胞通过既定机制逃避T细胞介导的细胞毒性(Zebley等人,2024)。有研究表明,幼稚肿瘤特异性T细胞在进入TME后的数小时内会迅速获得功能障碍表型(Philip等人,2017;Rudloff等人,2023)。在我们的研究中,传统mRNA疫苗扩增了大量祖细胞样抗原特异性T细胞,但这些细胞表现出有限的抗肿瘤活性。我们推测,类似的机制可能阻碍了传统mRNA疫苗产生的祖细胞T细胞分化为功能性效应细胞。在mRNA疫苗的启动阶段引入IL-12信号,有助于抗原特异性T细胞分化为预效应表型。有趣的是,这些预效应CD8+T细胞在循环中分泌的效应细胞因子很少,但在重新遇到肿瘤抗原时表现出更高的敏感性和更强的效应反应,这可能有助于解释其有限的外周毒性但增强的抗肿瘤效果。这些细胞还表现出较低水平的抑制性PD-1,并显示出对抑制性TME的抵抗力,增强了抗肿瘤能力。T cell dysfunction is characterized by established mechanisms by which tumor cells evade T cell-mediated cytotoxicity (Zebley et al., 2024). Studies have shown that naive tumor-specific T cells rapidly acquire a dysfunctional phenotype within hours of entering the TME (Philip et al., 2017; Rudloff et al., 2023). In our studies, conventional mRNA vaccines expanded a large number of progenitor-like antigen-specific T cells, but these cells exhibited limited antitumor activity. We speculate that similar mechanisms may hinder the differentiation of progenitor T cells generated by conventional mRNA vaccines into functional effector cells. Introducing IL-12 signaling during the priming phase of mRNA vaccines facilitates the differentiation of antigen-specific T cells into a pre-effector phenotype. Interestingly, these pre-effector CD8+ T cells secrete few effector cytokines in circulation but exhibit increased sensitivity and stronger effector responses upon re-encountering tumor antigens, which may help explain their limited peripheral toxicity but enhanced antitumor efficacy. These cells also express lower levels of inhibitory PD-1 and display resistance to the suppressive TME, enhancing their antitumor capacity.

总之,将膜结合型IL-12作为佐剂显著增强了mRNA肿瘤疫苗的治疗效果,同时最小化了毒性。这种策略适用于多种临床上相关的肿瘤抗原,并具有相当的临床转化潜力。In conclusion, the use of membrane-bound IL-12 as an adjuvant significantly enhanced the therapeutic efficacy of mRNA tumor vaccines while minimizing toxicity. This strategy is applicable to a variety of clinically relevant tumor antigens and has considerable potential for clinical translation.

本发明涉及的序列如下:The sequence involved in the present invention is as follows:

序列信息:


















Sequence information:


















Claims (10)

一种编码膜型IL-12细胞因子佐剂的mRNA肿瘤疫苗,其中,所述mRNA包含编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)三部分。A mRNA tumor vaccine encoding a membrane-type IL-12 cytokine adjuvant, wherein the mRNA comprises three parts: a sequence encoding IL-12 (IL12), a sequence encoding a membrane domain (MD), and a sequence encoding a tumor antigen (Ag). 如权利要求1所述的mRNA肿瘤疫苗,其中,The mRNA tumor vaccine according to claim 1, wherein 所述编码IL-12的序列(IL12)和所述编码膜结构域的序列(MD)位于一条mRNA上,所述编码肿瘤抗原的序列(Ag)位于另一条mRNA上;或者The IL-12 encoding sequence (IL12) and the membrane domain encoding sequence (MD) are located on one mRNA, and the tumor antigen encoding sequence (Ag) is located on another mRNA; or 所述编码IL-12的序列(IL12)、编码膜结构域的序列(MD)以及编码肿瘤抗原的序列(Ag)位于同一条mRNA上,所述三部分的排列顺序为IL12-L1-MD-L2-Ag;或Ag-L2-IL12-L1-MD;The sequence encoding IL-12 (IL12), the sequence encoding the membrane domain (MD), and the sequence encoding the tumor antigen (Ag) are located on the same mRNA, and the arrangement order of the three parts is IL12-L1-MD-L2-Ag; or Ag-L2-IL12-L1-MD; 其中L1为Glycine/Serine多肽连接子,L2为IRES序列或Glycine/Serine多肽连接子或2A多肽连接子,L2优选2A多肽连接子。Wherein L1 is a Glycine/Serine polypeptide linker, L2 is an IRES sequence or a Glycine/Serine polypeptide linker or a 2A polypeptide linker, and L2 is preferably a 2A polypeptide linker. 如权利要求1或2所述的mRNA肿瘤疫苗,其中,所述肿瘤抗原Ag包括突变来源的肿瘤新抗原、病毒来源的肿瘤特异性抗原或肿瘤高表达的相关抗原。The mRNA tumor vaccine according to claim 1 or 2, wherein the tumor antigen Ag includes a tumor neoantigen derived from a mutation, a tumor-specific antigen derived from a virus, or a tumor-related antigen that is highly expressed. 如权利要求3所述的mRNA肿瘤疫苗,其中,所述突变来源的肿瘤新抗原包括KARS G12C、KRASG12D、EGFRvIII或BRAFV600E;所述病毒来源的肿瘤特异性抗原包括HPV E6/E7、内源性逆转录病毒抗原hERT、LMP1或LMP2;所述肿瘤高表达的相关抗原包括WT1、MAGE-A3、GP100、NY-ESO-I、HER2/Neu、Claudin18.2、Mesothelin或MUC。The mRNA tumor vaccine according to claim 3, wherein the mutation-derived tumor neoantigens include KARS G12C, KRASG12D, EGFRvIII or BRAF V600E ; the viral-derived tumor-specific antigens include HPV E6/E7, endogenous retroviral antigen hERT, LMP1 or LMP2; the tumor-related antigens highly expressed include WT1, MAGE-A3, GP100, NY-ESO-I, HER2/Neu, Claudin18.2, Mesothelin or MUC. 如权利要求1或2所述的mRNA肿瘤疫苗,其中,所述编码IL-12细胞因子的序列包含编码IL-12B多肽和IL-12A多肽的ORF,其中IL-12B和IL-12A通过多肽接头(linker)连接,所述linker包括Gly/Serlinker,所述Gly/Serlinker包含(GnS)m,其中n为1、2、3、4、5、6、7、8、9、10、15或20且m为1、2、3、4、5、6、7、8、9、10、15或20;所述linker优选为(G4S)3linker。The mRNA tumor vaccine according to claim 1 or 2, wherein the sequence encoding the IL-12 cytokine comprises an ORF encoding an IL-12B polypeptide and an IL-12A polypeptide, wherein IL-12B and IL-12A are connected by a polypeptide linker, the linker comprising a Gly/Ser linker, the Gly/Ser linker comprising (GnS) m , wherein n is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20 and m is 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15 or 20; the linker is preferably a (G4S) 3 linker. 如权利要求1或2所述的mRNA肿瘤疫苗,其中,所述膜结构域包含跨膜结构域,优选包含跨膜结构域加胞内域,所述跨膜结构域包括CD80跨膜域、CD86跨膜域、CD4跨膜域、CD8跨膜域、VEGFR跨膜域、PDGF-RB跨膜域,优选为CD80跨膜域;所述胞内域包括CD80胞内域、CD86胞内域、CD4胞内域、CD8胞内域、VEGFR胞内域、PDGF-RB胞内域、PDGF-RB胞内域截短体,其中,所述膜结构域优选为CD80跨膜域加CD80胞内域的组合。The mRNA tumor vaccine according to claim 1 or 2, wherein the membrane domain comprises a transmembrane domain, preferably a transmembrane domain plus an intracellular domain, the transmembrane domain includes a CD80 transmembrane domain, a CD86 transmembrane domain, a CD4 transmembrane domain, a CD8 transmembrane domain, a VEGFR transmembrane domain, and a PDGF-RB transmembrane domain, preferably a CD80 transmembrane domain; the intracellular domain includes a CD80 intracellular domain, a CD86 intracellular domain, a CD4 intracellular domain, a CD8 intracellular domain, a VEGFR intracellular domain, a PDGF-RB intracellular domain, and a PDGF-RB intracellular domain truncation, wherein the membrane domain is preferably a combination of a CD80 transmembrane domain plus a CD80 intracellular domain. 如权利要求1或2所述的mRNA肿瘤疫苗,其中,所述mRNA肿瘤疫苗还包含负载mRNA的核酸载体,所述核酸载体包括脂质体、LPP或LPX,优选为LNP纳米脂质体。The mRNA tumor vaccine according to claim 1 or 2, wherein the mRNA tumor vaccine further comprises a nucleic acid vector loaded with mRNA, wherein the nucleic acid vector comprises a liposome, LPP or LPX, preferably an LNP nanoliposome. 一种药物组合物,其包含权利要求1至7中任一项所述的mRNA肿瘤疫苗和第二抗癌剂,所述第二抗癌剂包括免疫检查点抑制剂和/或双特异性T细胞衔接器(BiTE)。A pharmaceutical composition comprising the mRNA tumor vaccine according to any one of claims 1 to 7 and a second anticancer agent, wherein the second anticancer agent comprises an immune checkpoint inhibitor and/or a bispecific T cell engager (BiTE). 权利要求1至7中任一项所述的mRNA肿瘤疫苗或权利要求8所述的药物组合物在制备用于治疗或预防肿瘤的药物中的应用。Use of the mRNA tumor vaccine according to any one of claims 1 to 7 or the pharmaceutical composition according to claim 8 in the preparation of a medicament for treating or preventing tumors. 如权利要求9所述的应用,其中,所述肿瘤包括B细胞淋巴瘤、支气管癌、前列腺癌、膀胱癌、脑或中枢神经系统癌症、外周神经系统癌症、食道癌、宫颈癌、子宫或子宫内膜癌、头颈癌、口腔癌、喉癌、唾液腺癌、胸腺癌、肾上腺癌、骨肉瘤、软骨肉瘤、脂肪癌、睾丸癌、恶性纤维组织细胞瘤、结肠直肠癌、黑色素瘤、胃癌、胰腺癌、肺癌、肝癌、肾癌、胆管癌、小肠癌或阑尾癌、鳞状细胞癌、乳腺癌和卵巢癌。The use according to claim 9, wherein the tumor comprises B-cell lymphoma, bronchial cancer, prostate cancer, bladder cancer, brain or central nervous system cancer, peripheral nervous system cancer, esophageal cancer, cervical cancer, uterine or endometrial cancer, head and neck cancer, oral cancer, laryngeal cancer, salivary gland cancer, thymic cancer, adrenal cancer, osteosarcoma, chondrosarcoma, adipose cancer, testicular cancer, malignant fibrous histiocytoma, colorectal cancer, melanoma, gastric cancer, pancreatic cancer, lung cancer, liver cancer, kidney cancer, bile duct cancer, small intestine cancer or appendix cancer, squamous cell carcinoma, breast cancer and ovarian cancer.
PCT/CN2025/080420 2024-03-04 2025-03-04 Mrna tumor vaccine encoding membrane-bound il-12 cytokine adjuvant, and use thereof Pending WO2025185599A1 (en)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202410245327 2024-03-04
CN202410245327.9 2024-03-04

Publications (2)

Publication Number Publication Date
WO2025185599A1 true WO2025185599A1 (en) 2025-09-12
WO2025185599A8 WO2025185599A8 (en) 2025-10-02

Family

ID=96989972

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2025/080420 Pending WO2025185599A1 (en) 2024-03-04 2025-03-04 Mrna tumor vaccine encoding membrane-bound il-12 cytokine adjuvant, and use thereof

Country Status (1)

Country Link
WO (1) WO2025185599A1 (en)

Also Published As

Publication number Publication date
WO2025185599A8 (en) 2025-10-02

Similar Documents

Publication Publication Date Title
KR102351555B1 (en) Immunity enhancing therapeutic vaccine for hpv and related diseases
AU2014221143B2 (en) Vaccine composition
WO2021228105A1 (en) Oncolytic virus vaccine and drug for treating tumors by combining oncolytic virus vaccine with immune cells
JP2002515734A (en) Immunostimulation mediated by genetically modified dendritic cells
WO2021228106A1 (en) Oncolytic virus in combination with immune checkpoint inhibitor for treating tumors
Neukirch et al. Adenovirus based virus-like-vaccines targeting endogenous retroviruses can eliminate growing colorectal cancers in mice
US20250049916A1 (en) Use of ccl13
AU2023414262A1 (en) Polynucleotide molecule for preventing or treating hpv infection-related diseases
CA2694735A1 (en) Cell-penetrating peptides and use thereof bonded to biomolecules with therapeutic action
JP2005532298A (en) Non-immunosuppressive immunogen or vaccine comprising mutated E7 protein of HPV-16 virus
KR20240124919A (en) Lentiviral vector for expression of human papillomavirus antigen and its implementation in the treatment of HPV-induced cancer
WO2018058489A1 (en) Cacna1h-derived tumour antigen polypeptide and use thereof
TWI748349B (en) Tumor-specific polypeptide sequence and application thereof
CN100393878C (en) Vaccine
WO2025185599A1 (en) Mrna tumor vaccine encoding membrane-bound il-12 cytokine adjuvant, and use thereof
WO2025112755A1 (en) Method for treating tumor with combination of recombinant oncolytic virus and macromolecular antibody-based anti-cancer drug
Peng et al. Therapeutic mRNA vaccines targeting human papillomavirus type 16 E7 protein demonstrate significant antitumor efficacy in murine models of HPV-associated tumors
CN117797253A (en) A tumor vaccine capable of expanding T cell antigenic epitopes and its preparation method
WO2025246436A1 (en) Circular rna-based therapeutic vaccine for treating hpv positive cancer
US20190224291A1 (en) Col14a1-derived tumor antigen polypeptide and use thereof
US20250049903A1 (en) Therapy for Modulating Immune Response with Recombinant MVA Encoding IL-12
Nemeckova et al. Combination of intratumoral injections of vaccinia virus MVA expressing GM-CSF and immunization with DNA vaccine prolongs the survival of mice bearing HPV16 induced tumors with downregulated expression of MHC class I molecules
WO2001078769A2 (en) Regulation of systemic immune responses utilizing soluble cd40
WO2024222886A1 (en) Mrna tumor vaccines for mica/b target
JP2025527186A (en) Lentiviral vectors for the expression of human papillomavirus (HPV) antigens and their use in the treatment of HPV-induced cancers

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: 25767337

Country of ref document: EP

Kind code of ref document: A1