WO2025159614A1 - Nanostructure comprenant des peptides d'antigène associé au cancer conjugués à de l'acide désoxycholique et son utilisation - Google Patents
Nanostructure comprenant des peptides d'antigène associé au cancer conjugués à de l'acide désoxycholique et son utilisationInfo
- Publication number
- WO2025159614A1 WO2025159614A1 PCT/KR2025/099093 KR2025099093W WO2025159614A1 WO 2025159614 A1 WO2025159614 A1 WO 2025159614A1 KR 2025099093 W KR2025099093 W KR 2025099093W WO 2025159614 A1 WO2025159614 A1 WO 2025159614A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- phosphatidylethanolamine
- antigen peptide
- cancer antigen
- nanostructure
- 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
Classifications
-
- 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
- A61K39/39—Medicinal preparations containing antigens or antibodies characterised by the immunostimulating additives, e.g. chemical adjuvants
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K45/00—Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
- A61K45/06—Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/51—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
- A61K47/54—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K47/00—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
- A61K47/50—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
- A61K47/69—Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the conjugate being characterised by physical or galenical forms, e.g. emulsion, particle, inclusion complex, stent or kit
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K9/00—Medicinal preparations characterised by special physical form
- A61K9/48—Preparations in capsules, e.g. of gelatin, of chocolate
- A61K9/50—Microcapsules having a gas, liquid or semi-solid filling; Solid microparticles or pellets surrounded by a distinct coating layer, e.g. coated microspheres, coated drug crystals
- A61K9/51—Nanocapsules; Nanoparticles
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61P—SPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
- A61P35/00—Antineoplastic agents
- A61P35/04—Antineoplastic agents specific for metastasis
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/435—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
- C07K14/46—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates
- C07K14/47—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from vertebrates from mammals
Definitions
- One example of the present invention relates to a cancer antigen peptide nanostructure comprising a cancer antigen peptide assembly linked to deoxycholic acid; a method for preparing the same; and a use thereof.
- Immunotherapy is a strategy that utilizes the body's immune system to target and eliminate malignant cells in the fight against cancer.
- DCs dendritic cells
- Antigens presented by DCs are recognized by the immune system, and during this process, DCs migrate to lymph nodes and interact with immature T cells. DCs effectively control the immune response by regulating T cell activation.
- TAAs tumor-associated antigens
- the present invention aims to provide a cancer antigen peptide nanostructure comprising a cancer antigen peptide assembly linked to deoxycholic acid; and a lipid coating the same.
- the present invention comprises a step of linking a cancer antigen peptide with deoxycholic acid
- the purpose of the present invention is to provide a method for manufacturing a cancer antigen peptide nanostructure, comprising a step of coating the above assembly with lipid.
- the present invention aims to provide a vaccine composition for preventing or treating cancer, comprising the cancer antigen peptide nanostructure.
- the present invention aims to provide a method for preventing or treating cancer using the vaccine composition.
- the present invention aims to provide a use of the vaccine composition for preventing or treating cancer.
- the present invention provides a cancer antigen peptide nanostructure comprising a cancer antigen peptide assembly linked to deoxycholic acid; and a lipid coating the same.
- the cancer antigen peptide may be survivin.
- the cancer antigen peptide may be an MHC class I binding epitope of survivin.
- the cancer antigen peptide may be composed of an amino acid sequence represented by SEQ ID NO: 1.
- the lipid is selected from the group consisting of lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebroside, dicetylphosphate, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyloleoleoleoyl
- an immunomodulator may be additionally loaded onto the cancer antigen peptide nanostructure.
- the immunomodulator may be at least one selected from the group consisting of SD-208, Vactosertib, Galunisertib, LY3200882, Resiquimod, Imiquimod, Gardiqiomod, Motolimod, Alum (Aluminium salts), CpG ODNs, GM-CSF, IL-12, poly(I:C), MPL, AS01, IC31, and CFA01.
- the nanostructure may increase the expression of one or more selected from the group consisting of CD80/CD86, MHC I/II, CCR7, TNF- ⁇ , IL-6, and IL-12p70 in dendritic cells.
- the nanostructure may induce intratumoral infiltration of CD8 + T cells.
- the present invention comprises a step of linking a cancer antigen peptide with deoxycholic acid
- a method for producing a cancer antigen peptide nanostructure comprising a step of coating the above assembly with lipid.
- the cancer antigen peptide may be survivin.
- the cancer antigen peptide may be an MHC class I binding epitope of survivin.
- the cancer antigen peptide may be composed of an amino acid sequence represented by SEQ ID NO: 1.
- the lipid is selected from the group consisting of lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebroside, dicetylphosphate, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyloleoleoleoyl
- an immunomodulator may be additionally loaded into the assembly.
- the immunomodulator may be at least one selected from the group consisting of SD-208, Vactosertib, Galunisertib, LY3200882, Resiquimod, Imiquimod, Gardiqiomod, Motolimod, Alum (Aluminium salts), CpG ODNs, GM-CSF, IL-12, poly(I:C), MPL, AS01, IC31, and CFA01.
- the present invention provides a vaccine composition for preventing or treating cancer, comprising the cancer antigen peptide nanostructure.
- the cancer may be at least one selected from the group consisting of bladder cancer, brain cancer, breast cancer, cervical cancer, ovarian cancer, colon cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer (gastric cancer), head and neck cancer, testicular cancer, melanoma, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, T-cell lymphocytic leukemia, and B-cell lymphoma, and uterine cancer.
- the present invention provides a method for preventing or treating cancer using the vaccine composition.
- the present invention provides a use of the vaccine composition for preventing or treating cancer.
- the inventors of the present invention confirmed that the nanostructure of the present invention effectively stimulates dendritic cell maturation, lymph node migration, and T cell activation. Furthermore, in a cancer cell model, we observed a significant influx of cytotoxic T lymphocytes into the primary tumor, confirming an anti-metastatic effect. Therefore, the nanostructure of the present invention can be usefully utilized as a promising immunotherapy platform applicable to various intractable cancers.
- Figures 1a to 1e illustrate the manufacturing method and mechanism of action of DA-L-DSA.
- Figure 1a illustrates the synthetic procedure of a lipid-coated deoxycholic acid-survivin assembly (DA-L-DSA) loaded with dual adjuvants.
- Figure 1b shows that subcutaneously injected DA-L-DSA is effectively delivered to dendritic cells, inducing their maturation and migration to lymph nodes, where it activates na ⁇ ve T cells and induces antigen-specific killing effects.
- Figure 1c shows the chemical structure of the DS conjugate.
- Figure 1d shows the results of measuring the CMC of the DS conjugate.
- Figure 1e shows the hydrodynamic size and zeta potential of L-DSA.
- Figures 2a to 2e show the physicochemical properties of DA-L-DSA.
- Figure 2a shows the encapsulation efficiency and drug loading degree of SD-208.
- Figure 2b shows the encapsulation efficiency and drug loading degree of R848.
- Figure 2c shows the improvement of drug encapsulation efficiency through lipid coating.
- Figure 2d shows the morphology of DSA, L-DSA, and DA-L-DSA observed using a transmission electron microscope.
- Figure 2e shows the results of measuring the drug release profile of DA-L-DSA using a UV-vis spectrophotometer.
- Figures 3a to 3d show in vitro evaluation of bone marrow-derived dendritic cell (BMDC) maturation using DA-L-DSA.
- Figure 3a shows the cellular uptake efficiency of DSA and L-DSA into BMDCs.
- Figure 3b shows the results of a Western blot analysis for BMDC activation.
- Figure 3c shows the results of a flow cytometry analysis for BMDC activation.
- Figure 3d shows the results of an ELISA analysis.
- Figures 4a to 4d show the intracellular and intercellular delivery results of subcutaneously injected L-DSA in vivo .
- Figures 4a and 4b show the results 24 hours after injection of Cy5.5-labeled L-DSA.
- Figure 4a shows a fluorescence image of an ex vivo organ, and
- Figure 4b shows the quantitative results of the fluorescence signal.
- Figure 4c is an immunofluorescence image showing the distribution of dendritic cells in an inguinal lymph node.
- Figure 4d shows the distribution of dendritic cells in the lymph node confirmed by flow cytometry.
- Figures 5a to 5k show the antitumor immune response induced by DA-L-DSA in a survivin-expressing melanoma model.
- Figure 5a schematically shows the treatment schedule and average tumor growth profiles to verify the antitumor efficacy of DA-L-DSA.
- Figures 5c to 5d show flow cytometric analysis of dendritic cell maturation in inguinal lymph nodes.
- Figure 5e shows the quantification of IL-12p70 in lymph nodes by ELISA.
- Figures 5f to 5h show flow cytometric analysis of CD3 + tumor-infiltrating lymphocytes and CD8 + cytotoxic T cell recruitment in primary tumors.
- Figures 5i to 5k show flow cytometric analysis of regulatory T cells in primary tumors.
- Figures 6a to 6d demonstrate the antitumor and antimetastatic efficacy of DA-L-DSA in a spontaneous metastatic breast cancer model.
- Figure 6a presents a schematic of the treatment schedule to verify the antitumor and antimetastatic efficacy of DA-L-DSA.
- Figure 6b shows the average tumor growth profile and the growth profile of individual tumors.
- Figure 6c shows lung images and H&E staining images.
- Figure 6d presents the results comparing the number of metastatic nodules.
- Figures 7a to 7g demonstrate the synergistic antitumor efficacy of DA-L-DSA and immune checkpoint inhibitors.
- Figure 7a shows a schematic diagram of the treatment schedule to confirm the synergistic effect of DA-L-DSA and conventional immune checkpoint blockade, as well as the average tumor growth profile.
- Figure 7c shows an immunofluorescence image of intratumoral granzyme B + cells.
- Figure 7d shows the results of quantifying granzyme B+ cells.
- Figure 7e shows a flow cytometry image of a primary tumor to confirm T cell activation in the tumor microenvironment.
- Figure 7f shows a TUNEL assay image.
- Figure 7g shows the results of quantifying TUNEL + cells.
- the present invention provides a cancer antigen peptide nanostructure comprising a cancer antigen peptide assembly linked to deoxycholic acid; and a lipid coating the same.
- the term "assembly” refers to a structure formed by self-assembly of cancer antigen peptides linked to deoxycholic acid.
- the assembly is stably formed through hydrophobic bonds and intermolecular interactions, enhancing the structural stability of the cancer antigen peptides and enabling efficient antigen delivery.
- nanostructure refers to a nano-scale structure manufactured using biocompatible materials, which can be utilized for drug delivery, vaccine delivery, and the development of immunotherapeutic agents.
- the inventors of the present invention constructed a peptide containing an MHC class I binding epitope sequence derived from a tumor-associated antigen called survivin, and linked this peptide to deoxycholic acid to produce a single monomer.
- coating means covering the outer surface of an object with a thin film.
- deoxycholic acid refers to a hydrophobic molecule that can regulate the function of dendritic cells through the TGR5-cAMP-PKA pathway. It is linked to a peptide to increase the binding affinity of the nanostructure to the lipid membrane, while simultaneously contributing to maintaining a stable structure. Activation of TGR5 by deoxycholic acid suppresses excessive dendritic cell activation, thereby reducing excessive inflammatory responses. This suggests that deoxycholic acid may help regulate immune responses, and in particular, may be useful in preventing excessive inflammation, such as cytokine release syndrome, during cancer treatment by making dendritic cells more tolerant.
- cancer antigen peptide refers to a specific peptide sequence among the amino acid sequences of a tumor-specific or tumor-associated antigen that can induce an immune response.
- the peptide induces an immune response by binding to MHC class I or MHC class II molecules and presenting the antigen to T cells from antigen-presenting cells (APCs).
- APCs antigen-presenting cells
- Cancer antigen peptides can be derived from tumor-specific antigens (TSAs), which are generally overexpressed in cancer cells or created by mutations, or tumor-associated antigens (TAAs), which are also present in normal tissues but whose expression levels are increased in cancer cells.
- TSAs tumor-specific antigens
- TAAs tumor-associated antigens
- the above cancer antigen peptide can induce a cytotoxic T lymphocyte (CTL) response by CD8 + T cells presented by MHC class I, or can play an immune-assisting role through CD4+ T cells presented by MHC class II.
- CTL cytotoxic T lymphocyte
- the cancer antigen peptide may be survivin.
- the cancer antigen peptide may be an MHC class I binding epitope of survivin.
- survivin refers to a member of the anti-apoptotic protein family, also known as baculoviral inhibitor of apoptosis repeat-containing 5 (BIRC5). Survivin expression is involved in regulating functions such as survival, growth, division, and angiogenesis. It is overexpressed in various cancers, including melanoma and triple-negative breast cancer, but is rarely expressed in normal cells.
- BIRC5 baculoviral inhibitor of apoptosis repeat-containing 5
- epitope also known as an antigenic determinant, refers to a single region of an antigen to which an antibody or T-cell receptor (TCR) specifically binds.
- Epitopes are key elements of the immune response and are determined by a specific amino acid sequence of an antigenic protein or peptide. Epitopes serve as targets for antibodies or T cells in the immune response, and thus can be utilized in the development of vaccines, antibody therapeutics, and immunotherapies. Epitopes for tumor antigens, in particular, are crucial for inducing cancer-specific immune responses.
- Epitopes can be derived from tumor-specific antigens (TSAs) or tumor-associated antigens (TAAs).
- TSAs tumor-specific antigens
- TAAs tumor-associated antigens
- Epitopes are not limited in type but can be divided into B-cell epitopes and T-cell epitopes.
- B-cell epitopes are three-dimensional structural regions of an antigen that can be directly recognized and bound by antibodies, while T-cell epitopes are peptides presented by MHC molecules after the antigen is degraded and recognized by the T-cell receptor (TCR).
- the present inventors designed a cancer therapeutic vaccine that delivers the survivin peptide epitope (66-74) as a TAA-derived peptide capable of binding to MHC class I of dendritic cells.
- a method for delivering R848 (resiquimod) and SD-208 as adjuvants together with the antigen to promote DC maturation and enhance antigen presentation was devised.
- the cancer antigen peptide may be composed of an amino acid sequence represented by SEQ ID NO: 1.
- the peptide used herein may include not only the peptide but also derivatives thereof.
- the peptide of the present invention may exhibit at least 80% homology with the peptide of each corresponding sequence number, preferably at least 90%, more preferably at least 95% homology
- the derivative may include a peptide in which the N-terminus, C-terminus, etc. of the peptide is chemically modified or an amino acid is added, substituted, or deleted, and is not particularly limited thereto.
- lipid refers to a membrane-forming component used to coat cancer antigen peptide assemblies or to impart structural stability to nanostructures. These lipids are amphiphilic molecules possessing both hydrophilic (head) and hydrophobic (tail) portions, and their physicochemical properties aid in the self-assembly and stabilization of nanostructures.
- the lipid is selected from the group consisting of lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebroside, dicetylphosphate, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyloleoleoleoyl
- an immunomodulator may be additionally loaded onto the cancer antigen peptide nanostructure.
- bearing means mounted in any internal location.
- immunomodulator refers to a substance that affects the biological interaction between immune cells and cancer cells, and is a concept that includes immunosuppressants and immunopotentiators.
- the immunomodulator may be at least one selected from the group consisting of SD-208, Vactosertib, Galunisertib, LY3200882, Resiquimod, Imiquimod, Gardiqiomod, Motolimod, Alum (Aluminium salts), CpG ODNs, GM-CSF, IL-12, poly(I:C), MPL, AS01, IC31, and CFA01.
- the type of the immunosuppressant there is no limitation on the type of the immunosuppressant, and for example, it may be an immune checkpoint inhibitor that inhibits immune checkpoint proteins such as PD1, PDL-1, PD-L2, CTLA-4, LAG-3, BTLA, B7H3, B7H4, 4-1BB (CD137), TIM3, KIR, etc.
- an immune checkpoint inhibitor that inhibits immune checkpoint proteins such as PD1, PDL-1, PD-L2, CTLA-4, LAG-3, BTLA, B7H3, B7H4, 4-1BB (CD137), TIM3, KIR, etc.
- the type of the immunostimulant there is no limitation on the type of the immunostimulant, but it may be, for example, an immunostimulant such as monophosphoryl lipid A (MPLA), aluminum salt (alum), or CpG oligodeoxynucleotide (ODN).
- MPLA monophosphoryl lipid A
- alum aluminum salt
- ODN CpG oligodeoxynucleotide
- adjuvant refers to a substance that induces an anticancer effect by increasing a non-specific immune response to an antigen.
- the type of adjuvant is not limited, but examples thereof include inhibitors of TLR7, TLR8, and TGF- ⁇ (transforming growth factor- ⁇ receptor 1), preferably R848 or SD-208.
- R848 refers to a compound with immunomodulatory and antiviral properties that interacts with Toll-like receptor 7 (TLR7) and Toll-like receptor 8 (TLR8). These Toll-like receptors are pattern recognition receptors that detect pathogens or abnormalities in the external environment and regulate the immune response. In DCs, R848 activates TLR7 and TLR8, enhancing antigen expression and presentation.
- TLRs Toll-like receptors
- DCs dendritic cells
- the above R848 can enhance dendritic cell maturation and activation, antigen presentation, and T cell activation by activating TLR7 and TLR8. Accordingly, the above R848 can be used as an immunomodulatory agent that enhances the immune response.
- the TLR7/8 agonist such as R848 is not limited in type, but may be at least one selected from the group consisting of synthetic compounds, natural products, peptides, antibodies, aptamers, siRNA, shRNA, miRNA, ribozymes, DNAzymes, PNA (peptide nucleic acids), antisense oligonucleotides, and other TLR7/8 activators.
- TLR7/8 agonist enhances the antigen expression and immune regulatory function of DCs by activating the signal transduction pathways of TLR7 and TLR8, which can be usefully utilized in antiviral and anticancer immune responses.
- the above TLR7/8 agonist may be at least one selected from the group consisting of R848 (Resiquimod), Imiquimod, CL097, Gardiquimod, and other toll-like receptor 7/8 activators. More specifically, it may be R848 (Resiquimod).
- SD-208 is a kinase inhibitor targeting the TGF- ⁇ type 1 receptor (TGF- ⁇ R1), which blocks the downstream mechanisms that occur when TGF- ⁇ is delivered to cells. Previous studies have reported that TGF- ⁇ inhibits dendritic cell maturation, antigen presentation, and T cell activation.
- TGF- ⁇ transforming growth factor- ⁇
- TGF- ⁇ 1 TGF- ⁇ 2, and TGF- ⁇ 3, which are pleiotropic regulatory factors for immune responses, inflammatory responses, cell growth, and cell differentiation.
- TGF- ⁇ is a representative immunosuppressive factor for tumor sites and is known to induce epithelial-mesenchymal transition (EMT) in cancer cells.
- EMT epithelial-mesenchymal transition
- the TGF- ⁇ inhibitor is not limited in type as long as it inhibits the activity of the TGF- ⁇ protein or the expression of the protein or mRNA, but may be at least one selected from the group consisting of compounds, natural products, antibodies, aptamers, siRNA, shRNA, miRNA, ribozymes, DNAzymes, PNA (peptide nucleic acids), antisense oligonucleotides, and peptides that specifically bind to the TGF- ⁇ .
- the above TGF- ⁇ inhibitor may be a TGF- ⁇ receptor inhibitor.
- the TGF- ⁇ receptor inhibitor specifically binds to the TGF- ⁇ receptor and inhibits the TGF- ⁇ signaling pathway through the receptor.
- the above TGF- ⁇ receptor inhibitor is not limited in type as long as it inhibits the activity of the TGF- ⁇ receptor protein or the expression of the protein or mRNA, but may be at least one selected from the group consisting of compounds, natural products, antibodies, aptamers, siRNA, shRNA, miRNA, ribozymes, DNAzymes, PNA (peptide nucleic acids), antisense oligonucleotides, and peptides that specifically bind to the TGF- ⁇ receptor.
- TGF- ⁇ receptor inhibitor may be a TGF- ⁇ type 1 receptor (TGF- ⁇ R1) inhibitor (e.g., a TGF- ⁇ type 1 receptor kinase inhibitor), specifically, SD-208 (2-(5-chloro-2-fluorophenyl)pteridin-4-yl]pyridin-4-yl-amine), SB431542 (4-[4-(1,3-benzodioxol-5-yl)-5-(2-pyridinyl)-1H-imidazol-2-yl]benzamide), LY2109761 (4-(2-((4-(2-(pyridin-2-yl)-5,6-dihydro-4H-pyrrolo[1,2-b]pyrazol-3-yl)quinolin-7-yl)oxy)ethyl)morpholine), It may be at least one selected from the group consisting of IN-1130 (3-[[5-(6-methylpyridin-2-yl)-4-quinoxalin-6-yl
- the nanostructure may increase the expression of one or more selected from the group consisting of CD80/CD86, MHC I/II, CCR7, TNF- ⁇ , IL-6, and IL-12p70 in dendritic cells.
- the nanostructure may induce intratumoral infiltration of CD8 + T cells.
- the present invention comprises a step of linking a cancer antigen peptide with deoxycholic acid
- a method for producing a cancer antigen peptide nanostructure comprising a step of coating the above assembly with lipid.
- cancer antigen peptide deoxycholic acid, assembly, lipid, nanostructure, etc. are as described above.
- the cancer antigen peptide may be survivin.
- the cancer antigen peptide may be an MHC class I binding epitope of survivin.
- the cancer antigen peptide may be composed of an amino acid sequence represented by SEQ ID NO: 1.
- the lipid is selected from the group consisting of lecithin, phosphatidylethanolamine, lysolecithin, lysophosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, sphingomyelin, egg sphingomyelin (ESM), cephalin, cardiolipin, phosphatidic acid, cerebroside, dicetylphosphate, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoylphosphatidylethanolamine (DOPE), palmitoyloleoyl-phosphatidylcholine (POPC), palmitoyloleoleoleoyl
- an immunomodulator may be additionally loaded into the assembly.
- the immunomodulator may be at least one selected from the group consisting of SD-208, Vactosertib, Galunisertib, LY3200882, Resiquimod, Imiquimod, Gardiqiomod, Motolimod, Alum (Aluminium salts), CpG ODNs, GM-CSF, IL-12, poly(I:C), MPL, AS01, IC31, and CFA01.
- the present invention provides a vaccine composition for preventing or treating cancer, comprising the cancer antigen peptide nanostructure.
- cancer refers to a class of diseases characterized by the development of abnormal cells that multiply uncontrollably and have the ability to invade and destroy normal body tissues.
- prevention may mean any act of suppressing or delaying the onset of cancer in an individual by administering a pharmaceutical composition according to one aspect.
- treatment may mean any action that improves or beneficially changes the symptoms of cancer in an individual by administering a pharmaceutical composition according to one aspect.
- the cancer may be at least one selected from the group consisting of bladder cancer, brain cancer, breast cancer, cervical cancer, ovarian cancer, colon cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer (gastric cancer), head and neck cancer, testicular cancer, melanoma, acute myeloid leukemia, chronic myeloid leukemia, chronic lymphocytic leukemia, T-cell lymphocytic leukemia, and B-cell lymphoma, and uterine cancer.
- the above pharmaceutical composition may be provided as a pharmaceutical composition containing the active ingredient alone or including one or more pharmaceutically acceptable excipients or diluents.
- the above pharmaceutical composition when formulated, it can be prepared using diluents or excipients such as lubricants, sweeteners, flavoring agents, emulsifiers, suspending agents, preservatives, fillers, bulking agents, binders, wetting agents, disintegrants, and surfactants that are commonly used.
- Solid preparations for oral administration may include tablets, pills, powders, granules, capsules, etc., and such solid preparations can be prepared by mixing at least one excipient, such as starch, calcium carbonate, sucrose or lactose, gelatin, etc., with the composition.
- lubricants such as magnesium stearate and talc can also be used.
- Liquid preparations for oral administration include suspensions, solutions, emulsions, and syrups.
- simple diluents such as water and liquid paraffin, they may contain various excipients such as wetting agents, sweeteners, fragrances, and preservatives.
- Preparations for parenteral administration may include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, and suppositories.
- Non-aqueous solvents and suspensions may include propylene glycol, polyethylene glycol, vegetable oils such as olive oil, and injectable esters such as ethyl oleate.
- Suppositories may include witepsol, macrogol, tween 61, cacao butter, laurin butter, and glycerogelatin.
- known diluents or excipients may be used.
- the above pharmaceutical composition is administered in a pharmaceutically effective amount.
- pharmaceutically effective amount means an amount sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment.
- the effective dosage level may be determined based on the type and severity of the patient's disease, the activity and sensitivity of the drug, the time of administration, the route of administration, and the excretion rate, the duration of treatment, concomitant medications, and other factors well known in the medical field.
- the dosage may vary depending on the patient's condition and weight, the extent of the disease, the form of the drug, the route of administration, and the time of administration, but can be appropriately selected by a person skilled in the art.
- the nanostructure of the present invention can be administered alone or in combination with other therapeutic agents, and in the case of combination administration, administration can be sequential or simultaneous.
- administration can be sequential or simultaneous.
- other therapeutic agents for example, amino acids, vaccines, antiviral agents, gene transfer vectors, immune checkpoint inhibitors, immune enhancers, immunomodulators, interleukin inhibitors, neurotrophic factors, neuroprotective agents, antineoplastic agents, chemotherapeutic agents, polysaccharides, anticoagulants, antibiotics, analgesics, anesthetics, antihistamines, anti-inflammatory agents, viruses, Interferon beta-1a, Natalizumab, Daclizumab, Thalidomide, Glatiramer, Teriflunomide, Ocrelizumab, Ustekinumab, Fingolimod, Siponimod, Ozanimod, Dimethyl fumarate, Ponesimod, Darvadstrocel, etc. can be used.
- the nanostructure of the present invention may further comprise a formulation along with the immunomodulator.
- the formulation may be a therapeutic, prophylactic, or diagnostic agent.
- the formulation may be selected from the group consisting of peptides, proteins, carbohydrates, nucleic acid molecules, lipids, organic molecules, biologically active inorganic molecules, and combinations thereof.
- a wide range of drugs may be formulated for delivery using the present microneedle device and method.
- drug or “drug formulation” is broadly used to refer to any prophylactic, therapeutic, or diagnostic agent, or other substance suitable for introduction into biological tissue, including pharmaceutical excipients and substances for tattooing, cosmetics, and other uses.
- a drug may be a biologically active substance.
- Drug formulations may take various forms, such as liquid solutions, gels, solid particles (e.g., microparticles, nanoparticles), or combinations thereof. Drugs may include small molecules, large (i.e., macro-) molecules, or combinations thereof. Drugs may be selected from suitable proteins, peptides, and fragments thereof, which may be naturally occurring, synthetic, or recombinantly produced.
- DCA-survivin(66-74) (DS, DCA-Gly-Trp-Glu-Pro-Asp-Asp-Asn-Pro-Ile) was synthesized by GL Biochem Ltd. (Shanghai, China).
- 1,2-Dipalmitoyl- sn -glycero-3-phosphocholine (DPPC) and 1,2-distearoyl- sn -glycero-3-phosphoethanolamine-N-[amino(polyethyleneglycol)-2000] (DSPE-PEG2000) were purchased from Avanti Polar Lipids, Inc. (Alabaster, AL, USA).
- Anti-mouse CD45 antibody was purchased from BD Biosciences (Franklin Lakes, NJ, USA). ⁇ -Mercaptoethanol, Hanks' balanced salt solution (HBSS), and mouse ELISA kits (IL-12p70, IL-6, and TNF- ⁇ ) were obtained from Thermo Fisher Scientific (Waltham, MA, USA). The DeadEndTM fluorometric TUNEL system was obtained from Promega (Madison, WI, USA).
- CMC critical micelle concentration
- CMC critical micelle concentration
- a mixture of DSPE-PEG 2000 and DPPC was dissolved in chloroform at a molar ratio of 1:3 and stirred at room temperature for 30 min before evaporation.
- the prepared lipid mixture was then added to water (4% EtOH, 10 mL) at a concentration of 0.2 mg/mL -1 and stirred gently.
- DS (1 mg), R848 (0.15 mg), and SD-208 (0.15 mg) were dissolved in dichloromethane (1 mg mL -1 ).
- the premixed adjuvant/DS solution was slowly dropped into the lipid solution (1.25 mL), sonicated, and evaporated to remove the organic solvent.
- DA-L-DSA hydrodynamic size, zeta potential, and polydispersity index (PDI) of DA-L-DSA were measured using a Zetasizer-Nano ZS (Malvern Instrument, Worcestershire, UK). The morphological characteristics of DA-L-DSA were visualized by transmission electron microscopy.
- the drug loading and encapsulation efficiency of R848 and SD-208 were calculated by measuring the absorbance at 320 nm and 370 nm, respectively, using DA-L-DSA (0.25 mg). 1 mg mL1 DA-L-DSA was resuspended in PBS (with or without 10% serum), and the released media were collected by centrifugation at 2, 4, 6, 12, 24, 48, 72, and 120 h. The released media and nanoassemblies were lyophilized and resuspended, and R848 and SD-208 were detected at 320 nm and 370 nm, respectively.
- BMDCs mouse bone marrow-derived dendritic cells
- bone marrow cells were collected from the femurs and tibias of 6- to 10-week-old C57BL/6 mice (Orient Bio, Korea). Bone marrow contents were washed with HBSS. The harvested cells were washed twice with HBSS and cultured in RPMI1640 supplemented with 1% penicillin-streptomycin (100 U mL -1 ), 10% FBS, IL-4 (20 ng mL -1 ), GM-CSF (20 ng mL -1 ), and ⁇ -mercaptoethanol (55 nM) at 37°C for 6 days. On day 6, cells were sorted using a magnetic EasySep mouse CD11c positive selection kit (STEMCELL Technologies, USA).
- BMDCs Mouse bone marrow-derived dendritic cells
- BMDCs were seeded in 96-well plates and cultured at 37°C for 24 h. BMDCs were then treated with DA-L-DSA and other nanoassembly groups at concentrations of 0–20 ⁇ M for 24 h, followed by incubation with a CCK-8 solution for 30 min. Cell viability relative to the control group was then measured using a UV/Vis spectrophotometer (Tecan) at 450 nm.
- Tecan UV/Vis spectrophotometer
- BMDCs (2 ⁇ 10 5 cells mL -1 ) were incubated with Cy5.5-conjugated nanoassemblies at a concentration of 1.5 ⁇ g mL -1 for 2 h and then analyzed using a FACS Calibur (BD Biosciences, USA). Cells were stained with DAPI (Southern Biotech) and visualized using a confocal microscope (Leica).
- the membranes were incubated with secondary antibodies for 1 h at room temperature.
- Anti-rabbit HRP-linked antibody (1:2,000, Cell Signaling) was used as the secondary antibody.
- the membrane was captured using ChemiDoc XRS+ (Bio-Rad, CA, USA).
- mice C57BL/6 mice were subcutaneously administered 20 ⁇ g of DS (DCA-survivin (66-74) conjugate), and the experimental groups were divided into groups according to the drug loading (%) of DA-L-DSA.
- the mice were vaccinated three times at 5-day intervals.
- spleen cells from untreated healthy mice were harvested and treated with 1 ⁇ g of the survival peptide or PBS for approximately 4 hours.
- the surviving treated and PBS-treated spleen cells were labeled with 5 ⁇ M and 0.5 ⁇ M carboxyfluorescein succinimidyl ester (CFSE), respectively.
- Equal numbers of each spleen cell were then mixed and intravenously injected into each immunized mouse. After 18 hours, the percentages of CFSE [high] and CFSE [low] in the spleen of each mouse were detected by flow cytometry, and the antigen-specific killing ability was calculated according to the following formula:
- mice from each group were sacrificed to analyze tumor-infiltrating lymphocytes.
- Tumor tissue was collected and digested with DNase I solution and collagenase/hyaluronidase. The digested tumor tissue was filtered through a 70 ⁇ m strainer and centrifuged at 300 g for 10 minutes at room temperature.
- Red blood cells (RBCs) were lysed, and the cells were washed twice with phosphate-buffered saline (PBS). The cells were then fixed with 4% paraformaldehyde (Wako, Japan) for 15 minutes and permeabilized with cell permeabilization buffer (ThermoFisher Scientific, USA) for 10 minutes at room temperature.
- the cells were then stained with anti-mouse CD45, anti-mouse CD3, anti-mouse CD8, and anti-mouse IFN- ⁇ antibodies.
- the cells were washed twice and resuspended in ice-cold PBS supplemented with 10% fetal bovine serum (FBS) for analysis using a FACS Calibur (BD Biosciences, USA).
- Paraffin sections of the tumor tissue were prepared for immunofluorescence analysis. Paraffin sections were deparaffinized, permeabilized, and stained with anti-mouse CD3 and anti-mouse CD8 antibodies. After washing the sections twice, cell nuclei were stained with DAPI solution. Tumor cells were also stained according to the protocol using the DeadEnd ⁇ fluorescent TUNEL system to detect apoptosis. Slides were examined under a fluorescence microscope.
- FIG. 1a The fabrication process of a nano self-assembly system capable of delivering both a tumor-associated antigen (TAA) and an adjuvant is illustrated in Figure 1a.
- Deoxycholic acid composed of hydrophilic molecules and a hydrophobic cyclopentanophenanthrene nucleus, can form micelles in aqueous solutions. Therefore, when a relatively hydrophilic tumor-associated antigen (TAA) peptide binds to deoxycholic acid, it forms a micellar structure, exhibiting amphipathic properties.
- TAA tumor-associated antigen
- DA dual adjuvants
- DA-DSA deoxycholic acid-survivin
- SD-208 a transforming growth factor ⁇ receptor (TGF- ⁇ R) I kinase inhibitor, enhances dendritic cell and tumor-specific cytotoxic T cell (CTL) responses and stimulates antitumor natural killer cells.
- TGF- ⁇ R transforming growth factor ⁇ receptor
- CTL tumor-specific cytotoxic T cell
- the surface of this DA-DSA was coated with a DPPC/DSPE-PEG-based lipid membrane to complete DA-L-DSA.
- amphiphilic DS peptide monomers readily self-assembled to form core-shell peptide assemblies (DSA), which exhibited a critical micelle concentration (CMC) of 133.25 ⁇ g mL-1 in PBS (pH 7.4) (Fig. 1d).
- CMC critical micelle concentration
- the hydrodynamic size and zeta potential were measured at various weight ratios of lipid to DS. When the weight ratio of lipid to DS was 25%, the zeta potential was -13.50 ⁇ 2.40 mV and the Z-average size was 209.73 ⁇ 1.04 nm, which were increased compared to DSA at -29.4 ⁇ 1.20 mV and 177 ⁇ 0.96 nm, respectively (Fig. 1e).
- the drug loading and encapsulation efficiencies of R848 and SD-208 were measured at various weight ratios of adjuvants to L-DSA. Both adjuvants were loaded into L-DSA and detected at different wavelengths. As a result, it was found that 15% of adjuvant to L-DSA was optimal for maximum loading of both adjuvants (Figs. 2a and 2b).
- the drug encapsulation efficiency was further improved by coating the DSA surface with lipids.
- DA-L-DSA showed 1.8- and 3.8-fold higher encapsulation efficiencies than R848 and SD-208, respectively (Fig. 2c).
- Transmission electron microscopy (TEM) images confirmed the presence of a lipid layer on the surface of L-DSA compared to DSA. In addition, it was observed that the spherical shape was maintained even when the adjuvant was loaded (Fig. 2d).
- R848 and SD-208 were monitored in PBS (pH 7.4) and PBS containing 10% serum, with fresh media replaced at each time point.
- DA-L-DSA exhibited a slightly faster release in serum-containing media, demonstrating a sustained release pattern of both adjuvants without an initial burst.
- R848 and SD-208 were released from DA-L-DSA by 57.19 ⁇ 7.57% and 62.52 ⁇ 8.71%, respectively, after 120 h at 37°C in serum-containing media (Fig. 2e).
- the in vitro cytotoxicity of L-DSA and DA-L-DSA was measured using the CCK-8 assay using bone marrow-derived dendritic cells (BMDCs) cultured at various concentrations. Neither L-DSA nor DA-L-DSA exhibited cytotoxicity at concentrations up to 100 ⁇ M. Therefore, the treatment concentration was set at 100 ⁇ M for in vitro experiments.
- BMDCs were treated with DSA and L-DSA encapsulated with Cy5.5. Flow cytometry analysis revealed that the MFI value of L-DSA was 3.03-fold higher than that of DSA (Fig. 3a).
- BMDCs were treated with DA-L-DSA for 24 hours, and Western blot and immunohistochemical analyses were performed.
- CD80/86 expression in the DA-L-DSA-treated group was similar to that in the LPS-treated group used as a positive control (Fig. 3b).
- flow cytometry analysis confirmed that the expression of CD80, CD86, CD40, MHC I, MHC II, and CCR7 was significantly higher in the DA-L-DSA-treated group compared to the untreated group, the group treated with only the adjuvant, and the L-DSA-treated group (Fig. 3c).
- the DA-L-DSA treatment group showed the highest levels of IL-12p70, IL-6, and TNF- ⁇ release, which are known to activate na ⁇ ve T cells, as measured by ELISA, and was almost the same level as the positive control group, the LPS treatment group (Fig. 3d).
- L-DSA and DSA were injected subcutaneously into mice. After 24 hours, the biodistribution of Cy5.5-labeled L-DSA and DSA was investigated in vivo. We hypothesized that the subcutaneously injected nanoassemblies would be partially degraded and some would be delivered to antigen-presenting cells. Among these cells, dendritic cells would be activated and migrate to lymph nodes. Indeed, L-DSA exhibited significantly higher lymph node localization than DSA ( Figures 4a and 4b).
- DA-L-DSA was administered subcutaneously to mice on days 7 and 10 after tumor inoculation. All groups were sacrificed on day 16. Results showed that the primary tumor size in the DA-L-DSA-treated group was 3.52 times smaller than that in the untreated group ( Figures 5a and 5b).
- lymph nodes were collected from mice, and flow cytometry and ELISA were performed to determine the degree of maturation and activation of dendritic cells distributed within the lymph nodes.
- the flow cytometry results showed that the number of activated dendritic cells in the lymph nodes was 4.64 times higher in the dual adjuvant-treated group, 5.79 times higher in the DA-L-DSA-treated group, and 16.84 times higher in the DA-L-DSA-treated group than in the untreated group (Fig. 5c and 5d).
- IL-12p70 a T cell activation indicator derived from activated dendritic cells
- Fig. 5e primary tumors were collected from mice for flow cytometry and immunohistochemical analyses to determine the degree of T cell infiltration and immune regulation.
- Flow cytometry analysis confirmed immune activation due to an intratumoral increase in tumor-infiltrating lymphocytes (TILs) (Fig. 5g).
- TILs tumor-infiltrating lymphocytes
- DA-L-DSA increased the recruitment of CD8 + T cells into the tumor.
- the proportion of regulatory T cells (Treg) in the tumors of the DA-L-DSA-treated group was 3.33 times lower than in the untreated group (Figs. 5i and 5j).
- the proportion of CD8 + T cells and Tregs in the tumor microenvironment was significantly higher in the DA-L-DSA group than in the other groups (Fig. 5k).
- a metastatic breast cancer model was established by injecting 4T1 cells into the fourth mammary gland of Balb/c mice. After cancer cell inoculation, the cancer therapeutic vaccine DA-L-DSA was administered three times at three-day intervals, starting on day 6. Tumor tissue was surgically removed on day 16, and the lungs were examined on day 30 to assess the extent of metastasis (Fig. 6a).
- the primary tumor size in the DA-L-DSA-treated group in the metastatic breast cancer model was 3.60 times smaller than that in the untreated group (Fig. 6b). Furthermore, the anti-metastatic effect of DA-L-DSA treatment was also confirmed through the experiment. While the untreated group had an average of 56.75 metastatic nodules, the DA-L-DSA-treated group had 7.5 and 7.57 times fewer nodules, respectively (Figs. 6c and 6d).
- DA-L-DSA As DA-L-DSA administration has been shown to increase the influx of cytotoxic T cells and decrease Tregs in various cancers expressing survivin, we further investigated whether DA-L-DSA treatment could improve the response rate of immune checkpoint inhibitors by overcoming the limited response rate of existing immune checkpoint inhibitors.
- anti-PD-1 antibodies were administered every 3 days starting on day 4 after tumor inoculation in an allograft mouse melanoma model, followed by two doses of DA-L-DSA (Fig. 7a). As a result, the combination therapy of DA-L-DSA and anti-PD-1 antibodies barely inhibited tumor growth (Fig. 7b).
- DA-L-DSA/anti-PD-1 combination treatment group significantly produced granzyme B, indicating the activation of cytotoxic T cells within the tumor (Figs. 7c and 7d).
- Flow cytometry analysis results also showed that the DA-L-DSA/anti-PD-1 combination treatment group induced 1.98 times more IFN- ⁇ + CD8 + T cells than the anti-PD-1 monotherapy group, and the DA-L-DSA monotherapy group induced 1.7 times more IFN- ⁇ T cells, respectively.
- IFN- ⁇ secretion by CD8 + T cells promotes effector responses and enhances antitumor effects (Fig. 7e). Tumor cell apoptosis was confirmed through TUNEL analysis of tumor tissues.
Landscapes
- Health & Medical Sciences (AREA)
- Chemical & Material Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Medicinal Chemistry (AREA)
- General Health & Medical Sciences (AREA)
- Veterinary Medicine (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Pharmacology & Pharmacy (AREA)
- Epidemiology (AREA)
- Organic Chemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Engineering & Computer Science (AREA)
- General Chemical & Material Sciences (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Immunology (AREA)
- Mycology (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Microbiology (AREA)
- Toxicology (AREA)
- Biophysics (AREA)
- Zoology (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Molecular Biology (AREA)
- Oncology (AREA)
- Biochemistry (AREA)
- Gastroenterology & Hepatology (AREA)
- Genetics & Genomics (AREA)
- Physics & Mathematics (AREA)
- Biomedical Technology (AREA)
- Nanotechnology (AREA)
- Optics & Photonics (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
- Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)
- Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
Abstract
Un mode de réalisation de la présente invention concerne une nanostructure de peptide d'antigène associé au cancer comprenant un ensemble de peptides d'antigène associé au cancer conjugués à de l'acide désoxycholique ; son procédé de préparation ; et son utilisation. Les inventeurs de la présente invention ont confirmé que la nanostructure de la présente invention stimule efficacement la maturation des cellules dendritiques, migre vers les ganglions lymphatiques et active les lymphocytes T. De même, dans un modèle de cellules cancéreuses, une infiltration importante de lymphocytes T cytotoxiques dans des tumeurs primaires a été observée et un effet anti-métastatique a été confirmé. Par conséquent, la nanostructure selon la présente invention peut être efficacement utilisée comme plateforme d'immunothérapie prometteuse qui peut être appliquée à divers cancers réfractaires.
Applications Claiming Priority (4)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2024-0010423 | 2024-01-23 | ||
| KR20240010423 | 2024-01-23 | ||
| KR10-2024-0191253 | 2024-12-19 | ||
| KR1020240191253A KR20250115903A (ko) | 2024-01-23 | 2024-12-19 | 데옥시콜산과 연결된 암항원 펩타이드를 포함하는 나노구조체 및 이의 용도 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025159614A1 true WO2025159614A1 (fr) | 2025-07-31 |
Family
ID=96545260
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2025/099093 Pending WO2025159614A1 (fr) | 2024-01-23 | 2025-01-21 | Nanostructure comprenant des peptides d'antigène associé au cancer conjugués à de l'acide désoxycholique et son utilisation |
Country Status (1)
| Country | Link |
|---|---|
| WO (1) | WO2025159614A1 (fr) |
Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20100094664A (ko) * | 2009-02-19 | 2010-08-27 | 한국과학기술연구원 | 암 표적성이 우수한 단백질 복합체 및 이의 제조방법 |
| US7943138B2 (en) * | 2007-07-19 | 2011-05-17 | Health Research, Inc. | Survivin peptides as cancer vaccines |
| KR20170129841A (ko) * | 2015-07-20 | 2017-11-27 | 에스티팜 주식회사 | 경구투여가 어려운 약물의 경구투여를 위한 신규한 약물 전달체 및 이의 제조방법 |
| KR20230142467A (ko) * | 2020-12-18 | 2023-10-11 | 디펜스 테라퓨틱스 인코포레이티드 | 개선된 면역 반응 및/또는 안정성을 위한 공유결합적변형 항원 |
-
2025
- 2025-01-21 WO PCT/KR2025/099093 patent/WO2025159614A1/fr active Pending
Patent Citations (4)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| US7943138B2 (en) * | 2007-07-19 | 2011-05-17 | Health Research, Inc. | Survivin peptides as cancer vaccines |
| KR20100094664A (ko) * | 2009-02-19 | 2010-08-27 | 한국과학기술연구원 | 암 표적성이 우수한 단백질 복합체 및 이의 제조방법 |
| KR20170129841A (ko) * | 2015-07-20 | 2017-11-27 | 에스티팜 주식회사 | 경구투여가 어려운 약물의 경구투여를 위한 신규한 약물 전달체 및 이의 제조방법 |
| KR20230142467A (ko) * | 2020-12-18 | 2023-10-11 | 디펜스 테라퓨틱스 인코포레이티드 | 개선된 면역 반응 및/또는 안정성을 위한 공유결합적변형 항원 |
Non-Patent Citations (1)
| Title |
|---|
| AZUAR, A. ET AL.: "Cholic Acid-based Delivery System for Vaccine Candidates against Group A Streptococcus", ACS MEDICINAL CHEMISTRY LETTERS, vol. 10, 2019, pages 1253 - 1259, XP055947553, DOI: 10.1021/acsmedchemlett.9b00239 * |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| AU2019345104B2 (en) | Therapeutic nanoparticles and methods of use thereof | |
| US20210052724A1 (en) | Method for improving the efficacy of a survivin vaccine in the treatment of cancer | |
| CN104136614B (zh) | Toll‑样受体和激动剂用于治疗癌症的用途 | |
| Shariat et al. | P5 HER2/neu-derived peptide conjugated to liposomes containing MPL adjuvant as an effective prophylactic vaccine formulation for breast cancer | |
| WO2022191555A1 (fr) | Composition d'administration in vivo d'arn et son procédé de préparation | |
| CN117180442A (zh) | 用于治疗癌症的新型的基于重组质膜的囊泡 | |
| JP6443646B2 (ja) | Peg化リン脂質を担体とするポリペプチドワクチンミセル | |
| WO2018147710A1 (fr) | Nanoémulsion comprenant une matière à base d'imidazoquinoline et son utilisation | |
| CN114466659A (zh) | 免疫治疗组合物 | |
| US20210177802A1 (en) | Pharmaceutical composition for treating tumor | |
| WO2023163541A1 (fr) | Nouveaux lipides ionisables et nanoparticules lipidiques les comprenant | |
| Kim et al. | Dual adjuvant‐loaded peptide antigen self‐assembly potentiates dendritic cell‐mediated tumor Immunotherapy | |
| WO2017213328A1 (fr) | Liposome comprenant un conjugué récepteur synthétique-phospholipide, et composition pour le transfert d'un matériau fonctionnel qui peut se lier au récepteur synthétique et qui comprend, en tant que principe actif, un ligand auquel est lié le matériau fonctionnel | |
| WO2025159614A1 (fr) | Nanostructure comprenant des peptides d'antigène associé au cancer conjugués à de l'acide désoxycholique et son utilisation | |
| WO2023136689A1 (fr) | Lipide ionisable contenant une liaison ester biodégradable et nanoparticules lipidiques le comprenant | |
| JP7496623B2 (ja) | mRNAワクチン | |
| KR20250115903A (ko) | 데옥시콜산과 연결된 암항원 펩타이드를 포함하는 나노구조체 및 이의 용도 | |
| WO2021085696A1 (fr) | Petites nanoparticules lipidiques et vaccin contre le cancer les comprenant | |
| WO2023055078A1 (fr) | Composition vaccinale anticancéreuse comprenant un peptide antigénique hsp90 et son utilisation | |
| WO2025159411A1 (fr) | Nanoparticule comprenant des peptides de pénétration cellulaire conjugués à de l'acide désoxycholique et utilisation associée | |
| US20220143161A1 (en) | Mrna vaccine | |
| WO2025089649A1 (fr) | Composition de transfert d'acides nucléiques comprenant des liposomes cationiques et son utilisation | |
| WO2025029019A1 (fr) | Agent thérapeutique à base de cellules immunitaires recombinées pour le traitement du cancer via la multiplication polyclonale des lymphocytes t infiltrant la tumeur | |
| WO2024025059A1 (fr) | Nanoparticules mucoadhésives-plga | |
| WO2024177282A1 (fr) | Lipide ionisable comprenant du sulfure, et nanoparticules lipidiques le comprenant |
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: 25745279 Country of ref document: EP Kind code of ref document: A1 |