WO2024225764A1 - Immunodominant epitope peptides of fibroblast activating proteins, and uses thereof - Google Patents
Immunodominant epitope peptides of fibroblast activating proteins, and uses thereof Download PDFInfo
- Publication number
- WO2024225764A1 WO2024225764A1 PCT/KR2024/005587 KR2024005587W WO2024225764A1 WO 2024225764 A1 WO2024225764 A1 WO 2024225764A1 KR 2024005587 W KR2024005587 W KR 2024005587W WO 2024225764 A1 WO2024225764 A1 WO 2024225764A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- fap
- peptide
- present
- hla
- tumor
- 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
Links
Images
Classifications
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K7/00—Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
- C07K7/04—Linear peptides containing only normal peptide links
- C07K7/06—Linear peptides containing only normal peptide links having 5 to 11 amino acids
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K39/0005—Vertebrate antigens
- A61K39/0011—Cancer antigens
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/63—Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
-
- C—CHEMISTRY; METALLURGY
- C12—BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
- C12N—MICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
- C12N15/00—Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
- C12N15/09—Recombinant DNA-technology
- C12N15/87—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation
- C12N15/88—Introduction of foreign genetic material using processes not otherwise provided for, e.g. co-transformation using microencapsulation, e.g. using amphiphile liposome vesicle
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55555—Liposomes; Vesicles, e.g. nanoparticles; Spheres, e.g. nanospheres; Polymers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K39/00—Medicinal preparations containing antigens or antibodies
- A61K2039/555—Medicinal preparations containing antigens or antibodies characterised by a specific combination antigen/adjuvant
- A61K2039/55511—Organic adjuvants
- A61K2039/55561—CpG containing adjuvants; Oligonucleotide containing adjuvants
Definitions
- the present invention relates to an immunodominant epitope peptide of fibroblast activation protein and uses thereof, and more specifically, to an MHC I-restricted immunodominant epitope peptide of the fibroblast activation protein and uses thereof for the prevention or treatment of fibroblast activation protein alpha (FAP)-related diseases such as tumors, nonalcoholic steatohepatitis (NASH) or fibrosis, and uses thereof for inhibiting cancer metastasis.
- FAP fibroblast activation protein alpha
- neoantigen-based cancer vaccines induce patient-specific antitumor immunity and have shown good results in various clinical trials (Nature 2018, 555 (7696), 402-402; Nat Med 2021, 27 (3), 515 ⁇ ).
- neoantigen-based cancer vaccines are personalized vaccines, they require a lot of cost and time from neoantigen prediction for each patient to vaccine manufacturing, which increases the burden on patients. Therefore, the need for the development of a pan-cancer vaccine that can be applied to various types of cancer is emerging.
- the tumor microenvironment is composed of immune cells, cancer-associated fibroblasts (CAFs), signaling molecules, blood vessels, and the extracellular matrix (ECM), and acts as a powerful barrier that limits the effectiveness of conventional cancer treatments, such as chemotherapy and immunotherapy (Cell Commun Signal 2020, 18 (1), 59).
- cancer-associated fibroblasts present in the TME play a pivotal role in forming the tumor stroma by secreting extracellular matrix proteins (ECMs) such as collagen, laminin, and fibronectin (Nat Rev Immunol 2015, 15 (11), 669-682; Nat Rev Cancer 2016, 16 (9), 582-598; Nat Rev Clin Oncol 2021, 18 (12), 792-804).
- ECMs extracellular matrix proteins
- ECM proteins provide a structural network for tumor growth, mediate signaling and interactions between cells, and form a physical barrier between the tumor and the surrounding environment (Signal Transduct Target Ther 2021, 6 (1), 153).
- desmoplastic tumors such as pancreatic adenocarcinoma (PDAC) and colorectal cancer has been reported to be a major cause of the poor survival rate of patients with such tumors by strongly limiting the therapeutic effect (Proc Natl Acad Sci U S A 2019, 116 (22), 10674-10680; ACS Nano 2019, 13 (10), 11008-11021).
- PDAC pancreatic adenocarcinoma
- numerous attempts have been performed to inhibit or deplete CAFs in the TME.
- Fibroblast Activation Protein alpha is a transmembrane protein that is highly expressed in CAFs but at very low levels in normal adult tissues, and has been reported as a targeting marker for CAFs (Science 2010, 330 (6005), 827-830).
- FAP-expressing CAFs therapeutic approaches that inhibit the function of FAP-expressing CAFs, such as small molecule FAP inhibitors (J Clin Invest 2009, 119 (12), 3613-3625), FAP-specific antibodies and drug conjugates (Clin Cancer Res 2020, 26 (13), 3420-3430), and FAP-targeting CAR-T therapy (Cancer Immunol Res 2014, 2 (2), 154-166), have been developed and demonstrated that they promote specific types of tumor regression in vivo, suggesting the potential of this FAP therapeutic strategy (Front Biosci-Landmrk 2018, 23, 1933-1968).
- FAP-targeting CAR-T therapy Facer Immunol Res 2014, 2 (2), 154-166
- non-alcoholic steatohepatitis is a chronic liver disease characterized by hepatic steatosis and inflammation, and is a serious disease that can progress to fibrosis, cirrhosis, and hepatocellular carcinoma (Hepatology 77, 1335-1347(2023)).
- FAP is expressed in hepatocyte stellate cells and other fibroblasts in liver tissue (Nat Commun 13 (2022)) and is reported to cleave FGF21, a major regulator of lipids. Dysregulation of FGF21 by FAP activity is reported to contribute to the progression of NASH, but no NASH vaccine targeting FAP has been reported.
- the inventors of the present invention have made diligent efforts to develop a vaccine that exhibits excellent immune-inducing effects by inducing a strong fibroblast-activating protein (FAP)-specific T cell immune response, and as a result, have derived an immunodominant epitope peptide that exhibits excellent immune-inducing effects against FAP, and have confirmed that the immunodominant epitope peptide can effectively treat FAP-related diseases such as cancer, NASH, etc., thereby completing the present invention.
- FAP fibroblast-activating protein
- the purpose of the present invention is to provide an immunodominant epitope peptide exhibiting an excellent T cell inducing effect against fibroblast activation protein alpha (FAP) and its use for the prevention or treatment of diseases related to fibroblast activation protein alpha (FAP).
- FAP fibroblast activation protein alpha
- the purpose of the present invention is to provide a preparation that specifically binds to the above peptide and a use thereof.
- Another object of the present invention is to provide a nucleic acid encoding the peptide.
- Another object of the present invention is to provide a recombinant vector comprising the nucleic acid.
- Another object of the present invention is to provide a host cell into which the nucleic acid or recombinant vector has been introduced.
- the present invention provides a peptide which is an MHC I-restricted epitope peptide of fibroblast activation protein (FAP), wherein the peptide is composed of 8 to 10 amino acids.
- FAP fibroblast activation protein
- the present invention also provides a nanoparticle comprising the peptide.
- the present invention also provides a vaccine composition for preventing or treating a disease related to fibroblast activation protein alpha (FAP), comprising the peptide and/or nanoparticle as an active ingredient.
- FAP fibroblast activation protein alpha
- the present invention also provides a vaccination, prevention and/or treatment method for the prevention or treatment of a fibroblast activation protein alpha (FAP)-related disease comprising the step of administering to a subject the peptide and/or nanoparticle.
- a vaccination, prevention and/or treatment method for the prevention or treatment of a fibroblast activation protein alpha (FAP)-related disease comprising the step of administering to a subject the peptide and/or nanoparticle.
- the present invention also provides a use of the peptide and/or nanoparticle for the manufacture of a vaccine composition for the prevention or treatment of a disease associated with fibroblast activating protein alpha (FAP).
- FAP fibroblast activating protein alpha
- the present invention also provides a pharmaceutical composition for preventing or treating a disease associated with fibroblast activating protein alpha (FAP), comprising the peptide and/or lipid nanoparticle.
- FAP fibroblast activating protein alpha
- the present invention also provides a nucleic acid encoding the peptide.
- the present invention also provides a recombinant vector containing the nucleic acid.
- the present invention also provides a host cell into which the nucleic acid or recombinant vector has been introduced.
- the present invention also provides a method for producing a peptide, comprising a step of culturing the host cell.
- Figure 1 schematically illustrates the development process of a FAP immunodominant epitope peptide in an embodiment of the present invention.
- Figure 2 illustrates the gating method of flow cytometry used in ICS analysis.
- Figure 3 shows the upper epitope peptide sequences of FAP predicted by each program (netMHC, PREDEP, BIMAS).
- Figure 4 shows the immunogenicity and antitumor efficacy of the peptide candidates screened in vivo.
- Figure 4(a) summarizes the predicted FAP peptides presented by MHC Class I.
- Figure 4(c) and (d) show the representative flow cytometry results of IFN- ⁇ -secreting CD8+ T cells (c) and the percentage of CD8+ T cells producing IFN- ⁇ (d). The statistical significance between the control group and each experimental group was determined by two-tailed Student's t-test.
- Figure 4(e) and (f) show representative images of IFN- ⁇ -producing spots (e) and the average number of IFN- ⁇ spot-forming cells (f) determined by ELISpot analysis. The statistical significance between the control group and each experimental group was determined by two-tailed Student's t-test.
- Figure 4(g) shows the immunization schedule to evaluate the antitumor therapeutic efficacy of the predicted peptides.
- n 4 mice/group.
- Figure 4(h) shows the mean tumor growth curves of E.G7-OVA cells (left) and tumor volumes on day 25 (right).
- the statistical significance of the differences between the control group and each experimental group was determined by two-tailed Student's t-test. All data are expressed as mean ⁇ SEM. (*P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001).
- Figure 5 shows the ICS analysis results of the six screened peptides individually.
- Each panel represents: Control (unadministered); peptide administration; peptide and CFA co-administration
- the upper panel of each figure shows the results of no ex-vivo restimulation, and the lower panel shows the results of ex-vivo restimulation with peptides.
- Figure 6 shows the results of ELISpot analysis performed in the same manner as the ICS analysis.
- Figure 7 shows the ratio of FAP+CAF according to tumor volume.
- Figure 8 shows the design and antitumor efficacy of FAPPEP-SLNP nanovaccine in an established E.G7-OVA tumor model.
- Figure 8(a) shows the schematic diagram of FAPPEP-SLNP nanovaccine and its expected mechanism of action after internalization into APC.
- Figure 8(b) shows the hydrodynamic diameter of FAPPEP-SLNP measured by DLS.
- Figure 3(d) shows the immunization schedule to evaluate the antitumor efficacy of FAPPEP-SLNP nanovaccine against an established E.G7-OVA tumor model.
- Figure 9 schematically illustrates the structure of the DSPE-PEG2000-FAPPAP conjugate, one of the components of SLNP.
- Figure 10 shows the results of confirming the synthesis of DSPE-PEG2000-FAPPEP using HPLC (a, b) and MALDI-TOF (c, d).
- Figure 11 shows the results of ICS analysis confirming that cysteine (Cys) introduced to conjugate FAPPEP to DSPE-PEG2000-PDP does not affect immunogenicity.
- Figure 12 shows the results of ICS analysis performed on the spleen and lymph nodes of mice immunized with FAPPEP1-SLNP, confirming the presence of CD8+ T cells specific for FAPPEP1.
- Figure 13 shows the results of a tetramer assay using FAPPEP1-H-2Kb.
- Figure 14 shows the results confirming that FAPPEP-SLNP nanovaccine increases antigen-specific T cell immunity without inducing autoimmunity by mediating TME remodeling.
- Figure 14(a) shows the immunization schedule for tumor growth, mouse survival, tumor tissue, and spleen analysis.
- Figure 15 shows the results of confirming the distribution of blood vessels in a tumor (a and b) and the results of confirming metastasis in the lung (c and d).
- blood vessels are shown in brown
- metastatic cells are shown in red.
- Figure 16 shows the analysis of the ratio of FAP+ CAF among the cells forming the tumor by flow cytometry analysis of single cells dissociated from various types of tumors (E.G7-OVA, Panc02, MC38).
- Figure 17 shows the results of flow cytometry analysis to confirm whether FAP is expressed in cancer cells other than CAFs in MC38 tumors.
- Figure 18 is an experiment to confirm whether FAPPEP1-SLNP monotherapy shows therapeutic efficacy in Panc02 and MC38 tumors, which are connective tissue tumors.
- Figure 19 shows the results of an ICS assay performed to confirm whether CD8+ T cells (a) and CD4+ T cells (b) within the tumor in the MC38 tumor model have immune activity specific to FAP.
- Figure 20 shows the results confirming whether FAPPEP1-SLNP exhibits therapeutic efficacy even in large-sized tumors.
- Figure 21 shows the killing effect of ECM-rich MC38 tumors after combination treatment with FAPPEP1-SLNP nanovaccine and Dox.
- (a) Penetration of near-infrared dye cypate into MC38 tumors after FAPPEP1-SLNP immunization. Mice were immunized with FAPPEP1-SLNP nanovaccine and administered near-infrared dye cypate as a proxy for small molecule Dox. Penetration into tumors of different groups was compared using IVIS (n 5 mice/group). The left panels show representative images of intratumoral dye penetration and relative fluorescence signal intensity of the dye distributed in the tumors acquired by IVIS.
- Figure 21(c) shows the MC38 tumor growth in mice (left) and tumor volume on day 17 (right). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test.
- Figure 22 schematically illustrates the mechanism by which FAPPEP1-SLNP nanovaccine targets CAFs to kill tumors.
- Figure 23 schematically illustrates the mechanism of a tumor metastasis inhibition model targeting CAF of FAPPEP1 peptide vaccine or FAPPEP1-SLNP nanovaccine (top) and the therapeutic efficacy evaluation model of FAPPEP1 peptide vaccine on cancer metastasis used in the examples of the present invention (bottom).
- Figure 24 is a histological analysis of lung tissue showing the results of inhibiting cancer metastasis to the lung by the FAPPEP1 peptide vaccine.
- Figure 25 is a histological analysis of liver tissue showing the results of FAPPEP1 peptide vaccine inhibition of cancer metastasis to the liver.
- Figure 26 schematically illustrates the NASH treatment mechanism through targeting FAP expressing cells and preventing FGF21 degradation by FAPPEP1 peptide vaccine or FAPPEP1-SLNP nanovaccine.
- Figure 27 shows the results showing the therapeutic efficacy of the FAPPEP1 peptide vaccine for the NASH model.
- Figure 27 schematically shows the experimental method of the NASH establishment and therapeutic effect confirmation model performed in the example of the present invention.
- n 5 mice/group.
- Figure 27b shows the body weight monitoring of the mice (left) and the body weight of the mice at 45 days (right).
- Figure 27c shows the liver weight of the mice at 45 days.
- Figure 27d shows the liver-to-body weight ratio at 45 days.
- Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as the mean ⁇ SEM. (*P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001). (Illustration created with BioRender.com.)
- Figure 28 shows the results of serum analysis of mice after immunization with FAPPEP1 peptide vaccine in NASH model.
- Mouse sera were collected on day 45 and analyzed for the levels of damage-related markers as follows: ALT (a), AST (b), total bilirubin (c), and HDL (d).
- ALT a
- AST b
- c total bilirubin
- HDL d
- Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ⁇ SEM. (*P ⁇ 0.05, **P ⁇ 0.01, ***P ⁇ 0.001).
- Figure 29 shows the results of evaluating lipid deposition in liver tissue using Oil red O staining after immunization with FAPPEP1 peptide vaccine.
- Figure 30 shows the results of evaluating ballooning hepatocytes in liver tissue using H&E staining.
- nucleic acids and amino acids are listed in 5' to 3' and N-terminal to C-terminal orientations, respectively, from left to right. Numerical ranges listed within the specification are inclusive of the numbers defining the range and include each integer or any non-integer fraction within the defined range.
- amino acid sequence referred to in the present invention is interpreted to include variants or fragments thereof in which amino acid residues are conservatively substituted at specific amino acid residue positions.
- “conservative substitution” means a modification that includes replacing one or more amino acids with amino acids having similar biochemical properties that do not result in loss of biological or biochemical function of the protein.
- a “conservative amino acid substitution” refers to the substitution of an amino acid residue with an amino acid residue having a similar side chain.
- classes of amino acid residues having similar side chains are well known in the art. These classes include amino acids having basic side chains (e.g., lysine, arginine, histidine), amino acids having acidic side chains (e.g., aspartic acid, glutamic acid), amino acids having uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), amino acids having non-polar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), amino acids having beta-branched side chains (e.g., threonine, valine, isoleucine), and amino acids having aromatic side chains (e.g., tyrosine, pheny
- Fibroblast Activation Protein alpha is also known as prolyl endopeptidase FAP.
- FAP Fibroblast Activation Protein alpha
- FAP-targeting therapies that can induce long-term immune responses have rarely been reported, and some reported FAP-targeting vaccine therapies also have systemic toxicity or insufficient efficacy, requiring the development of new FAP-targeting vaccine therapeutics.
- FAP fibroblast activation protein
- the present invention relates, in one aspect, to an immunodominant epitope peptide of fibroblast activation protein (FAP).
- FAP fibroblast activation protein
- immunodominance refers to an immunological phenomenon in which an immune response is induced only to a small number of antigenic peptides among various antigenic peptides that can be derived from a specific protein or polypeptide (Clinical Immunology. 143 (2): 99-115). “Immunodominance” is clearly observed in both antibody-mediated immunity and cell-mediated immunity. The effect of immunodominance is caused by immunodominance.
- the difference in immunogenicity of hundreds to thousands of peptide antigens that can be produced from a pathogen represents a dominance hierarchy, and an antigen that stimulates a strong immune response is considered an “immunodominant epitope” or “immunodominant antigen.”
- the peptide may be characterized as being composed of 7 to 11 amino acids, preferably 8 to 10 amino acids.
- the peptide may be characterized as being an MHC I-restricted epitope peptide of fibroblast activation protein (FAP).
- FAP fibroblast activation protein
- MHC Major histocompatibility complex
- MHC is a complex that presents antigens on the cell surface and induces an immune response to the epitopes presented by MHC.
- MHC can be largely classified into type I and type II.
- Type I MHC induces a CD8+ T cell response
- type II MHC induces an immune response by presenting it to B cells or helper T cells through APC.
- the peptide may be characterized as being a human leukocyte antigen (HLA) restricted epitope.
- HLA human leukocyte antigen
- HLA Human leukocyte antigen
- HLA Human leukocyte antigen
- HLA is a major gene locus of human MHC and refers to an antigenic site that distinguishes whether the MHC presented by a cell is self or non-self.
- HLA is classified into various families according to its location within the human chromosome. For example, HLA of MHC I is classified into HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA-G, etc., and HLA of MHC II can be classified into HLA-DP, HLA-DQ, HLA-DR, etc., and each family group can be classified in more detail, but is not limited thereto.
- the peptide may be an epitope peptide restricted to one or more types of HLA among HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, and HLA-G, and more preferably, may be an epitope peptide restricted to one or more types of HLA among HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8, and HLA-B27, but is not limited thereto.
- MHC restricted epitope or "HLA restricted epitope” of the present invention means that the antigen can induce an immune cell response when presented by a specific type of MHC or HLA.
- the restriction of the epitope peptide can be exclusive (restricted to only one type) or non-exclusive (restricted to more than one type) with respect to other types of MHC or HLA. Such restriction of the epitope peptide not only allows more effective immunization depending on the patient's HLA typing, but also prevents toxicity and side effects due to induction of unintended excessive immune response.
- the peptide may be characterized by including an amino acid sequence selected from the group consisting of sequence numbers 1 to 149 of Tables 1 to 5 below.
- the peptide may be characterized by being composed of an amino acid sequence selected from the group consisting of sequence numbers 1 to 149 of Tables 1 to 5 below.
- the peptide may be characterized as being an epitope peptide restricted to HLA-A1.
- the epitope peptide restricted to HLA-A1 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
- the peptide may be characterized as being an epitope peptide restricted to HLA-A2.
- the epitope peptide restricted to HLA-A2 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, 4, 5, and 30 to 45.
- the peptide may be characterized as being an epitope peptide restricted to HLA-A3.
- the epitope peptide restricted to HLA-A3 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
- the peptide may be characterized as being an epitope peptide restricted to HLA-A24.
- the epitope peptide restricted to HLA-A24 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
- the peptide may be characterized as being an epitope peptide restricted to HLA-A26.
- the epitope peptide restricted to HLA-A26 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97.
- the peptide may be characterized as being an epitope peptide restricted to HLA-B7.
- the epitope peptide restricted to HLA-B7 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
- the peptide may be characterized as being an epitope peptide restricted to HLA-B8.
- the epitope peptide restricted to HLA-B8 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129.
- the peptide may be characterized as being an epitope peptide restricted to HLA-B27.
- the epitope peptide restricted to HLA-B27 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149.
- the peptide may preferably comprise or consist of an amino acid sequence of SEQ ID NO: 1, 2, 3, 4 or 5, more preferably SEQ ID NO: 1.
- the peptide may be characterized by inducing an immune response specific to fibroblast activation protein (FAP).
- FAP fibroblast activation protein
- the peptide may be characterized by, but is not limited to, stimulating a cytotoxic T cell response or a CD8+ T cell response against free FAP or FAP expressing cells.
- the FAP expressing cell may be characterized as being a FAP expressing fibroblast, but is not limited thereto.
- the peptide may additionally independently include amino acids at the N'-terminus and/or the C'-terminus, respectively.
- the peptide may additionally include 1 to 100 amino acids, preferably 1 to 50 amino acids, more preferably 1 to 25 or 1 to 10 amino acids, respectively, at the N'-terminus and/or the C'-terminus, but is not limited thereto, and any amino acid may be additionally included within a range in which the activity of the peptide of the present invention as an immunodominant epitope is substantially maintained.
- the peptide may additionally include amino acids at the N'-terminus and/or the C'-terminus of the amino acid sequence of SEQ ID NOs: 1 to 149. It is preferable that an amino acid according to a sequence of a known fibroblast activation protein (FAP), preferably a sequence of human fibroblast activation protein, is added, but is not limited thereto.
- FAP fibroblast activation protein
- the peptide includes a fragment in which a part of the N'-terminal and/or C'-terminal sequence of the amino acid sequence is deleted.
- it may be a fragment in which 1 to 5 amino acids, preferably 1 to 3 amino acid sequences are independently deleted from the N'-terminal and/or C'-terminal of the amino acid sequence of SEQ ID NOs: 1 to 149, but is not limited thereto, and includes a fragment in which any amino acid is deleted within a range in which the activity as an immunodominant epitope of the peptide having the amino acid sequence of SEQ ID NOs: 1 to 9 is substantially maintained.
- the peptide may be characterized by being obtained through cleavage of a full-length fibroblast activation protein (FAP) obtained from nature or a fragment thereof, or an artificially synthesized oligopeptide, but is not limited thereto.
- FAP fibroblast activation protein
- the peptide of the present invention can be produced and used in the form of a fusion protein by being fused with various functional peptides known in the art.
- the fusion protein may be produced and used in the form of a fusion protein in which one or more peptides of the same sequence of the present invention are fused (homo type) and/or peptides of different sequences are fused (hetero type), but is not limited thereto.
- it may be a fusion protein in which one or more of the amino acid sequences of SEQ ID NOs: 1 to 149 are fused, but is not limited thereto.
- the fusion protein may further include an Fc domain.
- the Fc domain may include the last two constant region immunoglobulin domains of IgA, IgD and IgG and a flexible hinge N-terminus for these domains.
- the Fc domain may include a J chain.
- the Fc domain may include immunoglobulin domains C ⁇ 2 and C ⁇ 3 and a hinge between C ⁇ 1 and C ⁇ 2.
- the boundaries of the Fc domain can vary, but the human IgG heavy chain Fc region is generally defined to include residues C226 or P230 of the carboxyl terminus, where the numbering of amino acids uses the Kabat numbering.
- the terms “Fc,” “Fc domain,” and “Fc region” of the present invention may be used interchangeably, and the Fc domain may refer to the region in isolation, or as part of an antibody, a fragment thereof, or a fusion protein. Polymorphisms in the Fc domain have been reported at various positions and can be used as the fusion domain of the fusion protein of the present invention without limitation.
- the peptide of the present invention can be prepared and used in the form of a fusion protein additionally including a membrane penetrating domain for presentation on the surface of a cell or an artificial membrane.
- the peptide of the present invention can be prepared and used in the form of a fusion protein additionally including at least one of a hinge domain, a coiled-coil domain, an immune regulatory domain, and an intracellular signaling domain, but is not limited thereto.
- transmembrane domain refers to a protein domain that spans the membrane of a cell.
- the transmembrane domain preferably has an alpha-helical structure, but is not limited thereto.
- honey domain refers to a series of amino acid sequences that exist between the transmembrane domain and the extracellular domain of a membrane anchored protein.
- coiled coil domain refers to a structural motif of a protein in which 2 to 7 alpha helices are coiled like a rope strand.
- the coiled coil domain may be characterized by having 2 or 3 alpha helices coiled.
- the peptide of the present invention can be fused without limitation with various domains known in the art for extending half-life, improving pharmacokinetic properties, etc., and it is preferable that the domain does not reduce, inhibit, or mask the function of the peptide of the present invention as an immunodominant epitope.
- the peptide of the present invention can be used as a conjugate by being conjugated to a molecule other than a peptide.
- Methods for producing protein-conjugates known in the art can be used without limitation, and preferably, it can be conjugated to another drug or adjuvant through chemical conjugation, but is not limited thereto.
- the present invention relates to a nucleic acid encoding a peptide or fusion protein of the present invention.
- nucleic acids used herein may be present in cells, a cell lysate, or may be present in a partially purified or substantially pure form.
- a nucleic acid is "isolated” or “rendered substantially pure” when purified away from other cellular components or other contaminants, such as other cellular nucleic acids or proteins, by standard techniques including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art.
- a nucleic acid of the present invention may be, for example, DNA or RNA.
- the present invention relates to a recombinant vector containing the nucleic acid of the present invention.
- any vector known in the art can be appropriately selected and used without limitation.
- a vector containing a T7 series (T7A1, T7A2, T7A3, etc.), lac, lacUV5, temperature-dependent ( ⁇ phoA, phoB, rmB, tac, trc, trp or 1PL promoter)
- yeast when yeast is used as a host, a vector containing an ADH1, AOX1, GAL1, GAL10, PGK or TDH3 promoter can be used, and in the case of Bacillus, a vector containing a P2 promoter can be used.
- vector in the present invention means a DNA preparation containing a DNA sequence operably linked to a suitable regulatory sequence capable of expressing the DNA in a suitable host.
- the vector may be a plasmid, a phage particle, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or in some cases may be integrated into the genome itself. Since plasmids are currently the most commonly used form of vector, the terms “plasmid” and “vector” are sometimes used interchangeably in the present description. However, the present invention includes other forms of vectors that have equivalent functions as known or become known in the art. Protein expression vectors used in E.
- coli include the pET series from Novagen (USA); the pBAD series from Invitrogen (USA); the pHCE or pCOLD series from Takara (Japan); the pACE series from Xenofocus (Korea); and the like.
- Bacillus subtilis protein expression can be achieved by inserting the target gene into a specific part of the genome, or by using vectors such as the pHT series from MoBiTech (Germany). Protein expression is also possible in molds and yeast by using genome insertion or self-replicating vectors.
- Plant protein expression vectors can be used using the T-DNA system of Agrobacterium tumefaciens or Agrobacterium rhizogenes. Typical expression vectors for expression in mammalian cell cultures are based on, for example, pRK5 (EP 307,247), pSV16B (WO 91/08291), and pVL1392 (Pharmingen).
- control sequence means a DNA sequence essential for the expression of an operably linked coding sequence in a particular host organism.
- control sequences include a promoter for initiating transcription, an optional operator sequence for regulating such transcription, a sequence encoding a suitable mRNA ribosome binding site, and a sequence for regulating the termination of transcription and translation.
- a control sequence suitable for a prokaryote includes a promoter, optionally an operator sequence, and a ribosome binding site.
- this includes a promoter, a polyadenylation signal, and an enhancer.
- the factor that most influences the amount of gene expression in a plasmid is the promoter.
- promoters for high expression the SR ⁇ promoter and a cytomegalovirus-derived promoter are preferably used.
- any of a wide variety of expression control sequences may be used in the vector to express the DNA sequence of the present invention.
- useful expression control sequences include, in addition to the promoters described above, the early and late promoters of SV40 or adenovirus, the lac system, the trp system, the TAC or TRC system, the T3 and T7 promoters, the major operator and promoter region of phage lambda, the regulatory region of the fd encoded protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of the phosphatases, e.g.
- Pho5 the promoter of the yeast alpha-mating system, and any other sequence of structure and inducibility known to control expression of genes in prokaryotes or eukaryotes or their viruses, and any combination thereof.
- the T7 RNA polymerase promoter ⁇ can be usefully used to express proteins in E. coli.
- a nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence.
- This can be a gene and regulatory sequence(s) that are linked in such a way that an appropriate molecule (e.g., a transcriptional activating protein) can cause gene expression when bound to the regulatory sequence(s).
- DNA for a pre-sequence or a secretory leader is operably linked to DNA for a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation.
- "operably linked" means that the linked DNA sequences are in contact, and in the case of a secretory leader, are in contact and are in reading frame. However, an enhancer need not be in contact. The connection of these sequences is carried out by ligation at convenient restriction enzyme sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers according to conventional methods are used.
- heterologous DNA refers to heterologous DNA, which is DNA that is not naturally found in the host cell. Once inside the host cell, the expression vector can replicate independently of the host chromosomal DNA, and multiple copies of the vector and its inserted (heterologous) DNA can be produced.
- the gene in order to increase the level of expression of a transfected gene in a host cell, the gene must be operably linked to transcriptional and translational expression control sequences that are functional in the selected expression host.
- the expression control sequences and the gene are contained in a single expression vector that also contains a bacterial selection marker and a replication origin. If the expression host is a eukaryotic cell, the expression vector may further contain expression markers that are useful in the eukaryotic expression host.
- the present invention relates to a nucleic acid encoding a peptide of the present invention; or a host cell into which the recombinant vector has been introduced.
- the host cell refers to an expression cell into which a gene or a recombinant vector, etc. has been introduced to produce a protein, etc.
- the host cell may be used without limitation as long as it is a cell capable of expressing the peptide of the present invention, and is preferably a eukaryotic cell, more preferably a yeast, an insect cell, an animal cell, and most preferably an animal cell.
- a CHO cell line or a HEK cell line which are mainly used for the expression of peptides, may be used, but is not limited thereto.
- Suitable expression vectors for eukaryotic hosts include, for example, expression control sequences derived from SV40, bovine papillomavirus, adenovirus, adeno-associated virus, cytomegalovirus and retroviruses.
- Expression vectors for use in bacterial hosts include, for example, bacterial plasmids obtained from E.
- coli such as pBluescript, pGEX2T, pUCvector, col E1, pCR1, pBR322, pMB9 and derivatives thereof, plasmids having a wider host range, such as RP4, phage DNA, such as a wide variety of phage lambda derivatives, such as ⁇ and ⁇ NM989, and other DNA phages, such as M13 and filamentous single-stranded DNA phages.
- Useful expression vectors for yeast cells are the 2 ⁇ plasmids and derivatives thereof.
- a useful vector for insect cells is pVL 941.
- the above recombinant vector can be introduced into a host cell by a method such as transformation or transfection.
- transformation means that DNA is introduced into a host so that the DNA becomes replicable as an extrachromosomal element or by chromosomal integration.
- transfection means that an expression vector is accepted by a host cell, regardless of whether any coding sequence is actually expressed.
- an expression control sequence several factors should also be considered, such as the relative strength of the sequences, their controllability, and their compatibility with the DNA sequences of the present invention, particularly with respect to possible secondary structures.
- the unicellular host should be selected by considering factors such as the selected vector, the toxicity of the product encoded by the DNA sequence of the present invention, secretion characteristics, the ability to accurately fold the protein, culture and fermentation requirements, and the ease of purifying the product encoded by the DNA sequence of the present invention from the host.
- factors such as the selected vector, the toxicity of the product encoded by the DNA sequence of the present invention, secretion characteristics, the ability to accurately fold the protein, culture and fermentation requirements, and the ease of purifying the product encoded by the DNA sequence of the present invention from the host.
- those skilled in the art can select various vector/expression control sequence/host combinations that can express the DNA sequence of the present invention in fermentation or large-scale animal culture.
- the binding method, the panning method, the film emulsion method, etc. can be applied.
- the above gene and recombinant vector can be introduced into a host cell through various methods known in the art.
- a gene encoding a nucleic acid encoding the peptide of the present invention can be directly introduced into the genome of a host cell and exist as a chromosomal element. It will be obvious to those skilled in the art that inserting the gene into the genomic chromosome of a host cell will have the same effect as introducing a recombinant vector into the host cell.
- the present invention relates to a method for producing a peptide, which comprises a step of culturing the host cell.
- the peptide can be produced by culturing the host cell for a period of time sufficient to express the peptide in the host cell or for a period of time sufficient to secrete the peptide into the culture medium in which the host cell is cultured.
- the expressed peptide can be obtained by separating from the host cell and purifying it to be homogeneous.
- the separation or purification of the peptide can be performed by a separation or purification method used for general proteins, for example, chromatography.
- the chromatography can be, for example, a combination of one or more selected from affinity chromatography, ion exchange chromatography, or hydrophobic chromatography, but is not limited thereto.
- filtration, ultrafiltration, salting out, dialysis, etc. can be additionally used in combination.
- the peptide of the present invention may be presented as a peptide itself, but is not limited thereto, and may be presented attached to the surface of or included within a delivery vehicle for various purposes such as effective delivery, targeting, and improved pharmacokinetic properties.
- peptide-based vaccines have several advantages, including cost-effective manufacturing, relatively easy quality control, and good safety profiles, but they exhibit low immunogenicity, require strong adjuvants, and have limited therapeutic efficacy due to low delivery efficiency to antigen-presenting cells (APCs) in vivo.
- APCs antigen-presenting cells
- a vaccine platform based on neutrally-charged small lipid nanoparticles was used to prepare lipid nanoparticles containing the peptide, and it was confirmed that when immunization was performed with the lipid nanoparticles, the number and activity of FAP-specific CD8+ T cells were significantly increased.
- the present invention from another aspect, relates to nanoparticles comprising the peptide.
- nanoparticle of the present invention is defined as a particle having a nanometer size, and each particle may have different physicochemical, biological, and immunogenic characteristics depending on the shape, size, components, surface characteristics, functional groups, etc.
- the nanoparticle may be characterized by including one or more types of the peptide.
- the nanoparticle may be characterized by including the peptide of the present invention including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more types of sequences.
- the nanoparticle may include any one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 1 to 149.
- the one or more peptides included in the vaccine composition of the present invention may be in various forms, such as an epitope peptide, a fusion protein including the same, a nanoparticle, etc., as described above.
- Nanoparticle platforms for delivering peptides are well known in the art.
- the nanoparticle may encapsulate a peptide and contain it inside, or may present the protein on the surface of the nanoparticle, but is not limited thereto.
- the nanoparticle may be selected from the group consisting of virus-like particles (VLPs), protein nanostructures, polymer nanoparticles, lipid nanoparticles, and inorganic nanoparticles, but is not limited thereto.
- VLPs virus-like particles
- protein nanostructures protein nanostructures
- polymer nanoparticles polymer nanoparticles
- lipid nanoparticles lipid nanoparticles
- inorganic nanoparticles but is not limited thereto.
- virus-like particle refers to a nanoparticle in the range of several to several hundred nanometers composed of self-assembled viral envelope or capsid proteins from which infectious elements such as genomes have been removed.
- Virus-like particles are the first nanoparticles for the in vivo delivery of peptide epitopes and are the delivery form for various clinically tested and commercially approved vaccines.
- the polymer nanoparticle may be a polymer-based nanoparticle selected from the group consisting of, for example, HPMA (N-(2-hydroxypropyl)-methacrylamide copolymer), SMA (polystyrene-maleic anhydride copolymer), PEG (polyethylene glycol), and PGA (poly-L-glutamic acid), but is not limited thereto.
- HPMA N-(2-hydroxypropyl)-methacrylamide copolymer
- SMA polystyrene-maleic anhydride copolymer
- PEG polyethylene glycol
- PGA poly-L-glutamic acid
- the inorganic nanoparticles include, but are not limited to, silica nanoparticles, gold nanoparticles, self-associating nanoparticles, etc.
- the protein nanostructure refers to a nanoparticle formed by self-assembly of a protein.
- the protein nanostructure is known to be a platform based on proteins such as, for example, ferritin, lumazine synrhase (LS), dihydrolipoyl acetyltransferase (E2p) nsp10 (nonstructural protein 10), but is not limited thereto.
- the nanoparticle is a lipid nanoparticle.
- the lipid nanoparticle may be selected from the group consisting of a nanodisk, a unilamellar vesicle, a multilamellar vesicle (MLV), a multivesicular vesicle (MV), a liposome, a LNP, an emulsion, and a lipopolyplex (LPP), but is not limited thereto.
- a nanodisk a unilamellar vesicle
- MMV multilamellar vesicle
- MV multivesicular vesicle
- LNP liposome
- LNP lipopolyplex
- the lipid nanoparticle may be characterized by further including at least one selected from the group consisting of a phospholipid, a cationic lipid, and an adjuvant.
- the phospholipid may be characterized by having an aliphatic carbon number of 10 to 30, preferably 12 to 25, and most preferably 14 to 22, but is not limited thereto.
- the phospholipid may be a DSPE-PEG derivative including 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPEPEG1000), a functionalized DSPE-PEG derivative including 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] (DSPE-PEG2000-PDP) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamineN-[maleimide(polyethylene glycol)-2000] (DSPE-PEG2000-Maleimide), and 1,2-d
- Fluorescently labeled phospholipids including 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl) (DPPE-Rhodamine), 1,2-Didecanoyl-sn-glycero-3-phosphocholine (DDPC), 1,2-Dierucoyl-sn-glycero-3-phosphate (DEPA), glycero-3-phosphocholine (DEPC), 1,2-Dierucoyl-sn-glycero-3-phosphoethanolamine (DEPE), 2-Dierucoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DEPG), 1,2-Dilinoleoyl-sn-glycero-3-phosphocholine
- DDPC 1,2-Didecanoyl-sn-glycero-3-phosphocholine
- DEPA 1,2-Dierucoyl-sn-glycero
- DLOPC 1,2-Dilauroyl-sn-glycero-3-phosphate
- DLPE 1,2-Dilauroyl-sn-glycero-3-phosphocholine
- DLPS 1,2-Dilauroyl-sn-glycero-3-phosphoserine
- DMPA 1,2-Dimyristoyl-sn-glycero-3-phosphate
- DMPC 1,2-Dimyristoyl-sn-glycero-3-phosphocholine
- DMPE 1,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine
- DMPG 1,2-Dimyristoyl-snglycero-3-Phospho-rac-(1-glycerol)
- DMPS 1,2-Dimyristoyl-sn-glycero-3-phosphoserine
- the cationic lipid is Dimethyldioctadecyl-ammoniumbromide (DDAB), Dimethyldioctadecylammonium (DDAB), (N,N-dimethyl-N-([2-sperminecarboxamido]ethyl)-2,3-bis(dioleyloxy)-1 -propaniminium pentahydrochloride) (DOSPA), (N-[1-(2,3-dioleyloxy)propyl]-N,N,Ntrimethylammonium) (DOTMA), (N-[1-(2,3-dioleoyloxy)propyl]- N,N,N-trimethylammonium) (DOTAP), 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol), N4-Cholesteryl-Spermine (GL67), 1,2-dioleyloxy -3-
- DOSPA
- the cationic lipid may preferably be a cationic cholesterol derivative, more preferably Monoarginine-cholesterol (MA-Chol).
- MA-Chol Monoarginine-cholesterol
- the adjuvant may be, but is not limited to, an immunostimulatory single-chain or double-chain oligonucleotide, poly(I:C), an immunostimulatory small-molecule compound, or a combination thereof.
- immunostimulatory single- or double-stranded oligonucleotides are known as useful adjuvants. They often contain a CpG motif (a dinucleotide sequence comprising an unmethylated cytosine linked to a guanosine). Oligonucleotides containing a TpG motif, a palindrome sequence, a plurality of consecutive thymidine nucleotides (e.g., TTTT), a plurality of consecutive cytosine nucleotides (e.g., CCCC) or a poly(dG) sequence are also known adjuvants, as are double-stranded RNAs. Any of these various immunostimulatory oligonucleotides can be used without limitation in conjunction with the present invention.
- the oligonucleotide may be characterized by having a length of, for example, 10 to 100 nucleotides, for example, 15 to 50 nucleotides, 20 to 30 nucleotides, or 25 to 28 nucleotides, but is not limited thereto.
- the oligonucleotide may be composed of a natural nucleotide or a non-natural nucleotide, or may be composed of a mixture of these.
- the oligonucleotide may be characterized by containing one or more phosphorothioate linkages and/or being modified with one or more 2'-O-methyl.
- the single-chain or double-chain oligonucleotide may be characterized as being a CpG oligonucleotide, a STING activating oligonucleotide, or a combination thereof.
- STING Stimulator of Interferon Genes
- CpG oligonucleotide (CpG oligodeoxynucleotide, or CpG oligodeoxynucleotide, CpG ODN) of the present invention is a short single-stranded synthetic DNA molecule containing unmethylated cytosine triphosphate deoxynucleotide ("C") and guanine triphosphate deoxynucleotide ("G"), which is known as an immunostimulant.
- C cytosine triphosphate deoxynucleotide
- G guanine triphosphate deoxynucleotide
- the CpG is included as a component of the nano vaccine of the present invention, it can function as an adjuvant that enhances the immune response of dendritic cells.
- the CpG oligonucleotide of SEQ ID NO: 151 was used, but is not limited thereto.
- the immunostimulatory small molecule compound is used interchangeably with a small molecule adjuvant, and includes a synthetic small molecule adjuvant and a natural small molecule adjuvant.
- the immunostimulatory small molecule compound or small molecule adjuvant include, but are not limited to, monophosphoryl lipid A, Muramyl dipeptide, Bryostatin-1, Mannide monooleate (Montanide ISA 720), Squalene, QS21, Bis-(3',5')-cyclic dimeric guanosine monophosphate, PAM2CSK4, PAM3CSK4, Imiquimod, Resiquimod, Gardiquimod, cl075, cl097, Levamisole, 48/80, Bupivacaine, Isatoribine, Bestatin, Sm360320, and Loxoribine.
- Small molecule adjuvants are described in Flower DR et al. (Expert Opin Drug Discov. 2012 Sep;7(9):807
- the nanoparticle may include an anionic drug in addition to an adjuvant.
- the anionic drug may be, but is not limited to, an oligonucleotide, an aptamer, mRNA, siRNA, miRNA, or a combination thereof.
- the peptides may include various forms, formulations or modifications well known in the field of peptide therapeutics in addition to the lipid nanoparticles, fusion proteins or conjugates.
- the peptide of the present invention is an epitope peptide of FAP having excellent immunodominance, and when the peptide of the present invention is administered, an immune response specific to FAP can be induced.
- Fibroblast activation protein alpha is a type II transmembrane serine protease exclusively expressed in the pathologic states of various non-neoplastic or neoplastic diseases, including fibrosis, arthritis, and tumors or cancer (Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803). It is reported to be expressed in activated stromal fibroblasts, also called activated cancer-associated fibroblasts (CAFs), in more than 90% of all human carcinomas (Oncogene. 37 (32) (August 2018): 4343-4357; rontiers in Bioscience. 23: 1933-1968 (June 2018)).
- activated stromal fibroblasts also called activated cancer-associated fibroblasts (CAFs)
- cancer-associated fibroblasts play an important role in the development, growth, and metastasis of cancer, and in NASH, FAP cleaves FGF21, inducing lipid accumulation and causing various diseases such as hepatic steatosis and nonalcoholic steatohepatitis (NASH).
- NASH nonalcoholic steatohepatitis
- lipid nanoparticles including the peptide were manufactured, and it was confirmed that when immunization is performed with the lipid nanoparticles, tumor growth can be effectively inhibited.
- cancer-associated fibroblasts can be effectively removed from the tumor microenvironment, the formation of ECM can be reduced, and CD8+ T cell responses and antigen-specific CD4+ T cell responses against tumor cells can be induced.
- the vaccination therapy based on the peptide of the present invention can exhibit excellent anticancer effects compared to previously reported FAP-targeting cancer vaccines without causing side effects such as induction of systemic toxicity and autoimmune response by not inducing an increase in Th17 cells related to autoimmunity. Furthermore, in another embodiment of the present invention, it was confirmed that it can prevent and inhibit cancer metastasis as well as inhibit and treat cancer growth.
- a vaccination therapy based on the peptide of the present invention exhibited an excellent therapeutic effect on nonalcoholic steatohepatitis (NASH), another FAP-related disease.
- NASH nonalcoholic steatohepatitis
- the excellent immunodominance of the peptide of the present invention can be usefully used for the prevention or treatment of FAP-associated diseases such as fibrosis, arthritis, non-alcoholic steatohepatitis, and tumors/cancer through the induction of a strong immune response to FAP.
- FAP-associated diseases such as fibrosis, arthritis, non-alcoholic steatohepatitis, and tumors/cancer through the induction of a strong immune response to FAP.
- the present invention in another aspect, relates to a vaccine composition for preventing or treating a disease associated with fibroblast activation protein alpha (FAP), comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
- FAP fibroblast activation protein alpha
- FAP-related disease in the present invention is used to collectively refer to diseases in which the expression of FAP is the cause or in which excessive expression of FAP is associated with pathological characteristics.
- FAP-related diseases include, but are not limited to, fibrosis, arthritis, tumors, nonalcoholic steatohepatitis, hepatic steatosis, arteriosclerosis, and myocardial infarction (Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803).
- the present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating cancer or a tumor, comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
- the present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating metastasis of cancer or tumor, comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
- the present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating nonalcoholic steatohepatitis (NASH) comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
- NASH nonalcoholic steatohepatitis
- the vaccine composition may be characterized by comprising one or more peptides of the present invention.
- the vaccine composition may be characterized by comprising a peptide of the present invention comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more kinds of sequences.
- the vaccine composition may comprise any one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 1 to 149.
- the one or more peptides included in the vaccine composition of the present invention may be in various forms, such as an epitope peptide, a fusion protein including the same, a nanoparticle, etc., as described above.
- the vaccine composition may be characterized by containing different peptides or a combination thereof depending on the HLA type of the subject to be administered.
- the HLA type of the subject to be administered can be performed using a genetic analysis method known in the art.
- the vaccine composition when the subject has MHC of the HLA-A1 type, may include one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
- the vaccine composition when the subject has MHC of the HLA-A2 type, may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of sequence numbers 1, 3, 4, 5, and 30 to 45.
- the vaccine composition when the subject has MHC of the HLA-A3 type, may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
- the vaccine composition when the subject has MHC of the HLA-A24 type, may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
- the vaccine composition when the subject has MHC of the HLA-A26 type, may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97 as a restricted epitope peptide.
- the vaccine composition when the subject has MHC of the HLA-B7 type, may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
- the subject when it has MHC of the HLA-B8 type, it may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129.
- the subject when the subject possesses MHC of the HLA-B27 type, it may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149.
- the vaccine composition according to the above HLA type is one embodiment, and the vaccine composition of the present invention is not limited thereto, and may include one or more different HLA type-restricted peptides.
- vaccine composition of the present invention means a composition containing a substance capable of inducing an immune response by acting as an antigen or immunogen in vivo or in vitro, and in the present invention, can be used interchangeably with “vaccine” or “immunogenic composition” in the same meaning.
- immune response in the present invention is a broad concept that includes both innate immune responses and adaptive immune responses, for example, complement-mediated immune responses, cell-mediated (T cell) immune responses, and/or antibody-mediated (B cell) responses.
- the vaccine composition of the present invention can induce or increase an immune response to FAP or FAP-expressing fibroblasts (CAFs) in a subject of administration, and thereby exhibit excellent preventive or therapeutic effects on FAP-related diseases such as fibrosis, arthritis, tumors, and NASH. More specific examples include, but are not limited to, effects of deficiency of FAP-expressing CAFs in a tumor microenvironment, inhibition of ECM formation, induction of CD8+ T cell and CD4+ T cell responses, inhibition of metastasis, and enhancement of tumor penetration ability of immune cells and anticancer agents.
- the vaccine composition may contain the peptide, nucleic acid, or nanoparticle of the present invention as an active ingredient, and when containing the peptide or nucleic acid, various means known in the art for effectively presenting and delivering it may be used.
- the nanoparticle may be understood as an example of a carrier for presenting the peptide or nucleic acid.
- the peptide may be included in the vaccine composition in various forms other than the form of the nanoparticle described above.
- it may be included in the form of a fusion protein or a conjugate, and for another example, it may be presented by being fixed to the surface of a biological membrane such as an exosome or an artificial membrane such as an oligomembrane, or presented by being encapsulated inside, but is not limited thereto.
- the peptide of the present invention may be delivered in vivo by being included in a vaccine composition in the form of a nucleic acid encoding the peptide of the present invention or a delivery vector including the same, and the delivered nucleic acid or vector may be expressed in vivo to synthesize and present the peptide of the present invention in vivo.
- various in vivo protein expression platforms for delivery of the nucleic acid can be used, including but not limited to viral vectors, naked DNA or RNA.
- the nucleic acid may be contained in a non-viral vector such as a viral vector or an expression plasmid and may be included in the vaccine composition of the present invention.
- a separate exogenous promoter may be additionally included for the expression of the nucleic acid, but is not limited thereto.
- Various vectors, promoters, delivery technologies, etc. that can be used for the in vivo delivery and expression of nucleic acids are well known in the art, and a person skilled in the art can perform this without limitation using a known technology depending on the target and purpose.
- the vaccine composition may be characterized by additionally comprising a phospholipid, a cationic lipid, and/or an adjuvant.
- adjuvant of the present invention is a concept that arose when Alexander Glenny discovered that aluminum salt increases immune response, and refers to an auxiliary component added to a vaccine composition or a subject to which a vaccine is administered to induce a stronger immune response.
- the adjuvant may include an emulsifier, muramyl dipeptide, abridin, an aqueous adjuvant, an oil, and more specifically, an aluminum salt such as aluminum phosphate or aluminum hydroxide, a squalene-containing emulsion such as MF59 or an analog thereof (MF59 like), AS03 or an analog thereof (AS03 like), AF03 or an analog thereof (AF03 like), SE or an analog thereof (SE like), a calcium salt, a dsRNA analogue, a lipopolysaccharide, a Lipid A analogue (MPL-A, GLA, etc.), Flagellin, imidazoquinolines, CpG ODN, mineral oil, an agonist of a Toll-like receptor (TLR), a C-type lectin ligand, Examples include, but are not limited to, CD1d ligands (such as ⁇ -galactosylceramide), detergents, liposomes, saponins such as
- the adjuvant may include, for example, Amphigen, LPS, bacterial cell wall extract, bacterial DNA, CpG sequence, poly(I:C), synthetic oligonucleotides and combinations thereof [see: Schijins et al. (2000) Curr. Opin. Immunol. 12:456], mycobacterial phlei (M. phlei) cell wall extract (MCWE) (U.S. Patent No. 4,744,984), M. phlei DNA (M-DNA) and M-DNA-M. phlei cell wall complex (MCC).
- Amphigen e. phlei
- MCWE mycobacterial phlei cell wall extract
- M-DNA M-DNA
- M-DNA-M. phlei cell wall complex MCC
- the adjuvant may include, for example, compounds which can be used as an emulsifier, natural and synthetic emulsifiers and anionic, cationic and nonionic compounds, and among the synthetic compounds, the anionic emulsifiers include, for example, calcium, sodium and aluminum salts of lauric acid and oleic acid, calcium, magnesium and aluminum salts of fatty acids, and organic sulfonates, for example, sodium lauryl sulfate, the synthetic cationic agents include, for example, cetyltriethylammonium bromide, and the synthetic nonionic agents include, but are not limited to, glyceryl esters (e.g., glyceryl monostearate), polyethylene glycol esters and ethers, and sorbitan fatty acid esters (e.g., sorbitan monopalmitate) and polyoxyethylene derivatives thereof (e.g., polyoxyethylene sorbitan monopalmitate).
- the anionic emulsifiers
- the adjuvant may be a natural emulsifier, and the natural emulsifier includes, but is not limited to, acacia, gelatin, lecithin, and cholesterol.
- oils may be mineral oils, vegetable oils or animal oils.
- Mineral oils are liquid hydrocarbons obtained from petroleum jelly by distillation techniques and are also referred to in the art as liquid paraffin, liquid petroleum jelly or white mineral oil.
- Suitable animal oils include, for example, cod liver oil, flounder oil, herring oil, orange roughy oil and shark liver oil, all of which are commercially available.
- Suitable vegetable oils include, for example, canola oil, almond oil, cottonseed oil, corn oil, olive oil, peanut oil, safflower oil, sesame oil, soybean oil and the like.
- FCA Freund's complete adjuvant
- FIA Freund's incomplete adjuvant
- immunomodulatory cytokines may also be included in the vaccine composition, for example as adjuvants, to enhance vaccine efficacy.
- cytokines include interferon alpha (IFN- ⁇ ), interleukin-2 (IL-2), and granulocyte macrophage-colony stimulating factor (GM-CSF) or combinations thereof, preferably but not limited to GM-CSF.
- immunization is performed by administering the peptide of the present invention using CFA as an adjuvant or by administering CpG ODN as an adjuvant together with nanoparticles comprising the peptide of the present invention, but is not limited thereto.
- the vaccine composition of the present invention can be manufactured in a unit dose form or can be manufactured by placing it in a multi-dose container by formulating it using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person having ordinary skill in the art to which the present invention pertains.
- the formulation can be formulated and used in the form of oral formulations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, and sterile injection solutions according to conventional methods.
- Suitable formulations known in the art can be used as disclosed in the literature (Remington's Pharmaceutical Science, Mack Publishing Company, Easton PA).
- Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations are prepared by mixing at least one excipient, such as starch, calcium carbonate, sucrose, lactose, gelatin, etc.
- Liquid preparations for oral administration include suspensions, solutions, emulsions, and syrups, and in addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, flavoring agents, and preservatives may be included.
- Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, and suppositories.
- Suppository bases include witepsol, macrogol, Tween 61, cacao butter, laurin butter, and glycerogelatin.
- the vaccine composition of the present invention can be administered orally or parenterally.
- the route of administration of the composition according to the present invention is not limited to these, but for example, intradermal, intravenous, subcutaneous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, intraperitoneal, enteral, sublingual, oral or topical administration is possible.
- the dosage of the composition according to the present invention varies depending on the patient's weight, age, sex, health condition, diet, administration time, method, excretion rate or disease severity, and can be easily determined by a person skilled in the art.
- the composition of the present invention can be formulated into a suitable dosage form using a known technique for clinical administration.
- the vaccine composition may be administered as a single dose or divided into several doses.
- the doses of the peptide, nucleic acid, nanoparticle and/or adjuvant of the present invention are equally distributed, but this is not limited thereto.
- the optimal dosage of the vaccine composition of the present invention can be determined by standard studies involving observation of an appropriate immune response in a subject. After the initial immunization, the subject may be administered one or more booster immunizations at appropriate intervals.
- the vaccine composition of the present invention can be administered in a pharmaceutically effective amount.
- pharmaceutically effective amount means an amount sufficient to induce or increase an immune response but not causing side effects or serious or excessive immune responses, and an appropriate dosage may be variously determined by factors such as the formulation method, administration method, patient’s age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and response sensitivity.
- the vaccine composition can be administered to a patient on any suitable schedule to induce and/or support a cytotoxic T lymphocyte response to induce and/or support protective immunity for the prevention or treatment of cancer.
- a booster can be administered to support and/or maintain protective immunity.
- the vaccine composition can be administered to the patient once, twice or more times per month, several months or several years.
- administration of the vaccine composition may continue, for example, over the course of several years.
- the vaccine schedule includes, but is not limited to, more frequent administrations at the beginning of the vaccine regimen, and less frequent administrations (e.g., boosters) for a period of time to maintain protective immunity.
- the vaccine composition may be administered in a lower dose at the beginning of the vaccine therapy and in a higher dose over time.
- the vaccine may be administered in a higher dose at the beginning of the vaccine therapy and in a lower dose over time, but is not limited thereto.
- fibrosis refers to pathological wound healing in which connective tissue is abnormally produced and replaces normal parenchymal tissue. Fibrosis can disrupt or block the normal structure and function of an organ or tissue by depositing connective tissue, and can cause various physical abnormalities as a result. Fibrosis can have excessive expression of fibroblast activation protein alpha as a cause or pathological symptom, and can include abnormal deposition of connective tissue due to proliferation and activation of fibroblasts.
- the tumor may be characterized as being a FAP-related tumor.
- the FAP-related tumor is used to mean all tumors characterized by an increase in the concentration of free FAP in tumor tissue or tumor microenvironment, the number of FAP-expressing cells, or the level of FAP expression in cells compared to normal tissue or environment. According to existing reports, in almost all tumors, the number of free FAP and/or FAP-expressing cells in the microenvironment increases compared to normal tissue. Therefore, the vaccination therapy and vaccine composition using the FAP peptide of the present invention can be used as an effective therapeutic agent without limitation on the type of tumor.
- tumor of the present invention includes all neoplasms or hyperplasias that are caused by cells that have escaped from the biological control mechanism and have autonomous and excessive proliferation.
- the above tumors include, for example, benign, premalignant, and malignant tumors, and more specifically, histiocytoma, glioma, astrocytoma, osteoma, various cancers, for example, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, lip cancer, oropharyngeal cancer, leukemia, basal cell carcinoma, ovarian cancer, breast cancer, brain tumor, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, retinoblastoma, melanoma, bladder cancer, peritoneal cancer, parathyroid cancer, adrenal cancer, paranasal sinus cancer, lung cancer, small cell lung cancer, non-small cell lung cancer
- the tumor may preferably be characterized as being a desmoplastic tumor.
- prevention means any act of suppressing or delaying the onset of a desired disease by administering a vaccine composition according to the present invention.
- treatment means any action by which the symptoms of a target disease are improved or beneficially changed by administration of a vaccine composition according to the present invention.
- the above vaccine composition may additionally contain suitable carriers, excipients and diluents commonly used in vaccine compositions.
- Carriers, excipients and diluents that may be included in the above vaccine composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methyl hydroxy benzoate, propyl hydroxy benzoate, talc, magnesium stearate and mineral oil.
- diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants and surfactants.
- the vaccine composition according to the present invention can be formulated and used in various forms according to conventional methods. Suitable formulations include, but are not limited to, oral formulations such as tablets, pills, powders, granules, sugar-coated tablets, hard or soft capsules, solutions, suspensions or emulsions, injections, aerosols, external preparations, suppositories, and sterile injection solutions.
- oral formulations such as tablets, pills, powders, granules, sugar-coated tablets, hard or soft capsules, solutions, suspensions or emulsions, injections, aerosols, external preparations, suppositories, and sterile injection solutions.
- the vaccine composition according to the present invention can be prepared into a suitable formulation using a pharmaceutically inactive organic or inorganic carrier. That is, when the formulation is a tablet, a coated tablet, a sugar-coated tablet, or a hard capsule, it can contain lactose, sucrose, starch or a derivative thereof, talc, calcium carbonate, gelatin, stearic acid, or a salt thereof. In addition, when the formulation is a soft capsule, it can contain vegetable oil, wax, fat, semi-solid, and liquid polyols. In addition, when the formulation is in the form of a solution or syrup, it can contain water, polyol, glycerol, and vegetable oil, etc.
- the vaccine composition may be formulated as a delayed release vehicle or a depot preparation.
- a long-acting preparation may be administered by inoculation or implantation (e.g., subcutaneously or intramuscularly) or by injection.
- the vaccine composition may be formulated with a suitable polymeric or hydrophobic material (e.g., as an emulsion in an acceptable oil) or with an ion exchange resin, or as a sparingly soluble derivative, e.g., a sparingly soluble salt.
- a suitable polymeric or hydrophobic material e.g., as an emulsion in an acceptable oil
- an ion exchange resin e.g., a sparingly soluble derivative
- Liposomes and emulsions are well known examples of delivery vehicles suitable for use as carriers.
- the vaccine composition according to the present invention may further include, in addition to the carrier described above, a preservative, a stabilizer, a wetting agent, an emulsifier, a solubilizer, a sweetener, a colorant, an osmotic pressure regulator, an antioxidant, etc.
- the vaccine composition according to the present invention is administered in a pharmaceutically effective amount.
- the "pharmaceutically effective amount” means an amount sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment, and the effective dosage level can be determined according to the type and severity of the patient's disease, the activity of the drug, the sensitivity to the drug, the time of administration, the route of administration and the excretion rate, the treatment period, the concurrently used drugs, and other factors well known in the medical field.
- the vaccine composition according to the present invention can be administered as an individual therapeutic agent or in combination with other therapeutic agents, can be administered sequentially or simultaneously with conventional therapeutic agents, and can be administered singly or in multiple doses. It is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects by considering all of the above factors, and this can be easily determined by those skilled in the art.
- the pharmaceutical composition of the present invention can be administered to a subject by various routes.
- the pharmaceutical composition can be administered orally or parenterally.
- parenteral administration it can be administered by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, intradermal administration, topical administration, intranasal administration, intrapulmonary administration, and rectal administration.
- the oral composition can be formulated to coat the active agent or protect it from decomposition in the stomach.
- the composition can be administered by any device that allows the active agent to travel to the target cell.
- the method of administration of the vaccine composition according to the present invention can be easily selected according to the formulation, and can be administered orally or parenterally.
- the dosage may vary depending on the patient's age, sex, weight, degree of disease, and route of administration.
- the vaccine composition may be characterized by inducing a FAP-specific cytotoxic T cell response or a FAP-specific CD8+ T cell response.
- the vaccine composition may be characterized by inducing an immune response to free FAP or FAP-expressing cells in a subject.
- the FAP-expressing cells may be characterized by being FAP-expressing fibroblasts.
- the vaccine composition may be characterized by inducing an anti-tumor response.
- the anti-tumor response may include, but is not limited to, induction of a tumor or tumor microenvironment-specific immune response, reduction in the number of tumor cells, reduction in tumor size, death of tumor cells, inhibition of tumor metastasis, etc.
- the vaccine composition can induce inhibition of lipid accumulation in the liver, inhibition of hepatic steatosis, inhibition of hepatic inflammation, inhibition of hepatic fibrosis, and reduction of the proportion of swollen cells in the liver, but is not limited thereto.
- the present invention relates to the use of the peptide, nucleic acid or nanoparticle of the present invention for the manufacture of a pharmaceutical composition for the prevention or treatment of a fibroblast activating protein alpha (FAP)-related disease.
- FAP fibroblast activating protein alpha
- the present invention relates to a method for preventing or treating a disease associated with fibroblast activation protein alpha (FAP), comprising administering to a subject a peptide, nucleic acid or nanoparticle of the present invention, or administering to a subject a vaccine composition of the present invention.
- FAP fibroblast activation protein alpha
- the preventive or therapeutic method of the present invention includes a vaccination method, a vaccination method, or an immunization method for preventing or treating a disease associated with fibroblast activation protein alpha (FAP).
- FAP fibroblast activation protein alpha
- a step of analyzing the HLA type of the subject may be additionally included prior to the step of administering to the subject the vaccine composition of the present invention comprising the peptide, nucleic acid, nanoparticle or any one or more of these of the present invention.
- a vaccine composition containing different peptides can be administered depending on the HLA type of the subject.
- the vaccine composition when the subject has MHC of the HLA-A1 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
- the vaccine composition when the subject possesses MHC of the HLA-A2 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, 4, 5, and 30 to 45.
- the vaccine composition when the subject possesses MHC of the HLA-A3 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
- the vaccine composition when the subject has MHC of the HLA-A24 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
- the vaccine composition when the subject has MHC of the HLA-A26 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97 as the restricted epitope peptide.
- the vaccine composition when the subject has MHC of the HLA-B7 type, may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
- one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129 may be administered.
- one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149 may be administered.
- HLA type is one embodiment and is not limited thereto, and one or more different HLA type restricted peptides may be administered regardless of the HLA type of the subject or without an analysis step for the HLA type.
- the present invention relates to a pharmaceutical composition for the prevention or treatment of tumors for co-administration with an anticancer agent comprising the peptide, nucleic acid or nanoparticle of the present invention from another aspect.
- the present invention provides a use of the peptide, nucleic acid or nanoparticle in combination with an anticancer agent for the prevention or treatment of tumors.
- the present invention provides a method for preventing or treating a tumor, comprising a step of co-administering the peptide, nucleic acid or nanoparticle; and an anticancer agent to a subject.
- the present invention relates from another aspect to the use of the peptide, nucleic acid or nanoparticle of the present invention for the preparation of a pharmaceutical composition for the prevention or treatment of tumors in combination with an anticancer agent.
- the peptides, nucleic acids and/or nanoparticles of the present invention inhibit the formation of ECM, which acts as a physical barrier to anticancer therapy, by reducing or depleting FAP-expressing CAFs in the tumor microenvironment, and consequently enhance the infiltration or penetration of anticancer agents and immune cells into the tumor, resulting in a more remarkable anti-tumor effect than when each drug is administered as a monotherapy. Therefore, the anticancer agent to be co-administered with the pharmaceutical composition of the present invention can be selected without limitation from various anticancer therapeutic agents or anticancer agents reported in the art.
- the anticancer agent may be, for example, a chemotherapy agent (cytotoxic anticancer agent), a targeted anticancer agent, an immunotherapy agent, a hormonal anticancer agent, a cell therapy agent, etc., but is not limited thereto.
- a chemotherapy agent cytotoxic anticancer agent
- a targeted anticancer agent a targeted anticancer agent
- an immunotherapy agent a hormonal anticancer agent
- a cell therapy agent etc., but is not limited thereto.
- the chemical anticancer agent is also called a cytotoxic anticancer agent, and is an anticancer agent that directly attacks cancer cells and exhibits an anticancer effect, and includes, but is not limited to, alkylating agents such as cyclophosphamide, ifosfamide, bendamustine, delphalan, and cisplatin; antimetabolites such as capecitadine, cytarabine, doxifluridine, fluorouracil, clofarabine, fludarabine, and decitamine; DNA rotator cuff inhibitors such as doxorubicin, daunorubicin, idarubicin, etoposide, and topotecan; microtubule inhibitors such as cabazitaxel, docetaxel, vincristine, and the like; and other chemical anticancer agents such as mitomycin C, bleomycin, and hydroxyurea.
- alkylating agents such as cyclophosphamide,
- the targeted anticancer agent is an anticancer agent containing as a main component a substance that targets and binds to or interacts with a specific antigen expressed by a cancer or an antigen related to a cancer, and examples thereof include, but are not limited to, antibody therapeutics such as cetuximab, trastuzumab, bevacizumab, rituximab, ibltumomab, alemtuzumab, brentuximab, and elotuzumab; and signal transduction inhibitors such as erlotinib, gefitinib, vandetanib, afatinib, rapanitib, axitinib, pazopanib, sunitinib, and sorafenib.
- antibody therapeutics such as cetuximab, trastuzumab, bevacizumab, rituximab, ibltumomab, alemtuzuma
- the immunotherapy anticancer agent includes, but is not limited to, a CTLA-4 antibody such as apilimumab, a PD-1 antibody such as pembrolizumab or nivolumab, a PD-L1 antibody such as atezolizumab, and an immune checkpoint inhibitor such as an IDO inhibitor.
- a CTLA-4 antibody such as apilimumab
- a PD-1 antibody such as pembrolizumab or nivolumab
- a PD-L1 antibody such as atezolizumab
- an immune checkpoint inhibitor such as an IDO inhibitor.
- the hormonal anticancer agent includes, but is not limited to, androgen suppressants such as bicalutamide and enzalutamide, and female hormone suppressants such as tamoxifen, anastrozole and letrozole.
- the cell therapy agent means a therapy agent containing living immune cells as an active ingredient, and includes, but is not limited to, cytotoxic T cell therapy agents, CAR-T cell therapy agents, and CAR-NK cell therapy agents.
- combined administration means administering two or more types of effective ingredients simultaneously or sequentially, or administering them simultaneously, sequentially, or at specific intervals so that an improved effect can be achieved compared to the effect expected when each effective ingredient is administered independently due to the action of the two or more types of effective ingredients.
- the combined administration may be characterized by administering the peptide, nucleic acid or nanoparticle of the present invention in combination with one or more anticancer agents, and may also be performed in parallel with other anticancer therapies.
- each of the effective ingredients to be co-administered may be characterized by being administered through an independent route.
- Each effective ingredient may be administered independently by a person skilled in the art with a suitable administration method and dosage.
- it may be characterized by preferably administering peptides, nucleic acids, or nanoparticles intradermally, and administering anticancer agents through intravenous injection, but is not limited thereto.
- the combined administration of the peptide, nucleic acid or nanoparticle and the anticancer agent may be characterized by being administered simultaneously.
- the combined administration of the peptide, nucleic acid or nanoparticle and the anticancer agent may be characterized by sequential administration.
- the anticancer agent may be administered after the administration of the peptide, nucleic acid or nanoparticle, or the peptide, nucleic acid or lipid nanoparticle may be administered after the administration of the anticancer agent.
- the peptide, nucleic acid or nanoparticle and the anticancer agent when administered sequentially, it can be characterized in that they are administered at a certain time interval, and for non-limiting examples, they can be sequentially administered at 1 minute intervals, 5 minutes intervals, 10 minutes intervals, 20 minutes intervals, 30 minutes intervals, 1 hour intervals, 1 day intervals, several days intervals, or 1 week to several weeks intervals, but are not limited thereto, and can be sequentially administered at an appropriate interval by a person skilled in the art.
- the administration of the peptide, nucleic acid or nanoparticle and the anticancer agent can be performed independently and repeatedly one or more times.
- each administration interval can be easily adjusted independently by a person skilled in the art according to the condition of the subject and the level of the desired effect.
- the peptide, nucleic acid or nanoparticle and the anticancer agent can be independently administered at 1 hour intervals, 6 hours intervals, 8 hours intervals, 12 hours intervals, 1 day intervals, 2 days intervals, 1 week intervals, 2 weeks intervals, or 1 month intervals, but is not limited thereto.
- the pharmaceutical composition may be administered in a formulation and dosage independent of the formulation of the anticancer agent to be administered in combination.
- the pharmaceutical composition comprising the peptide, nucleic acid or nanoparticle and the pharmaceutical composition comprising the anticancer agent may each be administered in an effective dosage, but is not limited thereto.
- amino acid sequence substantially identical to an enzyme to be implemented in the present invention and a base sequence encoding the same fall within the scope of the present invention.
- Substantially identical includes a case where the homology of the amino acid or base sequence is very high, and also means a protein that shares structural characteristics or has the same function as that used in the present invention regardless of the homology of the sequence.
- a protein in which a part of a sequence other than the sequence constituting the core of the present invention is deleted or a fragment of the base sequence encoding the same may also be included in the present invention, and therefore the present invention includes all amino acid or base sequences that have the same function as that used in the present invention regardless of the length of the fragment.
- 1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine DOPE
- 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] DSPE-PEG1000
- 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] DSPE-PEG2000-PDP) - Avanti Polar Lipids.
- CpG oligoDNA modified with phosphorothioate backbone (CpG ODN; 5’-TCC ATG ACG TTC CTG ACG TT-3’, SEQ ID NO: 151) - Genotech and Bioneer.
- Example 1-2 Animals and cell lines
- mice Female C57BL/6 mice (Orient Bio) were raised in a pathogen-free environment. Animal care and experimental procedures were approved by the Institutional Animal Care and Use Committee of the Korea Advanced Institute of Science and Technology (KAIST) (Accreditation number: KA2020-59).
- KAIST Korea Advanced Institute of Science and Technology
- E.G7-OVA cell line (ATCC; Manassas, VA, USA).
- E.G7-OVA cell line was cultured in RPMI-1640 medium (Welgene) supplemented with 10% heat-inactivated FBS, 1% penicillin/streptomycin, 2 mM L-glutamine, 4.5 g/l glucose, 10 mM HEPES, 1 mM sodium pyruvate, 50 ⁇ M 2-mercaptoethanol, and 0.5 mg/ml G418 (Gibco).
- Panc02 cell line was provided by Professor Seok-Jo Kang, KAIST. Panc02 cells were maintained in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin/streptomycin.
- the MC38 cell line was provided by Professor Chan-Hyuk Kim of the Korea Advanced Institute of Science and Technology. MC38 cells were maintained in DMEM medium supplemented with 10% FBS and 1% penicillin/streptomycin.
- EO771.lmb cell line ATCC; Manassas, VA, USA.
- EO771.lmb cell line was cultured in DMEM medium (Welgene) supplemented with 10% heat-inactivated FBS and 1% penicillin/streptomycin.
- Antibodies used were as follows: anti-CD16/CD32 (BioLegend; Catalog #101319, clone 2.4G2), anti-CD45 Pacific Blue (BioLegend; Catalog #103125, clone 30-F11), anti-CD45 PerCP/Cy5.
- Example 1-4 Selection of mouse FAP target peptides and vaccine design
- mice FAP target epitope peptides were predicted using epitope peptide prediction algorithms including NetMHC3.0, BIMAS, and PREDEP.
- the mouse FAP full-length sequence used is SEQ ID NO: 152 (Uniprot NO. P97321). All epitope peptides were selected as 9-mer peptide lengths recognized by MHC-I haplotype H-2Kb. The sequences of the selected peptides are shown in Table 1:
- Example 1-5 Evaluation of immunogenicity and antitumor effect
- mice For immunogenicity testing of the selected peptide candidates, 6-week-old female C57BL/6 mice were immunized twice using a homologous prime-boost regimen at 1-week intervals. Mice were immunized by subcutaneous injection of 100 ⁇ g of each peptide emulsified in Complete Freund's Adjuvant (CFA; Sigma Aldrich) into both footpads. Mice were sacrificed 10 days after the final immunization. Antigen-specific T cell responses were evaluated by isolating spleen cells from mice and restimulating them ex vivo with each peptide (10 ⁇ g/ml). IFN- ⁇ -producing CD8+ T cells were quantified by intracellular cytokine staining (ICS) (Fig.
- ICS intracellular cytokine staining
- IFN- ⁇ -producing cells were measured by enzyme-linked immunospot (ELISpot) assay.
- spleen cells (3 ⁇ 106 cells per tube) were restimulated with each peptide for 1 h.
- GolgiStop and GolgiPlug (BD Biosciences) were added to each tube to inhibit intracellular transport of cytokines, and the cells were cultured for an additional 5 h, immunostained with anti-CD3e and anti-CD8a antibodies, and then stained with Live/Dead cell dye for 20 min at 4°C.
- cytokine staining For intracellular cytokine staining, cells were permeabilized with Cytofix/Cytoperm solution (BD Biosciences) and incubated with anti-IFN- ⁇ antibody. Samples were then washed and analyzed by flow cytometry. To measure IFN- ⁇ -producing cells by ELISpot, spleen cells (3 ⁇ 10 5 cells/well) were seeded in 96-well microplates coated with mouse IFN- ⁇ -specific monoclonal antibodies and restimulated with each peptide for 30 h. IFN- ⁇ -producing spots were generated using the mouse IFN- ⁇ ELISpot kit (R&D Systems, Minneapolis, MN, USA) according to the manufacturer's protocol. Blue-black spots at the sites of cytokine localization were counted using an automated ELISpot reader (AID GmbH, France, Germany).
- mice that were inoculated subcutaneously with 2 ⁇ 10 5 E.G7-OVA cells into the right flank.
- the mice were randomly divided into groups such that the average tumor volumes of each group were approximately equal, and then immunized twice with each peptide at 1-week intervals using the same method as described above. Tumor growth was monitored every other day using digital calipers, and the tumor volumes were calculated as 0.5 ⁇ length ⁇ width. The mice were euthanized when the average tumor volume reached the ethical endpoint ( ⁇ 1,500 mm 3 ).
- Monoarginine-cholesterol (MA-Chol) and DSPE-PEG2000-FAP PEP were synthesized based on previously reported methods.
- Cysteinylated FAP PEP (Cys-FAP PEP ) prepared using two selected peptide candidates, CKLWRYSYTA and CYFRNVDYLL, was conjugated to DSPE-PEG2000-PDP by disulfide exchange reaction. Cys-FAP PEP and DSPE-PEG2000-PDP were dissolved in dimethyl sulfoxide and mixed at a molar ratio of 1:2. The solution was gently vortexed overnight at room temperature and the reaction was quenched by the addition of acetonitrile.
- the DSPE-PEG2000-FAP PEP conjugate was purified by high-performance liquid chromatography (HPLC; Agilent, Santa Clara, CA, USA) using a C4 column (Nomura Chemical, Japan) and characterized using MALDI-TOF spectroscopy.
- FAPPEP-SLNP nanovaccines containing MA-Chol:DOPE:DSPE-PEG1000:DSPE-PEG2000-FAP PEP (molar ratio, 48.625:48.625:2.25:0.5) were prepared using a thin film formation and rehydration method. After drying all lipid components dissolved in chloroform and methanol, the solvent was removed under vacuum, and the resulting lipid film was rehydrated with HEPES-buffered glucose (HBG) containing CpG ODN. The solution was sonicated for 10 min, magnetically stirred for more than 4 h at room temperature, and extruded more than 11 times using a mini extruder (Avanti Polar Lipids). The loading efficiency was close to 100% using 1.65 nmol of CpG ODN encapsulated in 8 ⁇ mol of SLNP.
- HEPES-buffered glucose HEPES-buffered glucose
- FAP PEP -SLNP For the characterization of FAP PEP -SLNP, the hydrodynamic size was determined by DLS at room temperature using a Zetasizer Nano range system (Malvern, Worcestershire, UK). Both the morphology and size of FAP PEP -SLNP were characterized by TEM using a Philips TECNAI F20 instrument (Philips Electronic Instrument Corp., Mahwah, NJ, USA) and 1% uranyl acetate solution was used for negative staining. The average size of the nanovaccines was measured using Gatan Microscopy Suite (GMS) software (Gatan, Pleasanton, CA, USA).
- GMS Gatan Microscopy Suite
- Example 1-7 Evaluation of antitumor effect of FAP PEP -SLNP nanovaccine
- E.G7-OVA treatment model 2 ⁇ 10 5 E.G7-OVA cancer cells were injected into the right flank of the mice.
- tumor-bearing mice were immunized three times at 4-day intervals with FAP PEP1 -SLNP or OVA PEP -SLNP nanovaccines (CpG, 0.4 nmol per mouse; FAPPEP, 5 nmol per mouse; OVAPEP, 5 nmol per mouse; SLNP, 2 ⁇ mol per mouse) by subcutaneous injection into both footpads at the indicated time points.
- Panc02 treatment model 1 ⁇ 106 Panc02 cancer cells were injected into the right flank of the mice. Sixteen days after Panc02 cell inoculation, the tumor-bearing mice were immunized three times with FAP PEP1 -SLNP at 4-day intervals using the same method as above.
- MC38 treatment model 1x10 5 MC38 cancer cells were injected into the right flank of the mice. Five days after MC38 cancer cell inoculation, the tumor-bearing mice were immunized three times with FAP PEP1 -SLNP at 4-day intervals using the same method as above.
- mice were injected intraperitoneally with Dox (10 mg/kg) every other day for a total of four times as indicated. Tumor growth was monitored every other day and mouse survival was monitored. Mice were euthanized when the mean tumor volume reached the ethical endpoint.
- Tumor tissues were resected, fixed in 10% formalin solution, embedded in paraffin, and then cut into 5- ⁇ m-thick sections. Collagen deposition in tumor tissues was assessed by Weigert's iron hematoxylin, Biebrich's scarlet-acid fuchsin solution, and Masson's trichrome staining with aniline blue. All section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). Masson's trichrome-stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
- Tumor-infiltrating CD8+ T cells and FAP+ CAFs were analyzed by dissociating tumor tissues into small pieces and then enzymatically and mechanically dissociating single-cell suspensions using a mouse tumor dissociation kit (Miltenyi Biotech, Auburn, CA, USA) and gentleMACS Dissociator (Miltenyi Biotech).
- the cell suspension was filtered through a cell strainer (70 ⁇ m) and washed with Dulbecco’s phosphate-buffered saline (DPBS). Red blood cells (RBCs) were removed by incubation with 2 ml RBC lysis buffer (BioLegend) for 2 min at room temperature with gentle shaking.
- the dissociated cells were immunostained with anti-CD45, anti-CD3e, anti-CD8a, and anti-FAP ⁇ antibodies, and stained with Live/Dead cell dye for 20 min at 4°C. The cells were then washed and subjected to flow cytometry.
- Spleens were harvested from immunized mice, and splenocytes were isolated.
- the isolated cells were stimulated with soluble anti-CD3e (BD Biosciences; Catalog #553057, clone 145-2C11) and anti-CD28 (BD Biosciences; Catalog #553294, clone 37.51) antibodies for 1 h at 37°C, and GolgiStop and GolgiPlug were added to inhibit intracellular transport of cytokines.
- the cells were incubated for an additional 5 h and then stained with anti-CD3e and anti-CD4 antibodies and Live/Dead cell dye for 20 min at 4°C.
- cytokine staining For intracellular cytokine staining, cells were permeabilized using Cytofix/Cytoperm solution and then incubated with anti-IL-17A antibody.
- spleen cells For analysis of antigen-specific splenic T cells, spleen cells were restimulated with FAPPEP or OVAPEP (10 ⁇ g/ml) for 1 h, followed by addition of GolgiStop and GolgiPlug and incubation for an additional 5 h as described above. Cells were then stained with anti-CD3e, anti-CD4 and anti-CD8a antibodies and Live/Dead cell dye for 20 min at 4°C. After surface marker staining, cells were permeabilized and incubated with anti-IFN- ⁇ antibody. All samples were analyzed by flow cytometry.
- cypate a near-infrared dye, as a surrogate for small-molecule chemotherapeutic drugs.
- Mice were injected with 1 ⁇ 10 5 MC38 cancer cells into the right flank and immunized three times at 4-day intervals with FAPPEP1-SLNPs 5 days after MC38 cancer cell inoculation according to the same schedule used in the MC38 tumor treatment study.
- cypate dye 2.5 mg kg-1 was injected intravenously via the retro-orbital route.
- Tumor tissues were harvested 2 h after cypate dye injection, and fluorescence signals were evaluated using an in vivo imaging system (IVIS; PerkinElmer, Waltham, MA, USA).
- Example 1-12 Evaluation of the cancer metastasis inhibition effect of FAP PEP1 peptide vaccine and tissue analysis
- the evaluation of the anti-cancer metastasis effect was tested in the form of a peptide vaccine using the FAPpep1 peptide.
- orthotopic tumor models were established in female C57BL/6 mice. 1 ⁇ 10 5 EO771.lmb cancer cells were injected into the right abdominal mammary glands (4th pair) of mice. Tumor growth was monitored daily using a digital caliper, and the tumor volume was calculated as (0.5 ⁇ length ⁇ width 2 ). Primary tumors were resected when the average tumor volume reached ⁇ 300 mm 3 . Four days after resection of the primary tumors, the mixture of FAPpep 1 (100 ⁇ g/head) and CpG-ODN (10 ⁇ g/head) was immunized twice at 1-week intervals via subcutaneous injection into both footpads. The mice were sacrificed and analyzed 1 week after the final vaccination.
- Lung and liver tissues of sacrificed mice were excised, fixed in 10% formalin solution, embedded in paraffin, and cut into 5- ⁇ m-thick slices.
- the lung and liver tissue samples were stained with H&E, and all section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). The stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
- Example 1-13 NASH therapeutic effect of FAP PEP1 peptide vaccine and serum and tissue analysis
- mice were fed a methionine/choline deficient (MCD) diet for 4 weeks, or a methionine/choline sufficient (MCS) diet as a control group to establish a model.
- MCD methionine/choline deficient
- MCS methionine/choline sufficient
- AST aspartate transaminase
- ALT alanine transferase
- HDL total bilirubin
- liver tissues were excised, fixed in 10% formalin solution, embedded in paraffin, and cut into 5- ⁇ m-thick slices.
- the prepared liver tissue samples were stained with H&E and analyzed, or stained with isopropanol and Oil Red O. All section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). The stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
- Example 2 Prediction and screening of immunodominant peptide epitopes of FAP.
- the immunodominant peptide epitopes of FAP were predicted using three epitope peptide prediction programs, netMHC3.0, BIMAS, and PREDEP (Fig. 3).
- Fig. 3 The immunodominant peptide epitopes of FAP were predicted using three epitope peptide prediction programs, netMHC3.0, BIMAS, and PREDEP (Fig. 3).
- Fig. 4a the identified epitope peptides.
- six of the nine selected peptides were tested and screened for in vivo immunogenicity.
- C57BL/6 mice were subcutaneously immunized twice at 1-week intervals with a mixture of each peptide and complete Freund's adjuvant (CFA).
- CFA complete Freund's adjuvant
- mice were sacrificed, and spleen cells were isolated to evaluate the antigen-specific T cell responses to each peptide (Fig. 4b).
- Isolated splenocytes were restimulated with each peptide, and interferon- ⁇ (IFN- ⁇ ) production by the cells was analyzed by intracellular cytokine staining (ICS) and enzyme-linked immunospot (ELISpot) assays.
- the ICS assay showed that immunization with each of the three peptides (KLWRYSYTA, GLFKCGIAV, and YFRNVDYLL) induced significantly higher frequencies of IFN- ⁇ -secreting CD8+ T cells than did immunization with the other three peptides (Figs.
- the three ICS-positive peptides were also selected as positive candidates in the ELISpot assay (Figs. 4e,f and 6a–f). Based on the ICS and ELISpot assays, these three peptides (KLWRYSYTA, GLFKCGIAV, and YFRNVDYLL) were ultimately selected for further evaluation of their antitumor efficacy.
- the murine lymphoma cell line E.G7-OVA is a popular model system to study MHC class I-restricted CD8+ T cell responses, and was used to induce tumor formation in syngeneic mice, and the presence of FAP+ CAFs was confirmed in the generated tumor tissues by flow cytometry (Fig. 7).
- mice When E.G7-OVA tumors reached an average volume of ⁇ 100 mm3, mice were randomly divided into four groups and immunized twice with a mixture of each peptide and CFA at 1-week intervals (Fig. 4g). Immunization with GLFKCGIAV + CFA was ineffective in suppressing tumor growth, whereas immunization with two other peptide candidates, KLWRYSYTA (designated FAP PEP1 ) and YFRNVDYLL (designated FAP PEP2 ), resulted in effective tumor growth inhibition compared to the control (Fig. 4h). These results imply that these two peptides are suitable epitope peptide candidates that enable potent induction of peptide epitope-specific CD8+ T cells and high antitumor efficacy.
- Example 3 Synthesis, characterization, and antitumor efficacy evaluation of FAP epitope peptide-presenting nanovaccine
- FAP peptide-labeled SLNPs were composed of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) to promote endosomal escape; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPE-PEG1000), which provides colloidal stability in vivo; and monoarginine-cholesterol (MA-Chol), which enables complexation with CpG adjuvants and provides mechanical stability (Figure 8).
- DOPE 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine
- DSPE-PEG1000 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000]
- MA-Chol monoarginine-cholesterol
- Peptide epitope peptides were introduced to the surface of SLNPs via cleavable disulfide bonds using cysteine-modified peptides at the N-terminus (N'-Cys-FAP PEP1 and N'-Cys-FAP PEP2 ) and DSPE-PEG2000 ( Figures 9 and 10).
- FAP PEP1 -SLNP is the most effective nanovaccine for treating tumors containing FAP+ CAFs in the TME.
- FAP PEP1 -H-2Kb tetramer an antibody that specifically binds to CD8+ cells specific for FAP PEP1 . was prepared using the 'QuickSwitchTM Quant H-2 Kb Tetramer Kit-PE' product (MBL Inc.) (MBL Inc.).
- MBL Inc. Quant H-2 Kb Tetramer Kit-PE' product
- FAP PEP1 -H-2Kb tetramer was treated, it was confirmed that the fluorescence reaction indicating that the tetramer was bound was very high in the group immunized with FAP PEP1 -SLNP (Fig. 13).
- E.G7-OVA tumor-bearing mice with an average tumor volume of ⁇ 100 mm 3 were immunized three times in total with FAP PEP1 -SLNP or OVA PEP -SLNP at 4-day intervals (Fig. 14a).
- Treatment with FAP PEP1 -SLNP significantly inhibited tumor growth, showing similar or slightly better efficacy compared to OVA PEP -SLNP nanovaccine targeting cancer cells (Fig. 14b). It also significantly prolonged survival compared to untreated controls (Fig. 14c).
- Vaccination with FAP PEP1 -SLNPs may affect ECM production in tumors because it induces a cytotoxic immune response against FAP+ CAFs in the TME.
- Masson's trichrome (MT) staining showed that immunization with FAP PEP1 -SLNPs significantly reduced the collagen-positive area in tumor tissues compared with the untreated control tumor tissues (Fig. 14d, e).
- OVA PEP -SLNPs showed similar antitumor efficacy as FAP PEP1 -SLNPs, the former failed to reduce ECM production, suggesting that the antitumor efficacy of the latter may be the result of the depletion of FAP+ CAFs in the TME.
- FAP PEP1 -SLNP is a nanovaccine that can induce not only FAP-specific CD8+ T cell responses but also CD4+ T cell responses, indicating that it can exhibit high antitumor efficacy with a low possibility of inducing autoimmune responses (J Exp Med 2013, 210 (6), 1137-1151; J Exp Med 2013, 210 (6), 1125-1135; Cancer Cell 2014, 25 (6), 719-734; Nat Rev Immunol 2018, 18 (10), 635-647; Immunity 2021, 54 (12), 2701-2711).
- FAP PEP1 -SLNP could act on other desmoplastic tumors.
- FAP + CAFs existed in MC38 and Panc02 tumors, which are known as desmoplastic tumors. After reaching an average of 300 mm 3 , E.G7-OVA, MC38, and Panc02 tumors were excised, dissociated into single cells, and analyzed by flow cytometry. As a result, we confirmed that the ratio of FAP + CAFs was high in both tumor models, including E.G7-OVA tumors (Fig. 16).
- FAP PEP1 -SLNP showed a continuous tumor suppression effect upon additional inoculation even after FAP + CAF were removed in the early stage of the tumor. Therefore, we conducted an experiment to confirm whether cancer cells themselves also express the FAP protein.
- the size of MC38 tumors reached 300 mm 3 , the tumors were excised and dissociated into single cells, and the abundance ratio of cancer cells and FAP + CAF was confirmed through flow cytometry.
- FAP was expressed in the cancer cells themselves in large tumors, and therefore, it was confirmed that 7FAP PEP1 -SLNP showed a continuous therapeutic efficacy upon additional inoculation even after FAP + CAF were removed in the early stage (Fig. 17).
- Example 6 Confirmation of antitumor effect through combination therapy of FAP PEP1 -SLNP nanovaccine and anticancer drug
- the dense and rigid ECM of connective tissue tumors acts as a powerful physical barrier not only to chemotherapeutic agents but also to biological agents such as antibodies and CAR-T cells, reducing the therapeutic efficacy of various anticancer therapies (Signal Transduct Target Ther 2021, 6 (1), 153, Proc Natl Acad Sci USA 2019, 116 (6), 2210-2219;, Nat Nanotechnol 2021, 16 (1), 25-36).
- FAP PEP1 -SLNP nanovaccine to deplete CAFs in the TME could reduce ECM density, thereby enhancing the penetration of chemotherapeutic drugs into connective tissue tumors.
- the murine pancreatic ductal adenocarcinoma cell line Panc02 and the colorectal cancer cell line MC38 were chosen as connective tissue tumor models (PLoS One 2013, 8 (11), e80580; Cancer Res 2018, 78 (5), 1321-1333; Nat Commun 2020, 11 (1), 515).
- FAP PEP1 -SLNP nanovaccine significantly slowed tumor growth and prolonged the survival period of immunized mice.
- Non-immunized mice were used as controls in both Panc02 (Fig. 20a-c) and MC38 tumor models (Fig. 20d-f), confirming that the FAP PEP1 -SLNP nanovaccine was also effective against these connective tissue tumors.
- Cypate dye was chosen as a model drug to visualize drug penetration in tumors.
- MC38 colorectal tumor-bearing mice were immunized with FAP PEP1 -SLNP nanovaccines for a total of three injections at 4-day intervals. After the tumor volume reached ⁇ 300 mm 3 , free Cypate was intravenously injected into the immunized mice, and the tumors were harvested for ex vivo fluorescence imaging 2 h later.
- the accumulation of dye in the tumors was significantly higher in the vaccinated group than in the unvaccinated control group ( Figure 21a ), suggesting that the drug penetration was enhanced due to the decrease in the density of the intratumoral ECM.
- FAP+CAF secrete extracellular vesicles from primary tumors to promote metastasis and protect cancer cells from anoikis and shear stress in the blood circulation by forming clusters with cancer cells. Therefore, the previously used tail vein injection metastasis model is not suitable for confirming the role of FAP+CAF.
- an orthotopic breast tumor model was constructed using the EO771.lmb metastatic breast cancer cell line by the method described in Examples 1-13. Since it was confirmed that the ratio of FAP+CAF was high at an average tumor size of 300 mm 3 in Example 5 (Fig. 16), the tumor was resected when the tumor size reached 300 mm 3 .
- the FAP PEP1 peptide vaccine and CpG ODN were immunized twice by subcutaneous injection into the footpad at one-week intervals to confirm the metastasis-suppressing effect of the FAP PEP1 peptide vaccine on metastasis (Fig. 23).
- Example 8 Confirmation of the efficacy of FAP PEP1 peptide vaccine in treating nonalcoholic steatohepatitis (NASH)
- Nonalcoholic steatohepatitis is a chronic liver disease characterized by hepatic steatosis and inflammation, which may progress to fibrosis, cirrhosis, and liver cancer.
- FAP cleaves fibroblast growth factor 21 (FGF21), an important regulator of lipids, and dysregulation thereof may contribute to the progression of NASH.
- FGF21 fibroblast growth factor 21
- Example 8-1 Confirmation of NASH therapeutic effect of FAP PEP1 peptide vaccine
- a NASH model was established using a methionine-choline deficient diet (MCD) (Nat Rev Gastro Hepat 16, 411-428 (2019)). The establishment of the NASH model was verified through monitoring of body weight and liver weight. Stages 2 and 3 NASH models and a control group model were prepared through 4 weeks of MCD diet and MCS diet therapy, and a mixture of the FAP PEP1 peptide vaccine and CpG ODN was subcutaneously injected twice into both heel pads of each mouse at 1-week intervals (Fig. 27a). After administration of the FAP PEP1 peptide vaccine, body weight was significantly recovered (Fig. 27b), and when the livers of the mice were removed one week after the final immunization and their weights were measured, it was confirmed that the liver weight was significantly recovered after administration of the FAP PEP1 peptide vaccine (Fig. 27c).
- Example 8-2 Confirmation of the liver damage alleviation effect of FAP PEP1 peptide vaccine
- mice were collected just before the sacrifice of mice.
- Mice of the NASH model immunized with the FAP PEP1 peptide vaccine showed a significant decrease in the levels of AST, ALT, and total bilirubin, which are indicators of liver damage (Figs. 28a to 28c).
- the level of HDL was increased (Fig. 28d), confirming that the FAP PEP1 peptide vaccine had a protective effect against liver damage induced by the MCD diet.
- Example 8-3 Liver tissue analysis results after immunization with FAP PEP1 peptide vaccine
- tissue analysis was performed using Oil Red O staining and H&E staining.
- H&E staining revealed balloon-shaped adipocytes in the liver tissue, and while unstained areas representing adipocytes were seen in the liver of the MCD diet-fed control group, a marked reduction in hepatocyte swelling was observed in mice immunized with the FAP PEP1 peptide vaccine (Figs. 30a and 30b).
- the histological analysis results indicate that the FAP PEP1 peptide vaccine exhibits an excellent therapeutic effect on hepatic steatosis, which can induce liver tissue recovery in the NASH model.
- These histological results support the results in the examples, such as body weight, liver weight, and serum analysis, implying an excellent therapeutic effect of the FAP PEP -SLNP nanovaccine in the treatment of NASH.
- Examples 2 to 8 demonstrate that immunotherapy using the predicted FAP epitope can exhibit excellent therapeutic effects not only for the treatment of cancer and inhibition of metastasis, but also for FAP-related fibrotic diseases such as NASH. It has already been well known through many studies that FAP is selectively overexpressed in cancer-associated fibroblasts present in human cancers and myofibroblasts or hepatic stellate cells in fibrotic diseases and that it promotes the growth and metastasis of cancer or mediates fibrotic diseases by the same mechanism as in mouse models. Therefore, it is suggested that the efficacy results of the FAP epitope vaccine shown in mouse cancer models and NASH models can be applied to humans.
- the similarity between the FAP protein of mice and the FAP protein of humans is about 94%, so there is a high possibility that the FAP PEP1 peptide epitope verified in mice has cross-reactivity with a specific HLA type in humans.
- the FAP PEP1 verified has a sequence identical to the peptide epitope candidate predicted from human HLA-A2.
- the epitope of human FAP (SEQ ID NO: 153, Uniprot No. B4DLR2) according to HLA Super type was additionally confirmed. Prediction of FAP target epitope peptides was performed using epitope peptide prediction algorithms including NetMHC3.0, BIMAS, and PREDEP. All epitope peptides were selected as 8-mer to 10-mer peptide lengths recognized by H-2Kb, which is an MHC-I haplotype. HLA Super types were analyzed for HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8, and HLA-B27. The final derived FAP epitope sequences are shown in Tables 2 to 5 below.
- a clinical trial of a FAP vaccine capable of targeting the FAP protein and eliminating cells expressing FAP can be performed as follows. 1) Analyze the HLA type of each patient, and select one or more FAP epitope peptides corresponding to the individual patient from the peptide sequences described in Tables 1 to 5. 2) The selected peptide sequence candidates can be administered together with an adjuvant (such as CpG or poly(I:C)), or 3) the selected peptide sequence candidates can be manufactured into a nanovaccine including nanoparticles and administered together with an adjuvant.
- an adjuvant such as CpG or poly(I:C)
- the vaccination therapy using the peptide sequences described in Tables 2 to 5 of the present invention will exhibit excellent effects in the prevention and treatment of FAP-related diseases such as cancer, cancer metastasis, NASH, etc.
- the peptide of the present invention has excellent immunodominance and exhibits a strong induction effect of CD8+ T cells specific for fibroblast activation protein (FAP) and FAP-expressing fibroblasts (FAP+ CAF).
- FAP fibroblast activation protein
- FAP+ CAF FAP-expressing fibroblasts
- the peptide of the present invention can exhibit excellent preventive and therapeutic effects on FAP-related diseases such as cancer, fibrosis, NASH, etc. through immunity induction against fibroblast activation protein, and has significantly lower toxicity compared to other FAP-expressing CAF targeting vaccines.
- FAP-related diseases such as cancer, fibrosis, NASH, etc.
- FAP-expressing fibroblasts FAP-expressing fibroblasts
- anti-tumor effects such as depletion of FAP-expressing CAF, reduction of ECM production in the TME, and inhibition of cancer metastasis.
- the peptide of the present invention When the peptide of the present invention is administered in combination with other anticancer agents, it increases the accumulation of anticancer agents in tumor cells and exhibits a remarkable enhancement of the antitumor effect, so that it can be usefully used as a pan-tumor vaccine.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Genetics & Genomics (AREA)
- Organic Chemistry (AREA)
- Engineering & Computer Science (AREA)
- General Health & Medical Sciences (AREA)
- General Engineering & Computer Science (AREA)
- Biotechnology (AREA)
- Zoology (AREA)
- Biomedical Technology (AREA)
- Wood Science & Technology (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Medicinal Chemistry (AREA)
- Biochemistry (AREA)
- Biophysics (AREA)
- Molecular Biology (AREA)
- Microbiology (AREA)
- Veterinary Medicine (AREA)
- Physics & Mathematics (AREA)
- Plant Pathology (AREA)
- Pharmacology & Pharmacy (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Immunology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- General Chemical & Material Sciences (AREA)
- Epidemiology (AREA)
- Mycology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Oncology (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Peptides Or Proteins (AREA)
Abstract
Description
본 발명은 섬유아세포 활성화 단백질의 면역우세 에피토프 펩타이드 및 이의 용도에 관한 것으로, 보다 구체적으로는 상기 섬유아세포 활성화 단백질의 MHC I 제한 면역우세 에피토프 펩타이드 및 이의 종양, 비알콜성 지방간염(NASH) 또는 섬유화증(fibrosis)과 같은 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용도, 및 암의 전이 억제 용도에 관한 것이다.The present invention relates to an immunodominant epitope peptide of fibroblast activation protein and uses thereof, and more specifically, to an MHC I-restricted immunodominant epitope peptide of the fibroblast activation protein and uses thereof for the prevention or treatment of fibroblast activation protein alpha (FAP)-related diseases such as tumors, nonalcoholic steatohepatitis (NASH) or fibrosis, and uses thereof for inhibiting cancer metastasis.
암 백신 또는 종양 백신은 개인 신생항원 예측 및 검출 기술의 발전에 기반하여 최근 종양의 치료 요법으로 각광받고 있다(Nature Reviews Clinical Oncology 2021, 18 (4), 215-229). 특히, 신생항원(neoantigen)에 기반한 암 백신은 환자 특이적 항 종양 면역을 유도하여, 다양한 임상시험에서 좋은 결과를 나타내고 있다(Nature 2018, 555 (7696), 402-402; Nat Med 2021, 27 (3), 515~). 그러나, 신생항원에 기반한 암 백신은 개인 맞춤형 백신이기 때문에, 환자 개개인에 대한 신생항원 예측부터 백신의 제조까지 비용 및 시간이 많이 소요되어 환자의 부담이 가중된다는 단점이 있다. 이에, 다양한 종류의 암에 적용가능한 범-암 백신(Pan-Cancer Vaccine)의 개발의 필요성이 대두되고 있다. Cancer vaccines or tumor vaccines have recently been spotlighted as a treatment for tumors based on the development of personalized neoantigen prediction and detection technology (Nature Reviews Clinical Oncology 2021, 18 (4), 215-229). In particular, neoantigen-based cancer vaccines induce patient-specific antitumor immunity and have shown good results in various clinical trials (Nature 2018, 555 (7696), 402-402; Nat Med 2021, 27 (3), 515~). However, since neoantigen-based cancer vaccines are personalized vaccines, they require a lot of cost and time from neoantigen prediction for each patient to vaccine manufacturing, which increases the burden on patients. Therefore, the need for the development of a pan-cancer vaccine that can be applied to various types of cancer is emerging.
한편, 종양 미세 환경(Tumor Microenvironment, TME)은 면역 세포, 암-연관 섬유아세포(Cancer-Associated Fibroblasts, CAF), 신호 분자, 혈관 및 세포외 매트릭스(Extracellular Matrix, ECM) 등으로 구성되며, 특히 화학요법 및 면역요법과 같은 기존의 암 치료 요법의 효과를 제한하는 강력한 장벽으로 작용한다(Cell Commun Signal 2020, 18 (1), 59). 특히, TME에 존재하는 암 연관 섬유아세포는 콜라겐, 라미닌 피브로넥틴과 같은 세포외 매트릭스 단백질(ECM)을 분비하여, 종양기질(Tumor Stroma)을 형성하는데 중추적인 역할을 수행한다(Nat Rev Immunol 2015, 15 (11), 669-682; Nat Rev Cancer 2016, 16 (9), 582-598; Nat Rev Clin Oncol 2021, 18 (12), 792-804). 이러한 ECM 단백질은 종양 성장을 위한 구조적 네트워크를 제공하며, 세포간의 신호 및 상호작용을 중재하고, 종양과 주변환경 사이의 물리적 장벽을 형성한다(Signal Transduct Target Ther 2021, 6 (1), 153). 특히, 췌장 선암(Pancreatic Adenocarcinoma, PDAC) 및 대장암과 같은 결합조직형성 종양(Desmoplastic Tumor)에서 형성되는 조밀하고 단단한 ECM 네트워크는 치료 효과를 강력하게 제한시킴으로써, 위와 같은 종양 환자의 낮은 생존율의 주요한 원인으로 보고되었다(Proc Natl Acad Sci U S A 2019, 116 (22), 10674-10680; ACS Nano 2019, 13 (10), 11008-11021). 이러한 관점에서, TME에서 CAF를 억제하거나 고갈시키려는 수많은 시도가 수행되었다. 그러나, 이러한 접근법은 종양 성장의 억제 및 종양 전이의 촉진이라는 복합적 결과를 나타내며, 이러한 결과는 다양한 종양에 존재하는 이질적인 CAF를 무작위로 표적화하는데서 기인하는 것으로 보고된다(Nat Rev Clin Oncol 2021, 18 (12), 792-804; Proc Natl Acad Sci U S A 2019, 116 (22), 10674-10680; Clin Cancer Res 2018, 24 (5), 1190-1201).Meanwhile, the tumor microenvironment (TME) is composed of immune cells, cancer-associated fibroblasts (CAFs), signaling molecules, blood vessels, and the extracellular matrix (ECM), and acts as a powerful barrier that limits the effectiveness of conventional cancer treatments, such as chemotherapy and immunotherapy (Cell Commun Signal 2020, 18 (1), 59). In particular, cancer-associated fibroblasts present in the TME play a pivotal role in forming the tumor stroma by secreting extracellular matrix proteins (ECMs) such as collagen, laminin, and fibronectin (Nat Rev Immunol 2015, 15 (11), 669-682; Nat Rev Cancer 2016, 16 (9), 582-598; Nat Rev Clin Oncol 2021, 18 (12), 792-804). These ECM proteins provide a structural network for tumor growth, mediate signaling and interactions between cells, and form a physical barrier between the tumor and the surrounding environment (Signal Transduct Target Ther 2021, 6 (1), 153). In particular, the dense and rigid ECM network formed in desmoplastic tumors such as pancreatic adenocarcinoma (PDAC) and colorectal cancer has been reported to be a major cause of the poor survival rate of patients with such tumors by strongly limiting the therapeutic effect (Proc Natl Acad Sci U S A 2019, 116 (22), 10674-10680; ACS Nano 2019, 13 (10), 11008-11021). In this regard, numerous attempts have been performed to inhibit or deplete CAFs in the TME. However, these approaches have shown mixed results, including inhibition of tumor growth and promotion of tumor metastasis, which are reported to result from randomly targeting heterogeneous CAFs present in various tumors (Nat Rev Clin Oncol 2021, 18 (12), 792-804; Proc Natl Acad Sci U S A 2019, 116 (22), 10674-10680; Clin Cancer Res 2018, 24 (5), 1190-1201).
섬유아세포 활성화 단백질 알파(Fibroblast Activation Protein alpha, FAP)는 CAF에서 높게 발현되나, 정상 성인 조직에서는 매우 낮은 수준으로 발현되는 막 관통 단백질이며, CAF의 표적화 마커로서 보고되었다(Science 2010, 330 (6005), 827-830). 이러한 관점에서, 소분자 FAP 억제제(J Clin Invest 2009, 119 (12), 3613-3625), FAP 특이적 항체 및 약물 접합체(Clin Cancer Res 2020, 26 (13), 3420-3430), 및 FAP 표적화 CAR-T 요법(Cancer Immunol Res 2014, 2 (2), 154-166)과 같은 FAP 발현 CAF의 기능을 억제하는 치료 요법이 개발되었으며, 이들은 생체 내에서 특정 유형의 종양 퇴행을 촉진하여, FAP 치료 전략이 유의미함을 입증하였다(Front Biosci-Landmrk 2018, 23, 1933-1968). 그러나, 상기한 것과 같은 FAP 발현 CAF에 단기간으로 작용하는 치료적 접근법 이외에, 장기적인 FAP 특이적 T 세포 반응을 유도할 수 있는 백신은 보고된 바 없다. FAP 암호화 DNA(. Clin Cancer Res 2018, 24 (5), 1190-1201) 또는 수지상 세포에 기반한 FAP 백신이 보고된 바 있으나, 불충분한 치료 효능 및 전신 독성(systemic toxicity)를 나타내어 임상에의 적용에 어려움이 있다(J Exp Med 2013, 210 (6), 1137-1151; J Exp Med 2013, 210 (6), 1125-1135; Cancer Cell 2014, 25 (6), 719-734). 이에, 낮은 독성을 가지면서도 뛰어난 치료효과를 나타내는 신규한 FAP 발현 CAF 표적 암 백신의 개발이 필요한 상황이다.Fibroblast Activation Protein alpha (FAP) is a transmembrane protein that is highly expressed in CAFs but at very low levels in normal adult tissues, and has been reported as a targeting marker for CAFs (Science 2010, 330 (6005), 827-830). In this regard, therapeutic approaches that inhibit the function of FAP-expressing CAFs, such as small molecule FAP inhibitors (J Clin Invest 2009, 119 (12), 3613-3625), FAP-specific antibodies and drug conjugates (Clin Cancer Res 2020, 26 (13), 3420-3430), and FAP-targeting CAR-T therapy (Cancer Immunol Res 2014, 2 (2), 154-166), have been developed and demonstrated that they promote specific types of tumor regression in vivo, suggesting the potential of this FAP therapeutic strategy (Front Biosci-Landmrk 2018, 23, 1933-1968). However, other than the short-term therapeutic approaches targeting FAP-expressing CAFs as described above, no vaccine has been reported that can induce long-term FAP-specific T cell responses. Although FAP vaccines based on FAP-encoding DNA (. Clin Cancer Res 2018, 24 (5), 1190-1201) or dendritic cells have been reported, they have insufficient therapeutic efficacy and systemic toxicity, making them difficult to apply in clinical practice (J Exp Med 2013, 210 (6), 1137-1151; J Exp Med 2013, 210 (6), 1125-1135; Cancer Cell 2014, 25 (6), 719-734). Therefore, there is a need to develop a novel FAP-expressing CAF-targeted cancer vaccine that has low toxicity but excellent therapeutic efficacy.
한편, 비알콜성 지방간염(Non-alcoholic steatohepatitis; NASH) 은 간 지방증과 염증을 특징으로 하는 만성 간질환으로, 섬유증, 간경변 및 간세포암 등으로 진행될 수 있는 심각한 질환이다(Hepatology 77, 1335-1347(2023)). FAP는 간세포 성상세포 및 간 조직내의 기타 섬유아세포에서 발현되며(Nat Commun 13 (2022)), 지질의 주요 조절자인 FGF21을 절단하는 것으로 보고된다. FAP 활성에 의한 FGF21 조절장애는 NASH의 진행에 기여할 수 있는 것으로 보고되나, FAP를 표적으로 하는 NASH 백신은 보고된 바 없다.Meanwhile, non-alcoholic steatohepatitis (NASH) is a chronic liver disease characterized by hepatic steatosis and inflammation, and is a serious disease that can progress to fibrosis, cirrhosis, and hepatocellular carcinoma (Hepatology 77, 1335-1347(2023)). FAP is expressed in hepatocyte stellate cells and other fibroblasts in liver tissue (Nat Commun 13 (2022)) and is reported to cleave FGF21, a major regulator of lipids. Dysregulation of FGF21 by FAP activity is reported to contribute to the progression of NASH, but no NASH vaccine targeting FAP has been reported.
이러한 배경기술 아래에서, 강력한 섬유아세포 활성화 단백질(FAP) 특이적 T 세포 면역 반응을 유도하여 뛰어난 면역 유도 효과를 나타내는 백신을 개발하기 위해 예의 노력한 결과, 본 발명자들은 FAP에 대해 뛰어난 면역 유도 효과를 나타내는 면역우세 에피토프 펩타이드를 도출하였으며, 상기 면역우세 에피토프 펩타이드가, 암, NASH 등과 같은 FAP 관련 질환을 효과적으로 치료할 수 있는 것을 확인하고, 본 발명을 완성하였다. Under these background technologies, the inventors of the present invention have made diligent efforts to develop a vaccine that exhibits excellent immune-inducing effects by inducing a strong fibroblast-activating protein (FAP)-specific T cell immune response, and as a result, have derived an immunodominant epitope peptide that exhibits excellent immune-inducing effects against FAP, and have confirmed that the immunodominant epitope peptide can effectively treat FAP-related diseases such as cancer, NASH, etc., thereby completing the present invention.
본 배경기술 부분에 기재된 상기 정보는 오직 본 발명의 배경에 대한 이해를 향상시키기 위한 것이며, 이에 본 발명이 속하는 기술분야에서 통상의 지식을 가지는 자에게 있어 이미 알려진 선행기술을 형성하는 정보를 포함하지 않을 수 있다.The above information set forth in this Background section is intended solely to enhance understanding of the background of the present invention and may not include information that constitutes prior art already known to a person skilled in the art to which the present invention pertains.
발명의 요약Summary of the invention
본 발명의 목적은 섬유아세포 활성화 단백질에 대한 뛰어난 T 세포 유도 효과를 나타내는 면역우세 에피토프 펩타이드 및 이의 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료 용도를 제공하는데 있다. The purpose of the present invention is to provide an immunodominant epitope peptide exhibiting an excellent T cell inducing effect against fibroblast activation protein alpha (FAP) and its use for the prevention or treatment of diseases related to fibroblast activation protein alpha (FAP).
본 발명의 목적은 상기 펩타이드에 특이적으로 결합하는 제제 및 이의 용도를 제공하는데 있다.The purpose of the present invention is to provide a preparation that specifically binds to the above peptide and a use thereof.
본 발명의 또 다른 목적은 상기 펩타이드를 코딩하는 핵산을 제공하는데 있다.Another object of the present invention is to provide a nucleic acid encoding the peptide.
본 발명의 또 다른 목적은 상기 핵산을 포함하는 재조합 벡터를 제공하는 데 있다.Another object of the present invention is to provide a recombinant vector comprising the nucleic acid.
본 발명의 또 다른 목적은 상기 핵산 또는 재조합 벡터가 도입된 숙주세포를 제공하는 데 있다.Another object of the present invention is to provide a host cell into which the nucleic acid or recombinant vector has been introduced.
상기 목적을 달성하기 위하여, 본 발명은 섬유아세포 활성화 단백질(FAP)의 MHC I 제한 에피토프 펩타이드로, 상기 펩타이드는 8 내지 10개의 아미노산으로 구성된, 펩타이드를 제공한다.To achieve the above purpose, the present invention provides a peptide which is an MHC I-restricted epitope peptide of fibroblast activation protein (FAP), wherein the peptide is composed of 8 to 10 amino acids.
본 발명은 또한, 상기 펩타이드를 포함하는 나노입자를 제공한다.The present invention also provides a nanoparticle comprising the peptide.
본 발명은 또한, 상기 펩타이드 및/또는 나노입자를 유효성분으로 포함하는 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용 백신 조성물을 제공한다.The present invention also provides a vaccine composition for preventing or treating a disease related to fibroblast activation protein alpha (FAP), comprising the peptide and/or nanoparticle as an active ingredient.
본 발명은 또한 상기 펩타이드 및/또는 나노입자를 대상에게 투여하는 단계를 포함하는 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료를 위한 백신접종(vaccination), 예방 및/또는 치료방법을 제공한다.The present invention also provides a vaccination, prevention and/or treatment method for the prevention or treatment of a fibroblast activation protein alpha (FAP)-related disease comprising the step of administering to a subject the peptide and/or nanoparticle.
본 발명은 또한, 상기 펩타이드 및/또는 나노입자의 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용 백신 조성물의 제조를 위한 용도를 제공한다.The present invention also provides a use of the peptide and/or nanoparticle for the manufacture of a vaccine composition for the prevention or treatment of a disease associated with fibroblast activating protein alpha (FAP).
본 발명은 또한, 상기 펩타이드 및/또는 지질 나노입자를 포함하는 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용 약학 조성물을 제공한다.The present invention also provides a pharmaceutical composition for preventing or treating a disease associated with fibroblast activating protein alpha (FAP), comprising the peptide and/or lipid nanoparticle.
본 발명은 또한, 상기 펩타이드를 코딩하는 핵산을 제공한다.The present invention also provides a nucleic acid encoding the peptide.
본 발명은 또한, 상기 핵산을 함유하는 재조합 벡터를 제공한다.The present invention also provides a recombinant vector containing the nucleic acid.
본 발명은 또한, 상기 핵산 또는 재조합 벡터가 도입된 숙주세포를 제공한다.The present invention also provides a host cell into which the nucleic acid or recombinant vector has been introduced.
본 발명은 또한, 상기 숙주세포를 배양하는 단계를 포함하는 펩타이드의 제조방법을 제공한다.The present invention also provides a method for producing a peptide, comprising a step of culturing the host cell.
도 1은 본 발명의 실시예에서의 FAP 면역우세 에피토프 펩타이드의 개발 과정을 개략적으로 나타낸 것이다.Figure 1 schematically illustrates the development process of a FAP immunodominant epitope peptide in an embodiment of the present invention.
도 2는 ICS 분석에서 사용된 유세포 계측의 게이팅 방법을 나타낸 것이다.Figure 2 illustrates the gating method of flow cytometry used in ICS analysis.
도 3은 각각의 프로그램(netMHC, PREDEP, BIMAS)으로 예측된 FAP의 상위 에피토프 펩타이드 서열을 나타낸 것이다.Figure 3 shows the upper epitope peptide sequences of FAP predicted by each program (netMHC, PREDEP, BIMAS).
도 4는 in vivo 스크리닝된 펩타이드 후보의 면역원성 및 항종양 효능을 나타낸 것이다. 도 4(a)는 MHC Class I에 의해 제시된 예측된 FAP 펩타이드를 요약한 것이다. 도 4(b)는 예측된 펩타이드의 면역원성을 평가하기 위한 며역화 일정을 나타낸 것이다(n=3 마우스/그룹). 도 4(c) 및 (d)는 IFN-γ 분비 CD8+T 세포의 대표 유세포 분석 결과(c) 및 IFN-γ를 생산하는 CD8+ T 세포의 비율(d) 을 나타낸 것이다. 대조군과 각 실험군 사이의 통계적 유의성은 two-tailed Student's t-test에 의해 결정되었다. 도 4(e) 및 (f) ELISpot 분석에 의해 결정된 IFN-γ 생성 반점(e) 및 IFN-γ 반점 형성 세포의 평균 수(f)의 대표 이미지를 나타낸 것이다. 대조군과 각 실험군 사이의 통계적 유의성은 two-tailed Student's t-test에 의해 결정되었다. 도 4(g)는 예측된 펩타이드의 항종양 치료 효능을 평가하기 위한 면역화 일정을 나타낸 것이다. E.G7-OVA 종양 접종 9일 후, C57BL/6 마우스를 1주 간격으로 각 펩티드로 2회 면역화하였다(n = 4 마우스/그룹). 도 4(h)는 E.G7-OVA 세포의 평균 종양 성장 곡선(왼쪽) 및 25일째 종양 부피(오른쪽)를 나타낸 것이다. 대조군과 각 실험군 사이의 차이의 통계적 유의성은 two-tailed Student's t-test에 의해 결정되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001). Figure 4 shows the immunogenicity and antitumor efficacy of the peptide candidates screened in vivo. Figure 4(a) summarizes the predicted FAP peptides presented by MHC Class I. Figure 4(b) shows the immunogenicity schedule of the predicted peptides (n=3 mice/group). Figure 4(c) and (d) show the representative flow cytometry results of IFN-γ-secreting CD8+ T cells (c) and the percentage of CD8+ T cells producing IFN-γ (d). The statistical significance between the control group and each experimental group was determined by two-tailed Student's t-test. Figure 4(e) and (f) show representative images of IFN-γ-producing spots (e) and the average number of IFN-γ spot-forming cells (f) determined by ELISpot analysis. The statistical significance between the control group and each experimental group was determined by two-tailed Student's t-test. Figure 4(g) shows the immunization schedule to evaluate the antitumor therapeutic efficacy of the predicted peptides. Nine days after E.G7-OVA tumor inoculation, C57BL/6 mice were immunized twice with each peptide at one-week intervals (n = 4 mice/group). Figure 4(h) shows the mean tumor growth curves of E.G7-OVA cells (left) and tumor volumes on day 25 (right). The statistical significance of the differences between the control group and each experimental group was determined by two-tailed Student's t-test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 5는 스크리닝된 6개 펩타이드의 ICS 분석 결과를 개별적으로 나타낸 것이다. (각 패널은 다음을 나타낸다: Control(미투여); 펩타이드 투여; 펩타이드 및 CFA 동시 투여) 각각의 도면의 상단 패널은 ex-vivo에서 재자극을 수행하지 않았으며, 하단 패널은 ex-vivo에서 펩타이드로 제자극을 한 결과이다.Figure 5 shows the ICS analysis results of the six screened peptides individually. (Each panel represents: Control (unadministered); peptide administration; peptide and CFA co-administration) The upper panel of each figure shows the results of no ex-vivo restimulation, and the lower panel shows the results of ex-vivo restimulation with peptides.
도 6은 ICS 분석과 동일한 방법으로 수행된 ELISpot 분석 결과를 나타낸 것이다.Figure 6 shows the results of ELISpot analysis performed in the same manner as the ICS analysis.
도 7은 종양 부피에 따른 FAP+CAF의 비율을 나타낸 것이다.Figure 7 shows the ratio of FAP+CAF according to tumor volume.
도 8은 확립된 E.G7-OVA 종양 모델에서 FAPPEP-SLNP 나노 백신의 설계 및 항종양 효능을 나타낸 것이다. 도 8(a)는 FAPPEP-SLNP 나노백신의 개략도 및 APC로의 내재화 이후 예상되는 작용 메커니즘을 나타낸 것이다. 도 8(b)는 DLS로 측정한 FAPPEP-SLNP의 유체역학적 직경을 나타낸 것이다. 도 8(c)는 FAPPEP-SLNP의 TEM 이미지로 형태 및 나노스케일의 크기를 나타낸다. 스케일 바=200nm. 도 3(d)는 확립된 E.G7-OVA 종양 모델에 대한 FAPPEP-SLNP 나노백신의 항종양 효능을 평가하기 위한 면역화 일정을 나타낸 것이다. E.G7-OVA 종양 접종 9일 후, C57BL/6 마우스에 각 백신 모달리티로 4일 간격으로 3회 주사하였다(n = 5 마우스/그룹). 도 8(e)는 E.G7-OVA 종양의 평균 성장 곡선(왼쪽) 및 21일째 종양 부피(오른쪽)를 나타낸 것이다. 대조군과 각 실험군 사이의 차이의 통계적 유의성은 two-tailed Student's t-test에 의해 결정되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 8 shows the design and antitumor efficacy of FAPPEP-SLNP nanovaccine in an established E.G7-OVA tumor model. Figure 8(a) shows the schematic diagram of FAPPEP-SLNP nanovaccine and its expected mechanism of action after internalization into APC. Figure 8(b) shows the hydrodynamic diameter of FAPPEP-SLNP measured by DLS. Figure 8(c) shows the TEM image of FAPPEP-SLNP showing the morphology and nanoscale size. Scale bar = 200 nm. Figure 3(d) shows the immunization schedule to evaluate the antitumor efficacy of FAPPEP-SLNP nanovaccine against an established E.G7-OVA tumor model. Nine days after E.G7-OVA tumor inoculation, C57BL/6 mice were injected three times with each vaccine modality at 4-day intervals (n = 5 mice/group). Figure 8(e) shows the average growth curve of E.G7-OVA tumors (left) and tumor volume on day 21 (right). The statistical significance of the differences between the control group and each experimental group was determined by two-tailed Student's t-test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 9는 SLNP의 구성요소중 하나인 DSPE-PEG2000-FAPPAP 접합체의 구조를 개략적으로 나타낸 것이다.Figure 9 schematically illustrates the structure of the DSPE-PEG2000-FAPPAP conjugate, one of the components of SLNP.
도 10은 HPLC(a, b) 및 MALDI-TOF(c, d)를 사용하여 DSPE-PEG2000-FAPPEP의 합성을 확인한 결과를 나타낸 것이다.Figure 10 shows the results of confirming the synthesis of DSPE-PEG2000-FAPPEP using HPLC (a, b) and MALDI-TOF (c, d).
도 11은 DSPE-PEG2000-PDP에 FAPPEP을 접합시키기 위해 도입된 시스테인(Cys) 면역원성에 영향을 미치지 않음을 ICS 분석을 통해 확인한 결과를 나타낸 것이다.Figure 11 shows the results of ICS analysis confirming that cysteine (Cys) introduced to conjugate FAPPEP to DSPE-PEG2000-PDP does not affect immunogenicity.
도 12는 FAPPEP1-SLNP를 투여하여 면역화를 수행한 마우스의 비장과 림프절에서 ICS 분석을 수행하여 FAPPEP1에 특이적인 CD8+ T 세포의 존재를 확인한 결과이다. Figure 12 shows the results of ICS analysis performed on the spleen and lymph nodes of mice immunized with FAPPEP1-SLNP, confirming the presence of CD8+ T cells specific for FAPPEP1.
도 13은 FAPPEP1-H-2Kb를 사용하여 테트라머 분석(tetramer assay)을 수행한 결과를 나타낸 것이다.Figure 13 shows the results of a tetramer assay using FAPPEP1-H-2Kb.
도 14는 FAPPEP-SLNP 나노백신의 TME의 리모델링을 매개하여, 자가면역을 유도하지 않고 항원 특이적 T 세포 면역을 증가시킴을 확인한 결과이다. 도 14(a)는 종양 성장, 마우스 생존, 종양 조직 및 비장 분석을 위한 면역화 일정을 나타낸 것이다. E.G7-OVA 종양 접종 9일 후, C57BL/6 마우스를 각 백신 방식으로 4일 간격으로 3회 면역화하였다. 도 14(b)는 면역화된 마우스에서 종양 성장을 모니터링한 결과를 나타낸 것으로 E.G7-OVA 종양의 평균 성장 곡선(왼쪽) 및 21일째 E.G7-OVA 종양의 평균 부피(오른쪽)를 나타낸다(n = 9 마우스/그룹). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 도 14(c)는 E.G7-OVA 종양 보유 마우스의 생존 곡선을 나타낸 것이다(n = 10 마우스/그룹). 대조군과 각 실험군 간의 생존율 차이의 통계적 유의성은 log-rank (Mantel-Cox) test로 결정하였다. 도 14(d)는 Masson’s trichrome 염색을 사용하여 종양 조직의 콜라겐 침착을 평가한 결과로, 각 그룹의 Masson’s trichrome 염색 조직의 대표 이미지를 나타낸 것이다. 청색 염색은 콜라겐; 흑색 염색은 핵; 빨간색 백그라운드는 세포질 및 근육; 검은색 점선은 콜라겐 침착을 나타낸다. 축척 막대 = 100μm. 도 14(e)는 각 종양 그룹의 섹션에서 3 개의 무작위 필드에서 콜라겐 염색 영역의 백분율을 나타낸 것이다(n = 각 염색에 대해 3 마리의 마우스에서 종양 당 3 섹션). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 도 14(f) 및 (g)는 세포 수준에서 종양 조직 분석을 수행한 결과(n = 4 마우스/그룹)로, 유세포 계측법에 의해 결정된 종양에서의 FAP+ CAF의 비율(f) 및 CD8+ TIL의 비율(g)을 나타낸다. 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 도 14(h) 및 (i)는 항원 특이적 T 세포 반응을 평가하기 위한 비장 세포 분석결과를 나타낸 것이다(n = 4 마우스/그룹). ICS에 의해 결정된 IFN-γ 분비 CD8+ T 세포(h) 및 CD4+ T 세포(i)의 비율을 나타낸다. 통계적 유의성은 two-tailed Student's t-test에 의해 결정되었다. 도 4(j)는 FAPPEP1-SLNP로 면역화한 후 자가면역 유도 여부를 확인하기 위한 비장 세포 분석결과를 나타낸 것이다(n = 4 마우스/그룹). IL-17A-분비 CD4+ T 세포의 비율은 ICS에 의해 결정되었다. 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 14 shows the results confirming that FAPPEP-SLNP nanovaccine increases antigen-specific T cell immunity without inducing autoimmunity by mediating TME remodeling. Figure 14(a) shows the immunization schedule for tumor growth, mouse survival, tumor tissue, and spleen analysis. Nine days after E.G7-OVA tumor inoculation, C57BL/6 mice were immunized three times with each vaccine method at 4-day intervals. Figure 14(b) shows the results of monitoring tumor growth in immunized mice, showing the average growth curve of E.G7-OVA tumors (left) and the average volume of E.G7-OVA tumors on day 21 (right) (n = 9 mice/group). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. Figure 14(c) shows the survival curve of E.G7-OVA tumor-bearing mice (n = 10 mice/group). The statistical significance of the survival difference between the control group and each experimental group was determined by the log-rank (Mantel-Cox) test. Figure 14(d) shows the representative images of Masson’s trichrome-stained tissues from each group as a result of evaluating collagen deposition in tumor tissues using Masson’s trichrome staining. Blue staining indicates collagen; black staining indicates nuclei; red background indicates cytoplasm and muscle; and black dotted lines indicate collagen deposition. Scale bar = 100 μm. Figure 14(e) shows the percentage of collagen-stained areas in three random fields from sections from each tumor group (n = 3 sections per tumor from 3 mice for each staining). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. Figure 14(f) and (g) show the results of tumor tissue analysis at the cellular level (n = 4 mice/group), showing the percentage of FAP+ CAFs (f) and CD8+ TILs (g) in tumors as determined by flow cytometry. Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. Figure 14(h) and (i) show the results of spleen cell analysis to evaluate antigen-specific T cell responses (n = 4 mice/group). The percentages of IFN-γ-secreting CD8+ T cells (h) and CD4+ T cells (i) determined by ICS are shown. Statistical significance was determined by two-tailed Student's t-test. Figure 4(j) shows the results of spleen cell analysis to determine whether autoimmunity was induced after immunization with FAPPEP1-SLNP (n = 4 mice/group). The percentages of IL-17A-secreting CD4+ T cells were determined by ICS. Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 15는 종양에서 혈관의 분포를 확인한 결과(a 및 b) 및 폐에서의 전이를 확인한 결과(c 및 d)를 나타낸 것이다. 도 15(a) 및 (b)에서 혈관은 갈색으로 표시되며, 도 15(c) 및 (d)에서 전이된 세포는 붉은색으로 표시된다.Figure 15 shows the results of confirming the distribution of blood vessels in a tumor (a and b) and the results of confirming metastasis in the lung (c and d). In Figures 15(a) and (b), blood vessels are shown in brown, and in Figures 15(c) and (d), metastatic cells are shown in red.
도 16은 다양한 종류의 tumor (E.G7-OVA, Panc02, MC38)를 single cell로 dissociation 하여 flow cytometry analysis로 종양을 이루는 세포들 중 FAP+ CAF의 비율을 분석한 것이다.Figure 16 shows the analysis of the ratio of FAP+ CAF among the cells forming the tumor by flow cytometry analysis of single cells dissociated from various types of tumors (E.G7-OVA, Panc02, MC38).
도 17은 MC38 tumor에서 CAF 외에 cancer cell에서도 FAP를 발현하는지 flow cytometry analysis로 확인한 결과이다.Figure 17 shows the results of flow cytometry analysis to confirm whether FAP is expressed in cancer cells other than CAFs in MC38 tumors.
도 18은 결합조직성 종양인 Panc02와 MC38 종양에서 FAPPEP1-SLNP의 단독요법으로 치료 효능를 보이는지 확인한 실험이다. Panc02 모델에서의 실험 방법 (a)과 종양 성장 (b), Kaplan-Meier 생존 분석 결과(c), 그리고 MC38 model에서의 실험 방법 (d)과 종양성장 (e), Kaplan-Meier 생존분석 결과(f)이다.Figure 18 is an experiment to confirm whether FAPPEP1-SLNP monotherapy shows therapeutic efficacy in Panc02 and MC38 tumors, which are connective tissue tumors. Experimental method (a) and tumor growth (b) in the Panc02 model, and Kaplan-Meier survival analysis result (c), and experimental method (d) and tumor growth (e) and Kaplan-Meier survival analysis result (f) in the MC38 model.
도 19는 MC38 tumor model에서 종양 안에 있는 CD8+ T cell (a) 과 CD4+ T cell (b) 이 FAP에 특이적인 면역활성을 갖는지 확인하기 위하여 ICS assay를 진행한 결과이다.Figure 19 shows the results of an ICS assay performed to confirm whether CD8+ T cells (a) and CD4+ T cells (b) within the tumor in the MC38 tumor model have immune activity specific to FAP.
도 20은 큰 사이즈의 종양에서도 FAPPEP1-SLNP가 치료 효능을 나타내는지 확인한 결과이다. Figure 20 shows the results confirming whether FAPPEP1-SLNP exhibits therapeutic efficacy even in large-sized tumors.
도 21은 FAPPEP1-SLNP 나노백신 및 Dox와의 병용 치료 후, ECM이 풍부한 MC38 종양의 사멸효과를 나타낸 것이다. (a) FAPPEP1-SLNP 면역화 후 MC38 종양에서 근적외선 염료 사이페이트(near-infrared dye cypate)의 침투를 확인한 결과이다. 마우스를 FAPPEP1-SLNP 나노백신으로 면역화하고 소분자 Dox에 대한 프록시로서 근적외선 염료 사이페이트를 투여하였다. 상이한 그룹의 종양으로의 침투를 IVIS를 사용하여 비교하였다(n = 5 마우스/그룹). 왼쪽 패널은 IVIS에 의해 획득된 염료의 종양 내 침투 및 종양에 분포된 염료의 상대적 형광 신호 강도의 대표적인 이미지를 나타낸 것이다. 오른쪽 패널의 데이터는 사이페이트 투여 마우스/사이페이트 비투여 마우스의 종양의 형광 강도 비율을 나타낸 것이다. 도 21(b)는 확립된 MC38 종양 모델에 대한 FAPPEP1-SLNP 나노백신 및 Dox와의 병용 요법을 위한 면역화 일정을 나타낸 것이다. MC38 종양 접종 5일 후, C57BL/6 마우스는 FAPPEP1-SLNP 나노백신으로 4일 간격으로 3회 면역화되었으며, Dox로 처리 또는 비처리되었다(n = 7 마우스/그룹). 도 21(c)는 마우스에서 MC38 종양 성장(왼쪽) 및 17일째 종양 부피(오른쪽)를 나타낸 것이다. 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 도 21(d) 및 (e)는 세포 수준에서 MC38 종양 조직 분석을 나타낸 것이다(n = 4 마우스/그룹). 유세포 계측법에 의해 결정된 종양에서 FAP+ CAF의 비율(d) 및 CD8+ TIL의 비율(e)을 나타낸다. 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 21 shows the killing effect of ECM-rich MC38 tumors after combination treatment with FAPPEP1-SLNP nanovaccine and Dox. (a) Penetration of near-infrared dye cypate into MC38 tumors after FAPPEP1-SLNP immunization. Mice were immunized with FAPPEP1-SLNP nanovaccine and administered near-infrared dye cypate as a proxy for small molecule Dox. Penetration into tumors of different groups was compared using IVIS (n = 5 mice/group). The left panels show representative images of intratumoral dye penetration and relative fluorescence signal intensity of the dye distributed in the tumors acquired by IVIS. The data in the right panels show the ratio of fluorescence intensities of tumors of cyphate-treated mice/cyphate-nontreated mice. Figure 21(b) shows the immunization schedule for combination therapy with FAPPEP1-SLNP nanovaccines and Dox for the established MC38 tumor model. Five days after MC38 tumor inoculation, C57BL/6 mice were immunized three times at 4-day intervals with FAPPEP1-SLNP nanovaccines and treated or not with Dox (n = 7 mice/group). Figure 21(c) shows the MC38 tumor growth in mice (left) and tumor volume on day 17 (right). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. Figure 21(d) and (e) show MC38 tumor tissue analysis at the cellular level (n = 4 mice/group). The percentage of FAP+ CAFs (d) and the percentage of CD8+ TILs (e) in the tumors determined by flow cytometry are shown. Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are presented as mean ± SEM (*P < 0.05, **P < 0.01, ***P < 0.001).
도 22는 FAPPEP1-SLNP 나노백신이 CAF를 표적으로 하여 종양을 사멸시키는 메커니즘을 개략적으로 나타낸 것이다. Figure 22 schematically illustrates the mechanism by which FAPPEP1-SLNP nanovaccine targets CAFs to kill tumors.
도 23은 FAPPEP1 펩타이드 백신 또는 FAPPEP1-SLNP 나노백신의 CAF를 표적으로 하는 종양의 전이 억제 모델의 메커니즘(상단) 및 본 발명의 실시예에서 사용된 암의 전이에 대한 FAPPEP1 펩타이드 백신의 치료 효능 평가 모델(하단)을 개략적으로 나타낸 것이다.Figure 23 schematically illustrates the mechanism of a tumor metastasis inhibition model targeting CAF of FAPPEP1 peptide vaccine or FAPPEP1-SLNP nanovaccine (top) and the therapeutic efficacy evaluation model of FAPPEP1 peptide vaccine on cancer metastasis used in the examples of the present invention (bottom).
도 24는 FAPPEP1 펩타이드 백신의 폐로의 암 전이를 억제하는 결과를 보여주는 폐 조직의 조직학적 분석 결과이다. 도 24a는 H&E로 염색된 폐조직의 이미지이다. 빨간색 화살표는 폐로의 암 전이 영역을 나타낸다. 스케일바 = 50 nm. 도 24b는 각 폐 그룹의 섹션 내 무작위 3개 부위(field)의 암 전이 영역 비율을 나타낸 것이다. (각 염색당 5마리 마우스로부터의 폐당 3개의 섹션 n = 3). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 24 is a histological analysis of lung tissue showing the results of inhibiting cancer metastasis to the lung by the FAPPEP1 peptide vaccine. Figure 24a is an image of lung tissue stained with H&E. The red arrow indicates the area of cancer metastasis to the lung. Scale bar = 50 nm. Figure 24b shows the percentage of cancer metastasis area in three random fields within the sections of each lung group. (n = 3 for three sections per lung from five mice for each staining). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 25는 FAPPEP1 펩타이드 백신의 간으로의 암 전이를 억제하는 결과를 보여주는 간 조직의 조직학적 분석 결과이다. 도 25a는 H&E로 염색된 간조직의 이미지이다. 빨간색 화살표는 폐로의 암 전이 영역을 나타낸다. 스케일바 = 50 nm. (b) 도 24b는 각 간 그룹의 섹션 내 무작위 3개 부위(field)의 암 전이 영역 비율을 나타낸 것이다 (각 염색당 5마리 마우스로부터의 간당 3개의 섹션 n = 3). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 25 is a histological analysis of liver tissue showing the results of FAPPEP1 peptide vaccine inhibition of cancer metastasis to the liver. Figure 25a is an image of liver tissue stained with H&E. The red arrow indicates the area of cancer metastasis to the lung. Scale bar = 50 nm. (b) Figure 24b shows the percentage of cancer metastasis area in three random fields within the sections of each liver group (n = 3 for three sections per liver from five mice for each staining). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 26은 FAPPEP1 펩타이드 백신 또는 FAPPEP1-SLNP 나노백신의 FAP 발현 세포를 표적화 및 FGF21 분해 방지를 통한 NASH 치료 메커니즘을 개략적으로 나타낸 것이다. Figure 26 schematically illustrates the NASH treatment mechanism through targeting FAP expressing cells and preventing FGF21 degradation by FAPPEP1 peptide vaccine or FAPPEP1-SLNP nanovaccine.
도 27은 NASH 모델에 대한 FAPPEP1 펩타이드 백신의 치료효능을 나타내는 결과이다. 도 27은 본 발명의 실시예에서 수행된 NASH 확립 및 치료 효과 확인 모델의 실험 방법을 개략적으로 나타낸다. MCD/MCS 식이요법 4주 후 C51BL/6 마우스에 FAPPEP1 펩타이드 백신을 사용하여 일주일 간격으로 2회 면역화를 수행하였다. (n = 5 mice/group). 도 27b는 마우스의 체중 모니터링(왼쪽) 및 45일에서의 마우스 체중(오른쪽)을 나타낸 것이다. 도 27c는 45일에서의 마우스의 간 무게를 나타낸 것이다. 도 27d는 45일에서의 간 대 체중의 비율을 나타낸 것이다. 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001). (Illustration created with BioRender.com.)Figure 27 shows the results showing the therapeutic efficacy of the FAPPEP1 peptide vaccine for the NASH model. Figure 27 schematically shows the experimental method of the NASH establishment and therapeutic effect confirmation model performed in the example of the present invention. After 4 weeks of MCD/MCS diet therapy, C51BL/6 mice were immunized twice with the FAPPEP1 peptide vaccine at one-week intervals. (n = 5 mice/group). Figure 27b shows the body weight monitoring of the mice (left) and the body weight of the mice at 45 days (right). Figure 27c shows the liver weight of the mice at 45 days. Figure 27d shows the liver-to-body weight ratio at 45 days. Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as the mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001). (Illustration created with BioRender.com.)
도 28은 NASH 모델을 FAPPEP1 펩타이드 백신으로 면역화 후 마우스의 혈청을 분석한 결과이다. 마우스 혈청은 45일에 수집되었으며 다음과 같은 같은 손상 관련 마커의 수준을 분석하였다: ALT (a), AST (b), 총 빌리루빈(c), 및 HDL (d). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 28 shows the results of serum analysis of mice after immunization with FAPPEP1 peptide vaccine in NASH model. Mouse sera were collected on
도 29는 FAPPEP1 펩타이드 백신으로 면역화 후 Oil red O 염색을 사용하여 간 조직에서의 지질 침착을 평가한 결과이다. 도 29는 각 그룹의 Oil red O 염색 조직의 이미지를 나타낸 것이다. 붉은색 염색 부위는 콜라겐; 검은색 염색 부위는 핵; 빨간색 배경 염색은 세포질 및 근육; 흰색 화살표는 지질 침착을 나타낸다. 스케일바 = 50 μm. 도 29b는 각 간 그룹의 섹션 내 무작위 3개 부위(field)의 지질 염색 영역 비율을 나타낸 것이다 (각 염색당 5마리 마우스로부터의 간당 3개의 섹션 n = 3). Percentage of lipid-stained area in three random fields in sections from each liver group (n = 3 sections per tumor from 5 mice for each stain). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 29 shows the results of evaluating lipid deposition in liver tissue using Oil red O staining after immunization with FAPPEP1 peptide vaccine. Figure 29 shows the images of Oil red O-stained tissues in each group. Red staining area is collagen; black staining area is nucleus; red background staining is cytoplasm and muscle; white arrows indicate lipid deposition. Scale bar = 50 μm. Figure 29b shows the percentage of lipid-stained area in three random fields in sections from each liver group (n = 3 sections per tumor from 5 mice for each stain). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
도 30은 H&E 염색을 사용하여 간조직의 팽창 간세포(ballooning hepatocyte)를 평가한 결과이다. 도 30a는 H&E로 염색된 간 조직의 조직학적 이미지를 나타낸 것이다. 빈 영역은 간 조직의 지방세포 팽창을 나타낸다. 스케일 바 = 100 nm. 도 30b는 각 간 그룹의 섹션 내 무작위 3개 부위(field)의 지방세포 팽창 영역 비율을 나타낸 것이다 (각 염색당 5마리 마우스로부터의 간당 3개의 섹션 n = 3). 통계적 유의성은 post hoc Tukey’s test를 사용한 one-way ANOVA로 계산되었다. 모든 데이터는 평균 ± SEM으로 표시된다. (*P < 0.05, **P < 0.01, ***P < 0.001).Figure 30 shows the results of evaluating ballooning hepatocytes in liver tissue using H&E staining. Figure 30a shows a histological image of liver tissue stained with H&E. The blank area represents ballooning adipocytes in the liver tissue. Scale bar = 100 nm. Figure 30b shows the percentage of ballooning adipocyte areas in three random fields within the sections of each liver group (n = 3 for three sections per liver from five mice for each staining). Statistical significance was calculated by one-way ANOVA with post hoc Tukey’s test. All data are expressed as mean ± SEM. (*P < 0.05, **P < 0.01, ***P < 0.001).
발명의 상세한 설명 및 바람직한 구현예Detailed description of the invention and preferred embodiments
이하, 본 발명을 상세히 설명하기로 한다. 다만, 하기 상세한 설명은 본 발명에 대한 예시로 제시되는 것으로, 이에 의해 본 발명이 제한되지는 않으며 본 발명은 후술하는 특허청구범위의 기재 및 그로부터 해석되는 균등 범주 내에서 다양한 변형 및 응용이 가능하다. Hereinafter, the present invention will be described in detail. However, the following detailed description is provided as an example of the present invention, and the present invention is not limited thereby, and the present invention can be variously modified and applied within the scope of the following claims and equivalents interpreted therefrom.
달리 지시되지 않는 한, 핵산 및 아미노산은 좌측에서 우측으로 각각 5'에서 3' 및 N-말단에서 C-말단 배향으로 기록된다. 명세서 내에서 열거된 수치 범위는 범위를 정의하는 숫자를 포함하고, 정의된 범위 내의 각각의 정수 또는 임의의 비-정수 분획을 포함한다.Unless otherwise indicated, nucleic acids and amino acids are listed in 5' to 3' and N-terminal to C-terminal orientations, respectively, from left to right. Numerical ranges listed within the specification are inclusive of the numbers defining the range and include each integer or any non-integer fraction within the defined range.
달리 정의되지 않는 한, 본원에서 사용된 모든 기술적 및 과학적 용어는 본 발명이 속하는 분야의 당업자가 통상적으로 이해하는 것과 동일한 의미를 갖는다. 본원에 기술된 것들과 유사하거나 등가인 임의의 방법 및 재료가 본 발명을 테스트하기 위한 실행에서 사용될 수 있지만, 바람직한 재료 및 방법이 본원에서 기술된다.Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any methods and materials similar or equivalent to those described herein can be used in the practice of testing the present invention, the preferred materials and methods are described herein.
본 발명 명세서에서 언급되는 아미노산 서열은, 특정 아미노산 잔기 위치에서 아미노산 잔기가 보존적으로 치환된 변이체 또는 이의 절편들도 포함하는 의미로 해석된다.The amino acid sequence referred to in the present invention is interpreted to include variants or fragments thereof in which amino acid residues are conservatively substituted at specific amino acid residue positions.
본 명세서에서 “보존적 치환”이란 1개 이상의 아미노산을 해당 단백질의 생물학적 또는 생화학적 기능의 손실을 야기하지 않는 유사한 생화학적 특성을 갖는 아미노산으로 치환하는 것을 포함하는 변형을 의미한다.As used herein, “conservative substitution” means a modification that includes replacing one or more amino acids with amino acids having similar biochemical properties that do not result in loss of biological or biochemical function of the protein.
“보존적 아미노산 치환”은 아미노산 잔기를 유사한 측쇄를 갖는 아미노산 잔기로의 치환을 의미한다. 예를 들어, 유사한 측쇄를 갖는 아미노산 잔기 부류는 해당 기술분야에 잘 알려져 있다. 이들 부류는 염기성 측쇄를 갖는 아미노산(예를 들어, 라이신, 아르기닌, 히스티딘), 산성 측쇄를 갖는 아미노산(예를 들어, 아스파르트산, 글루탐산), 대전되지 않은 극성 측쇄를 갖는 아미노산(예를 들어, 글리신, 아스파라진, 글루타민, 세린, 트레오닌, 티로신, 시스테인), 비-극성 측쇄를 갖는 아미노산(예를 들어, 알라닌, 발린, 류신, 이소류신, 프롤린, 페닐알라닌, 메티오닌, 트립토판), 베타-분지된 측쇄를 갖는 아미노산(예를 들어, 트레오닌, 발린, 이소류신) 및 방향족 측쇄를 갖는 아미노산(예를 들어, 티로신, 페닐알라닌, 트립토판, 히스티딘)을 포함한다.A “conservative amino acid substitution” refers to the substitution of an amino acid residue with an amino acid residue having a similar side chain. For example, classes of amino acid residues having similar side chains are well known in the art. These classes include amino acids having basic side chains (e.g., lysine, arginine, histidine), amino acids having acidic side chains (e.g., aspartic acid, glutamic acid), amino acids having uncharged polar side chains (e.g., glycine, asparagine, glutamine, serine, threonine, tyrosine, cysteine), amino acids having non-polar side chains (e.g., alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), amino acids having beta-branched side chains (e.g., threonine, valine, isoleucine), and amino acids having aromatic side chains (e.g., tyrosine, phenylalanine, tryptophan, histidine).
섬유아세포 활성화 단백질 알파(Fibroblast Activation Protein, FAP)은 프롤릴 엔도펩티다아제 FAP로도 알려져 있다. 일반적으로 성인 조직에서 FAP의 발현이 매우 낮은 것으로 보고되나, FAP는 암, 섬유증, NASH 등의 다양한 질환에서 질환의 발병 및 진행에 관여하는 주요 인자로 알려져 있다. 이에, FAP를 표적으로하는 다양한 요법이 연구 및 보고되었으나, 장기적인 면역반응을 유도할 수 있는 FAP 표적화 요법은 거의 보고된 바 없으며, 일부 보고된 FAP 표적 백신 요법 또한 전신성 독성 또는 충분치 않은 효능으로 새로운 FAP 표적화 백신 치료제의 개발이 요구된다. Fibroblast Activation Protein alpha (FAP) is also known as prolyl endopeptidase FAP. Although the expression of FAP is generally reported to be very low in adult tissues, FAP is known to be a major factor involved in the onset and progression of various diseases such as cancer, fibrosis, and NASH. Accordingly, various therapies targeting FAP have been studied and reported, but FAP-targeting therapies that can induce long-term immune responses have rarely been reported, and some reported FAP-targeting vaccine therapies also have systemic toxicity or insufficient efficacy, requiring the development of new FAP-targeting vaccine therapeutics.
본 발명의 일 실시예에서, in silico 분석을 통해 8-mer 내지 10-mer의 섬유아세포 활성화 단백질(FAP)의 MHC(Major histocompatibility complex) I 제한 에피토프 펩타이드를 스크리닝하였으며, 이러한 에피토프 펩타이드가 뛰어난 FAP 특이적 T 세포의 생성을 유도하고, 종양의 치료, 전이 억제, NASH의 치료에 있어서 뛰어난 효과를 나타낼 수 있음을 확인하였다.In one embodiment of the present invention, 8-mer to 10-mer MHC (Major histocompatibility complex) I-restricted epitope peptides of fibroblast activation protein (FAP) were screened through in silico analysis, and it was confirmed that these epitope peptides can induce the production of excellent FAP-specific T cells and exhibit excellent effects in the treatment of tumors, inhibition of metastasis, and treatment of NASH.
따라서, 본 발명은 일 관점에서, 섬유아세포 활성화 단백질(FAP)의 면역우성 에피토프 펩타이드에 관한 것이다. Accordingly, the present invention relates, in one aspect, to an immunodominant epitope peptide of fibroblast activation protein (FAP).
본 발명의 용어, “면역우성(Immunodominance)”은 특정 단백질 또는 폴리펩타이드로부터 유래할 수 있는 다양한 항원성 펩타이드 중, 일부 소수의 항원성 펩타이드에 대해서만 면역 반응이 유도되는 면역학적 현상을 의미한다(Clinical Immunology. 143 (2): 99-115). “면역 우성”은 항체 매개성 면역과 세포 매개성 면역 모두에 대해 명확하게 나타난다. 면역우성의 효과는 면역우세(Immunodomination)에 의해 발생한다. 일반적으로 병원체로부터 생성될 수 있는 수백 내지 수천개의 펩타이드 항원의 면역원성의 차이는 우성 계층(dominance hierarchy)을 나타내며, 강력한 면역 반응을 자극하는 항원이 “면역우세 에피토프” 또는 “면역우세 항원”으로 간주된다.The term “immunodominance” of the present invention refers to an immunological phenomenon in which an immune response is induced only to a small number of antigenic peptides among various antigenic peptides that can be derived from a specific protein or polypeptide (Clinical Immunology. 143 (2): 99-115). “Immunodominance” is clearly observed in both antibody-mediated immunity and cell-mediated immunity. The effect of immunodominance is caused by immunodominance. In general, the difference in immunogenicity of hundreds to thousands of peptide antigens that can be produced from a pathogen represents a dominance hierarchy, and an antigen that stimulates a strong immune response is considered an “immunodominant epitope” or “immunodominant antigen.”
본 발명에 있어서, 상기 펩타이드는 7 내지 11개, 바람직하게는 8 내지 10개의 아미노산으로 구성된 것을 특징으로 할 수 있다.In the present invention, the peptide may be characterized as being composed of 7 to 11 amino acids, preferably 8 to 10 amino acids.
본 발명에 있어서, 상기 펩타이드는 섬유아세포 활성화 단백질(FAP)의 MHC I 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다.In the present invention, the peptide may be characterized as being an MHC I-restricted epitope peptide of fibroblast activation protein (FAP).
MHC(Major histocompatibility complex)는 항원을 세포 표면에 제시하는 복합체로, MHC가 제시하는 에피토프에 대한 면역반응을 유도한다. MHC는 크게 I형과 II형으로 분류될 수 있으며, I형 MHC의 경우 CD8+ T 세포 반응을 유도하고, II형 MHC의 경우 APC를 통해 B 세포 또는 helper T 세포 등에 제시하여 면역반응을 유도한다.MHC (Major histocompatibility complex) is a complex that presents antigens on the cell surface and induces an immune response to the epitopes presented by MHC. MHC can be largely classified into type I and type II. Type I MHC induces a CD8+ T cell response, while type II MHC induces an immune response by presenting it to B cells or helper T cells through APC.
본 발명에 있어서, 상기 펩타이드는 인간 백혈구 항원(Human leukocyte antigen; HLA) 제한 에피토프인 것을 특징으로 할 수 있다. In the present invention, the peptide may be characterized as being a human leukocyte antigen (HLA) restricted epitope.
인간 백혈구 항원(HLA)는 인간 MHC의 주요한 유전자 좌위로서 세포가 제시하는 MHC가 자가(self) 또는 비자가(non-Self)인지를 구분하게 하는 항원 부위를 의미한다. HLA는 인간 염색체 내에서 위치에 따라 다양한 패밀리로 분류된다. 예를들어, MHC I의 HLA는 HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA-G 등으로 분류되며, MHC II의 HLA는 HLA-DP, HLA-DQ, HLA-DR 등으로 분류될 수 있고, 각각의 패밀리 그룹은 보다 세부적으로 분류될 수 있으나, 이에 제한되는 것은 아니다.Human leukocyte antigen (HLA) is a major gene locus of human MHC and refers to an antigenic site that distinguishes whether the MHC presented by a cell is self or non-self. HLA is classified into various families according to its location within the human chromosome. For example, HLA of MHC I is classified into HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, HLA-G, etc., and HLA of MHC II can be classified into HLA-DP, HLA-DQ, HLA-DR, etc., and each family group can be classified in more detail, but is not limited thereto.
본 발명에 있어서, 상기 펩타이드는 HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, 및 HLA-G 중 어느 하나 이상의 유형의 HLA에 제한 에피토프 펩타이드일 수 있으며, 보다 바람직하게는 HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8 및 HLA-B27 중 어느 하나 이상 유형의 HLA에 제한 에피토프 펩타이드일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the peptide may be an epitope peptide restricted to one or more types of HLA among HLA-A, HLA-B, HLA-C, HLA-E, HLA-F, and HLA-G, and more preferably, may be an epitope peptide restricted to one or more types of HLA among HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8, and HLA-B27, but is not limited thereto.
본 발명의 용어, 특정 유형의 “MHC 제한(되는)(restricted) 에피토프” 또는 “HLA 제한(되는)(restricted) 에피토프”는 해당 항원이 특정 유형의 MHC 또는 HLA에 의해 결합되어 제시된 경우에 면역세포의 반응을 유도할 수 있음을 의미한다. 에피토프 펩타이드의 제한성은 다른 유형의 MHC 또는 HLA에 대해 배타적(하나의 유형에만 제한) 또는 비-배타적(하나 이상의 유형에 지한)일 수 있다. 에피토프 펩타이드의 이러한 제한(restriction)은 환자의 HLA 타입 분석에 따라 보다 효과적인 면역화를 가능하게 할 뿐만 아니라, 의도하지 않은 과도한 면역반응의 유도로 인한 독성 및 부작용을 방지할 수 있다.The term "MHC restricted epitope" or "HLA restricted epitope" of the present invention means that the antigen can induce an immune cell response when presented by a specific type of MHC or HLA. The restriction of the epitope peptide can be exclusive (restricted to only one type) or non-exclusive (restricted to more than one type) with respect to other types of MHC or HLA. Such restriction of the epitope peptide not only allows more effective immunization depending on the patient's HLA typing, but also prevents toxicity and side effects due to induction of unintended excessive immune response.
본 발명에 있어서, 상기 펩타이드는 하기 표 1 내지 표 5의 서열번호 1 내지 149로 구성된 군에서 선택되는 아미노산 서열을 포함하는 것을 특징으로 할 수 있다.In the present invention, the peptide may be characterized by including an amino acid sequence selected from the group consisting of sequence numbers 1 to 149 of Tables 1 to 5 below.
본 발명에 있어서, 상기 펩타이드는 하기 표 1 내지 표 5의 서열번호 1 내지 149로 구성된 군에서 선택되는 아미노산 서열로 구성되는 것을 특징으로 할 수 있다.In the present invention, the peptide may be characterized by being composed of an amino acid sequence selected from the group consisting of sequence numbers 1 to 149 of Tables 1 to 5 below.
본 발명에 있어서, 상기 펩타이드는 HLA-A1에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-A1에 제한 에피토프 펩타이드는 서열번호 10 내지 29로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-A1. For example, the epitope peptide restricted to HLA-A1 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
본 발명에 있어서, 상기 펩타이드는 HLA-A2에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-A2에 제한 에피토프 펩타이드는 서열번호 1, 3, 4, 5, 및 30 내지 45로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-A2. For example, the epitope peptide restricted to HLA-A2 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, 4, 5, and 30 to 45.
본 발명에 있어서, 상기 펩타이드는 HLA-A3에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-A3에 제한 에피토프 펩타이드는 서열번호 46 내지 65로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-A3. For example, the epitope peptide restricted to HLA-A3 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
본 발명에 있어서, 상기 펩타이드는 HLA-A24에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-A24에 제한 에피토프 펩타이드는 서열번호 66 내지 85로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-A24. For example, the epitope peptide restricted to HLA-A24 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
본 발명에 있어서, 상기 펩타이드는 HLA-A26에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-A26에 제한 에피토프 펩타이드는 서열번호 11, 12, 13, 14, 19, 21, 28, 58 및 86 내지 97로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-A26. For example, the epitope peptide restricted to HLA-A26 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97.
본 발명에 있어서, 상기 펩타이드는 HLA-B7에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-B7에 제한 에피토프 펩타이드는 서열번호 30, 93 및 98 내지 115로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-B7. For example, the epitope peptide restricted to HLA-B7 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
본 발명에 있어서, 상기 펩타이드는 HLA-B8에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-B8에 제한 에피토프 펩타이드는 서열번호 30, 91, 93, 99, 100, 103 및 116 내지 129로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-B8. For example, the epitope peptide restricted to HLA-B8 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129.
본 발명에 있어서, 상기 펩타이드는 HLA-B27에 제한 에피토프 펩타이드인 것을 특징으로 할 수 있다. 예를 들어, HLA-B27에 제한 에피토프 펩타이드는 서열번호 130 내지 149로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may be characterized as being an epitope peptide restricted to HLA-B27. For example, the epitope peptide restricted to HLA-B27 may include or be composed of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149.
본 발명에 있어서, 상기 펩타이드는 바람직하게는 서열번호 1, 2, 3, 4 또는 5,보다 바람직하게는 서열번호 1의 아미노산 서열을 포함하거나 구성될 수 있다.In the present invention, the peptide may preferably comprise or consist of an amino acid sequence of SEQ ID NO: 1, 2, 3, 4 or 5, more preferably SEQ ID NO: 1.
본 발명에 있어서, 상기 펩타이드는 섬유아세포 활성화 단백질(FAP) 특이적인 면역반응을 유도하는 것을 특징으로 할 수 있다.In the present invention, the peptide may be characterized by inducing an immune response specific to fibroblast activation protein (FAP).
본 발명에 있어서, 상기 펩타이드는 유리 FAP 또는 FAP 발현 세포에 대한 세포독성 T 세포 반응 또는 CD8+ T 세포 반응을 자극하는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the peptide may be characterized by, but is not limited to, stimulating a cytotoxic T cell response or a CD8+ T cell response against free FAP or FAP expressing cells.
본 발명에 있어서, 상기 FAP 발현 세포는 FAP 발현 섬유아세포인 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the FAP expressing cell may be characterized as being a FAP expressing fibroblast, but is not limited thereto.
본 발명에 있어서, 상기 펩타이드는 N’ 말단 및/또는 C’ 말단에 각각 독립적으로 아미노산을 추가로 포함할 수 있다. 예를 들어, N’ 말단 및/또는 C’ 말단에 각각 독립적으로 1 내지 100개의 아미노산, 바람직하게는 1 내지 50개의 아미노산, 보다 바람직하게는 1 내지 25개 또는 1 내지 10개의 아미노산을 추가로 포함할 수 있으나, 이에 제한되는 것이 아니며 본 발명의 펩타이드의 면역우세 에피토프로서의 활성이 실질적으로 유지되는 범위에서 임의의 아미노산을 추가로 포함할 수 있다. 예를 들어, 본 발명에 있어서, 상기 펩타이드의 서열번호 1 내지 149의 아미노산 서열의 N’ 말단 및/또는 C’ 말단에 아미노산을 추가로 포함할 수 있다. 공지된 섬유아세포 활성화 단백질(FAP)의 서열, 바람직하게는 인간 섬유아세포 활성화 단백질의 서열에 따른 아미노산이 추가되는 것이 바람직하나, 이에 제한되는 것은 아니다.In the present invention, the peptide may additionally independently include amino acids at the N'-terminus and/or the C'-terminus, respectively. For example, the peptide may additionally include 1 to 100 amino acids, preferably 1 to 50 amino acids, more preferably 1 to 25 or 1 to 10 amino acids, respectively, at the N'-terminus and/or the C'-terminus, but is not limited thereto, and any amino acid may be additionally included within a range in which the activity of the peptide of the present invention as an immunodominant epitope is substantially maintained. For example, in the present invention, the peptide may additionally include amino acids at the N'-terminus and/or the C'-terminus of the amino acid sequence of SEQ ID NOs: 1 to 149. It is preferable that an amino acid according to a sequence of a known fibroblast activation protein (FAP), preferably a sequence of human fibroblast activation protein, is added, but is not limited thereto.
본 발명에 있어서, 상기 펩타이드는 아미노산 서열의 N’ 말단 및/또는 C’ 말단의 일부 서열이 결실된 단편을 포함한다. 예를 들어, 서열번호 1 내지 149의 아미노산 서열의 N’ 말단 및/또는 C’ 말단에 각각 독립적으로 1 내지 5개의 아미노산, 바람직하게는 독립적으로 1 내지 3개의 아미노산 서열이 결실된 단편일 수 있으나, 이에 제한되는 것은 아니며 서열번호 1 내지 9의 아미노산 서열을 갖는 펩타이드의 면역우세 에피토프로서의 활성이 실질적으로 유지되는 범위에서 임의의 아미노산이 결실된 단편을 포함한다.In the present invention, the peptide includes a fragment in which a part of the N'-terminal and/or C'-terminal sequence of the amino acid sequence is deleted. For example, it may be a fragment in which 1 to 5 amino acids, preferably 1 to 3 amino acid sequences are independently deleted from the N'-terminal and/or C'-terminal of the amino acid sequence of SEQ ID NOs: 1 to 149, but is not limited thereto, and includes a fragment in which any amino acid is deleted within a range in which the activity as an immunodominant epitope of the peptide having the amino acid sequence of SEQ ID NOs: 1 to 9 is substantially maintained.
본 발명에 있어서, 상기 펩타이드는 자연으로부터 얻어진 전장 섬유아세포 활성화 단백질(FAP) 또는 이의 단편의 절단을 통해 수득되거나, 인공 합성된 올리고펩타이드인 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the peptide may be characterized by being obtained through cleavage of a full-length fibroblast activation protein (FAP) obtained from nature or a fragment thereof, or an artificially synthesized oligopeptide, but is not limited thereto.
본 발명의 펩타이드는 당업계에 알려진 다양한 기능성 펩타이드와 융합되어 융합 단백질의 형태로 제조 및 사용될 수 있다.The peptide of the present invention can be produced and used in the form of a fusion protein by being fused with various functional peptides known in the art.
본 발명에 있어서, 상기 융합 단백질은 하나 이상의 본 발명의 동일한 서열의 펩타이드의 융합(homo 형) 및/또는 상이한 서열의 펩타이드가 융합(hetero 형)된 융합단백질의 형태로 제조 및 사용될 수 있으나 이에 제한되는 것은 아니다. 예를 들어, 서열번호 1 내지 149의 아미노산 서열 중 하나 이상이 융합된 융합 단백질일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the fusion protein may be produced and used in the form of a fusion protein in which one or more peptides of the same sequence of the present invention are fused (homo type) and/or peptides of different sequences are fused (hetero type), but is not limited thereto. For example, it may be a fusion protein in which one or more of the amino acid sequences of SEQ ID NOs: 1 to 149 are fused, but is not limited thereto.
대표적인 예를 들어, 본 발명의 펩타이드 이외에도, 당업계에 알려진 또 다른 FAP 에피토프 펩타이드 또는 암 항원과 융합될 수 있거나, 약물동태학적 특성 또는 ADME 특성의 개선을 위해 Fc 도메인, PEG, 히알루론산, 알부민, 혈청 결합 도메인 등과 융합될 수 있으나 이에 제한되는 것은 아니다. 본 발명에 있어서, 상기 융합단백질은 Fc 도메인을 추가로 포함할 수 있다. 본 발명에 있어서, 상기 Fc 도메인은 IgA, IgD 및 IgG의 마지막 두개의 불변 영역 면역 글로불린 도메인 및 이들 도메인에 대한 유연한 힌지 N-말단을 포함할 수 있다. IgA, 및 IgM의 경우 Fc 도메인은 J 사슬을 포함할 수 있다. IgG의 경우, Fc 도메인은 이뮤노 글로불린 도메인 Cγ2 및 Cγ3 및 Cγ1와 Cγ2 사이의 힌지를 포함할 수 있다. Fc 도메인의 경계는 다양할 수 있으나, 인간 IgG 중쇄 Fc 영역은 일반적으로 카르복실 말단의 잔기 C226 또는 P230을 포함하도록 정의되며, 여기서 아미노산의 번호는 Kabat 넘버링을 사용한다. 본 발명의 용어, “Fc”, “Fc 도메인” 및 “Fc 영역”은 상호호환적으로 사용될 수 있으며, 상기 Fc 도메인은 상기한 영역을 분리하여 지칭하거나, 항체, 이의 단편, 융합 단백질의 일부로서 지칭될 수 있다. Fc 도메인의 다형성은 다양한 위치에서 보고되며, 제한 없이 본 발명의 융합 단백질의 융합 도메인으로 사용될 수 있다.For example, in addition to the peptide of the present invention, it may be fused with another FAP epitope peptide or cancer antigen known in the art, or may be fused with an Fc domain, PEG, hyaluronic acid, albumin, serum binding domain, etc. to improve pharmacokinetic properties or ADME properties, but is not limited thereto. In the present invention, the fusion protein may further include an Fc domain. In the present invention, the Fc domain may include the last two constant region immunoglobulin domains of IgA, IgD and IgG and a flexible hinge N-terminus for these domains. In the case of IgA and IgM, the Fc domain may include a J chain. In the case of IgG, the Fc domain may include immunoglobulin domains Cγ2 and Cγ3 and a hinge between Cγ1 and Cγ2. The boundaries of the Fc domain can vary, but the human IgG heavy chain Fc region is generally defined to include residues C226 or P230 of the carboxyl terminus, where the numbering of amino acids uses the Kabat numbering. The terms “Fc,” “Fc domain,” and “Fc region” of the present invention may be used interchangeably, and the Fc domain may refer to the region in isolation, or as part of an antibody, a fragment thereof, or a fusion protein. Polymorphisms in the Fc domain have been reported at various positions and can be used as the fusion domain of the fusion protein of the present invention without limitation.
본 발명에 있어서, 본 발명의 펩타이드는 세포의 표면 또는 인공 막의 표면에 제시되기 위해 막 관통 도메인을 추가로 포함하는 융합단백질의 형태로 제조 및 사용될 수 있다. In the present invention, the peptide of the present invention can be prepared and used in the form of a fusion protein additionally including a membrane penetrating domain for presentation on the surface of a cell or an artificial membrane.
본 발명에 있어서, 본 발명의 펩타이드는 힌지 도메인, 코일드 코일 도메인 면역 조절 도메인 및 세포 내 신호전달 도메인 중 어느 하나 이상을 추가로 포함하는 융합단백질의 형태로 제조 및 사용될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the peptide of the present invention can be prepared and used in the form of a fusion protein additionally including at least one of a hinge domain, a coiled-coil domain, an immune regulatory domain, and an intracellular signaling domain, but is not limited thereto.
본 발명의 용어, “막 관통 도메인”은 세포의 막에 걸쳐있는 단백질 도메인을 의미한다. 본 발명에 있어서, 상기 막 관통 도메인은 알파-나선 구조를 갖는 것이 바람직하나, 이에 제한되는 것은 아니다.The term “transmembrane domain” of the present invention refers to a protein domain that spans the membrane of a cell. In the present invention, the transmembrane domain preferably has an alpha-helical structure, but is not limited thereto.
본 발명의 용어, “힌지 도메인”은 막 고정 단백질의 막 관통 도메인과 세포 외 도메인 사이에 존재하는 일련의 아미노산 서열을 의미한다.The term “hinge domain” of the present invention refers to a series of amino acid sequences that exist between the transmembrane domain and the extracellular domain of a membrane anchored protein.
본 발명의 용어, “코일드 코일 도메인(coiled coil domain)”은 2개 내지 7개의 알파 나선이 로프 가닥과 같이 감겨있는 단백질의 구조적 모티프를 의미한다. 바람직하네는 상기 코일드 코일 도메인은 2개 또는 3개의 알파 나선이 감겨있는 것을 특징으로 할 수 있다.The term “coiled coil domain” of the present invention refers to a structural motif of a protein in which 2 to 7 alpha helices are coiled like a rope strand. Preferably, the coiled coil domain may be characterized by having 2 or 3 alpha helices coiled.
이외에도 본 발명의 펩타이드는 반감기 연장, 약물 동태학적 특성의 개선 등을 위해 당업계에 알려진 다양한 도메인과 제한 없이 융합될 수 있으며, 상기 도메인은 본 발명의 펩타이드의 면역우세 에피토프로서의 기능을 감소, 억제 또는 차폐시키지 않는 것이 바람직하다.In addition, the peptide of the present invention can be fused without limitation with various domains known in the art for extending half-life, improving pharmacokinetic properties, etc., and it is preferable that the domain does not reduce, inhibit, or mask the function of the peptide of the present invention as an immunodominant epitope.
본 발명의 펩타이드는 펩타이드 외의 분자와 접합되어, 접합체로 사용될 수 있다. 본 기술분야에서 알려진 단백질-접합체의 제조방법이 제한없이 사용될 수 있으며, 바람직하게는 화학적 접합을 통해 다른 약물, 또는 어쥬번트와 접합될 수 있으나, 이에 제한되는 것은 아니다.The peptide of the present invention can be used as a conjugate by being conjugated to a molecule other than a peptide. Methods for producing protein-conjugates known in the art can be used without limitation, and preferably, it can be conjugated to another drug or adjuvant through chemical conjugation, but is not limited thereto.
본 발명은 또 다른 관점에서, 본 발명의 펩타이드 또는 융합단백질을 코딩하는 핵산에 관한 것이다.In another aspect, the present invention relates to a nucleic acid encoding a peptide or fusion protein of the present invention.
본 명세서에서 사용되는 핵산은 세포, 세포 용해물(lysate) 중에 존재하거나, 또는 부분적으로 정제된 형태 또는 실질적으로 순수한 형태로 존재할 수도 있다. 핵산은 알칼리/SDS 처리, CsCl 밴드화(banding), 컬럼 크로마토그래피, 아가로스 겔 전기 영동 및 해당 기술분야에 잘 알려진 기타의 것을 포함하는 표준 기술에 의해 다른 세포 성분 또는 기타 오염 물질, 예를 들어 다른 세포의 핵산 또는 단백질로부터 정제되어 나올 경우 "단리"되거나 "실질적으로 순수하게 된" 것이다. 본 발명의 핵산은 예를 들어 DNA 또는 RNA일 수 있다.The nucleic acids used herein may be present in cells, a cell lysate, or may be present in a partially purified or substantially pure form. A nucleic acid is "isolated" or "rendered substantially pure" when purified away from other cellular components or other contaminants, such as other cellular nucleic acids or proteins, by standard techniques including alkaline/SDS treatment, CsCl banding, column chromatography, agarose gel electrophoresis, and others well known in the art. A nucleic acid of the present invention may be, for example, DNA or RNA.
본 발명은 또 다른 관점에서, 본 발명의 핵산을 함유하는 재조합 벡터에 관한 것이다.In another aspect, the present invention relates to a recombinant vector containing the nucleic acid of the present invention.
본 발명에 있어서, 상기 재조합 벡터는 상기 펩타이드를 코딩하는 핵산의 단백질 발현을 유도할 수 있는 벡터라면 당업계에 공지된 벡터를 적절히 선택하여 제한없이 사용할 수 있다. 예를 들어, 대장균을 숙주로 사용할 경우 T7계열 (T7A1, T7A2, T7A3 등), lac, lacUV5, 온도의존형 (λphoA, phoB, rmB, tac, trc, trp 또는 1PL 프로모터를 포함하는 벡터를 사용할 수 있고, 효모를 숙주로 사용할 경우 ADH1, AOX1, GAL1, GAL10, PGK 또는 TDH3 프로모터를 포함하는 벡터를 사용할 수 있으며, 바실러스의 경우 P2 프로모터를 포함하는 벡터를 사용할 수 있으나, 이는 일부 실시 양태를 나열한 것으로, 상기 프로모터를 포함하는 벡터 이외에도 본 발명에 따른 펩타이드의 발현을 유도하기 위한 프로모터를 포함하는 벡터로써 숙주에 적합한 것이라면 제한없이, 당업계에 공지된 다양한 벡터를 당업자가 적절히 선택하여 사용할 수 있다.In the present invention, as long as the recombinant vector can induce protein expression of a nucleic acid encoding the peptide, any vector known in the art can be appropriately selected and used without limitation. For example, when E. coli is used as a host, a vector containing a T7 series (T7A1, T7A2, T7A3, etc.), lac, lacUV5, temperature-dependent (λphoA, phoB, rmB, tac, trc, trp or 1PL promoter) can be used, and when yeast is used as a host, a vector containing an ADH1, AOX1, GAL1, GAL10, PGK or TDH3 promoter can be used, and in the case of Bacillus, a vector containing a P2 promoter can be used. However, this is only a listing of some embodiments, and in addition to the vector containing the promoter, as long as it is suitable for the host as a vector containing a promoter for inducing expression of the peptide according to the present invention, various vectors known in the art can be appropriately selected and used by those skilled in the art.
본 발명에서 용어 "벡터 (vector)"는 적합한 숙주 내에서 DNA를 발현시킬 수 있는 적합한 조절 서열에 작동 가능하게 연결된 DNA 서열을 함유하는 DNA 제조물을 의미한다. 벡터는 플라스미드, 파지 입자, 또는 간단하게 잠재적 게놈 삽입물일 수 있다. 적당한 숙주로 형질전환되면, 벡터는 숙주 게놈과 무관하게 복제하고 기능할 수 있거나, 또는 일부 경우에 게놈 그 자체에 통합될 수 있다. 플라스미드가 현재 벡터의 가장 통상적으로 사용되는 형태이므로, 본 발명의 명세서에서 "플라스미드 (plasmid)" 및 "벡터 (vector)"는 때로 상호 교환적으로 사용된다. 그러나, 본 발명은 당업계에 알려진 또는 알려지게 되는 바와 동등한 기능을 갖는 벡터의 다른 형태를 포함한다. 대장균에서 사용되는 단백질 발현 벡터로는 Novagen (미국) 사의 pET 계열; Invitrogen (미국)의 pBAD 계열; Takara (일본)의 pHCE 나 pCOLD; 제노포커스 (대한민국)의 pACE 계열; 등을 사용할 수 있다. 고초균에서는 게놈의 특정부분에 목적 유전자를 삽입하여 단백질 발현을 구현하거나, MoBiTech (독일)의 pHT 계열의 벡터, 등을 사용할 수 있다. 곰팡이나 효모에서도 게놈 삽입이나 자가복제 벡터들 이용하여 단백질 발현이 가능하다. Agrobacterium tumefaciens나 Agrobacterium rhizogenes 등의 T-DNA 시스템을 이용하여 식물용 단백질 발현 벡터를 사용할 수 있다. 포유동물 세포 배양물 발현을 위한 전형적인 발현 벡터는 예를 들면 pRK5 (EP 307,247호), pSV16B (WO 91/08291호) 및 pVL1392 (Pharmingen) 등을 기초로 한다.The term "vector" in the present invention means a DNA preparation containing a DNA sequence operably linked to a suitable regulatory sequence capable of expressing the DNA in a suitable host. The vector may be a plasmid, a phage particle, or simply a potential genomic insert. Once transformed into a suitable host, the vector may replicate and function independently of the host genome, or in some cases may be integrated into the genome itself. Since plasmids are currently the most commonly used form of vector, the terms "plasmid" and "vector" are sometimes used interchangeably in the present description. However, the present invention includes other forms of vectors that have equivalent functions as known or become known in the art. Protein expression vectors used in E. coli include the pET series from Novagen (USA); the pBAD series from Invitrogen (USA); the pHCE or pCOLD series from Takara (Japan); the pACE series from Xenofocus (Korea); and the like. In Bacillus subtilis, protein expression can be achieved by inserting the target gene into a specific part of the genome, or by using vectors such as the pHT series from MoBiTech (Germany). Protein expression is also possible in molds and yeast by using genome insertion or self-replicating vectors. Plant protein expression vectors can be used using the T-DNA system of Agrobacterium tumefaciens or Agrobacterium rhizogenes. Typical expression vectors for expression in mammalian cell cultures are based on, for example, pRK5 (EP 307,247), pSV16B (WO 91/08291), and pVL1392 (Pharmingen).
"발현 조절 서열 (expression control sequence)"이라는 표현은 특정한 숙주 생물에서 작동가능하게 연결된 코딩 서열의 발현에 필수적인 DNA 서열을 의미한다. 그러한 조절 서열은 전사를 실시하기 위한 프로모터, 그러한 전사를 조절하기 위한 임의의 오퍼레이터 서열, 적합한 mRNA 리보좀 결합 부위를 코딩하는 서열 및 전사 및 해독의 종결을 조절하는 서열을 포함한다. 예를 들면, 원핵생물에 적합한 조절 서열은 프로모터, 임의로 오퍼레이터 서열 및 리보좀 결합 부위를 포함한다. 진핵세포는 프로모터, 폴리아데닐화 시그날 및 인핸서가 이에 포함된다. 플라스미드에서 유전자의 발현 양에 가장 영향을 미치는 인자는 프로모터이다. 고 발현용의 프로모터로서 SRα 프로모터와 사이토메갈로바이러스 (cytomegalovirus) 유래 프로모터 등이 바람직하게 사용된다.The term "expression control sequence" means a DNA sequence essential for the expression of an operably linked coding sequence in a particular host organism. Such control sequences include a promoter for initiating transcription, an optional operator sequence for regulating such transcription, a sequence encoding a suitable mRNA ribosome binding site, and a sequence for regulating the termination of transcription and translation. For example, a control sequence suitable for a prokaryote includes a promoter, optionally an operator sequence, and a ribosome binding site. For a eukaryote, this includes a promoter, a polyadenylation signal, and an enhancer. The factor that most influences the amount of gene expression in a plasmid is the promoter. As promoters for high expression, the SRα promoter and a cytomegalovirus-derived promoter are preferably used.
본 발명의 DNA 서열을 발현시키기 위하여, 매우 다양한 발현 조절 서열 중 어느 것이라도 벡터에 사용될 수 있다. 유용한 발현 조절서열의 예에는, 상기 기술한 프로모터 이외에도, 예를 들어, SV40 또는 아데노바이러스의 초기 및 후기 프로모터들, lac 시스템, trp 시스템, TAC 또는 TRC 시스템, T3 및 T7 프로모터들, 파지 람다의 주요 오퍼레이터 및 프로모터 영역, fd 코드 단백질의 조절 영역, 3-포스포글리세레이트 키나제 또는 다른 글리콜분해 효소에 대한 프로모터, 상기 포스파타제의 프로모터들, 예를 들어 Pho5, 효모 알파-교배 시스템의 프로모터 및 원핵세포 또는 진핵세포 또는 이들의 바이러스의 유전자의 발현을 조절하는 것으로 알려진 구성과 유도의 기타 다른 서열 및 이들의 여러 조합이 포함된다. T7 RNA 폴리메라아제 프로모터 Φ은 E. coli에서 단백질을 발현시키는데 유용하게 사용될 수 있다.Any of a wide variety of expression control sequences may be used in the vector to express the DNA sequence of the present invention. Examples of useful expression control sequences include, in addition to the promoters described above, the early and late promoters of SV40 or adenovirus, the lac system, the trp system, the TAC or TRC system, the T3 and T7 promoters, the major operator and promoter region of phage lambda, the regulatory region of the fd encoded protein, the promoter for 3-phosphoglycerate kinase or other glycolytic enzymes, the promoters of the phosphatases, e.g. Pho5, the promoter of the yeast alpha-mating system, and any other sequence of structure and inducibility known to control expression of genes in prokaryotes or eukaryotes or their viruses, and any combination thereof. The T7 RNA polymerase promoter Φ can be usefully used to express proteins in E. coli.
핵산은 다른 핵산 서열과 기능적 관계로 배치될 때 "작동가능하게 연결 (operably linked)"된다. 이것은 적절한 분자 (예를 들면, 전사 활성화 단백질)은 조절 서열(들)에 결합될 때 유전자 발현을 가능하게 하는 방식으로 연결된 유전자 및 조절 서열(들)일 수 있다. 예를 들면, 전서열(pre-sequence) 또는 분비 리더 (leader)에 대한 DNA는 폴리펩타이드의 분비에 참여하는 전단백질로서 발현되는 경우 폴리펩타이드에 대한 DNA에 작동가능하게 연결되고; 프로모터 또는 인핸서는 서열의 전사에 영향을 끼치는 경우 코딩서열에 작동가능하게 연결되거나; 또는 리보좀 결합 부위는 서열의 전사에 영향을 끼치는 경우 코딩 서열에 작동가능하게 연결되거나; 또는 리보좀 결합 부위는 번역을 용이하게 하도록 배치되는 경우 코딩 서열에 작동가능하게 연결된다. 일반적으로, "작동가능하게 연결된"은 연결된 DNA 서열이 접촉하고, 또한 분비 리더의 경우 접촉하고 리딩 프레임 내에 존재하는 것을 의미한다. 그러나, 인핸서 (enhancer)는 접촉할 필요가 없다. 이들 서열의 연결은 편리한 제한 효소 부위에서 라이게이션(접합)에 의해 수행된다. 그러한 부위가 존재하지 않는 경우, 통상의 방법에 따른 합성 올리고뉴클레오티드 어댑터 (oligonucleotide adaptor) 또는 링커(linker)를 사용한다.A nucleic acid is "operably linked" when it is placed into a functional relationship with another nucleic acid sequence. This can be a gene and regulatory sequence(s) that are linked in such a way that an appropriate molecule (e.g., a transcriptional activating protein) can cause gene expression when bound to the regulatory sequence(s). For example, DNA for a pre-sequence or a secretory leader is operably linked to DNA for a polypeptide if it is expressed as a pre-protein that participates in the secretion of the polypeptide; a promoter or enhancer is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it affects the transcription of the sequence; or a ribosome binding site is operably linked to a coding sequence if it is positioned so as to facilitate translation. In general, "operably linked" means that the linked DNA sequences are in contact, and in the case of a secretory leader, are in contact and are in reading frame. However, an enhancer need not be in contact. The connection of these sequences is carried out by ligation at convenient restriction enzyme sites. If such sites do not exist, synthetic oligonucleotide adaptors or linkers according to conventional methods are used.
본원 명세서에 사용된 용어 "발현 벡터"는 통상 이종의 DNA의 단편이 삽입된 재조합 캐리어 (recombinant carrier)로서 일반적으로 이중 가닥의 DNA의 단편을 의미한다. 여기서, 이종 DNA는 숙주세포에서 천연적으로 발견되지 않는 DNA인 이형 DNA를 의미한다. 발현 벡터는 일단 숙주 세포 내에 있으면 숙주 염색체 DNA와 무관하게 복제할 수 있으며 벡터의 수 개의 카피 및 그의 삽입된 (이종) DNA가 생성될 수 있다.The term "expression vector" as used herein generally refers to a recombinant carrier into which a fragment of heterologous DNA has been inserted, typically a fragment of double-stranded DNA. Here, heterologous DNA refers to heterologous DNA, which is DNA that is not naturally found in the host cell. Once inside the host cell, the expression vector can replicate independently of the host chromosomal DNA, and multiple copies of the vector and its inserted (heterologous) DNA can be produced.
당업계에 주지된 바와 같이, 숙주세포에서 형질감염 유전자의 발현 수준을 높이기 위해서는, 해당 유전자가, 선택된 발현 숙주 내에서 기능을 발휘하는 전사 및 해독 발현 조절 서열에 작동가능하도록 연결되어야만 한다. 바람직하게는 발현 조절서열 및 해당 유전자는 세균 선택 마커 및 복제 개시점 (replication origin)을 같이 포함하고 있는 하나의 발현 벡터 내에 포함되게 된다. 발현 숙주가 진핵세포인 경우에는, 발현 벡터는 진핵 발현 숙주 내에서 유용한 발현 마커를 더 포함할 수 있다.As is well known in the art, in order to increase the level of expression of a transfected gene in a host cell, the gene must be operably linked to transcriptional and translational expression control sequences that are functional in the selected expression host. Preferably, the expression control sequences and the gene are contained in a single expression vector that also contains a bacterial selection marker and a replication origin. If the expression host is a eukaryotic cell, the expression vector may further contain expression markers that are useful in the eukaryotic expression host.
본 발명은 또 다른 관점에서, 본 발명의 펩타이드를 코딩하는 핵산; 또는 상기 재조합 벡터가 도입된 숙주세포에 관한 것이다.In another aspect, the present invention relates to a nucleic acid encoding a peptide of the present invention; or a host cell into which the recombinant vector has been introduced.
본 발명에 있어서, 상기 숙주세포는 단백질 등을 생산하기 위해, 유전자 또는 재조합 벡터 등이 도입된 발현용 세포를 의미한다. 상기 숙주세포는 본 발명의 펩타이드를 발현할 수 있는 세포라면 제한없이 사용될 수 있으며, 바람직하게는 진핵세포, 더욱 바람직하게는 효모, 곤충세포, 동물세포, 가장 바람직하게는 동물세포일 수 있다. 예를 들어 펩타이드의 발현에 주로 사용되는 CHO 세포주 또는 HEK 세포주 등이 사용될 수 있으나, 이에 제한되는 것은 아니다. In the present invention, the host cell refers to an expression cell into which a gene or a recombinant vector, etc. has been introduced to produce a protein, etc. The host cell may be used without limitation as long as it is a cell capable of expressing the peptide of the present invention, and is preferably a eukaryotic cell, more preferably a yeast, an insect cell, an animal cell, and most preferably an animal cell. For example, a CHO cell line or a HEK cell line, which are mainly used for the expression of peptides, may be used, but is not limited thereto.
본 발명의 상기 펩타이드를 발현시키기 위해 매우 다양한 발현 숙주/벡터 조합이 이용될 수 있다. 진핵 숙주에 적합한 발현 벡터에는, 예를 들어 SV40, 소 유두종바이러스, 아네노바이러스, 아데노-연관 바이러스(adeno-associated virus), 사이토메갈로바이러스 및 레트로바이러스로부터 유래된 발현 조절 서열을 포함한다. 세균 숙주에 사용할 수 있는 발현 벡터에는 pBluescript, pGEX2T, pUC벡터, col E1, pCR1, pBR322, pMB9 및 이들의 유도체와 같이 E. coli에서 얻는 것을 예시할 수 있는 세균성 플라스미드, RP4와 같이 보다 넓은 숙주 범위를 갖는 플라스미드, λ과 λNM989와 같은 매우 다양한 파지 람다(phage lambda) 유도체로 예시될 수 있는 파지 DNA, 및 M13과 필라멘트성 단일가닥의 DNA 파지와 같은 기타 다른 DNA 파지가 포함된다. 효모 세포에 유용한 발현 벡터는 2μ 플라스미드 및 그의 유도체이다. 곤충 세포에 유용한 벡터는 pVL 941이다.A wide variety of expression host/vector combinations can be utilized to express the peptides of the present invention. Suitable expression vectors for eukaryotic hosts include, for example, expression control sequences derived from SV40, bovine papillomavirus, adenovirus, adeno-associated virus, cytomegalovirus and retroviruses. Expression vectors for use in bacterial hosts include, for example, bacterial plasmids obtained from E. coli, such as pBluescript, pGEX2T, pUCvector, col E1, pCR1, pBR322, pMB9 and derivatives thereof, plasmids having a wider host range, such as RP4, phage DNA, such as a wide variety of phage lambda derivatives, such as λ and λNM989, and other DNA phages, such as M13 and filamentous single-stranded DNA phages. Useful expression vectors for yeast cells are the 2μ plasmids and derivatives thereof. A useful vector for insect cells is pVL 941.
상기 재조합 벡터는 형질전환 또는 형질감염 등의 방법으로 숙주세포에 도입될 수 있다. 본원 명세서에 사용된 용어 "형질전환"은 DNA를 숙주로 도입하여 DNA가 염색체 외 인자로서 또는 염색체 통합완성에 의해 복제 가능하게 되는 것을 의미한다. 본원 명세서에 사용된 용어 "형질감염"은 임의의 코딩 서열이 실제로 발현되든 아니든 발현 벡터가 숙주 세포에 의해 수용되는 것을 의미한다. The above recombinant vector can be introduced into a host cell by a method such as transformation or transfection. The term "transformation" as used in the present specification means that DNA is introduced into a host so that the DNA becomes replicable as an extrachromosomal element or by chromosomal integration. The term "transfection" as used in the present specification means that an expression vector is accepted by a host cell, regardless of whether any coding sequence is actually expressed.
물론 모든 벡터와 발현 조절 서열이 본 발명의 DNA 서열을 발현하는데 모두 동등하게 기능을 발휘하지는 않는다는 것을 이해하여야만 한다. 마찬가지로 모든 숙주가 동일한 발현 시스템에 대해 동일하게 기능을 발휘하지는 않는다. 그러나, 당업자라면 과도한 실험적 부담 없이 본 발명의 범위를 벗어나지 않는 채로 여러 벡터, 발현 조절 서열 및 숙주 중에서 적절한 선택을 할 수 있다. 예를 들어, 벡터를 선택함에 있어서는 숙주를 고려하여야 하는데, 이는 벡터가 그 안에서 복제되어야만 하기 때문이다. 벡터의 복제 수, 복제 수를 조절할 수 있는 능력 및 당해 벡터에 의해 코딩되는 다른 단백질, 예를 들어 항생제 마커의 발현도 또한 고려되어야만 한다. 발현 조절 서열을 선정함에 있어서도, 여러 가지 인자들을 고려하여야만 한다. 예를 들어, 서열의 상대적 강도, 조절 가능성 및 본 발명의 DNA 서열과의 상용성 등, 특히 가능성 있는 이차 구조와 관련하여 고려하여야 한다. 단세포 숙주는 선정된 벡터, 본 발명의 DNA 서열에 의해 코딩되는 산물의 독성, 분비 특성, 단백질을 정확하게 폴딩시킬 수 있는 능력, 배양 및 발효 요건들, 본 발명 DNA 서열에 의해 코딩되는 산물을 숙주로부터 정제하는 것의 용이성 등의 인자를 고려하여 선정되어야만 한다. 이들 변수의 범위내에서, 당업자는 본 발명의 DNA 서열을 발효 또는 대규모 동물 배양에서 발현시킬 수 있는 각종 벡터/발현 조절 서열/숙주 조합을 선정할 수 있다. 발현 클로닝에 의해 cDNA를 클로닝 하려고 할 때의 스크리닝 법으로서 바인딩법(binding법), 페닝법(panning법), 필름 에멀션법(film emulsion 법)등이 적용될 수 있다.It should be understood that not all vectors and expression control sequences are equally effective in expressing the DNA sequences of the present invention. Likewise, not all hosts are equally effective in the same expression system. However, one skilled in the art can make appropriate selections among the various vectors, expression control sequences, and hosts without undue experimental burden and without departing from the scope of the present invention. For example, in selecting a vector, one should consider the host, since the vector must be replicated in it. The copy number of the vector, the ability to control the copy number, and the expression of other proteins encoded by the vector, such as antibiotic markers, should also be considered. In selecting an expression control sequence, several factors should also be considered, such as the relative strength of the sequences, their controllability, and their compatibility with the DNA sequences of the present invention, particularly with respect to possible secondary structures. The unicellular host should be selected by considering factors such as the selected vector, the toxicity of the product encoded by the DNA sequence of the present invention, secretion characteristics, the ability to accurately fold the protein, culture and fermentation requirements, and the ease of purifying the product encoded by the DNA sequence of the present invention from the host. Within the range of these variables, those skilled in the art can select various vector/expression control sequence/host combinations that can express the DNA sequence of the present invention in fermentation or large-scale animal culture. As a screening method when attempting to clone cDNA by expression cloning, the binding method, the panning method, the film emulsion method, etc. can be applied.
상기 유전자 및 재조합 벡터는 종래에 공지된 다양한 방법을 통해, 숙주세포에 도입될 수 있다. 본 발명의 펩타이드를 코딩하는 핵산를 코딩하는 유전자가 직접적으로 숙주세포의 게놈에 도입되어 염색체 상 인자로서 존재할 수 있다. 본 발명이 속하는 기술분야의 당업자에게 있어 상기 유전자를 숙주세포의 게놈 염색체에 삽입하여서도 재조합 벡터를 숙주세포에 도입한 경우와 동일한 효과를 가질 것은 자명하다 할 것이다.The above gene and recombinant vector can be introduced into a host cell through various methods known in the art. A gene encoding a nucleic acid encoding the peptide of the present invention can be directly introduced into the genome of a host cell and exist as a chromosomal element. It will be obvious to those skilled in the art that inserting the gene into the genomic chromosome of a host cell will have the same effect as introducing a recombinant vector into the host cell.
본 발명은 또 다른 관점에서, 상기 숙주세포를 배양하는 단계를 포함하는 펩타이드의 제조방법에 관한 것이다.In another aspect, the present invention relates to a method for producing a peptide, which comprises a step of culturing the host cell.
상기 펩타이드를 발현할 수 있는 재조합 발현 벡터가 포유류 숙주세포 내로 도입될 경우, 펩타이드가 숙주세포에서 발현되기에 충분한 기간 동안, 또는 숙주세포가 배양되는 배양 배지 내로 펩타이드가 분비되게 하기에 충분한 기간 동안 숙주세포를 배양함으로써 제조될 수 있다.When a recombinant expression vector capable of expressing the above peptide is introduced into a mammalian host cell, the peptide can be produced by culturing the host cell for a period of time sufficient to express the peptide in the host cell or for a period of time sufficient to secrete the peptide into the culture medium in which the host cell is cultured.
경우에 따라서, 발현된 펩타이드는 숙주세포로부터 분리하여 균일하도록 정제하여 수득될 수 있다. 상기 펩타이드의 분리 또는 정제는 통상의 단백질에서 사용되고 있는 분리, 정제 방법, 예를 들어 크로마토그래피에 의해 수행될 수 있다. 상기 크로마토그래피는 예를 들어, 친화성 크로마토그래피, 이온교환 크로마토그래피 또는 소수성 크로마토그래피에서 선택된 하나 이상의 조합일 수 있지만, 이에 한정되지는 않는다. 상기 크로마토그래피 이외에, 추가로 여과, 초여과, 염석, 투석 등을 조합되어 사용될 수 있다.In some cases, the expressed peptide can be obtained by separating from the host cell and purifying it to be homogeneous. The separation or purification of the peptide can be performed by a separation or purification method used for general proteins, for example, chromatography. The chromatography can be, for example, a combination of one or more selected from affinity chromatography, ion exchange chromatography, or hydrophobic chromatography, but is not limited thereto. In addition to the chromatography, filtration, ultrafiltration, salting out, dialysis, etc. can be additionally used in combination.
본 발명의 펩타이드는 펩타이드 자체로 제시될 수 있으나, 이에 제한되는 것은 아니며, 효과적인 전달, 표적화, 약물 동태학적 특성 개선 등 다양한 목적을 위해 전달 비히클의 표면에 부착 또는 내부에 포함되어 제시될 수 있다.The peptide of the present invention may be presented as a peptide itself, but is not limited thereto, and may be presented attached to the surface of or included within a delivery vehicle for various purposes such as effective delivery, targeting, and improved pharmacokinetic properties.
예를 들어, 펩타이드 기반 백신은 비용 효율적인 제조, 상대적으로 쉬운 품질 관리 및 우수한 안전성 프로파일을 포함하여 몇 가지 장점이 있지만 낮은 면역원성을 나타내며, 강력한 보조제를 필요로 한다. 또한 생체 내에서 항원 제시 세포(APC)로의 낮은 전달 효율로 인해 치료 효능이 제한된다.For example, peptide-based vaccines have several advantages, including cost-effective manufacturing, relatively easy quality control, and good safety profiles, but they exhibit low immunogenicity, require strong adjuvants, and have limited therapeutic efficacy due to low delivery efficiency to antigen-presenting cells (APCs) in vivo.
이러한 한계를 극복하기 위해 본 발명자들은 중성-하전된 작은 지질 나노입자 (small lipid nanoparticle, SLNP) 기반의 항원 전달체 및 이를 이용한 백신 시스템을 개발하고 특허등록받은 바 있다(대한민국 특허 제10-2425028호 Theranostics 2016, 6 (2), 192-203, 및 Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638).To overcome these limitations, the present inventors have developed and patented an antigen carrier based on neutrally charged small lipid nanoparticles (SLNPs) and a vaccine system using the same (Korean Patent No. 10-2425028 Theranostics 2016, 6 (2), 192-203, and Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638).
본 발명의 일 실시예에서, 중성-하전된 작은 지질 나노입자 (small lipid nanoparticle, SLNP) 기반의 백신 플랫폼을 이용하여, 상기 펩타이드를 포함하는 지질 나노입자를 제조하고, 상기 지질 나노입자로 면역화를 하는 경우 FAP 특이적인 CD8+ T세포의 수 및 활성이 현저히 증가함을 확인하였다.In one embodiment of the present invention, a vaccine platform based on neutrally-charged small lipid nanoparticles (SLNPs) was used to prepare lipid nanoparticles containing the peptide, and it was confirmed that when immunization was performed with the lipid nanoparticles, the number and activity of FAP-specific CD8+ T cells were significantly increased.
따라서, 본 발명은 다른 관점에서, 상기 펩타이드를 포함하는 나노입자에 관한 것이다.Therefore, the present invention, from another aspect, relates to nanoparticles comprising the peptide.
본 발명의 용어 나노입자는 나노미터 크기를 갖는 입자로 정의되며, 각 입자들은 모양, 크기, 성분, 표면의 특성, 작용기 등에 따라 물리화학적, 생물학적, 면역원성 특징이 달라질 수 있다.The term nanoparticle of the present invention is defined as a particle having a nanometer size, and each particle may have different physicochemical, biological, and immunogenic characteristics depending on the shape, size, components, surface characteristics, functional groups, etc.
본 발명에 있어서, 상기 나노입자는 하나 이상의 종류의 상기 펩타이드를 포함하는 것을 특징으로 할 수 있다. 본 발명에 있어서, 상기 나노입자는 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 또는 그 이상의 종류의 서열을 포함하는 본 발명의 펩타이드를 포함하는 것을 특징으로 할 수 있다. 예를 들어, 상기 나노입자는 서열번호 1 내지 149로 구성된 군에서 선택되는 어느 하나 이상의 아미노산 서열을 포함할 수 있다. 본 발명의 백신 조성물에 포함된 하나 이상의 펩타이드는 앞서 설명한 것과 같이 에피토프 펩타이드, 이를 포함하는 융합 단백질, 나노입자 등의 다양한 형태일 수 있다. In the present invention, the nanoparticle may be characterized by including one or more types of the peptide. In the present invention, the nanoparticle may be characterized by including the peptide of the present invention including 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more types of sequences. For example, the nanoparticle may include any one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 1 to 149. The one or more peptides included in the vaccine composition of the present invention may be in various forms, such as an epitope peptide, a fusion protein including the same, a nanoparticle, etc., as described above.
본 발명의 기술 분야에서 펩타이드를 전달하기 위한 나노입자 플랫폼이 잘 알려져 있다. 예를 들어, 상기 나노입자는 펩타이드를 캡슐화(encapsulation) 하여 내부에 포함하거나, 나노입자의 표면에 상기 단백질을 제시할 수 있으나, 이에 제한되는 것은 아니다.Nanoparticle platforms for delivering peptides are well known in the art. For example, the nanoparticle may encapsulate a peptide and contain it inside, or may present the protein on the surface of the nanoparticle, but is not limited thereto.
본 발명에 있어서, 예를 들어 상기 나노입자는 바이러스 유사 입자(Virus-like particle, VLPs), 단백질 나노구조체, 폴리머 나노입자, 지질 나노입자, 무기 나노입자로 구성된 군에서 선택되는 것일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, for example, the nanoparticle may be selected from the group consisting of virus-like particles (VLPs), protein nanostructures, polymer nanoparticles, lipid nanoparticles, and inorganic nanoparticles, but is not limited thereto.
본 발명의 용어 바이러스 유사 입자(VLP)는 유전체와 같은 감염요소가 제거된 자가 조립된 바이러스 외피 또는 캡시드 단백질로 구성된 수~수백 나노미터 범위의 나노입자를 의미한다. 바이러스 유사입자는 펩타이드 에피토프의 체내 전달을 위한 최초의 나노입자로, 다양한 임상시험 및 상업적으로 승인된 백신의 전달 형태이다.The term virus-like particle (VLP) of the present invention refers to a nanoparticle in the range of several to several hundred nanometers composed of self-assembled viral envelope or capsid proteins from which infectious elements such as genomes have been removed. Virus-like particles are the first nanoparticles for the in vivo delivery of peptide epitopes and are the delivery form for various clinically tested and commercially approved vaccines.
본 발명에 있어서, 상기 폴리머 나노입자는 예를 들어, HPMA (N-(2-hydroxypropyl)-methacrylamide copolymer), SMA (polystyrene-maleic anhydride copolymer), PEG(polyethylene glycol), 및 PGA(poly-L-glutamic acid) 로 구성된 군에서 선택되는 폴리머 기반의 나노입자일 수 있으나 이에 제한되는 것은 아니다. 또 다른 예를 들어, 상기 폴리머 나노입자는 블록 공중합체, 덴드리머 등일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the polymer nanoparticle may be a polymer-based nanoparticle selected from the group consisting of, for example, HPMA (N-(2-hydroxypropyl)-methacrylamide copolymer), SMA (polystyrene-maleic anhydride copolymer), PEG (polyethylene glycol), and PGA (poly-L-glutamic acid), but is not limited thereto. For another example, the polymer nanoparticle may be, but is not limited to, a block copolymer, a dendrimer, or the like.
본 발명에 있어서, 상기 무기 나노입자는 예를 들어, 실리카 나노입자(silica nanoparticle), 금 나노입자, 자기결합 나노입자 등이 있으나, 이에 제한되는 것은 아니다.In the present invention, the inorganic nanoparticles include, but are not limited to, silica nanoparticles, gold nanoparticles, self-associating nanoparticles, etc.
본 발명에 있어서, 상기 단백질 나노구조체는 단백질의 자가조립으로 형성되는 나노입자를 의미한다. 본 발명에 있어서, 상기 단백질 나노구조체는 예를 들어 페리틴(Ferritin), 루마진 합성효소(Lumazine synrhase, LS), 디하이드로리포일 아세틸트랜스퍼라아제(dihydrolipoyl acetyltransferase, E2p) nsp10(nonstructural protein 10) 등의 단백질에 기반한 플랫폼이 알려져 있으나 이에 제한되는 것은 아니다.In the present invention, the protein nanostructure refers to a nanoparticle formed by self-assembly of a protein. In the present invention, the protein nanostructure is known to be a platform based on proteins such as, for example, ferritin, lumazine synrhase (LS), dihydrolipoyl acetyltransferase (E2p) nsp10 (nonstructural protein 10), but is not limited thereto.
본 발명에 있어서, 바람직하게는 상기 나노입자는 지질 나노입자인 것을 특징으로 할 수 잇다.In the present invention, it is preferably characterized in that the nanoparticle is a lipid nanoparticle.
본 발명에 있어서, 상기 지질 나노입자는 나노디스크, 유니라멜라 비지클(unilamellar vesicle), 멀티라멜라 비지클(multilamellar vesicle; MLV), 멀티베지큘라 비지클(multivesicular vesicle; MV), 리포좀, LNP, 에멀젼 및 LPP(Lipopolyplex)로 구성된 군에서 선택되는 것일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the lipid nanoparticle may be selected from the group consisting of a nanodisk, a unilamellar vesicle, a multilamellar vesicle (MLV), a multivesicular vesicle (MV), a liposome, a LNP, an emulsion, and a lipopolyplex (LPP), but is not limited thereto.
본 발명에 있어서, 상기 지질 나노입자는 인지질, 양이온성 지질 및 어쥬번트로 구성된 군에서 선택되는 어느 하나 이상을 추가로 포함하는 것을 특징으로 할 수 있다.In the present invention, the lipid nanoparticle may be characterized by further including at least one selected from the group consisting of a phospholipid, a cationic lipid, and an adjuvant.
본 발명에 있어서, 예를 들어 상기 인지질은 지방족 탄소수가 10~30, 바람직하게는 12~25, 가장 바람직하게는 14 내지 22인 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다. 보다 구체적인 예를 들어, 상기 인지질은 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPEPEG1000) 를 포함하는 DSPE-PEG 유도체, 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] (DSPE-PEG2000-PDP) 및 1,2-distearoyl-sn-glycero-3-phosphoethanolamineN-[maleimide(polyethylene glycol)-2000] (DSPE-PEG2000-Maleimide) 등을 포함하는 기능화된 (functionalized) DSPE-PEG 유도체, 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) 및 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl) (DPPE-Rhodamine)를 포함하는 형광 표지된 인지질, 1,2-Didecanoyl-sn-glycero-3-phosphocholine (DDPC), 1,2-Dierucoyl-sn-glycero-3-phosphate (DEPA), 1,2-Dierucoyl-sn-glycero-3-phosphocholine (DEPC), 1,2-Dierucoyl-sn-glycero-3-phosphoethanolamine (DEPE), 2-Dierucoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DEPG), 1,2-Dilinoleoyl-sn-glycero-3-phosphocholineIn the present invention, for example, the phospholipid may be characterized by having an aliphatic carbon number of 10 to 30, preferably 12 to 25, and most preferably 14 to 22, but is not limited thereto. For more specific examples, the phospholipid may be a DSPE-PEG derivative including 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine (DSPE), 1,2-Distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPEPEG1000), a functionalized DSPE-PEG derivative including 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] (DSPE-PEG2000-PDP) and 1,2-distearoyl-sn-glycero-3-phosphoethanolamineN-[maleimide(polyethylene glycol)-2000] (DSPE-PEG2000-Maleimide), and 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE). Fluorescently labeled phospholipids including 1,2-dipalmitoyl-sn-glycero-3-phosphoethanolamine-N-(lissamine rhodamine B sulfonyl) (DPPE-Rhodamine), 1,2-Didecanoyl-sn-glycero-3-phosphocholine (DDPC), 1,2-Dierucoyl-sn-glycero-3-phosphate (DEPA), glycero-3-phosphocholine (DEPC), 1,2-Dierucoyl-sn-glycero-3-phosphoethanolamine (DEPE), 2-Dierucoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DEPG), 1,2-Dilinoleoyl-sn-glycero-3-phosphocholine
(DLOPC), 1,2-Dilauroyl-sn-glycero-3-phosphate (DLPA), 1,2-Dilauroyl-sn-glycero-3-phosphocholine (DLPE), 1,2-Dilauroyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DLPG), 1,2-Dilauroyl-sn-glycero-3-phosphoserine (DLPS), 1,2-Dimyristoyl-sn-glycero-3-phosphate (DMPA), 1,2-Dimyristoyl-sn-glycero-3- phosphocholine (DMPC), 1,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE), 1,2-Dimyristoyl-snglycero-3-Phospho-rac-(1-glycerol) (DMPG), 1,2-Dimyristoyl-sn-glycero-3-phosphoserine (DMPS), 1,2-Dioleoyl-sn-glycero-3-phosphate (DOPA), 1,2-Dioleoyl-sn-glycero-3-phosphocholine (DOPC), 1,2-Dioleoylsn-glycero-3-Phospho-rac-(1-glycerol) (DOPG), 1,2-Dioleoyl-sn-glycero-3-phosphoserine (DOPS), 1,2-Dipalmitoyl-sn-glycero-3-phosphate (DPPA), 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-Dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE), 1,2-Dipalmitoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DPPG), 1,2-Dipalmitoyl-sn-glycero-3-phosphoserine (DPPS), 1,2-Distearoyl-sn-glycero-3-phosphate (DSPA), 1,2-Distearoyl-sn-glycero-3-phosphocholine (DSPC), 1,2-Distearoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DSPG), 1,2-Distearoyl-sn-glycero-3-phosphoserine (DSPS), Egg-PC (EPC), Hydrogenated Egg PC (HEPC), Hydrogenated Soy PC (HSPC), 1-Myristoyl-sn-glycero-3-phosphocholine (LYSOPC MYRISTIC), 1-Palmitoyl-sn-glycero-3-phosphocholine (LYSOPC PALMITIC), 1-Stearoyl-sn-glycero-3-phosphocholine (LYSOPC STEARIC), 1-Myristoyl-2-palmitoyl-sn-glycero 3-phosphocholine (Milk Sphingomyelin MPPC), 1-Myristoyl-2-stearoyl-sn-glycero-3-phosphocholine (MSPC), 1-Palmitoyl-2-myristoyl-sn-glycero-3-phosphocholine (PMPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE), 1-Palmitoyl-2-oleoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (POPG), 1-Palmitoyl-2-stearoyl-sn-glycero-3-phosphocholine (PSPC), 1-Stearoyl-2-myristoyl-sn-glycero-3-phosphocholine (SMPC), 1-Stearoyl-2-oleoyl-sn-glycero-3-phosphocholine (SOPC) 및 1-Stearoyl-2-palmitoyl-sn-glycero-3-phosphocholine (SPPC) 로 이루어진 군으로부터 선택되는 1이상의 인지질일 수 있으나, 이에 한정되는 것은 아니다.(DLOPC), 1,2-Dilauroyl-sn-glycero-3-phosphate (DLPA), 1,2-Dilauroyl-sn-glycero-3-phosphocholine (DLPE), 1,2-Dilauroyl-sn-glycero-3- Phospho-rac-(1-glycerol) (DLPG), 1,2-Dilauroyl-sn-glycero-3-phosphoserine (DLPS), 1,2-Dimyristoyl-sn-glycero-3-phosphate (DMPA), 1,2 -Dimyristoyl-sn-glycero-3-phosphocholine (DMPC), 1,2-Dimyristoyl-sn-glycero-3-phosphoethanolamine (DMPE), 1,2-Dimyristoyl-snglycero-3-Phospho-rac-(1-glycerol) (DMPG), 1,2-Dimyristoyl-sn-glycero-3-phosphoserine (DMPS), 1,2-Dioleoyl-sn-glycero-3-phosphate (DOPA), 1,2-Dioleoyl-sn-glycero-3-phosphocholine (DOPC) , 1,2-Dioleoylsn-glycero-3-Phospho-rac-(1-glycerol) (DOPG), 1,2-Dioleoyl-sn-glycero-3-phosphoserine (DOPS), 1,2-Dipalmitoyl-sn-glycero -3-phosphate (DPPA), 1,2-Dipalmitoyl-sn-glycero-3-phosphocholine (DPPC), 1,2-Dipalmitoyl-sn-glycero-3-phosphoethanolamine (DPPE), 1,2-Dipalmitoyl-sn- glycero-3-Phospho-rac-(1-glycerol) (DPPG), 1,2-Dipalmitoyl-sn-glycero-3-phosphoserine (DPPS), 1,2-Distearoyl-sn-glycero-3-phosphate (DSPA), 1,2-Distearoyl-sn-glycero-3- phosphocholine (DSPC), 1,2-Distearoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (DSPG), 1,2-Distearoyl-sn-glycero-3-phosphoserine (DSPS), Egg-PC (EPC), Hydrogenated Egg PC (HEPC), Hydrogenated Soy PC (HSPC), 1-Myristoyl-sn-glycero-3-phosphocholine (LYSOPC MYRISTIC), 1-Palmitoyl-sn-glycero-3-phosphocholine (LYSOPC PALMITIC), 1-Stearoyl-sn-glycero-3-phosphocholine (LYSOPC STEARIC), 1-Myristoyl-2-palmitoyl-sn-glycero 3-phosphocholine (Milk Sphingomyelin MPPC), 1-Myristoyl-2-stearoyl-sn-glycero-3-phosphocholine (MSPC), 1-Palmitoyl-2- myristoyl-sn-glycero-3-phosphocholine (PMPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphocholine (POPC), 1-Palmitoyl-2-oleoyl-sn-glycero-3-phosphoethanolamine (POPE) , 1-Palmitoyl-2-oleoyl-sn-glycero-3-Phospho-rac-(1-glycerol) (POPG), 1-Palmitoyl-2-stearoyl-sn-glycero-3-phosphocholine (PSPC), 1-Stearoyl-2-myristoyl-sn-glycero-3-phosphocholine (SMPC), 1-Stearoyl-2-oleoyl-sn-glycero-3-phosphocholine (SOPC) and 1-Stearoyl-2-palmitoyl-sn-glycero- It may be one or more phospholipids selected from the group consisting of, but is not limited to, 3-phosphocholine (SPPC).
본 발명에 있어서, 상기 양이온성 지질은 Dimethyldioctadecyl-ammoniumbromide (DDAB), Dimethyldioctadecylammonium (DDAB), (N,N-dimethyl-N-([2-sperminecarboxamido]ethyl)-2,3-bis(dioleyloxy)-1-propaniminium pentahydrochloride) (DOSPA), (N-[1-(2,3-dioleyloxy)propyl]-N,N,Ntrimethylammonium) (DOTMA), (N-[1-(2,3-dioleoyloxy)propyl]-N,N,N-trimethylammonium) (DOTAP), 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol), N4-Cholesteryl-Spermine (GL67), 1,2-dioleyloxy-3-dimethylaminopropane (DODMA), 1,2-dilauroyl-sn-glycero-3-ethylphosphocholine (12:0 EPC)를 포함하는 O-alkyl phosphatidylcholines 유도체, N-(4-carboxybenzyl)-N,N-dimethyl-2,3-bis(oleoyloxy)propan-1-aminium (DOBAQ) 및 1,2-distearoyl-3-dimethylammonium-propane (18:0 DAP)을 포함하는 DAP 유도체로 이루어진 군으로부터 선택되는 1 이상의 양이온성 지질일 수 있으나, 이에 한정되는 것은 아니다.In the present invention, the cationic lipid is Dimethyldioctadecyl-ammoniumbromide (DDAB), Dimethyldioctadecylammonium (DDAB), (N,N-dimethyl-N-([2-sperminecarboxamido]ethyl)-2,3-bis(dioleyloxy)-1 -propaniminium pentahydrochloride) (DOSPA), (N-[1-(2,3-dioleyloxy)propyl]-N,N,Ntrimethylammonium) (DOTMA), (N-[1-(2,3-dioleoyloxy)propyl]- N,N,N-trimethylammonium) (DOTAP), 3ß-[N-(N',N'-dimethylaminoethane)-carbamoyl]cholesterol (DC-Chol), N4-Cholesteryl-Spermine (GL67), 1,2-dioleyloxy -3-dimethylaminopropane (DODMA), O-alkyl phosphatidylcholines derivatives containing 1,2-dilauroyl-sn-glycero-3-ethylphosphocholine (12:0 EPC), N-(4-carboxybenzyl)-N,N-dimethyl-2,3-bis(oleoyloxy) It may be at least one cationic lipid selected from the group consisting of DAP derivatives including propan-1-aminium (DOBAQ) and 1,2-distearoyl-3-dimethylammonium-propane (18:0 DAP), but is not limited thereto. no.
본 발명에 있어서, 상기 나노입자는 위에서 기재된 다양한 예들 이외에도 당업계에 펩타이드를 제시 또는 캡슐화 하기 위해 알려진 다양한 나노입자가 제한없이 사용될 수 있다.In the present invention, in addition to the various examples described above, various nanoparticles known in the art for presenting or encapsulating peptides can be used without limitation.
본 발명에 있어서, 상기 양이온성 지질은 바람직하게는 양이온성 콜레스테롤 유도체, 보다 바람직하게는 Monoarginine-cholesterol (MA-Chol)일 수 잇다.In the present invention, the cationic lipid may preferably be a cationic cholesterol derivative, more preferably Monoarginine-cholesterol (MA-Chol).
본 발명에 있어서, 상기 어쥬번트는 면역 자극성 단일사슬 또는 이중사슬 올리고뉴클레오타이드, poly(I:C), 면역자극성 저분자 화합물, 또는 이들의 조합일 수 있으나, 이에 제한되는 것은 아니다. In the present invention, the adjuvant may be, but is not limited to, an immunostimulatory single-chain or double-chain oligonucleotide, poly(I:C), an immunostimulatory small-molecule compound, or a combination thereof.
본 기술분야에서, 상기 면역 자극성 단일사슬 또는 이중사슬 올리고뉴클레오타이드는 유용한 어쥬번트(면역보조제)로서 알려져 있다. 이들은 종종 CpG 모티프(구아노신에 연결된 메틸화되어 있지 않은 시토신을 포함한 디뉴클레오타이드 서열)를 포함한다. TpG 모티프, 회문(palindrome) 배열, 복수의 연속한 티미딘 뉴클레오타이드(예를 들면 TTTT), 복수의 연속한 시토신 뉴클레오타이드(예를 들면 CCCC) 또는 폴리(dG) 배열을 포함한 올리고뉴클레오타이 드도 또한 이중가닥 RNA와 같이 공지의 어쥬번트이다. 이들이 다양한 면역 자극성 올리고뉴클레오타이드 중 하나를 본 발명과 함께 제한 없이 사용할 수 있다.In the art, immunostimulatory single- or double-stranded oligonucleotides are known as useful adjuvants. They often contain a CpG motif (a dinucleotide sequence comprising an unmethylated cytosine linked to a guanosine). Oligonucleotides containing a TpG motif, a palindrome sequence, a plurality of consecutive thymidine nucleotides (e.g., TTTT), a plurality of consecutive cytosine nucleotides (e.g., CCCC) or a poly(dG) sequence are also known adjuvants, as are double-stranded RNAs. Any of these various immunostimulatory oligonucleotides can be used without limitation in conjunction with the present invention.
본 발명에 있어서, 상기 올리고뉴클레오타이드는 예를 들어 10~100뉴클레오타이드, 예를 들면15~50뉴클레오타이드, 20~30뉴클레오타이드 또는 25~28뉴클레오타이드의 길이를 갖는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the oligonucleotide may be characterized by having a length of, for example, 10 to 100 nucleotides, for example, 15 to 50 nucleotides, 20 to 30 nucleotides, or 25 to 28 nucleotides, but is not limited thereto.
본 발명에 있어서, 상기 올리고뉴클레오타이드는 천연 뉴클레오타이드 또는 비천연 뉴클레오타이드로 구성되거나, 이들이 혼합되어 구성될 수 있다. 예를 들어, 상기 올리고뉴클레오타이드는 하나 이상의 포스포로티오에이트 결합을 포함하고, 및/또는 하나 이상의 2'-O-메틸 변형된 것을 특징으로 할 수 있다.In the present invention, the oligonucleotide may be composed of a natural nucleotide or a non-natural nucleotide, or may be composed of a mixture of these. For example, the oligonucleotide may be characterized by containing one or more phosphorothioate linkages and/or being modified with one or more 2'-O-methyl.
본 발명에 있어서, 상기 단일사슬 또는 이중사슬 올리고뉴클레오타이드는 CpG 올리고뉴클레오타이드, STING 활성 올리고뉴클레오타이드, 또는 이들의 조합인 것을 특징으로 할 수 있다.In the present invention, the single-chain or double-chain oligonucleotide may be characterized as being a CpG oligonucleotide, a STING activating oligonucleotide, or a combination thereof.
본 발명의 용어 "Stimulator of Interferon Genes (STING)"은 선천성 면역 반응에서 주요한 역할을 수행하는 분자인 인터페론 유전자의 자극인자(STING)를 의미한다. The term "Stimulator of Interferon Genes (STING)" of the present invention refers to stimulator of interferon genes (STING), a molecule that plays a major role in the innate immune response.
본 발명의 용어, CpG 올리고뉴클레오타이드(CpG oligodeoxynucleotide, 또는 CpG oligodeoxynucleotide, CpG ODN)는 메틸화되지 않은 시토신 트리포스페이트 디옥시뉴클레오타이드("C")와 구아닌 트리포스페이트 디옥시뉴클레오타이드("G")를 포함하는 짧은 단일-가닥 합성 DNA 분자로서, 면역자극제(immunostimulant)로 알려져 있다. 상기 CpG는 본 발명의 나노 백신의 구성성분으로 포함되는 경우 수지상 세포의 면역반응을 강화하는 어쥬번트로서의 역할을 수행할 수 있다. 본 발명의 실시예에서는 서열번호 151의 CpG 올리고뉴클레오타이드가 사용되었으나 이에 제한되는 것은 아니다.The term CpG oligonucleotide (CpG oligodeoxynucleotide, or CpG oligodeoxynucleotide, CpG ODN) of the present invention is a short single-stranded synthetic DNA molecule containing unmethylated cytosine triphosphate deoxynucleotide ("C") and guanine triphosphate deoxynucleotide ("G"), which is known as an immunostimulant. When the CpG is included as a component of the nano vaccine of the present invention, it can function as an adjuvant that enhances the immune response of dendritic cells. In the embodiment of the present invention, the CpG oligonucleotide of SEQ ID NO: 151 was used, but is not limited thereto.
본 발명에 있어서, 상기 면역자극성 저분자 화합물은 저분자 어쥬번트(small molecule adjuvant)와 상호호환적으로 사용되며, 합성 저분자 어쥬번트와 천연 저분자 어쥬번트를 포함한다. 상기 면역자극성 저분자 화합물 또는 저분자 어쥬번트의 예로는 monophosphoryl lipid A, Muramyl dipeptide, Bryostatin-1, Mannide monooleate (Montanide ISA 720), Squalene, QS21, Bis-(3',5')-cyclic dimeric guanosine monophosphate, PAM2CSK4, PAM3CSK4, Imiquimod, Resiquimod, Gardiquimod, cl075, cl097, Levamisole, 48/80, Bupivacaine, Isatoribine, Bestatin, Sm360320, 및 Loxoribine 등이 있으나, 이에 제한되는 것은 아니다. 저분자 어쥬번트에 대해서는 Flower DR et al. (Expert Opin Drug Discov. 2012 Sep;7(9):807-17.)에 기술되어 있다.In the present invention, the immunostimulatory small molecule compound is used interchangeably with a small molecule adjuvant, and includes a synthetic small molecule adjuvant and a natural small molecule adjuvant. Examples of the immunostimulatory small molecule compound or small molecule adjuvant include, but are not limited to, monophosphoryl lipid A, Muramyl dipeptide, Bryostatin-1, Mannide monooleate (Montanide ISA 720), Squalene, QS21, Bis-(3',5')-cyclic dimeric guanosine monophosphate, PAM2CSK4, PAM3CSK4, Imiquimod, Resiquimod, Gardiquimod, cl075, cl097, Levamisole, 48/80, Bupivacaine, Isatoribine, Bestatin, Sm360320, and Loxoribine. Small molecule adjuvants are described in Flower DR et al. (Expert Opin Drug Discov. 2012 Sep;7(9):807-17.).
본 발명에 있어서, 상기 나노입자는 어쥬번트 외에도 음이온성 약물을 포함할 수 있다. 예를 들어, 상기 음이온성 약물은 올리고뉴클레오타이드, 압타머, mRNA, siRNA, miRNA 또는 이들의 조합일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the nanoparticle may include an anionic drug in addition to an adjuvant. For example, the anionic drug may be, but is not limited to, an oligonucleotide, an aptamer, mRNA, siRNA, miRNA, or a combination thereof.
본 발명의 상기 나노입자의 제조예는 본 발명자들의 공개 문헌 Theranostics 2016, 6 (2), 192-203, 및 Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638와, 대한민국 특허 제10-2425028호에 예시되어 있다.Examples of the preparation of the nanoparticles of the present invention are exemplified in the inventors' published documents Theranostics 2016, 6 (2), 192-203, and Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638, and Korean Patent No. 10-2425028.
통상의 기술자는 상기 펩타이드의 면역원성을 심각하게 저해하지 않는 수준에서의 변형, 융합, 접합 또는 제형화가 수행될 수 있으며, 상기한 나노입자, 융합단백질 및 접합체가 본 발명의 펩타이드를 사용하는 일 실시 형태임을 자명하게 이해할 수 있다. 따라서, 상기 펩타이드는 상기 지질 나노입자, 융합단백질 또는 접합체 이외에도 펩타이드 치료제 분야에 잘 알려진 다양한 형태, 제형 또는 변형(modification)을 포함할 수 있다. Those skilled in the art will readily appreciate that modification, fusion, conjugation or formulation of the peptide may be performed at a level that does not significantly impair the immunogenicity of the peptide, and that the nanoparticles, fusion proteins and conjugates described above are embodiments of the peptides of the present invention. Accordingly, the peptides may include various forms, formulations or modifications well known in the field of peptide therapeutics in addition to the lipid nanoparticles, fusion proteins or conjugates.
본 발명의 펩타이드는 뛰어난 면역우성을 갖는 FAP의 에피토프 펩타이드로서, 본 발명의 펩타이드를 투여하는 경우 FAP에 특이적인 면역 반응을 유도할 수 있다.The peptide of the present invention is an epitope peptide of FAP having excellent immunodominance, and when the peptide of the present invention is administered, an immune response specific to FAP can be induced.
섬유아세포 활성화 단백질 알파(FAP)는 섬유증(fibrosis), 관절염(arthritis) 및 종양 또는 암을 포함하는 다양한 비종양성 또는 종양성 질환의 병리학적 상태에 대해 독점적으로 발현되는 II형 막관통 세린 프로테아제이다(Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803). 모든 인간 암종의 90% 이상에서 활성화된 암-연관 섬유아세포(CAF)로도 명명되는 활성화된 기질 섬유아세포(activiated stromal fibroblast)에서 발현되는 것으로 보고된다(Oncogene. 37 (32) (August 2018): 4343-4357; rontiers in Bioscience. 23: 1933-1968 (June 2018)). 암-연관 섬유아세포는 암의 발달, 성장 전이에 중요한 역할을 하는 것으로 보고되고 있으며, NASH에서 FAP는 FGF21을 절단하여, 지질의 축적을 유도하고 간지방증, 비알콜성 지방간염(NASH)등 다양한 질환을 유발할 수 있다.Fibroblast activation protein alpha (FAP) is a type II transmembrane serine protease exclusively expressed in the pathologic states of various non-neoplastic or neoplastic diseases, including fibrosis, arthritis, and tumors or cancer (Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803). It is reported to be expressed in activated stromal fibroblasts, also called activated cancer-associated fibroblasts (CAFs), in more than 90% of all human carcinomas (Oncogene. 37 (32) (August 2018): 4343-4357; rontiers in Bioscience. 23: 1933-1968 (June 2018)). It has been reported that cancer-associated fibroblasts play an important role in the development, growth, and metastasis of cancer, and in NASH, FAP cleaves FGF21, inducing lipid accumulation and causing various diseases such as hepatic steatosis and nonalcoholic steatohepatitis (NASH).
본 발명의 일 실시예에서, 중성-하전된 작은 지질 나노입자 (small lipid nanoparticle, SLNP) 기반의 백신 플랫폼을 이용하여, 상기 펩타이드를 포함하는 지질 나노입자를 제조하고, 상기 지질 나노입자로 면역화를 하는 경우 종양 성장을 효과적으로 억제할 수 있음을 확인하였으며, 특히 종양 미세환경에서 암-연관 섬유아세포를 효과적으로 제거하여, ECM의 형성을 감소시킬 수 있고, 종양세포에 대한 CD8+ T세포 반응 및 항원 특이적 CD4+T 세포 반응을 유도할 수 있음을 확인하였다.In one embodiment of the present invention, by using a vaccine platform based on neutrally-charged small lipid nanoparticles (SLNPs), lipid nanoparticles including the peptide were manufactured, and it was confirmed that when immunization is performed with the lipid nanoparticles, tumor growth can be effectively inhibited. In particular, it was confirmed that cancer-associated fibroblasts can be effectively removed from the tumor microenvironment, the formation of ECM can be reduced, and CD8+ T cell responses and antigen-specific CD4+ T cell responses against tumor cells can be induced.
본 발명의 다른 실시예에서는 본 발명의 펩타이드에 기반한 백신화 요법이 자가 면역과 관련한 Th17세포의 증가를 유도하지 않아 전신성 독성 및 자가면역 반응의 유발과 같은 부작용이 발생하지 않으면서, 기존에 보고된 FAP 표적화 암 백신에 비해 뛰어난 항암 효과를 나타낼 수 있음을 확인하였다. 나아가, 본 발명의 또 다른 실시예에서는 암의 성장 억제 및 치료 뿐만 아니라, 암의 전이를 예방 및 억제할 수 있음을 확인하였다.In another embodiment of the present invention, it was confirmed that the vaccination therapy based on the peptide of the present invention can exhibit excellent anticancer effects compared to previously reported FAP-targeting cancer vaccines without causing side effects such as induction of systemic toxicity and autoimmune response by not inducing an increase in Th17 cells related to autoimmunity. Furthermore, in another embodiment of the present invention, it was confirmed that it can prevent and inhibit cancer metastasis as well as inhibit and treat cancer growth.
본 발명의 또 다른 실시예에서는 본 발명의 펩타이드에 기반한 백신화 요법이 다른 FAP 관련 질환인 비알콜성 지방간염(NASH)에 대해 뛰어난 치료효과를 나타내는 것은 확인하였다.In another embodiment of the present invention, it was confirmed that a vaccination therapy based on the peptide of the present invention exhibited an excellent therapeutic effect on nonalcoholic steatohepatitis (NASH), another FAP-related disease.
따라서, 본 발명의 펩타이드가 갖는 뛰어난 면역우성은 FAP에 대한 강력한 면역 반응의 유도를 통해 상기 섬유증, 관절염, 비알콜성 지방간염 및 종양/암과 같은 FAP 연관 질환의 예방 또는 치료에 유용하게 사용될 수 있다.Therefore, the excellent immunodominance of the peptide of the present invention can be usefully used for the prevention or treatment of FAP-associated diseases such as fibrosis, arthritis, non-alcoholic steatohepatitis, and tumors/cancer through the induction of a strong immune response to FAP.
따라서, 본 발명은 또 다른 관점에서, 상기 펩타이드(및/또는 융합 단백질), 핵산 또는 나노입자를 포함하는 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용 백신 조성물에 관한 것이다.Therefore, the present invention, in another aspect, relates to a vaccine composition for preventing or treating a disease associated with fibroblast activation protein alpha (FAP), comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
본 발명의 용어 “FAP 관련 질환”은 FAP의 발현이 원인 또는 FAP의 과도한 발현이 병리학적 특징으로 연관되는 질환을 총칭하는 의미로 사용된다. 예를 들어, FAP 관련 질환은 섬유증, 관절염, 종양, 비알콜성 지방간염, 간 지방증, 동맥경화증 및 심근 경색 (Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803) 포함하나 이에 제한되는 것은 아니다.The term “FAP-related disease” in the present invention is used to collectively refer to diseases in which the expression of FAP is the cause or in which excessive expression of FAP is associated with pathological characteristics. For example, FAP-related diseases include, but are not limited to, fibrosis, arthritis, tumors, nonalcoholic steatohepatitis, hepatic steatosis, arteriosclerosis, and myocardial infarction (Cancer Metastasis Rev. 2020 Sep; 39(3): 783-803).
본 발명은 보다 구체적인 관점에서, 상기 펩타이드(및/또는 융합 단백질), 핵산 또는 나노입자를 포함하는 암 또는 종양의 예방 또는 치료용 백신 조성물에 관한 것이다.The present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating cancer or a tumor, comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
본 발명은 보다 구체적인 관점에서, 상기 펩타이드(및/또는 융합 단백질), 핵산 또는 나노입자를 포함하는 암 또는 종양의 전이 예방 또는 치료용 백신 조성물에 관한 것이다.The present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating metastasis of cancer or tumor, comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
본 발명은 보다 구체적인 관점에서, 상기 펩타이드(및/또는 융합 단백질), 핵산 또는 나노입자를 포함하는 비알콜성 지방간염(NASH)의 예방 또는 치료용 백신 조성물에 관한 것이다.The present invention relates, in a more specific aspect, to a vaccine composition for preventing or treating nonalcoholic steatohepatitis (NASH) comprising the peptide (and/or fusion protein), nucleic acid or nanoparticle.
본 발명에 있어서, 상기 백신 조성물은 하나 이상의 본 발명의 펩타이드를 포함하는 것을 특징으로 할 수 있다. 본 발명에 있어서, 상기 백신 조성물은 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 또는 그 이상의 종류의 서열을 포함하는 본 발명의 펩타이드를 포함하는 것을 특징으로 할 수 있다. 예를 들어, 상기 백신 조성물은 서열번호 1 내지 149로 구성된 군에서 선택되는 어느 하나 이상의 아미노산 서열을 포함할 수 있다. 본 발명의 백신 조성물에 포함된 하나 이상의 펩타이드는 앞서 설명한 것과 같이 에피토프 펩타이드, 이를 포함하는 융합 단백질, 나노입자 등의 다양한 형태일 수 있다. In the present invention, the vaccine composition may be characterized by comprising one or more peptides of the present invention. In the present invention, the vaccine composition may be characterized by comprising a peptide of the present invention comprising 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, or more kinds of sequences. For example, the vaccine composition may comprise any one or more amino acid sequences selected from the group consisting of SEQ ID NOs: 1 to 149. The one or more peptides included in the vaccine composition of the present invention may be in various forms, such as an epitope peptide, a fusion protein including the same, a nanoparticle, etc., as described above.
본 발명에 있어서, 상기 백신 조성물은 투여되는 대상의 HLA 타입에 따라 상이한 펩타이드 또는 이들의 조합을 포함하는 것을 특징으로 할 수 있다. 투여되는 대상의 HLA 타입은 당업계에 공지된 유전자 분석 방법을 사용하여 수행될 수 있다.In the present invention, the vaccine composition may be characterized by containing different peptides or a combination thereof depending on the HLA type of the subject to be administered. The HLA type of the subject to be administered can be performed using a genetic analysis method known in the art.
예를 들어, 본 발명에 있어서, 상기 대상이 HLA-A1 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 10 내지 29로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다. For example, in the present invention, when the subject has MHC of the HLA-A1 type, the vaccine composition may include one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
본 발명에 있어서, 상기 대상이 HLA-A2 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 1, 3, 4, 5, 및 30 내지 45로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다In the present invention, when the subject has MHC of the HLA-A2 type, the vaccine composition may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of
본 발명에 있어서, 상기 대상이 HLA-A3 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 46 내지 65로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다In the present invention, when the subject has MHC of the HLA-A3 type, the vaccine composition may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
본 발명에 있어서, 상기 대상이 HLA-A24 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 66 내지 85로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다. In the present invention, when the subject has MHC of the HLA-A24 type, the vaccine composition may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
본 발명에 있어서, 상기 대상이 HLA-A26 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 제한 에피토프 펩타이드는 서열번호 11, 12, 13, 14, 19, 21, 28, 58 및 86 내지 97로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다.In the present invention, when the subject has MHC of the HLA-A26 type, the vaccine composition may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97 as a restricted epitope peptide.
본 발명에 있어서, 상기 대상이 HLA-B7 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 30, 93 및 98 내지 115로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다In the present invention, when the subject has MHC of the HLA-B7 type, the vaccine composition may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
본 발명에 있어서, 상기 대상이 HLA-B8 타입의 MHC를 보유하는 경우, 서열번호 30, 91, 93, 99, 100, 103 및 116 내지 129로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다In the present invention, when the subject has MHC of the HLA-B8 type, it may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129.
본 발명에 있어서, 상기 대상이 HLA-B27 타입의 MHC를 보유하는 경우, 서열번호 130 내지 149로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드를 하나 이상 포함할 수 있다.In the present invention, when the subject possesses MHC of the HLA-B27 type, it may include at least one peptide comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149.
상기 HLA 타입에 따른 백신 조성물은 하나의 구현 예로서, 본 발명의 백신 조성물이 이에 제한되는 것은 아니며, 상이한 HLA 타입 제한 펩타이드를 하나 이상 포함할 수 있다.The vaccine composition according to the above HLA type is one embodiment, and the vaccine composition of the present invention is not limited thereto, and may include one or more different HLA type-restricted peptides.
본 발명의 용어, “백신 조성물”은 in vivo 또는 in vitro에서 항원 또는 면역원으로 작용하여, 면역반응을 이끌어 낼 수 있는 물질을 포함하는 조성물을 의미하며, 본 발명에서, “백신” 또는 “면역원성 조성물”과 동일한 의미에서 상호 호환적으로 사용될 수 있다.The term “vaccine composition” of the present invention means a composition containing a substance capable of inducing an immune response by acting as an antigen or immunogen in vivo or in vitro, and in the present invention, can be used interchangeably with “vaccine” or “immunogenic composition” in the same meaning.
본 발명에서의 용어 "면역 반응"은 선천 면역 반응 및 적응 면역 반응, 예를 들어, 보체 매개 면역 반응, 세포 매개된 (T 세포) 면역 반응 및/또는 항체 매개된 (B 세포) 반응을 모두 포함하는 최광의의 개념이다.The term “immune response” in the present invention is a broad concept that includes both innate immune responses and adaptive immune responses, for example, complement-mediated immune responses, cell-mediated (T cell) immune responses, and/or antibody-mediated (B cell) responses.
본 발명의 백신 조성물은 투여 대상에서 FAP 또는 FAP 발현 섬유아세포(CAF)에 대한 면역 반응을 유도 또는 증가시킬 수 있으며, 이를 통해, 섬유화증, 관절염, 종양, NASH과 같은 FAP 관련 질환에 대한 뛰어난 예방 또는 치료효과를 나타낼 수 있다. 보다 구체적인 예를 들어, 종양 미세환경에서의 FAP 발현 CAF의 결핍, ECM의 형성 억제, CD8+ T 세포 및 CD4+ T 세포 반응의 유도, 전이 억제, 면역세포와 항암제의 종양 침투능력 향상 효과를 포함하나, 이에 제한되는 것은 아니다.The vaccine composition of the present invention can induce or increase an immune response to FAP or FAP-expressing fibroblasts (CAFs) in a subject of administration, and thereby exhibit excellent preventive or therapeutic effects on FAP-related diseases such as fibrosis, arthritis, tumors, and NASH. More specific examples include, but are not limited to, effects of deficiency of FAP-expressing CAFs in a tumor microenvironment, inhibition of ECM formation, induction of CD8+ T cell and CD4+ T cell responses, inhibition of metastasis, and enhancement of tumor penetration ability of immune cells and anticancer agents.
본 발명에 있어서, 상기 백신 조성물은 본 발명의 펩타이드, 핵산, 또는 나노입자를 유효성분으로 포함할 수 있으며, 펩타이드 또는 핵산을 포함하는 경우 이를 효과적으로 제시 및 전달하기 위한 당업계에 알려진 다양한 수단이 사용될 수 있다. 본 발명에 있어서, 상기 나노입자는 펩타이드 또는 핵산을 제시하기 위한 전달체로서의 예로 이해될 수 있다.In the present invention, the vaccine composition may contain the peptide, nucleic acid, or nanoparticle of the present invention as an active ingredient, and when containing the peptide or nucleic acid, various means known in the art for effectively presenting and delivering it may be used. In the present invention, the nanoparticle may be understood as an example of a carrier for presenting the peptide or nucleic acid.
본 발명에 있어서, 상기 펩타이드는 위에서 설명한 나노입자의 형태 외에도 다양한 형태로 백신 조성물에 포함될 수 있다. 예를 들어, 융합 단백질, 접합체의 형태로 포함될 수 있으며, 또 다른 예를 들어, 엑소좀과 같은 생체 유래 막 또는 올리고멤브레인과 같은 인공막의 표면에 고정되어 제시되거나 내부에 캡슐화 되어 제시될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the peptide may be included in the vaccine composition in various forms other than the form of the nanoparticle described above. For example, it may be included in the form of a fusion protein or a conjugate, and for another example, it may be presented by being fixed to the surface of a biological membrane such as an exosome or an artificial membrane such as an oligomembrane, or presented by being encapsulated inside, but is not limited thereto.
본 발명에 있어서, 상기 펩타이드 또는 나노입자의 직접적인 전달의 대안적 방법으로서, 본 발명의 펩타이드를 코딩하는 핵산 또는 이를 포함하는 전달 벡터의 형태로 백신 조성물에 포함되어 in vivo 전달될 수 있으며, 전달된 핵산 또는 벡터는 생체 내에서 발현되어 생체 내에서 본 발명의 펩타이드를 합성 및 제시하도록 할 수 있다.In the present invention, as an alternative method of direct delivery of the peptide or nanoparticle, the peptide of the present invention may be delivered in vivo by being included in a vaccine composition in the form of a nucleic acid encoding the peptide of the present invention or a delivery vector including the same, and the delivered nucleic acid or vector may be expressed in vivo to synthesize and present the peptide of the present invention in vivo.
본 발명에 있어서, 상기 핵산의 전달을 위한 다양한 생체 내 단백질 발현 플랫폼이 사용될 수 있으며, 예를 들어 바이러스 벡터, 네이키드 DNA 또는 RNA를 포함하나, 이에 제한되는 것은 아니다.In the present invention, various in vivo protein expression platforms for delivery of the nucleic acid can be used, including but not limited to viral vectors, naked DNA or RNA.
본 발명에 있어서, 상기 핵산은 바이러스 벡터 또는 발현 플라스미드와 같은 비 바이러스 벡터에 포함되어 본 발명의 백신 조성물에 함유될 수 있다. 본 발명에 있어서, 상기 핵산의 발현을 위해 별도의 외인성 프로모터를 추가로 포함할 수 있으나, 이에 제한되는 것은 아니다. 핵산의 생체 내 전달 및 발현에 사용될 수 있는 다양한 벡터, 프로모터, 전달 기술 등이 당업계에 잘 알려져 있으며, 통상의 기술자는 대상 및 목적 등에 따라 제한 없이 알려진 기술을 사용하여 이를 수행할 수 있다.In the present invention, the nucleic acid may be contained in a non-viral vector such as a viral vector or an expression plasmid and may be included in the vaccine composition of the present invention. In the present invention, a separate exogenous promoter may be additionally included for the expression of the nucleic acid, but is not limited thereto. Various vectors, promoters, delivery technologies, etc. that can be used for the in vivo delivery and expression of nucleic acids are well known in the art, and a person skilled in the art can perform this without limitation using a known technology depending on the target and purpose.
본 발명에 있어서, 상기 백신 조성물은 인지질, 양이온성 지질 및/또는 어쥬번트(adjuvant)를 추가로 포함하는 것을 특징으로 할 수 있다.In the present invention, the vaccine composition may be characterized by additionally comprising a phospholipid, a cationic lipid, and/or an adjuvant.
본 발명의 용어 “어쥬번트(adjuvant)”는 Alexander Glenny에 의해 aluminum salt가 면역반응을 증가시킨다는 것이 발견되면서 생겨난 개념으로 백신 조성물 또는 백신을 투여하는 대상에서 더 강력한 면역반응을 유도하기 위해 넣는 보조 성분을 의미한다. 예를 들어, 상기 어쥬번트는, 유화제, 무라밀 디펩타이드, 아브리딘, 수성 어쥬번트, 오일 등을 포함할 수 있으며, 보다 구체적으로는, 알루미늄 포스페이트 또는 알루미늄 하이드록사이드와 같은 알루미늄 염, MF59 또는 이의 유사체(MF59 like), AS03 또는 이의 유사체(AS03 like), AF03 또는 이의 유사체(AF03 like), SE 또는 이의 유사체(SE like)와 같은 스쿠알렌(squalene) 함유 에멀젼(Emulsion), 칼슘 염, dsRNA 유사체, 지방다당질(Lipopolysaccharide), Lipid A 유사체 (MPL-A, GLA 등), 플라젤린(Flagellin), 이미다조퀴놀린(imidazoquinolines), CpG ODN, 미네랄 오일(mineral oil), 톨유사수용체(Toll-like receptor, TLR)의 길항제(agonist), C-형 렉틴 리간드, CD1d 리간드(α-galactosylceramide 등), 계면 활성제(detergent), 리포좀(liposome), QS21과 같은 사포닌(Saponin), 사이토카인(cytokine), 펩타이드 등이 있으나, 이에 제한되는 것은 아니다.The term “adjuvant” of the present invention is a concept that arose when Alexander Glenny discovered that aluminum salt increases immune response, and refers to an auxiliary component added to a vaccine composition or a subject to which a vaccine is administered to induce a stronger immune response. For example, the adjuvant may include an emulsifier, muramyl dipeptide, abridin, an aqueous adjuvant, an oil, and more specifically, an aluminum salt such as aluminum phosphate or aluminum hydroxide, a squalene-containing emulsion such as MF59 or an analog thereof (MF59 like), AS03 or an analog thereof (AS03 like), AF03 or an analog thereof (AF03 like), SE or an analog thereof (SE like), a calcium salt, a dsRNA analogue, a lipopolysaccharide, a Lipid A analogue (MPL-A, GLA, etc.), Flagellin, imidazoquinolines, CpG ODN, mineral oil, an agonist of a Toll-like receptor (TLR), a C-type lectin ligand, Examples include, but are not limited to, CD1d ligands (such as α-galactosylceramide), detergents, liposomes, saponins such as QS21, cytokines, and peptides.
본 발명에 있어서, 상기 어쥬번트는 또 다른 예를 들어, 암피겐 (Amphigen), LPS, 세균 세포벽 추출물, 세균 DNA, CpG 서열, poly(I:C), 합성 올리고뉴클레오타이드 및 이의 배합물 [참조: Schijins et al. (2000) Curr. Opin. Immunol. 12:456], 미코박테리알 플레이 (M. phlei) 세포벽 추출물 (MCWE) (미국 특허 번호 4,744,984), 엠.플레이 DNA (M-DNA) 및 M-DNA-엠.플레이 세포벽 복합체 (MCC)를 포함할 수 있다. In the present invention, the adjuvant may include, for example, Amphigen, LPS, bacterial cell wall extract, bacterial DNA, CpG sequence, poly(I:C), synthetic oligonucleotides and combinations thereof [see: Schijins et al. (2000) Curr. Opin. Immunol. 12:456], mycobacterial phlei (M. phlei) cell wall extract (MCWE) (U.S. Patent No. 4,744,984), M. phlei DNA (M-DNA) and M-DNA-M. phlei cell wall complex (MCC).
본 발명에 있어서, 상기 어쥬번트는 또 다른 예를 들어, 유화제로서 사용될 수 있는 화합물은 천연 및 합성 유화제 및 음이온성, 양이온성 및 비이온성 화합물을 포함할 수 있으며, 합성 화합물 중, 음이온성 유화제는, 예를 들어 라우르산 및 올레산의 칼슘, 나트륨 및 알루미늄 염, 지방산의 칼슘, 마그네슘 및 알루미늄 염, 및 유기 설포네이트, 예를 들어 나트륨 라우릴 설페이트를 포함하고, 상기 합성 양이온성 작용제는, 예를 들어 세틸트리에틸암모늄 브로마이드를 포함하며, 상기 합성 비이온성 작용제는 글리세릴에스테르 (예: 글리세릴 모노스테아레이트), 폴리에틸렌 글리콜 에스테르 및 에테르, 및 소르비탄 지방산 에스테르 (예: 소르비탄 모노팔미테이트) 및 이들의 폴리옥시에틸렌 유도체 (예: 폴리옥시에틸렌 소르비탄 모노팔미테이트)를 포함하나, 이에 제한되는 것은 아니다.In the present invention, the adjuvant may include, for example, compounds which can be used as an emulsifier, natural and synthetic emulsifiers and anionic, cationic and nonionic compounds, and among the synthetic compounds, the anionic emulsifiers include, for example, calcium, sodium and aluminum salts of lauric acid and oleic acid, calcium, magnesium and aluminum salts of fatty acids, and organic sulfonates, for example, sodium lauryl sulfate, the synthetic cationic agents include, for example, cetyltriethylammonium bromide, and the synthetic nonionic agents include, but are not limited to, glyceryl esters (e.g., glyceryl monostearate), polyethylene glycol esters and ethers, and sorbitan fatty acid esters (e.g., sorbitan monopalmitate) and polyoxyethylene derivatives thereof (e.g., polyoxyethylene sorbitan monopalmitate).
본 발명에 있어서, 상기 어쥬번트는 천연 유화제일 수 있으며, 상기 천연 유화제는 아카시아, 젤라틴, 레시틴 및 콜레스테롤을 포함하나 이에 제한되는 것은 아니다.In the present invention, the adjuvant may be a natural emulsifier, and the natural emulsifier includes, but is not limited to, acacia, gelatin, lecithin, and cholesterol.
본 발명에 있어서, 기타 적합한 어쥬번트는 예를 들어 오일 성분, 예를 들어 단일 오일, 오일 혼합물, 유중수 에멀젼 또는 수중유 에멀젼으로 형성될 수 있다. 오일은 광유, 식물성유 또는 동물성유일 수 있다. 광유는 증류 기술을 통해 바셀린로부터 수득되는 액체 탄화수소이며, 또한 당해 기술 분야에서 액체 파라핀, 액체 바셀린 또는 화이트 광유로도 언급된다. 적합한 동물성유는, 예를 들어 간유, 가자미유, 큰청어유, 오렌지 러기유 (orange roughy oil) 및 상어간유를 포함하며, 이들 모두 시판된다. 적합한 식물성유는, 예를 들어 카놀라유, 아몬드유, 면실유, 옥수 수유, 올리브유, 낙화생유, 홍화유, 참기름, 대두유 등을 포함한다. 프로인트 완전 어쥬번트 (FCA, 또는 CFA) 및 프로인트 불완전 어쥬번트 (FIA)는 백신 제제에 통상 사용되는 2가지의 일반적인 어쥬번트며, 또한 본 발명에서 사용하기에도 적합하다. FCA 및 FIA 모두 광유중수 에멀젼이다; 그러나, FCA는 사멸된 미코박테리움 종 (Mycobacterium sp.)도 또한 포함한다.In the present invention, other suitable adjuvants may be formed, for example, of oil components, for example, single oils, oil mixtures, water-in-oil emulsions or oil-in-water emulsions. The oils may be mineral oils, vegetable oils or animal oils. Mineral oils are liquid hydrocarbons obtained from petroleum jelly by distillation techniques and are also referred to in the art as liquid paraffin, liquid petroleum jelly or white mineral oil. Suitable animal oils include, for example, cod liver oil, flounder oil, herring oil, orange roughy oil and shark liver oil, all of which are commercially available. Suitable vegetable oils include, for example, canola oil, almond oil, cottonseed oil, corn oil, olive oil, peanut oil, safflower oil, sesame oil, soybean oil and the like. Freund's complete adjuvant (FCA, or CFA) and Freund's incomplete adjuvant (FIA) are two common adjuvants commonly used in vaccine formulations and are also suitable for use in the present invention. Both FCA and FIA are water-in-mineral-oil emulsions; however, FCA also contains killed Mycobacterium sp.
본 발명에 있어서, 면역조절 사이토카인도 백신 효능을 향상시키기 위해, 예를 들어 어쥬번트로서 백신 조성물에 포함될 수 있다. 이러한 사이토카인의 비제한적 예는 인터페론 알파 (IFN-α), 인터루킨-2 (IL-2) 및 과립구 대식세포-콜로니 자극 인자 (GM-CSF) 또는 이의 배합물을 포함하며, 바람직하게는 GM-CSF일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, immunomodulatory cytokines may also be included in the vaccine composition, for example as adjuvants, to enhance vaccine efficacy. Non-limiting examples of such cytokines include interferon alpha (IFN-α), interleukin-2 (IL-2), and granulocyte macrophage-colony stimulating factor (GM-CSF) or combinations thereof, preferably but not limited to GM-CSF.
바람직한 예를 들어, 본 발명의 실시예에서는 본 발명의 펩타이드를 CFA를 어쥬번트로 사용하여 투여하거나 본 발명의 펩타이드를 포함하는 나노입자와 함께CpG ODN을 어쥬번트로 사용하여 투여하여 면역화하였으나, 이에 제한되는 것은 아니다.For example, in a preferred embodiment of the present invention, immunization is performed by administering the peptide of the present invention using CFA as an adjuvant or by administering CpG ODN as an adjuvant together with nanoparticles comprising the peptide of the present invention, but is not limited thereto.
본 발명의 백신 조성물은 당해 발명이 속하는 기술분야에서 통상의 지식을 가진 자가 용이하게 실시할 수 있는 방법에 따라, 약제학적으로 허용되는 담체 및/또는 부형제를 이용하여 제형화함으로써 단위 용량 형태로 제조되거나 또는 다용량 용기 내에 내입시켜 제조될 수 있다. The vaccine composition of the present invention can be manufactured in a unit dose form or can be manufactured by placing it in a multi-dose container by formulating it using a pharmaceutically acceptable carrier and/or excipient according to a method that can be easily performed by a person having ordinary skill in the art to which the present invention pertains.
이 때의 제형은 통상의 방법에 따라 산제, 과립제, 정제, 캡슐제, 현탁액, 에멀젼, 시럽, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액의 형태로 제형화하여 사용될 수 있다. 당해 기술분야에 알려진 적합한 제제는 문헌(Remington's Pharmaceutical Science, Mack Publishing Company, EastonPA)에 개시되어 있는 것을 사용할 수 있다. 경구투여를 위한 고형제제에는 정제, 환제, 산제, 과립제, 캡슐제 등이 포함되며, 이러한 고형제제는 적어도 하나 이상의 부형제 예를 들면, 전분, 칼슘카보네이트, 수크로오스, 락토오스, 젤라틴 등을 섞어 조제된다. 또한 단순한 부형제 이외에 마그네슘 스테아레이트, 탈크 같은 윤활제들도 사용된다. 경구투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데 흔히 사용되는 단순 희석제인 물, 리퀴드 파라핀 이외에 여러 가지 부형제, 예를 들면 습윤제, 감미제, 방향제, 보존제 등이 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성용제, 현탁제, 유제, 동결건조 제제, 좌제가 포함된다. 좌제의 기제로는 위텝솔(witepsol), 마크로골, 트윈(tween) 61, 카카오지, 라우린지, 글리세로제라틴 등이 사용될 수 있다.At this time, the formulation can be formulated and used in the form of oral formulations such as powders, granules, tablets, capsules, suspensions, emulsions, syrups, aerosols, etc., external preparations, suppositories, and sterile injection solutions according to conventional methods. Suitable formulations known in the art can be used as disclosed in the literature (Remington's Pharmaceutical Science, Mack Publishing Company, Easton PA). Solid preparations for oral administration include tablets, pills, powders, granules, capsules, etc., and these solid preparations are prepared by mixing at least one excipient, such as starch, calcium carbonate, sucrose, lactose, gelatin, etc. In addition to simple excipients, lubricants such as magnesium stearate and talc are also used. Liquid preparations for oral administration include suspensions, solutions, emulsions, and syrups, and in addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, flavoring agents, and preservatives may be included. Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, and suppositories. Suppository bases include witepsol, macrogol, Tween 61, cacao butter, laurin butter, and glycerogelatin.
본 발명의 백신 조성물은 경구 또는 비경구 투여가 가능하다. 본 발명에 따른 조성물의 투여경로는 이들로 한정되는 것은 아니지만, 예를 들면, 피내, 정맥 내, 피하, 근육 내, 동맥 내, 골수 내, 경막 내, 심장 내, 경피, 복강 내, 장관, 설하, 구강 또는 국소 투여가 가능하다. 본 발명에 따른 조성물의 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 방법, 배설율 또는 질병의 중증도 등에 따라 그 범위가 다양하며, 본 기술분야의 통상의 전문가가 용이하게 결정할 수 있다. 또한, 임상 투여를 위해 공지의 기술을 이용하여 본 발명의 조성물을 적합한 제형으로 제제화할 수 있다.The vaccine composition of the present invention can be administered orally or parenterally. The route of administration of the composition according to the present invention is not limited to these, but for example, intradermal, intravenous, subcutaneous, intramuscular, intraarterial, intramedullary, intrathecal, intracardiac, transdermal, intraperitoneal, enteral, sublingual, oral or topical administration is possible. The dosage of the composition according to the present invention varies depending on the patient's weight, age, sex, health condition, diet, administration time, method, excretion rate or disease severity, and can be easily determined by a person skilled in the art. In addition, the composition of the present invention can be formulated into a suitable dosage form using a known technique for clinical administration.
본 발명에 있어서, 상기 백신 조성물의 투여는 단일 용량으로 투여되거나, 수회로 나누어 분할 투여될 수 있다. 분할 투여되는 경우, 본 발명의 펩타이드, 핵산, 나노입자 및/또는 어쥬번트의 용량은 동등하게 할당되는 것이 바람직하나, 이에 제한되는 것은 아니다.In the present invention, the vaccine composition may be administered as a single dose or divided into several doses. In the case of divided administration, it is preferable that the doses of the peptide, nucleic acid, nanoparticle and/or adjuvant of the present invention are equally distributed, but this is not limited thereto.
본 발명의 백신 조성물의 최적의 투여량은 피검체에서 적합한 면역 반응의 관찰을 포함하는 표준 연구에 의해 확인될 수 있다. 초기 면역화 후, 피검체는 적당한 간격을 두고 1회 또는 수 차례의 부스터 면역화 처리될 수 있다. The optimal dosage of the vaccine composition of the present invention can be determined by standard studies involving observation of an appropriate immune response in a subject. After the initial immunization, the subject may be administered one or more booster immunizations at appropriate intervals.
본 발명의 백신 조성물은 약제학적으로 유효한 양으로 투여될 수 있다. 본 발명의 용어 “약제학적으로 유효한 양”은 면역반응을 유도 또는 증가시킬 수 있을 정도로 충분하면서도 부작용이나 심각하거나 과도한 면역반응을 일으키지 않을 정도의 양을 의미하며, 적합한 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하게 결정될 수 있다. "약제학적으로 유효한 양" 결정시 고려되는 다양한 일반적인 사항들은 당업자에게 공지되어 있으며, 예를 들어, 문헌 [Gilman et al., eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed.,Pergamon Press, 1990] 및 [Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990]에 기재되어 있다.The vaccine composition of the present invention can be administered in a pharmaceutically effective amount. The term “pharmaceutically effective amount” as used herein means an amount sufficient to induce or increase an immune response but not causing side effects or serious or excessive immune responses, and an appropriate dosage may be variously determined by factors such as the formulation method, administration method, patient’s age, weight, sex, pathological condition, food, administration time, administration route, excretion rate, and response sensitivity. Various general considerations for determining a “pharmaceutically effective amount” are known to those skilled in the art and are described in, for example, the literature [Gilman et al., eds., Goodman And Gilman's: The Pharmacological Bases of Therapeutics, 8th ed., Pergamon Press, 1990] and [Remington's Pharmaceutical Sciences, 17th ed., Mack Publishing Co., Easton, Pa., 1990].
본 발명에 있어서, 상기 백신 조성물은 암의 예방 또는 치료에 대해 보호 면역을 유도 및/또는 지지하기 위하여, 세포독성 T 림프구 반응을 유도 및/또는 지지하기 위하여 적합한 임의의 스케줄로 환자에게 투여될 수 있다. 예를 들어, 일차 예방투여로 백신 조성물을 환자에게 투여한 후, 보호 면역을 지지 및/또는 유지하기 위해 부스터를 투여할 수 있다. 일부 양상에서, 백신 조성물은 1개월, 수개월 또는 수년당 1회, 2회 또는 그 이상의 횟수로 환자에게 투여될 수 있다. In the present invention, the vaccine composition can be administered to a patient on any suitable schedule to induce and/or support a cytotoxic T lymphocyte response to induce and/or support protective immunity for the prevention or treatment of cancer. For example, after the vaccine composition is administered to the patient as a primary prophylactic administration, a booster can be administered to support and/or maintain protective immunity. In some aspects, the vaccine composition can be administered to the patient once, twice or more times per month, several months or several years.
본 발명에 있어서, 상기 백신 조성물의 투여는 예를 들어 수년의 과정에 걸쳐 계속될 수 있다. 일반적으로, 백신 스케줄은 백신 요법의 시작시에 보다 빈번한 투여를 포함하며, 보호 면역을 유지하기 위한 시간 동안은 보다 덜 빈번한 투여 (예: 부스터)를 포함하나, 이에 제한되는 것은 아니다.In the present invention, administration of the vaccine composition may continue, for example, over the course of several years. Typically, the vaccine schedule includes, but is not limited to, more frequent administrations at the beginning of the vaccine regimen, and less frequent administrations (e.g., boosters) for a period of time to maintain protective immunity.
본 발명에 있어서, 상기 백신 조성물은 백신 요법의 시작시에는 보다 저용량이 투여되고, 시간이 지남에 따라 보다 고용량이 투여될 수 있다. 또한, 백신은 백신 요법의 시작시에는 보다 고용량이 투여되고, 시간이 지남에 따라 보다 저용량이 투여될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the vaccine composition may be administered in a lower dose at the beginning of the vaccine therapy and in a higher dose over time. In addition, the vaccine may be administered in a higher dose at the beginning of the vaccine therapy and in a lower dose over time, but is not limited thereto.
본 발명의 용어, “섬유화증(fibrosis)”은 결합 조직이 비정상적으로 생성되어 정상 실질 조직(normal parenchymal tissue)를 대체하는 병리학적 상처 치유를 의미한다. 섬유화증은 결합조직을 침착시킴으로써 기관 또는 조직의 정상적인 구조 및 기능을 방해 또는 차폐시킬 수 있으며, 이로 인한 다양한 신체적 이상을 야기할 수 있다. 섬유화증은 섬유아세포 활성화 단백질 알파의 과도한 발현을 원인 또는 병리학적 증상으로 할 수 있으며, 섬유아세포의 증식 및 활성화에 의한 결합조직의 비정상적인 침착을 포함할 수 있다.The term “fibrosis” of the present invention refers to pathological wound healing in which connective tissue is abnormally produced and replaces normal parenchymal tissue. Fibrosis can disrupt or block the normal structure and function of an organ or tissue by depositing connective tissue, and can cause various physical abnormalities as a result. Fibrosis can have excessive expression of fibroblast activation protein alpha as a cause or pathological symptom, and can include abnormal deposition of connective tissue due to proliferation and activation of fibroblasts.
본 발명에 있어서, 상기 종양은 FAP 관련 종양인 것을 특징으로 할 수 있다. In the present invention, the tumor may be characterized as being a FAP-related tumor.
본 발명에 있어서, 상기 FAP 관련 종양은 종양 조직 또는 종양미세환경에서의 유리 FAP 농도, FAP 발현 세포의 수 또는 세포의 FAP 발현 수준이 정상 조직 또는 환경에 비해 증가한 것을 특징으로 하는 종양을 모두 포함하는 의미로 사용된다. 기존의 보고에 따를 때, 거의 모든 종양에서 미세환경 내 유리 FAP 및/또는 FAP 발현 세포의 수가 정상 조직에 비해 증가한다. 따라서, 본 발명의 FAP 펩타이드를 사용한 백신화 요법 및 백신 조성물은 종양의 종류에 제한없이 효과적인 치료제로 사용될 수 있다.In the present invention, the FAP-related tumor is used to mean all tumors characterized by an increase in the concentration of free FAP in tumor tissue or tumor microenvironment, the number of FAP-expressing cells, or the level of FAP expression in cells compared to normal tissue or environment. According to existing reports, in almost all tumors, the number of free FAP and/or FAP-expressing cells in the microenvironment increases compared to normal tissue. Therefore, the vaccination therapy and vaccine composition using the FAP peptide of the present invention can be used as an effective therapeutic agent without limitation on the type of tumor.
본 발명의 용어, “종양” 은 생체 조절기구에서 이탈하여 세포가 자율성을 지니고 과잉 증식하는 현상 또는 이로 인해 생성된 신생물, 또는 과형성물을 모두 포함한다. 상기 종양은 예를 들어, 양성, 전악성, 악성 종양을 모두 포함하며, 보다 구체적인 예로, 조직세포종, 신경교종, 성상세포종, 골종, 각종 암, 예를 들어, 간암, 갑상선암, 결장암, 고환암, 골수이형성증후군, 교모세포종, 구강암, 구순암, 구인두암, 백혈병, 기저세포암, 난소암, 유방암, 뇌종양, 뇌하수체선종, 다발성골수종, 담낭암, 담도암, 대장암, 망막모세포종, 흑색종, 방광암, 복막암, 부갑상선암, 부신암, 비부비동암, 폐암, 소세포 폐암, 비소세포 폐암, 비호지킨림프종, 설암, 식도암, 소장암, 신우암, 신장암, 심장암, 십이지장암, 림프종(호지킨 림프종, FL, MCL, MZBL, CLL, T-ALL, AML, ALL 등), 외음부암, 요관암, 요도암, 위암, 위장관암, 장암, 육종, 골육종, 건선, 자궁경부암, 자궁암, 전립선암, 직장암, 질암, 췌장암, 피부암, 및 후두암 일 수 있으나, 이에 제한되는 것은 아니다.The term “tumor” of the present invention includes all neoplasms or hyperplasias that are caused by cells that have escaped from the biological control mechanism and have autonomous and excessive proliferation. The above tumors include, for example, benign, premalignant, and malignant tumors, and more specifically, histiocytoma, glioma, astrocytoma, osteoma, various cancers, for example, liver cancer, thyroid cancer, colon cancer, testicular cancer, myelodysplastic syndrome, glioblastoma, oral cancer, lip cancer, oropharyngeal cancer, leukemia, basal cell carcinoma, ovarian cancer, breast cancer, brain tumor, pituitary adenoma, multiple myeloma, gallbladder cancer, biliary tract cancer, colon cancer, retinoblastoma, melanoma, bladder cancer, peritoneal cancer, parathyroid cancer, adrenal cancer, paranasal sinus cancer, lung cancer, small cell lung cancer, non-small cell lung cancer, non-Hodgkin's lymphoma, tongue cancer, esophageal cancer, small intestine cancer, renal pelvis cancer, kidney cancer, heart cancer, duodenal cancer, lymphoma (Hodgkin's lymphoma, FL, MCL, MZBL, CLL, T-ALL, AML, ALL, etc.), vulvar cancer, These may include, but are not limited to, ureteral cancer, urethral cancer, stomach cancer, gastrointestinal cancer, intestinal cancer, sarcoma, osteosarcoma, psoriasis, cervical cancer, uterine cancer, prostate cancer, rectal cancer, vaginal cancer, pancreatic cancer, skin cancer, and laryngeal cancer.
본 발명에 있어서, 상기 종양은 바람직하게는 결합조직성 종양(Desmoplastic tumor)인 것을 특징으로 할 수 있다.In the present invention, the tumor may preferably be characterized as being a desmoplastic tumor.
본 발명에서 사용되는 용어, "예방"은 본 발명에 따른 백신 조성물의 투여에 의해 목적하는 질환을 억제시키거나 발병을 지연시키는 모든 행위를 의미한다.The term “prevention” as used in the present invention means any act of suppressing or delaying the onset of a desired disease by administering a vaccine composition according to the present invention.
본 발명에서 사용되는 용어, "치료"는 본 발명에 따른 백신 조성물의 투여에 의해 목적하는 질환에 대한 증세가 호전되거나 이롭게 변경되는 모든 행위를 의미한다.The term "treatment" as used in the present invention means any action by which the symptoms of a target disease are improved or beneficially changed by administration of a vaccine composition according to the present invention.
상기 백신 조성물은 통상적으로 백신 조성물에 사용되는 적절한 담체, 부형제 및 희석제를 추가로 포함할 수 있다.The above vaccine composition may additionally contain suitable carriers, excipients and diluents commonly used in vaccine compositions.
상기 백신 조성물에 포함될 수 있는 담체, 부형제 및 희석제는 락토오스, 덱스트로오스, 수크로오스, 소르비톨, 만니톨, 자일리톨, 에리스리톨, 말티톨, 전분, 아카시아 고무, 알지네이트, 젤라틴, 칼슘 포스페이트, 칼슘 실리케이트, 셀룰로오스, 메틸 셀룰로오스, 미정질 셀룰로오스, 폴리비닐 피롤리돈, 물, 메틸히드록시 벤조에이트, 프로필히드록시 벤조에이트, 탈크, 마그네슘 스테아레이트 및 광물유 등이 있다. 상기 조성물을 제제화할 경우에는 보통 사용하는 충진제, 증량제, 결합제, 습윤제, 붕해제, 계면활성제 등의 희석제 또는 부형제를 사용하여 조제된다.Carriers, excipients and diluents that may be included in the above vaccine composition include lactose, dextrose, sucrose, sorbitol, mannitol, xylitol, erythritol, maltitol, starch, acacia gum, alginate, gelatin, calcium phosphate, calcium silicate, cellulose, methyl cellulose, microcrystalline cellulose, polyvinyl pyrrolidone, water, methyl hydroxy benzoate, propyl hydroxy benzoate, talc, magnesium stearate and mineral oil. When formulating the above composition, it is prepared using diluents or excipients such as commonly used fillers, extenders, binders, wetting agents, disintegrants and surfactants.
본 발명에 따른 백신 조성물은 통상의 방법에 따라 다양한 형태로 제형화하여 사용될 수 있다. 적합한 제형으로는 정제, 환제, 산제, 과립제, 당의정, 경질 또는 연질의 캡슐제, 용액제, 현탁제 또는 유화액제, 주사제, 에어로졸 등의 경구형 제형, 외용제, 좌제 및 멸균 주사용액 등이 있으나, 이에 한정되는 것은 아니다.The vaccine composition according to the present invention can be formulated and used in various forms according to conventional methods. Suitable formulations include, but are not limited to, oral formulations such as tablets, pills, powders, granules, sugar-coated tablets, hard or soft capsules, solutions, suspensions or emulsions, injections, aerosols, external preparations, suppositories, and sterile injection solutions.
본 발명에 따른 백신 조성물은 약학적으로 불활성인 유기 또는 무기 담체를 이용하여 적합한 제형으로 제조할 수 있다. 즉, 제형이 정제, 코팅된 정제, 당의정 및 경질 캡슐제인 경우 락토스, 수크로스, 전분 또는 그 유도체, 탈크, 칼슘 카보네이트, 젤라틴, 스테아르산 또는 그 염을 포함할 수 있다. 또한, 제형이 연질 캡슐제인 경우에는 식물성 오일, 왁스, 지방, 반고체 및 액체의 폴리올을 포함할 수 있다. 또한, 제형이 용액 또는 시럽 형태인 경우, 물, 폴리올, 글리세롤, 및 식물성 오일 등을 포함할 수 있다.The vaccine composition according to the present invention can be prepared into a suitable formulation using a pharmaceutically inactive organic or inorganic carrier. That is, when the formulation is a tablet, a coated tablet, a sugar-coated tablet, or a hard capsule, it can contain lactose, sucrose, starch or a derivative thereof, talc, calcium carbonate, gelatin, stearic acid, or a salt thereof. In addition, when the formulation is a soft capsule, it can contain vegetable oil, wax, fat, semi-solid, and liquid polyols. In addition, when the formulation is in the form of a solution or syrup, it can contain water, polyol, glycerol, and vegetable oil, etc.
본 발명에 있어서, 상기 백신 조성물은 지연 방출 비히클 또는 데포 (depot) 제제로 제형화될 수 있다. 이러한 장기간 작용 제형은 접종 또는 이식 (예를 들어 피하 또는 근육내)에 의하거나 주사에 의해 투여될 수 있다. 따라서, 예를 들어, 백신 조성물은 적합한 중합체성 또는 소수성 물질 (예를 들어, 허용되는 오일 중의 에멀젼으로서) 또는 이온 교환 수지와 함께, 또는 난용성 유도체, 예를 들어 난용성 염으로서 제형화될 수 있다. 리포좀 및 에멀젼은 담체로서 사용하기에 적합한 전달 비히클로서 널리 알려진 예이다.In the present invention, the vaccine composition may be formulated as a delayed release vehicle or a depot preparation. Such a long-acting preparation may be administered by inoculation or implantation (e.g., subcutaneously or intramuscularly) or by injection. Thus, for example, the vaccine composition may be formulated with a suitable polymeric or hydrophobic material (e.g., as an emulsion in an acceptable oil) or with an ion exchange resin, or as a sparingly soluble derivative, e.g., a sparingly soluble salt. Liposomes and emulsions are well known examples of delivery vehicles suitable for use as carriers.
본 발명에 따른 백신 조성물은 상기의 담체 외에도 보존제, 안정화제, 습윤제, 유화제, 용해제, 감미제, 착색제, 삼투압 조절제, 산화방지제 등을 더 포함할 수 있다.The vaccine composition according to the present invention may further include, in addition to the carrier described above, a preservative, a stabilizer, a wetting agent, an emulsifier, a solubilizer, a sweetener, a colorant, an osmotic pressure regulator, an antioxidant, etc.
본 발명에 따른 백신 조성물은 약학적으로 유효한 양으로 투여한다. 본 발명에 있어서, "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분한 양을 의미하며, 유효용량 수준은 환자의 질환의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명에 따른 백신 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와는 순차적 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 상기한 요소들을 모두 고려하여 부작용 없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The vaccine composition according to the present invention is administered in a pharmaceutically effective amount. In the present invention, the "pharmaceutically effective amount" means an amount sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment, and the effective dosage level can be determined according to the type and severity of the patient's disease, the activity of the drug, the sensitivity to the drug, the time of administration, the route of administration and the excretion rate, the treatment period, the concurrently used drugs, and other factors well known in the medical field. The vaccine composition according to the present invention can be administered as an individual therapeutic agent or in combination with other therapeutic agents, can be administered sequentially or simultaneously with conventional therapeutic agents, and can be administered singly or in multiple doses. It is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects by considering all of the above factors, and this can be easily determined by those skilled in the art.
본 발명의 약학 조성물은 개체에 다양한 경로로 투여될 수 있다. 상기 약학 조성물은 경구 또는 비경구로 투여할 수 있다. 비경구 투여인 경우에는 정맥내 주입, 피하 주입, 근육 주입, 복강 주입, 내피 투여, 국소 투여, 비내 투여, 폐내 투여 및 직장내 투여 등으로 투여할 수 있다. 경구 투여 시, 단백질 또는 펩타이드는 소화가 되기 때문에 경구용 조성물은 활성 약제를 코팅하거나 위에서의 분해로부터 보호되도록 제형화될 수 있다. 또한, 상기 조성물은 활성 물질이 표적 세포로 이동할 수 있는 임의의 장치에 의해 투여될 수 있다.The pharmaceutical composition of the present invention can be administered to a subject by various routes. The pharmaceutical composition can be administered orally or parenterally. In the case of parenteral administration, it can be administered by intravenous injection, subcutaneous injection, intramuscular injection, intraperitoneal injection, intradermal administration, topical administration, intranasal administration, intrapulmonary administration, and rectal administration. When administered orally, since proteins or peptides are digested, the oral composition can be formulated to coat the active agent or protect it from decomposition in the stomach. In addition, the composition can be administered by any device that allows the active agent to travel to the target cell.
본 발명에 따른 백신 조성물의 투여방법은 제형에 따라 용이하게 선택될 수 있으며, 경구 또는 비경구 투여될 수 있다. 투여량은 환자의 나이, 성별, 체중, 병증의 정도, 투여경로에 따라 달라질 수 있다.The method of administration of the vaccine composition according to the present invention can be easily selected according to the formulation, and can be administered orally or parenterally. The dosage may vary depending on the patient's age, sex, weight, degree of disease, and route of administration.
본 발명에 있어서, 상기 백신 조성물은 FAP 특이적 세포독성 T 세포 반응 또는 FAP 특이적 CD8+ T 세포 반응을 유도하는 것을 특징으로 할 수 있다. 본 발명에 있어서, 상기 백신 조성물은 대상에서 유리 FAP 또는 FAP 발현 세포에 대한 면역 반응을 유도하는 것을 특징으로 할 수 있다. 본 발명에 있어서, 상기 FAP 발현 세포는 FAP 발현 섬유아세포인 것을 특징으로 할 수 있다.In the present invention, the vaccine composition may be characterized by inducing a FAP-specific cytotoxic T cell response or a FAP-specific CD8+ T cell response. In the present invention, the vaccine composition may be characterized by inducing an immune response to free FAP or FAP-expressing cells in a subject. In the present invention, the FAP-expressing cells may be characterized by being FAP-expressing fibroblasts.
본 발명에 있어서, 상기 백신 조성물은 항종양 반응을 유도하는 것을 특징으로 할 수 있다.In the present invention, the vaccine composition may be characterized by inducing an anti-tumor response.
본 발명에 있어서, 상기 항종양 반응은 종양 또는 종양미세환경 특이적 면역반응의 유도, 종양 세포의 수 감소, 종양 크기 감소, 종양 세포의 사멸, 종양의 전이 억제 등일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the anti-tumor response may include, but is not limited to, induction of a tumor or tumor microenvironment-specific immune response, reduction in the number of tumor cells, reduction in tumor size, death of tumor cells, inhibition of tumor metastasis, etc.
본 발명에 있어서, 상기 백신 조성물은 간에서의 지질 축적 억제, 간 지방증 억제, 간의 염증 억제, 간의 섬유화 억제, 간에서의 팽창 세포 비율 감소를 유도할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the vaccine composition can induce inhibition of lipid accumulation in the liver, inhibition of hepatic steatosis, inhibition of hepatic inflammation, inhibition of hepatic fibrosis, and reduction of the proportion of swollen cells in the liver, but is not limited thereto.
본 발명은 또 다른 관점에서, 본 발명의 펩타이드, 핵산 또는 나노입자의 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료용 약학 조성물의 제조를 위한 용도에 관한 것이다. In another aspect, the present invention relates to the use of the peptide, nucleic acid or nanoparticle of the present invention for the manufacture of a pharmaceutical composition for the prevention or treatment of a fibroblast activating protein alpha (FAP)-related disease.
본 발명은 또 다른 관점에서, 본 발명의 펩타이드, 핵산 또는 나노입자를 대상에게 투여하거나, 본 발명의 백신 조성물을 대상에게 투여하는 단계를 포함하는 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료 방법에 관한 것이다.In another aspect, the present invention relates to a method for preventing or treating a disease associated with fibroblast activation protein alpha (FAP), comprising administering to a subject a peptide, nucleic acid or nanoparticle of the present invention, or administering to a subject a vaccine composition of the present invention.
본 발명의 예방 또는 치료 방법은 섬유아세포 활성화 단백질 알파(FAP) 관련 질환의 예방 또는 치료를 위한, 백신 접종 방법, 백신화 방법, 또는 면역화 방법을 포함한다.The preventive or therapeutic method of the present invention includes a vaccination method, a vaccination method, or an immunization method for preventing or treating a disease associated with fibroblast activation protein alpha (FAP).
본 발명에 있어서, 상기 섬유아세포 활성화 단백질 알파 관련 질환의 예방 또는 치료방법에서, 상기 본 발명의 펩타이드, 핵산, 나노입자 또는 이들 중 어느 하나 이상을 포함하는 본 발명의 백신 조성물을 대상에게 투여하는 단계 이전에 대상의 HLA 타입을 분석하는 단계를 추가로 포함할 수 있다.In the present invention, in the method for preventing or treating a disease related to fibroblast activation protein alpha, a step of analyzing the HLA type of the subject may be additionally included prior to the step of administering to the subject the vaccine composition of the present invention comprising the peptide, nucleic acid, nanoparticle or any one or more of these of the present invention.
본 발명에 있어서, 상기 대상의 HLA 타입에 따라 다른 펩타이드를 포함하는 백신 조성물이 투여될 수 있다.In the present invention, a vaccine composition containing different peptides can be administered depending on the HLA type of the subject.
예를 들어, 본 발명에 있어서, 상기 대상이 HLA-A1 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 10 내지 29로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다. For example, in the present invention, when the subject has MHC of the HLA-A1 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 10 to 29.
본 발명에 있어서, 상기 대상이 HLA-A2 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 1, 3, 4, 5, 및 30 내지 45로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject possesses MHC of the HLA-A2 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 1, 3, 4, 5, and 30 to 45.
본 발명에 있어서, 상기 대상이 HLA-A3 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 46 내지 65로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject possesses MHC of the HLA-A3 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 46 to 65.
본 발명에 있어서, 상기 대상이 HLA-A24 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 66 내지 85로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject has MHC of the HLA-A24 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 66 to 85.
본 발명에 있어서, 상기 대상이 HLA-A26 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 제한 에피토프 펩타이드는 서열번호 11, 12, 13, 14, 19, 21, 28, 58 및 86 내지 97로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject has MHC of the HLA-A26 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 11, 12, 13, 14, 19, 21, 28, 58, and 86 to 97 as the restricted epitope peptide.
본 발명에 있어서, 상기 대상이 HLA-B7 타입의 MHC를 보유하는 경우, 상기 백신 조성물은 서열번호 30, 93 및 98 내지 115로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject has MHC of the HLA-B7 type, the vaccine composition may be administered with one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 93, and 98 to 115.
본 발명에 있어서, 상기 대상이 HLA-B8 타입의 MHC를 보유하는 경우, 서열번호 30, 91, 93, 99, 100, 103 및 116 내지 129로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다. In the present invention, when the subject possesses MHC of the HLA-B8 type, one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 30, 91, 93, 99, 100, 103, and 116 to 129 may be administered.
본 발명에 있어서, 상기 대상이 HLA-B27 타입의 MHC를 보유하는 경우, 서열번호 130 내지 149로 구성된 군에서 선택되는 아미노산 서열을 포함하거나 구성되는 펩타이드가 하나 이상 투여될 수 있다.In the present invention, when the subject possesses MHC of the HLA-B27 type, one or more peptides comprising or consisting of an amino acid sequence selected from the group consisting of SEQ ID NOs: 130 to 149 may be administered.
상기 HLA 타입에 따른 펩타이드 또는 백신 조성물의 투여는 하나의 구현 예로서, 이에 제한되는 것은 아니며, 대상의 HLA 타입에 관계 없이, 또는 HLA 타입에 대한 분석 단계 없이, 상이한 HLA 타입 제한 펩타이드가 하나 이상 투여될 수 있다.Administration of peptide or vaccine compositions according to the above HLA type is one embodiment and is not limited thereto, and one or more different HLA type restricted peptides may be administered regardless of the HLA type of the subject or without an analysis step for the HLA type.
따라서, 본 발명은 다른 관점에서 본 발명의 펩타이드, 핵산 또는 나노입자를 포함하는 항암제와의 병용 투여를 위한 종양의 예방 또는 치료용 약학적 조성물에 관한 것이다.Therefore, the present invention relates to a pharmaceutical composition for the prevention or treatment of tumors for co-administration with an anticancer agent comprising the peptide, nucleic acid or nanoparticle of the present invention from another aspect.
본 발명은 또 다른 관점에서, 상기 펩타이드, 핵산 또는 나노입자의 종양의 예방 또는 치료를 위한 항암제와의 병용투여 용도를 제공한다.In another aspect, the present invention provides a use of the peptide, nucleic acid or nanoparticle in combination with an anticancer agent for the prevention or treatment of tumors.
본 발명은 다른 관점에서, 상기 펩타이드, 핵산 또는 나노입자; 및 항암제를 대상에게 병용 투여하는 단계를 포함하는 종양의 예방 또는 치료방법을 제공한다.In another aspect, the present invention provides a method for preventing or treating a tumor, comprising a step of co-administering the peptide, nucleic acid or nanoparticle; and an anticancer agent to a subject.
본 발명은 다른 관점에서 본 발명의 펩타이드, 핵산 또는 나노입자의 항암제와의 병용 투여를 위한 종양의 예방 또는 치료용 약학적 조성물의 제조를 위한 용도에 관한 것이다.The present invention relates from another aspect to the use of the peptide, nucleic acid or nanoparticle of the present invention for the preparation of a pharmaceutical composition for the prevention or treatment of tumors in combination with an anticancer agent.
본 발명의 펩타이드, 핵산 및/또는 나노입자는 종양 미세 환경에서, FAP 발현 CAF를 감소 또는 고갈시켜, 항암요법의 물리적 장벽으로 작용하는 ECM의 형성을 억제하고, 결과적으로 항암제 및 면역세포의의 종양 내 침윤 또는 침투를 강화하며, 결과적으로는 각각의 약물을 단일요법으로 투여하는 것보다 현저한 항-종양효과를 나타낸다. 따라서, 본 발명의 약학적 조성물과 병용투여되는 항암제는 당업계에 보고된 다양한 항암요법제 또는 항암제로부터 제한 없이 선택될 수 있다.The peptides, nucleic acids and/or nanoparticles of the present invention inhibit the formation of ECM, which acts as a physical barrier to anticancer therapy, by reducing or depleting FAP-expressing CAFs in the tumor microenvironment, and consequently enhance the infiltration or penetration of anticancer agents and immune cells into the tumor, resulting in a more remarkable anti-tumor effect than when each drug is administered as a monotherapy. Therefore, the anticancer agent to be co-administered with the pharmaceutical composition of the present invention can be selected without limitation from various anticancer therapeutic agents or anticancer agents reported in the art.
본 발명에 있어서, 상기 항암제는 예를 들어, 화학항암제(세포독성 항암제), 표적항암제, 면역항암제, 호르몬성 항암제, 세포치료제 등일 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the anticancer agent may be, for example, a chemotherapy agent (cytotoxic anticancer agent), a targeted anticancer agent, an immunotherapy agent, a hormonal anticancer agent, a cell therapy agent, etc., but is not limited thereto.
본 발명에 있어서, 상기 화학항암제는 세포독성 항암제로도 불리며, 암세포를 직접 공격하여 항암효과를 나타내는 항암제로 예를 들어, 사이클로포스파미드, 이포스파미드, 벤다무스틴, 델팔란 및 시스플라틴 등과 같은 알킬화제; 카페시타딘, 시타라빈, 독시플루리딘, 플루오로우라실, 클로파라빈, 플루다라빈 및 데시타민 등과 같은 대사 길항제; 독소루비신, 다우노루비신, 이다루비신, 에토포시드 및 토포테칸과 같은 DNA 회전효소 저해제; 카바지탁셀, 도세탁셀, 빈크리스틴 등과 같은 미세소관 저해제; 및 미토마이신 C, 블레오마이신, 히드록시우레아와 같은 기타 화학항암제등을 포함하나, 이에 제한되는 것은 아니다.In the present invention, the chemical anticancer agent is also called a cytotoxic anticancer agent, and is an anticancer agent that directly attacks cancer cells and exhibits an anticancer effect, and includes, but is not limited to, alkylating agents such as cyclophosphamide, ifosfamide, bendamustine, delphalan, and cisplatin; antimetabolites such as capecitadine, cytarabine, doxifluridine, fluorouracil, clofarabine, fludarabine, and decitamine; DNA rotator cuff inhibitors such as doxorubicin, daunorubicin, idarubicin, etoposide, and topotecan; microtubule inhibitors such as cabazitaxel, docetaxel, vincristine, and the like; and other chemical anticancer agents such as mitomycin C, bleomycin, and hydroxyurea.
본 발명에 있어서, 표적 항암제는 암이 발현하는 특정 항원 또는 암과 관련된 항원을 표적으로 하여 이에 결합 또는 상호작용하는 물질을 주요성분으로 하는 항암제로, 예를들어, 세툭시맙, 트라스트주맙, 베바시주맙, 리툭시맙, 이블리투모맙, 알렘투주맙, 브렌툭시맙 및 엘로투주맙과 같은 항체 치료제; 및 엘로티닙, 게피티닙, 반데탄닙, 아파티닙, 라파니팁, 엑시티닙, 파조파닙, 수니티닙, 소라페닙 등과 같은 신호전달 억제제를 포함하나 이에 제한되는 것은 아니다.In the present invention, the targeted anticancer agent is an anticancer agent containing as a main component a substance that targets and binds to or interacts with a specific antigen expressed by a cancer or an antigen related to a cancer, and examples thereof include, but are not limited to, antibody therapeutics such as cetuximab, trastuzumab, bevacizumab, rituximab, ibltumomab, alemtuzumab, brentuximab, and elotuzumab; and signal transduction inhibitors such as erlotinib, gefitinib, vandetanib, afatinib, rapanitib, axitinib, pazopanib, sunitinib, and sorafenib.
본 발명에 있어, 상기 면역항암제는 아필리무맙과 같은 CTLA-4 항체, 펨브롤리주맙, 니볼루맙과 같은 PD-1 항체, 아테졸리주맙과 같은 PD-L1 항체, IDO 억제제와 같은 면역 체크포인트 억제제를 포함하나 이에 제한되는 것은 아니다.In the present invention, the immunotherapy anticancer agent includes, but is not limited to, a CTLA-4 antibody such as apilimumab, a PD-1 antibody such as pembrolizumab or nivolumab, a PD-L1 antibody such as atezolizumab, and an immune checkpoint inhibitor such as an IDO inhibitor.
본 발명에 있어서, 상기 호르몬성 항암제는, 비칼루타미드, 엔잘루타미드와 같은 남성호르몬 억제제 및 타목시펜, 아나스트로졸 레트로졸 과 같은 여성 호르몬 억제제등을 포함하나 이에 제한되는 것은 아니다.In the present invention, the hormonal anticancer agent includes, but is not limited to, androgen suppressants such as bicalutamide and enzalutamide, and female hormone suppressants such as tamoxifen, anastrozole and letrozole.
본 발명에 있어서, 상기 세포치료제는 살아있는 면역세포를 유효성분으로 하는 치료제를 의미하며, 세포독성 T 세포 치료제, CAR-T 세포치료제, CAR-NK 세포 치료제 등이 포함되나, 이에 제한되는 것은 아니다.In the present invention, the cell therapy agent means a therapy agent containing living immune cells as an active ingredient, and includes, but is not limited to, cytotoxic T cell therapy agents, CAR-T cell therapy agents, and CAR-NK cell therapy agents.
본 발명의 용어 "병용 투여"는 두 종류 이상의 유효 성분을 동시 또는 순차적으로 투여하거나, 두 종류 이상의 유효 성분의 작용으로 각각의 유효 성분을 독립적으로 투여하였을 때 기대되는 효과보다 향상된 효과를 나타낼 수 있도록 동시, 순차 또는 특정한 간격을 두어 투여하는 것을 의미한다.The term "combined administration" of the present invention means administering two or more types of effective ingredients simultaneously or sequentially, or administering them simultaneously, sequentially, or at specific intervals so that an improved effect can be achieved compared to the effect expected when each effective ingredient is administered independently due to the action of the two or more types of effective ingredients.
본 발명에 있어서, 상기 병용 투여는 본 발명의 펩타이드, 핵산 또는 나노입자와 하나 이상의 항암제를 병용하여 투여하는 것을 특징으로 할 수 있으며, 이외에도 다른 항종양요법과 병행하여 수행될 수 있다.In the present invention, the combined administration may be characterized by administering the peptide, nucleic acid or nanoparticle of the present invention in combination with one or more anticancer agents, and may also be performed in parallel with other anticancer therapies.
본 발명에 있어서, 상기 병용 투여되는 각각의 유효성분은 독립적인 경로를 통해 투여되는 것을 특징으로 할 수 있다. 각각의 유효성분은 독립적으로 통상의 기술자에 의해 적합한 투여용법 및 용량으로 투여될 수 있다. 예를 들어, 본 발명의 일 실시예에서와 같이 바람직하게는 펩타이드, 핵산 또는 나노입자는 피내 투여하고, 항암제는 정맥 주사를 통해 투여하는 것을 특징으로 할 수 있으나, 이에 제한되는 것은 아니다.In the present invention, each of the effective ingredients to be co-administered may be characterized by being administered through an independent route. Each effective ingredient may be administered independently by a person skilled in the art with a suitable administration method and dosage. For example, as in one embodiment of the present invention, it may be characterized by preferably administering peptides, nucleic acids, or nanoparticles intradermally, and administering anticancer agents through intravenous injection, but is not limited thereto.
본 발명에 있어서, 상기 펩타이드, 핵산 또는 나노입자와 항암제의 병용 투여는 동시에 투여되는 것을 특징으로 할 수 있다.In the present invention, the combined administration of the peptide, nucleic acid or nanoparticle and the anticancer agent may be characterized by being administered simultaneously.
본 발명에 있어서, 상기 펩타이드, 핵산 또는 나노입자와 항암제의 병용 투여는 순차적으로 투여되는 것을 특징으로 할 수 있다. 예를 들어, 상기 펩타이드, 핵산 또는 나노입자의 투여 후, 항암제가 투여되거나, 항암제의 투여 후 상기 펩타이드, 핵산 또는 지질 나노입자가 투여될 수 있다.In the present invention, the combined administration of the peptide, nucleic acid or nanoparticle and the anticancer agent may be characterized by sequential administration. For example, the anticancer agent may be administered after the administration of the peptide, nucleic acid or nanoparticle, or the peptide, nucleic acid or lipid nanoparticle may be administered after the administration of the anticancer agent.
본 발명에 있어서, 상기 펩타이드, 핵산 또는 나노입자와 항암제가 순차적으로 투여되는 경우, 일정한 시간적 간격을 두고 투여되는 것을 특징으로 할 수 있으며, 비제한적인 예를 들어, 1분 간격, 5분 간격, 10분 간격, 20분 간격, 30분 간격, 1시간 간격, 1일 간격, 수일 간격, 또는 1주 내지 수주 간격으로 순차적으로 투여될 수 있으나, 이에 제한되는 것은 아니며, 통상의 기술자에 의해 적절한 간격으로 순차적으로 투여될 수 있다.In the present invention, when the peptide, nucleic acid or nanoparticle and the anticancer agent are administered sequentially, it can be characterized in that they are administered at a certain time interval, and for non-limiting examples, they can be sequentially administered at 1 minute intervals, 5 minutes intervals, 10 minutes intervals, 20 minutes intervals, 30 minutes intervals, 1 hour intervals, 1 day intervals, several days intervals, or 1 week to several weeks intervals, but are not limited thereto, and can be sequentially administered at an appropriate interval by a person skilled in the art.
본 발명에 있어서, 상기 펩타이드, 핵산 또는 나노입자와 항암제의 투여는 각각 독립적으로 반복하여 1회 이상 수행될 수 있다. 본 발명에 있어서, 상기 펩타이드, 핵산 또는 나노입자와 항암제가 반복하여 투여되는 경우, 각각의 투여 간격은 독립적으로, 대상의 상태, 목적하는 효과의 수준에 따라 통상의 기술자에게 용이하게 조절될 수 있다. 예를 들어, 상기 펩타이드, 핵산 또는 나노입자와 항암제는 각각 독립적으로 1시간 간격, 6시간 간격, 8시간 간격, 12시간 간격, 1일 간격, 2일 간격, 일주일 간격, 2주일 간격, 또는 한달 간격으로 투여될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the administration of the peptide, nucleic acid or nanoparticle and the anticancer agent can be performed independently and repeatedly one or more times. In the present invention, when the peptide, nucleic acid or nanoparticle and the anticancer agent are repeatedly administered, each administration interval can be easily adjusted independently by a person skilled in the art according to the condition of the subject and the level of the desired effect. For example, the peptide, nucleic acid or nanoparticle and the anticancer agent can be independently administered at 1 hour intervals, 6 hours intervals, 8 hours intervals, 12 hours intervals, 1 day intervals, 2 days intervals, 1 week intervals, 2 weeks intervals, or 1 month intervals, but is not limited thereto.
본 발명에 있어서, 상기 약학적 조성물은 병용 투여되는 항암제의 제형과 독립적인 제형 및 용량로 투여될 수 있다. 바람직하게는, 상기 펩타이드, 핵산 또는 나노입자를 포함하는 약학 조성물과 항암제를 포함하는 약학 조성물이 각각 유효한 용량으로 투여될 수 있으나, 이에 제한되는 것은 아니다.In the present invention, the pharmaceutical composition may be administered in a formulation and dosage independent of the formulation of the anticancer agent to be administered in combination. Preferably, the pharmaceutical composition comprising the peptide, nucleic acid or nanoparticle and the pharmaceutical composition comprising the anticancer agent may each be administered in an effective dosage, but is not limited thereto.
본 발명에서는 특정 아미노산 서열 및 염기 서열을 기재하였으나, 본 발명에서 실시하고자 하는 효소와 실질적으로 동일한 아미노산 서열 및 이를 코딩하는 염기 서열이 본 발명의 권리범위에 속하는 것은 당업자에게 자명할 것이다. 실질적으로 동일하다는 것은, 아미노산 또는 염기서열의 상동성이 매우 높은 경우를 포함하고, 그 밖에도 서열의 상동성과는 무관하게 구조적 특징을 공유하거나 본 발명에서 사용된 것과 동일한 기능을 가지는 단백질을 의미한다. 본 발명의 핵심을 구성하는 서열을 제외한 다른 서열이 일부 결실된 단백질 또는 이를 코딩하는 염기서열의 단편도 본 발명에 포함될 수 있으며, 따라서 본 발명은 단편의 길이와는 무관하게 본 발명에서 사용된 것과 동일한 기능을 가지는 아미노산 또는 염기 서열을 모두 포함한다.Although specific amino acid sequences and base sequences are described in the present invention, it will be apparent to those skilled in the art that an amino acid sequence substantially identical to an enzyme to be implemented in the present invention and a base sequence encoding the same fall within the scope of the present invention. Substantially identical includes a case where the homology of the amino acid or base sequence is very high, and also means a protein that shares structural characteristics or has the same function as that used in the present invention regardless of the homology of the sequence. A protein in which a part of a sequence other than the sequence constituting the core of the present invention is deleted or a fragment of the base sequence encoding the same may also be included in the present invention, and therefore the present invention includes all amino acid or base sequences that have the same function as that used in the present invention regardless of the length of the fragment.
실시예Example
하기의 실시예를 통하여 본 발명을 보다 상세하게 설명한다. 그러나 하기 실시예는 본 발명의 내용을 구체화하기 위한 것일 뿐 이에 의해 본 발명이 한정되는 것은 아니다.The present invention will be described in more detail through the following examples. However, the following examples are only intended to concretize the contents of the present invention and the present invention is not limited thereto.
실시예 1: 재료 및 방법Example 1: Materials and Methods
실시예 1-1: 시약Example 1-1: Reagents
1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPE-PEG1000) 및 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] (DSPE-PEG2000-PDP) - Avanti Polar Lipids.1,2-Dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE), 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPE-PEG1000) and 1, 2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[PDP(polyethylene glycol)-2000] (DSPE-PEG2000-PDP) - Avanti Polar Lipids.
Boc-Arg(Pbf)-OH 및 콜레스테롤 - Sigma Aldrich Boc-Arg(Pbf)-OH and Cholesterol - Sigma Aldrich
포스포로티오에이트 백본으로 변형된 CpG 올리고DNA(CpG ODN; 5’-TCC ATG ACG TTC CTG ACG TT-3’, 서열번호 151) - 제노테크 및 바이오니아.CpG oligoDNA modified with phosphorothioate backbone (CpG ODN; 5’-TCC ATG ACG TTC CTG ACG TT-3’, SEQ ID NO: 151) - Genotech and Bioneer.
이외에 사용된 모든 펩타이드는 애니진에서 합성되었으며, 별도로 기재되지 않는 모든 시약은 Sigma Aldrich에서 구매하였다.All peptides used were synthesized in Anigene, and all reagents unless otherwise stated were purchased from Sigma Aldrich.
실시예 1-2: 동물 및 세포주Example 1-2: Animals and cell lines
암컷 C57BL/6 마우스(Female C57BL/6 mice, Orient Bio)는 무병원체 환경에서 사육되었다. 동물 관리 및 실험절차는 한국과학기술원(KAIST)의 동물관리 및 사용위원회의 승인을 받았다 (Accreditation number: KA2020-59). Female C57BL/6 mice (Orient Bio) were raised in a pathogen-free environment. Animal care and experimental procedures were approved by the Institutional Animal Care and Use Committee of the Korea Advanced Institute of Science and Technology (KAIST) (Accreditation number: KA2020-59).
E.G7-OVA 세포주 (ATCC; Manassas, VA, USA).E.G7-OVA cell line (ATCC; Manassas, VA, USA).
E.G7-OVA 세포주는 10% 열 불활성화 FBS, 1% 페니실린/스트렙토마이신, 2mM L-글루타민, 4.5 g/l 글루코스, 10 mM HEPES, 1 mM sodium pyruvate, 50 μM 2-메르캅토에탄올(2-mercaptoethanol), 및 0.5mg/ml G418(Gibco)로 보충된 RPMI-1640 배지(Welgene)에서 배양되었다. E.G7-OVA cell line was cultured in RPMI-1640 medium (Welgene) supplemented with 10% heat-inactivated FBS, 1% penicillin/streptomycin, 2 mM L-glutamine, 4.5 g/l glucose, 10 mM HEPES, 1 mM sodium pyruvate, 50 μM 2-mercaptoethanol, and 0.5 mg/ml G418 (Gibco).
Panc02 세포주는 한국과학기술원의 강석조 교수로부터 제공받았다. Panc02 세포는 10% FBS 및 1% 페니실린/스트렙토마이신으로 보충된 RPMI-1640 배지에서 유지되었다.Panc02 cell line was provided by Professor Seok-Jo Kang, KAIST. Panc02 cells were maintained in RPMI-1640 medium supplemented with 10% FBS and 1% penicillin/streptomycin.
MC38 세포주는 한국과학기술원의 김찬혁 교수로부터 제공받았다. MC38 세포는 10% FBS 및 1% 페니실린/스트렙토마이신으로 보충된 DMEM 배지에서 유지되었다.The MC38 cell line was provided by Professor Chan-Hyuk Kim of the Korea Advanced Institute of Science and Technology. MC38 cells were maintained in DMEM medium supplemented with 10% FBS and 1% penicillin/streptomycin.
EO771.lmb 세포주(ATCC; Manassas, VA, USA).EO771.lmb cell line (ATCC; Manassas, VA, USA).
EO771.lmb 세포주는 10% 열 불활성화 FBS, 1% 페니실린/스트렙토마이신으로 보충된 DMEM 배지(Welgene)에서 배양되었다. EO771.lmb cell line was cultured in DMEM medium (Welgene) supplemented with 10% heat-inactivated FBS and 1% penicillin/streptomycin.
모든 세포는 가습된 5% CO2 및 37℃에서 유지되었다.All cells were maintained at 37°C in a humidified atmosphere of 5% CO2 .
실시예 1-3: 유세포 분석 (Flow Cytometry)Example 1-3: Flow Cytometry
모든 항체는 BioLegend(San Diego, CA, USA), eBioscience(San Diego, CA, USA) 또는 Bioss Antibodies(Woburn, MA, USA)에서 구입하였다. All antibodies were purchased from BioLegend (San Diego, CA, USA), eBioscience (San Diego, CA, USA), or Bioss Antibodies (Woburn, MA, USA).
사용된 항체는 다음과 같다: anti-CD16/CD32 (BioLegend; Catalog #101319, clone 2.4G2), anti-CD45 Pacific Blue (BioLegend; Catalog #103125, clone 30-F11), anti-CD45 PerCP/Cy5.5 (eBioscience; Catalog #45-0451-82, clone 30-F11), anti-CD3e PerCP/Cy5.5 (BioLegend; Catalog #100327, clone 145-2C11), anti-CD3e PE/Cy7 (BioLegend; Catalog #100219, clone 145-2C11), anti-CD3e FITC (BioLegend; Catalog #100306, clone 145-2C11), anti-CD8a PE (BioLegend; Catalog #100708, clone 53-6.7), anti-CD8a PE/Cy7 (BioLegend; Catalog #100722, clone 53-6.7), anti-CD8a APC (BioLegend; Catalog #100711, clone 53-6.7), anti-CD8a APC/Cy7 (BioLegend; Catalog #100713, clone 53-6.7), anti-CD4 FITC (eBioscience; Catalog # 11-0041-82, clone GK1.5), anti-IFN-γ PE (eBioscience; Catalog #12-7311-82, clone XMG1.2), anti-IL-17A PE (eBioscience; Catalog # 12-7177-81, clone eBio17B7), 및 anti-FAPα Alexa Fluor 647 (Bioss Antibodies; Catalog # bs-5758R-A647). Antibodies used were as follows: anti-CD16/CD32 (BioLegend; Catalog #101319, clone 2.4G2), anti-CD45 Pacific Blue (BioLegend; Catalog #103125, clone 30-F11), anti-CD45 PerCP/Cy5. 5 (eBioscience; Catalog #45-0451-82, clone 30-F11), anti-CD3e PerCP/Cy5.5 (BioLegend; Catalog #100327, clone 145-2C11), anti-CD3e PE/Cy7 (BioLegend; Catalog # 100219, clone 145-2C11), anti-CD3e FITC (BioLegend; Catalog #100306, clone 145-2C11), anti-CD8a PE (BioLegend; Catalog #100708, clone 53-6.7), anti-CD8a PE/Cy7 (BioLegend; Catalog #100722, clone 53-6.7), anti-CD8a APC (BioLegend; Catalog #100711, clone 53-6.7), anti -CD8a APC/Cy7 (BioLegend; Catalog #100713, clone 53-6.7), anti-CD4 FITC (eBioscience; Catalog # 11-0041-82, clone GK1.5), anti-IFN-γ PE (eBioscience; Catalog # 12-7311-82, clone XMG1.2), anti-IL-17A PE (eBioscience; Catalog # 12-7177-81, clone eBio17B7), and anti-FAPα Alexa Fluor 647 (Bioss Antibodies; Catalog #bs-5758R-A647).
유세포 분석에 추가적으로 사용된 시약은 다음과 같다: 7-AAD 생존율 염색 용액 (BioLegend; Catalog #420403), Ghost Dye Violet 450 (Tonbo Biosciences, San Diego, CA, USA; Catalog #13-0863) 및 Fixable Viability Dye eFluor 450. Additional reagents used for flow cytometry analysis were: 7-AAD Viability Stain Solution (BioLegend; Catalog #420403), Ghost Dye Violet 450 (Tonbo Biosciences, San Diego, CA, USA; Catalog #13-0863), and Fixable Viability Dye eFluor 450.
세포는 4℃에서 10분간 항-CD16/CD32 항체와 배양하여 차단되었으며, 4℃에서 20분동안 다른 항체로 면역염색되었다. 죽은 세포는 7AAD 생존율 염색 용액, Ghost Dye Violet 450 또는 및 Fixable Viability Dye eFluor 450으로 염색하여 제외시켰다. LSRFortessa 유세포 분석기(BD Biosciences, San Jose, CA, USA)를 사용하여 유세포 분석을 수행하고 데이터를 획득하였다. FlowJo 소프트웨어(TreeStar)를 사용하여 분석하였다.Cells were blocked by incubation with anti-CD16/CD32 antibodies for 10 min at 4°C and immunostained with different antibodies for 20 min at 4°C. Dead cells were excluded by staining with 7AAD viability stain solution, Ghost Dye Violet 450, or Fixable Viability Dye eFluor 450. Flow cytometry was performed and data were acquired using an LSRFortessa flow cytometer (BD Biosciences, San Jose, CA, USA). Analysis was performed using FlowJo software (TreeStar).
실시예 1-4: 마우스 FAP 표적 펩타이드의 선택 및 백신 설계Example 1-4: Selection of mouse FAP target peptides and vaccine design
마우스 FAP 표적 에피토프 펩타이드의 예측은 NetMHC3.0, BIMAS 및 PREDEP을 포함하는 에피토프 펩타이드 예측 알고리즘을 사용하여 수행되었다. 사용된 마우스 FAP 전장서열은 서열번호 152(Uniprot NO. P97321)와 같다. 모든 에피토프 펩타이드는 MHC-Ⅰ 일배체형인 H-2Kb에 의해 인식되는 9-mer 펩타이드 길이로 선택되었다. 선택된 펩타이드의 서열은 표 1과 같다: Prediction of mouse FAP target epitope peptides was performed using epitope peptide prediction algorithms including NetMHC3.0, BIMAS, and PREDEP. The mouse FAP full-length sequence used is SEQ ID NO: 152 (Uniprot NO. P97321). All epitope peptides were selected as 9-mer peptide lengths recognized by MHC-I haplotype H-2Kb. The sequences of the selected peptides are shown in Table 1:
[표 1][Table 1]
실시예 1-5: 면역원성 및 항종양 효과 평가Example 1-5: Evaluation of immunogenicity and antitumor effect
선택된 펩타이드 후보의 면역원성 시험을 위해, 6주령 암컷 C57BL/6 마우스를 1주 간격으로 상동 프라임-부스트 요법을 사용하여 2회 면역화하였다. Complete Freund's Adjuvant(CFA; Sigma Aldrich)에 유화된 각 펩타이드 100μg을 양쪽 발바닥에 피하 주사하여 마우스를 면역화하였다. 마지막 면역화 후 10일 뒤 마우스를 희생시켰다. 항원 특이적 T 세포 반응은 마우스에서 비장 세포를 분리하고 각 펩타이드(10 μg/ml)로 생체 외에서 재자극하여 평가하였다. IFN-γ 생성 CD8+ T 세포는 세포내 사이토카인 염색법(intracellular cytokine staining, ICS)으로 정량하였으며(도 2), IFN-γ 생성 세포의 수는 ELISpot(enzyme-linked immunospot) 분석법으로 측정하였다. ICS 분석의 경우, 비장 세포(튜브당 3x106개 세포)를 각 펩타이드로 1시간 동안 재자극하였다. GolgiStop 및 GolgiPlug(BD Biosciences)를 각 튜브에 첨가하여 사이토카인의 세포내 수송을 억제한 후, 세포를 추가로 5시간 동안 배양하고, 항-CD3e 및 항-CD8a 항체로 면역염색한 뒤, 4℃에서 20분 동안 Live/Dead 세포 염료로 염색하였다. 세포내 사이토카인 염색을 위해 Cytofix/Cytoperm 용액(BD Biosciences)을 사용하여 세포를 투과화 한 뒤, 항-IFN-γ 항체와 함께 배양하였다. 이후, 샘플을 세척하고 유세포 분석법으로 분석하였다. ELISpot으로 IFN-γ 생성 세포를 측정하기 위해 마우스 IFN-γ 특이적 단일클론 항체로 코팅된 96-웰 마이크로플레이트에 비장 세포(3 × 105개 세포/웰)를 접종하고 각 펩타이드로 30시간 동안 재자극하였다. IFN-γ 생성 스팟은 제조업체의 프로토콜에 따라 마우스 IFN-γ ELISpot 키트(R&D Systems, Minneapolis, MN, USA)를 사용하여 생성하였다. 사이토카인 국소화 부위의 흑청색 반점을 자동 ELISpot 판독기(AID GmbH, Strasbourg, Germany)를 사용하여 계수하였다.For immunogenicity testing of the selected peptide candidates, 6-week-old female C57BL/6 mice were immunized twice using a homologous prime-boost regimen at 1-week intervals. Mice were immunized by subcutaneous injection of 100 μg of each peptide emulsified in Complete Freund's Adjuvant (CFA; Sigma Aldrich) into both footpads. Mice were sacrificed 10 days after the final immunization. Antigen-specific T cell responses were evaluated by isolating spleen cells from mice and restimulating them ex vivo with each peptide (10 μg/ml). IFN-γ-producing CD8+ T cells were quantified by intracellular cytokine staining (ICS) (Fig. 2 ), and the number of IFN-γ-producing cells was measured by enzyme-linked immunospot (ELISpot) assay. For ICS analysis, spleen cells (3 × 106 cells per tube) were restimulated with each peptide for 1 h. GolgiStop and GolgiPlug (BD Biosciences) were added to each tube to inhibit intracellular transport of cytokines, and the cells were cultured for an additional 5 h, immunostained with anti-CD3e and anti-CD8a antibodies, and then stained with Live/Dead cell dye for 20 min at 4°C. For intracellular cytokine staining, cells were permeabilized with Cytofix/Cytoperm solution (BD Biosciences) and incubated with anti-IFN-γ antibody. Samples were then washed and analyzed by flow cytometry. To measure IFN-γ-producing cells by ELISpot, spleen cells (3 × 10 5 cells/well) were seeded in 96-well microplates coated with mouse IFN-γ-specific monoclonal antibodies and restimulated with each peptide for 30 h. IFN-γ-producing spots were generated using the mouse IFN-γ ELISpot kit (R&D Systems, Minneapolis, MN, USA) according to the manufacturer's protocol. Blue-black spots at the sites of cytokine localization were counted using an automated ELISpot reader (AID GmbH, Strasbourg, Germany).
펩타이드의 항종양 치료 효능은 오른쪽 옆구리에 2 x 105 E.G7-OVA 세포를 피하 접종한 마우스에서 평가하였다. E.G7-OVA 세포 접종 9일 후, 각 그룹의 평균 종양 부피가 대략 동일하도록 마우스를 무작위로 그룹으로 나눈 다음, 상기한 것과 동일한 방법으로 각 펩타이드를 1주 간격으로 2회 면역화하였다. 디지털 캘리퍼스를 사용하여 격일로 종양 성장을 모니터링하고 종양 부피를 0.5×길이×너비로 계산하였다. 평균 종양 부피가 윤리적 종점(~1,500 mm3)에 도달했을 때 마우스를 안락사시켰다.The antitumor therapeutic efficacy of the peptides was evaluated in mice that were inoculated subcutaneously with 2 × 10 5 E.G7-OVA cells into the right flank. Nine days after E.G7-OVA cell inoculation, the mice were randomly divided into groups such that the average tumor volumes of each group were approximately equal, and then immunized twice with each peptide at 1-week intervals using the same method as described above. Tumor growth was monitored every other day using digital calipers, and the tumor volumes were calculated as 0.5 × length × width. The mice were euthanized when the average tumor volume reached the ethical endpoint (~1,500 mm 3 ).
실시예 1-6: FAPPEP-SLNP 나노백신의 제조Example 1-6: Preparation of FAP PEP -SLNP nanovaccine
모노아르기닌-콜레스테롤(MA-Chol) 및 DSPE-PEG2000-FAPPEP는 기존에 보고된 방법을 기반으로 합성되었다. Monoarginine-cholesterol (MA-Chol) and DSPE-PEG2000-FAP PEP were synthesized based on previously reported methods.
CKLWRYSYTA 및 CYFRNVDYLL의 두 가지 선택된 펩타이드 후보를 사용하여 제조된 시스테닐화 FAPPEP(Cys-FAPPEP)를 디설파이드 교환 반응에 의해 DSPE-PEG2000-PDP에 접합시켰다. Cys-FAPPEP 및 DSPE-PEG2000-PDP를 디메틸 설폭사이드에 용해시키고 1:2의 몰비로 혼합하였다. 용액을 실온에서 밤새 부드럽게 와류시키고 아세토니트릴을 첨가하여 반응을 퀀칭하였다. Cysteinylated FAP PEP (Cys-FAP PEP ) prepared using two selected peptide candidates, CKLWRYSYTA and CYFRNVDYLL, was conjugated to DSPE-PEG2000-PDP by disulfide exchange reaction. Cys-FAP PEP and DSPE-PEG2000-PDP were dissolved in dimethyl sulfoxide and mixed at a molar ratio of 1:2. The solution was gently vortexed overnight at room temperature and the reaction was quenched by the addition of acetonitrile.
DSPE-PEG2000-FAPPEP 접합체는 C4 컬럼(Nomura Chemical, Japan)을 사용하여 고성능 액체 크로마토그래피(HPLC; Agilent, Santa Clara, CA, USA)로 정제되었으며 MALDI-TOF 분광법을 사용하여 특성화되었다. The DSPE-PEG2000-FAP PEP conjugate was purified by high-performance liquid chromatography (HPLC; Agilent, Santa Clara, CA, USA) using a C4 column (Nomura Chemical, Japan) and characterized using MALDI-TOF spectroscopy.
MA-Chol:DOPE:DSPE-PEG1000:DSPE-PEG2000-FAPPEP(몰비, 48.625:48.625:2.25:0.5)를 포함하는 FAPPEP-SLNP 나노백신을 박막 형성 및 재수화 방법을 사용하여 제조하였다. 클로로포름과 메탄올에 용해된 모든 지질 성분을 건조하고 진공 하에서 용매를 제거한 후 생성된 지질막을 CpG ODN을 포함하는 HEPES-buffered glucose(HBG)로 재수화시켰다. 용액을 10분 동안 초음파 처리하고, 실온에서 4시간 이상 마그네틱 바로 교반하고, 미니 압출기(Avanti Polar Lipids)를 사용하여 11회 이상 압출하였다. 8μmol의 SLNP에 캡슐화된 1.65nmol의 CpG ODN을 사용하여 로딩 효율이 100%에 근접하도록 하였다.FAPPEP-SLNP nanovaccines containing MA-Chol:DOPE:DSPE-PEG1000:DSPE-PEG2000-FAP PEP (molar ratio, 48.625:48.625:2.25:0.5) were prepared using a thin film formation and rehydration method. After drying all lipid components dissolved in chloroform and methanol, the solvent was removed under vacuum, and the resulting lipid film was rehydrated with HEPES-buffered glucose (HBG) containing CpG ODN. The solution was sonicated for 10 min, magnetically stirred for more than 4 h at room temperature, and extruded more than 11 times using a mini extruder (Avanti Polar Lipids). The loading efficiency was close to 100% using 1.65 nmol of CpG ODN encapsulated in 8 μmol of SLNP.
FAPPEP-SLNP의 특성화를 위해 Zetasizer Nano 범위 시스템(Malvern, Worcestershire, UK)을 사용하여 상온에서 DLS에 의해 유체역학적 크기를 결정하였다. FAPPEP-SLNP의 형태와 크기는 모두 Philips TECNAI F20 기기(Philips Electronic Instrument Corp., 미국 뉴저지주 마화)를 사용하여 TEM에 의해 평가되었으며 1% 우라닐 아세테이트 용액을 음성 염색에 사용하였다. 나노백신의 평균 크기는 Gatan Microscopy Suite(GMS) 소프트웨어(Gatan, Pleasanton, CA, USA)를 사용하여 측정하였다.For the characterization of FAP PEP -SLNP, the hydrodynamic size was determined by DLS at room temperature using a Zetasizer Nano range system (Malvern, Worcestershire, UK). Both the morphology and size of FAP PEP -SLNP were characterized by TEM using a Philips TECNAI F20 instrument (Philips Electronic Instrument Corp., Mahwah, NJ, USA) and 1% uranyl acetate solution was used for negative staining. The average size of the nanovaccines was measured using Gatan Microscopy Suite (GMS) software (Gatan, Pleasanton, CA, USA).
실시예 1-7: FAPPEP-SLNP나노백신의 항종양 효과 평가Example 1-7: Evaluation of antitumor effect of FAP PEP -SLNP nanovaccine
FAPPEP1-SLNP나노백신의 항종양 효과 평가를 위해, 6주령 암컷 C57BL/6 마우스에서 3개의 피하 종양 모델을 구축하였다.To evaluate the antitumor effect of FAP PEP1 -SLNP nanovaccine, three subcutaneous tumor models were established in 6-week-old female C57BL/6 mice.
E.G7-OVA 치료 모델의 경우, 마우스 오른쪽 옆구리에 2 x 105 E.G7-OVA 암세포를 주사하였다. E.G7-OVA 암 세포 접종 9일 후, 표시된 시점에서 양쪽 발바닥에 피하 주사하여 종양 보유 마우스를 FAPPEP1-SLNP 또는 OVAPEP-SLNP 나노백신(CpG, 마우스당 0.4nmol; FAPPEP, 마우스당 5nmol; OVAPEP, 마우스당 5nmol; SLNP, 마우스당 2μmol)으로 4일 간격으로 3회 면역화하였다.For the E.G7-OVA treatment model, 2 × 10 5 E.G7-OVA cancer cells were injected into the right flank of the mice. Nine days after E.G7-OVA cancer cell inoculation, tumor-bearing mice were immunized three times at 4-day intervals with FAP PEP1 -SLNP or OVA PEP -SLNP nanovaccines (CpG, 0.4 nmol per mouse; FAPPEP, 5 nmol per mouse; OVAPEP, 5 nmol per mouse; SLNP, 2 μmol per mouse) by subcutaneous injection into both footpads at the indicated time points.
Panc02 치료 모델의 경우, 1×106 Panc02 암 세포를 마우스 오른쪽 옆구리에 주사하였다. Panc02 세포 접종 16일 후, 종양 보유 마우스를 위와 동일한 방법으로 4일 간격으로 FAPPEP1-SLNP를 사용하여 3회 면역화하였다. For the Panc02 treatment model, 1× 106 Panc02 cancer cells were injected into the right flank of the mice. Sixteen days after Panc02 cell inoculation, the tumor-bearing mice were immunized three times with FAP PEP1 -SLNP at 4-day intervals using the same method as above.
MC38 치료 모델의 경우, 마우스의 오른쪽 옆구리에 1x105 MC38 암세포를 주사하였다. MC38 암 세포 접종 5일 후, 종양 보유 마우스를 위와 동일한 방법으로 4일 간격으로 FAPPEP1-SLNP를 사용하여 3회 면역화하였다. For the MC38 treatment model, 1x10 5 MC38 cancer cells were injected into the right flank of the mice. Five days after MC38 cancer cell inoculation, the tumor-bearing mice were immunized three times with FAP PEP1 -SLNP at 4-day intervals using the same method as above.
병용 요법의 평가를 위해, 표시된 것과 같이, 마우스에 총 4번 격일로 Dox(10mg/kg)를 복강내 주사하였다. 종양 성장을 격일로 모니터링하고 마우스의 생존율을 모니터링하였다. 평균 종양 부피가 윤리적 종점에 도달했을 때 마우스를 안락사시켰다.For evaluation of combination therapy, mice were injected intraperitoneally with Dox (10 mg/kg) every other day for a total of four times as indicated. Tumor growth was monitored every other day and mouse survival was monitored. Mice were euthanized when the mean tumor volume reached the ethical endpoint.
실시예 1-8: 종양 조직의 조직학적 분석Example 1-8: Histological analysis of tumor tissue
종양 조직을 절제하고 10% 포르말린 용액에 고정하고 파라핀에 포매한 다음 5μm 두께의 절편으로 절단하였다. 종양 조직의 콜라겐 침착은 Weigert의 철 헤마톡실린, Biebrich scarlet-acid fuchsin 용액 및 Aniline blue를 사용하는 Masson의 trichrome 염색으로 평가하였다. 모든 절편 슬라이드는 도립 현미경 (Eclipse Ti2; Nikon, Tokyo, Japan)을 사용하여 이미지화 하였다. Masson의 trichrome으로 염색된 조직 절편은 ImageJ 소프트웨어(National Institutes of Health, Bethesda, MD, USA)를 사용하여 분석하였다.Tumor tissues were resected, fixed in 10% formalin solution, embedded in paraffin, and then cut into 5-μm-thick sections. Collagen deposition in tumor tissues was assessed by Weigert's iron hematoxylin, Biebrich's scarlet-acid fuchsin solution, and Masson's trichrome staining with aniline blue. All section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). Masson's trichrome-stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
실시예 1-9: TIL 및 CAF 분석Example 1-9: TIL and CAF analysis
종양 침윤성 CD8+ T 세포 및 FAP+ CAF는 종양 조직을 작은 조각으로 절단한 다음 마우스 종양 해리 키트(Miltenyi Biotech, Auburn, CA, USA) 및 gentleMACS Dissociator(Miltenyi Biotech)를 효소 및 기계적으로 단일 세포 현탁액으로 분리하여 분석하였다. 세포 현탁액을 세포 여과기(70 μm)를 통해 여과하고 DPBS(Dulbecco’s phosphate-buffered saline)로 세척하였다. 적혈구(RBC)를 2ml RBC 용해 완충액(BioLegend)과 함께 실온에서 2분 동안 부드럽게 흔들면서 배양하여 제거하였다. 해리된 세포를 항-CD45, 항-CD3e, 항-CD8a 및 항-FAPα 항체로 면역염색하고, 4℃에서 20분 동안 Live/Dead 세포 염료로 염색하였다. 이후, 세포를 세척하고 유세포 분석을 수행하였다.Tumor-infiltrating CD8+ T cells and FAP+ CAFs were analyzed by dissociating tumor tissues into small pieces and then enzymatically and mechanically dissociating single-cell suspensions using a mouse tumor dissociation kit (Miltenyi Biotech, Auburn, CA, USA) and gentleMACS Dissociator (Miltenyi Biotech). The cell suspension was filtered through a cell strainer (70 μm) and washed with Dulbecco’s phosphate-buffered saline (DPBS). Red blood cells (RBCs) were removed by incubation with 2 ml RBC lysis buffer (BioLegend) for 2 min at room temperature with gentle shaking. The dissociated cells were immunostained with anti-CD45, anti-CD3e, anti-CD8a, and anti-FAPα antibodies, and stained with Live/Dead cell dye for 20 min at 4°C. The cells were then washed and subjected to flow cytometry.
실시예 1-10: 비장 T 세포 분석Example 1-10: Splenic T cell analysis
면역화된 마우스로부터 비장을 회수하고, 비장세포를 분리하였다. 비장 Th17 세포의 분석을 위해, 분리된 세포를 가용성 항-CD3e(BD Biosciences; Catalog #553057, clone 145-2C11) 및 항-CD28 (BD Biosciences; Catalog #553294, clone 37.51) 항체로 37℃에서 1시간 동안 자극하였으며, GolgiStop 및 GolgiPlug를 추가하여 사이토카인의 세포내 수송을 억제하였다. 세포를 추가로 5시간 동안 배양한 다음 항-CD3e 및 항-CD4 항체 및 Live/Dead 세포 염료로 4°C에서 20분 동안 염색하였다. 세포내 사이토카인 염색을 위해 세포를 Cytofix/Cytoperm 용액을 사용하여 투과화한 다음 항-IL-17A 항체와 함께 배양하였다. 항원 특이적 비장 T 세포의 분석을 위해 비장 세포를 FAPPEP 또는 OVAPEP(10 μg/ml)로 1시간 동안 재자극한 후, GolgiStop 및 GolgiPlug를 첨가하고 세포를 위에서 설명한 대로 추가로 5시간 동안 배양하였다. 이어서 세포를 항-CD3e, 항-CD4 및 항-CD8a 항체 및 Live/Dead 세포 염료로 4℃서 20분 동안 염색하였다. 표면 마커 염색 후, 세포를 투과화시키고 항-IFN-γ 항체와 함께 배양하였다. 모든 샘플은 유세포 분석을 통해 분석되었다.Spleens were harvested from immunized mice, and splenocytes were isolated. For analysis of splenic Th17 cells, the isolated cells were stimulated with soluble anti-CD3e (BD Biosciences; Catalog #553057, clone 145-2C11) and anti-CD28 (BD Biosciences; Catalog #553294, clone 37.51) antibodies for 1 h at 37°C, and GolgiStop and GolgiPlug were added to inhibit intracellular transport of cytokines. The cells were incubated for an additional 5 h and then stained with anti-CD3e and anti-CD4 antibodies and Live/Dead cell dye for 20 min at 4°C. For intracellular cytokine staining, cells were permeabilized using Cytofix/Cytoperm solution and then incubated with anti-IL-17A antibody. For analysis of antigen-specific splenic T cells, spleen cells were restimulated with FAPPEP or OVAPEP (10 μg/ml) for 1 h, followed by addition of GolgiStop and GolgiPlug and incubation for an additional 5 h as described above. Cells were then stained with anti-CD3e, anti-CD4 and anti-CD8a antibodies and Live/Dead cell dye for 20 min at 4°C. After surface marker staining, cells were permeabilized and incubated with anti-IFN-γ antibody. All samples were analyzed by flow cytometry.
실시예 1-11: Cypate 염료의 종양 침투Example 1-11: Tumor Penetration of Cypate Dye
소분자 화학요법 약물의 대용물로서 근적외선 염료인 cypate를 사용하여 종양 침투를 평가하였다. 마우스의 오른쪽 옆구리에 1×105 MC38 암세포를 주사하고 MC38 종양 치료 연구에서 사용된 것과 동일한 일정에 따라 MC38 암세포 접종 5일 후 FAPPEP1-SLNP로 4일 간격으로 3회 면역화하였다. 최종 면역화 후 종양이 ~300 mm3의 평균 크기에 도달한 후, cypate 염료(2.5 mg kg-1)를 안와후 경로(retro-orbital route)를 통해 정맥 주사하였다. 종양 조직을 cypate 염료 주입 2시간 후에 수확하고, 생체 내 이미징 시스템(IVIS; PerkinElmer, Waltham, MA, USA)을 사용하여 형광 신호를 평가하였다.Tumor penetration was assessed using cypate, a near-infrared dye, as a surrogate for small-molecule chemotherapeutic drugs. Mice were injected with 1 × 10 5 MC38 cancer cells into the right flank and immunized three times at 4-day intervals with FAPPEP1-
실시예 1-12: FAPPEP1 펩타이드 백신의 암 전이 억제 효과 평가 및 조직 분석Example 1-12: Evaluation of the cancer metastasis inhibition effect of FAP PEP1 peptide vaccine and tissue analysis
암의 전이 억제 효과 평가는 FAPpep1 펩타이드를 사용한 펩타이드 백신의 형태로 시험되었다. The evaluation of the anti-cancer metastasis effect was tested in the form of a peptide vaccine using the FAPpep1 peptide.
FAPPEP1 펩타이드 백신의 전이 억제효능을 평가하기 위해 Female C57BL/6 마우스에 동소 종양 모델(orthotopic tumor models)을 구축하였다. 마우스의 오른쪽 복부 유방선(abdominal mammary glands, 4th pair)에 1x105 EO771.lmb 암세포를 주사하였다. 종양의 성장은 디지털 캘리퍼를 이용하여 매일 진행성을 모니터링하였으며, 종양의 부피는 (0.5 x 길이 x 너비2)으로 계산하였다. 평균 종양부피가 ~300mm3에 도달한 뒤 원발 종양을 절제하였다. 원발 종양의 절제 4일 후 양쪽 발바닥에 피하주사를 통해 FAPpep1(100 μg/head) 및 CpG-ODN(10μg/head)의 혼합물을 1주 간격으로 2회 면역화하였다. 마지막 접종 1주일 뒤 마우스를 희생하여 분석하였다. To evaluate the metastatic inhibition efficacy of the FAP PEP1 peptide vaccine, orthotopic tumor models were established in female C57BL/6 mice. 1 × 10 5 EO771.lmb cancer cells were injected into the right abdominal mammary glands (4th pair) of mice. Tumor growth was monitored daily using a digital caliper, and the tumor volume was calculated as (0.5 × length × width 2 ). Primary tumors were resected when the average tumor volume reached ~300 mm 3 . Four days after resection of the primary tumors, the mixture of FAPpep 1 (100 μg/head) and CpG-ODN (10 μg/head) was immunized twice at 1-week intervals via subcutaneous injection into both footpads. The mice were sacrificed and analyzed 1 week after the final vaccination.
희생된 마우스의 폐 및 간 조직을 절제하고, 10% 포르말린 용액에 고정환 뒤 파라핀에 포매하고, 5μm 두께의 슬라이스로 절단하였다. FAPpep 백신의 전이 억제 효과 확인을 위해 상기 폐 및 간 조직 샘플을 H&E로 염색하고, 모든 절편 슬라이드는 도립 현미경 (Eclipse Ti2; Nikon, Tokyo, Japan)을 사용하여 이미지화 하였다. 염색된 조직 절편은 ImageJ 소프트웨어(National Institutes of Health, Bethesda, MD, USA)를 사용하여 분석하였다.Lung and liver tissues of sacrificed mice were excised, fixed in 10% formalin solution, embedded in paraffin, and cut into 5-μm-thick slices. To confirm the metastatic inhibition effect of FAPpep vaccine, the lung and liver tissue samples were stained with H&E, and all section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). The stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
실시예 1-13: FAPPEP1 펩타이드 백신의 NASH 치료 효과 및 혈청, 조직 분석Example 1-13: NASH therapeutic effect of FAP PEP1 peptide vaccine and serum and tissue analysis
NASH에 대한 FAPPEP1 펩타이드 백신의 치료 효과를 평가하기 위해, 6주령의 암컷 C57BL/6 마우스에 4주간 메티오닌/콜린 결핍 식이요법(methionine/choline deficient(MCD) diet)을 실시하고, 대조군에는 메티오닌/콜린 충분 식이요법(methionine/choline sufficient(MCS) diet)을 수행하여 모델을 구축하였다. 체중은 주당 3회 모니터링 되었다. 4주간의 MCD/MCS 식이요법의 실시 후, FAPpep1(100 μg/head) 및 CpG-ODN(10μg/head)의 혼합물을 1주 간격으로 2회 발바닥에 피하 투여하여 면역화하였다. 백신의 투여기간에도 식이요법은 계속 수행하였다. 마지막 면역화 1주후 마우스를 희생시켜 간을 적출하고 간의 무게를 측정하였다.To evaluate the therapeutic effect of FAP PEP1 peptide vaccine on NASH, 6-week-old female C57BL/6 mice were fed a methionine/choline deficient (MCD) diet for 4 weeks, or a methionine/choline sufficient (MCS) diet as a control group to establish a model. Body weight was monitored three times per week. After 4 weeks of MCD/MCS diet, mice were immunized by subcutaneous administration of a mixture of FAPpep1 (100 μg/head) and CpG-ODN (10 μg/head) twice at 1-week intervals into the footpad. The diet was continued during the vaccine administration period. One week after the last immunization, the mice were sacrificed, the livers were removed, and the liver weights were measured.
마우스의 희생 직전 혈액을 채취하고, 원심분리기를 통해 혈청을 분리하고 AST(aspartate transaminase), ALT(alanine transferase), 총 빌리루빈 및 HDL의 양을 측정하였다.Blood was collected immediately before sacrifice of the mice, serum was separated by centrifugation, and the levels of aspartate transaminase (AST), alanine transferase (ALT), total bilirubin, and HDL were measured.
폐 및 간 조직을 절제하고, 10% 포르말린 용액에 고정환 뒤 파라핀에 포매하고, 5μm 두께의 슬라이스로 절단하였다. 준비된 간 조직 샘플은 H&E로 염색하여 분석하거나, 이소프로판올 및 오일 레드 O 염색하여 분석하였다. 모든 절편 슬라이드는 도립 현미경 (Eclipse Ti2; Nikon, Tokyo, Japan)을 사용하여 이미지화 하였다. 염색된 조직 절편은 ImageJ 소프트웨어(National Institutes of Health, Bethesda, MD, USA)를 사용하여 분석하였다.Lung and liver tissues were excised, fixed in 10% formalin solution, embedded in paraffin, and cut into 5-μm-thick slices. The prepared liver tissue samples were stained with H&E and analyzed, or stained with isopropanol and Oil Red O. All section slides were imaged using an inverted microscope (Eclipse Ti2; Nikon, Tokyo, Japan). The stained tissue sections were analyzed using ImageJ software (National Institutes of Health, Bethesda, MD, USA).
실시예 1-12: 통계 분석Example 1-12: Statistical Analysis
데이터는 평균 ± 평균의 표준 오차(S.E.M.)로 표시하였다. GraphPad Prism 5(GraphPad Software)를 사용하여 적절하게 two-tailed Student’s t-test or one-way analysis of variance (ANOVA)으로 그룹을 비교하였다. Log-rank(Mantel-Cox) 테스트를 사용하여 생존율의 통계 분석을 수행하였다. P < 0.05에서 통계적으로 유의한 것으로 정의하였다.Data are expressed as mean ± standard error of the mean (S.E.M.). Groups were compared using two-tailed Student’s t-test or one-way analysis of variance (ANOVA), as appropriate, using GraphPad Prism 5 (GraphPad Software). Statistical analysis of survival was performed using the log-rank (Mantel-Cox) test. Statistically significant was defined as P < 0.05.
실시예 2: FAP의 면역우세 펩타이드 에피토프의 예측 및 스크리닝.Example 2: Prediction and screening of immunodominant peptide epitopes of FAP.
FAP의 면역우세 펩타이드 에피토프는 netMHC3.0, BIMAS 및 PREDEP의 세 가지 에피토프 펩타이드 예측 프로그램을 사용하여 예측되었다(도 3). 확인된 에피토프 펩타이드 중에서 H-2Kb 일배체형의 MHC 클래스 I에 대해 점수가 높고 8-9개의 아미노산 펩타이드 서열을 공유하는 에피토프 펩타이드를 선택하였다(도 4a). 궁극적으로, 9개의 선택된 펩타이드 중 6개가 생체내 면역원성에 대해 테스트 및 스크리닝 하였다. C57BL/6 마우스는 각 펩타이드와 완전 프로인트 보조제(CFA)의 혼합물로 1주 간격으로 2회 피하 면역화되었다. 마지막 면역화 1주일 후, 마우스를 희생시키고 각 펩타이드의 항원 특이적 T 세포 반응을 평가하기 위해 비장 세포를 분리하였다(도 4b). 분리된 비장 세포를 각 펩타이드로 재자극하고 세포에 의한 인터페론-γ(IFN-γ) 생산을 세포내 사이토카인 염색(ICS) 및 효소 결합 면역스팟(ELISpot) 분석으로 분석하였다. ICS 분석은 세 가지 펩타이드(KLWRYSYTA, GLFKCGIAV 및 YFRNVDYLL) 각각으로 면역화가 다른 세 가지 펩타이드로 면역화한 것보다 IFN-γ 분비 CD8+ T 세포의 빈도가 현저히 높은 것으로 나타났다(도 4c,d 및 도 5a-f). 3개의 ICS-양성 펩타이드는 또한 ELISpot 분석에서 양성 후보로 선택되었다(도 4e, f 및 도 6a-f). ICS 및 ELISpot 분석을 기반으로 항종양 효능의 추가 평가를 위해 궁극적으로 이 세 가지 펩타이드(KLWRYSYTA, GLFKCGIAV 및 YFRNVDYLL)를 선택하였다. 쥐 림프종 세포주 E.G7-OVA는 MHC 클래스 I 제한 CD8+ T 세포 반응을 연구하기 위한 인기 있는 모델 시스템으로 동계(syngeneic) 쥐의 종양 형성에 사용되었으며, FAP+ CAF의 존재는 유세포 분석에 의해 생성된 종양 조직에서 확인하였다 (도 7). E.G7-OVA 종양의 평균 부피가 ~100 mm3에 도달하면 마우스를 무작위로 4개의 그룹으로 나누고 각 펩타이드와 CFA의 혼합물로 1주 간격으로 2회 면역화하였다(도 4g). GLFKCGIAV + CFA를 사용한 면역화는 종양 성장 억제에 비효율적인 반면, 2개의 다른 펩타이드 후보인 KLWRYSYTA(FAPPEP1로 지정됨) 및 YFRNVDYLL(FAPPEP2로 지정됨)로 면역화하면 대조군에 비해 효과적인 종양 성장 억제가 나타났다(도 4h). 이러한 결과는 이들 2개의 펩타이드가 펩타이드 에피토프 펩타이드-특이적 CD8+ T 세포의 강력한 유도 및 높은 항종양 효능을 가능하게 하는 적합한 에피토프 펩타이드 후보임을 의미한다.The immunodominant peptide epitopes of FAP were predicted using three epitope peptide prediction programs, netMHC3.0, BIMAS, and PREDEP (Fig. 3). Among the identified epitope peptides, those with high scores for MHC class I of the H-2Kb haplotype and sharing 8–9 amino acid peptide sequences were selected (Fig. 4a). Ultimately, six of the nine selected peptides were tested and screened for in vivo immunogenicity. C57BL/6 mice were subcutaneously immunized twice at 1-week intervals with a mixture of each peptide and complete Freund's adjuvant (CFA). One week after the last immunization, the mice were sacrificed, and spleen cells were isolated to evaluate the antigen-specific T cell responses to each peptide (Fig. 4b). Isolated splenocytes were restimulated with each peptide, and interferon-γ (IFN-γ) production by the cells was analyzed by intracellular cytokine staining (ICS) and enzyme-linked immunospot (ELISpot) assays. The ICS assay showed that immunization with each of the three peptides (KLWRYSYTA, GLFKCGIAV, and YFRNVDYLL) induced significantly higher frequencies of IFN-γ-secreting CD8+ T cells than did immunization with the other three peptides (Figs. 4c,d and 5a–f). The three ICS-positive peptides were also selected as positive candidates in the ELISpot assay (Figs. 4e,f and 6a–f). Based on the ICS and ELISpot assays, these three peptides (KLWRYSYTA, GLFKCGIAV, and YFRNVDYLL) were ultimately selected for further evaluation of their antitumor efficacy. The murine lymphoma cell line E.G7-OVA is a popular model system to study MHC class I-restricted CD8+ T cell responses, and was used to induce tumor formation in syngeneic mice, and the presence of FAP+ CAFs was confirmed in the generated tumor tissues by flow cytometry (Fig. 7). When E.G7-OVA tumors reached an average volume of ~100 mm3, mice were randomly divided into four groups and immunized twice with a mixture of each peptide and CFA at 1-week intervals (Fig. 4g). Immunization with GLFKCGIAV + CFA was ineffective in suppressing tumor growth, whereas immunization with two other peptide candidates, KLWRYSYTA (designated FAP PEP1 ) and YFRNVDYLL (designated FAP PEP2 ), resulted in effective tumor growth inhibition compared to the control (Fig. 4h). These results imply that these two peptides are suitable epitope peptide candidates that enable potent induction of peptide epitope-specific CD8+ T cells and high antitumor efficacy.
실시예 3: FAP 에피토프 펩타이드 제시 나노 백신의 합성, 특성화 및 항종양 효능 평가Example 3: Synthesis, characterization, and antitumor efficacy evaluation of FAP epitope peptide-presenting nanovaccine
본 발명자들은 이전의 연구에서 보고한 작은 지질 나노입자 플랫폼을 사용하여, FAP의 두 가지 에피토프 펩타이드를 표시하는 나노백신을 설계하였다(Theranostics 2016, 6 (2), 192-203; Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638;대한민국 특허 제10-2425028호).Using the small lipid nanoparticle platform reported in our previous study, we designed a nanovaccine displaying two epitope peptides of FAP (Theranostics 2016, 6 (2), 192-203; Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638; Korean Patent No. 10-2425028).
이러한 FAP 펩티드 표시 SLNP(FAPPEP-SLNP)는 엔도좀 탈출을 촉진하기 위해 1,2-디올레오일-sn-글리세로-3-포스포에탄올아민(DOPE)으로 구성되었다. 생체 내에서 콜로이드 안정성을 부여하는 1,2-디스테아로일-sn-글리세로-3-포스포에탄올아민-N-[카르복시(폴리에틸렌 글리콜)-1000](DSPE-PEG1000); 및 모노아르기닌-콜레스테롤(MA-Chol)은 CpG 보조제와의 복합체 형성을 가능하게 하고 기계적 안정성을 제공한다(도 8). 펩타이드 에피토프 펩타이드는 N-말단(N'-Cys-FAPPEP1 및 N'-Cys-FAPPEP2) 및 DSPE-PEG2000(도 9 및 10)에서 시스테인 변형된 펩타이드를 사용하여 절단 가능한 이황화 결합을 통해 SLNP의 표면에 도입되었다.These FAP peptide-labeled SLNPs (FAPPEP-SLNPs) were composed of 1,2-dioleoyl-sn-glycero-3-phosphoethanolamine (DOPE) to promote endosomal escape; 1,2-distearoyl-sn-glycero-3-phosphoethanolamine-N-[carboxy(polyethylene glycol)-1000] (DSPE-PEG1000), which provides colloidal stability in vivo; and monoarginine-cholesterol (MA-Chol), which enables complexation with CpG adjuvants and provides mechanical stability (Figure 8). Peptide epitope peptides were introduced to the surface of SLNPs via cleavable disulfide bonds using cysteine-modified peptides at the N-terminus (N'-Cys-FAP PEP1 and N'-Cys-FAP PEP2 ) and DSPE-PEG2000 (Figures 9 and 10).
상기 두 시스테인 변형 펩타이드가 생체 내에서 면역원성을 유지한다는 것을 확인하였다(도 11a,b). 우리는 일단 FAPPEP-SLNP가 APC에 의해 흡수되면 세포질의 환원 환경에서 이황화 결합의 절단을 통해 유리 FAPPEP가 방출되고 세포막의 MHC 클래스 I에 의해 제시되는 것으로 예측된다(도 8a). 박막 재수화 방법을 사용하여 제조된 FAPPEP-SLNP의 유체역학적 크기는 동적 광산란(DLS)에 의해 ~144nm로 측정되었다(도 8b). 투과 전자 현미경(TEM)은 FAPPEP-SLNP가 평균 직경이 ~104 nm인 구형 형태를 가지는 것으로 나타내었다(도 8c).We confirmed that the two cysteine-modified peptides above maintained immunogenicity in vivo (Fig. 11a,b). We predict that once FAP PEP -SLNPs are taken up by APCs, free FAP PEP is released through cleavage of disulfide bonds in the reducing environment of the cytoplasm and presented by MHC class I on the cell membrane (Fig. 8a). The hydrodynamic size of FAP PEP -SLNPs prepared using the thin film rehydration method was measured to be ~144 nm by dynamic light scattering (DLS) (Fig. 8b). Transmission electron microscopy (TEM) showed that FAP PEP -SLNPs had a spherical morphology with an average diameter of ~104 nm (Fig. 8c).
다음으로 E.G7-OVA 종양 모델에서 FAPPEP1-SLNP 및 FAPPEP2-SLNP의 치료 효능을 평가하였다. E.G7-OVA 종양 보유 마우스를 각각의 나노백신 방식으로 4일 간격으로 3회 면역화하였다(도 8d). 예상대로, 표면에 FAPPEP 에피토프 펩타이드가 없는 대조군 SLNP는 종양 성장을 억제하지 못했다(도 8e). FAPPEP1-SLNP 처리는 FAPPEP2-SLNP로 처리한 것보다 훨씬 더 큰 항종양 효능을 나타냈으며, FAPPEP2-SLNP는 중간 정도의 종양 퇴행 효과를 나타냈다. 이러한 결과는 FAPPEP1-SLNP를 TME에 FAP+ CAF를 포함하는 종양을 치료하는 데 가장 효과적인 나노백신임을 의미한다. Next, we evaluated the therapeutic efficacy of FAP PEP1 -SLNP and FAP PEP2 -SLNP in the E.G7-OVA tumor model. E.G7-OVA tumor-bearing mice were immunized three times with each nanovaccine method at 4-day intervals (Fig. 8d). As expected, the control SLNP without the FAP PEP epitope peptide on the surface did not inhibit tumor growth (Fig. 8e). Treatment with FAP PEP1 -SLNP showed significantly greater antitumor efficacy than treatment with FAP PEP2 -SLNP, and FAP PEP2 -SLNP showed moderate tumor regression effect. These results imply that FAP PEP1 -SLNP is the most effective nanovaccine for treating tumors containing FAP+ CAFs in the TME.
FAPPEP1에 특이적인 CD8+ T 세포의 존재여부를 확인하기 위해 FAPPEP1-SNLP로 면역화된 마우스의 비장과 림프절을 적출하여 ICS 분석을 진행하였다. 그 결과, FAPPEP1과 동일한 아미노산을 가지지만, 서열이 무작위로 존재하는 대조군 펩타이드로 재자극하는 경우 IFN-ν의 분비가 낮은 것에 비해, FAPPEP1으로 재자극하는 경우에는 IFN-ν 활성이 크게 증가하여, 면역화를 통해 FAPPEP1에 특이적인 CD8+ 세포가 형성되었음을 확인하였다(도 12).To confirm the presence of CD8+ T cells specific for FAP PEP1 , ICS analysis was performed on the spleen and lymph nodes of mice immunized with FAP PEP1 -SNLP. As a result, when restimulated with a control peptide having the same amino acid sequence as FAP PEP1 but with a random sequence, IFN-ν secretion was low, whereas when restimulated with FAP PEP1 , IFN-ν activity was greatly increased, confirming that CD8+ cells specific for FAP PEP1 were formed through immunization (Fig. 12).
FAPPEP1에 특이적인 CD8+ 세포의 형성에 대한 추가적 확인을 위해 FAPPEP1-SNLP로 면역화된 마우스의 비장과 림프절을 적출하여 FAPPEP1에 특이적인 CD8+ 세포에 특이적으로 결합하는 항체인 FAPPEP1-H-2Kb 4중체를 ‘QuickSwitchTM Quant H-2 Kb Tetramer Kit-PE’ 제품(MBL Inc.)을 사용하여 제조하였다(MBL Inc). FAPPEP1-H-2Kb 4중체를 처리하는 경우, FAPPEP1-SLNP으로 면역화를 진행한 그룹에서 4중체가 붙었음을 나타내는 형광 반응이 매우 높게 나타남을 확인하였다(도 13).To further confirm the formation of CD8+ cells specific for FAP PEP1, the spleen and lymph nodes of mice immunized with FAP PEP1 -SNLP were extracted, and FAP PEP1 -H-2Kb tetramer, an antibody that specifically binds to CD8+ cells specific for FAP PEP1 , was prepared using the 'QuickSwitchTM Quant H-2 Kb Tetramer Kit-PE' product (MBL Inc.) (MBL Inc.). When FAP PEP1 -H-2Kb tetramer was treated, it was confirmed that the fluorescence reaction indicating that the tetramer was bound was very high in the group immunized with FAP PEP1 -SLNP (Fig. 13).
실시예 4: FAPPEP1-SLNP 나노백신의 종양 미세 환경에서의 CAF 제거 효과에 의한 치료 효능 확인Example 4: Confirmation of therapeutic efficacy of FAP PEP1 -SLNP nanovaccine by CAF removal effect in tumor microenvironment
다음으로 종양 성장을 억제하는 FAPPEP1-SLNP 나노백신의 작동 메커니즘을 확인하였다. 비교를 위해 OVA(ovalbumin) 특이적 CD8+ T 세포 반응을 유도하고 암세포를 제거함으로써 E.G7-OVA 종양에 대해 높은 항종양 효능을 나타내는 에피토프 펩타이드(SIINFEKL, 서열번호 150)를 표시하는 OVAPEP-SLNP 암세포 표적 나노백신(Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638)을 사용하였다. 평균 종양 부피가 ~100 mm3인 E.G7-OVA 종양 보유 마우스를 FAPPEP1-SLNP 또는 OVAPEP-SLNP로 4일 간격으로 총 3회 면역화하였다(도 14a). FAPPEP1-SLNP를 사용한 치료는 종양 성장을 상당히 억제하여 암세포를 표적으로 하는 OVAPEP-SLNP 나노백신과 비교하여 유사하거나 약간 더 나은 효능을 보였다(도 14b). 또한 처리되지 않은 대조군에 비해 생존 기간이 상당히 연장되었다(도 14c). Next, we investigated the mechanism of action of FAP PEP1 -SLNP nanovaccine to inhibit tumor growth. For comparison, we used an OVA PEP -SLNP cancer cell-targeted nanovaccine (Angew Chem Int Ed Engl 2020, 59 (34), 14628-14638) displaying an epitope peptide (SIINFEKL, SEQ ID NO: 150) that exhibits high antitumor efficacy against E.G7-OVA tumors by inducing OVA (ovalbumin)-specific CD8 + T cell responses and eliminating cancer cells. E.G7-OVA tumor-bearing mice with an average tumor volume of ~100 mm 3 were immunized three times in total with FAP PEP1 -SLNP or OVA PEP -SLNP at 4-day intervals (Fig. 14a). Treatment with FAP PEP1 -SLNP significantly inhibited tumor growth, showing similar or slightly better efficacy compared to OVA PEP -SLNP nanovaccine targeting cancer cells (Fig. 14b). It also significantly prolonged survival compared to untreated controls (Fig. 14c).
FAPPEP1-SLNP를 사용한 백신접종은 TME에서 FAP+ CAF에 대한 세포독성 면역 반응을 유도하기 때문에 종양에서 ECM 생산에 영향을 미칠 수 있다. Masson's trichrome (MT) 염색은 FAPPEP1-SLNP로 면역화하면 치료되지 않은 대조군 종양 조직과 비교하여 종양 조직 내 콜라겐 양성 영역이 크게 감소한다는 것을 나타내었다 (도 14d, e). 그러나 OVAPEP-SLNP가 FAPPEP1-SLNP와 유사한 항종양 효능을 보였지만 전자는 ECM 생산을 줄이는 데 실패했으며 이는 후자의 항종양 효능이 TME에서 FAP+ CAF 고갈의 결과일 수 있음을 의미한다. 유동 세포 계측법 분석은 FAPPEP1-SLNP로 면역화하면 종양 조직에서 FAP + CAF의 비율이 크게 감소하고 CD8 + 종양 침윤 림프구 (TIL)가 증가한다는 것을 확인하였다 (도 14f, g).Vaccination with FAP PEP1 -SLNPs may affect ECM production in tumors because it induces a cytotoxic immune response against FAP+ CAFs in the TME. Masson's trichrome (MT) staining showed that immunization with FAP PEP1 -SLNPs significantly reduced the collagen-positive area in tumor tissues compared with the untreated control tumor tissues (Fig. 14d, e). However, although OVA PEP -SLNPs showed similar antitumor efficacy as FAP PEP1 -SLNPs, the former failed to reduce ECM production, suggesting that the antitumor efficacy of the latter may be the result of the depletion of FAP+ CAFs in the TME. Flow cytometry analysis confirmed that immunization with FAP PEP1 -SLNPs significantly reduced the proportion of FAP+ CAFs and increased CD8+ tumor-infiltrating lymphocytes (TILs) in tumor tissues (Fig. 14f, g).
다음으로 FAPPEP1-SLNP 또는 OVAPEP-SLNP로 면역화된 마우스에서 분리한 비장 세포의 항원 특이적 CD8+ T 세포 반응을 평가하였다. 비장 세포에서 IFN-γ 분비 CD8+ T 세포의 빈도는 FAPPEP1 또는 OVAPEP로 생체 외 재자극 후 실질적으로 증가했으며 이는 항원 특이적 CD8+ T 세포 반응의 성공적인 유도를 나타내었다(도 14h). 흥미롭게도, FAPPEP1이 MHC 클래스 I의 에피토프로 예측되었지만 FAPPEP1-SLNP로 처리된 비장 세포 중에서 IFN-γ 분비 CD4+ T 세포의 빈도가 유의하게 증가하는 것을 관찰확인하였다. 그러나 OVAPEP-SLNP 나노백신은 항원 특이적 CD4+ T 세포 반응을 생성하지 못하는 것으로 나타났다(도 14i). Next, we evaluated antigen-specific CD8+ T cell responses in splenocytes isolated from mice immunized with FAP PEP1 -SLNP or OVA PEP -SLNP. The frequency of IFN-γ-secreting CD8+ T cells in splenocytes was substantially increased after ex vivo restimulation with FAP PEP1 or OVA PEP , indicating successful induction of antigen-specific CD8+ T cell responses (Fig. 14h). Interestingly, although FAP PEP1 is predicted to be a MHC class I epitope, we observed a significant increase in the frequency of IFN-γ-secreting CD4+ T cells among splenocytes treated with FAP PEP1 -SLNP. However, OVA PEP -SLNP nanovaccine failed to generate antigen-specific CD4+ T cell responses (Fig. 14i).
한편, FAP는 골수 간질 세포, 췌장 알파 세포 및 자궁 간질에서도 선택적으로 발현되기 때문에 우리의 FAP 표적 나노백신은 자가면역 관련 부작용 또는 전신 독성을 유발할 수 있다. 이러한 부작용 및 독성 여부를 평가하기 위해 FAPPEP1-SLNP로 면역화한 후 인터루킨-17A(IL-17A) 분비 T 헬퍼 17(Th17) 세포의 생성을 분석하였다. FAPPEP1-SLNP와 OVAPEP-SLNP 면역 사이의 Th17 세포 생성 빈도에서 유의미한 차이를 발견하지 못했으며(도 14j), 이는 FAPPEP1-SLNP가 자가 면역 반응을 일으키지 않는다는 것을 의미한다. 결과적으로, FAPPEP1-SLNP는 FAP 특이적 CD8+ T 세포 반응뿐만 아니라 CD4+ T 세포 반응을 유도할 수 있는 나노백신으로, 자가면역 반응을 유도할 가능성이 낮으면서도 높은 항종양 효능을 나타낼 수 있음을 의미한다(J Exp Med 2013, 210 (6), 1137-1151; J Exp Med 2013, 210 (6), 1125-1135; Cancer Cell 2014, 25 (6), 719-734; Nat Rev Immunol 2018, 18 (10), 635-647; Immunity 2021, 54 (12), 2701-2711).Meanwhile, since FAP is also selectively expressed in bone marrow stromal cells, pancreatic α cells, and uterine stroma, our FAP-targeted nanovaccine may induce autoimmune-related adverse effects or systemic toxicity. To evaluate such adverse effects and toxicity, we analyzed the generation of interleukin-17A (IL-17A)-secreting T helper 17 (Th17) cells after immunization with FAP PEP1 -SLNP. We found no significant difference in the frequency of Th17 cell generation between FAP PEP1 -SLNP and OVA PEP -SLNP immunization (Fig. 14j), indicating that FAP PEP1 -SLNP does not induce autoimmune responses. In conclusion, FAP PEP1 -SLNP is a nanovaccine that can induce not only FAP-specific CD8+ T cell responses but also CD4+ T cell responses, indicating that it can exhibit high antitumor efficacy with a low possibility of inducing autoimmune responses (J Exp Med 2013, 210 (6), 1137-1151; J Exp Med 2013, 210 (6), 1125-1135; Cancer Cell 2014, 25 (6), 719-734; Nat Rev Immunol 2018, 18 (10), 635-647; Immunity 2021, 54 (12), 2701-2711).
다음으로, FAPPEP1-SLNP를 사용하여 면역화한 마우스 그룹의 종양에서 혈관을 확인하였다. 그 결과, 대조군을 투여한 경우에 비해 FAPPEP1-SLNP로 면역화한 마우스의 종양 내 혈관이 현저히 감소됨을 확인하였다(도 15a 및 b). 이는 CAF가 VEGF 같은 혈관 생성인자를 분비하는 것에 기초하여, CAF의 감소로 인한 신생혈관 형성이 감소하였음을 의미한다. Next, blood vessels were identified in the tumors of the mice immunized with FAP PEP1 -SLNP. As a result, it was confirmed that blood vessels in the tumors of the mice immunized with FAP PEP1 -SLNP were significantly reduced compared to the control group (Fig. 15a and b). This suggests that angiogenesis was reduced due to the reduction of CAFs, based on the fact that CAFs secrete angiogenic factors such as VEGF.
FAPPEP1-SLNP를 사용하여 면역화한 마우스 그룹의 폐에서 종양의 전이를 확인하였다. 그 결과 대조군을 투여한 경우에 비해 FAPPEP1-SLNP로 면역화한 마우스의 종양 전이가 현저히 감소한 것을 확인하였다(도 15c 및 d). 이는 CAF가 전-전이성 니케(pre-metastatic niche)를 형성하는 것에 큰 역할을 수행하는 것으로 알려진 것에 기초하여, CAF의 감소로 인한 전이 억제효과를 나타낸 것으로 해석될 수 있다.We confirmed tumor metastasis in the lungs of the mice immunized with FAP PEP1 -SLNP. As a result, we confirmed that tumor metastasis in the mice immunized with FAP PEP1 -SLNP was significantly reduced compared to the control group (Fig. 15c and d). Based on the fact that CAFs are known to play a major role in forming the pre-metastatic niche, this can be interpreted as an inhibitory effect of metastasis due to the reduction of CAFs.
실시예 5: FAPPEP1-SLNP 나노백신의 다양한 종양에 대한 효과 확인Example 5: Confirmation of the efficacy of FAP PEP1 -SLNP nanovaccine against various tumors
FAPPEP1-SLNP가 다른 결합조직성 종양(desmoplastic tumor)에도 작용할 수 있을지 확인하기 위해, 결합조직성 종양으로 알려진 MC38, Panc02 종양에서 FAP+ CAF가 존재하는지 여부를 확인하였다. 평균 300 mm3 에 도달한 E.G7-OVA, MC38, Panc02 종양을 적출한 뒤에 single cell로 dissociation을 진행하고, 유세포 분석을 통하여 분석하였다. 그 결과, E.G7-OVA 종양을 포함하여 나머지 두 종양 모델 모두에서 FAP+ CAF의 비율이 높음을 확인하였다(도 16).To determine whether FAP PEP1 -SLNP could act on other desmoplastic tumors, we examined whether FAP + CAFs existed in MC38 and Panc02 tumors, which are known as desmoplastic tumors. After reaching an average of 300 mm 3 , E.G7-OVA, MC38, and Panc02 tumors were excised, dissociated into single cells, and analyzed by flow cytometry. As a result, we confirmed that the ratio of FAP + CAFs was high in both tumor models, including E.G7-OVA tumors (Fig. 16).
또한, FAPPEP1-SLNP가 종양의 초기에 FAP+ CAF를 없앤 뒤에도 추가 접종 시에 지속적인 종양 억제 효과를 나타내는 것을 확인하였다. 이에 암세포(Cancer cell) 자체도 FAP 단백질을 발현하는지 확인해보고자 실험을 진행했다. MC38 종양의 크기가 300 mm3 에 도달했을 때 종양을 적출하여 single cell로 dissociation을 진행했고, 암세포와 FAP+ CAF의 존재 비율을 유세포 분석을 통해 확인했다. 그 결과, 큰 크기의 종양에서는 암세포 자체에서도 FAP가 발현함을 알 수 있었고, 그렇기에 7FAPPEP1-SLNP가 초기에 FAP+ CAF를 제거한 뒤에도 추가 접종에 따른 지속적인 치료 효능을 보이는 것과 일치함을 확인하였다(도 17).In addition, we confirmed that FAP PEP1 -SLNP showed a continuous tumor suppression effect upon additional inoculation even after FAP + CAF were removed in the early stage of the tumor. Therefore, we conducted an experiment to confirm whether cancer cells themselves also express the FAP protein. When the size of MC38 tumors reached 300 mm 3 , the tumors were excised and dissociated into single cells, and the abundance ratio of cancer cells and FAP + CAF was confirmed through flow cytometry. As a result, it was found that FAP was expressed in the cancer cells themselves in large tumors, and therefore, it was confirmed that 7FAP PEP1 -SLNP showed a continuous therapeutic efficacy upon additional inoculation even after FAP + CAF were removed in the early stage (Fig. 17).
이러한 결과를 근거로, ECM이 많은 결합조직성 종양인 PDAC(pancreatic ductal adenocarcinoma) 세포주인 Panc02와 대장암(colon cancer) 세포주인 MC38 종양 모델에서 FAPPEP1-SLNP 나노백신이 치료효능를 가지는지 확인하였다. 두 종양 모델 모두 종양의 평균 사이즈가 100 mm3 일 때 면역화를 시작했고, 이전 실험과 동일한 스케쥴인 4일 간격으로 총 3번의 면역화를 진행하였다. 그 결과, Panc02 종양 모델과 (도 18a-c) MC38 종양 모델 (도 18d-f) 모두에서 FAPPEP1-SLNP의 접종을 진행한 쥐들의 종양크기가 더디게 자람은 물론 증가된 생존율을 나타내는 것을 확인하였다(도 18). Based on these results, we confirmed whether the FAP PEP1 -SLNP nanovaccine had therapeutic efficacy in Panc02, a pancreatic ductal adenocarcinoma (PDAC) cell line with abundant ECM, and MC38, a colon cancer cell line. In both tumor models, immunization was started when the average tumor size was 100 mm 3 , and a total of three immunizations were performed at 4-day intervals, the same schedule as the previous experiment. As a result, in both the Panc02 tumor model (Fig. 18a-c) and the MC38 tumor model (Fig. 18d-f), it was confirmed that the mice vaccinated with FAP PEP1 -SLNP showed slower tumor growth and increased survival rate (Fig. 18).
추가적으로, MC38 종양 모델에서 종양 안에 있는 CD8 T 세포 및 CD4 T 세포가 FAP+ 세포에 대한 면역활성을 가지는지 확인해보고자 MC38 tumor를 적출 후 single cell로 dissociation하여 ICS 분석을 수행하였다. 그 결과 FAPPEP1-SLNP를 접종한 쥐들의 TIL (tumor infiltrating lymphocyte)의 경우, 대조군에 비하여 유의미한 IFN-γ의 분비 증가를 확인할 수 있었고, 이는 실제로 FAPPEP1-SLNP에 의하여 만들어진 FAP에 특이적인 CD8+ T cell, CD4+ T cell이 tumor 내에 존재함을 확인하였으며(도 19), 이는 종양 내의 FAP+ 세포에도 효과적으로 작용을 함을 의미한다.Additionally, to determine whether CD8 T cells and CD4 T cells within the tumor have immune activity against FAP + cells in the MC38 tumor model, MC38 tumors were extracted, dissociated into single cells, and ICS analysis was performed. As a result, in the case of TILs (tumor infiltrating lymphocytes) of mice vaccinated with FAP PEP1 -SLNP, a significant increase in the secretion of IFN-γ was confirmed compared to the control group. This confirmed that FAP-specific CD8 + T cells and CD4 + T cells produced by FAP PEP1 -SLNP are present within the tumor (Fig. 19), which means that they effectively act on FAP + cells within the tumor.
이러한 결과(도 16 내지 도 19)은 본 발명의 에피토프 펩타이드 및 이를 포함하는 백신이 다른 결합조직성 종양 또는 FAP 발현 종양에 대한 범-종양 백신으로 사용될 수 있음을 의미한다.These results (Figs. 16 to 19) imply that the epitope peptide of the present invention and the vaccine comprising it can be used as a pan-tumor vaccine against other connective tissue tumors or FAP expressing tumors.
실시예 6: FAPPEP1-SLNP 나노백신과 항암제 병용요법을 통한 항종양 효과 확인 Example 6: Confirmation of antitumor effect through combination therapy of FAP PEP1 -SLNP nanovaccine and anticancer drug
결합조직성 종양의 조밀하고 단단한 ECM은 화학요법제뿐만 아니라, 항체 및 CAR-T 세포와 같은 생물학적 제제에 대한 강력한 물리적 장벽으로 작용하며, 다양한 항암 요법의 치료효과를 감소시킨다(Signal Transduct Target Ther 2021, 6 (1), 153, Proc Natl Acad Sci U S A 2019, 116 (6), 2210-2219;, Nat Nanotechnol 2021, 16 (1), 25-36). 본 발명자들은 FAPPEP1-SLNP 나노백신의 TME에서의 CAF 고갈 능력이 ECM 밀도를 감소시켜, 결합조직성 종양 내 화학 요법 약물의 침투를 향상시킬 수 있을 것으로 예측하였다.The dense and rigid ECM of connective tissue tumors acts as a powerful physical barrier not only to chemotherapeutic agents but also to biological agents such as antibodies and CAR-T cells, reducing the therapeutic efficacy of various anticancer therapies (Signal Transduct Target Ther 2021, 6 (1), 153, Proc Natl Acad Sci USA 2019, 116 (6), 2210-2219;, Nat Nanotechnol 2021, 16 (1), 25-36). We predicted that the ability of FAP PEP1 -SLNP nanovaccine to deplete CAFs in the TME could reduce ECM density, thereby enhancing the penetration of chemotherapeutic drugs into connective tissue tumors.
이를 검증하기 위해 뮤린 췌관 선암종 세포주 Panc02와 결장직장암 세포주 MC38이 결합조직성 종양 모델로 선택되었다(PLoS One 2013, 8 (11), e80580; Cancer Res 2018, 78 (5), 1321-1333; Nat Commun 2020, 11 (1), 515). 먼저, FAPPEP1-SLNP 나노백신을 사용한 면역화가 종양 성장을 현저히 늦추고 면역화된 마우스의 생존 기간을 연장함을 확인하였다. Panc02(도 20a-c) 및 MC38 종양 모델(도 20d-f) 모두에서 면역화되지 않은 마우스를 대조군으로 사용하여 FAPPEP1-SLNP 나노백신이 이러한 결합조직성 종양에도 효과적임을 확인하였다. To verify this, the murine pancreatic ductal adenocarcinoma cell line Panc02 and the colorectal cancer cell line MC38 were chosen as connective tissue tumor models (PLoS One 2013, 8 (11), e80580; Cancer Res 2018, 78 (5), 1321-1333; Nat Commun 2020, 11 (1), 515). First, we confirmed that immunization with FAP PEP1 -SLNP nanovaccine significantly slowed tumor growth and prolonged the survival period of immunized mice. Non-immunized mice were used as controls in both Panc02 (Fig. 20a-c) and MC38 tumor models (Fig. 20d-f), confirming that the FAP PEP1 -SLNP nanovaccine was also effective against these connective tissue tumors.
다음으로 FAP+ CAF 표적 나노백신이 소분자 항암제의 종양 침투를 향상시킬 수 있는지 확인하였다. Cypate 염료는 종양에서 약물 침투를 시각화하기 위한 모델 약물로 선택되었다. MC38 결장직장 종양 보유 마우스를 4일 간격으로 총 3회 주사하는 FAPPEP1-SLNP 나노백신으로 면역화시켰다. 종양 부피가 ~300 mm3에 도달한 후, 자유 Cypate를 면역화된 마우스에 정맥 주사하고, 2시간 후에 생체외 형광 이미징을 위해 종양을 수확하였다. 종양 내 염료의 축적은 백신 접종되지 않은 대조군보다 백신 접종 그룹에서 훨씬 더 높았으며 (도 21a), 이는 종양 내 ECM 밀도 감소로 인해 약물 침투가 향상되었음을 의미한다. Next, we investigated whether the FAP+ CAF-targeted nanovaccines could enhance the tumor penetration of small molecule anticancer drugs. Cypate dye was chosen as a model drug to visualize drug penetration in tumors. MC38 colorectal tumor-bearing mice were immunized with FAP PEP1 -SLNP nanovaccines for a total of three injections at 4-day intervals. After the tumor volume reached ~300 mm 3 , free Cypate was intravenously injected into the immunized mice, and the tumors were harvested for ex vivo fluorescence imaging 2 h later. The accumulation of dye in the tumors was significantly higher in the vaccinated group than in the unvaccinated control group ( Figure 21a ), suggesting that the drug penetration was enhanced due to the decrease in the density of the intratumoral ECM.
나아가, FAPPEP1-SLNP 면역 요법과 독소루비신(Dox)을 사용한 병용 요법을 수행하여 예측된 것과 같은 시너지 효과를 나타낼 수 있는지 확인하였다. MC38 유래 결장직장 종양이 ~100 mm3 크기에 도달한 후, 마우스를 FAPPEP1-SLNP로 4일 간격으로 3회 면역화하고 격일로 Dox를 복강내로 총 4회 주사하였다(도 21b). FAPPEP1-SLNP 나노백신과 Dox 단일 요법 모두 상당한 항종양 효능을 보였지만, 병용 요법은 단독 요법보다 종양 성장 억제에 훨씬 더 효과적이었다(도 21c). 또한, 처리된 종양의 유세포 분석 결과 FAP+ CAF의 비율이 FAPPEP1-SLNP 나노백신 및 조합 그룹 모두에서 유의하게 감소했지만 Dox 단일 처리 그룹에서는 그렇지 않은 것으로 나타났다(도 21d). 또한 병용 투여 그룹에서 CD8+ TIL의 비율이 증가하는 경향이 있었지만 이 차이는 통계적으로 유의하지 않았다(도 21e). 종합하면, 이러한 결과는 FAPPEP1-SLNP 나노백신이 결합조직성 종양의 성장을 억제하는 데 효과적일 뿐만 아니라, 종양 내에서 약물 침투를 증가시켜 화학요법 약물과 함께 그 효능이 실질적으로 더 향상된다는 것을 의미한다(도 22).Furthermore, we performed combination therapy with FAP PEP1 -SLNP immunotherapy and doxorubicin (Dox) to determine whether the combination therapy could exhibit the predicted synergistic effects. After MC38-derived colorectal tumors reached a size of ~100 mm 3 , mice were immunized three times with FAP PEP1 -SLNP at 4-day intervals followed by intraperitoneal injection of Dox every other day for a total of 4 times (Fig. 21b). Although both FAP PEP1 -SLNP nanovaccine and Dox monotherapy exhibited significant antitumor efficacy, the combination therapy was much more effective in inhibiting tumor growth than either monotherapy (Fig. 21c). In addition, flow cytometry analysis of the treated tumors showed that the proportion of FAP+ CAFs was significantly reduced in both the FAP PEP1 -SLNP nanovaccine and combination groups, but not in the Dox monotherapy group (Fig. 21d). In addition, although the proportion of CD8+ TILs tended to increase in the combination treatment group, this difference was not statistically significant (Fig. 21e). Taken together, these results suggest that the FAP PEP1 -SLNP nanovaccine is not only effective in inhibiting the growth of connective tissue tumors, but also substantially enhances its efficacy in combination with chemotherapeutic drugs by increasing drug penetration within the tumor (Figure 22).
실시예 7: FAPPEP1 펩타이드 백신의 암 전이 억제 효과 확인Example 7: Confirmation of the cancer metastasis inhibition effect of FAP PEP1 peptide vaccine
FAP+CAF는 원발성 종양에서 세포외 소포를 분비하여, 전이를 촉진하고 암세포와의 클러스터 형성을 통해 혈액 순환 중 아노이키스(anoikis) 및 전단 스트레스(shear stress)로부터 암세포를 보호한다. 따라서, 기존에 사용되던 꼬리 정맥 주입에 의한 전이 모델은 FAP+CAF의 역할을 확인하는데 적합하지 않다. FAP+CAF secrete extracellular vesicles from primary tumors to promote metastasis and protect cancer cells from anoikis and shear stress in the blood circulation by forming clusters with cancer cells. Therefore, the previously used tail vein injection metastasis model is not suitable for confirming the role of FAP+CAF.
이러한 이유로 본 실시예에서는, FAP+CAF의 연관성 및 FAPPEP1 펩타이드 백신의 암 전이 억제 효과를 확인하기 위해, EO771.lmb 전이성 유방암 세포주를 사용하여 실시예 1-13에 기재된 방법으로 동소 유방 종양 모델을 구축하였다. 실시예 5에서 평균 300 mm3의 종양 크기에서 FAP+ CAF의 비율이 높음을 확인하였기 때문에(도 16), 종양 크기가 300 mm3에 도달할 때 종양을 절제하였다.For this reason, in this example, to confirm the association of FAP+CAF and the metastasis inhibition effect of the FAP PEP1 peptide vaccine, an orthotopic breast tumor model was constructed using the EO771.lmb metastatic breast cancer cell line by the method described in Examples 1-13. Since it was confirmed that the ratio of FAP+CAF was high at an average tumor size of 300 mm 3 in Example 5 (Fig. 16), the tumor was resected when the tumor size reached 300 mm 3 .
절제 4일 후, FAPPEP1 펩타이드 백신와 CpG ODN의 혼합물을 1주일 간격으로 2회 발바닥에 피하주사를 통해 면역화하여 전이에 대한 FAPPEP1 펩타이드 백신의 전이 억제 효과를 확인하였다(도 23).Four days after resection, the FAP PEP1 peptide vaccine and CpG ODN were immunized twice by subcutaneous injection into the footpad at one-week intervals to confirm the metastasis-suppressing effect of the FAP PEP1 peptide vaccine on metastasis (Fig. 23).
EO771.lmb 종양은 주로 폐와 간으로 전이되는 것으로 알려져 있기 때문에, 면역화 주사의 1주일 뒤 폐 및 간 조직을 적출하여 조직학적 분석을 실시하였다.Since EO771.lmb tumors are known to metastasize primarily to the lungs and liver, lung and liver tissues were removed one week after the immunization injection and subjected to histological analysis.
폐 조직의 H&E 염색 분석 결과 FAPPEP1 펩타이드 백신을 투여한 경우 전이가 현저히 감소한 반면, 대조군에서는 전이 면적이 증가한 것으로 나타났다(도 24). H&E staining analysis of lung tissue showed that administration of the FAP PEP1 peptide vaccine significantly reduced metastasis, whereas the metastatic area increased in the control group (Fig. 24).
또한, 간 조직의 H&E 염색에서도 FAPPEP1 펩타이드 백신을 투여한 경우 대조군에 비해 전이가 유의하게 감소한 것으로 나타났다(도 25). In addition, H&E staining of liver tissue showed that metastasis was significantly reduced when the FAP PEP1 peptide vaccine was administered compared to the control group (Fig. 25).
이러한 결과는 본 발명의 FAPPEP1 백신이 전이에서 FAP+CAF를 표적화함으로써 암의 전이를 효과적으로 예방할 수 있음을 입증한다.These results demonstrate that the FAP PEP1 vaccine of the present invention can effectively prevent cancer metastasis by targeting FAP+CAF in metastasis.
실시예 8: FAPPEP1 펩타이드 백신의 비알콜성 지방간염(NASH) 치료 효과 확인Example 8: Confirmation of the efficacy of FAP PEP1 peptide vaccine in treating nonalcoholic steatohepatitis (NASH)
비알콜성 지방간염(NASH)는 간 지방증과 염증을 특징으로 하는 만성 간 질환으로, 섬유증, 간경화 등을 거쳐 간암으로 진행될 수 있다. FAP는 지질의 중요한 조절자인 섬유아세포 성장인자 21(FGF21)을 절단하며, 이에 의한 조절장애는 NASH의 진행에 기여할 수 있다. Nonalcoholic steatohepatitis (NASH) is a chronic liver disease characterized by hepatic steatosis and inflammation, which may progress to fibrosis, cirrhosis, and liver cancer. FAP cleaves fibroblast growth factor 21 (FGF21), an important regulator of lipids, and dysregulation thereof may contribute to the progression of NASH.
실시예 8-1: FAPPEP1 펩타이드 백신의 NASH 치료효과 확인Example 8-1: Confirmation of NASH therapeutic effect of FAP PEP1 peptide vaccine
본 발명의 FAPPEP1 펩타이드 백신의 비알콜성 지방간염의 치료 효과를 확인하기 위해, 메타오닌-콜린 결핍 식이(MCD)를 사용하여 NASH 모델을 구축하였다(Nat Rev Gastro Hepat 16, 411-428 (2019)). NASH 모델 구축은 체중과 간 중량 모니터링을 통해 검증하였다. 4주간의 MCD 식이요법 및 MCS 식이요법을 통해 2 내지 3 스테이지의 NASH 모델과 비교군 모델을 준비하였으며, 1주 간격으로 각 마우스에 FAPPEP1 펩타이드 백신과 CpG ODN의 혼합물을 양쪽 발박닥에 1주 간격으로 2회 피하 주사하였다(도 27a). FAPPEP1 펩타이드 백신의 투여 후, 체중이 현저히 회복되었으며(도 27b), 최종 면역화 1주일 후 마우스의 간을 적출하여 무게를 측정한 결과, FAPPEP1 펩타이드 백신의 투여 후 간 무게가 현저히 회복되는 것을 확인하였다(도 27c).To confirm the therapeutic effect of the FAP PEP1 peptide vaccine of the present invention on nonalcoholic steatohepatitis, a NASH model was established using a methionine-choline deficient diet (MCD) (Nat Rev Gastro Hepat 16, 411-428 (2019)). The establishment of the NASH model was verified through monitoring of body weight and liver weight.
또한, FAPPEP1 펩타이드 백신으로 면역화한 MCD 식이 유발 NASH 모델에서, 간 중량 대 체중 비율이 눈에 띄게 증가하여(도 27d), FAPPEP1 펩타이드 백신이 NASH의 치료에도 뛰어난 효과를 나타내는 것을 확인하였다.In addition, in the MCD diet-induced NASH model immunized with the FAP PEP1 peptide vaccine, the liver weight-to-body weight ratio was markedly increased (Fig. 27d), confirming that the FAP PEP1 peptide vaccine also exhibited excellent effects in the treatment of NASH.
실시예 8-2: FAPPEP1 펩타이드 백신의 간 손상 완화 효과 확인Example 8-2: Confirmation of the liver damage alleviation effect of FAP PEP1 peptide vaccine
간 손상의 완화를 확인하기 위해, 마우스의 희생직전에 혈청을 수집하였다. FAPPEP1 펩타이드 백신으로 면역화된 NASH 모델의 마우스는 간손상 지표인 AST, ALT 및 총 빌리루빈 수치가 크게 감소한 것으로 나타났다(도 28a 내지 28c). 또한, HDL의 수준이 증가하여(도 28d), FAPPEP1 펩타이드 백신이 MCD 식이로 유발된 간손상에 대한 보호효과가 있음을 확인하였다.To confirm the alleviation of liver damage, serum was collected just before the sacrifice of mice. Mice of the NASH model immunized with the FAP PEP1 peptide vaccine showed a significant decrease in the levels of AST, ALT, and total bilirubin, which are indicators of liver damage (Figs. 28a to 28c). In addition, the level of HDL was increased (Fig. 28d), confirming that the FAP PEP1 peptide vaccine had a protective effect against liver damage induced by the MCD diet.
실시예 8-3: FAPPEP1 펩타이드 백신에 의한 면역화 후 간조직 분석 결과Example 8-3: Liver tissue analysis results after immunization with FAP PEP1 peptide vaccine
NASH 모델에서 간조직에 대한 FAPPEP1 펩타이드 백신의 효과를 검증하기 위해 Oil Red O 염색 및 H&E 염색을 사용하여 조직 분석을 수행하였다.To verify the effect of the FAP PEP1 peptide vaccine on liver tissue in the NASH model, tissue analysis was performed using Oil Red O staining and H&E staining.
FAPPEP1 펩타이드 백신으로 면역화된 NASH 모델에서 Oil Red O 염색은 간 지방증(hepatic steatosis)과 관련된 뛰어난 효과를 나타내었다. 구체적으로 도 29a 및 29b에 도시된 것과 같이, FAPPEP 펩타이드 백신으로 면역화된 마우스의 간은 Oil Red O 염색의 강도가 현저히 감소하여, 세포 내 지질 축적이 크게 감소하였음을 확인하였다.In the NASH model immunized with the FAP PEP1 peptide vaccine, Oil Red O staining showed a remarkable effect related to hepatic steatosis. Specifically, as shown in Figures 29a and 29b, the livers of mice immunized with the FAP PEP peptide vaccine showed a marked decrease in the intensity of Oil Red O staining, confirming a significant decrease in intracellular lipid accumulation.
H&E 염색은 간 조직에서 풍선 모양의 지방세포를 나타내는데, MCD 식이요법을 실시한 대조군에서 간에 지방세포를 나타내는 미염색 부분이 나타나는 반면, FAPPEP1 펩타이드 백신으로 면역화된 마우스에서는 간세포 팽창이 현저히 감소한 것을 확인하였다(도 30a 및 30b).H&E staining revealed balloon-shaped adipocytes in the liver tissue, and while unstained areas representing adipocytes were seen in the liver of the MCD diet-fed control group, a marked reduction in hepatocyte swelling was observed in mice immunized with the FAP PEP1 peptide vaccine (Figs. 30a and 30b).
결과적으로, 조직학적 분석 결과는 FAPPEP1 펩타이드 백신이 간 지방증에 뛰어난 치료효과를 나타내어, NASH 모델에서 간 조직 회복을 유도할 수 있음을 나타낸다. 이러한 조직학적 결과는 체중, 간 중량 및 혈청 분석과 같은 실시예에서의 결과를 뒷받침하여, NASH 치료에서 FAPPEP-SLNP 나노백신의 뛰어난 치료효과를 의미한다.As a result, the histological analysis results indicate that the FAP PEP1 peptide vaccine exhibits an excellent therapeutic effect on hepatic steatosis, which can induce liver tissue recovery in the NASH model. These histological results support the results in the examples, such as body weight, liver weight, and serum analysis, implying an excellent therapeutic effect of the FAP PEP -SLNP nanovaccine in the treatment of NASH.
실시예 9: 인간 FAP 에피토프 및 나노백신Example 9: Human FAP epitope and nanovaccine
실시예 2 내지 8의 결과는 예측된 FAP 에피토프를 사용한 면역화 요법이 암의 치료, 전이 억제뿐만 아니라, NASH와 같은 FAP 관련 섬유화 질환에 뛰어난 치료효과를 나타낼 수 있음을 입증한다. 인간의 암에 존재하는 암연관 섬유아세포(cancer-associated fibroblast)와 섬유화질환에서의 myofibroblasts 또는 hepatic stellate cells에는 FAP가 선택적으로 과다발현되어있고 마우스 모델에서와 동일한 기작으로 암을 성장 및 전이시키거나 섬유화질환을 매개한다는 것은 많은 연구를 통해 이미 잘 알려져 있다. 따라서, 마우스 암 모델 및 NASH 모델에서 보여주었던 FAP 에피토프 백신의 효능 결과가 인간에게도 적용될 수 있을 것임을 말해준다. 또한, 생쥐의 FAP 단백질과 인간의 FAP 단백질 서열의 유사도는 약 94% 수준으로서 마우스에서 검증한 FAPPEP1 펩타이드 에피토프가 인간의 특정 HLA type과 cross-reactivity가 존재할 가능성이 높다. 본 실시예에서 검증된 FAPPEP1이 인간 HLA-A2에서 예측한 펩타이드 에피트포 후보군과 서열이 일치한다.The results of Examples 2 to 8 demonstrate that immunotherapy using the predicted FAP epitope can exhibit excellent therapeutic effects not only for the treatment of cancer and inhibition of metastasis, but also for FAP-related fibrotic diseases such as NASH. It has already been well known through many studies that FAP is selectively overexpressed in cancer-associated fibroblasts present in human cancers and myofibroblasts or hepatic stellate cells in fibrotic diseases and that it promotes the growth and metastasis of cancer or mediates fibrotic diseases by the same mechanism as in mouse models. Therefore, it is suggested that the efficacy results of the FAP epitope vaccine shown in mouse cancer models and NASH models can be applied to humans. In addition, the similarity between the FAP protein of mice and the FAP protein of humans is about 94%, so there is a high possibility that the FAP PEP1 peptide epitope verified in mice has cross-reactivity with a specific HLA type in humans. In this example, the FAP PEP1 verified has a sequence identical to the peptide epitope candidate predicted from human HLA-A2.
본 실시예에서는 HLA Super type에 따른 인간 FAP(서열번호 153, Uniprot No. B4DLR2)의 에피토프를 추가로 확인하였다. FAP 표적 에피토프 펩타이드의 예측은 NetMHC3.0, BIMAS 및 PREDEP을 포함하는 에피토프 펩타이드 예측 알고리즘을 사용하여 수행되었다. 모든 에피토프 펩타이드는 MHC-Ⅰ 일배체형인 H-2Kb에 의해 인식되는 8-mer 내지 10-mer 펩타이드 길이로 선택되었다. HLA Super type은 HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8 및 HLA-B27에 대해 분석되었다. 최종 도출된 FAP 에피토프 서열은 다음 표 2 내지 표 5과 같다.In this example, the epitope of human FAP (SEQ ID NO: 153, Uniprot No. B4DLR2) according to HLA Super type was additionally confirmed. Prediction of FAP target epitope peptides was performed using epitope peptide prediction algorithms including NetMHC3.0, BIMAS, and PREDEP. All epitope peptides were selected as 8-mer to 10-mer peptide lengths recognized by H-2Kb, which is an MHC-I haplotype. HLA Super types were analyzed for HLA-A1, HLA-A2, HLA-A3, HLA-A24, HLA-A26, HLA-B7, HLA-B8, and HLA-B27. The final derived FAP epitope sequences are shown in Tables 2 to 5 below.
[표 2][Table 2]
[표 3][Table 3]
[표 4][Table 4]
[표 5][Table 5]
FAP 단백질을 표적하여 FAP를 발현하는 세포를 제거할 수 있는 FAP 백신 임상시험은 다음과 같이 수행될 수 있다. 1) 각 환자의 HLA type을 분석하여 환자 개인에 해당하는 하나 이상의 FAP 에피토프 펩타이드를 표1 내지 표5에서 기재된 펩타이드 서열에서 선정한다. 2) 선정된 펩타이드 서열 후보군을 어쥬번트(CpG 또는 poly(I:C) 등)와 같이 투여하거나, 3) 선정된 펩타이드 서열 후보군들을 나노입자에 포함한 나노백신으로 제조하여 어쥬번트와 같이 투여할 수 있다.상기 실시예들에서 확인한 효과를 토대로, 본 발명의 표 2 내지 표 5에 기재된 펩타이드 서열을 사용한 백신화 요법으로, 암, 암의 전이, NASH 등과 같은 FAP 관련 질환의 예방 및 치료에 뛰어난 효과를 나타낼 것임은 자명하게 예측될 수 있다.A clinical trial of a FAP vaccine capable of targeting the FAP protein and eliminating cells expressing FAP can be performed as follows. 1) Analyze the HLA type of each patient, and select one or more FAP epitope peptides corresponding to the individual patient from the peptide sequences described in Tables 1 to 5. 2) The selected peptide sequence candidates can be administered together with an adjuvant (such as CpG or poly(I:C)), or 3) the selected peptide sequence candidates can be manufactured into a nanovaccine including nanoparticles and administered together with an adjuvant. Based on the effects confirmed in the above examples, it can be clearly predicted that the vaccination therapy using the peptide sequences described in Tables 2 to 5 of the present invention will exhibit excellent effects in the prevention and treatment of FAP-related diseases such as cancer, cancer metastasis, NASH, etc.
본 발명의 펩타이드는 뛰어난 면역우세성을 가지며, 섬유아세포 활성화 단백질(FAP) 및 FAP 발현 섬유아세포(FAP+ CAF) 특이적인 CD8+ T 세포의 강력한 유도 효과를 나타낸다. 본 발명의 펩타이드는 섬유아세포 활성화 단백질에 대한 면역 유도를 통해 암, 섬유화증, NASH 등과 같읕 FAP 관련 질환에 뛰어난 예방 및 치료효과를 나타낼 수 있으며, 다른 FAP 발현 CAF 표적 백신에 비해 현저하게 낮은 독성을 갖는다. 특히, 암의 경우 FAP 발현 CAF의 고갈, TME에서의 ECM 생산 감소, 암의 전이 억제와 같은 뛰어난 항-종양 효과를 나타낸다. 본 발명의 펩타이드를 다른 항암제와 병용투여하는 경우에, 항암제의 종양 세포로의 축적을 증가시키고, 현저한 항 종양 효과의 향상을 나타내어, 범-종양(Pan-tumor) 백신으로서 유용하게 사용될 수 있다.The peptide of the present invention has excellent immunodominance and exhibits a strong induction effect of CD8+ T cells specific for fibroblast activation protein (FAP) and FAP-expressing fibroblasts (FAP+ CAF). The peptide of the present invention can exhibit excellent preventive and therapeutic effects on FAP-related diseases such as cancer, fibrosis, NASH, etc. through immunity induction against fibroblast activation protein, and has significantly lower toxicity compared to other FAP-expressing CAF targeting vaccines. In particular, in the case of cancer, it exhibits excellent anti-tumor effects such as depletion of FAP-expressing CAF, reduction of ECM production in the TME, and inhibition of cancer metastasis. When the peptide of the present invention is administered in combination with other anticancer agents, it increases the accumulation of anticancer agents in tumor cells and exhibits a remarkable enhancement of the antitumor effect, so that it can be usefully used as a pan-tumor vaccine.
이상으로 본 발명의 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.While the specific parts of the present invention have been described in detail above, it will be apparent to those skilled in the art that such specific descriptions are merely preferred embodiments and that the scope of the present invention is not limited thereby. Accordingly, the actual scope of the present invention will be defined by the appended claims and their equivalents.
전자파일 첨부하였음.Electronic file attached.
Claims (38)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR20230054746 | 2023-04-26 | ||
| KR10-2023-0054746 | 2023-04-26 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2024225764A1 true WO2024225764A1 (en) | 2024-10-31 |
Family
ID=93256833
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2024/005587 Pending WO2024225764A1 (en) | 2023-04-26 | 2024-04-25 | Immunodominant epitope peptides of fibroblast activating proteins, and uses thereof |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR20240158819A (en) |
| WO (1) | WO2024225764A1 (en) |
Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080058272A1 (en) * | 2006-08-29 | 2008-03-06 | Juergen Becker | Nonamer Peptides for Cancer Treatment |
| KR20160106192A (en) * | 2010-03-19 | 2016-09-09 | 이매틱스 바이오테크놀로지스 게엠베하 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
| CN105949302A (en) * | 2016-05-27 | 2016-09-21 | 郑州大学 | FAP(fibroblast activation protein)-sourced anti-tumor CTL (cytotoxic T lymphocyte) epitope peptide P639 and application thereof |
| KR20180089522A (en) * | 2015-12-22 | 2018-08-08 | 이매틱스 바이오테크놀로지스 게엠베하 | Combinations of peptides and peptides for use in immunotherapy for breast cancer and other cancers |
| KR20210052924A (en) * | 2019-11-01 | 2021-05-11 | 한국과학기술원 | Small lipid nanoparticle and cancer vaccine comprising the same |
-
2024
- 2024-04-25 WO PCT/KR2024/005587 patent/WO2024225764A1/en active Pending
- 2024-04-25 KR KR1020240055414A patent/KR20240158819A/en active Pending
Patent Citations (5)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US20080058272A1 (en) * | 2006-08-29 | 2008-03-06 | Juergen Becker | Nonamer Peptides for Cancer Treatment |
| KR20160106192A (en) * | 2010-03-19 | 2016-09-09 | 이매틱스 바이오테크놀로지스 게엠베하 | Novel immunotherapy against several tumors including gastrointestinal and gastric cancer |
| KR20180089522A (en) * | 2015-12-22 | 2018-08-08 | 이매틱스 바이오테크놀로지스 게엠베하 | Combinations of peptides and peptides for use in immunotherapy for breast cancer and other cancers |
| CN105949302A (en) * | 2016-05-27 | 2016-09-21 | 郑州大学 | FAP(fibroblast activation protein)-sourced anti-tumor CTL (cytotoxic T lymphocyte) epitope peptide P639 and application thereof |
| KR20210052924A (en) * | 2019-11-01 | 2021-05-11 | 한국과학기술원 | Small lipid nanoparticle and cancer vaccine comprising the same |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20240158819A (en) | 2024-11-05 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| JP6548579B2 (en) | Conjugate compound | |
| EP3852727A1 (en) | Therapeutic nanoparticles and methods of use thereof | |
| WO2019156365A1 (en) | Peptide nucleic acid complex having endosomal escape capacity, and use thereof | |
| WO2019066535A1 (en) | Novel recombinant plasma membrane-based vesicle, for treating cancer | |
| Chang et al. | Development of Th1-mediated CD8+ effector T cells by vaccination with epitope peptides encapsulated in pH-sensitive liposomes | |
| EP3440096A1 (en) | A peptide with ability to penetrate cell membrane | |
| US20170087232A1 (en) | Medicament for use in a method of inducing or extending a cellular cytotoxic immune response | |
| WO2022191555A1 (en) | Composition for in vivo delivery of rna and preparation method therefor | |
| WO2022191377A1 (en) | Vaccine composition for preventing sars-cov-2 | |
| CN114466659A (en) | Immunotherapeutic compositions | |
| WO2017003267A1 (en) | Peptide having anti-viral effect and composition containing same | |
| WO2024225764A1 (en) | Immunodominant epitope peptides of fibroblast activating proteins, and uses thereof | |
| WO2023229446A1 (en) | Method for constructing gram-negative bacterial outer membrane-derived nanovesicles and use thereof | |
| EP2368899B1 (en) | Avian influenza vaccine | |
| EP2519263B1 (en) | Lipopeptide- and lipoprotein-conjugates and its use | |
| WO2021085696A1 (en) | Small lipid nanoparticles, and cancer vaccine including same | |
| US20150343089A1 (en) | Hepatitis c virus liposome vaccine | |
| WO2015002512A1 (en) | Dengue virus-specific sirna, double helix oligo-rna structure comprising sirna, and composition for suppressing proliferation of dengue virus comprising rna structure | |
| WO2025159411A1 (en) | Nanoparticle comprising cell-penetrating peptides conjugated with deoxycholic acid and use thereof | |
| WO2023003380A1 (en) | Novel cell-penetrating peptide and use thereof | |
| WO2023075421A1 (en) | Novel peptide targeting dendritic cells, and composition for treating cancer comprising same | |
| WO2022034946A1 (en) | Immunomodulatory protein-sirna complex having anticancer activity | |
| WO2024195922A1 (en) | Novel ionizable lipid and lipid nanoparticle composition using same | |
| WO2017176076A1 (en) | A peptide with ability to penetrate cell membrane | |
| WO2022250518A1 (en) | Vaccine for prevention or treatment of viral infection |
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: 24797419 Country of ref document: EP Kind code of ref document: A1 |