WO2025127849A1 - Anticancer composition for co-administration with anticancer agent, comprising novel peptide as active ingredient - Google Patents
Anticancer composition for co-administration with anticancer agent, comprising novel peptide as active ingredient Download PDFInfo
- Publication number
- WO2025127849A1 WO2025127849A1 PCT/KR2024/096873 KR2024096873W WO2025127849A1 WO 2025127849 A1 WO2025127849 A1 WO 2025127849A1 KR 2024096873 W KR2024096873 W KR 2024096873W WO 2025127849 A1 WO2025127849 A1 WO 2025127849A1
- Authority
- WO
- WIPO (PCT)
- Prior art keywords
- cancer
- cells
- cell
- tumor
- antibody
- 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
- 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
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
- C07K14/70503—Immunoglobulin superfamily
- C07K14/7051—T-cell receptor (TcR)-CD3 complex
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K38/00—Medicinal preparations containing peptides
- A61K38/16—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/11—T-cells, e.g. tumour infiltrating lymphocytes [TIL] or regulatory T [Treg] cells; Lymphokine-activated killer [LAK] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/10—Cellular immunotherapy characterised by the cell type used
- A61K40/15—Natural-killer [NK] cells; Natural-killer T [NKT] cells
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/31—Chimeric antigen receptors [CAR]
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/33—Antibodies; T-cell engagers
-
- A—HUMAN NECESSITIES
- A61—MEDICAL OR VETERINARY SCIENCE; HYGIENE
- A61K—PREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
- A61K40/00—Cellular immunotherapy
- A61K40/30—Cellular immunotherapy characterised by the recombinant expression of specific molecules in the cells of the immune system
- A61K40/34—Antigenic peptides
-
- 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
- A61K47/68—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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
-
- 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
- A61K47/68—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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6801—Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
- A61K47/6803—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
- A61K47/68031—Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
-
- 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
- A61K47/68—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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
- A61K47/6835—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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
- A61K47/6849—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 antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a receptor, a cell surface antigen or a cell surface determinant
-
- 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
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K14/00—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
- C07K14/001—Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof by chemical synthesis
-
- 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
- C07K14/705—Receptors; Cell surface antigens; Cell surface determinants
-
- 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
- C07K7/08—Linear peptides containing only normal peptide links having 12 to 20 amino acids
-
- G—PHYSICS
- G01—MEASURING; TESTING
- G01N—INVESTIGATING OR ANALYSING MATERIALS BY DETERMINING THEIR CHEMICAL OR PHYSICAL PROPERTIES
- G01N33/00—Investigating or analysing materials by specific methods not covered by groups G01N1/00 - G01N31/00
- G01N33/48—Biological material, e.g. blood, urine; Haemocytometers
- G01N33/50—Chemical analysis of biological material, e.g. blood, urine; Testing involving biospecific ligand binding methods; Immunological testing
- G01N33/53—Immunoassay; Biospecific binding assay; Materials therefor
- G01N33/574—Immunoassay; Biospecific binding assay; Materials therefor for cancer
-
- C—CHEMISTRY; METALLURGY
- C07—ORGANIC CHEMISTRY
- C07K—PEPTIDES
- C07K2319/00—Fusion polypeptide
Definitions
- the present invention relates to a combination therapy of a TB511 peptide that specifically binds to CD18 and has an anticancer effect and an anticancer agent.
- anticancer immunotherapy is a method of treating cancer using the body's immune system, which can minimize side effects.
- anticancer immunotherapy techniques there are cell therapy methods that activate therapeutic immune cells such as T cells (including CAR-T), dendritic cells, and natural killer cells outside the body and then directly inject them into the body, and methods for anticancer vaccines that directly activate immune cells existing in the body by injecting cancer antigens and immune-activating substances into the body, thereby increasing anticancer efficacy.
- T cells including CAR-T
- anticancer vaccines that directly activate immune cells existing in the body by injecting cancer antigens and immune-activating substances into the body, thereby increasing anticancer efficacy.
- these cell therapy agents or cancer vaccines are mainly used for diseases related to blood cancer, and most of them have a disadvantage in that their therapeutic efficacy is very low for solid cancers.
- One of the reasons for this is due to microenvironmental factors that suppress immune function around solid cancers.
- tumor microenvironment myeoloid-derived stromal cells
- Treg regulatory T cells
- TAM tumor-assocaited macrophages
- immunosuppressive cytokines and metabolites actively work, rapidly reducing the activity of immune-activating substances and therapeutic immune cells. Therefore, it is becoming important to develop a treatment that has an anticancer effect by controlling only the microenvironment around tumor cells without directly affecting tumor cells and immune cells, thereby blocking the supply of nutrients to tumor cells and angiogenesis around tumor cells.
- the tumor microenvironment is greatly considered as a treatment target because it contributes to the proliferation and survival of malignant cells, angiogenesis, metastasis, abnormal adaptive immunity, and decreased response to hormones and chemotherapeutics.
- TAMs tumor-associated macrophages
- M1 macrophages have a potent antigen-presenting ability, are generally activated by interferon- ⁇ , lipopolysaccharide (LPS), and tumor necrosis factor (TNF)- ⁇ , and exhibit proinflammatory and bactericidal effects.
- M2 macrophages are known to promote immunosuppression, tumorigenesis, and angiogenesis by releasing various extracellular matrix components, angiogenic and chemotactic factors. In general, they are induced by IL-4 and IL-13, and are distinguished from M1 type tumor-associated macrophages by expressing markers such as arginase-1, mannose (MMR, CD206), scavenger receptors (SR-A, CD204), CD163, and IL-10.
- MMR mannose
- SR-A scavenger receptors
- Tumor-associated macrophages present around tumors are closely related to the growth and metastasis of tumor cells, and it has been reported that when a large number of M2 type tumor-associated macrophages exist around tumors in cancer patients, the prognosis and survival rate of the patients are poor.
- M2 type macrophages produce cytokines such as IL-10, TGF ⁇ , and CCL18 that promote cancer growth, and receptors such as PDL1 and B7-1/2 present on the surface of M2 type tumor-associated macrophages have been reported to suppress the antitumor activity of T cells and NK cells. Since tumor growth, differentiation, and metastasis occur actively in a microenvironment where M2 type tumor-associated macrophages exist in large numbers, development of targeted therapeutics targeting M2 type tumor-associated macrophages is necessary.
- cancer cells express immune checkpoint proteins on their cell surface, which are used by normal cells to suppress immune cell activation in order to avoid the killing mechanism by immune cells.
- immune checkpoint inhibitor proteins as a method for treating cancer has been actively conducted. It has been reported in academic circles that among immune checkpoint inhibitor proteins, blocking PD-1/PD-L1 binding is particularly effective in cancer treatment and has fewer side effects than other immune checkpoint inhibitor proteins (J. Naidoo et al. (2015) Annals of Oncology, Lucia Gelao et al. (2014) Toxins, Gorge K. Philips et al (2015) International Immunology).
- the PD-1 receptor is expressed on the surface of activated immune cell types, including T cells, B cells, and natural killer (NK)/natural killer T (NKT) cells (Goodman, Patel & Kurzrock, PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas, Nature Reviews Clinical Oncology, 14:203-220, 2017.).
- PD-1 is a negative regulator of T cell activation, and the interaction of PD-1 with one of its ligands, PD-L1, on the tumor surface represents an immune checkpoint blockade that reduces the ability of activated T cells to generate effective immune responses.
- Therapeutic antibodies targeting PD-1 or PD-L1 block the ligand-receptor interaction and restore immune function to the tumor microenvironment.
- mAbs targeting PD-1 and PD-L1 are on the market, and anti-PD1 antibody treatments that inhibit the binding of PD-1 and PD-L1, such as Opdivo (nivolumab) from BMS, Keytruda (Pembrolizumab) from Merck, and Libtayo (Cemiplimab) developed by Regeneron, and anti-PD-L1 antibody treatments such as Tecentriq (Atezolizumab) from Roche, Imfinzi (Durvalumab) from AstraZeneca, and Bavencio (Avelumab) from Merck Sereno, have received US FDA approval and are revolutionizing the treatment of intractable cancers in clinical trials.
- An object of the present invention is to provide a chimeric antigen receptor comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
- T cell engager comprising a peptide of sequence number 1 or sequence number 2.
- an object of the present invention is to provide a composition for cancer diagnosis.
- the purpose of the present invention is to provide a method for preventing or treating cancer.
- the present invention provides a chimeric antigen receptor comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
- the present invention provides a recombinant vector comprising a gene encoding the chimeric antigen receptor.
- the present invention provides a chimeric antigen receptor expressing cell transformed with the recombinant vector.
- the present invention provides a T cell engager comprising a peptide of sequence number 1 or sequence number 2.
- the present invention provides a pharmaceutical composition for preventing or treating cancer, comprising a peptide of the sequence number 1 or sequence number 2, a chimeric antigen receptor, a chimeric antigen receptor expressing cell, or a T cell engager as an active ingredient.
- the present invention provides a combination for preventing or treating cancer, comprising a peptide of the above sequence number 1 or sequence number 2, a chimeric antigen receptor, a chimeric antigen receptor expressing cell, or a T cell engager; and an immunotherapy agent, a chemotherapy agent, or an antibody-drug conjugate (ADC).
- a combination for preventing or treating cancer comprising a peptide of the above sequence number 1 or sequence number 2, a chimeric antigen receptor, a chimeric antigen receptor expressing cell, or a T cell engager; and an immunotherapy agent, a chemotherapy agent, or an antibody-drug conjugate (ADC).
- ADC antibody-drug conjugate
- the present invention provides a composition for diagnosing cancer, comprising a peptide of the sequence number 1 or sequence number 2, a chimeric antigen receptor, a chimeric antigen receptor expressing cell, or a T cell engager as an effective ingredient.
- the present invention provides a method for preventing or treating cancer, comprising a step of administering the pharmaceutical composition or the combination to a subject.
- TB511 of the present invention induces apoptosis by specifically binding to CD18 of activated form of M2-Tumor-associated macrophages (M2-TAMs) that contribute to tumor progression in the tumor microenvironment (TME), and does not exhibit toxicity to other tissue-resident macrophages other than M2-TAM, and increases the infiltration of tumor killer cells and exhibits antitumor effects in various cancer models and humanized mouse models, and in particular, exhibits synergistic antitumor effects when used in combination with other immunotherapy drugs, and therefore can be utilized as an immunotherapy drug or a combination thereof.
- M2-TAMs M2-Tumor-associated macrophages
- Figures 1 to 11 are diagrams analyzing the target of TB511 and analyzing the relationship between CD18, the target of TB511, and the progression of solid tumors:
- FIG. 1 Localization analysis of TAMpep and CD18 in M2 macrophages
- Figures 8 to 11 Results of CD18 expression analysis according to macrophage subtype.
- FIGs 12 to 15 are diagrams showing the results of analysis of membrane proteins of M0, M1, and M2 macrophages binding to TAMpep.
- FIG. 12 Proteins extracted from M0 macrophages
- FIG. 13 Proteins extracted from M1 macrophages
- FIG. 14 Proteins extracted from M2 macrophages.
- Figure 15 Ratios of M2/M0 and M2/M1.
- Figures 16 and 17 are diagrams showing the results of analyzing the binding affinity of TAMpep to CD18:
- Figures 18 to 26 are diagrams analyzing the cytotoxicity induction effect through CD18 structure-specific targeting of TB511:
- Figures 20 and 21 Cytotoxicity analysis results of TB511 depending on CD18 activation
- Figure 22 Results of analysis of CD18 activation level according to tumor microenvironment induction
- Figure 23 Results of analysis of CD18 activation levels according to macrophage subtypes
- Figure 24 and Figure 25 Results of apoptosis analysis of TB511 according to macrophage subtype.
- Figure 26 Results of analysis of CD18 activation level in tumor tissue.
- Figures 27 to 34 show the analysis of internalization and endosomal escape of TB511 in M2 macrophages:
- Figure 27 Results of analysis of the distribution of TB511 in M2 macrophages
- Figure 31 Results of analysis of the localization of early endosome antigen 1 (EEA1) and TB511-Cy5 in endosomes following treatment with CPZ (chlorpromazine), M*?*CD (methyl-_-cyclodextrin), or CyD (cytochalasin D);
- Figure 32 Results of TB511-induced apoptosis analysis according to treatment with CPZ (chlorpromazine), M_CD (methyl-_-cyclodextrin) or CyD (cytochalasin D);
- Figure 33 Results of localization analysis of early endosome antigen 1 and TB511-Cy5 in endosomes according to treatment.
- Figure 34 Results of TB511-induced apoptosis analysis following Baf-A1 (bafilomycin A1) or CQ (chloroquine) treatment.
- Figures 35 to 53 are diagrams analyzing the antitumor effect of TB511:
- Figures 35-37 Antitumor effect of TB511 in a mouse model of cold tumors that are nonresponsive to immunotherapy;
- Figures 38-41 Antitumor effect of TB511 in a mouse model of immunotherapy-responsive tumors (hot tumors);
- Figures 42 to 47 Results of selective targeting analysis of TB511 on macrophage subtypes in tumor tissue.
- Figures 49 to 51 Results of macrophage count analysis in normal and tumor tissues.
- Figures 52 to 55 are diagrams analyzing the tumor targeting of TB511 through the biological distribution analysis of TB511 in a breast cancer mouse model.
- Figures 56 to 72 are diagrams analyzing immune cell changes caused by TB511 in TME:
- Figures 56 and 57 Results of analysis of changes in CD8+ T and NK cells in prostate cancer (PC) specimens;
- Figures 58 and 59 Results of analysis of the effects of CD8+ T and NK cells on the antitumor effect of TB511 in prostate cancer (PC) specimens;
- Figure 60 Process for preparing NK or CD8 deficient colon cancer mouse model
- Figures 61 to 63 Results of cell viability analysis in the control group (isotype control), CD8+ T cell depletion group (CD8 depletion), and NK cell depletion group (NK depletion) in a prostate cancer cell line injection mouse model;
- Figures 64 and 65 Analysis of the number of tumor-infiltrating CD8+ T cells and the number of exhausted CD8+ T cells in a lung cancer cells (LLC) injection mouse model;
- Figures 66 and 67 Results of analysis of the number of anti-tumor immune cells in colorectal cancer (CRC) cells;
- Figures 68 and 69 Results of CD8+ T and Granzyme B+ cell count analysis
- Figure 70 Manufacturing process of CD8 deficient colon cancer mouse model
- Fig. 71 Antitumor effect of TB511 in control mice.
- Figure 72 Antitumor effect of TB511 in a CD8 deficient colon cancer mouse model.
- Figures 73 to 76 are diagrams analyzing the number of CD8+ T and NK cells in the blood of a CD8 deficient mouse model (a) or an NK deficient mouse model (b).
- Figures 77 to 93 are diagrams evaluating the clinical potential of TB511:
- Figure 77 Process for manufacturing a humanized prostate cancer mouse model
- Figure 79 Antitumor effect of TB511 in humanized prostate cancer mice
- FIGS. 80 to 82 Analysis of human immune cells infiltrating the TME
- Figure 83 Process for manufacturing a humanized lung cancer mouse model
- Figure 84 Antitumor effect of TB511 in immunodeficient mice
- Figure 85 Antitumor effects of TB511 alone, anti-PD-1 antibody alone, and TB511 and anti-PD-1 antibody combination in humanized lung cancer mice;
- FIGS 86 to 88 Cell count analysis results of M2-TAMs (CD86+CD11b+/CD45+ cells);
- FIGS 89 to 91 Cell count analysis results of exhausted CD8+ T-cells (PD-1+CD8+/CD45+ cells).
- Figure 92 and Figure 93 CD8+ cell counts in each group determined by immunohistochemical staining in humanized non-small cell lung cancer (NSCLC) tumor tissues.
- NSCLC non-small cell lung cancer
- Figures 94 to 99 are diagrams analyzing the synergistic effect of combined administration of TB511 and an immune checkpoint inhibitor.
- Figure 94 Process for manufacturing hPD-L1 MC38 tumor-humanized PD-1 mouse model
- Figure 96 Tumor volume in the anti-PD-1 monotherapy group
- Figure 97 Tumor volume in the TB511 monotherapy group
- Figure 98 Tumor volume of the group administered with combination of TB511 and anti-PD-1 antibody.
- Figures 100 to 105 are diagrams analyzing the anticancer effect of combined administration of TB511 and oxaliplatin.
- Figure 100 Manufacturing process of colon cancer mouse model and drug administration
- Figure 101 Tumor volume in the control group
- Figure 102 Tumor volume of the control group
- Figure 103 Tumor volume in the TB511 monotherapy group
- Figure 104 Tumor volume in the oxaliplatin monotherapy group.
- Figure 105 Tumor volume of the combination therapy group of TB511 and oxaliplatin.
- Figures 106 to 108 are diagrams analyzing tumor weight and body weight changes according to combined administration of TB511 and oxaliplatin.
- Fig. 106 Size of tumor tissue
- Fig. 107 Changes in the weight of tumor tissue
- Fig. 108 Weight change of the mouse.
- FIGS 109 to 113 are diagrams analyzing the decrease in M2-like tumor associated macrophages (TAMs) in the tumor microenvironment (TME) following combined administration of TB511 and oxaliplatin.
- TAMs M2-like tumor associated macrophages
- Figures 109 and 110 Flow cytometry analysis results for macrophage populations within the TME
- Figure 111 M1-like TAMs (CD86+ F4/80+ cells within CD45+ CD11b+ cells);
- FIG. 112 M2-like TAMs (CD206+ F4/80+ cells within CD45+ CD11b+);
- Fig. 113 M1/M2 ratio.
- Figures 114 to 117 are diagrams analyzing the induction of infiltration of cytotoxic T cells into the TME by combined administration of TB511 and oxaliplatin.
- Figure 114 Flow cytometry analysis results for lymphocyte distribution within the TME
- FIG. 115 CD4 T cells (CD45+ CD3+ CD4+ cells);
- FIG. 116 CD8 T cells (CD45+ CD3+ CD8+ cells).
- Figures 150 to 153 Pie charts showing the proportion of cell types in cell clusters within tumor tissue.
- Figures 154 and 155 Cell population (%) by each cell type.
- Figures 156 to 159 illustrate the results of analysis of the effect of TB511 on the activation of CD8 T cells within a tumor microenvironment.
- Figures 156 and 157 CD8 T cell exhausted genes (white arrows) and activator genes (red arrows) in tumor tissue;
- Figures 158 and 159 CTLA4, FOXP3, LAG3, GZMB, IFNG and PDCD1 gene expression levels.
- amino acids referred to by abbreviations in the present invention are described according to the IUPAC-IUB nomenclature as follows:
- the present invention relates to a chimeric antigen receptor (CAR) comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
- CAR chimeric antigen receptor
- the peptide of SEQ ID NO: 1 or SEQ ID NO: 2 can specifically bind to M2 macrophages or M2 tumor associated macrophages (M2 TAM).
- the peptide of SEQ ID NO: 1 or SEQ ID NO: 2 can specifically bind to CD18.
- the peptide may comprise a variant thereof or an analog thereof.
- the peptide has the amino acid sequence described above, but is not limited thereto. According to a preferred embodiment of the present invention, it is preferable that the peptide has a high ratio of the amino acid of 50% or more, preferably 60% or more, more preferably 70% or more, more preferably 80% or more, more preferably 90% or more, and most preferably 100%.
- the peptide may also include additional amino acid sequences designed for a specific purpose, such as a targeting sequence, a tag, a labeled residue, a half-life or to increase the stability of the peptide.
- the peptide of the present invention may be linked to coupling partners, such as effectors, drugs, prodrugs, toxins, peptides, delivery molecules, etc.
- the peptide can be prepared in the form of a pharmaceutically acceptable salt.
- a salt can be formed by adding an acid, for example, an inorganic acid (e.g., hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, etc.), an organic carboxylic acid (e.g., acetic acid, haloacetic acids such as trifluoroacetic acid, propionic acid, maleic acid, succinic acid, malic acid, citric acid, tartaric acid, salicylic acid), and an organic sulfonic acid including an acidic sugar (glucuronic acid, galacturonic acid, gluconic acid, ascorbic acid), an acidic polysaccharide (e.g., hyaluronic acid, chondroitin sulfate, arginic acid), a sulfonic acid sugar ester such as chondroitin sulfate (e.g., methanesulfonic
- an acid for
- peptide used in the present invention refers to an amino acid polymer, which may include not only natural amino acids but also non-protein amino acids as components.
- peptide variant refers to a corresponding amino acid sequence that contains at least one amino acid difference (substitution, insertion or deletion) compared to a reference sequence.
- a “variant” has high amino acid sequence homology and/or conservative amino acid substitutions, deletions and/or insertions compared to a reference sequence.
- peptide analogue may include analogues in which the side chain or the alpha-amino acid backbone of an amino acid is substituted with one or more other functional groups.
- side chain or backbone modified peptide analogues include, but are not limited to, hydroxyproline in which the pyrrolidine ring is substituted with a hydroxy group, or N-methyl glycine "peptoids”. Types of peptide analogues are well known in the art.
- the term “conservative amino acid substitution” refers to a modification of a variant 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 peptide.
- a “conservative amino acid substitution” is a substitution that replaces 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 and defined in the art.
- 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
- amino acids having aromatic side chains e.g., tyrosine, phenylalanine, tryptophan, histidine
- Peptides according to the present invention can be prepared by standard synthetic methods, recombinant expression systems, or any other method known in the art.
- peptides according to the present invention can be synthesized by a number of methods, including, for example, methods including:
- CAR chimeric antigen receptor
- Ectodomain extracellular domain
- Ectodomain transmembrane domain
- Ectodomain intracellular domain
- transmembrane domain when the above-mentioned transmembrane domain is synthetic, it may mainly include hydrophobic amino acid residues such as leucine and valine, and for example, a triplet of phenylalanine, tryptophan and valine may be present in the synthetic transmembrane domain, but is not limited thereto. Sequence information on such transmembrane domain can be obtained from literature known in the art regarding synthetic transmembrane domains, but is not limited thereto.
- the intracellular domain is a part of the domain of the CAR existing within a cell, and is connected to the transmembrane domain.
- the intracellular domain of the present invention may include an intracellular signaling domain which is characterized by causing T cell activation, preferably T cell proliferation, when an antigen binds to the antigen binding site of the CAR.
- the intracellular signaling domain is not particularly limited in its type as long as it is a part that transmits a signal capable of causing T cell activation when an antibody binds to the antigen binding site existing outside the cell, and various types of intracellular signaling domains may be used.
- intracellular signaling domain examples include an immunoreceptor tyrosine-based activation motif or ITAM
- ITAM includes, but is not limited to, those derived from CD3 zeta ( ⁇ , zeta), FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CDS, CD22, CD79a, CD79b, CD66d or Fc ⁇ RI ⁇ .
- CARs comprise a single-chain fragment variable region (scFv) of an antibody specific for a tumor-associated antigen (TAA) that is coupled via the hinge and transmembrane regions to the cytoplasmic domain of a T-cell signaling molecule.
- Most common lymphocyte activating moieties comprise a T-cell costimulatory (e.g., CD28, CD137, OX40, ICOS, and CD27) domain in tandem with a T-cell triggering (e.g., CD3 ⁇ ) moiety.
- TAA-associated antigen e.g., CD28, CD137, OX40, ICOS, and CD27
- the present invention relates to a recombinant vector comprising a gene encoding the chimeric antigen receptor.
- the present invention relates to a chimeric antigen receptor expressing cell transformed with the recombinant vector.
- the chimeric antigen receptor expressing cell can be a chimeric antigen receptor expressing macrophage (CAR-macrophage), a chimeric antigen receptor expressing T (CAR-T) cell, a chimeric antigen receptor expressing-gamma-delta T (CAR-Gamma-delta T) cell, or a natural killer (CAR-NK) cell.
- CAR-macrophage a chimeric antigen receptor expressing macrophage
- CAR-T chimeric antigen receptor expressing T
- CAR-Gamma-delta T a chimeric antigen receptor expressing-gamma-delta T
- CAR-NK natural killer
- chimeric antigen receptor expressing T (CAR-T) cell refers to a T cell expressing a CAR.
- the chimeric antigen receptor expressing T (CAR-T) cell has the advantage of: i) recognizing a cancer antigen in a manner independent of HLA (human leukocyte antigen), and thus capable of treating cancers that evade the action of anticancer agents by reducing HLA expression on the cell surface; ii) being independent of the HLA type, and thus capable of being used for treatment regardless of the patient's HLA type; and iii) capable of producing a large amount of cancer-specific T cells in a short period of time, and thus capable of exhibiting an excellent anticancer effect.
- HLA human leukocyte antigen
- T cells include CD4 + T cells (helper T cells, TH cells), CD8 + T cells (cytotoxic T cells, CTLs), memory T cells, regulatory T cells (Treg cells), natural killer T cells, etc., and the T cells into which the CAR is introduced in the present invention are preferably CD8 + T cells, but are not limited thereto.
- the present invention relates to a T-cell engager comprising a peptide having sequence number 1 or sequence number 2.
- the T cell engager can be, for example, a bispecific T-cell engager (BiTE).
- the BiTE is a class of artificial bispecific monoclonal antibodies, which are fusion proteins consisting of amino acid sequences from four different genes or two single chain variable region fragments (scFv) of different antibodies on a single peptide chain of about 55 kilodaltons.
- scFv single chain variable region fragments
- the BiTE forms a link between the T cell and the tumor cell. It causes the T cell to exert cytotoxic activity on the tumor cell independently of the presence of MHC I or costimulatory molecules by producing proteins such as perforin and granzymes. These proteins enter the tumor cell and initiate apoptosis of the cell.
- the present invention relates to a pharmaceutical composition for preventing or treating cancer, comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2, a chimeric antigen receptor of the present invention, a cell expressing a chimeric antigen receptor of the present invention, or a T cell engager of the present invention as an active ingredient.
- the cancer can be a non-responsive or responsive cancer to an immunotherapy agent, can be a solid cancer, and is preferably, but not limited to, breast cancer, colon cancer, or pancreatic cancer.
- treatment means any act of killing cancer cells or improving or beneficially changing the symptoms of cancer by administering the composition of the present invention.
- anyone with ordinary knowledge in the technical field to which the present invention pertains will be able to refer to materials presented by the Korean Medical Association, etc. to know the exact criteria for diseases to which the composition of the present invention is effective, and to determine the degree of improvement, enhancement, and treatment.
- a diluent, a dispersant, a surfactant, a binder and a lubricant may be additionally added to formulate the composition into a main-use dosage form such as an aqueous solution, a suspension, an emulsion, a pill, a capsule, a granule or a tablet.
- a main-use dosage form such as an aqueous solution, a suspension, an emulsion, a pill, a capsule, a granule or a tablet.
- it can be preferably formulated according to each disease or ingredient using an appropriate method in the field or the method disclosed in Remington's Pharmaceutical Science (Mack Publishing Company, Easton PA, 18th, 1990).
- the present invention relates to a combination for preventing or treating cancer, comprising a peptide having sequence number 1 or sequence number 2 of the present invention, a chimeric antigen receptor of the present invention, a cell expressing a chimeric antigen receptor of the present invention, or a T cell engager of the present invention; and an immunotherapy agent, a chemotherapeutic agent, or an antibody-drug conjugate (ADC).
- a combination for preventing or treating cancer comprising a peptide having sequence number 1 or sequence number 2 of the present invention, a chimeric antigen receptor of the present invention, a cell expressing a chimeric antigen receptor of the present invention, or a T cell engager of the present invention; and an immunotherapy agent, a chemotherapeutic agent, or an antibody-drug conjugate (ADC).
- ADC antibody-drug conjugate
- the chemotherapeutic agent may be a pro-apoptotic peptide, an immunogenic apoptosis inducer or an anticancer agent, and may be selected from the group consisting of SN-38 (7-Ethyl-10-hydroxy-camptothecin), daunorubicin, doxorubicin, epirubicin, idarubicin, pixantrone, sabarubicin, valrubicin, paclitaxel, docetaxel, mechloethamine, chlorambucil, phenylalanine, mustard, cyclophosphamide, Ifosfamide, carmustine (BCNU), lomustine (CCNU), streptozotocin, busulfan, thiotepa, cisplatin, carboplatin, dactinomycin (actinomycin D), plicamycin, mitomycin C, vincristine, vinblastine, teniposide, topotecan,
- the pharmaceutical composition or combination of the present invention may be administered at a concentration of TB511 of 100 to 300 nmol/kg, and oxaliplatin may be administered at a concentration of 1 to 2 mg/kg, but is not limited thereto.
- subject used in the present invention means a subject that requires a method for preventing, controlling or treating a disease, and can be used without limitation as a human, dog, monkey, cat, rodent, such as a mouse, a genetically engineered mouse, etc. More specifically, it means a mammal, such as a human or non-human primate, mouse, rat, dog, cat, horse, cow, etc.
- TAMs tumor-associated macrophages
- TAMpep TAM-targeting peptide
- dKLA pro-apoptotic peptide
- THP-1 monocyte cells were differentiated into M0, M1, or M2 macrophages, and proteomic analysis was performed. Specifically, THP-1 was differentiated into M0 macrophages by culturing in complete media with 100 nM PMA (phorbol 12-myristate 13-acetate; Sigma-Aldrich, St. Louis, MO, USA) for 24 h.
- PMA phorbol 12-myristate 13-acetate
- M0 cells were cultured with 20 ng/mL recombinant human interferon-gamma (rhIFN- ⁇ ; Prospec, Rehovot, Israel) and 100 ng/mL lipopolysaccharides (LPS; Sigma-Aldrich), or 20 ng/mL recombinant human interleukin-4 (rhIL-4; Prospec) and 20 ⁇ g/mL rhIL-13 (Prospec) in RPMI 1640 medium supplemented with 5% FBS for 72 h.
- rhIFN- ⁇ human interferon-gamma
- LPS lipopolysaccharides
- rhIL-4 human interleukin-4
- Prospec 20 ⁇ g/mL rhIL-13
- PC3 or A549 cells were grown in complete medium to 80% confluence, washed and cultured in serum-free medium for 24 h, and then filtered through a 0.2 ⁇ m syringe filter (Sartorius, Göttingen, Germany) to prepare tumor-conditioned medium (TCM), and M0 cells were cultured with 10% or 50% tumor-conditioned medium for 72 h to polarize to M2-TAM. Afterwards, THP-1 macrophages differentiated into M0, M1, and M2 were disrupted using Mem-PER Plus Membrane Protein Extraction Kit (Thermo Fisher Scientific, Waltham, MA, USA), and the cell lysates were incubated with biotinylated TAMpep for 1 h at room temperature.
- a total of 1,450 proteins were identified in M0, M1, and M2 macrophages, and 54 of them preferentially bound to biotin-labeled TAMpep in M2 macrophages compared to M0 and M1 macrophages (Figs. 12 to 14).
- CD18 was particularly highly expressed in myeloid lineage cell lines such as macrophages, and was weakly expressed only in lymphoid (Fig. 15).
- CD18-targeting sgRNA (sgRNA/CD18; GTTCAACGTGACCTTCCGGC) was transfected into M2-differentiated cas9 stable THP-1 cells to perform CD18/sgRNA knockdown, and then CD18 mRNA levels were analyzed by qRT-PCR to verify the generation of knockdown cells (Fig. 2). After that, the cells were treated with TB511 (3 ⁇ M) for 24 h, and cell viability was confirmed by MTS assay.
- TAMpep peptide (0, 0.001, 0.01, 0.1, 1, or 10 ⁇ g)
- biotinylated TAMpep and performing PAGE analysis of the eluted proteins to separate peptide-bound membrane proteins using Streptavidin Dynabeads (Thermo Fisher Scientific).
- immunofluorescence staining analysis was performed using FITC-labeled TAMpep and APC-conjugated anti-rabbit IgG secondary antibody (1:500, Invitrogen).
- the binding affinity of TAMpep to CD18 was analyzed.
- CD18 is associated with the progression of solid tumors, especially with the infiltration of M2 macrophages into tumor tissues.
- tumor tissue arrays paraffin was removed from normal and tumor tissue slides of breast, colon, lung, liver, kidney, and skin using EZ prep (Ventana Medical Systems, #950-102) at 76°C for 4 min, followed by incubation with cell conditioning solution 1 (Ventana Medical Systems, #950-124) at 100°C for 24 min for antigen retrieval.
- the slides were treated with OptiView peroxidase inhibitor (Ventana Medical Systems, #760-700) for 4 min at 37°C, followed by incubation with anti-CD18 antibody (1:500, LSbio). Slides were then visualized using the OptiView DAB IHC Detection Kit (Ventana Medical Systems, #760-700), and images were scanned and analyzed using Aperio ImageScope software (Leica, Wetzlar, Germany).
- the expression level of CD18 was found to be significantly higher in breast, colon, lung, liver, kidney, and skin tumor tissues than in breast, colon, lung, liver, kidney, and skin normal tissues (Figs. 6 and 7).
- THP-1 monocytes were differentiated into M0, M1, and M2 macrophages, and polarized with M2-TAM.
- the relative mRNA expression level of CD18 was analyzed by qRT-PCR, and the protein expression level was confirmed by Western blot analysis and immunofluorescence staining.
- the binding of TB511 to CD18 was analyzed using protein-peptide interaction modeling. Specifically, based on the sequence of the cysteine-rich region of CD18 (Uniport _p05107, cysteine-rich tandem repeat region: 449-617), the 3D structure was constructed using the trRosetta algorithm ( https:/yanglab.nankai.edu.cn/trRosetta/ ). Using the CD18 PDB file generated by trRosetta and the amino acid sequence of TB511, docking simulations of TB511 and CD18 were performed using the CABS-dock server ( http:/biocomp.chem.uw.edu.pl/CABSdock/ ). Ten models were generated.
- CD18 protein Abbexa
- FITC-conjugated TAMpep and alanine-substituted library peptides of TAMpep were reacted at room temperature for 2 hours, and then the binding affinity between the peptides was analyzed using ELISA. Based on the results of the above binding affinity analysis, a representative model was selected from among the 10 models generated through the above docking simulation.
- Protein-peptide interaction modeling analysis results showed that TB511 binding to total CD18 in the bent (closed) conformation was not predicted, whereas the open conformation cysteine-rich domain was strongly predicted to bind to TB511 (Fig. 18), suggesting that lysine and tryptophan residues of TB511 are the major binding elements.
- binding affinity analysis using the alanine substitution library of TAMpep showed that the A6 peptide, in which lysine was substituted with alanine, and the A12 peptide, in which tryptophan was substituted with alanine, exhibited significantly reduced binding to CD18 protein (Fig. 19).
- THP-1 cells were activated in the resting state by treating them with 1 mM manganese chloride tetrahydrate (Mn 2+ , Sigma-Aldrich) for 1 h. Then, TB511 (1 ⁇ M) was treated to the activated THP-1 cells and the inactivated control THP-1 cells, respectively. Cell viability was determined by staining with Annexin V (APC) and 7AAD and analyzing the stained cells with a FACS Lyric System (BD Bioscience, NJ, USA).
- APC Annexin V
- 7AAD FACS Lyric System
- THP-1 cells were treated with A549 tumor-conditioned medium, and the activation level of CD18 was analyzed by FACS using FITC-conjugated mAb KIM127 (Leinco Technologies, Pantone, MO, USA).
- THP-1 cells were differentiated into M0, M1, M2, and M2-TAM subtypes, respectively, and the degree of CD18 activation was analyzed.
- the activation level of CD18 was found to be significantly increased in a concentration-dependent manner by A549 tumor-conditioned medium treatment (Fig. 22).
- the activation level of CD18 was found to be the highest in polarized M2-TAM cultured with tumor-conditioned medium (Fig. 23), confirming that M2-TAM maintains the activated form of CD18 in the TME.
- THP-1 cells were differentiated into M0, M1, M2, or M2-TAM and treated with MitoTracker RedROX (Invitrogen) and caspase 3/7 reagents for 30 min, followed by TB511 (1 ⁇ M), and live-cell imaging was performed immediately using restoration fluorescence microscopy (DeltaVison, NJ, USA).
- caspase 3/7 activation by TB511 started in M2-TAM within 5 min of incubation with TB511 and reached a peak at 15 min.
- M2 macrophages were affected by TB511 to a lesser extent, but no caspase 3/7 activation was observed in M0 or M1 macrophages (Figs. 24 and 25). This confirmed that the conformational-specific binding between CD18 and TB511 resulted in selective cytotoxicity toward M2-TAM among macrophage subtypes.
- Example 7 Analysis of M2 macrophage membrane dynamics and intracellular transport of TB511
- TB511 was conjugated to carboxylic acid-functionalized gold nanorods (AuNRs; Nanopartz, Salt Lake City, UT, USA) using 1-ethyl-3-3-(dimethylaminopropyl)carbodiimide hydrochloride (Pierce, Rockford, IL, USA) and sulfo-N-hydroxysulfosuccinimide (Pierce).
- the cell counts of M2-TAM (the percentage of F4/80 + CD206 + among the CD45 + gated cells) and M1-TAM (the percentage of CD206 + F4/80 + among the CD45 + gated cells) in PC and CRC tumor tissues were analyzed by FACS, and the M1/M2 ratio was calculated.
- the cell counts of M1-TAM and M2-TAM in CRC tumor tissues, and the number of tissue-resident macrophages present in normal tissues including brain, skin, kidney, and liver after treatment with TB511 were analyzed by immunohistochemistry (IHC).
- the number of M2-TAM cells in PC and CRC tumor tissues was significantly reduced in the TB511-administered group compared to the control group, whereas the number of M1-TAM cells did not change significantly, indicating that the M1/M2 ratio was significantly increased by TB511 administration (Figs. 42 to 47).
- the number of M2-TAM cells was reduced and the number of M1-TAM cells was not changed by TB511 treatment in tumor tissues, whereas the tissue-resident macrophage population (F4/80 + ) in normal tissues was not affected by TB511 treatment (Figs. 48 to 51).
- Cy5-labeled TB511 was injected into an orthotopic breast cancer mouse model, and the bio-distribution of TB511 was investigated. Specifically, 4T1 breast cancer cells were injected into the mammary fat pad of mice at a density of 5 ⁇ 10 5 cells/mouse to prepare an orthotopic breast cancer mouse model, and 7 days later, Cy5-labeled TB511 or scrambled peptides (control) were intravenously injected at 2.5 mg/kg. The mice were anesthetized, and the distribution of TB511 in vivo was photographed and analyzed using IVIS Lumina II (PerkinElmer, Waltham, MA, USA).
- the fluorescent signal of TB511 was observed only in the injected breast tissue, not in the liver, lung, heart, kidney, or muscle, and showed a higher biological distribution in the tumor tissue compared to the control group (Figs. 52 to 55).
- TB511 specifically accumulates at tumor sites and selectively removes M2-TAM in the TME, but does not affect tissue-resident macrophages within normal tissues.
- the NK cell population in the PC was significantly increased, whereas the CD8 + T cell population did not change statistically significantly (Figs. 56 and 57).
- the number of NCAM1 + NK cells was found to increase in the PC specimens after TB511 treatment (Figs. 58 and 59).
- the numbers of CD8 + T and NK cells in the blood of NK or CD8 deficient mouse models were significantly decreased by antibody treatment (Figs. 73 to 76).
- TB511 showed an antitumor effect in both the control group and the CD8 + T cell deficient group, whereas it did not show an antitumor effect in the NK cell deficient group (Figs. 61 to 63).
- the number of tumor-infiltrated CD8 + T cells (CD45 + CD8 + ) and exhausted CD8 + T cells (PD-1 + CD8 + T) by TB511 treatment in immunotherapy-responsive tumors such as NSCLC and CRC were determined by FACS analysis, and the number of CD8 + T and Granzyme B + cells were determined by IHC analysis.
- a CRC mouse model was created by injecting CT26 cells, a colon cancer cell line, and anti-CD8 antibody (Fig. 70), and the antitumor effect by TB511 treatment was analyzed.
- the number of tumor-infiltrating CD8 + T cells in NSCLC increased in the group administered TB511, while the number of depleted CD8 + T cells decreased (Figs. 64 and 65).
- the antitumor immune cells only CD8 + T cells were significantly increased in CRC (Figs. 66 and 67).
- the IHC analysis results showed that the number of CD8 + T and Granzyme B + cells increased by TB511 treatment (Figs. 68 and 69), and the antitumor effect of TB511 was not observed only in CD8 + -depleted cells in the CRC mouse model (Figs. 70 to 72).
- mice which are immunodeficient mice, were irradiated with 1.5 Gy of gamma irradiation and intravenously injected with human CD34 + hematopoietic stem cells (Lonza, Basel, Switzerland) at 2 ⁇ 10 5 cells/mouse to generate a humanized mouse model. Eleven weeks after stem cell injection, peripheral blood was collected, and flow cytometry was used to confirm the engraftment of mature human leukocytes (human CD45 + cells).
- PC3 human prostate cancer cell line
- A549 human lung cancer cell line
- TB511 was administered every 3 days from day 11 after tumor cell injection (Figs. 77 and 85), and the tumor growth in the mice and the changes in human immune cells infiltrating the TME were investigated by t-SNE analysis.
- mice injected with A549 cells were administered TB511 alone, anti-PD-1 antibody (pembrolizumab) alone, or a combination of TB511 and anti-PD-1 antibody, and the antitumor effect was analyzed by measuring tumor volume and analyzing changes in tumor-infiltrating immune cell populations.
- the antitumor effects of the TB511 monotherapy group and the anti-PD-1 antibody monotherapy group were found to be similar, while the antitumor effect of the group administered in combination was found to be significantly increased (Fig. 85).
- the cell number of M1-TAMs (CD206 + CD11b + /CD45 + cells) was found to be significantly increased in the group administered in combination with TB511 and anti-PD-1 antibody, while the cell number of M2-TAMs (CD86 + CD11b + /CD45 + cells) was found to be significantly decreased (Figs. 86 to 88).
- Granzyme B-positive CD8 + T cells were significantly increased by co-administration of TB511 and anti-PD-1 antibody, indicating increased infiltration into the TME, while the depleted CD8 + T-cell (PD-1 + CD8 + /CD45 + cell) population was significantly decreased in the TB511 monotherapy group and the TB511 and anti-PD-1 antibody co-administration group (Figs. 89 to 91).
- a hPD-L1 MC38 tumor-humanized PD-1 mouse model was generated by subcutaneously injecting human PD-L1-expressing mouse colon cancer MC38 cells (hPD-L1 MC38 cells) (Shanghai Model Organisms Center, Shanghai, China) stably expressing human PD-L1 into the dorsal skin of genetically modified C57BL/6J mice (humanized PD-1 knock-in mice) (Shanghai Model Organisms Center, Shanghai, China) ( Figure 96).
- mice were divided into groups administered PBS, TB511, anti-PD-1 antibody (Selleckchem), or TB511 and anti-PD-1 antibody, and then PBS or TB511 (200 nmol/kg) was administered subcutaneously, and anti-PD-1 antibody (2.5 mg/kg) was administered intraperitoneally twice a week for 32 days to confirm the synergistic effect of co-administration of TB511 and anti-PD-1 antibody (Figs. 94 to 99).
- Example 11 Analysis of anticancer effects of combined administration of TB511 and chemotherapy (oxaliplatin) in a colon cancer animal model
- Mouse colon cancer cells were cultured in RPMI1640 medium (Welgene, Gyeongsan, Korea) supplemented with 10% fetal bovine serum (FBS; Welgene, Gyeongsan, Korea), 100 U/ml penicillin, and 100 ⁇ g/ml streptomycin (Invitrogen, CA, USA). Cells were seeded every 3 days when 80% confluent was reached, and cultured at 37°C and 5% CO 2 .
- FBS fetal bovine serum
- penicillin 100 ⁇ g/ml streptomycin
- CT-26 cells were harvested and mixed with Matrigel at a 1:1 ratio.
- the mixed cells were injected subcutaneously into the right flank of the mice at a concentration of 3 ⁇ 10 ⁇ 5 cells/mouse.
- the mice were divided into four groups: control, TB511, Oxaliplatin (Oxp), and combination treatment group.
- the mice in each group were injected subcutaneously with PBS or TB511 (200 nmol/kg) every 3 days, and Oxp (1.5 mg/kg) was injected intraperitoneally as a single dose.
- the tumor volume was measured every 3 days using a digital caliper.
- the animal experiments were approved by the Institutional Animal Care and Use Committee of Kyung Hee University (KHSAP(SE)-24-332). All mice were housed in a specific pathogen-free environment with free access to food and water under a 12-h light/dark cycle. After the analysis, all animals were euthanized using isoflurane and cervical dislocation. Tumor tissue was isolated and used for further analysis.
- Tumor tissues were minced and digested in serum-free cell culture medium containing DNase I (1 U/mL) and collagenase D (1 mg/mL). The samples were incubated for 1 h at 37°C with gentle agitation. The samples were filtered through a 100- ⁇ m nylon mesh sieve. Red blood cells were lysed with Pharmlyse buffer (BD Bioscience, CA, USA). Single cells were filtered through a 40- ⁇ m nylon mesh sieve and stained with the following antibodies.
- anti-CD45-APCcy7 (clone 30-F11; BD Bioscience, #557659), anti-CD4-BB700 (clone RM4-5; BD Bioscience, #566407), anti-CD8-PE-cy7 (clone 53-6.7; BD Bioscience, #552877), anti-NKp46-BV711 (clone 29A1.4; BD Bioscience, #740822);
- anti-CD45-APCcy7 clone 30-F11; BD Bioscience, #557659
- anti-CD11b-BV510 (clone M1/70; BD Bioscience, #566117)
- anti-F4/80-PE (clone BM8; Biolegend, #123110)
- anti-CD206-APC (clone C068C2; Biolegend, #141708)
- anti-CD86-BV786 (clone GL1; BD Bioscience, #740
- Example 11-2 TB511 and oxaliplatin were administered alone or in combination to a colon cancer mouse model.
- the mice were injected intraperitoneally with a single dose of oxaliplatin on the 7th day, and TB511 was injected every 3 days (Fig. 100).
- the tumor volume was significantly reduced in the TB511 and oxaliplatin groups, and the combination group showed the highest anticancer efficacy compared to the other groups, and tumor growth was significantly inhibited compared to the control and TB511 groups (Figs. 101 to 105).
- the weight changes of tumor tissues isolated from euthanized mice were measured according to TB511 and oxaliplatin treatment. As a result, it was shown that the tumor size and weight were significantly reduced in the combined treatment group compared to the control group (Fig. 106 and Fig. 108). In addition, when the weight change due to TB511 or oxaliplatin administration was checked, no weight change was observed in any group (Fig. 109).
- TB511 selectively targets M2-like TAM among macrophage subtypes in the TME.
- the distribution of macrophage subtypes in the TME was analyzed by flow cytometry.
- the flow cytometry results showed that the distribution of M1-like TAM (CD86 + F4/80 + cells within CD45 + CD11b + cells) did not show significant changes in any group.
- the distribution of M2-like TAM CD206 + F4/80 + cells within CD45 + CD11b +
- significantly decreased in the combination treatment group compared to the control group Figs. 109 to 112).
- lymphocytes were analyzed by flow cytometry.
- the distribution of CD4 T cells and NK cells did not show significant differences in any group.
- CD8 T cells significantly increased in the combination treatment group.
- Example 12 Analysis of anticancer effects of combined administration of TB511 and chemotherapy (paclitaxel) in a triple-negative breast cancer animal model
- mice Female mice (6–8 weeks old).
- Balb/c is a strain suitable for transplantation of the triple-negative breast cancer cell line 4T1 and was selected because it is widely used in immunology and tumor experiments.
- Mice were housed in an SPF environment at 22 ⁇ 1°C, 55 ⁇ 15% relative humidity, and 12-h day/night lighting cycle (lights on at 07:00, lights off at 19:00). Up to five mice were housed in cages (W 260 X L 420 X H 180 mm), and individual animals were identified with a permanent marker on the tail. Food and water were provided ad libitum through solid food for laboratory animals and water bottles, respectively. This experiment was approved by the Animal Ethics Committee of Kyung Hee University (KHUAP(SE)-20-398) in accordance with the Animal Protection Act.
- mice Female Balb/c mice (BALB/cAnNTac, Taconic Bioscience) were injected with triple-negative breast cancer cells, 4T1 (ATCC), at a density of 1 ⁇ 10 cells per fourth nipple.
- the 4T1 cells were suspended in serum-free media and mixed with Matrigel (Corning) to promote tumor formation.
- a group treated with PBS was set as a control group, and drug administration and analysis were performed on 18 mice per group.
- TB511 and paclitaxel were administered as a single administration or in combination at 3-day intervals starting 5 days after tumor cell injection.
- TB511 was administered by subcutaneous injection, and paclitaxel was administered by intraperitoneal injection (Fig. 118).
- the tumor size was measured using a digital caliper to calculate the volume.
- Example 12-2 when TB511 or paclitaxel was administered, the change in tumor size was measured, and through this, the anticancer effect of combined administration of TB511 and paclitaxel was confirmed.
- the analysis results are shown in Fig. 119.
- the tumor size in the group administered combined administration of TB511 and paclitaxel was significantly smaller compared to the group administered alone of TB511 and paclitaxel and the control group.
- Example 13 Analysis of anticancer effects of combined administration of TB511 and PADCEV (anti-Nectin-4 antibody antibody drug conjugate) in a humanized pancreatic cancer mouse model
- SID mice (NOD-Prkdcem1BeakIL2rgem1Break, 4 weeks old) were purchased from GemBioscience (Chungbuk, South Korea). Mice were irradiated with 1.5 Gy of ⁇ -rays and then intravenously injected with human CD34+ hematopoietic stem cell 2 ⁇ 10 cells (Lonza, Basel, Switzerland). SID mice were selected because they are a suitable humanized mouse model that can express humanized immune cells, are small in size and easy to handle, are convenient for experiments, and have immunodeficiency characteristics that do not generate an immune response for transplantation of human-derived hHSC cells. Mice were 16 weeks old at the start of administration, and a total of 20 female humanized mice were used.
- mice 4-week-old male immunodeficient mice (SID) were irradiated with 100 cGy for 1 minute, and human hematopoietic stem cells (hHSCs) were injected into the tail vein using a 30G insulin syringe within one hour.
- hHSCs human hematopoietic stem cells
- Table 2 shows the immune cell expression rate of the mice.
- the humanized mice used in the analysis were supplied by Lonza.
- human lung cancer cell lines (PANC1; ATCC) were prepared at a concentration of 2 ⁇ 10 cells per mouse, mixed 1:1 with Matrigel (Corning), and 100 ⁇ L of the mixture was injected subcutaneously into the right dorsal area of the mice using a 30G syringe.
- the dosage form of TB511 was prepared by the sponsor at a concentration of 5 mg/ml in D-PBS and diluted to 200 nmol/kg for administration.
- the administration dose of TB511 was set to 200 nmol/kg based on the results of the previous efficacy evaluation in a lung cancer mouse model, and PADCEV (anti-Nectin-4 antibody drug conjugate) was set to 5 mg/kg with reference to external data.
- the mice were divided into four groups for the study: G1 (PBS administration group), G2 (anti-nectin-4 administration group, 5 mg/kg), G3 (TB511 administration group, 200 nmol/kg), and G4 (combination administration group, PADCEV 5 mg/kg + TB511 200 nmol/kg).
- TB511 was administered subcutaneously into the back of mice using an insulin syringe after disinfecting with a 70% alcohol swab, and PADCEV was injected intraperitoneally using the same method.
- the adjuvant volume for both drugs was the same at 100 ⁇ L, and the administration interval was every 3 days.
- Brain, breast, colon, kidney, liver, lung, skin, and tumor tissues were cut into thin pieces using a MACS digester (Milteny Biotec, Auburn, CA, USA) and incubated in RPMI 1640 serum-free medium containing 1 U/mL DNase (Cincinnati, Indianapolis, IN, USA) and 1 mg/mL collagenase D (Roche) at 37°C for 30 min. Tissues were filtered through a 100 ⁇ m nylon mesh filter. Red blood cells in the filtered single cells were lysed with BD Pharm lyse buffer (BD Bioscience, CA, USA).
- Single cells were then stained with the following antibodies in BD Pharmingen Stain Buffer (BD Bioscience): anti-CD45-FITC, anti-CD11b-BV510, anti-CD18-PE, and anti-KIM127-BV786 (BD Bioscience) for human macrophages.
- Cells were detected using a FACS Lyric system (BD Bioscience) and analyzed using FlowJo software (BD Bioscience).
- tumor tissues were fixed overnight in 10% neutral buffered formalin.
- the tumor tissues embedded in paraffin were then sectioned at 5 ⁇ m thickness.
- the slides were deparaffinized with xylene and dehydrated with ethanol and deionized water.
- the slides were incubated in sodium citrate buffer (pH 6.0) for 15 min in a microwave oven.
- the slides were then treated with peroxidase blocking solution (Dako, Glostrup, Denmark) for 15 min and blocked with 5% bovine serum albumin.
- the slides were incubated with primary antibodies overnight, washed with TBS containing 0.1% Tween 20, and incubated with an avidin-biotin complex kit (Vector Laboratories, Burlingame, CA, USA).
- tumor tissues were sectioned at 5 ⁇ m thickness.
- the slides were deparaffinized with xylene and dehydrated with ethanol and deionized water.
- the slides were incubated in sodium citrate buffer (pH 6.0) for 15 min in a microwave oven.
- the slides were then treated with peroxidase-blocking solution (Dako, Glostrup, Denmark) for 15 min and blocked with 5% bovine serum albumin.
- the slides were incubated with primary antibodies overnight and then washed with TBS containing 0.1% Tween 20.
- Paraffin-embedded blocks were precisely sectioned into 5 ⁇ m thickness and mounted on Xenium slides.
- the sectioned slides were dried in a thermal cycler (C1000 96-well, Bio-Rad) at 42°C for 3 h. Deparaffinization and de-crosslinking were then performed. After prime hybridization and RNase treatment, the sectioned slides were hybridized overnight ( ⁇ 24 h) at 50°C with Xenium human 5k pan-tissue probe (10x Genomics). The slides were then washed and ligated at 42°C for 30 min. Amplification enhancement and amplification were immediately processed in a thermal cycler (C1000 96-well, Bio-Rad). The slides were washed again, and the cell division staining probe was applied overnight ( ⁇ 24 h) at 4°C. After staining enhancement and washing, autofluorescence suppression and nuclear staining were performed.
- the cut slides were then loaded onto the Xenium analyzer (XOA v3.0.2, 10x Genomics) for fluorescence signal detection. After running Xenium, the raw data were preprocessed with xeniumranger v3.0.0.
- a tumor mouse model was established by subcutaneously injecting human pancreatic cancer cell line (PANC1) into humanized mice, and the drug was administered to the mice twice a week.
- PANC1 human pancreatic cancer cell line
- the tumor size was significantly reduced in the TB511 or PADCEV administration groups compared to the PBS group, and in particular, the combination administration group showed the most significant reduction (Figs. 120 and 121).
- the expression level of PCNA was significantly reduced in the TB511, PADCEV, and combination groups compared to the PBS group (Figs. 122 and 123).
- E-cadherin an epithelial cell marker
- PADCEV PADCEV
- vimentin a mesenchymal cell marker
- TB511 reduces M2 macrophages in the tumor microenvironment
- tumor tissues were analyzed using immunohistochemical staining and flow cytometry.
- the results showed that the TB511 and combination treatment groups significantly decreased cells expressing CD163, a marker of M2 macrophages, in tumor tissues compared to the PBS group, whereas no effect was observed in the PADCEV group (Figs. 126 and 127).
- TB511 was shown to selectively bind to activated CD18 in M2 macrophages.
- CD18-positive cells in tumor tissues were shown to significantly decrease in the TB511 and combination treatment groups (Figs. 128 and 129).
- TB511 reduced M2 macrophages using the Kim127 antibody that binds to activated CD18 (Figs. 130 to 133).
- Figs. 130 to 133 we confirmed that TB511 selectively targets activated CD18 of M2 macrophages in tumor tissues and induces a decrease in these cells.
- TB511 The association of TB511 with activated CD18-expressing macrophages and CD8 T cells in normal tissues was shown in Fig. 20, where TB511 decreased activated CD18-expressing macrophages.
- TB511 decreased activated CD18-expressing macrophages.
- single cells were obtained from spleen, liver, lung, and brain tissues, stained for activated CD18 using Kim127 antibody, and analyzed by flow cytometry.
- TB511, PADCEV, and combination treatment groups showed no effect on Kim127-positive macrophages and CD8 T cells compared to the PBS group (Figs. 134 to 141). These results confirmed that TB511 could specifically target activated CD18-expressing M2 macrophages in tumor tissues.
- Figures 142 to 145 show the cell distribution of the PBS, TB511, PADCEV, and combination treatment groups.
- PBS group epithelial cells, endothelial cells, stromal cells, fibroblasts, and neural cells were evenly distributed near the tumor cells, whereas in the TB511, PADCEV, and combination treatment groups, the cell clusters were reduced to 4 or 3, respectively ( Figures 142 to 145).
- the above cell populations were analyzed using UMAP.
- the proportions of immune cells in the PBS, TB511, PADCEV, and combination administration groups were 18%, 23%, 17%, and 26%, respectively. That is, it can be confirmed that the groups treated with TB511 showed a higher proportion of immune cells compared to the other groups (Figs. 146 to 153).
- the proportion of epithelial cells which are pancreatic cancer cells, decreased in the TB511, PADCEV, and combination administration groups, and M1 macrophages increased, M2 macrophages decreased, and CD8 T cells and NK cells increased in the TB511 and combination administration groups.
- Treg cells did not appear in any drug group (Figs. 154 and 155).
- TB511 and PADCEV induced a decrease in tumor cells, and TB511 specifically targeted M2 macrophages to induce changes in immune cells within the microenvironment.
- PADCEV since it directly targets pancreatic cancer cells, it did not show any induction or changes in immune cells.
- CD8 T cells expressing exhausted T cell genes are indicated by white arrows, and cells expressing activated T cell genes are indicated by red arrows.
- the analysis results showed that the number of activated T cells increased in the TB511 and combination treatment groups compared to the PBS and PADCEV groups (Figs. 156 and 157).
- TB511 induces the infiltration and activation of CD8 T cells by targeting and reducing M2 macrophages in pancreatic cancer tumor tissues, and PADCEV directly targets nectin-4 of tumor cells to reduce tumor growth, but does not contribute to the regulation of immune cells in the tumor microenvironment.
Landscapes
- Health & Medical Sciences (AREA)
- Life Sciences & Earth Sciences (AREA)
- Chemical & Material Sciences (AREA)
- General Health & Medical Sciences (AREA)
- Public Health (AREA)
- Animal Behavior & Ethology (AREA)
- Veterinary Medicine (AREA)
- Organic Chemistry (AREA)
- Epidemiology (AREA)
- Medicinal Chemistry (AREA)
- Immunology (AREA)
- Engineering & Computer Science (AREA)
- Molecular Biology (AREA)
- Biochemistry (AREA)
- Bioinformatics & Cheminformatics (AREA)
- Proteomics, Peptides & Aminoacids (AREA)
- Biophysics (AREA)
- Pharmacology & Pharmacy (AREA)
- Genetics & Genomics (AREA)
- Gastroenterology & Hepatology (AREA)
- Cell Biology (AREA)
- Hematology (AREA)
- Urology & Nephrology (AREA)
- Chemical Kinetics & Catalysis (AREA)
- Biomedical Technology (AREA)
- General Chemical & Material Sciences (AREA)
- Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
- Toxicology (AREA)
- Zoology (AREA)
- Analytical Chemistry (AREA)
- Biotechnology (AREA)
- Oncology (AREA)
- Physics & Mathematics (AREA)
- Hospice & Palliative Care (AREA)
- Food Science & Technology (AREA)
- Pathology (AREA)
- General Physics & Mathematics (AREA)
- Microbiology (AREA)
- Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
Abstract
Description
본 발명은 CD18에 특이적으로 결합하여 항암 효과를 가지는 TB511 펩타이드 및 항암제의 병용 요법에 관한 것이다.The present invention relates to a combination therapy of a TB511 peptide that specifically binds to CD18 and has an anticancer effect and an anticancer agent.
기존의 항암치료는 암세포를 직접 공격하거나 암세포를 공격하는 체내 면역세포의 활성을 강화시키는 방향으로 연구되어 왔다. 그러나 이러한 항암제는 암세포 외의 다른 정상세포 또한 공격하여 탈모, 오심, 구토등의 많은 부작용을 가져왔고, 면역세포의 과도한 증가로 인한 부가적 반응을 초래한다. 기존의 화학적 요법이나 방사선 치료방법에 비하여 부작용을 최소화할 수 있는 항암면역치료방법은 체내의 면역시스템을 이용하여 암을 치료하는 방법이며, 이러한 항암면역치료기법 중에는 치료용 면역세포인 T 세포 (CAR-T 포함), 수지상세포(Dendritic Cells), 자연살해세포(Natural Killer Cells) 등을 체외에서 활성화 시킨 후에 체내에 직접 주입하는 세포치료제방법과 암 항원과 면역활성화 물질을 체내에 주입함으로써, 체내에 존재하는 면역세포를 직접적으로 활성화함으로써 항암효능을 높이는 항암백신에 대한 방법 등이 활발하게 진행되고 있다. 하지만, 이러한 세포치료제나 암백신은 주로 혈액암 관련 질병에 주로 사용되고 있고, 고형암에서는 대부분 그 치료효능이 매우 낮다는 단점을 갖고 있다. 이러한 이유 중의 하나는 고형암 주위에서 면역기능을 억제하는 미세환경 요인에 기인한다. 실제로, 종양미세환경에서 면역세포의 기능을 저하시키는 세포 (MDSC: myeoloid-derived stromal cells, Treg: regulatory T cell, TAM: tumor-assocaited macrophages)나 면역억제유발 사이토카인, 대사체 등이 활발하게 작용함으로써, 면역활성화 물질과 치료용 면역세포의 활성을 급격하게 저하시키는 것이다. 따라서 종양세포 및 면역세포에 직접적인 영향없이 종양세포의 주변 미세환경만을 조절하여 종양세포로의 영양분 공급, 종양세포 주변의 혈관신생등을 차단하여 항암효과를 갖는 치료제 개발이 중요해지고 있다. 종양 미세 환경은 악성 세포의 증식 및 생존, 혈관 신생, 전이, 비정상 적응 면역 및 호르몬 및 화학요법제에 대한 반응 감소에 기여하여 치료 목표로 크게 고려되고 있다. Existing cancer treatments have been studied in the direction of directly attacking cancer cells or enhancing the activity of the body's immune cells that attack cancer cells. However, these anticancer drugs attack normal cells in addition to cancer cells, resulting in many side effects such as hair loss, nausea, and vomiting, and causing additional reactions due to excessive increase in immune cells. Compared to existing chemotherapy or radiation therapy, anticancer immunotherapy is a method of treating cancer using the body's immune system, which can minimize side effects. Among these anticancer immunotherapy techniques, there are cell therapy methods that activate therapeutic immune cells such as T cells (including CAR-T), dendritic cells, and natural killer cells outside the body and then directly inject them into the body, and methods for anticancer vaccines that directly activate immune cells existing in the body by injecting cancer antigens and immune-activating substances into the body, thereby increasing anticancer efficacy. However, these cell therapy agents or cancer vaccines are mainly used for diseases related to blood cancer, and most of them have a disadvantage in that their therapeutic efficacy is very low for solid cancers. One of the reasons for this is due to microenvironmental factors that suppress immune function around solid cancers. In fact, cells that reduce the function of immune cells in the tumor microenvironment (MDSC: myeoloid-derived stromal cells, Treg: regulatory T cells, TAM: tumor-assocaited macrophages) or immunosuppressive cytokines and metabolites actively work, rapidly reducing the activity of immune-activating substances and therapeutic immune cells. Therefore, it is becoming important to develop a treatment that has an anticancer effect by controlling only the microenvironment around tumor cells without directly affecting tumor cells and immune cells, thereby blocking the supply of nutrients to tumor cells and angiogenesis around tumor cells. The tumor microenvironment is greatly considered as a treatment target because it contributes to the proliferation and survival of malignant cells, angiogenesis, metastasis, abnormal adaptive immunity, and decreased response to hormones and chemotherapeutics.
종양 주변의 미세환경(microenvironment)은 내피세포, 염증성 세포 및 섬유아세포로 구성되어 있으며, 1970년대에 종양 관련 대식세포(tumor-associated macrophage, TAM)가 종양의 성장에 있어서 중요한 역할을 한다는 것이 밝혀졌다. 종양 관련 대식세포는 암의 성장, 전이 등 전반적인 종양 미세환경과 관련하여 중요한 역할을 담당하며, 종양억제 M1형 대식세포 또는 종양지지 M2형 대식세포의 두 가지 표현형으로 분류된다. M1형 대식세포는 항원을 제시하는 강력한 능력을 갖고, 일반적으로 인터페론-γ, 지방당류(LPS), 종양 괴사 인자(TNF)-α에 의하여 활성화 되며, 전염증 작용 및 살균 작용을 한다. M2형 대식세포는 다양한 세포 외 기질성분, 혈관 신생 및 주화성 인자를 방출함으로써 면역억제, 종양형성 및 혈관형성을 촉진하는 것으로 알려져 있다. 일반적으로, IL-4와 IL-13에 의해 유도되며, 아르기나제-1, mannose(MMR, CD206), 스캐빈저 수용체(SR-A, CD204), CD163, IL-10과 같은 마커를 발현함으로써 M1형 종양 관련 대식세포와 구별된다. 종양 주변에 존재하는 종양 관련 대식세포는 종양 세포의 성장, 전이와 밀접하게 관련이 되어 있으며, 암 환자에서 많은 숫자의 M2형 종양 관련 대식세포가 종양 주변에 존재하면 환자의 예후 및 생존률이 좋지 못한 것으로 보고되고 있다. M2형 대식세포는 암의 성장을 촉진하는 IL-10, TGFβ 및 CCL18과 같은 사이토카인을 생성하며, M2형 종양 관련 대식세포의 표면에 존재하는 PDL1 및 B7-1/2와 같은 수용체들은 T 세포, NK 세포의 항종양 활성을 억제하는 것으로 보고되고 있다. M2형 종양 관련 대식세포가 다량으로 존재하는 미세환경에서는 종양의 성장, 분화 및 전이가 활발하게 이루어지므로, M2형 종양 관련 대식세포를 표적으로 하는 표적 치료제의 개발이 필요하다.The microenvironment around the tumor is composed of endothelial cells, inflammatory cells, and fibroblasts, and in the 1970s, tumor-associated macrophages (TAMs) were found to play a critical role in tumor growth. TAMs play an important role in the overall tumor microenvironment, including cancer growth and metastasis, and are classified into two phenotypes: tumor-suppressive M1 macrophages and tumor-supportive M2 macrophages. M1 macrophages have a potent antigen-presenting ability, are generally activated by interferon-γ, lipopolysaccharide (LPS), and tumor necrosis factor (TNF)-α, and exhibit proinflammatory and bactericidal effects. M2 macrophages are known to promote immunosuppression, tumorigenesis, and angiogenesis by releasing various extracellular matrix components, angiogenic and chemotactic factors. In general, they are induced by IL-4 and IL-13, and are distinguished from M1 type tumor-associated macrophages by expressing markers such as arginase-1, mannose (MMR, CD206), scavenger receptors (SR-A, CD204), CD163, and IL-10. Tumor-associated macrophages present around tumors are closely related to the growth and metastasis of tumor cells, and it has been reported that when a large number of M2 type tumor-associated macrophages exist around tumors in cancer patients, the prognosis and survival rate of the patients are poor. M2 type macrophages produce cytokines such as IL-10, TGFβ, and CCL18 that promote cancer growth, and receptors such as PDL1 and B7-1/2 present on the surface of M2 type tumor-associated macrophages have been reported to suppress the antitumor activity of T cells and NK cells. Since tumor growth, differentiation, and metastasis occur actively in a microenvironment where M2 type tumor-associated macrophages exist in large numbers, development of targeted therapeutics targeting M2 type tumor-associated macrophages is necessary.
한편, 암세포들은 면역세포들에 의한 살상 작용기작을 회피하기 위해 정상세포들이 면역세포 활성화를 억제할 때 이용되는 면역관문(immune checkpoint) 단백질을 세포 표면에 발현하고 있어, 최근 암을 치료하기 위한 방법으로써 면역관문 억제 단백질에 대한 연구가 활발히 진행되고 있다. 면역관문 억제 단백질 중 특히 PD-1/PD-L1 결합의 차단이 암치료에 큰 효과가 있으며, 다른 면역관문 억제 단백질에 비해 부작용이 적다는 결과가 학계에 보고되었다 (J. Naidoo et al. (2015) Annals of Oncology, Lucia Gelao et al. (2014) Toxins, Gorge K. Philips et al (2015) International Immunology). PD-1 수용체는 T 세포, B 세포, 자연 살해(natural killer; NK)/자연 살해 T(natural killer T; NKT) 세포 등을 포함한 활성화된 면역세포 타입들의 표면에서 발현된다 (Goodman, Patel & Kurzrock, PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas, Nature Reviews Clinical Oncology, 14:203-220,2017.). PD-1은 T 세포 활성의 음성 조절인자이고, 종양 표면에서 PD-1과 이의 리간드들 중 하나인 PD-L1의 상호작용은 효과적인 면역 반응을 생성하는 활성화된 T 세포의 능력을 감소시키는 면역관문 차단을 나타낸다. PD-1 또는 PD-L1을 표적하는 치료용 항체는 리간드-수용체 상호작용을 차단하고 종양 미세 환경으로 면역 기능을 회복시킨다. PD-1 및 PD-L1을 표적하는 여러 치료용 모노클로날 항체(monoclonal antibodies; mAb)들이 시판되고 있으며, PD-1과 PD-L1 결합력을 억제하는 Anti-PD1 항체 치료제들인 BMS사의 Opdivo (nivolumab), Merck사의 Keytruda (Pembrolizumab), Regeneron사에서 개발된 Libtayo (Cemiplimab)들과 Anti-PD-L1 항체 치료제들인 Roche사의 Tecentriq (Atezolizumab), AstraZeneca사의 Imfinzi (Durvalumab), Merck Sereno사의 Bavencio (Avelumab)들이 US FDA 승인을 받아서 임상에서 난치성 암치료에 혁신을 가져오고 있다. Meanwhile, cancer cells express immune checkpoint proteins on their cell surface, which are used by normal cells to suppress immune cell activation in order to avoid the killing mechanism by immune cells. Recently, research on immune checkpoint inhibitor proteins as a method for treating cancer has been actively conducted. It has been reported in academic circles that among immune checkpoint inhibitor proteins, blocking PD-1/PD-L1 binding is particularly effective in cancer treatment and has fewer side effects than other immune checkpoint inhibitor proteins (J. Naidoo et al. (2015) Annals of Oncology, Lucia Gelao et al. (2014) Toxins, Gorge K. Philips et al (2015) International Immunology). The PD-1 receptor is expressed on the surface of activated immune cell types, including T cells, B cells, and natural killer (NK)/natural killer T (NKT) cells (Goodman, Patel & Kurzrock, PD-1-PD-L1 immune-checkpoint blockade in B-cell lymphomas, Nature Reviews Clinical Oncology, 14:203-220, 2017.). PD-1 is a negative regulator of T cell activation, and the interaction of PD-1 with one of its ligands, PD-L1, on the tumor surface represents an immune checkpoint blockade that reduces the ability of activated T cells to generate effective immune responses. Therapeutic antibodies targeting PD-1 or PD-L1 block the ligand-receptor interaction and restore immune function to the tumor microenvironment. Several therapeutic monoclonal antibodies (mAbs) targeting PD-1 and PD-L1 are on the market, and anti-PD1 antibody treatments that inhibit the binding of PD-1 and PD-L1, such as Opdivo (nivolumab) from BMS, Keytruda (Pembrolizumab) from Merck, and Libtayo (Cemiplimab) developed by Regeneron, and anti-PD-L1 antibody treatments such as Tecentriq (Atezolizumab) from Roche, Imfinzi (Durvalumab) from AstraZeneca, and Bavencio (Avelumab) from Merck Sereno, have received US FDA approval and are revolutionizing the treatment of intractable cancers in clinical trials.
본 발명의 목적은 서열번호 1 또는 서열번호 2의 펩타이드를 항원 결합 도메인으로 포함하는 키메라 항원 수용체를 제공하는 것이다.An object of the present invention is to provide a chimeric antigen receptor comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
또한, 본 발명의 목적은 키메라 항원 수용체를 암호화하는 유전자를 포함하는 재조합 벡터를 제공하는 것이다. It is also an object of the present invention to provide a recombinant vector comprising a gene encoding a chimeric antigen receptor.
또한, 본 발명의 목적은 재조합 벡터로 형질전환된 키메라 항원 수용체 발현 세포를 제공하는 것이다.Furthermore, it is an object of the present invention to provide a chimeric antigen receptor expressing cell transformed with a recombinant vector.
또한, 본 발명의 목적은 서열번호 1 또는 서열번호 2의 펩타이드를 포함하는 T 세포 관여자를 제공하는 것이다.Furthermore, it is an object of the present invention to provide a T cell engager comprising a peptide of
또한, 본 발명의 목적은 암의 예방 또는 치료용 약학적 조성물을 제공하는 것이다.In addition, it is an object of the present invention to provide a pharmaceutical composition for preventing or treating cancer.
또한, 본 발명의 목적은 암의 예방 또는 치료용 조합물을 제공하는 것이다.Furthermore, it is an object of the present invention to provide a combination for preventing or treating cancer.
또한, 본 발명의 목적은 암 진단용 조성물을 제공하는 것이다.In addition, an object of the present invention is to provide a composition for cancer diagnosis.
아울러, 본 발명의 목적은 암의 예방 또는 치료방법을 제공하는 것이다.In addition, the purpose of the present invention is to provide a method for preventing or treating cancer.
상기 과제를 해결하기 위하여, 본 발명은 서열번호 1 또는 서열번호 2의 펩타이드를 항원 결합 도메인으로 포함하는 키메라 항원 수용체를 제공한다.To solve the above problem, the present invention provides a chimeric antigen receptor comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
또한, 본 발명은 상기 키메라 항원 수용체를 암호화하는 유전자를 포함하는 재조합 벡터를 제공한다.Additionally, the present invention provides a recombinant vector comprising a gene encoding the chimeric antigen receptor.
또한, 본 발명은 상기 재조합 벡터로 형질전환된 키메라 항원 수용체 발현 세포를 제공한다.In addition, the present invention provides a chimeric antigen receptor expressing cell transformed with the recombinant vector.
또한, 본 발명은 서열번호 1 또는 서열번호 2의 펩타이드를 포함하는 T 세포 관여자를 제공한다.Additionally, the present invention provides a T cell engager comprising a peptide of
또한, 본 발명은 상기 서열번호 1 또는 서열번호 2의 펩타이드, 키메라 항원 수용체, 키메라 항원 수용체 발현 세포, 또는 T 세포 관여자를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물을 제공한다.In addition, the present invention provides a pharmaceutical composition for preventing or treating cancer, comprising a peptide of the
또한, 본 발명은 상기 서열번호 1 또는 서열번호 2의 펩타이드, 키메라 항원 수용체, 키메라 항원 수용체 발현 세포, 또는 T 세포 관여자; 및 면역항암제, 화학항암제 또는 항체-약물 접합체(Antibody-Drug Conjugate, ADC);를 포함하는 암의 예방 또는 치료용 조합물을 제공한다.In addition, the present invention provides a combination for preventing or treating cancer, comprising a peptide of the
또한, 본 발명은 상기 서열번호 1 또는 서열번호 2의 펩타이드, 키메라 항원 수용체, 키메라 항원 수용체 발현 세포, 또는 T 세포 관여자를 유효성분으로 포함하는 암 진단용 조성물을 제공한다.In addition, the present invention provides a composition for diagnosing cancer, comprising a peptide of the
아울러, 본 발명은 상기 약학적 조성물 또는 상기 조합물을 개체에게 투여하는 단계를 포함하는 암의 예방 또는 치료방법을 제공한다.In addition, the present invention provides a method for preventing or treating cancer, comprising a step of administering the pharmaceutical composition or the combination to a subject.
본 발명의 TB511은 종양 미세환경(tumor microenvironment, TME)에서 종양 진행에 기여하는 M2-TAM(M2-Tumor-associated macrophages, M2-TAMs)의 활성화된 형태의 CD18에 특이적으로 결합함으로써 세포사멸을 유도하며, M2-TAM이 아닌 다른 조직-상주 대식세포에는 독성을 나타내지 않고, 다양한 암 모델 및 인간화된 마우스 모델에서 종양 살해 세포의 침윤을 증가시키고 항종양 효과를 나타냈으며, 특히, 다른 면역항암제와 병용 처리시 시너지적인 항종양 효과를 나타냈으므로, 이를 면역항암제 또는 이와의 조합물로 활용할 수 있다.TB511 of the present invention induces apoptosis by specifically binding to CD18 of activated form of M2-Tumor-associated macrophages (M2-TAMs) that contribute to tumor progression in the tumor microenvironment (TME), and does not exhibit toxicity to other tissue-resident macrophages other than M2-TAM, and increases the infiltration of tumor killer cells and exhibits antitumor effects in various cancer models and humanized mouse models, and in particular, exhibits synergistic antitumor effects when used in combination with other immunotherapy drugs, and therefore can be utilized as an immunotherapy drug or a combination thereof.
도 1 내지 11은 TB511의 표적을 분석하고, TB511의 표적인 CD18의 고형 종양의 진행 관련성을 분석한 도이다:Figures 1 to 11 are diagrams analyzing the target of TB511 and analyzing the relationship between CD18, the target of TB511, and the progression of solid tumors:
도 1: TB511의 표적 단백질 분석 과정;Figure 1: Target protein analysis process of TB511;
도 2: CD18 녹다운 세포의 검증 결과;Figure 2: Validation results of CD18 knockdown cells;
도 3: 세포생존율 분석 결과;Figure 3: Cell viability analysis results;
도 4: TB511 및 CD18의 직접적인 결합 분석 결과;Figure 4: Results of direct binding analysis of TB511 and CD18;
도 5: M2 대식세포에서 TAMpep 및 CD18의 위치 분석 결과;Figure 5: Localization analysis of TAMpep and CD18 in M2 macrophages;
도 6 및 도 7: 고형 종양에서 CD18의 발현 분석 결과; 및Figures 6 and 7: Results of CD18 expression analysis in solid tumors; and
도 8 내지 11: 대식세포 서브타입에 따른 CD18의 발현 분석 결과.Figures 8 to 11: Results of CD18 expression analysis according to macrophage subtype.
도 12 내지 15는 TAMpep과 결합하는 M0, M1 및 M2 대식세포의 막 단백질의 분석 결과를 나타낸 도이다.Figures 12 to 15 are diagrams showing the results of analysis of membrane proteins of M0, M1, and M2 macrophages binding to TAMpep.
도 12: M0 대식세포로부터 추출된 단백질;Figure 12: Proteins extracted from M0 macrophages;
도 13: M1 대식세포로부터 추출된 단백질;Figure 13: Proteins extracted from M1 macrophages;
도 14: M2 대식세포로부터 추출된 단백질; 및Figure 14: Proteins extracted from M2 macrophages; and
도 15: M2/M0 및 M2/M1의 비율.Figure 15: Ratios of M2/M0 and M2/M1.
도 16 및 도 17은 TAMpep의 CD18에 대한 결합 친화도를 분석한 결과를 나타낸 도이다:Figures 16 and 17 are diagrams showing the results of analyzing the binding affinity of TAMpep to CD18:
a: TAMpep의 CD18에 대한 결합 친화도; 및 a: Binding affinity of TAMpep to CD18; and
b: 항-CD18 항체의 CD18에 대한 결합 친화도.b: Binding affinity of anti-CD18 antibody to CD18.
도 18 내지 26은 TB511의 CD18 구조 특이적 표적화를 통한 세포독성 유도 효과를 분석한 도이다:Figures 18 to 26 are diagrams analyzing the cytotoxicity induction effect through CD18 structure-specific targeting of TB511:
도 18: 단백질-펩타이드 상호작용 모델링 분석 결과;Figure 18: Results of protein-peptide interaction modeling analysis;
도 19: TB511의 CD18 결합 부위 분석 결과;Figure 19: Results of CD18 binding site analysis of TB511;
도 20 및 도 21: CD18의 활성화 여부에 따른 TB511의 세포독성 분석 결과;Figures 20 and 21: Cytotoxicity analysis results of TB511 depending on CD18 activation;
도 22: 종양미세환경 유도에 따른 CD18의 활성화 수준 분석 결과;Figure 22: Results of analysis of CD18 activation level according to tumor microenvironment induction;
도 23: 대식세포의 서브타입에 따른 CD18의 활성화 수준 분석 결과;Figure 23: Results of analysis of CD18 activation levels according to macrophage subtypes;
도 24 및 도 25: 대식세포 서브타입에 따른 TB511의 세포사멸 분석 결과; 및Figure 24 and Figure 25: Results of apoptosis analysis of TB511 according to macrophage subtype; and
도 26: 종양 조직 내 CD18의 활성화 수준 분석 결과.Figure 26: Results of analysis of CD18 activation level in tumor tissue.
도 27 내지 34는 M2 대식세포에서 TB511의 내재화 및 엔도좀 탈출을 분석한 도이다: Figures 27 to 34 show the analysis of internalization and endosomal escape of TB511 in M2 macrophages:
도 27: TB511의 M2 대식세포 내 분포 분석 결과;Figure 27: Results of analysis of the distribution of TB511 in M2 macrophages;
도 28 내지 30: TB511의 3D 공간궤적 분석 결과Figures 28 to 30: Results of 3D spatial trajectory analysis of TB511
도 31: CPZ(chlorpromazine), M*?*CD(methyl-_-cyclodextrin) 또는 CyD(cytochalasin D) 처리에 따른 엔도좀에서 초기 엔도좀 항원 1(early endosome antigen 1 (EEA1)) 및 TB511-Cy5의 위치 분석 결과; Figure 31: Results of analysis of the localization of early endosome antigen 1 (EEA1) and TB511-Cy5 in endosomes following treatment with CPZ (chlorpromazine), M*?*CD (methyl-_-cyclodextrin), or CyD (cytochalasin D);
도 32: CPZ(chlorpromazine), M_CD(methyl-_-cyclodextrin) 또는 CyD(cytochalasin D) 처리에 따른 TB511-유도 세포사멸 분석 결과; Figure 32: Results of TB511-induced apoptosis analysis according to treatment with CPZ (chlorpromazine), M_CD (methyl-_-cyclodextrin) or CyD (cytochalasin D);
도 33: 처리에 따른 엔도좀에서 초기 엔도좀 항원 1 및 TB511-Cy5의 위치 분석 결과; 및Figure 33: Results of localization analysis of
도 34: Baf-A1(bafilomycin A1) 또는 CQ(chloroquine) 처리에 따른 TB511-유도 세포사멸 분석 결과.Figure 34: Results of TB511-induced apoptosis analysis following Baf-A1 (bafilomycin A1) or CQ (chloroquine) treatment.
도 35 내지 도 53은 TB511의 항종양 효과를 분석한 도이다:Figures 35 to 53 are diagrams analyzing the antitumor effect of TB511:
도 35 내지 37: 면역항암제 비반응성 종양(Cold tumors) 마우스 모델에서 TB511의 항종양 효과;Figures 35-37: Antitumor effect of TB511 in a mouse model of cold tumors that are nonresponsive to immunotherapy;
도 38 내지 41: 면역항암제 반응성 종양(Hot tumors) 마우스 모델에서 TB511의 항종양 효과;Figures 38-41: Antitumor effect of TB511 in a mouse model of immunotherapy-responsive tumors (hot tumors);
도 42 내지 47: 종양 조직에서 TB511의 대식세포 서브타입에 대한 선택적인 표적화 분석 결과; 및Figures 42 to 47: Results of selective targeting analysis of TB511 on macrophage subtypes in tumor tissue; and
도 49 내지 51: 정상 조직 및 종양 조직에서 대식세포 수 분석 결과.Figures 49 to 51: Results of macrophage count analysis in normal and tumor tissues.
도 52 내지 55는 유방암 마우스 모델에서 TB511의 생물학적 분포 분석을 통해 TB511의 종양 표적화를 분석한 도이다.Figures 52 to 55 are diagrams analyzing the tumor targeting of TB511 through the biological distribution analysis of TB511 in a breast cancer mouse model.
도 56 내지 72는 TME 에서 TB511에 의한 면역세포 변화를 분석한 도이다:Figures 56 to 72 are diagrams analyzing immune cell changes caused by TB511 in TME:
도 56 및 도 57: 전립선암(PC) 검체에서 CD8+ T 및 NK 세포의 변화 분석 결과;Figures 56 and 57: Results of analysis of changes in CD8+ T and NK cells in prostate cancer (PC) specimens;
도 58 및 도 59: 전립선암(PC) 검체에서 TB511의 항종양 효과에 대한 CD8+ T 및 NK 세포의 영향 분석 결과;Figures 58 and 59: Results of analysis of the effects of CD8+ T and NK cells on the antitumor effect of TB511 in prostate cancer (PC) specimens;
도 60: NK 또는 CD8 결핍 대장암 마우스 모델 제조 프로세스;Figure 60: Process for preparing NK or CD8 deficient colon cancer mouse model;
도 61 내지 63: 전립선암 세포주 주입 마우스 모델에서 대조군 (isotype control) 및 CD8+ T 세포 결핍 그룹 (CD8 depletion) 및 NK 세포가 결핍된 그룹 (NK depletion)에서 세포생존율 분석 결과;Figures 61 to 63: Results of cell viability analysis in the control group (isotype control), CD8+ T cell depletion group (CD8 depletion), and NK cell depletion group (NK depletion) in a prostate cancer cell line injection mouse model;
도 64 및 도 65: 폐암세포(lung cancer cells, LLC) 주입 마우스 모델에서 종양-침윤 CD8+ T 세포의 수 및 고갈된 CD8+ T 세포의 수 분석 결과;Figures 64 and 65: Analysis of the number of tumor-infiltrating CD8+ T cells and the number of exhausted CD8+ T cells in a lung cancer cells (LLC) injection mouse model;
도 66 및 도 67: 대장암 세포(colorectal cancer, CRC) 항종양 면역세포의 수 분석 결과;Figures 66 and 67: Results of analysis of the number of anti-tumor immune cells in colorectal cancer (CRC) cells;
도 68 및 도 69: CD8+ T 및 Granzyme B+ 세포수 분석 결과;Figures 68 and 69: Results of CD8+ T and Granzyme B+ cell count analysis;
도 70: CD8 결핍 대장암 마우스 모델 제조 프로세스;Figure 70: Manufacturing process of CD8 deficient colon cancer mouse model;
도 71: 대조군 마우스에서 TB511의 항종양 효과; 및Fig. 71: Antitumor effect of TB511 in control mice; and
도 72: CD8 결핍 대장암 마우스 모델에서 TB511의 항종양 효과.Figure 72: Antitumor effect of TB511 in a CD8 deficient colon cancer mouse model.
도 73 내지 76은 CD8 결핍 마우스 모델 (a) 또는 NK 결핍 마우스 모델 (b)의 혈액에서 CD8+ T 및 NK 세포수를 분석한 도이다.Figures 73 to 76 are diagrams analyzing the number of CD8+ T and NK cells in the blood of a CD8 deficient mouse model (a) or an NK deficient mouse model (b).
도 77 내지 93은 TB511의 임상적 가능성을 평가한 도이다:Figures 77 to 93 are diagrams evaluating the clinical potential of TB511:
도 77: 인간화된 전립선암 마우스 모델 제조 프로세스;Figure 77: Process for manufacturing a humanized prostate cancer mouse model;
도 78: 면역결핍 마우스에서 TB511의 항종양 효과;Figure 78: Antitumor effect of TB511 in immunodeficient mice;
도 79: 인간화된 전립선암 마우스에서 TB511의 항종양 효과;Figure 79: Antitumor effect of TB511 in humanized prostate cancer mice;
도 80 내지 82: TME에 침투하는 인간 면역세포 분석 결과;Figures 80 to 82: Analysis of human immune cells infiltrating the TME;
도 83: 인간화된 폐암 마우스 모델 제조 프로세스;Figure 83: Process for manufacturing a humanized lung cancer mouse model;
도 84: 면역결핍 마우스에서 TB511의 항종양 효과;Figure 84: Antitumor effect of TB511 in immunodeficient mice;
도 85: 인간화된 폐암 마우스에서 TB511 단독, 항-PD-1 항체 단독, 및 TB511 및 항-PD-1 항체 조합 (combination)의 항종양 효과;Figure 85: Antitumor effects of TB511 alone, anti-PD-1 antibody alone, and TB511 and anti-PD-1 antibody combination in humanized lung cancer mice;
도 86 내지 88: M2-TAMs (CD86+CD11b+/CD45+ 세포)의 세포수 분석 결과; Figures 86 to 88: Cell count analysis results of M2-TAMs (CD86+CD11b+/CD45+ cells);
도 89 내지 91: 고갈된 CD8+ T-세포 (PD-1+CD8+/CD45+ 세포)의 세포수 분석 결과; 및Figures 89 to 91: Cell count analysis results of exhausted CD8+ T-cells (PD-1+CD8+/CD45+ cells); and
도 92 및 도 93: 인간화된 비소세포 폐암(NSCLC) 종양 조직에서 면역조직화학 염색을 통해 결정된 각 그룹의 CD8+ 세포 수.Figure 92 and Figure 93: CD8+ cell counts in each group determined by immunohistochemical staining in humanized non-small cell lung cancer (NSCLC) tumor tissues.
도 94 내지 99는 TB511 및 면역 체크포인트 억제제의 병용 투여에 의한 시너지 효과를 분석한 도이다Figures 94 to 99 are diagrams analyzing the synergistic effect of combined administration of TB511 and an immune checkpoint inhibitor.
도 94: hPD-L1 MC38 종양-인간화된 PD-1 마우스 모델 제조 프로세스;Figure 94: Process for manufacturing hPD-L1 MC38 tumor-humanized PD-1 mouse model;
도 95: 대조군의 종양 부피;Figure 95: Tumor volume in the control group;
도 96: 항-PD-1 단독 투여 그룹의 종양 부피;Figure 96: Tumor volume in the anti-PD-1 monotherapy group;
도 97: TB511 단독 투여 그룹의 종양 부피;Figure 97: Tumor volume in the TB511 monotherapy group;
도 98: TB511 및 항-PD-1 항체의 병용 투여 그룹의 종양 부피; 및Figure 98: Tumor volume of the group administered with combination of TB511 and anti-PD-1 antibody; and
도 99: 각 그룹의 마우스의 생존율.Figure 99: Survival rate of mice in each group.
도 100 내지 105는 TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 따른 항암효과를 분석한 도이다.Figures 100 to 105 are diagrams analyzing the anticancer effect of combined administration of TB511 and oxaliplatin.
도 100: 대장암 마우스 모델 제조 및 약물 투여 프로세스;Figure 100: Manufacturing process of colon cancer mouse model and drug administration;
도 101: 대조군의 종양 부피;Figure 101: Tumor volume in the control group;
도 102: 대조군의 종양 부피;Figure 102: Tumor volume of the control group;
도 103: TB511 단독 투여 그룹의 종양 부피;Figure 103: Tumor volume in the TB511 monotherapy group;
도 104:옥살리플라틴(oxaliplatin) 단독 투여 그룹의 종양 부피;및Figure 104: Tumor volume in the oxaliplatin monotherapy group; and
도 105: TB511 및 옥살리플라틴 의 병용 투여 그룹의 종양 부피.Figure 105: Tumor volume of the combination therapy group of TB511 and oxaliplatin.
도 106 내지 108은 TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 따른 종양 무게 및 체중 변화를 분석한 도이다.Figures 106 to 108 are diagrams analyzing tumor weight and body weight changes according to combined administration of TB511 and oxaliplatin.
도 106: 종양 조직의 크기;Fig. 106: Size of tumor tissue;
도 107: 종양 조직의 무게 변화;및Fig. 107: Changes in the weight of tumor tissue; and
도 108: 마우스의 체중 변화.Fig. 108: Weight change of the mouse.
도 109 내지 113은 TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 따른 종양 미세 환경(Tumor Micro Environment, TME) 내 M2 유사 TAM(Tumor Associated Macrophage)의 감소를 분석한 도이다.Figures 109 to 113 are diagrams analyzing the decrease in M2-like tumor associated macrophages (TAMs) in the tumor microenvironment (TME) following combined administration of TB511 and oxaliplatin.
도 109 및 도 110: TME 내 대식세포 집단에 대한 유세포분석 결과;Figures 109 and 110: Flow cytometry analysis results for macrophage populations within the TME;
도 111: M1 유사 TAM(CD45+ CD11b+ 세포 내의 CD86+ F4/80+ 세포);Figure 111: M1-like TAMs (CD86+ F4/80+ cells within CD45+ CD11b+ cells);
도 112: M2 유사 TAM(CD45+ CD11b+ 내의 CD206+ F4/80+ 세포);및Figure 112: M2-like TAMs (CD206+ F4/80+ cells within CD45+ CD11b+); and
도 113: M1/M2 비율.Fig. 113: M1/M2 ratio.
도 114 내지 117은 TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 의한 세포독성 T세포의 TME로의 침투 유도 분석한 도이다.Figures 114 to 117 are diagrams analyzing the induction of infiltration of cytotoxic T cells into the TME by combined administration of TB511 and oxaliplatin.
도 114: TME 내 림프구 분포에 대한 유세포분석 결과;Figure 114: Flow cytometry analysis results for lymphocyte distribution within the TME;
도 115: CD4 T 세포 (CD45+ CD3+ CD4+ 세포);Figure 115: CD4 T cells (CD45+ CD3+ CD4+ cells);
도 116: CD8 T 세포(CD45+ CD3+ CD8+ 세포); 및Figure 116: CD8 T cells (CD45+ CD3+ CD8+ cells); and
도 117: NK세포(CD45+ CD3+ NKp46+ 세포).Figure 117: NK cells (CD45+ CD3+ NKp46+ cells).
도 118은 삼중음성유방암 마우스 모델 설정 및 약물 프로세스를 나타낸 도이다.Figure 118 is a diagram showing the triple-negative breast cancer mouse model setup and drug process.
도 119은 TB511 및 파클리탁셀(paclitaxel)의 병용투여에 따른 항암 효과 분석 결과를 나타낸 도이다.Figure 119 is a diagram showing the results of an analysis of the anticancer effect according to combined administration of TB511 and paclitaxel.
도 120 내지 125는 인간 췌장암 마우스 모델에서 TB511 및 PADCEV의 병용투여에 따른 종양 억제 분석 결과를 나타낸 도이다.Figures 120 to 125 are diagrams showing the results of tumor inhibition analysis according to combined administration of TB511 and PADCEV in a human pancreatic cancer mouse model.
도 120: 종양 접종 후 43일이 도과하였을 때의 종양 샘플;Figure 120:
도 121: 종양 접종 후 시간 경과에 따른 종양의 부피;Figure 121: Tumor volume over time after tumor inoculation;
도 122 및 도 123: 종양 조직에 대한 Ki67의 면역조직화학 염색 및 Ki67 양성 세포 정량화한 그래프;및Figures 122 and 123: Graphs showing immunohistochemical staining of Ki67 for tumor tissue and quantification of Ki67 positive cells; and
도 124 및 도 125: 종양 조직에서 E-카드헤린(E-cadherin) 및 비멘틴(Vimentin)의 면역형광염색 분석 결과.Figures 124 and 125: Results of immunofluorescence staining analysis of E-cadherin and Vimentin in tumor tissues.
도 126 내지 131은 TB511 및 PADCEV의 병용투여에 따른 종양 조직에서 M2 대식세포의 감소 분석 결과를 나타낸 도이다.Figures 126 to 131 are diagrams showing the results of analysis of the reduction of M2 macrophages in tumor tissues following combined administration of TB511 and PADCEV.
도 126 및 도 127: 종양조직에서 CD163 및 액틴(actin)의 면역조직화학 염색 결과 및 CD163 양성세포 정량화한 그래프;Figures 126 and 127: Graphs showing the results of immunohistochemical staining of CD163 and actin in tumor tissue and quantification of CD163 positive cells;
도 128 및 도 129: 종양조직에서 CD18의 면역조직화학 염색 결과 및 CD18 양성세포 정량화;및Figure 128 and Figure 129: Results of immunohistochemical staining of CD18 in tumor tissue and quantification of CD18 positive cells; and
도 130 및 도 131: 종양조직에서 Kim127 및 CD11b의 면역조직화학 염색 결과 및 Kim127 및 CD11b 양성세포 정량화한 그래프.Figures 130 and 131: Graphs showing the results of immunohistochemical staining of Kim127 and CD11b in tumor tissue and the quantification of Kim127 and CD11b positive cells.
도 132 및 도 133은 TB511 및 PADCEV의 병용투여에 따른 종양 조직에서 CD8 T 세포의 증가 분석 결과를 나타낸 도이다.Figures 132 and 133 are diagrams showing the results of analysis of the increase in CD8 T cells in tumor tissues following combined administration of TB511 and PADCEV.
도 132: 종양 조직에서 CD8의 면역형광 염색분석 결과;및Figure 132: Results of immunofluorescence staining analysis of CD8 in tumor tissue; and
도 133: CD8 양성 세포의 수를 정량화한 그래프.Figure 133: Graph quantifying the number of CD8 positive cells.
도 134 내지 141은 정상 조직에서 활성화된 CD18을 발현하는 대식세포 및 CD8 T 세포와 TB511의 연관성 분석 결과를 나타낸 도이다.Figures 134 to 141 are diagrams showing the results of an association analysis of TB511 with macrophages and CD8 T cells expressing activated CD18 in normal tissues.
도 134 내지 137: 항-Kim127, 항-CD18, 항-CD11b 및 항-CD45 항체를 이용한 대식세포에 대한 유세포분석 결과;및 Figures 134 to 137: Flow cytometry analysis results for macrophages using anti-Kim127, anti-CD18, anti-CD11b and anti-CD45 antibodies; and
도 138 내지 141: 항-Kim127, 항-CD18, 항-CD8 및 항-CD45 항체를 이용한 CD8 T 세포에 대한 유세포분석 결과.Figures 138 to 141: Flow cytometry analysis of CD8 T cells using anti-Kim127, anti-CD18, anti-CD8, and anti-CD45 antibodies.
도 142 내지 156은 TB511 및 PADCEV의 병용투여에 따른 종양 미세환경 내 면역 세포 조절에 대한 분석결과를 나타낸 도이다.Figures 142 to 156 are diagrams showing the results of analysis on immune cell regulation within the tumor microenvironment following combined administration of TB511 and PADCEV.
도 142 내지 145: 종양조직 내 세포 클러스터; 및Figures 142 to 145: Cell clusters within tumor tissue; and
도 146 내지 149: 종양조직 내 세포 클러스터에 대한 UMAP 분석 결과.Figures 146 to 149: UMAP analysis results for cell clusters within tumor tissue.
도 150 내지 153: 종양조직 내 세포 클러스터에서 세포 유형의 비율을 나타낸 파이 차트;및Figures 150 to 153: Pie charts showing the proportion of cell types in cell clusters within tumor tissue; and
도 154 및 도 155: 각 세포 유형별 세포집단(cell population)(%).Figures 154 and 155: Cell population (%) by each cell type.
도 156 내지 159는 종양 미세환경 내에서 CD8 T 세포의 활성화에서 TB511의 효과 분석결과를 나타낸 도이다.Figures 156 to 159 illustrate the results of analysis of the effect of TB511 on the activation of CD8 T cells within a tumor microenvironment.
도 156 및 도 157: 종양 조직에서 CD8 T 세포의 소진(exhausted) 유전자(흰색 화살표) 및 활성화 인자 유전자(빨간색 화살표); 및Figures 156 and 157: CD8 T cell exhausted genes (white arrows) and activator genes (red arrows) in tumor tissue; and
도 158 및 도 159: CTLA4, FOXP3, LAG3, GZMB, IFNG 및 PDCD1 유전자 발현 수준.Figures 158 and 159: CTLA4, FOXP3, LAG3, GZMB, IFNG and PDCD1 gene expression levels.
이하, 첨부된 도면을 참조하여 본 발명의 구현예로 본 발명을 상세히 설명하기로 한다. 다만, 하기 구현예는 본 발명에 대한 예시로 제시되는 것으로, 당업자에게 주지 저명한 기술 또는 구성에 대한 구체적인 설명이 본 발명의 요지를 불필요하게 흐릴 수 있다고 판단되는 경우에는 그 상세한 설명을 생략할 수 있고, 이에 의해 본 발명이 제한되지는 않는다. 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 with reference to the attached drawings as embodiments of the present invention. However, the following embodiments are presented as examples of the present invention, and if it is judged that a detailed description of a technology or configuration well known to those skilled in the art may unnecessarily obscure the gist of the present invention, the detailed description thereof may be omitted, and the present invention is not limited thereby. The present invention is capable of various modifications and applications within the scope of the following claims and equivalents interpreted therefrom.
또한, 본 명세서에서 사용되는 용어(terminology)들은 본 발명의 바람직한 실시예를 적절히 표현하기 위해 사용된 용어들로서, 이는 사용자, 운용자의 의도 또는 본 발명이 속하는 분야의 관례 등에 따라 달라질 수 있다. 따라서, 본 용어들에 대한 정의는 본 명세서 전반에 걸친 내용을 토대로 내려져야 할 것이다. 명세서 전체에서, 어떤 부분이 어떤 구성요소를 "포함"한다고 할 때, 이는 특별히 반대되는 기재가 없는 한 다른 구성요소를 제외하는 것이 아니라 다른 구성 요소를 더 포함할 수 있는 것을 의미한다.In addition, the terminology used in this specification is a terminology used to appropriately express the preferred embodiment of the present invention, and this may vary depending on the intention of the user or operator, or the custom of the field to which the present invention belongs. Therefore, the definition of these terms should be determined based on the contents throughout this specification. Throughout the specification, when a part is said to "include" a certain component, this does not mean that other components are excluded, but rather that other components can be included, unless specifically stated otherwise.
본 발명에서 사용되는 모든 기술용어는, 달리 정의되지 않는 이상, 본 발명의 관련 분야에서 통상의 당업자가 일반적으로 이해하는 바와 같은 의미로 사용된다. 또한 본 명세서에는 바람직한 방법이나 시료가 기재되나, 이와 유사하거나 동등한 것들도 본 발명의 범주에 포함된다. 본 명세서에 참고문헌으로 기재되는 모든 간행물의 내용은 본 발명에 통합된다.All technical terms used in the present invention, unless otherwise defined, are used with the same meaning as commonly understood by those skilled in the art in the relevant field of the present invention. In addition, although preferred methods or samples are described in this specification, similar or equivalent ones are also included in the scope of the present invention. The contents of all publications mentioned as references in this specification are incorporated into the present invention.
본 명세서 전반을 통하여, 천연적으로 존재하는 아미노산에 대한 통상의 1문자 및 3문자 코드가 사용될 뿐만 아니라 Aib(α아미노이소부티르산), Sar(N-methylglycine) 등과 같은 다른 아미노산에 대해 일반적으로 허용되는 3문자 코드가 사용된다. 또한 본 발명에서 약어로 언급된 아미노산은 하기와 같이 IUPAC-IUB 명명법에 따라 기재되었다:Throughout this specification, the conventional one-letter and three-letter codes for naturally occurring amino acids are used, as well as the generally accepted three-letter codes for other amino acids, such as Aib (α-aminoisobutyric acid), Sar (N-methylglycine), etc. In addition, amino acids referred to by abbreviations in the present invention are described according to the IUPAC-IUB nomenclature as follows:
알라닌: A, 아르기닌: R, 아스파라긴: N, 아스파르트산: D, 시스테인: C, 글루탐산: E, 글루타민: Q, 글리신: G, 히스티딘: H, 이소류신: I, 류신: L, 리신: K, 메티오닌: M, 페닐알라닌: F, 프롤린: P, 세린: S, 트레오닌: T, 트립토판: W, 티로신: Y 및 발린: V. Alanine: A, arginine: R, asparagine: N, aspartic acid: D, cysteine: C, glutamic acid: E, glutamine: Q, glycine: G, histidine: H, isoleucine: I, leucine: L, lysine: K, methionine: M, phenylalanine: F, proline: P, serine: S, threonine: T, tryptophan: W, tyrosine: Y, and valine: V.
일 측면에서, 본 발명은 서열번호 1 또는 서열번호 2의 펩타이드를 항원 결합 도메인으로 포함하는 키메라 항원 수용체(Chimeric antigen receptor: CAR)에 관한 것이다.In one aspect, the present invention relates to a chimeric antigen receptor (CAR) comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2 as an antigen binding domain.
일 구현예에서, 상기 서열번호 1 또는 서열번호 2의 펩타이드는 M2 대식세포 또는 M2 종양 관련 대식세포(M2 tumor associated macrophages, M2 TAM)에 특이적으로 결합할 수 있다.In one embodiment, the peptide of SEQ ID NO: 1 or SEQ ID NO: 2 can specifically bind to M2 macrophages or M2 tumor associated macrophages (M2 TAM).
일 구현예에서, 상기 서열번호 1 또는 서열번호 2의 펩타이드는 CD18과 특이적으로 결합할 수 있다.In one embodiment, the peptide of SEQ ID NO: 1 or SEQ ID NO: 2 can specifically bind to CD18.
일 구현예에서, 상기 펩타이드는 이의 변이체 또는 이의 유사체를 포함할 수 있다.In one embodiment, the peptide may comprise a variant thereof or an analog thereof.
본 발명에 있어서, 상기 펩타이드는 상기 아미노산 서열을 갖는 것이 바람직하지만 이에 한정되는 것은 아니다. 본 발명의 바람직한 구현예에 따르면, 상기 펩타이드는 상기 아미노산의 비율이 50% 이상, 바람직하게는 60% 이상, 보다 바람직하게는 70% 이상, 보다 바람직하게는 80% 이상, 보다 바람직하게는 90% 이상, 가장 바람직하게는 100%로 높은 것이 바람직하다.In the present invention, it is preferable that the peptide has the amino acid sequence described above, but is not limited thereto. According to a preferred embodiment of the present invention, it is preferable that the peptide has a high ratio of the amino acid of 50% or more, preferably 60% or more, more preferably 70% or more, more preferably 80% or more, more preferably 90% or more, and most preferably 100%.
본 발명에 있어서, 상기 펩타이드는 표적화 서열, 태그(tag), 표지된 잔기, 반감기 또는 펩타이드의 안정성을 증가시키기 위한 특정 목적으로 고안된 추가의 아미노산 서열도 포함할 수 있다. 또한, 본 발명의 펩타이드는 이펙터(effectors), 약물, 프로드럭, 독소, 펩타이드, 전달 분자 등의 커플링 파트너와 연결될 수 있다.In the present invention, the peptide may also include additional amino acid sequences designed for a specific purpose, such as a targeting sequence, a tag, a labeled residue, a half-life or to increase the stability of the peptide. In addition, the peptide of the present invention may be linked to coupling partners, such as effectors, drugs, prodrugs, toxins, peptides, delivery molecules, etc.
본 발명에 있어서, 상기 펩타이드는 약학적으로 허용 가능한 염의 형태로 제조될 수 있다. 구체적으로 산을 첨가함으로써 염을 형성할 수 있고, 예를 들어 무기산(예: 염산, 히드로브롬산, 인산, 질산, 황산 등), 유기 카르복실산(예: 아세트산, 트리플루오로아세트산과 같은 할로 아세트산, 프로피온산, 말레산, 숙신산, 말산, 시트르산, 타르타르산, 살리실산), 및 산성 당(글루쿠론산, 갈락투론산, 글루콘산, 아스코르브산), 산성 폴리사카리드 (예: 히알우론산, 콘드로이틴 술페이트, 아르기닌산), 콘드로이틴 술페이트와 같은 술폰산 당 에스테르를 포함하는 유기 술폰산(예: 메탄술폰산, p-톨루엔 술폰산) 등을 첨가하여 염을 형성할 수 있다.In the present invention, the peptide can be prepared in the form of a pharmaceutically acceptable salt. Specifically, a salt can be formed by adding an acid, for example, an inorganic acid (e.g., hydrochloric acid, hydrobromic acid, phosphoric acid, nitric acid, sulfuric acid, etc.), an organic carboxylic acid (e.g., acetic acid, haloacetic acids such as trifluoroacetic acid, propionic acid, maleic acid, succinic acid, malic acid, citric acid, tartaric acid, salicylic acid), and an organic sulfonic acid including an acidic sugar (glucuronic acid, galacturonic acid, gluconic acid, ascorbic acid), an acidic polysaccharide (e.g., hyaluronic acid, chondroitin sulfate, arginic acid), a sulfonic acid sugar ester such as chondroitin sulfate (e.g., methanesulfonic acid, p-toluene sulfonic acid), etc.
본 발명에서 사용된 용어, "펩타이드"는 아미노산 중합체로서, 천연 아미노산 뿐 아니라, 비단백질성 아미노산도 구성요소로 포함할 수 있다.The term "peptide" used in the present invention refers to an amino acid polymer, which may include not only natural amino acids but also non-protein amino acids as components.
본 발명에서 사용된 용어, "펩타이드 변이체"는 기준 물질과 비교하였을 때 최소한 한개의 아미노산 차이(치환, 삽입 또는 결손)를 포함하는 대응하는 아미노산 서열을 말한다. 특정 구체예들에 있어서 "변이체"는 기준 서열과 비교하였을 때 높은 아미노산 서열 상동성(homology) 및/또는 보존적 아미노산 치환, 결손 및/또는 삽입을 가진다. The term "peptide variant" as used herein refers to a corresponding amino acid sequence that contains at least one amino acid difference (substitution, insertion or deletion) compared to a reference sequence. In certain embodiments, a "variant" has high amino acid sequence homology and/or conservative amino acid substitutions, deletions and/or insertions compared to a reference sequence.
본 발명에서 사용된 용어, "펩타이드 유사체"는 아미노산의 측쇄 또는 알파-아미노산 백본에 대하여 하나 이상의 다른 기능기로 치환된 유사체를 포함할 수 있다. 측쇄 또는 백본 개질화 펩타이드 유사체의 예로는 피롤리딘 고리가 하이드록시기로 치환된 하이드록시프롤린이나, N-메틸 글리신 "펩토이드"를 들 수 있으나, 이로 제한되지 않는다. 펩타이드 유사체의 종류에 대해서는 당업계에 공지되어 있다.The term "peptide analogue" as used herein may include analogues in which the side chain or the alpha-amino acid backbone of an amino acid is substituted with one or more other functional groups. Examples of side chain or backbone modified peptide analogues include, but are not limited to, hydroxyproline in which the pyrrolidine ring is substituted with a hydroxy group, or N-methyl glycine "peptoids". Types of peptide analogues are well known in the art.
본 발명에서 사용된 용어, "보존적 아미노산 치환"이란 1개 이상의 아미노산을 해당 펩타이드의 생물학적 또는 생화학적 기능의 손실을 야기하지 않는 유사한 생화학적 특성을 갖는 아미노산으로 치환하는 것을 포함하는 변이체의 변형을 의미한다. "보존적 아미노산 치환"은 아미노산 잔기를 유사한 측쇄를 갖는 아미노산 잔기로 대체시키는 치환이다. 유사한 측쇄를 갖는 아미노산 잔기 부류는 해당 기술분야에 규정되어 있으며, 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.As used herein, the term "conservative amino acid substitution" refers to a modification of a variant 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 peptide. A "conservative amino acid substitution" is a substitution that replaces 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 and defined 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).
본 발명에 따른 펩타이드들은 표준 합성 방법, 재조합 발현 시스템, 또는 임의의 다른 당해 분야의 방법에 의해 제조될 수 있다. 따라서, 본 발명에 따른 펩타이드들은, 예를 들어 하기를 포함하는 방법을 포함하는 다수의 방법으로 합성될 수 있다:Peptides according to the present invention can be prepared by standard synthetic methods, recombinant expression systems, or any other method known in the art. Thus, peptides according to the present invention can be synthesized by a number of methods, including, for example, methods including:
(a) 펩타이드를 고체상 또는 액체상 방법의 수단으로 단계적으로 또는 단편 조립에 의해 합성하고, 최종 펩타이드 생성물을 분리 및 정제하는 방법; 또는(a) a method of synthesizing a peptide stepwise or by fragment assembly by means of solid-phase or liquid-phase methods, and isolating and purifying the final peptide product; or
(b) 펩타이드를 인코딩하는 핵산 작제물을 숙주세포 내에서 발현시키고, 발현 생성물을 숙주 세포 배양물로부터 회수하는 방법; 또는(b) a method of expressing a nucleic acid construct encoding a peptide in a host cell and recovering the expression product from the host cell culture; or
(c) 펩타이드를 인코딩하는 핵산 작제물의 무세포 시험관 내 발현을 수행하고, 발현 생성물을 회수하는 방법; 또는(c) a method of performing cell-free in vitro expression of a nucleic acid construct encoding a peptide and recovering the expression product; or
(a), (b) 및 (c)의 임의의 조합으로 펩타이드의 단편을 수득하고, 이어서 단편을 연결시켜 펩타이드를 수득하고, 당해 펩타이드를 회수하는 방법.A method of obtaining fragments of a peptide by any combination of (a), (b), and (c), then linking the fragments to obtain a peptide, and recovering the peptide.
본 발명에서 사용된 용어, "CAR(chimeric antigen receptor)"는, 면역 효과기 세포에 특정 항원에 대한 특이성을 부여할 수 있는, 자연적으로 존재하지 않는 수용체를 의미한다. CAR는 대개 세포 외 도메인(Ectodomain), 막투과성 도메인(transmembrane domain) 및 세포 내 도메인(Ectodomain)으로 구성된다.The term "CAR (chimeric antigen receptor)" used in the present invention refers to a non-naturally occurring receptor capable of conferring specificity for a specific antigen to immune effector cells. CAR is usually composed of an extracellular domain (Ectodomain), a transmembrane domain, and an intracellular domain (Ectodomain).
상기 세포 외 도메인은 항원 결합 부위(antigen recognition region)를 포함하며, CAR의 막통과 도메인은 세포 외 도메인과 연결된 형태로, 자연적 또는 합성된 것에서 유래한 것일 수 있다. 자연적으로 존재하는 것에 유래한 경우, 막 결합 또는 막투과성 단백질에서 유래한 것일 수 있으며, T 세포 수용체의 알파, 베타 또는 제타 체인, CD28, CD3 엡실론, CD45, CD4, CD5, CDS, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137, CD 154 또는 CD8 등 다양한 단백질의 막 투과성 영역에서 유래한 부분일 수 있다. 이러한 막통과 도메인의 서열은 막투과성 단백질의 막투과성 영역 부분을 공지하고 있는 통상의 기술분야에 공지된 문헌 등으로부터 얻을 수 있으나, 이에 제한되는 것은 아니다.The above extracellular domain includes an antigen recognition region, and the transmembrane domain of the CAR is in a form connected to the extracellular domain, and may be derived from natural or synthetic sources. If derived from a naturally existing source, it may be derived from a membrane-bound or membrane-permeable protein, and may be a portion derived from a membrane-permeable region of various proteins such as the alpha, beta or zeta chain of the T cell receptor, CD28, CD3 epsilon, CD45, CD4, CD5, CDS, CD9, CD16, CD22, CD33, CD37, CD64, CD80, CD86, CD134, CD137,
또한, 상기 막통과 도메인이 합성된 것일 경우, 이는 류신 및 발린과 같은 소수성 아미노산 잔기를 주로 포함할 수 있으며, 그 예로 페닐알라닌, 트립토판 및 발린의 트리플렛(triplet)이 합성된 막통과 도메인에 존재할 수 있으나, 이에 제한되는 것은 아니다. 이러한 막통과 도메인에 대한 서열정보는 합성된 막통과 도메인에 대한 통상의 기술분야에 공지된 문헌으로부터 얻을 수 있으나, 이에 제한되는 것은 아니다. In addition, when the above-mentioned transmembrane domain is synthetic, it may mainly include hydrophobic amino acid residues such as leucine and valine, and for example, a triplet of phenylalanine, tryptophan and valine may be present in the synthetic transmembrane domain, but is not limited thereto. Sequence information on such transmembrane domain can be obtained from literature known in the art regarding synthetic transmembrane domains, but is not limited thereto.
본 발명의 CAR에서 상기 세포 내 도메인은 세포 내에 존재하는 CAR의 도메인 일부로서, 막통과 도메인과 연결된 형태이다. 본 발명의 상기 세포 내 도메인은 CAR의 항원 결합 부위에 항원이 결합하면 T 세포 활성화, 바람직하게는 T 세포 증식을 가져오는 것이 특징인, 세포 내 신호전달 도메인을 포함할 수 있다. 상기 세포 내 신호전달 도메인은 세포 외에 존재하는 항원 결합 부위에 항체가 결합하면, T 세포 활성화를 가져올 수 있는 신호를 전달하는 부분이라면 특별히 그 종류에 제한되지 않으며, 다양한 종류의 세포 내 신호전달 도메인이 사용될 수 있으며, 그 예로 면역수용체 티로신-기초한 활성화 모티프(tyrosine-based activation motif) 또는 ITAM일 수 있으며, 상기 ITAM은 CD3 제타(ξ, zeta), FcR 감마, FcR 베타, CD3 감마, CD3 델타, CD3 엡실론, CDS, CD22, CD79a, CD79b, CD66d 또는 FcεRIγ에서 유래한 것을 포함하나, 이에 제한되는 것은 아니다.In the CAR of the present invention, the intracellular domain is a part of the domain of the CAR existing within a cell, and is connected to the transmembrane domain. The intracellular domain of the present invention may include an intracellular signaling domain which is characterized by causing T cell activation, preferably T cell proliferation, when an antigen binds to the antigen binding site of the CAR. The intracellular signaling domain is not particularly limited in its type as long as it is a part that transmits a signal capable of causing T cell activation when an antibody binds to the antigen binding site existing outside the cell, and various types of intracellular signaling domains may be used. Examples of the intracellular signaling domain include an immunoreceptor tyrosine-based activation motif or ITAM, and the ITAM includes, but is not limited to, those derived from CD3 zeta (ξ, zeta), FcR gamma, FcR beta, CD3 gamma, CD3 delta, CD3 epsilon, CDS, CD22, CD79a, CD79b, CD66d or FcεRIγ.
CAR는 T-세포 신호화 분자의 세포질 도메인에 경첩 및 막관통 영역을 통해 커플링되는 종양 연계 항원 (TAA)에 특이적인 항체의 단쇄 단편 가변부 (scFv)를 포함한다. 대부분의 통상적인 림프구 활성화 모이어티는 T-세포 촉발 (예를 들어 CD3ζ) 모이어티를 갖는 탠덤(tandem)에서 T-세포 공자극 (예를 들어 CD28, CD137, OX40, ICOS, 및 CD27) 도메인을 포함한다. CAR-매개 입양 면역요법은 CAR-이식된 세포가 비-HLA-제한 방식으로 표적 종양 세포 상의 TAA를 직접 인식하게 한다.CARs comprise a single-chain fragment variable region (scFv) of an antibody specific for a tumor-associated antigen (TAA) that is coupled via the hinge and transmembrane regions to the cytoplasmic domain of a T-cell signaling molecule. Most common lymphocyte activating moieties comprise a T-cell costimulatory (e.g., CD28, CD137, OX40, ICOS, and CD27) domain in tandem with a T-cell triggering (e.g., CD3ζ) moiety. CAR-mediated adoptive immunotherapy allows CAR-engrafted cells to directly recognize TAAs on target tumor cells in a non-HLA-restricted manner.
일 측면에서, 본 발명은 상기 키메라 항원 수용체를 암호화하는 유전자를 포함하는 재조합 벡터에 관한 것이다.In one aspect, the present invention relates to a recombinant vector comprising a gene encoding the chimeric antigen receptor.
일 측면에서, 본 발명은 상기 재조합 벡터로 형질전환된 키메라 항원 수용체 발현 세포에 관한 것이다.In one aspect, the present invention relates to a chimeric antigen receptor expressing cell transformed with the recombinant vector.
일 구현예에서, 키메라 항원 수용체 발현 세포는 키메라 항원 수용체 발현 대식세포(CAR-macrophage), 키메라 항원 수용체 발현 T(CAR-T) 세포, 키메라 항원 수용체 발현-감마-델타 T(CAR-Gamma-delta T) 세포 또는 자연살해(CAR-NK) 세포일 수 있다.In one embodiment, the chimeric antigen receptor expressing cell can be a chimeric antigen receptor expressing macrophage (CAR-macrophage), a chimeric antigen receptor expressing T (CAR-T) cell, a chimeric antigen receptor expressing-gamma-delta T (CAR-Gamma-delta T) cell, or a natural killer (CAR-NK) cell.
본 발명에서 사용된 용어, "키메라 항원 수용체 발현 T(CAR-T) 세포"는 CAR을 발현하는 T 세포를 의미한다. 상기 키메라 항원 수용체 발현 T(CAR-T) 세포는 ⅰ)HLA(human leukocyte antigen)에 비의존적인 방식으로 암 항원을 인지하므로, 세포 표면에 HLA 발현을 감소시켜 항암제의 작용을 회피하는 암들을 치료할 수 있는 이점을 지니며, ⅱ)HLA 타입과 무관하므로, 환자의 HLA 타입과 관계없이 치료에 이용할 수 있으며, ⅲ)짧은 시간 내에 많은 양의 암 특이적 T 세포를 만들어 낼 수 있으므로, 우수한 항암 효과를 나타낼 수 있는 이점을 지닌다.The term "chimeric antigen receptor expressing T (CAR-T) cell" used in the present invention refers to a T cell expressing a CAR. The chimeric antigen receptor expressing T (CAR-T) cell has the advantage of: i) recognizing a cancer antigen in a manner independent of HLA (human leukocyte antigen), and thus capable of treating cancers that evade the action of anticancer agents by reducing HLA expression on the cell surface; ii) being independent of the HLA type, and thus capable of being used for treatment regardless of the patient's HLA type; and iii) capable of producing a large amount of cancer-specific T cells in a short period of time, and thus capable of exhibiting an excellent anticancer effect.
상기 T 세포는 CD4+ T세포(도움 T 세포, TH 세포), CD8+ T세포(세포독성 T 세포, CTL), 기억 T 세포, 조절 T 세포(Treg 세포) 자연살해 T 세포 등 있으며, 본 발명에서 CAR이 도입되는 T 세포는 바람직하게는 CD8+ T세포 이나, 이에 제한되지 않는다.The above T cells include CD4 + T cells (helper T cells, TH cells), CD8 + T cells (cytotoxic T cells, CTLs), memory T cells, regulatory T cells (Treg cells), natural killer T cells, etc., and the T cells into which the CAR is introduced in the present invention are preferably CD8 + T cells, but are not limited thereto.
일 측면에서, 본 발명은 서열번호 1 또는 서열번호 2의 펩타이드를 포함하는 T 세포 관여자(T-cell engager)에 관한 것이다.In one aspect, the present invention relates to a T-cell engager comprising a peptide having
일 구현예에서, 상기 T 세포 관여자는 예를 들어, 이중-특이적 T-세포 관여자 (bispecific T-cell engager, BiTE)일 수 있다. 상기 BiTE는 인공 이중특이적 모노클로날 항체의 부류로, 약 55 킬로달톤의 단일 펩티드 사슬 상에 4 개의 상이한 유전자로부터 아미노산 서열 또는 상이한 항체의 2 개의 단쇄 가변부 단편(scFv) 으로 구성되는 융합 단백질이다. scFv 중 하나는 CD3 수용체를 통해 T 세포에 결합하고, 다른 하나는 종양 특이적인 분자를 통해 종양 세포에 결합한다. 다른 이중특이적 항체와 유사하게, 그리고 보통의 모노클로날 항체와는 달리, BiTE는 T 세포와 종양 세포 사이에서 연결을 형성한다. 이는 퍼포린 및 그랜자임과 같은 단백질을 생산함으로써 MHC I 또는 공자극 분자의 존재와는 독립적으로 T 세포가 종양 세포 상에서 세포독성 활성을 발휘하게 한다. 이들 단백질은 종양 세포로 진입하고 세포의 세포사멸을 개시한다.In one embodiment, the T cell engager can be, for example, a bispecific T-cell engager (BiTE). The BiTE is a class of artificial bispecific monoclonal antibodies, which are fusion proteins consisting of amino acid sequences from four different genes or two single chain variable region fragments (scFv) of different antibodies on a single peptide chain of about 55 kilodaltons. One of the scFv binds to the T cell via the CD3 receptor, and the other binds to the tumor cell via a tumor-specific molecule. Similar to other bispecific antibodies, and unlike conventional monoclonal antibodies, the BiTE forms a link between the T cell and the tumor cell. It causes the T cell to exert cytotoxic activity on the tumor cell independently of the presence of MHC I or costimulatory molecules by producing proteins such as perforin and granzymes. These proteins enter the tumor cell and initiate apoptosis of the cell.
일 측면에서, 본 발명은 서열번호 1 또는 서열번호 2의 펩타이드, 본 발명의 키메라 항원 수용체, 본 발명의 키메라 항원 수용체 발현 세포, 또는 본 발명의 T 세포 관여자를 유효성분으로 포함하는 암의 예방 또는 치료용 약학적 조성물에 관한 것이다.In one aspect, the present invention relates to a pharmaceutical composition for preventing or treating cancer, comprising a peptide of SEQ ID NO: 1 or SEQ ID NO: 2, a chimeric antigen receptor of the present invention, a cell expressing a chimeric antigen receptor of the present invention, or a T cell engager of the present invention as an active ingredient.
일 구현예에서, 암은 면역항암제에 대해 비반응성 또는 반응성 암일 수 있으며, 고형암일 수 있으며, 바람직하게는 유방암, 대장암, 또는 췌장암인 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the cancer can be a non-responsive or responsive cancer to an immunotherapy agent, can be a solid cancer, and is preferably, but not limited to, breast cancer, colon cancer, or pancreatic cancer.
일 구현예에서, 상기 조성물은 면역항암제일 수 있다.In one embodiment, the composition may be an immunotherapy agent.
일 구현예에서, 상기 조성물은 화학항암제 또는 항체-약물 접합체(Antibody-Drug Conjugate, ADC)와 병용투여하였을 때, 항암 효과가 상승되는 것을 특징으로 할 수 있으나, 이에 제한되지는 않는다.In one embodiment, the composition may be characterized by, but is not limited to, an enhanced anticancer effect when co-administered with a chemotherapeutic agent or an antibody-drug conjugate (ADC).
일 구현예에서, 상기 항체-약물 접합체(Antibody-Drug Conjugate, ADC)는 PADCEV(anti-Nectin-4 antibody drug conjugate)인 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the antibody-drug conjugate (ADC) may be, but is not limited to, an anti-Nectin-4 antibody drug conjugate (PADCEV).
본 발명의 약학적 조성물은 단독의 요법으로 이용될 수 있으나, 다른 통상적인 생물학적 요법, 화학 요법 또는 방사 요법과 함께 이용될 수도 있으며, 이러한 병행 요법을 실시하는 경우에는 보다 효과적으로 암을 치료할 수 있다. 본 발명을 암의 예방 및 치료에 이용하는 경우 상기 조성물과 함께 이용될 수 있는 화학 요법제는 시스플라틴(cisplatin), 카르보플라틴(carboplatin), 프로카르바진(procarbazine), 메클로레타민(mechlorethamine), 시클로포스파미드(cyclophosphamide), 이포스파미드(ifosfamide), 멜팔란(melphalan), 클로라부실(chlorambucil), 비술판(bisulfan), 니트로소우레아(nitrosourea), 디악티노마이신(dactinomycin), 다우노루비신(daunorubicin), 독소루비신(doxorubicin), 블레오마이신(bleomycin), 플리코마이신(plicomycin), 미토마이신(mitomycin), 에토포시드(etoposide), 탁목시펜(tamoxifen), 택솔(taxol), 트랜스플라티눔(transplatinum), 5-플루오로우라실(5-fluorouracil), 빈크리스틴(vincristin), 빈블라스틴(vinblastin) 및 메토트렉세이트(methotrexate) 등을 포함한다. 본 발명의 조성물과 함께 이용될 수 있는 방사 요법은 X-선 조사 및 γ-선 조사 등이다.The pharmaceutical composition of the present invention can be used as a single therapy, but can also be used in combination with other conventional biological therapies, chemotherapy, or radiotherapy, and when such combination therapy is performed, cancer can be treated more effectively. When the present invention is used for the prevention and treatment of cancer, chemotherapeutic agents that can be used together with the composition include cisplatin, carboplatin, procarbazine, mechlorethamine, cyclophosphamide, ifosfamide, melphalan, chlorambucil, bisulfan, nitrosourea, dactinomycin, daunorubicin, doxorubicin, bleomycin, plicomycin, mitomycin, etoposide, tamoxifen, taxol, transplatinum, 5-fluorouracil, vincristine, vinblastine, and methotrexate, etc. Radiation therapy that can be used with the composition of the present invention includes X-ray irradiation and γ-ray irradiation, etc.
일 구현예에서, 본 발명의 조성물은 면역원성 세포사멸 유도제를 추가로 포함할 수 있으며, 면역원성 세포사멸 유도제는 안트라사이클린계열 항암제, 탁산 계열 항암제, 항-EGFR 항체, BK 채널 작용제, 보르테조밉(Bortezomib), 강심성 배당체(cardiac glycoside), 사이클로포스마이드 계열 항암제, GADD34/PP1 저해제, LV-tSMAC, Measles 바이러스, 블레오마이신(bleomycin), 미토잔트론(mitoxantrone) 또는 옥살리플라틴(oxaliplatin)으로 이루어진 군으로부터 선택되는 어느 하나 이상일 수 있으며, 안트라사이클린 계열 항암제는 다우노루비신(daunorubicin), 독소루비신(doxorubicin), 에피루비신(epirubicin), 이다루비신(idarubicin), 픽산트론(pixantrone), 사바루비신(sabarubicin) 또는 발루비신(valrubicin)일 수 있고, 탁산계열 항암제는 파클리탁셀(paclitaxel) 또는 도세탁셀(docetaxel)일 수 있다.In one embodiment, the composition of the present invention may further comprise an immunogenic apoptosis inducer, wherein the immunogenic apoptosis inducer may be at least one selected from the group consisting of an anthracycline series anticancer agent, a taxane series anticancer agent, an anti-EGFR antibody, a BK channel agonist, bortezomib, a cardiac glycoside, a cyclophosphamide series anticancer agent, a GADD34/PP1 inhibitor, LV-tSMAC, Measles virus, bleomycin, mitoxantrone, or oxaliplatin, and the anthracycline series anticancer agent may be daunorubicin, doxorubicin, epirubicin, idarubicin, pixantrone, sabarubicin, or It could be valrubicin, and the taxane family of chemotherapy agents could be paclitaxel or docetaxel.
본 발명의 암의 예방 또는 치료용 약학적 조성물은 화학적 항암 약물(항암제) 등과 함께 투여함으로써, 암세포의 사멸 효과를 통해 종래의 항암제의 암치료 효과를 증가시킬 수 있다. 병용 투여는 상기 항암제와 동시에 또는 순차적으로 이루어질 수 있다. 상기 항암제의 예시에는 DNA 알킬화제(DNA alkylating agents)로 메클로에타민(mechloethamine), 클로람부칠(chlorambucil), 페닐알라닌(phenylalanine), 무스타드(mustard), 사이클로포스파미드(cyclophosphamide), 이포스파미드(ifosfamide), 카르무스틴(carmustine: BCNU), 로무스틴(lomustine: CCNU), 스트렙토조토신(streptozotocin), 부술판(busulfan), 티오테파(thiotepa), 시스플라틴(cisplatin) 및 카보플라틴(carboplatin); 항암 항생제(anti-cancer antibiotics)로 닥티노마이신(dactinomycin: actinomycin D), 플리카마이신(plicamycin) 및 마이토마이신 C(mitomycin C); 및 식물 알카로이드(plant alkaloids)로 빈크리스틴(vincristine), 빈블라스틴(vinblastine), 에토포시드(etoposide), 테니포시드(teniposide), 토포테칸(topotecan) 및 이리도테칸(iridotecan) 등이 포함되지만, 이에 한정되는 것은 아니다.The pharmaceutical composition for preventing or treating cancer of the present invention can increase the cancer treatment effect of conventional anticancer drugs through the cancer cell killing effect by administering it together with a chemical anticancer drug (anticancer agent), etc. The combined administration can be performed simultaneously with or sequentially with the anticancer agent. Examples of the anticancer agent include DNA alkylating agents such as mechloethamine, chlorambucil, phenylalanine, mustard, cyclophosphamide, ifosfamide, carmustine (BCNU), lomustine (CCNU), streptozotocin, busulfan, thiotepa, cisplatin, and carboplatin; Anti-cancer antibiotics include, but are not limited to, dactinomycin (actinomycin D), plicamycin, and mitomycin C; and plant alkaloids include, but are not limited to, vincristine, vinblastine, etoposide, teniposide, topotecan, and iridotecan.
본 발명에서, 용어 "예방"이란 본 발명에 따른 약학적 조성물의 투여에 의해 암의 발생, 확산 및 재발을 억제 또는 지연시키는 모든 행위를 의미한다.In the present invention, the term “prevention” means any act of inhibiting or delaying the occurrence, spread, and recurrence of cancer by administering a pharmaceutical composition according to the present invention.
본 발명에서 사용되는 용어 "치료"란 본 발명의 조성물의 투여로 암세포의 사멸 또는 암의 증세를 호전시키거나 이롭게 변경하는 모든 행위를 의미한다. 본 발명이 속하는 기술분야에서 통상의 지식을 가진 자라면, 대한의학협회 등에서 제시된 자료를 참조하여 본원의 조성물이 효과가 있는 질환의 정확한 기준을 알고, 개선, 향상 및 치료된 정도를 판단할 수 있을 것이다.The term "treatment" used in the present invention means any act of killing cancer cells or improving or beneficially changing the symptoms of cancer by administering the composition of the present invention. Anyone with ordinary knowledge in the technical field to which the present invention pertains will be able to refer to materials presented by the Korean Medical Association, etc. to know the exact criteria for diseases to which the composition of the present invention is effective, and to determine the degree of improvement, enhancement, and treatment.
본 발명에서 유효성분과 결합하여 사용된 "치료학적으로 유효한 양"이란 용어는 대상 질환을 예방 또는 치료하는데 유효한 조성물의 약학적으로 허용가능한 염의 양을 의미하며, 본 발명의 조성물의 치료적으로 유효한 양은 여러 요소, 예를 들면 투여방법, 목적부위, 환자의 상태 등에 따라 달라질 수 있다. 따라서, 인체에 사용 시 투여량은 안전성 및 효율성을 함께 고려하여 적정량으로 결정되어야 한다. 동물실험을 통해 결정한 유효량으로부터 인간에 사용되는 양을 추정하는 것도 가능하다. 유효한 양의 결정시 고려할 이러한 사항은, 예를 들면 Hardman and Limbird, eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed.(2001), Pergamon Press; 및 E.W. Martin ed., Remington's Pharmaceutical Sciences, 18th ed.(1990), Mack Publishing Co.에 기술되어 있다.The term "therapeutically effective amount" used in combination with the active ingredient in the present invention means the amount of a pharmaceutically acceptable salt of the composition effective for preventing or treating a target disease, and the therapeutically effective amount of the composition of the present invention may vary depending on various factors, such as the administration method, the target site, the condition of the patient, etc. Therefore, the dosage when used in humans should be determined as an appropriate amount by considering both safety and efficacy. It is also possible to estimate the amount used in humans from the effective amount determined through animal testing. Such considerations when determining the effective amount are described in, for example, Hardman and Limbird, eds., Goodman and Gilman's The Pharmacological Basis of Therapeutics, 10th ed.(2001), Pergamon Press; and E.W. Martin ed., Remington's Pharmaceutical Sciences, 18th ed.(1990), Mack Publishing Co.
본 발명의 약학적 조성물은 약학적으로 유효한 양으로 투여한다. 본 발명에서 사용되는 용어, "약학적으로 유효한 양"은 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미하며, 유효용량 수준은 환자의 건강상태, 암의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료기간, 배합 또는 동시 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 결정될 수 있다. 본 발명의 조성물은 개별 치료제로 투여하거나 다른 치료제와 병용하여 투여될 수 있고, 종래의 치료제와 순차적으로 또는 동시에 투여될 수 있으며, 단일 또는 다중 투여될 수 있다. 상기한 요소들을 모두 고려하여, 부작용없이 최소한의 양으로 최대 효과를 얻을 수 있는 양을 투여하는 것이 중요하며, 이는 당업자에 의해 용이하게 결정될 수 있다.The pharmaceutical composition of the present invention is administered in a pharmaceutically effective amount. The term "pharmaceutically effective amount" as used in the present invention means an amount sufficient to treat a disease at a reasonable benefit/risk ratio applicable to medical treatment and not causing side effects, and the effective dosage level can be determined according to factors including the patient's health condition, the type and severity of cancer, the activity of the drug, the sensitivity to the drug, the administration method, the administration time, the administration route and the excretion rate, the treatment period, the drug used in combination or simultaneously, and other factors well known in the medical field. The composition of 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. Considering all of the above factors, it is important to administer an amount that can obtain the maximum effect with the minimum amount without side effects, and this can be easily determined by those skilled in the art.
본 발명의 약학적 조성물은 약학적으로 허용 가능한 첨가제를 더 포함할 수 있으며, 이때 약학적으로 허용 가능한 첨가제로는 전분, 젤라틴화 전분, 미결정셀룰로오스, 유당, 포비돈, 콜로이달실리콘디옥사이드, 인산수소칼슘, 락토스, 만니톨, 엿, 아라비아고무, 전호화전분, 옥수수전분, 분말셀룰로오스, 히드록시프로필셀룰로오스, 오파드라이, 전분글리콜산나트륨, 카르나우바 납, 합성규산알루미늄, 스테아린산, 스테아린산마그네슘, 스테아린산알루미늄, 스테아린산칼슘, 백당, 덱스트로스, 소르비톨 및 탈크 등이 사용될 수 있다. 본 발명에 따른 약학적으로 허용 가능한 첨가제는 상기 조성물에 대해 0.1 중량부 내지 90 중량부 포함되는 것이 바람직하나, 이에 한정되는 것은 아니다.The pharmaceutical composition of the present invention may further contain a pharmaceutically acceptable additive. At this time, the pharmaceutically acceptable additive may include starch, gelatinized starch, microcrystalline cellulose, lactose, povidone, colloidal silicon dioxide, calcium hydrogen phosphate, lactose, mannitol, taffy, gum arabic, pregelatinized starch, corn starch, powdered cellulose, hydroxypropyl cellulose, opadry, sodium starch glycolate, carnauba wax, synthetic aluminum silicate, stearic acid, magnesium stearate, aluminum stearate, calcium stearate, sucrose, dextrose, sorbitol, and talc. The pharmaceutically acceptable additive according to the present invention is preferably contained in an amount of 0.1 to 90 parts by weight with respect to the composition, but is not limited thereto.
본 발명의 조성물은 또한 생물학적 제제에 통상적으로 사용되는 담체, 희석제, 부형제 또는 둘 이상의 이들의 조합을 포함할 수 있다. 약학적으로 허용 가능한 담체는 조성물을 생체 내 전달에 적합한 것이면 특별히 제한되지 않으며, 예를 들면, Merck Index, 13th ed., Merck & Co. Inc. 에 기재된 화합물, 식염수, 멸균수, 링거액, 완충 식염수, 덱스트로스 용액, 말토 덱스트린 용액, 글리세롤, 에탄올 및 이들 성분 중 1 성분 이상을 혼합하여 이용할 수 있으며, 필요에 따라 항산화제, 완충액, 정균제 등 다른 통상의 첨가제를 첨가할 수 있다. 또한, 희석제, 분산제, 계면활성제, 결합제 및 윤활제를 부가적으로 첨가하여 수용액, 현탁액, 유탁액 등과 같은 주이용 제형, 환약, 캡슐, 과립 또는 정제로 제제화할 수 있다. 더 나아가 당 분야의 적정한 방법으로 또는 Remington's Pharmaceutical Science(Mack Publishing Company, Easton PA, 18th, 1990)에 개시되어 있는 방법을 이용하여 각 질환에 따라 또는 성분에 따라 바람직하게 제제화할 수 있다.The composition of the present invention may also include a carrier, a diluent, an excipient or a combination of two or more thereof commonly used in biological preparations. The pharmaceutically acceptable carrier is not particularly limited as long as it is suitable for delivering the composition in vivo, and for example, compounds described in Merck Index, 13th ed., Merck & Co. Inc., saline, sterile water, Ringer's solution, buffered saline, dextrose solution, maltodextrin solution, glycerol, ethanol and one or more of these components may be mixed and used, and other common additives such as antioxidants, buffers, and bacteriostatic agents may be added as necessary. In addition, a diluent, a dispersant, a surfactant, a binder and a lubricant may be additionally added to formulate the composition into a main-use dosage form such as an aqueous solution, a suspension, an emulsion, a pill, a capsule, a granule or a tablet. Furthermore, it can be preferably formulated according to each disease or ingredient using an appropriate method in the field or the method disclosed in Remington's Pharmaceutical Science (Mack Publishing Company, Easton PA, 18th, 1990).
본 발명의 조성물은 목적하는 방법에 따라 비 경구 투여(예를 들어 정맥 내, 피하, 복강 내 또는 국소에 주사 제형으로 적용)하거나 경구 투여할 수 있으며, 투여량은 환자의 체중, 연령, 성별, 건강상태, 식이, 투여시간, 투여방법, 배설률 및 질환의 중증도 등에 따라 그 범위가 다양하다. 본 발명에 따른 조성물의 일일 투여량은 0.0001 ~ 10 ㎎/㎖이며, 바람직하게는 0.0001 ~ 5 ㎎/㎖이며, 하루 일 회 내지 수회에 나누어 투여하는 것이 더욱 바람직하다. The composition of the present invention can be administered non-orally (for example, intravenously, subcutaneously, intraperitoneally, or locally as an injection formulation) or orally, depending on the intended method, and the dosage range varies depending on the patient's weight, age, sex, health condition, diet, administration time, administration method, excretion rate, and severity of disease. The daily dosage of the composition according to the present invention is 0.0001 to 10 mg/ml, preferably 0.0001 to 5 mg/ml, and it is more preferable to administer once a day or in divided doses several times.
본 발명의 조성물의 경구 투여를 위한 액상 제제로는 현탁제, 내용액제, 유제, 시럽제 등이 해당되는데, 통상적으로 사용되는 단순 희석제인 물, 액체 파라핀 이외에 다양한 부형제, 예컨대 습윤제, 감미제, 방향제, 보존제 등이 함께 포함될 수 있다. 비경구 투여를 위한 제제에는 멸균된 수용액, 비수성 용제, 현탁제, 유제, 동결건조 제제, 좌제 등이 포함된다.Liquid preparations for oral administration of the composition of the present invention include suspensions, solutions, emulsions, syrups, etc., and in addition to commonly used simple diluents such as water and liquid paraffin, various excipients such as wetting agents, sweeteners, flavoring agents, preservatives, etc. may be included. Preparations for parenteral administration include sterile aqueous solutions, non-aqueous solvents, suspensions, emulsions, lyophilized preparations, suppositories, etc.
일 측면에서, 본 발명은 본 발명의 서열번호 1 또는 서열번호 2의 펩타이드, 본 발명의 키메라 항원 수용체, 본 발명의 키메라 항원 수용체 발현 세포, 또는 본 발명의 T 세포 관여자; 및 면역항암제, 화학항암제 또는 항체-약물 접합체(Antibody-Drug Conjugate, ADC);를 포함하는 암의 예방 또는 치료용 조합물에 관한 것이다.In one aspect, the present invention relates to a combination for preventing or treating cancer, comprising a peptide having
일 구현예에서, 면역항암제는 면역관문 억제제(Immune checkpoint inhibitors), 면역억제인자 제어 약물, 암 백신, 면역아주번트(immunoadjuvant), 암 치료용 면역세포, 면역세포 활성 보조인자, 암 치료용 항체 또는 암 치료용 면역세포의 활성 유지에 필요한 사이토카인일 수 있다.In one embodiment, the immunotherapy agent may be an immune checkpoint inhibitor, an immunosuppressant modulating drug, a cancer vaccine, an immunoadjuvant, an immune cell for cancer treatment, an immune cell activation cofactor, an antibody for cancer treatment, or a cytokine required for maintaining the activity of an immune cell for cancer treatment.
일 구현예에서, 면역관문 억제제는 CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, BTLA, B7H3, B7H4, TIM3, KIR, TIGIT, CD47, VISTA 또는 A2aR의 억제제일 수 있으며, 항-PD-1 항체, 항-PDL-1 항체, 항-CTLA4, 또는 PD-1, PDL-1, 또는 CTLA4에 특이적으로 결합하는 상기 항체의 항원-결합 단편, 또는 이들의 조합물일 수 있다.In one embodiment, the immune checkpoint inhibitor can be an inhibitor of CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, BTLA, B7H3, B7H4, TIM3, KIR, TIGIT, CD47, VISTA or A2aR, an anti-PD-1 antibody, an anti-PDL-1 antibody, an anti-CTLA4, or an antigen-binding fragment of such an antibody that specifically binds to PD-1, PDL-1, or CTLA4, or a combination thereof.
일 구현예에서, 면역관문 억제제는 항-PD1 항체인 니볼루맙 또는 펨브롤리주맙, 항-PD-L1 항체인 아테졸리주맙, 아벨루맙 또는 더발루맙, 항-CTLA-4 항체인 트레멜리무맙 또는 이필리무맙, 또는 이들의 조합물일 수 있다.In one embodiment, the immune checkpoint inhibitor can be an anti-PD1 antibody nivolumab or pembrolizumab, an anti-PD-L1 antibody atezolizumab, avelumab or durvalumab, an anti-CTLA-4 antibody tremelimumab or ipilimumab, or a combination thereof.
일 구현예에서, 상기 조합물은 화학치료제, 또는 방사선을 추가로 포함할 수 있다.In one embodiment, the combination may additionally comprise a chemotherapeutic agent, or radiation.
일 구현예에서 상기 화학치료제는 전세포사멸성(pro-apoptotic) 펩타이드, 면역원성 세포사멸 유도제 또는 항암제일 수 있으며, SN-38(7-에틸-10-히드록시-캠토테신, 7-Ethyl-10-hydroxy-camptothecin), 다우노루비신(daunorubicin), 독소루비신(doxorubicin), 에피루비신(epirubicin), 이다루비신(idarubicin), 픽산트론(pixantrone), 사바루비신(sabarubicin), 발루비신(valrubicin), 파클리탁셀(paclitaxel), 도세탁셀(docetaxel), 메클로에타민(mechloethamine), 클로람부실(chlorambucil), 페닐알라닌(phenylalanine), 무스타드(mustard), 사이클로포스파미드(cyclophosphamide), 이포스파미드(ifosfamide), 카르무스틴(carmustine: BCNU), 로무스틴(lomustine: CCNU), 스트렙토조토신(streptozotocin), 부설판(busulfan), 티오테파(thiotepa), 시스플라틴(cisplatin), 카보플라틴(carboplatin), 닥티노마이신(dactinomycin: actinomycin D), 플리카마이신(plicamycin), 마이토마이신 C(mitomycin C), 빈크리스틴(vincristine), 빈블라스틴(vinblastine), 테니포사이드(teniposide), 토포테칸(topotecan), 이리도테칸(iridotecan), 우라무스틴(uramustine), 멜파란(melphalan), 벤다무스틴(bendamustine), 다카바진(dacarbazine), 테모졸로마이드(temozolomide), 알트레타민(altretamine), 듀오카르마이신(duocarmycin), 네다플라틴(nedaplatin), 옥사리플라틴(oxaliplatin), 사트라플라틴(satraplatin), 트리플라틴 테트라나이트레이트(triplatin tetranitrate), 5-플루오로우라실(5-fluorouracil), 6-머캅토퓨린(6-mercaptopurine), 카페시타빈(capecitabine), 클라드리빈(cladribine), 클로파라빈(clofarabine), 시스타르빈(cystarbine), 플록스유리딘(floxuridine), 플루다라빈(fludarabine), 겜시타빈(gemcitabine), 하이드록시우레아(hydroxyurea), 메토트렉세이트(methotrexate), 페메트렉세드(pemetrexed), 펜토스타틴(pentostatin), 티오구아닌(thioguanine), 에토포사이드(etoposide), 미토산트론(mitoxantrone), 이자베필론(izabepilone), 빈데신(vindesine), 비노렐빈(vinorelbine), 에스트라머스틴(estramustine), 메이탄신(maytansine), DM1(mertansine, 메르탄신), DM4, 돌라스타틴(dolastatin), 아우리스타틴 E(auristatin E), 아우리스타틴 F(auristatin F), 모노메틸 아우리스타틴 E(monomethyl auristatin E, MMAE), 모노메틸 아우리스타틴 F(monomethyl auristatin F) 및 이들의 유도체로 이루어진 군으로부터 선택될 수 있으며, 바람직하게는 옥살리플라틴(Oxaliplatin), 페메트렉시드 (Pemetrexed), 시스플라틴(Cisplatin), 젬시타빈(Gemcitabine), 카보플라틴(Carboplatin), 플루오로우라실(5-FU), 시클로포스파미드(Cyclophosphamide), 파클리탁셀(Paclitaxel), 빈크리스틴(Vincristine), 에토포사이드(Etoposide) 및 독소루비신(Doxorubicin)으로 이루어진 군에서 선택된 하나 이상의 것일 수 있고, 가장 바람직하게는 옥살리플라틴(Oxaliplatin) 또는 파클리탁셀(Paclitaxel)일 수 있다.In one embodiment, the chemotherapeutic agent may be a pro-apoptotic peptide, an immunogenic apoptosis inducer or an anticancer agent, and may be selected from the group consisting of SN-38 (7-Ethyl-10-hydroxy-camptothecin), daunorubicin, doxorubicin, epirubicin, idarubicin, pixantrone, sabarubicin, valrubicin, paclitaxel, docetaxel, mechloethamine, chlorambucil, phenylalanine, mustard, cyclophosphamide, Ifosfamide, carmustine (BCNU), lomustine (CCNU), streptozotocin, busulfan, thiotepa, cisplatin, carboplatin, dactinomycin (actinomycin D), plicamycin, mitomycin C, vincristine, vinblastine, teniposide, topotecan, iridotecan, uramustine, melphalan, bendamustine, dacarbazine, temozolomide, altretamine, Duocarmycin, nedaplatin, oxaliplatin, satraplatin, triplatin tetranitrate, 5-fluorouracil, 6-mercaptopurine, capecitabine, cladribine, clofarabine, cystarbine, floxuridine, fludarabine, gemcitabine, hydroxyurea, methotrexate, pemetrexed, pentostatin, thioguanine, etoposide, mitoxantrone, It may be selected from the group consisting of izabepilone, vindesine, vinorelbine, estramustine, maytansine, DM1 (mertansine), DM4, dolastatin, auristatin E, auristatin F, monomethyl auristatin E (MMAE), monomethyl auristatin F, and derivatives thereof, preferably oxaliplatin, pemetrexed, cisplatin, gemcitabine, carboplatin, fluorouracil (5-FU), cyclophosphamide, paclitaxel, It may be one or more selected from the group consisting of Vincristine, Etoposide and Doxorubicin, most preferably Oxaliplatin or Paclitaxel.
일 구현예에서, 상기 항체-약물 접합체는 PADCEV(anti-Nectin-4 antibody drug conjugate)인 것일 수 있다.In one embodiment, the antibody-drug conjugate may be an anti-Nectin-4 antibody drug conjugate (PADCEV).
본 발명의 PADCEV는 항체-약물 접합체(ADC: Antibody-Drug Conjugate)이다. 구체적으로, PADCEV는 FDA에서 승인된 엔포투맙 베도틴(enfortumab vedotin)이라는 약물로, 방광암을 포함한 요로상피암(urothelial carcinoma) 치료에 사용된다.The PADCEV of the present invention is an antibody-drug conjugate (ADC). Specifically, PADCEV is a drug called enfortumab vedotin, which is approved by the FDA and is used to treat urothelial carcinoma, including bladder cancer.
Nectin-4는 방광암 세포와 같은 암세포에서 과발현되는 단백질이며, 항Nectin-4 항체가 Nectin-4에 결합하면, 약물이 암세포 내부로 들어가게된다. 세포 내부에서 MMAE가 방출되어 미세소관(microtubule)을 파괴함으로써 암세포를 사멸시키는 작용을 한다. 대게의 경우 요로상피암 환자에 사용하며, 특히 이전의 면역치료제(예: PD-1/PD-L1 억제제)나 화학요법에 반응하지 않는 환자들에게 사용하는 것이 일반적이다.Nectin-4 is a protein that is overexpressed in cancer cells such as bladder cancer cells, and when anti-Nectin-4 antibodies bind to Nectin-4, the drug enters the cancer cells. MMAE is released inside the cells, destroying microtubules and killing cancer cells. It is usually used for patients with urothelial cancer, and is especially common for patients who do not respond to previous immunotherapy (e.g., PD-1/PD-L1 inhibitors) or chemotherapy.
일 구현예에서, 본 발명의 서열번호 1 또는 서열번호 2의 펩타이드, 본 발명의 키메라 항원 수용체, 본 발명의 키메라 항원 수용체 발현 세포, 또는 본 발명의 T 세포 관여자;와 면역항암제, 화학항암제 또는 항체-약물 접합체(Antibody-Drug Conjugate, ADC);는 동시에, 별도로, 또는 순차적으로 병용 투여될 수 있다.In one embodiment, the peptide of SEQ ID NO: 1 or SEQ ID NO: 2 of the present invention, the chimeric antigen receptor of the present invention, the chimeric antigen receptor expressing cell of the present invention, or the T cell engager of the present invention; and an immunotherapy agent, a chemotherapeutic agent, or an antibody-drug conjugate (ADC); may be administered simultaneously, separately, or sequentially.
일 측면에서, 본 발명은 본 발명의 서열번호 1 또는 서열번호 2의 펩타이드, 본 발명의 키메라 항원 수용체, 본 발명의 키메라 항원 수용체 발현 세포, 또는 본 발명의 T 세포 관여자를 유효성분으로 포함하는 항암 보조제에 관한 것이다.In one aspect, the present invention relates to an anticancer adjuvant comprising a peptide of the present invention having
일 구현예에서, 본 발명의 항암 보조제는 면역항암제와 동시에, 별도로, 또는 순차적으로 병용 투여될 수 있다.In one embodiment, the anticancer adjuvant of the present invention may be administered concurrently, separately, or sequentially with an immunotherapy agent.
일 구현예에서, 면역항암제는 면역관문 억제제(Immune checkpoint inhibitors), 면역억제인자 제어 약물, 암 백신, 면역아주번트(immunoadjuvant), 암 치료용 면역세포, 면역세포 활성 보조인자, 암 치료용 항체 또는 암 치료용 면역세포의 활성 유지에 필요한 사이토카인일 수 있다.In one embodiment, the immunotherapy agent may be an immune checkpoint inhibitor, an immunosuppressant modulating drug, a cancer vaccine, an immunoadjuvant, an immune cell for cancer treatment, an immune cell activation cofactor, an antibody for cancer treatment, or a cytokine required for maintaining the activity of an immune cell for cancer treatment.
일 구현예에서, 면역관문 억제제는 CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, BTLA, B7H3, B7H4, TIM3, KIR, TIGIT, CD47, VISTA 또는 A2aR의 억제제일 수 있으며, 항-PD-1 항체, 항-PDL-1 항체, 항-CTLA4, 또는 PD-1, PDL-1, 또는 CTLA4에 특이적으로 결합하는 상기 항체의 항원-결합 단편, 또는 이들의 조합물일 수 있다.In one embodiment, the immune checkpoint inhibitor can be an inhibitor of CTLA-4, PD-1, PD-L1, PD-L2, LAG-3, BTLA, B7H3, B7H4, TIM3, KIR, TIGIT, CD47, VISTA or A2aR, an anti-PD-1 antibody, an anti-PDL-1 antibody, an anti-CTLA4, or an antigen-binding fragment of such an antibody that specifically binds to PD-1, PDL-1, or CTLA4, or a combination thereof.
일 구현예에서, 면역관문 억제제는 항-PD1 항체인 니볼루맙 또는 펨브롤리주맙, 항-PD-L1 항체인 아테졸리주맙, 아벨루맙 또는 더발루맙, 항-CTLA-4 항체인 트레멜리무맙 또는 이필리무맙, 또는 이들의 조합물일 수 있다.In one embodiment, the immune checkpoint inhibitor can be an anti-PD1 antibody nivolumab or pembrolizumab, an anti-PD-L1 antibody atezolizumab, avelumab or durvalumab, an anti-CTLA-4 antibody tremelimumab or ipilimumab, or a combination thereof.
일 구현예에서, 본 발명의 약학적 조성물 또는 조합물의 대상이 되는 암은 난소암, 유방암, 폐암, 비소세포성 폐암, 위암, 간암, 전립선암, 두경부암, 방광암, 대장암, 결장암, 췌장암, 신장암, 골수암, 자궁암, 멜라닌종, 뇌암 및 갑상선암으로 이루어진 군으로부터 선택되는 것일 수 있으나, 바람직하게는 유방암, 폐암, 비소세포성 폐암, 간암, 대장암, 결장암, 췌장암 및 뇌암으로 이루어진 군에서 선택되는 것일 수 있으며, 가장 바람직하게는 유방암, 대장암 또는 유방암인 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the cancer targeted by the pharmaceutical composition or combination of the present invention may be selected from the group consisting of ovarian cancer, breast cancer, lung cancer, non-small cell lung cancer, stomach cancer, liver cancer, prostate cancer, head and neck cancer, bladder cancer, colon cancer, colon cancer, pancreatic cancer, kidney cancer, bone marrow cancer, uterine cancer, melanoma, brain cancer and thyroid cancer, but is preferably selected from the group consisting of breast cancer, lung cancer, non-small cell lung cancer, liver cancer, colon cancer, colon cancer, pancreatic cancer and brain cancer, and is most preferably breast cancer, colon cancer or breast cancer, but is not limited thereto.
일 구현예에서, 본 발명의 약학적 조성물 또는 조합물은 TB511을 본 발명의 약학적 조성물 또는 조합물은 TB511을 100 내지 300 nmol/kg의 농도로 투여되는것일 수 있으며, 옥살리플라틴은 1 내지 2 mg/kg 농도로 투여되는 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the pharmaceutical composition or combination of the present invention may be administered at a concentration of TB511 of 100 to 300 nmol/kg, and oxaliplatin may be administered at a concentration of 1 to 2 mg/kg, but is not limited thereto.
일 구현예에서, 본 발명의 약학적 조성물 또는 조합물은 TB511을 본 발명의 약학적 조성물 또는 조합물은 TB511을 100 내지 300 nmol/kg의 농도로 투여되는것일 수 있으며, 파클리탁셀은 3 내지 7 mg/kg 농도로 투여되는 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the pharmaceutical composition or combination of the present invention may be administered at a concentration of TB511 of 100 to 300 nmol/kg, and paclitaxel may be administered at a concentration of 3 to 7 mg/kg, but is not limited thereto.
일 구현예에서, 본 발명의 약학적 조성물 또는 조합물은 TB511을 본 발명의 약학적 조성물 또는 조합물은 TB511을 100 내지 300 nmol/kg의 농도로 투여되는것일 수 있으며, PADCEV(anti-Nectin-4 antibody drug conjugate)은 3 내지 7 mg/kg 농도로 투여되는 것일 수 있으나, 이에 제한되지는 않는다.In one embodiment, the pharmaceutical composition or combination of the present invention may be administered at a concentration of TB511 of 100 to 300 nmol/kg, and PADCEV (anti-Nectin-4 antibody drug conjugate) may be administered at a concentration of 3 to 7 mg/kg, but is not limited thereto.
일 측면에서, 본 발명은 본 발명의 서열번호 1 또는 서열번호 2의 펩타이드, 본 발명의 키메라 항원 수용체, 본 발명의 키메라 항원 수용체 발현 세포, 또는 본 발명의 T 세포 관여자를 유효성분으로 포함하는 암 진단용 조성물에 관한 것이다.In one aspect, the present invention relates to a composition for diagnosing cancer, comprising a peptide of
일 구현예에서, 상기 조성물은 표지를 추가로 포함할 수 있으며, 표지는 발색효소, 방사성 동위원소, 크로모포어(chromopore), 발광물질, 형광물질, 프로브 또는 택(tag)일 수 있고, 형광물질은 Cy(cyanine) 계열, 로다민(Rhodamine) 계열, 알렉사(Alexa) 계열, BODIPY 계열 또는 ROX 계열의 형광물질일 수 있으며, 나일 레드 (Nile Red), 보디피 (BODIPY, 4,4-difluoro-4-bora-3a,4a-diaza-s-indacene), 시아닌 (cyanine), 플루오레세인 (fluorescein), 로다민 (rhodamine), 쿠마린 (coumarine) 또는 알렉사 (Alexa)일 수 있다.In one embodiment, the composition can further comprise a label, and the label can be a chromogenic enzyme, a radioisotope, a chromopore, a luminescent substance, a fluorescent substance, a probe or a tag, and the fluorescent substance can be a fluorescent substance of the Cy (cyanine) series, Rhodamine series, Alexa series, BODIPY series or ROX series, and can be Nile Red, BODIPY (4,4-difluoro-4-bora-3a,4a-diaza-s-indacene), cyanine, fluorescein, rhodamine, coumarine or Alexa.
일 측면에서, 본 발명은 본 발명의 약학적 조성물 또는 조합물을 개체에게 투여하는 단계를 포함하는 암의 예방 또는 치료방법에 관한 것이다.In one aspect, the present invention relates to a method for preventing or treating cancer, comprising administering to a subject a pharmaceutical composition or combination of the present invention.
본 발명에서 사용되는 용어 "개체"란 질병의 예방, 조절 또는 치료방법을 필요로 하는 대상을 의미하고, 사람, 개, 원숭이, 고양이, 설치류, 예컨대 마우스, 유전자 조작된 마우스 등 제한 없이 사용될 수 있다. 보다 구체적으로는, 인간 또는 비-인간인 영장류, 생쥐 (mouse), 쥐 (rat), 개, 고양이, 말, 소 등의 포유류를 의미한다.The term "subject" used in the present invention means a subject that requires a method for preventing, controlling or treating a disease, and can be used without limitation as a human, dog, monkey, cat, rodent, such as a mouse, a genetically engineered mouse, etc. More specifically, it means a mammal, such as a human or non-human primate, mouse, rat, dog, cat, horse, cow, etc.
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. TB511 펩타이드 합성Example 1. Synthesis of TB511 peptide
종양미세환경(tumor microenvironment, TME)에서 면역억제 환경을 구축하여 종양 진행에 기여하고, 주로 M2-유사 표현형으로 존재하는 종양-관련 대식세포(Tumor-associated macrophages, TAM)를 표적화하여 사멸시키는 펩타이드를 합성하기 위해, TAM 표적화 펩타이드인 TAMpep (서열번호 2)과 세포사멸 촉진(pro-apoptotic) 펩타이드인 dKLA (서열번호 3)를 포함하고, 이들의 상호작용 및 폴드를 최소화하기 위한 4개의 글리신 및 1개의 세린으로 구성된 링커를 사이에 배치한 TB511 펩타이드 (서열번호 1)를 GenScript (Piscataway, NJ, USA)에서 합성하여 정제하였다. 이 때, KLA는 체내 분해를 최소화 하기 위해 L형이 아닌 D형 이성질체를 사용하였다. 또한, FITC 또는 Cy5(cyanine 5)는 펩타이드의 N-말단에서 아마이드 결합을 통해 접합하였고, 비오틴은 펩타이드의 C-말단의 라이신 잔기에서 아마이드 결합으로 접합하였다. 또한, 펩타이드의 아미노 말단에 PEG화(PEGylation)하였다. 모든 펩타이드는 95% 이상의 순도로 정제되었다. TB511의 페이로드인 d(KLA)는 음으로 하전된 미토콘드리아 막에 결합하여 이를 파괴할 수 있는 양이온성(cationic) 및 양친매성(amphipathic) α-나선 펩타이드이며, 양쪽 이온성(zwitterionic) 진핵세포를 통과할 수 없으므로 진핵세포에는 독성을 나타내지 않는다. To synthesize a peptide that targets and kills tumor-associated macrophages (TAMs), which mainly exist in an M2-like phenotype and contribute to tumor progression by establishing an immunosuppressive environment in the tumor microenvironment (TME), a TAM-targeting peptide, TAMpep (SEQ ID NO: 2) and a pro-apoptotic peptide, dKLA (SEQ ID NO: 3), with a linker consisting of four glycines and one serine to minimize their interaction and folding, TB511 peptide (SEQ ID NO: 1) was synthesized and purified by GenScript (Piscataway, NJ, USA). In this case, the D-isomer, not the L-form, of KLA was used to minimize degradation in the body. In addition, FITC or Cy5 (cyanine 5) was conjugated via an amide bond at the N-terminus of the peptide, and biotin was conjugated via an amide bond at the lysine residue of the C-terminus of the peptide. In addition, PEGylation was performed at the amino terminus of the peptide. All peptides were purified with a purity of more than 95%. The payload of TB511, d(KLA), is a cationic and amphipathic α-helical peptide that can bind to and disrupt negatively charged mitochondrial membranes, and cannot pass through zwitterionic eukaryotic cells, so it does not exhibit toxicity to eukaryotic cells.
실시예 2. TB511의 표적 단백질 분석Example 2. Target protein analysis of TB511
TB511의 표적 단백질을 확인하기 위해, THP-1 단핵구(monocyte) 세포를 M0, M1 또는 M2 대식세포로 분화시키고 단백질체학(Proteomic analysis) 분석을 수행하였다. 구체적으로, THP-1를 100 nM PMA (phorbol 12-myristate 13-acetate; Sigma-Aldrich, St. Louis, MO, USA)와 함께 완전 배지(complete media)에서 24시간 동안 배양하여 M0 대식세포로 분화시켰다. 또한, M1 또는 M2 세포로 분극화(polarize)하기 위해, M0 세포를 5% FBS로 보충된 RPMI 1640 배지에서 20 ng/mL 재조합 인간 인터페론-감마(recombinant human interferon-gamma, rhIFN-γ; Prospec, Rehovot, Israel) 및 100 ng/mL LPS(lipopolysaccharides; Sigma-Aldrich), 또는 20 ng/mL 재조합 인간 인터루킨-4(recombinant human interleukin-4, rhIL-4; Prospec) 및 20 μg/mL rhIL-13 (Prospec)와 함께 72시간 동안 배양하였다. 또한, PC3 또는 A549 세포를 완전 배지에서 80% 합류에 도달할 때까지 성장시킨 다음 세포를 세척하고 무혈청 배지로 24시간 동안 배양한 뒤, 0.2 μm 주사기 필터 (Sartorius, Gottingen, ermany)로 여과하여 종양 조절 배지(tumor-conditioned medium, TCM)를 제조하고, M0 세포를 72시간 동안 10% 또는 50% 종양 조절 배지와 함께 배양하여 M2-TAM으로 분극화하였다. 이 후, M0, M1 및 M2로 분화된 THP-1 대식세포를 Mem-PER쪠 Plus Membrane Protein Extraction Kit (Thermo Fisher Scientific, Waltham, MA, USA)로 파쇄한 뒤, 세포 파쇄물을 실온에서 1시간 동안 비오틴화된 TAMpep와 함께 인큐베이션하였다. 스트렙타비딘 아가로스 레진 (Thermo Fisher Scientific)과 실온에서 30분 동안 인큐베이션한 뒤, 레진을 세척하고 막-결합된 단백질을 레진으로부터 용출하였다. 용출된 단백질 시료를 Q-Exactive 질량 분석기 (Thermo Fisher Scientific)를 사용하여 LC-MS/MS로 분석하였다. 분석된 각 시료의 LC-MS/MS 데이터를 Proteome Discoverer를 이용하여 분석하고, Label-Free Quantification를 위해 UniProt 인간 데이터베이스를 사용하였다. ExDEGA(Excel-based Differentially Expressed Gene Analysis) 툴 (eBiogen, Seoul, Korea)을 이용하여 단백질체학 분석을 수행하였다 (도 1).To identify the target protein of TB511, THP-1 monocyte cells were differentiated into M0, M1, or M2 macrophages, and proteomic analysis was performed. Specifically, THP-1 was differentiated into M0 macrophages by culturing in complete media with 100 nM PMA (phorbol 12-myristate 13-acetate; Sigma-Aldrich, St. Louis, MO, USA) for 24 h. Additionally, to polarize into M1 or M2 cells, M0 cells were cultured with 20 ng/mL recombinant human interferon-gamma (rhIFN-γ; Prospec, Rehovot, Israel) and 100 ng/mL lipopolysaccharides (LPS; Sigma-Aldrich), or 20 ng/mL recombinant human interleukin-4 (rhIL-4; Prospec) and 20 μg/mL rhIL-13 (Prospec) in RPMI 1640 medium supplemented with 5% FBS for 72 h. Additionally, PC3 or A549 cells were grown in complete medium to 80% confluence, washed and cultured in serum-free medium for 24 h, and then filtered through a 0.2 μm syringe filter (Sartorius, Göttingen, Germany) to prepare tumor-conditioned medium (TCM), and M0 cells were cultured with 10% or 50% tumor-conditioned medium for 72 h to polarize to M2-TAM. Afterwards, THP-1 macrophages differentiated into M0, M1, and M2 were disrupted using Mem-PER Plus Membrane Protein Extraction Kit (Thermo Fisher Scientific, Waltham, MA, USA), and the cell lysates were incubated with biotinylated TAMpep for 1 h at room temperature. After incubation with streptavidin agarose resin (Thermo Fisher Scientific) at room temperature for 30 min, the resin was washed, and the membrane-bound proteins were eluted from the resin. The eluted protein samples were analyzed by LC-MS/MS using a Q-Exactive mass spectrometer (Thermo Fisher Scientific). The LC-MS/MS data of each analyzed sample were analyzed using Proteome Discoverer, and the UniProt human database was used for label-free quantification. Proteomic analysis was performed using the ExDEGA (Excel-based Differentially Expressed Gene Analysis) tool (eBiogen, Seoul, Korea) (Fig. 1).
그 결과, M0, M1 및 M2 대식세포에서 총 1,450개의 단백질이 식별되었으며, 이 중 54개는 M0 및 M1 대식세포에 비해 M2 대식세포에서 비오틴-표지된 TAMpep에 우선적으로 결합하였다 (도 12 내지 14). 이 단백질들 중 CD18이 특히, 대식세포와 같은 골수(myeloid) 계통의 세포주에서 높게 발현되어 있었으며, 림프계(lymphoid)에서만 약하게 발현되어 있는 것으로 나타났다 (도 15). As a result, a total of 1,450 proteins were identified in M0, M1, and M2 macrophages, and 54 of them preferentially bound to biotin-labeled TAMpep in M2 macrophages compared to M0 and M1 macrophages (Figs. 12 to 14). Among these proteins, CD18 was particularly highly expressed in myeloid lineage cell lines such as macrophages, and was weakly expressed only in lymphoid (Fig. 15).
실시예 3. TB511의 CD18 표적화 분석Example 3. CD18 targeting analysis of TB511
CD18이 TB511의 표적 단백질임을 확인하기 위해, M2-분화된 cas9 스테이블 THP-1 세포(cas9 stable THP-1 cells)에 CD18 표적화 sgRNA(sgRNA/CD18; GTTCAACGTGACCTTCCGGC)를 트랜스펙션하여 CD18/sgRNA 녹다운(CD18/sgRNA knockdown) 을 수행한 뒤, CD18 mRNA 수준을 qRT-PCR로 분석하여 녹다운 세포의 생성을 검증하였다 (도 2). 이 후, 세포에 TB511 (3 μM)을 24시간 동안 처리하고 MTS 분석으로 세포 생존율을 확인하였다. 또한, 세포의 파쇄물을 스크램블 또는 TAMpep 펩타이드 (0, 0.001, 0.01, 0.1, 1 또는 10 μg)와 인큐베이션한 뒤, 비오틴화된(biotinylated) TAMpep와 인큐베이션하고, 스트렙타비딘 다이나비즈 (Streptavidin Dynabeads)(Thermo Fisher Scientific)를 사용하여 펩타이드-결합된 막 단백질을 용출한 단백질을 PAGE 분석하여, TB511의 CD18에 대한 직접적 결합을 분석하였다. 또한, FITC-표지된 TAMpep 및 APC-접합된 항-래빗 IgG 이차 항체 (1:500, Invitrogen)를 이용하여 면역형광 염색 분석을 수행하였다. 아울러, TAMpep의 CD18에 대한 결합 친화도(binding affinity)을 분석하였다.To confirm that CD18 is the target protein of TB511, CD18-targeting sgRNA (sgRNA/CD18; GTTCAACGTGACCTTCCGGC) was transfected into M2-differentiated cas9 stable THP-1 cells to perform CD18/sgRNA knockdown, and then CD18 mRNA levels were analyzed by qRT-PCR to verify the generation of knockdown cells (Fig. 2). After that, the cells were treated with TB511 (3 μM) for 24 h, and cell viability was confirmed by MTS assay. Additionally, direct binding of TB511 to CD18 was analyzed by incubating cell lysates with scrambled or TAMpep peptide (0, 0.001, 0.01, 0.1, 1, or 10 μg), followed by incubation with biotinylated TAMpep, and performing PAGE analysis of the eluted proteins to separate peptide-bound membrane proteins using Streptavidin Dynabeads (Thermo Fisher Scientific). In addition, immunofluorescence staining analysis was performed using FITC-labeled TAMpep and APC-conjugated anti-rabbit IgG secondary antibody (1:500, Invitrogen). In addition, the binding affinity of TAMpep to CD18 was analyzed.
세포 생존율 분석 결과, TB511 처리에 의해 CD18/sgRNA 녹다운 세포에서 대조군 세포(mock-transfected cells)에 비해 세포 생존율이 현저히 감소한 것으로 나타났다 (도 3). 또한, 면역침강(immunoprecipitation) 분석 결과, CD18가 비오틴화된 TAMpep과 면역침강되었으며, 이 결합은 비오틴화되지 않은 TAMpep과 미리 인큐베이션한 경우 농도-의존적으로 차단된 것으로 나타나 (도 4), TB511과 CD18가 직접적으로 결합하는 것을 확인하였다. 또한, 면역형광 염색 분석 결과, M2 대식세포에서 TAMpep이 CD18과 같은 위치에 존재하는 것을 확인하였다 (도 5). 아울러, TAMpep의 CD18에 대한 결합 친화도를 분석한 결과, 1.963 Х 10-6 의 KD 값을 나타내 (도 16 및 도 17), 항-CD18 항체의 결합 친화도와 유사한 수준임을 확인하였다.Cell viability analysis results showed that cell viability was significantly reduced in CD18/sgRNA knockdown cells treated with TB511 compared to mock-transfected cells (Fig. 3). In addition, immunoprecipitation analysis results showed that CD18 was immunoprecipitated with biotinylated TAMpep, and this binding was blocked in a concentration-dependent manner when pre-incubated with non-biotinylated TAMpep (Fig. 4), confirming the direct binding of TB511 to CD18. In addition, immunofluorescence staining analysis results confirmed that TAMpep was present at the same location as CD18 in M2 macrophages (Fig. 5). In addition, analysis of the binding affinity of TAMpep to CD18 showed a K D value of 1.963 Х 10 -6 (Figs. 16 and 17), confirming that it is at a similar level to the binding affinity of anti-CD18 antibody.
실시예 4. CD18의 종양 관련성 분석Example 4. Analysis of tumor relevance of CD18
CD18이 고형 종양의 진행, 특히 M2 대식세포의 종양 조직으로의 침투와 연관되어 있는지 확인하기 위해, 종양 조직 어레이(tumor tissue array)를 이용하여 CD18발현과 종양의 진행 간의 상관관계를 확인하였다. 구체적으로, EZ prep(Ventana Medical Systems, #950-102)을 사용하여 76℃에서 4분간 유방, 결장(colon), 폐, 간, 신장 및 피부의 정상 조직과 종양 조직 슬라이드에서 파라핀을 제거하고 항원 회수를 위해 cell conditioning solution 1 (Ventana Medical Systems, #950-124)과 함께 100℃에서 24분간 인큐베이션하였다. 슬라이드를 OptiView peroxidase inhibitor (Ventana Medical Systems, #760-700)로 4분간 37℃에서 처리한 뒤, 항-CD18 항체 (1:500, LSbio)와 인큐베이션하였다. 이 후, 슬라이드를 OptiView DAB IHC Detection Kit (Ventana Medical Systems, #760-700)를 사용하여 시각화하였으며, 이미지는 Aperio ImageScope 소프트웨어 (Leica, Wetzlar, Germany)를 사용하여 스캔하고 분석하였다. To determine whether CD18 is associated with the progression of solid tumors, especially with the infiltration of M2 macrophages into tumor tissues, the correlation between CD18 expression and tumor progression was determined using tumor tissue arrays. Specifically, paraffin was removed from normal and tumor tissue slides of breast, colon, lung, liver, kidney, and skin using EZ prep (Ventana Medical Systems, #950-102) at 76°C for 4 min, followed by incubation with cell conditioning solution 1 (Ventana Medical Systems, #950-124) at 100°C for 24 min for antigen retrieval. The slides were treated with OptiView peroxidase inhibitor (Ventana Medical Systems, #760-700) for 4 min at 37°C, followed by incubation with anti-CD18 antibody (1:500, LSbio). Slides were then visualized using the OptiView DAB IHC Detection Kit (Ventana Medical Systems, #760-700), and images were scanned and analyzed using Aperio ImageScope software (Leica, Wetzlar, Germany).
그 결과, CD18의 발현 수준이 유방, 결장, 폐, 간, 신장 및 피부 정상 조직에 비해 유방, 결장, 폐, 간, 신장 및 피부 종양 조직에서 현저하게 높은 것으로 나타났다 (도 6 및 도 7). As a result, the expression level of CD18 was found to be significantly higher in breast, colon, lung, liver, kidney, and skin tumor tissues than in breast, colon, lung, liver, kidney, and skin normal tissues (Figs. 6 and 7).
실시예 5. 대식세포 서브타입에 따른 CD18의 발현 분석Example 5. Analysis of CD18 expression according to macrophage subtype
상기 실시예 2에서와 같이 THP-1 단핵구를 M0, M1 및 M2 대식세포로 분화시키고, M2-TAM으로 분극화시킨 뒤, CD18의 상대적 mRNA 발현 수준을 qRT-PCR로 분석하고, 단백질 발현 수준을 웨스턴 블롯 분석 및 면역형광염색으로 확인하였다.As in Example 2 above, THP-1 monocytes were differentiated into M0, M1, and M2 macrophages, and polarized with M2-TAM. The relative mRNA expression level of CD18 was analyzed by qRT-PCR, and the protein expression level was confirmed by Western blot analysis and immunofluorescence staining.
그 결과, CD18의 유전자 및 단백질의 발현 수준이 다른 서브타입의 대식세포에 비해 M2 대식세포 및 M2-TAM에서 현저히 높은 것으로 나타났다 (도 8 내지 11). As a result, the expression levels of CD18 genes and proteins were found to be significantly higher in M2 macrophages and M2-TAM than in other subtypes of macrophages (Figs. 8 to 11).
실시예 6. 활성화된 구조의 CD18에 대한 TB511의 결합 및 효과 분석Example 6. Binding and effect analysis of TB511 to CD18 in activated conformation
6-1. TB511 및 CD18의 결합 모델링6-1. Combination modeling of TB511 and CD18
TB511의 CD18에 대한 결합을 단백질-펩타이드 상호작용 모델링을 사용해 분석하였다. 구체적으로, CD18의 시스테인-풍부 영역(cysteine-rich region)의 서열 (Uniport _p05107, cysteine-rich tandem repeat region: 449-617)에 기반하여, trRosetta algorithm (https:/yanglab.nankai.edu.cn/trRosetta/)을 사용해 3차 구조를 구축하였다. trRosetta로 생성한 CD18 PDB 파일과 TB511의 아미노산 서열을 이용해 CABS-dock server (http:/biocomp.chem.uw.edu.pl/CABSdock/)를 사용하여 TB511 및 CD18의 도킹 시뮬레이션을 수행하였다. 이를 통해 10가지 모델이 생성되었다. 또한, 5 μg/mL CD18 단백질 (Abbexa)과 FITC-접합된 TAMpep 및 TAMpep의 알라닌 치환 라이브러리 펩타이드를 상온에서 2시간 동안 반응시킨 뒤, 펩타이드들 간의 결합 친화력을 ELISA를 사용해 분석하였다. 상기의 결합 친화도 분석 결과를 기반으로 상기의 도킹 시뮬레이션을 통해 생성된 10가지 모델 중에 대표적인 모델을 선택하였다.The binding of TB511 to CD18 was analyzed using protein-peptide interaction modeling. Specifically, based on the sequence of the cysteine-rich region of CD18 (Uniport _p05107, cysteine-rich tandem repeat region: 449-617), the 3D structure was constructed using the trRosetta algorithm ( https:/yanglab.nankai.edu.cn/trRosetta/ ). Using the CD18 PDB file generated by trRosetta and the amino acid sequence of TB511, docking simulations of TB511 and CD18 were performed using the CABS-dock server ( http:/biocomp.chem.uw.edu.pl/CABSdock/ ). Ten models were generated. Additionally, 5 μg/mL CD18 protein (Abbexa) and FITC-conjugated TAMpep and alanine-substituted library peptides of TAMpep were reacted at room temperature for 2 hours, and then the binding affinity between the peptides was analyzed using ELISA. Based on the results of the above binding affinity analysis, a representative model was selected from among the 10 models generated through the above docking simulation.
단백질-펩타이드 상호작용 모델링 분석 결과, 굽은 (닫힌) (bent (closed)) 형태의 총 CD18에 대한 TB511의 결합은 예측되지 않은 반면, 열린 (open)형태의 시스테인-풍부 도메인은 TB511과 결합할 것으로 강하게 예측되어 (도 18), TB511의 라이신 및 트립토판 잔기들이 주요한 결합 요소인 것으로 유추되었다. 또한, TAMpep의 알라닌 치환 라이브러리를 이용하여 분석한 결합 친화력 분석 결과, 라이신이 알라닌으로 치환된 A6 펩타이드 및 트립토판이 알리닌으로 치환된 A12 펩타이드가 CD18 단백질에 대해 현저히 저하된 결합을 나타냈다 (도 19).Protein-peptide interaction modeling analysis results showed that TB511 binding to total CD18 in the bent (closed) conformation was not predicted, whereas the open conformation cysteine-rich domain was strongly predicted to bind to TB511 (Fig. 18), suggesting that lysine and tryptophan residues of TB511 are the major binding elements. In addition, binding affinity analysis using the alanine substitution library of TAMpep showed that the A6 peptide, in which lysine was substituted with alanine, and the A12 peptide, in which tryptophan was substituted with alanine, exhibited significantly reduced binding to CD18 protein (Fig. 19).
6-2. TB511의 활성화 CD18 특이적 결합 및 이를 통한 세포사멸 분석 6-2. Analysis of CD18-specific binding and apoptosis through activation of TB511
TB511이 CD18의 활성화된(active) 열린 구조(conformation)를 인지하는지, 그리고 이를 통해 세포독성을 유발하는지 확인하기 위해, THP-1 세포에서 CD18를 활성화시키고 TB511 처리로 인한 세포독성을 비활성화된 경우와 비교하였다. 구체적으로, THP-1 세포에 1 mM manganese chloride tetrahydrate (Mn2+, Sigma-Aldrich)를 1시간 동안 처리하여 휴지 상태(resting state)의 THP-1 세포를 활성화하였다. 이 후, 활성화된 THP-1 세포 및 비활성화된 대조군 THP-1 세포에 TB511 (1 μM)를 각각 처리하였다. Annexin V (APC) 및 7AAD로 염색하고 FACS Lyric System (BD Bioscience, NJ, USA)로 염색된 세포를 분석하여 세포생존율을 확인하였다.To determine whether TB511 recognizes the activated open conformation of CD18 and induces cytotoxicity through this, CD18 was activated in THP-1 cells and the cytotoxicity induced by TB511 treatment was compared with that of the inactivated case. Specifically, THP-1 cells were activated in the resting state by treating them with 1 mM manganese chloride tetrahydrate (Mn 2+ , Sigma-Aldrich) for 1 h. Then, TB511 (1 μM) was treated to the activated THP-1 cells and the inactivated control THP-1 cells, respectively. Cell viability was determined by staining with Annexin V (APC) and 7AAD and analyzing the stained cells with a FACS Lyric System (BD Bioscience, NJ, USA).
그 결과, TB511 처리시 대조군 세포에 비해 CD18이 활성화된 THP-1세포에서 세포사멸이 현저하게 증가한 것으로 나타났다 (도 20 및 도 21). 따라서, TB511이 CD18의 활성화된 열린 구조를 인지하여 세포독성을 유도하는 것을 확인하였다. As a result, it was shown that apoptosis significantly increased in CD18-activated THP-1 cells compared to the control cells when treated with TB511 (Figs. 20 and 21). Therefore, it was confirmed that TB511 recognizes the activated open structure of CD18 and induces cytotoxicity.
6-3. 종양미세환경 및 대식세포 서브타입에 따른 CD18의 활성화 분석6-3. Analysis of CD18 activation according to tumor microenvironment and macrophage subtype
종양미세환경의 M2-TAM에서 CD18이 활성화된 구조를 유지하는지 확인하기 위해, THP-1 세포에 A549 종양 조절 배지를 처리하고, FITC-접합된 mAb KIM127 (Leinco Technologies, Pantone, MO, USA)를 이용하여 CD18의 활성화 수준을 FACS로 분석하였다. 또한, THP-1 세포를 M0, M1, M2 및 M2-TAM의 서브타입으로 각각 분화시킨 뒤, CD18의 활성화 정도를 분석하였다.To determine whether CD18 maintains the activated structure in M2-TAM of the tumor microenvironment, THP-1 cells were treated with A549 tumor-conditioned medium, and the activation level of CD18 was analyzed by FACS using FITC-conjugated mAb KIM127 (Leinco Technologies, Pantone, MO, USA). In addition, THP-1 cells were differentiated into M0, M1, M2, and M2-TAM subtypes, respectively, and the degree of CD18 activation was analyzed.
그 결과, CD18의 활성화 수준이 A549 종양 조절 배지 처리에 의해 농도 의존적으로 현저하게 증가하는 것으로 나타났다 (도 22). 또한, 대식세포의 서브타입들 중 종양 조절 배지와 함께 배양하여 분극화된 M2-TAM에서 CD18의 활성화 수준이 가장 높게 나타나 (도 23), M2-TAM이 TME에서 활성화된 형태의 CD18 을 유지하는 것을 확인하였다.As a result, the activation level of CD18 was found to be significantly increased in a concentration-dependent manner by A549 tumor-conditioned medium treatment (Fig. 22). In addition, among the subtypes of macrophages, the activation level of CD18 was found to be the highest in polarized M2-TAM cultured with tumor-conditioned medium (Fig. 23), confirming that M2-TAM maintains the activated form of CD18 in the TME.
6-4. 대식세포 서브타입에 따른 TB511에 의한 세포사멸 분석6-4. Analysis of cell death by TB511 according to macrophage subtype
각 대식세포 서브타입에서 TB511에 의한 세포사멸 정도를 비교하기 위해, THP-1 세포를 M0, M1, M2 또는 M2-TAM으로 분화시키고 MitoTracker RedROX (Invitrogen) 및 caspase 3/7 시약을 30분 동안 처리한 뒤 TB511 (1 μM)를 처리하고 바로 restoration fluorescence microscopy (DeltaVison, NJ, USA)를 이용하여 라이브-세포 이미징을 수행하였다. To compare the extent of TB511-induced apoptosis in each macrophage subtype, THP-1 cells were differentiated into M0, M1, M2, or M2-TAM and treated with MitoTracker RedROX (Invitrogen) and
그 결과, TB511에 의한 caspase 3/7의 활성화가 TB511과의 인큐베이션 후 5분 안에 M2-TAM에서 시작되었으며, 15분에 최고점에 도달하였다. M2 대식세포는 이보다는 덜하지만 TB511에 영향을 받는 것으로 나타났으나, M0 또는 M1 대식세포에서는 caspase 3/7의 활성화가 관찰되지 않았다 (도 24 및 도 25). 이를 통해, CD18 및 TB511간의 구조 특이적 결합이 대식세포 서브타입들 중 M2-TAM에 대한 선택적 세포독성으로 나타난 것을 확인하였다.As a result,
6-5. 활성화된 CD18의 종양 관련성 분석6-5. Analysis of tumor relevance of activated CD18
종양 조직 어레이(tumor tissue array)를 이용하여 CD18의 활성화된 열린 구조의 수준을 분석한 결과, 전립선, 간, 유방, 폐, 및 결장의 정상조직에 비해 종양 조직에서 CD18의 활성화 (pan CD18 및 KIM127-이중 양성) 수준이 현저히 증가되어 있어 (도 26), 정상 세포에서 대부분의 조직-상주(tissue-resident) 대식세포에서 CD18가 비활성화된 형태로 존재하지만, 이는 TME에서 활성화된 형태로 변화하는 것을 알 수 있다.Analysis of the level of activated open conformation of CD18 using tumor tissue array revealed that the level of CD18 activation (pan CD18 and KIM127-double positive) was significantly increased in tumor tissues compared to normal tissues of prostate, liver, breast, lung, and colon (Fig. 26), indicating that CD18 exists in an inactive form in most tissue-resident macrophages in normal cells, but changes to an activated form in the TME.
실시예 7. TB511의 M2 대식세포 막 역학 및 세포 내 수송 분석Example 7. Analysis of M2 macrophage membrane dynamics and intracellular transport of TB511
TB511의 세포막 역학 및 세포내 수송을 추적하기 위해, 1-ethyl-3-3-(dimethylaminopropyl)carbodiimide hydrochloride (Pierce, Rockford, IL, USA) 및 sulfo-N-hydroxysulfosuccinimide (Pierce)를 이용하여 카복실산-기능화된(carboxylic acid-functionalized) 금 나노로드 (AuNRs; Nanopartz, Salt Lake City, UT, USA)와 TB511를 접합하였다. THP-1를 분화시킨 M2 대식세포에 약 1.5 Х 109 particles/mL의 TB511-AuNRs 20 μL를 처리하여 인큐베이션하고 MitoTracker Green (Invitrogen) 및 Hoechst 33342 (Invitrogen)를 처리하여 0.5시간 동안 인큐베이션한 뒤, iMLSM(integrated multi-dimensional light-sheet microscopy)를 이용하여 10 ms의 간격으로 단일 입자 추적 이미지를 획득하였다. ImageJ (NIH)와 MATLAB (Mathworks, Torrance, CA, USA)를 사용하여 획득한 이미지를 분석하고, 개별 펩타이드-AuNR의 3D 공간궤적 (x, y, z)은 astigmatism 방법으로 초-국소화되었다(super-localized). 또한, 개별 펩타이드-AuNR의 회전(rotation) 및 방향(orientation)은 TSPR(transverse surface plasmon resonance)는 cos2φ로 피팅하여, LSPR(longitudinal surface plasmon resonance)는 sin2θ로 피팅하여 계산하였다. 아울러, 엔도사이토시스(endocytosis) 억제를 위해, clathrin-매개된 엔도사이토시스(clathrin-mediated endocytosis, CME) 억제제인 CPZ(chlorpromazine), clathrin-매개된 엔도사이토시스 및 caveolae-매개된 엔도사이토시스를 모두 억제하는 억제제인 MβCD(methyl-β-cyclodextrin), 또는 거대음세포증(macropinocytosis, MP)을 억제하는 억제제인 CyD(cytochalasin D)를 처리하거나, 초기 엔도좀의 산성화(acidification)의 억제, 또는 엔도좀과 라이소좀의 성숙 및 결합을 억제하기 위해, Baf-A1(bafilomycin A1) 또는 CQ(chloroquine)를 처리하고, 엔도좀에서 초기 엔도좀 항원 1(early endosome antigen 1 (EEA1)) 및 TB511-Cy5의 위치를 면역형광염색 분석으로 확인하였다.To track the membrane dynamics and intracellular transport of TB511, TB511 was conjugated to carboxylic acid-functionalized gold nanorods (AuNRs; Nanopartz, Salt Lake City, UT, USA) using 1-ethyl-3-3-(dimethylaminopropyl)carbodiimide hydrochloride (Pierce, Rockford, IL, USA) and sulfo-N-hydroxysulfosuccinimide (Pierce). THP-1-differentiated M2 macrophages were treated with 20 μL of TB511-AuNRs (about 1.5 Х 10 9 particles/mL) and incubated with MitoTracker Green (Invitrogen) and Hoechst 33342 (Invitrogen) for 0.5 h. Single-particle tracking images were acquired at 10-ms intervals using iMLSM (integrated multi-dimensional light-sheet microscopy). The acquired images were analyzed using ImageJ (NIH) and MATLAB (Mathworks, Torrance, CA, USA), and the 3D spatial trajectories (x, y, z) of individual peptide-AuNRs were super-localized by the astigmatism method. Additionally, the rotation and orientation of individual peptide-AuNR were calculated by fitting the transverse surface plasmon resonance (TSPR) with cos2φ and the longitudinal surface plasmon resonance (LSPR) with sin2θ. In addition, for endocytosis inhibition, CPZ (chlorpromazine), an inhibitor of clathrin-mediated endocytosis (CME), MβCD (methyl-β-cyclodextrin), an inhibitor that inhibits both clathrin-mediated endocytosis and caveolae-mediated endocytosis, or CyD (cytochalasin D), an inhibitor that inhibits macropinocytosis (MP), or Baf-A1 (bafilomycin A1) or CQ (chloroquine) were treated to inhibit acidification of early endosomes, or maturation and association of endosomes with lysosomes, and the locations of early endosome antigen 1 (EEA1) and TB511-Cy5 in endosomes were confirmed by immunofluorescence staining analysis.
그 결과, M2의 DIC(Differential interference contrast) 및 SRRF(super-resolution radial fluctuations) 이미지를 통해 TB511-AuNRs가 세포 내 미토콘드리아 및 핵에 분포하는 것으로 나타났다 (도 27). 개별 펩타이드-AuNR의 3D 공간궤적 분석 결과, x, y 및 z 방향으로의 이동(displacements) 및 산란 강도 변동(scattering intensity fluctuations)이 단계적으로 손실되어 (도 28 내지 30), TB511-AuNR가 M2 대식세포의 세포막 수용체에 결합하는 것을 확인하였다. 또한, 20-41초 정도의 특정 시간에 TB511-AuNR가 더 적은 공간적 이동 및 느린 방향 전환을 나타내, 세포막에 포장되어 엔도사이토시스 장치들로 조립되는 것을 확인하였으며, 이 후, x, y 및 z 방향으로의 명확한 움직임이 관찰되어 TB511-AuNR가 세포막으로부터 분리되어 세포내로 수송된 것을 확인하였다. 또한, 세포의 흡수를 억제하는 억제제를 처리한 결과, CPZ 및 CyD 는 TB511의 흡수를 억제해 TB511-유도된 세포사멸을 감소시킨 반면, MβCD는 세포사멸이 일어나지 않는 것으로 나타났다 (도 31 및 도 32). 아울러, Baf-A1 및 CQ의 처리는 엔도좀에서 EEA1 및 TB511-Cy5의 공동 국소화를 증가시켰으며, TB511-유도된 세포사멸을 현저하게 감소시켰다 (도 33 및 도 34). As a result, DIC (differential interference contrast) and SRRF (super-resolution radial fluctuations) images of M2 showed that TB511-AuNRs were distributed in the mitochondria and nucleus within the cells (Fig. 27). Analysis of the 3D spatial trajectories of individual peptide-AuNRs showed that displacements and scattering intensity fluctuations in the x, y, and z directions were gradually lost (Figs. 28 to 30), confirming the binding of TB511-AuNRs to the membrane receptors of M2 macrophages. In addition, at a specific time of approximately 20-41 s, TB511-AuNRs showed less spatial movement and slower directional changes, confirming that they were packed into the membrane and assembled into endocytosis apparatuses, after which clear movements in the x, y, and z directions were observed, confirming that TB511-AuNRs were separated from the membrane and transported into the cell. In addition, when treated with inhibitors that inhibit cellular uptake, CPZ and CyD inhibited the uptake of TB511 and reduced TB511-induced apoptosis, whereas MβCD did not cause apoptosis (Figs. 31 and 32). In addition, treatment with Baf-A1 and CQ increased the co-localization of EEA1 and TB511-Cy5 in endosomes, and significantly reduced TB511-induced apoptosis (Figs. 33 and 34).
이를 통해, TB511가 M2 대식세포에 엔도사이토시스를 통해 세포내로 내부화(internalized)되고, 엔도좀에서 나와 세포질로 들어갔음을 알 수 있다.This suggests that TB511 is internalized into M2 macrophages through endocytosis, exits the endosomes, and enters the cytoplasm.
실시예 8. TB511의 Example 8. TB511 항종양 효과 분석Antitumor effect analysis
8-1. 면역항암제 비반응성 종양(Cold tumors)에 대한 항종양 효과8-1. Antitumor effect on cold tumors that are not responsive to immunotherapy
면역요법(immunotherapy)에 낮은 반응성을 나타내며, 종양 항원이 없는 Cold tumors에 대한 TB511의 항종양 효과를 확인하기 위해, 피하(subcutaneous) 전립선암(prostate cancer, PC) 마우스 모델 및 정위(orthotopic) 간세포암(hepatocellular carcinoma, HCC) 마우스 모델을 제조하고 TB511의 항종양 효과를 평가하였다. 구체적으로, 피하 종양 마우스 모델을 제조하기 위해, PC 종양 세포를 마트리젤 (Corning, NY, USA)과 1:1로 혼합한 뒤, 6-8주령 C57BL/6 또는 BALB/c 마우스 (Taconic Biosciences, NY, USA)의 오른쪽 옆구리에 피하 주입하였다. 종양 부피가 50-100 mm3에 도달하면 종양이 있는 마우스를 두 그룹으로 나눠, TB511 투여군에는 TB511을 3일마다 200 nmol/kg의 용량으로 피하 주사하였다. 정위 종양 마우스 모델을 제조하기 위해, 마우스의 복부를 절개하고 hepa1-6-Luc 세포를 맹장 또는 간의 장막하 평면에 주입한 뒤 봉합하였다. 종양 주입 7일 또는 14일 후에 마우스를 두 그룹으로 나누고 TB511를 3일마다 피하 주사하였으며, D-luciferin (BioVision, Milpitas, CA, USA)을 마우스 한 마리당 3mg의 용량으로 복강내 주사하여 종양 성장을 추적하였다. 종양 직경은 디지털 캘리퍼를 사용하여 측정하였으며, 종양의 부피는 하기 수학식 1로 계산하였다. 마우스는 NightOWL LB 983 in vivo imaging system (Berthold Technologies, Bad Wildbad, Germany)을 사용하여 일주일에 한두 번 평가하였으며, 종양 주입 30일 후에 안락사하고 종양 조직을 분리하였다.To determine the antitumor effect of TB511 on Cold tumors, which have low responsiveness to immunotherapy and lack tumor antigens, a subcutaneous prostate cancer (PC) mouse model and an orthotopic hepatocellular carcinoma (HCC) mouse model were prepared and the antitumor effect of TB511 was evaluated. Specifically, to prepare a subcutaneous tumor mouse model, PC tumor cells were mixed 1:1 with Matrigel (Corning, NY, USA) and then subcutaneously injected into the right flank of 6-8 week-old C57BL/6 or BALB/c mice (Taconic Biosciences, NY, USA). When the tumor volume reached 50-100 mm 3 , the tumor-bearing mice were divided into two groups, and TB511 was injected subcutaneously into the TB511 group at a dose of 200 nmol/kg every 3 days. To prepare orthotopic tumor mouse models, the abdomens of mice were incised, and hepa1-6-Luc cells were injected into the cecum or the subserosal plane of the liver, and then sutured. Seven or 14 days after tumor injection, the mice were divided into two groups and injected subcutaneously with TB511 every 3 days, and D-luciferin (BioVision, Milpitas, CA, USA) was injected intraperitoneally at a dose of 3 mg per mouse to track tumor growth. Tumor diameters were measured using a digital caliper, and tumor volumes were calculated by the following
그 결과, TB511은 면역항암제 비반응성 종양(Cold tumors) 마우스 모델에서 종양 성장을 현저하게 억제하는 것으로 나타났다 (도 35 내지 37).As a result, TB511 was shown to significantly inhibit tumor growth in a mouse model of immunotherapy-unresponsive tumors (Cold tumors) (Figs. 35 to 37).
8-2. 면역항암제 반응성 종양(Hot tumors)에 대한 항종양 효과8-2. Antitumor effect on hot tumors responsive to immunotherapy
면역요법에 높은 반응성을 나타내며, 종양 항원의 면역 활성화 및 침투한 T 세포로 특징지어지는 Hot tumors에 대한 TB511의 항종양 효과를 확인하기 위해, 피하 비소세포성 폐암(non-small cell lung cancer, NSCLC) 또는 신세포암종(renal cell carcinoma, RCC) 마우스 모델과 정위 대장암(colorectal cancer, CRC) 마우스 모델을 상기 실시예 8-1에서와 같이 제조하고 TB511를 투여한 뒤 항종양 효과를 분석하였다.To confirm the antitumor effect of TB511 on hot tumors, which exhibit high responsiveness to immunotherapy and are characterized by immune activation of tumor antigens and infiltrated T cells, subcutaneous non-small cell lung cancer (NSCLC) or renal cell carcinoma (RCC) mouse models and orthotopic colorectal cancer (CRC) mouse models were prepared as in Example 8-1, and TB511 was administered thereto, and the antitumor effect was analyzed.
그 결과, TB511은 면역항암제 반응성 종양(Hot tumors) 마우스 모델에서 종양 성장을 현저하게 억제하는 것으로 나타났다 (도 38 내지 41).As a result, TB511 was shown to significantly inhibit tumor growth in a mouse model of immunotherapy-responsive tumors (Hot tumors) (Figs. 38 to 41).
이를 통해, TB511이 면역항암제에 대한 반응성과 상관없이, 항종양 효과를 나타내는 것을 확인하였다. Through this, it was confirmed that TB511 exhibited antitumor effects regardless of responsiveness to immunotherapy.
8-3. 종양미세환경에서 TB511의 M2-TAM 표적화 분석8-3. Analysis of M2-TAM targeting of TB511 in tumor microenvironment
TB511의 항종양 효과가 TME에서 M2-TAM을 표적화 함으로써 야기된 것인지 확인하기 위해, PC 및 CRC 종양 조직에서 M2-TAM (CD45+ 게이팅된 세포 중 F4/80+CD206+의 백분율) 및 M1-TAM (CD45+ 게이팅된 세포 중 CD206+F4/80+의 백분율)의 세포수를 FACS로 분석하고 M1/M2비율을 계산하였다. 또한, TB511의 대식세포 서브타입에 대한 선택적인 표적화를 확인하기 위해, TB511의 처리 후 CRC 종양 조직에서 M1-TAM 및 M2-TAM의 세포수, 뇌, 피부, 신장 및 간을 포함하는 정상 조직에 존재하는 조직-상주 대식세포의 수를 면역조직화학(immunohistochemistry)(IHC)으로 분석하였다.To determine whether the antitumor effect of TB511 was caused by targeting M2-TAM in the TME, the cell counts of M2-TAM (the percentage of F4/80 + CD206 + among the CD45 + gated cells) and M1-TAM (the percentage of CD206 + F4/80 + among the CD45 + gated cells) in PC and CRC tumor tissues were analyzed by FACS, and the M1/M2 ratio was calculated. In addition, to confirm the selective targeting of TB511 to macrophage subtypes, the cell counts of M1-TAM and M2-TAM in CRC tumor tissues, and the number of tissue-resident macrophages present in normal tissues including brain, skin, kidney, and liver after treatment with TB511 were analyzed by immunohistochemistry (IHC).
그 결과, PC 및 CRC 종양 조직에서 M2-TAM 세포수가 대조군에 비해 TB511 투여군에서 현저히 감소한 반면, M1-TAM 세포수는 유의미하게 변하지 않아, TB511 투여에 의해 M1/M2비율이 현저히 증가한 것으로 나타났다 (도 42 내지 47). 또한, 종양 조직에서 TB511 처리에 의해 M2-TAM 세포수가 감소하고, M1-TAM 세포수는 변화가 없었던 반면, 정상 조직에서 조직-상주 대식세포 집단 (F4/80+)은 TB511 처리에 의해 영향을 받지 않는 것으로 나타났다 (도 48 내지 51). As a result, the number of M2-TAM cells in PC and CRC tumor tissues was significantly reduced in the TB511-administered group compared to the control group, whereas the number of M1-TAM cells did not change significantly, indicating that the M1/M2 ratio was significantly increased by TB511 administration (Figs. 42 to 47). In addition, the number of M2-TAM cells was reduced and the number of M1-TAM cells was not changed by TB511 treatment in tumor tissues, whereas the tissue-resident macrophage population (F4/80 + ) in normal tissues was not affected by TB511 treatment (Figs. 48 to 51).
8-4. TB511의 종양 표적화 분석8-4. Tumor targeting analysis of TB511
정위 유방암 마우스 모델에 Cy5-표지된 TB511을 주사하고 TB511의 생물학적 분포(bio-distribution)를 조사하였다. 구체적으로, 4T1 유방암 세포를 마우스의 유방 지방 패드에 5Х105 cells/mouse의 밀도로 주입하여 정위 유방암 마우스 모델을 제조하고, 7일 후 Cy5-표지된 TB511 또는 스크램블된 펩타이드(scrambled peptides) (대조군)을 2.5 mg/kg로 정맥 주사하였다. 마우스를 마취하고 생체 내 TB511의 분포를 IVIS Lumina II (PerkinElmer, Waltham, MA, USA)로 촬영하고 분석하였다.Cy5-labeled TB511 was injected into an orthotopic breast cancer mouse model, and the bio-distribution of TB511 was investigated. Specifically, 4T1 breast cancer cells were injected into the mammary fat pad of mice at a density of 5Х10 5 cells/mouse to prepare an orthotopic breast cancer mouse model, and 7 days later, Cy5-labeled TB511 or scrambled peptides (control) were intravenously injected at 2.5 mg/kg. The mice were anesthetized, and the distribution of TB511 in vivo was photographed and analyzed using IVIS Lumina II (PerkinElmer, Waltham, MA, USA).
그 결과, 주사 48시간 후 TB511의 형광 신호가 간, 폐, 심장, 신장, 또는 근육이 아닌 종양 조직을 주사한 유방 조직에서만 관찰되었으며, 대조군에 비해 종양 조직에서 더 높은 생물학적 분포를 나타냈다 (도 52 내지 55). As a result, 48 hours after injection, the fluorescent signal of TB511 was observed only in the injected breast tissue, not in the liver, lung, heart, kidney, or muscle, and showed a higher biological distribution in the tumor tissue compared to the control group (Figs. 52 to 55).
이를 통해, TB511이 종양 부위(sites)에 특이적으로 축적되고TME에서 M2-TAM을 선택적으로 제거하지만 정상 조직 내의 조직-상주 대식세포에는 영향을 주지 않음을 확인하였다.Through this, we confirmed that TB511 specifically accumulates at tumor sites and selectively removes M2-TAM in the TME, but does not affect tissue-resident macrophages within normal tissues.
실시예 9. TME 에서 TB511에 의한 면역세포 변화 분석Example 9. Analysis of immune cell changes in TME caused by TB511
9-1. 면역세포 재프로그래밍 분석9-1. Immune cell reprogramming analysis
TB511에 의한 M2-TAM의 제거에 의해 면역세포들이 재프로그래밍(reprogramming)되는지 확인하기 위해, PC 검체에 TB511을 처리하고, CD4+, CD8+ 및 NK 세포와 같은 TME 내의 항종양 면역세포 집단 변화를 FACS로 분석하고 이 데이터를 t-SNE 분석하여 확인하였다. 또한, TB511의 항종양 효과에 대한 CD8+ T 및 NK 세포의 영향을 확인하기 위해, 항-NK1.1 항체 (clone PK136; BioXcell) 또는 항-CD8 항체 (clone 2.43; BioXcell, Lebanon, NH, USA)를 종양 주입 3일 전 및 1일 전에 복강내 주사하고 상기 실시예 8-1에서와 같이 대장암 세포인 TRAMP-C2 세포를 마트리젤과 혼합하여 주입 후 상기 항체는 매주 주사하여 (대조군: IgGa isotype (C1.18.4, BioXcell) 주사) NK 또는 CD8 결핍 마우스 모델을 제조하였다 (도 60). 이 후, 마우스의 말초 혈액(peripheral blood) 단핵 세포(mononuclear cells)들 중 CD8+ T 및 NK 세포의 숫자를 분석하였다. 아울러, NK 또는 CD8 결핍 마우스 모델에서 TB511의 항종양 효과를 분석하였다.To determine whether the removal of M2-TAM by TB511 reprograms immune cells, PC specimens were treated with TB511, and changes in anti-tumor immune cell populations within the TME, such as CD4 + , CD8 + , and NK cells, were analyzed by FACS, and the data were confirmed by t-SNE analysis. In addition, to confirm the influence of CD8 + T and NK cells on the antitumor effect of TB511, anti-NK1. 1 antibody (clone PK136; BioXcell) or anti-CD8 antibody (clone 2.43; BioXcell, Lebanon, NH, USA) was injected intraperitoneally 3 days and 1 day before tumor injection, and TRAMP-C2 cells, which are colon cancer cells, were mixed with Matrigel and injected as in Example 8-1, and then the antibodies were injected weekly (control group: injection of IgGa isotype (C1.18.4, BioXcell)) to prepare NK or CD8 deficient mouse models (Fig. 60). Thereafter, the numbers of CD8 + T and NK cells in the peripheral blood mononuclear cells of the mice were analyzed. In addition, the antitumor effect of TB511 was analyzed in the NK or CD8 deficient mouse models.
그 결과, PC에서 NK세포 집단은 현저하게 증가한 반면, CD8+ T세포 집단은 통계적으로 유의미하게 변화하지 않았다 (도 56 및 도 57). 또한, TB511처리 후 PC 표본 (specimens)에서 NCAM1+ NK 세포의 숫자가 증가하는 것으로 나타났다 (도 58 및 도 59). 또한, NK 또는 CD8 결핍 마우스 모델의 혈액에서 CD8+ T 및 NK 세포의 숫자는 항체 처리에 의해 현저하게 감소하였다 (도 73 내지 76). 아울러, TB511는 대조군 및 CD8+ T 세포 결핍 그룹 모두에서 항종양 효과를 나타낸 반면, NK 세포가 결핍된 그룹에서는 항종양 효과를 나타내지 않았다 (도 61 내지 63).As a result, the NK cell population in the PC was significantly increased, whereas the CD8 + T cell population did not change statistically significantly (Figs. 56 and 57). In addition, the number of NCAM1 + NK cells was found to increase in the PC specimens after TB511 treatment (Figs. 58 and 59). In addition, the numbers of CD8 + T and NK cells in the blood of NK or CD8 deficient mouse models were significantly decreased by antibody treatment (Figs. 73 to 76). In addition, TB511 showed an antitumor effect in both the control group and the CD8 + T cell deficient group, whereas it did not show an antitumor effect in the NK cell deficient group (Figs. 61 to 63).
9-2. 종양-침윤 세포수 분석9-2. Analysis of tumor-infiltrating cell counts
NSCLC 및 CRC와 같은 면역항암제 반응성 종양에서 TB511 처리에 의한 종양-침윤된(tumor-Infiltrated) CD8+ T 세포 (CD45+ CD8+)의 수 및 고갈된(exhausted) CD8+ T 세포 (PD-1+ CD8+ T)의 수를 FACS 분석으로 확인하였으며, CD8+ T 및 Granzyme B+ 세포의 수를 IHC 분석으로 확인하였다. 또한, 대장암 세포인 CT26 세포를 주입하고 항-CD8 항체를 주입한 CRC 마우스 모델을 제조하고 (도 70), TB511 처리에 의한 항종양 효과를 분석하였다. The number of tumor-infiltrated CD8 + T cells (CD45 + CD8 + ) and exhausted CD8 + T cells (PD-1 + CD8 + T) by TB511 treatment in immunotherapy-responsive tumors such as NSCLC and CRC were determined by FACS analysis, and the number of CD8 + T and Granzyme B + cells were determined by IHC analysis. In addition, a CRC mouse model was created by injecting CT26 cells, a colon cancer cell line, and anti-CD8 antibody (Fig. 70), and the antitumor effect by TB511 treatment was analyzed.
그 결과, NSCLC에서 종양-침윤 CD8+ T 세포의 수가 TB511를 투여한 군에 증가하였으며, 고갈된 CD8+ T 세포의 수는 감소하는 것으로 나타났다 (도 64 및 도 65). 또한, 항종양 면역세포들 중에서, CD8+ T 세포만이 CRC에서 유의미하게 증가한 것으로 나타났다 (도 66 및 도 67). 또한, IHC 분석 결과, TB511 처리에 의해 CD8+ T 및 Granzyme B+ 세포수가 증가하는 것으로 나타났으며 (도 68 및 도 69), TB511의 항종양 효과는 CRC 마우스 모델에서 오직 CD8+-고갈된 세포에서만 나타나지 않았다 (도 70 내지 72).As a result, the number of tumor-infiltrating CD8 + T cells in NSCLC increased in the group administered TB511, while the number of depleted CD8 + T cells decreased (Figs. 64 and 65). In addition, among the antitumor immune cells, only CD8 + T cells were significantly increased in CRC (Figs. 66 and 67). In addition, the IHC analysis results showed that the number of CD8 + T and Granzyme B + cells increased by TB511 treatment (Figs. 68 and 69), and the antitumor effect of TB511 was not observed only in CD8 + -depleted cells in the CRC mouse model (Figs. 70 to 72).
이를 통해, TME의 종양-침윤 세포는 면역항암제 반응성 및 비반응성 종양 모두에서 TB511 처리 후 현저하게 변화하지만, 각 종양 종류에 따라 침윤 세포의 종류는 다른 것을 확인하였다.Through this, we confirmed that tumor-infiltrating cells in the TME significantly changed after TB511 treatment in both immunotherapy-responsive and non-responsive tumors, but the types of infiltrating cells differed depending on each tumor type.
실시예 10.Example 10. TB511의 임상적 가능성 평가Clinical feasibility assessment of TB511
10-1. TB511의 면역 시스템을 통한 항종양 효과 분석10-1. Analysis of antitumor effect of TB511 through immune system
TB511의 임상적 가능성을 평가하기 위해, 면역결핍(Immunodeficient) 마우스인 4주령의 SID 마우스 (NOD-Prkdcem1BeakIL2rgem1Break)에 1.5 Gy의 감마선을 조사하고 인간 CD34+ 조혈줄기세포(hematopoietic stem cells) (Lonza, 바젤, 스위스) 2Х105 세포/마우스로 정맥주사하여 인간화된 마우스 모델을 제조하였다. 줄기세포 주입 11주 후에 말초 혈액을 채취하고 유세포 분석을 사용하여 성숙한 인간 백혈구(인간 CD45+ 세포)의 이식(engraftment)을 확인한 결과, hCD45+ 세포 집단은 이식 후 11주 후에 수컷 마우스 (n=16)에서 37.32 ± 13.44%, 암컷 마우스 (n=15)에서 42.63 ± 10.41%를 차지하여, 말초 혈액에 25% 이상의 인간 CD45 세포가 있는 인간화 마우스 모델의 조건을 충족하였다. 이 후, 인간화된 마우스 모델에 PC3 (human prostate cancer cell line) 및 A549 (human lung cancer cell line)를 피하 주사하고 3일마다 부피를 측정하였다. 종양 세포 주입 11일차부터 3일마다 TB511을 투여하고 (도 77 및 85), 마우스에서의 종양 성장, 및 TME에 침투하는 인간 면역세포의 변화를 t-SNE 분석으로 조사하였다. To evaluate the clinical potential of TB511, 4-week-old SID mice (NOD-Prkdc em1Beak IL2rg em1Break ), which are immunodeficient mice, were irradiated with 1.5 Gy of gamma irradiation and intravenously injected with human CD34 + hematopoietic stem cells (Lonza, Basel, Switzerland) at 2Х10 5 cells/mouse to generate a humanized mouse model. Eleven weeks after stem cell injection, peripheral blood was collected, and flow cytometry was used to confirm the engraftment of mature human leukocytes (human CD45 + cells). The hCD45 + cell population accounted for 37.32 ± 13.44% in male mice (n = 16) and 42.63 ± 10.41% in female mice (n = 15) 11 weeks after transplantation, which met the condition of a humanized mouse model with more than 25% human CD45 cells in the peripheral blood. Then, PC3 (human prostate cancer cell line) and A549 (human lung cancer cell line) were injected subcutaneously into the humanized mouse model, and the volume was measured every 3 days. TB511 was administered every 3 days from
그 결과, PC3 종양 세포 또는 A549 세포가 주입된 면역결핍 마우스에서 TB511의 항종양 효과가 완전히 사라진 것으로 나타난 반면 (도 78 및 도 84), CD34+ 이식된 마우스에서는 TB511가 종양 성장을 억제하는 것으로 나타나 (도 79 및 85), 온전한 면역 시스템이 TB511의 항종양 효과에 필수적임을 확인하였다. 또한, TME에 침투하는 인간 면역세포를 분석한 결과, TB511처리 후 M2-TAM (CD11b+ CD206+/CD45+ 세포)의 숫자는 현저하게 감소하였으며, M1-TAM (CD11b+ CD86+/CD45+ 세포)의 숫자는 변화하지 않아, M1/M2 비율이 증가한 것으로 나타났다 (도 80 내지 82). 또한, CD335+ NK 및 CD8+ T 세포를 포함하는, 종양-살해 세포(tumor-killer cells)들은 TB511처리 후 현저하게 TME에 침윤하는 것으로 나타났다 (도 80 내지 82). As a result, the antitumor effect of TB511 was completely lost in immunodeficient mice injected with PC3 tumor cells or A549 cells (Figs. 78 and 84), whereas TB511 inhibited tumor growth in CD34 + transplanted mice (Figs. 79 and 85), confirming that an intact immune system is essential for the antitumor effect of TB511. In addition, as a result of analyzing human immune cells infiltrating the TME, the number of M2-TAM (CD11b + CD206 + /CD45 + cells) was significantly reduced after TB511 treatment, while the number of M1-TAM (CD11b + CD86 + /CD45 + cells) did not change, indicating that the M1/M2 ratio increased (Figs. 80 to 82). Additionally, tumor-killer cells, including CD335 + NK and CD8 + T cells, were shown to significantly infiltrate the TME after TB511 treatment (Figs. 80 to 82).
10-2. TB511 및 면역 체크포인트 억제제의 단독 또는 조합 효과 분석10-2. Analysis of the effects of TB511 and immune checkpoint inhibitors alone or in combination
TB511의 면역 체크포인트 억제제와의 병용 처리에 의한 효과를 확인하기 위해, A549 세포를 주입한 인간화 마우스에 TB511 단독, 항-PD-1 항체 (pembrolizumab) 단독, 또는 TB511 및 항-PD-1 항체의 병용 투여하고 종양 부피를 측정하여 항종양 효과를 분석하고, 종양-침윤 면역세포 집단의 변화를 분석하였다.To determine the effect of combined treatment with immune checkpoint inhibitors of TB511, humanized mice injected with A549 cells were administered TB511 alone, anti-PD-1 antibody (pembrolizumab) alone, or a combination of TB511 and anti-PD-1 antibody, and the antitumor effect was analyzed by measuring tumor volume and analyzing changes in tumor-infiltrating immune cell populations.
항종양 효과를 분석한 결과, TB511 단독 투여 그룹과 항-PD-1 항체 단독 투여 그룹의 항종양 효과는 유사한 정도인 것으로 나타났으며, 이들을 병용 투여한 그룹의 항종양 효과는 현저하게 증가한 것으로 나타났다 (도 85). 또한, 종양-침윤 면역세포 집단 변화를 분석한 결과, TB511및 항-PD-1 항체를 병용 투여한 그룹에서 M1-TAMs (CD206+ CD11b+/CD45+ 세포)의 세포수가 현저하게 증가하였으며, M2-TAMs (CD86+ CD11b+/CD45+ 세포)의 세포수가 현저하게 감소한 긋으로 나타났다 (도 86 내지 88). 또한, Granzyme B-양성 CD8+ T 세포가 TB511 및 항-PD-1 항체를 병용 투여에 의해 현저히 증가하여 TME에 침윤이 증가한 것으로 확인되었으며, 고갈된 CD8+ T-세포 (PD-1+ CD8+/CD45+ 세포) 집단은 TB511 단독 투여 그룹, 및 TB511및 항-PD-1 항체 병용 투여 그룹에서 현저히 감소하였다 (도 89 내지 91).As a result of analyzing the antitumor effect, the antitumor effects of the TB511 monotherapy group and the anti-PD-1 antibody monotherapy group were found to be similar, while the antitumor effect of the group administered in combination was found to be significantly increased (Fig. 85). In addition, as a result of analyzing the change in the tumor-infiltrating immune cell population, the cell number of M1-TAMs (CD206 + CD11b + /CD45 + cells) was found to be significantly increased in the group administered in combination with TB511 and anti-PD-1 antibody, while the cell number of M2-TAMs (CD86 + CD11b + /CD45 + cells) was found to be significantly decreased (Figs. 86 to 88). In addition, Granzyme B-positive CD8 + T cells were significantly increased by co-administration of TB511 and anti-PD-1 antibody, indicating increased infiltration into the TME, while the depleted CD8 + T-cell (PD-1 + CD8 + /CD45 + cell) population was significantly decreased in the TB511 monotherapy group and the TB511 and anti-PD-1 antibody co-administration group (Figs. 89 to 91).
10-3. TB511 및 면역 체크포인트 억제제의 병용 투여에 의한 시너지 효과 분석10-3. Analysis of synergistic effects by combined administration of TB511 and immune checkpoint inhibitors
TB511 및 항-PD-1 항체 (pembrolizumab)의 조합에 의한 시너지(synergistic) 효과를 확인하기 위해, 인간의 전장 PD-1 단백질을 발현하는 유전자 변형된 C57BL/6J 마우스 (humanized PD-1 knock-in mice) (Shanghai Model Organisms Center, Shanghai, China)의 등 피부 피하에 인간 PD-L1을 안정적으로 발현하는 마우스 대장암 MC38 세포 (hPD-L1 MC38 세포) (Shanghai Model Organisms Center, Shanghai, China)를 주입하여 hPD-L1 MC38 종양-인간화된 PD-1 마우스 모델을 제조하였다 (도 96). 마우스를 PBS, TB511, 항-PD-1 항체 (Selleckchem), 또는 TB511 및 항-PD-1 항체 투여군으로 나눈 뒤, PBS 또는 TB511 (200 nmol/kg)을 피하 주사로 투여하고, 항-PD-1 항체 (2.5 mg/kg)를 주 2회 복강 주사로 32일 동안 투여하여, TB511및 항-PD-1 항체 병용 투여에 의한 시너지 효과를 확인하였다 (도 94 내지 99).To confirm the synergistic effect of the combination of TB511 and anti-PD-1 antibody (pembrolizumab), a hPD-L1 MC38 tumor-humanized PD-1 mouse model was generated by subcutaneously injecting human PD-L1-expressing mouse colon cancer MC38 cells (hPD-L1 MC38 cells) (Shanghai Model Organisms Center, Shanghai, China) stably expressing human PD-L1 into the dorsal skin of genetically modified C57BL/6J mice (humanized PD-1 knock-in mice) (Shanghai Model Organisms Center, Shanghai, China) (Figure 96). Mice were divided into groups administered PBS, TB511, anti-PD-1 antibody (Selleckchem), or TB511 and anti-PD-1 antibody, and then PBS or TB511 (200 nmol/kg) was administered subcutaneously, and anti-PD-1 antibody (2.5 mg/kg) was administered intraperitoneally twice a week for 32 days to confirm the synergistic effect of co-administration of TB511 and anti-PD-1 antibody (Figs. 94 to 99).
실시예 11. 대장암 동물모델에서 TB511와 화학항암제(oxaliplatin)의 병용투여에 따른 항암 효과 분석Example 11. Analysis of anticancer effects of combined administration of TB511 and chemotherapy (oxaliplatin) in a colon cancer animal model
11-1.세포배양11-1. Cell culture
RPMI1640 배지(Welgene, Gyeongsan, Korea)에서 마우스 대장암 세포(CT-26)를 배양하였으며, 상기 배지에는 10% 우태아혈청(FBS; Welgene, Gyeongsan, Korea)과 100U/ml 페니실린, 100μg/ml 스트렙토마이신(Invitrogen, CA, USA)이 첨가되었다. 세포는 80% 수렴에 도달하면 3일마다 세포를 분주하였으며, 37℃ 5% CO2에서 배양하였다.Mouse colon cancer cells (CT-26) were cultured in RPMI1640 medium (Welgene, Gyeongsan, Korea) supplemented with 10% fetal bovine serum (FBS; Welgene, Gyeongsan, Korea), 100 U/ml penicillin, and 100 μg/ml streptomycin (Invitrogen, CA, USA). Cells were seeded every 3 days when 80% confluent was reached, and cultured at 37°C and 5% CO 2 .
11-2. 대장암 마우스 모델 설정11-2. Establishment of colon cancer mouse model
대장암 마우스 모델을 확립하기 위해 CT-26 세포를 수확하여 매트리젤(matrigel)과 1:1 비율로 혼합하였다. 혼합된 세포는 3 Х 10^5 세포/마우스의 농도로 마우스의 오른쪽 옆구리에 피하 주입되었다. 종양 부피가 50-100 mm³에 도달하면, 마우스는 대조군, TB511, 옥살리플라틴(Oxaliplatin, Oxp) 및 병용처리군의 네 그룹으로 분류하였다. 각 그룹의 마우스에 PBS 또는 TB511 (200 nmol/kg)을 3일마다 피하 주사하고, OxP(1.5 mg/kg)를 단일 용량으로 복강 내 주사하였다. 종양 부피는 디지털 캘리퍼스를 사용하여 3일마다 측정하였다. 종양 부피 계산 공식은 V = (폭 Х 폭 Х 길이) / 2이다. 동물 실험은 경희대학교 기관 동물 관리 및 사용 위원회(KHSAP(SE)-24-332)의 승인을 받았다. 모든 마우스는 특정 병원체가 없는 환경에서 12시간 주기적인 빛/어두운 환경에서 사료와 물에 자유롭게 접근할 수 있도록 사육되었다. 분석을 수행한 후 모든 동물은 이소플루란과 경추 탈구를 사용하여 안락사 처리되었다. 종양 조직은 분리되어 추가적인 분석에 사용하였다.To establish a mouse model of colon cancer, CT-26 cells were harvested and mixed with Matrigel at a 1:1 ratio. The mixed cells were injected subcutaneously into the right flank of the mice at a concentration of 3
11-3. 유세포분석(Flow cytometry)11-3. Flow cytometry
종양 조직은 잘게 자르고 DNase I (1U/mL)와 콜라겐분해효소 D (1mg/mL)를 포함한 혈청이 없는 세포 배양 배지에서 분해하였다. 샘플은 37℃에서 부드럽게 교반하며 1시간동안 배양하였다. 샘플은 100-μm 나일론 메쉬 체를 통해 여과하였다. 적혈구는 Pharmlyse 버퍼 (BD Bioscience, CA, USA)로 용해시켰다. 단일 세포는 40-μm 나일론 메쉬 체를 통해 걸러낸 후, 다음 항체들로 염색하였다. 마우스 림프구의 경우, anti-CD45-APCcy7 (클론 30-F11; BD Bioscience, #557659), anti-CD4-BB700 (클론 RM4-5; BD Bioscience, #566407), anti-CD8-PE-cy7 (클론 53-6.7; BD Bioscience, #552877), anti-NKp46-BV711 (클론 29A1.4; BD Bioscience, #740822); 마우스 대식세포의 경우, anti-CD45-APCcy7 (클론 30-F11; BD Bioscience, #557659), anti-CD11b-BV510 (클론 M1/70; BD Bioscience, #566117), anti-F4/80-PE (클론 BM8; Biolegend, #123110), anti-CD206-APC (클론 C068C2; Biolegend, #141708), anti-CD86-BV786 (클론 GL1; BD Bioscience, #740900)였다. 세포는 FACS Lyric 시스템 (BD Bioscience)에서 검출하였으며, FlowJo 소프트웨어 (BD Bioscience)를 사용하여 분석하였다.Tumor tissues were minced and digested in serum-free cell culture medium containing DNase I (1 U/mL) and collagenase D (1 mg/mL). The samples were incubated for 1 h at 37°C with gentle agitation. The samples were filtered through a 100-μm nylon mesh sieve. Red blood cells were lysed with Pharmlyse buffer (BD Bioscience, CA, USA). Single cells were filtered through a 40-μm nylon mesh sieve and stained with the following antibodies. For mouse lymphocytes, anti-CD45-APCcy7 (clone 30-F11; BD Bioscience, #557659), anti-CD4-BB700 (clone RM4-5; BD Bioscience, #566407), anti-CD8-PE-cy7 (clone 53-6.7; BD Bioscience, #552877), anti-NKp46-BV711 (clone 29A1.4; BD Bioscience, #740822); For mouse macrophages, the following antibodies were used: anti-CD45-APCcy7 (clone 30-F11; BD Bioscience, #557659), anti-CD11b-BV510 (clone M1/70; BD Bioscience, #566117), anti-F4/80-PE (clone BM8; Biolegend, #123110), anti-CD206-APC (clone C068C2; Biolegend, #141708), anti-CD86-BV786 (clone GL1; BD Bioscience, #740900). Cells were detected on a FACS Lyric system (BD Bioscience) and analyzed using FlowJo software (BD Bioscience).
11-4. 통계 분석11-4. Statistical Analysis
모든 데이터는 평균 ± 표준 오차 (SEMs)로 제시되었다. 통계 분석은 Prism 10.1.2 소프트웨어(GraphPad Software Inc., CA, USA)를 사용하여 그룹 비교를 위한 일원 배치 분산 분석(one-way ANOVA) 후 Tukey의 사후 분석 또는 이원 배치 분산 분석(two-way ANOVA) 후 Bonferroni의 사후 분석으로 수행되었다.All data are presented as means ± standard errors of means (SEMs). Statistical analyses were performed using Prism 10.1.2 software (GraphPad Software Inc., CA, USA) using one-way ANOVA followed by Tukey's post hoc analysis for group comparisons or two-way ANOVA followed by Bonferroni's post hoc analysis.
11-5. 대장암 마우스 모델에서 TB511 및 옥살리플라틴(oxaliplatin) 병용투여시 항암 효과 분석11-5. Analysis of anticancer effects of combined administration of TB511 and oxaliplatin in a colon cancer mouse model
상기 실시예 11-2에 따라 대장암 마우스 모델에 TB511 및 옥살리플라틴(oxaliplatin)을 단독 또는 병용투여하였다.마우스는 7일째에 옥살리플라틴을 단일 용량으로 복강 내 주사하고, 3일마다 TB511을 주사하였다(도 100). 대조군 대비 TB511과 옥살리플라틴 군에서 종양 부피가 유의미하게 감소한 것으로 나타났으며, 병용투여군은 다른 군들보다 가장 높은 항암 효능을 나타내었고, 대조군과 TB511 군 대비 종양 성장이 유의적으로 억제되었다(도 101 내지 105).According to the above Example 11-2, TB511 and oxaliplatin were administered alone or in combination to a colon cancer mouse model. The mice were injected intraperitoneally with a single dose of oxaliplatin on the 7th day, and TB511 was injected every 3 days (Fig. 100). Compared to the control group, the tumor volume was significantly reduced in the TB511 and oxaliplatin groups, and the combination group showed the highest anticancer efficacy compared to the other groups, and tumor growth was significantly inhibited compared to the control and TB511 groups (Figs. 101 to 105).
11-6. TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 의한 종양 무게 및 마우스 체중 변화 분석11-6. Analysis of changes in tumor weight and mouse body weight by combined administration of TB511 and oxaliplatin
안락사 시킨 마우스에서 분리된 종양 조직의 무게 변화를 TB511과 옥살리플라틴 처리에 따라 측정하였다. 그 결과, 병용처리군에서 대조군 대비 종양 크기 및 무게가 유의적으로 감소하는 것으로 나타났다(도 106 및 도 108). 또한, TB511 또는 옥살리플라틴 투여로 인한 체중 변화를 확인한 결과, 어떤 군에서도 체중 변화는 나타나지 않았다(도 109).The weight changes of tumor tissues isolated from euthanized mice were measured according to TB511 and oxaliplatin treatment. As a result, it was shown that the tumor size and weight were significantly reduced in the combined treatment group compared to the control group (Fig. 106 and Fig. 108). In addition, when the weight change due to TB511 or oxaliplatin administration was checked, no weight change was observed in any group (Fig. 109).
11-7. TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 의한 종양 미세 환경(Tumor Micro Environment, TME)에서 M2 유사 TAM(Tumor Associated Macrophage)의 감소11-7. Reduction of M2-like TAM (Tumor Associated Macrophage) in the Tumor Microenvironment (TME) by Combination Administration of TB511 and Oxaliplatin
TB511이 TME에서 대식세포 하위 유형 중 M2 유사 TAM을 선택적으로 표적하는지 확인하였다. 종양 조직을 단일 세포로 만든 후, TME 내 대식세포 하위 유형의 분포를 유세포분석을 통해 분석하였다. 유세포분석 결과, M1 유사 TAM(CD45+ CD11b+ 세포 내의 CD86+ F4/80+ 세포)의 분포는 모든 군에서 유의적인 변화가 나타나지 않았다. 다만, M2 유사 TAM(CD45+ CD11b+ 내의 CD206+ F4/80+ 세포)의 분포는 TB511 군에서 대조군 대비 유의적으로 감소하였다. 또한, 병용처리군에서 M2 유사 TAM이 대조군 대비 유의적으로 감소하였다(도 109 내지 112). 또한, 병용처리군의 경우, TME에서 M2 유사 TAM의 감소가 M1/M2 비율을 증가시켰음을 확인하였다(도 113). 상기 결과들을 통해, TB511 및 chemotherapy 병용투여시 TME에서 M2 유사 TAM을 제거하여, 항종양 환경으로 변화를 유도하는 것을 확인하였다.We confirmed that TB511 selectively targets M2-like TAM among macrophage subtypes in the TME. After making tumor tissues into single cells, the distribution of macrophage subtypes in the TME was analyzed by flow cytometry. The flow cytometry results showed that the distribution of M1-like TAM (CD86 + F4/80 + cells within CD45 + CD11b + cells) did not show significant changes in any group. However, the distribution of M2-like TAM (CD206 + F4/80 + cells within CD45 + CD11b + ) significantly decreased in the TB511 group compared to the control group. In addition, M2-like TAM significantly decreased in the combination treatment group compared to the control group (Figs. 109 to 112). In addition, in the combination treatment group, it was confirmed that the decrease in M2-like TAM in the TME increased the M1/M2 ratio (Fig. 113). Through the above results, we confirmed that combined administration of TB511 and chemotherapy eliminated M2-like TAMs in the TME, thereby inducing a change to an anti-tumor environment.
11-7. TB511 및 옥살리플라틴(oxaliplatin) 병용투여에 의한 세포독성 T세포의 TME로의 침투 유도 분석11-7. Analysis of induction of cytotoxic T cell infiltration into TME by combined administration of TB511 and oxaliplatin
TB511 유도 M2 유사 TAM의 제거에 인한 TME 내 림프구 세포의 분포 변화를 분석하였다. CD4 T 세포(CD45+ CD3+ CD4+ 세포), CD8 T 세포(CD45+ CD3+ CD8+ 세포) 및 NK세포(CD45+ CD3+ NKp46+ 세포)를 포함한 림프구 세포의 분포는 유세포분석을 통해 분석하였다. CD4 T 세포와 NK 세포의 분포는 모든 군에서 유의적인 차이를 나타내지 않았다. 그러나, CD8 T 세포의 경우, 병용투여군에서 유의적으로 증가하였다. 상기 결과를 통해, TB511 처리에 따른 M2 유사 TAM 감소가 CD8 T 세포의 종양 내 침투를 증가시켜, 종양 세포를 직접 제거하는 활성을 나타냄을 확인하였다.We analyzed the changes in the distribution of lymphocytes in the TME due to the removal of TB511-induced M2-like TAMs. The distribution of lymphocytes, including CD4 T cells (CD45 + CD3 + CD4 + cells), CD8 T cells (CD45 + CD3 + CD8 + cells), and NK cells (CD45 + CD3 + NKp46 + cells), was analyzed by flow cytometry. The distribution of CD4 T cells and NK cells did not show significant differences in any group. However, CD8 T cells significantly increased in the combination treatment group. Through the above results, we confirmed that the decrease in M2-like TAMs due to TB511 treatment increased the infiltration of CD8 T cells into tumors, thereby exhibiting the activity of directly removing tumor cells.
상기 실시예들을 종합할때, TB511에 의한 M2 유사 TAM 제거가 옥살리플라틴의 항암 효능을 향상시키고, TB511 및 oxaliplatin 을 병용투여하였을 때 각각을 단독투여한 경우 대비 T 세포의 종양 침윤을 증가시켜 항종양 면역 환경을 촉진하는 것으로 나타났는바, TB511 및 oxaliplatin 병용투여에 따른 상승효과가 있음을 확인하였다.In summary of the above examples, it was shown that M2-like TAM elimination by TB511 enhanced the anticancer efficacy of oxaliplatin, and that combined administration of TB511 and oxaliplatin promoted an antitumor immune environment by increasing tumor infiltration of T cells compared to when each was administered alone, confirming the synergistic effect of combined administration of TB511 and oxaliplatin.
실시예 12. 삼중음성유방암 동물모델에서 TB511와 화학항암제(paclitaxel)의 병용투여에 따른 항암 효과 분석Example 12. Analysis of anticancer effects of combined administration of TB511 and chemotherapy (paclitaxel) in a triple-negative breast cancer animal model
12-1. 삼중음성유방암 마우스 모델 설정12-1. Establishment of triple-negative breast cancer mouse model
하기 분석은 Balb/c (BALB/cAnNTac, Taconic Bioscience) 암컷 마우스(6~8주)를 사용하여 수행하였다. Balb/c는 삼중음성 유방암 세포주 4T1의 이식에 적합한 strain으로, 면역학 및 종양 실험에서 널리 사용되기에 선정하였다. 마우스는 SPF 환경에서 22±1℃, 상대습도 55±15%, 12시간 주야조명(07:00 점등, 19:00 소등) 조건 하에 사육하였으며, W 260 Х L 420 Х H 180 mm의 사육상자에 최대 5마리씩 수용하고 개체 식별을 위해 꼬리에 유성펜 표식을 사용하였다. 사료와 물은 각각 실험동물용 고형사료와 물병을 통해 자유롭게 섭취하도록 하였다. 본 실험은 동물보호법에 의거하여 경희대학교 동물윤리위원회의 승인(KHUAP(SE)-20-398)을 받았다.The following analyses were performed using Balb/c (BALB/cAnNTac, Taconic Bioscience) female mice (6–8 weeks old). Balb/c is a strain suitable for transplantation of the triple-negative breast cancer cell line 4T1 and was selected because it is widely used in immunology and tumor experiments. Mice were housed in an SPF environment at 22±1℃, 55±15% relative humidity, and 12-h day/night lighting cycle (lights on at 07:00, lights off at 19:00). Up to five mice were housed in cages (W 260 X L 420
12-2. 종양 부피 측정12-2. Tumor volume measurement
암컷 Balb/c 마우스(BALB/cAnNTac, Taconic Bioscience)를 사용하여 삼중음성 유방암 세포인 4T1(ATCC)을 네 번째 젖꼭지에 1Х10개 세포씩 주사하였다. 4T1 세포는 serum-free media에 현탁 후 Matrigel(Corning)과 혼합하여 종양 형성을 촉진하였다. 대조군으로서 PBS를 처리한 그룹을 설정하였으며, 그룹당 18마리의 마우스에 대해 분석을 약물투여 및 분석을 수행하였다. 종양세포 주입 후 5일이 되는 시점부터 TB511 및 파클리탁셀(paclitaxel)을 3일 간격으로 단일투여 또는 병용투여하였다. TB511은 피하주사를 통해 투여하였으며, 파클리탁셀(paclitaxel)은 복강주사를 통해 투여하였다(도 118). 종양의 크기는 디지털 캘리퍼로 측정하여 부피를 산출하였다. Female Balb/c mice (BALB/cAnNTac, Taconic Bioscience) were injected with triple-negative breast cancer cells, 4T1 (ATCC), at a density of 1Х10 cells per fourth nipple. The 4T1 cells were suspended in serum-free media and mixed with Matrigel (Corning) to promote tumor formation. A group treated with PBS was set as a control group, and drug administration and analysis were performed on 18 mice per group. TB511 and paclitaxel were administered as a single administration or in combination at 3-day intervals starting 5 days after tumor cell injection. TB511 was administered by subcutaneous injection, and paclitaxel was administered by intraperitoneal injection (Fig. 118). The tumor size was measured using a digital caliper to calculate the volume.
12-3. 통계학적 방법12-3. Statistical Methods
측정 결과에 대한 유의성 차이 분석은 Graphpad Prism software (Version 5, CA, USA)에서 평균값을 분산분석 한 후, unpaired T test에 따라 P-value가 0.05보다 작을 때 통계적으로 유의한 차이를 나타낸다고 판단하였다. 모든 실험은 맹검법으로 수행되었으며 동일한 조건 하에서 독립적으로 반복되었다.The significance difference analysis for the measurement results was performed by analyzing the mean values using Graphpad Prism software (
12-4. TB511와 화학항암제(paclitaxel)의 병용투여에 따른 항암 효과 분석12-4. Analysis of anticancer effects of combined administration of TB511 and chemotherapy (paclitaxel)
상기 실시예 12-2에 따라 TB511 또는 파클리탁셀(paclitaxel)을 투여하였을때의 종양크기 변화를 측정하였고, 이를 통해 TB511 및 파클리탁셀(paclitaxel)을 병용투여하였을때의 항암 효과를 확인하였다. 분석 결과를 도 119에 나타내었다. 도 119를 살펴보면, TB511 및 파클리탁셀(paclitaxel)을 병용투여한 그룹에서 TB511 및 paclitaxel을 단독으로 투여한 그룹, 대조군 대비 종양의 크기가 유의적으로 작게 나타났다. 상기 결과를 통해, TB511 및 파클리탁셀(paclitaxel)을 병용투여한 경우, 상승된 항암효과를 나타내는 것을 확인하였다.According to the above Example 12-2, when TB511 or paclitaxel was administered, the change in tumor size was measured, and through this, the anticancer effect of combined administration of TB511 and paclitaxel was confirmed. The analysis results are shown in Fig. 119. As can be seen in Fig. 119, the tumor size in the group administered combined administration of TB511 and paclitaxel was significantly smaller compared to the group administered alone of TB511 and paclitaxel and the control group. Through the above results, it was confirmed that when TB511 and paclitaxel were administered combinedly, an enhanced anticancer effect was exhibited.
실시예 13. 인간화 췌장암 마우스모델에서 TB511 및 PADCEV(anti-Nectin-4 antibody antibody drug conjugate)의 병용투여에 따른 항암 효과 분석Example 13. Analysis of anticancer effects of combined administration of TB511 and PADCEV (anti-Nectin-4 antibody antibody drug conjugate) in a humanized pancreatic cancer mouse model
13-1. 동물모델 설정13-1. Setting up the animal model
SID 마우스(NOD-Prkdcem1BeakIL2rgem1Break, 4주령)를 GemBioscience(충북, 대한민국)에서 구매하여 사용하였다. 마우스는 1.5 Gy의 γ-선으로 조사된 후, human CD34+ 조혈모세포(Lonza, 스위스 바젤) 2Х10 세포를 정맥으로 주사하였다. SID 마우스는 인간화된 면역세포를 발현할 수 있는 인간화 마우스 모델로 적합하며, 크기가 작고 다루기 쉬워 실험에 용이하고, 인간 유래 hHSC 세포의 이식을 위해 면역반응을 생성하지 않는 면역결핍 특성을 가지고 있어 선택되었다. 투여 시작 시 마우스는 16주령이었으며, 총 20마리의 암컷 인간화 마우스가 사용되었다. 동물은 W 200 Х L 260 Х H 130(mm)의 폴리카보네이트 케이지에 방사선 조사된 깔개를 사용하여 최대 5마리씩 사육되었으며, 꼬리에 표식을 하여 개체를 식별하고, 케이지에는 시험 번호 및 군 정보를 기재한 라벨을 부착하였다. 사육 조건은 100% HEPA 필터를 통과한 공기, 시간당 최소 10회의 공기 교체, 20.2-23.8℃의 온도, 41.9-55.4%의 상대습도, 150-300 Lux의 광도로 12시간 주간 및 야간 주기로 유지하였다. 모든 동물에게 표준 방사선 조사된 펠렛형 실험용 사료(Purina Rodent Chow 38057)와 물을 자유롭게 제공하였다. 분석은 경희대학교 동물윤리위원회(KHUASP(SE)-20-308)의 승인을 받아 동물보호법 및 관련 규정에 따라 수행하였다..SID mice (NOD-Prkdcem1BeakIL2rgem1Break, 4 weeks old) were purchased from GemBioscience (Chungbuk, South Korea). Mice were irradiated with 1.5 Gy of γ-rays and then intravenously injected with human CD34+ hematopoietic stem cell 2Х10 cells (Lonza, Basel, Switzerland). SID mice were selected because they are a suitable humanized mouse model that can express humanized immune cells, are small in size and easy to handle, are convenient for experiments, and have immunodeficiency characteristics that do not generate an immune response for transplantation of human-derived hHSC cells. Mice were 16 weeks old at the start of administration, and a total of 20 female humanized mice were used. Animals were housed in polycarbonate
13-2. 인간화 마우스 모델 설정13-2. Setting up a humanized mouse model
인간화 마우스 모델을 확립하기 위해, 4주령 수컷 면역결핍 마우스(SID)를 100 cGy로 1분간 방사선 조사하였고, 한 시간 이내에 human hematopoietic stem cells(hHSCs)을 30G 인슐린 주사기를 이용하여 꼬리정맥으로 주입하였다. 인간 면역세포의 발현은 10주차 및 16주차에 유세포 분석법(flow cytometry)을 통해 확인하였으며, hCD45+ 세포 발현율이 25% 이상인 개체를 실험에 사용하였다. 하기 표 2에 마우스의 면역세포 발현 비율을 나타내었다. 분석에 사용된 인간화 마우스는 Lonza에서 공급받았다.To establish a humanized mouse model, 4-week-old male immunodeficient mice (SID) were irradiated with 100 cGy for 1 minute, and human hematopoietic stem cells (hHSCs) were injected into the tail vein using a 30G insulin syringe within one hour. The expression of human immune cells was confirmed by flow cytometry at 10 and 16 weeks, and individuals with hCD45+ cell expression rate of 25% or higher were used in the experiment. Table 2 shows the immune cell expression rate of the mice. The humanized mice used in the analysis were supplied by Lonza.
또한, 폐암 인간화 마우스 모델을 확립하기 위해, 인간 폐암 세포주(PANC1; ATCC)를 마우스 당 2Х10 세포 농도로 준비한 후, Matrigel(Corning)과 1:1로 혼합하고 30G 주사기를 이용해 혼합액 100 μL를 마우스의 오른쪽 등 부위 피하에 주사하였다.In addition, to establish a humanized mouse model of lung cancer, human lung cancer cell lines (PANC1; ATCC) were prepared at a concentration of 2Х10 cells per mouse, mixed 1:1 with Matrigel (Corning), and 100 μL of the mixture was injected subcutaneously into the right dorsal area of the mice using a 30G syringe.
13-3. 약물 투여 방법13-3. Drug administration method
TB511의 용량 제형은 스폰서에 의해 D-PBS에서 5 mg/ml의 농도로 준비되었으며, 200 nmol/kg로 희석하여 투여하였다. TB511의 투여 용량은 폐암 마우스 모델에서 이전에 수행된 효능 평가 결과를 기반으로 200 nmol/kg로 설정되었고, PADCEV(anti-Nectin-4 antibody drug conjugate)는 외부 데이터를 참고하여 5 mg/kg로 설정되었다. 마우스는 4개의 그룹으로 나뉘어 연구를 진행하였다: G1 (PBS 투여군), G2 (anti-nectin-4 투여군, 5 mg/kg), G3 (TB511 투여군, 200 nmol/kg), G4 (병용 투여군, PADCEV 5 mg/kg + TB511 200 nmol/kg).The dosage form of TB511 was prepared by the sponsor at a concentration of 5 mg/ml in D-PBS and diluted to 200 nmol/kg for administration. The administration dose of TB511 was set to 200 nmol/kg based on the results of the previous efficacy evaluation in a lung cancer mouse model, and PADCEV (anti-Nectin-4 antibody drug conjugate) was set to 5 mg/kg with reference to external data. The mice were divided into four groups for the study: G1 (PBS administration group), G2 (anti-nectin-4 administration group, 5 mg/kg), G3 (TB511 administration group, 200 nmol/kg), and G4 (combination administration group,
TB511은 70% 알코올 솜으로 소독한 뒤, 인슐린 주사기를 이용해 마우스의 등 부위에 피하주사로 투여되었으며, PADCEV는 같은 방법으로 복강 내에 주사하였다. 두 약물 모두 보조제의 용량은 100 μL로 동일하며, 투여 간격은 3일마다 이루어졌다.TB511 was administered subcutaneously into the back of mice using an insulin syringe after disinfecting with a 70% alcohol swab, and PADCEV was injected intraperitoneally using the same method. The adjuvant volume for both drugs was the same at 100 μL, and the administration interval was every 3 days.
13-4. 종양 부피 측정13-4. Tumor volume measurement
종양 부피는 공식 V = (너비(2) Х 길이)/2를 사용해서 계산하였다. 가이드라인에 따르면, 마우스는 종양 크기가 접종 후 최대 직경 1~1.5 cm에 도달하면 희생시켰다.Tumor volume was calculated using the formula V = (width (2) Х length)/2. According to the guidelines, mice were sacrificed when tumor size reached a maximum diameter of 1–1.5 cm after inoculation.
13-5. 유세포분석 (Flow Cytometry) 분석13-5. Flow Cytometry Analysis
뇌, 유방, 대장, 신장, 간, 폐, 피부, 종양 조직은 MACS 분해기(Milteny Biotec, Auburn, CA, USA)를 사용해서 얇은 조각으로 자르고, 1 U/mL DNase 캧ndianapolis, IN, USA)과 1 mg/mL 콜라겐분해효소 D (Roche)를 RPMI 1640 무혈청 배지에서 37℃에서 30분 동안 배양하였다. 조직은 100 μm 나일론 메쉬 필터로 걸러졌다. 필터링된 단일 세포에서 적혈구는 BD Pharm lyse buffer (BD Bioscience, CA, USA)로 용해하였다. 단일 세포는 이후 BD Pharmingen Stain Buffer (BD Bioscience)와 함께 다음 항체들로 염색하였다. 인간 대식세포의 경우, 항-CD45-FITC, 항-CD11b-BV510, 항-CD18-PE, 항-KIM127-BV786(BD Bioscience)였다. 세포는 FACS Lyric 시스템(BD Bioscience)을 이용하여 탐지하였고, FlowJo 소프트웨어(BD Bioscience)를 사용하여 분석하였다.Brain, breast, colon, kidney, liver, lung, skin, and tumor tissues were cut into thin pieces using a MACS digester (Milteny Biotec, Auburn, CA, USA) and incubated in RPMI 1640 serum-free medium containing 1 U/mL DNase (Cincinnati, Indianapolis, IN, USA) and 1 mg/mL collagenase D (Roche) at 37°C for 30 min. Tissues were filtered through a 100 μm nylon mesh filter. Red blood cells in the filtered single cells were lysed with BD Pharm lyse buffer (BD Bioscience, CA, USA). Single cells were then stained with the following antibodies in BD Pharmingen Stain Buffer (BD Bioscience): anti-CD45-FITC, anti-CD11b-BV510, anti-CD18-PE, and anti-KIM127-BV786 (BD Bioscience) for human macrophages. Cells were detected using a FACS Lyric system (BD Bioscience) and analyzed using FlowJo software (BD Bioscience).
13-6. 면역조직화학(Immunohistochemistry, IHC)13-6. Immunohistochemistry (IHC)
조직학적 분석을 위해 종양 조직은 10% 중성 완충 포르말린에 하룻밤 동안 고정하였다. 그 후 파라핀에 포함시킨 종양 조직은 5 μm 두께로 절단하였다. 슬라이드는 자일렌으로 탈파라핀화하고, 에탄올과 탈이온수로 탈수하였다. 항원 회복을 위해 슬라이드는 나트륨 시트르산 완충액(pH 6.0)에 15분간 전자레인지로 인큐베이션하였다. 그 후 슬라이드는 15분 동안 과산화효소 차단 용액(Dako, Glostrup, Denmark)으로 처리하고, 5% 소의 혈청알부민으로 차단하였다. 슬라이드는 1차 항체와 함께 하룻밤 동안 인큐베이션 한 후, 0.1% Tween20이 포함된 TBS로 세척하고, avidin-biotin complex 키트(Vector Laboratories, Burlingame, CA, USA)와 함께 인큐베이션 하였으며, diaminobenzidine-HCL(Vector Laboratories)로 시각화하였다. 핵은 헤마톡실린으로 반대 염색하였다. 슬라이드는 밝은 필드 현미경(Nikon, Tokyo, Japan)으로 관찰하고, ImageJ 소프트웨어(NIH)를 사용하여 분석하였다. IHC 염색에 사용된 항체는 항-KI67 및 항-CD18 항체(1:200; Abcam, Cambridge, UK)였다.For histological analysis, tumor tissues were fixed overnight in 10% neutral buffered formalin. The tumor tissues embedded in paraffin were then sectioned at 5 μm thickness. The slides were deparaffinized with xylene and dehydrated with ethanol and deionized water. For antigen retrieval, the slides were incubated in sodium citrate buffer (pH 6.0) for 15 min in a microwave oven. The slides were then treated with peroxidase blocking solution (Dako, Glostrup, Denmark) for 15 min and blocked with 5% bovine serum albumin. The slides were incubated with primary antibodies overnight, washed with TBS containing 0.1
13-7. 면역형광 염색(Immunofluorescence staining)13-7. Immunofluorescence staining
파라핀에 포함시킨 후, 종양 조직은 5 μm 두께로 절단하였다. 슬라이드는 자일렌으로 탈파라핀화하고, 에탄올과 탈이온수로 탈수하였다. 항원 회복을 위해 슬라이드는 나트륨 시트르산 완충액(pH 6.0)에 15분간 전자레인지로 인큐베이션하였다. 그 후 슬라이드는 15분 동안 peroxidase-blocking 용액(Dako, Glostrup, Denmark)으로 처리하고, 5% bovine serum albumin으로 차단하였다. 슬라이드는 1차 항체와 함께 하룻밤 동안 배양 한 후, 0.1% Tween20이 포함된 TBS로 세척하였다. 슬라이드는 항-비멘틴, 항-E-카드헤린, 항-액틴, 항-CD163, 항-CD11b, 항-Kim127, 항-CD8 항체(1:500; Abcam)와 함께 4℃에서 하룻밤 동안 배양하였다. 그 후, FITC로 표지된 TAMpep와 APC 결합 항-토끼 IgG 2차 항체(1:500, Invitrogen)로 37℃에서 1시간 동안 염색하였다. 핵을 시각화하기 위해, 커버글라스는 4‘,6-diamidino-2-phenylindole(DAPI)을 포함한 Vectashield 장착 매체(Vector Laboratories)로 장착하였다. 이미지는 LSM 800 공초점 레이저 스캐닝 현미경(Carl Zeiss, Oberkochen, Germany)을 사용하여 촬영하였다.After embedding in paraffin, tumor tissues were sectioned at 5 μm thickness. The slides were deparaffinized with xylene and dehydrated with ethanol and deionized water. For antigen retrieval, the slides were incubated in sodium citrate buffer (pH 6.0) for 15 min in a microwave oven. The slides were then treated with peroxidase-blocking solution (Dako, Glostrup, Denmark) for 15 min and blocked with 5% bovine serum albumin. The slides were incubated with primary antibodies overnight and then washed with TBS containing 0.1
13-8. 공간 전사체학 (Spatial Transcriptomics)13-8. Spatial Transcriptomics
파라핀에 포함된 블록은 5μm 두께로 정밀하게 절단되어 Xenium 슬라이드에 장착하였다. 절단된 슬라이드는 42℃에서 3시간 동안 열 순환기(C1000 96-well, Bio-Rad)에서 건조하였다. 그 후, 탈파라핀화(Deparaffinization) 및 탈교차결합(de-crosslinking)을 수행하였다. 혼성화(prime hybridization)와 RNase 처리가 완료된 후, 절단된 슬라이드는 Xenium 인간 5k 팬-티슈 프로브(10x Genomics)와 함께 50℃에서 하룻밤동안(~24시간) 혼성화를 수행하였다. 그 후 슬라이드는 세척되고, 42℃에서 30분 동안 결합(ligation)을 수행하였다. 증폭 강화 및 증폭은 열 순환기(C1000 96-well, Bio-Rad)에서 즉시 처리되었다. 슬라이드는 다시 세척되고, 세포 분할 염색 프로브를 4℃에서 하룻밤 동안 (~24시간) 적용하였다. 염색 강화 및 세척 후, 자가형광 억제 및 핵 염색을 수행하였다.Paraffin-embedded blocks were precisely sectioned into 5 μm thickness and mounted on Xenium slides. The sectioned slides were dried in a thermal cycler (C1000 96-well, Bio-Rad) at 42°C for 3 h. Deparaffinization and de-crosslinking were then performed. After prime hybridization and RNase treatment, the sectioned slides were hybridized overnight (~24 h) at 50°C with Xenium human 5k pan-tissue probe (10x Genomics). The slides were then washed and ligated at 42°C for 30 min. Amplification enhancement and amplification were immediately processed in a thermal cycler (C1000 96-well, Bio-Rad). The slides were washed again, and the cell division staining probe was applied overnight (~24 h) at 4°C. After staining enhancement and washing, autofluorescence suppression and nuclear staining were performed.
그 후, 절단된 슬라이드는 Xenium 분석기(XOA v3.0.2, 10x Genomics)에 로딩하여 형광 신호 감지를 수행하였다. Xenium 실행 후, 원시 데이터는 xeniumranger v3.0.0으로 전처리하였다.The cut slides were then loaded onto the Xenium analyzer (XOA v3.0.2, 10x Genomics) for fluorescence signal detection. After running Xenium, the raw data were preprocessed with xeniumranger v3.0.0.
13-9. 통계 분석13-9. Statistical Analysis
모든 데이터는 세 가지 또는 네 가지 독립적인 실험의 평균 ± 표준편차로 표현되었다. 비교는 두 표본 Mann-Whitney U-test, 일원 및 이원 분산 분석(ANOVA) (IBM SPSS Statistics 버전 21; IBM Inc., Chicago, IL, USA)을 사용해서 수행하였으며, P값 < 0.05는 통계적으로 유의미한 것으로 간주하였다.All data are expressed as the mean ± standard deviation of three or four independent experiments. Comparisons were performed using the two-sample Mann-Whitney U-test, one- and two-way analysis of variance (ANOVA) (IBM
13-10. 인간 췌장암 마우스 모델에서 TB511 및 PADCEV의 병용투여에 따른 종양 억제 분석13-10. Tumor suppression analysis of combined administration of TB511 and PADCEV in a human pancreatic cancer mouse model
TB511 및 PADCEV(항-nectin4 항체)을 병용투여할 때, 인간화된 췌장암 마우스 모델에서 종양 성장을 억제할 수 있는지 확인하기 위해, 인간 췌장암 세포주(PANC1)를 인간화된 마우스에 피하 주입하여 종양 마우스 모델을 설정하고, 마우스에 약물을 주 2회 투여했다. 종양 크기는 PBS 그룹 대비 TB511 또는 PADCEV 투여 그룹에서 유의적으로 감소하였으며, 특히, 병용 투여 그룹에서 가장 유의적인 감소를 나타내었다(도 120 및 도 121). 종양 조직에서는 증식 인자인 PCNA의 발현 수준이 PBS 그룹에 비해 TB511, PADCEV, 병용 그룹에서 유의적으로 감소한 것으로 나타났다(도 122 및 도 123). 또한, 상피세포 마커인 E-cadherin의 발현은 PBS 그룹에 비해 TB511, PADCEV, 병용 그룹에서 유의적으로 증가했으며, 간엽세포 마커인 vimentin의 발현은 E-cadherin과 반대되는 패턴을 나타내었고, TB511, PADCEV, 병용 그룹에서 PBS 그룹에 비해 유의적으로 감소한 것으로 나타났다(도 124 및 도 125). 상기 결과를 통해, TB511 및 PADCEV을 병용투여할 경우, 각각을 별도로 투여할 때 대비 인간화된 췌장암 마우스 모델에서 종양 진행을 억제하는 활성을 나타냄을 확인하였다.To determine whether combined administration of TB511 and PADCEV (anti-nectin4 antibody) could inhibit tumor growth in a humanized pancreatic cancer mouse model, a tumor mouse model was established by subcutaneously injecting human pancreatic cancer cell line (PANC1) into humanized mice, and the drug was administered to the mice twice a week. The tumor size was significantly reduced in the TB511 or PADCEV administration groups compared to the PBS group, and in particular, the combination administration group showed the most significant reduction (Figs. 120 and 121). In tumor tissues, the expression level of PCNA, a proliferation factor, was significantly reduced in the TB511, PADCEV, and combination groups compared to the PBS group (Figs. 122 and 123). In addition, the expression of E-cadherin, an epithelial cell marker, was significantly increased in the TB511, PADCEV, and combination groups compared to the PBS group, and the expression of vimentin, a mesenchymal cell marker, showed an opposite pattern to E-cadherin and was significantly decreased in the TB511, PADCEV, and combination groups compared to the PBS group (Figs. 124 and 125). Through the above results, it was confirmed that when TB511 and PADCEV were administered together, they exhibited an activity of inhibiting tumor progression in a humanized pancreatic cancer mouse model compared to when each was administered separately.
13-11. TB511 및 PADCEV의 병용투여에 따른 종양 조직에서 M2 대식세포의 감소13-11. Reduction of M2 macrophages in tumor tissues following combined administration of TB511 and PADCEV
TB511이 종양 미세환경 내 M2 대식세포를 감소시키는지 여부를 확인하기 위해, 종양 조직을 면역조직화학 염색과 유세포분석(flow cytometry)을 사용하여 분석하였다. 분석결과, TB511 및 병용 치료 그룹은 PBS 그룹에 비해 종양 조직에서 M2 대식세포 마커로 알려진 CD163을 발현하는 세포가 유의적으로 감소한 반면, PADCEV 그룹에서는 효과가 나타나지 않았다(도 126 및 도 127). 특히, TB511은 M2 대식세포에서 활성화된 CD18에 선택적으로 결합하는 것으로 나타났다. 종양 조직에서 CD18 양성 세포는 TB511 및 병용 치료 그룹에서 유의적으로 감소한 것으로 나타났다(도 128 및 도 129). 또한, TB511이 활성화된 CD18에 결합하는 Kim127 항체를 사용하여 M2 대식세포를 감소시키는 것으로 나타났다(도 130 내지 133). 상기 결과를 통해, TB511이 종양 조직 내 M2 대식세포의 활성화된 CD18을 선택적으로 표적으로 하여 이들 세포의 감소를 유도하는 것을 확인하였다.To determine whether TB511 reduces M2 macrophages in the tumor microenvironment, tumor tissues were analyzed using immunohistochemical staining and flow cytometry. The results showed that the TB511 and combination treatment groups significantly decreased cells expressing CD163, a marker of M2 macrophages, in tumor tissues compared to the PBS group, whereas no effect was observed in the PADCEV group (Figs. 126 and 127). In particular, TB511 was shown to selectively bind to activated CD18 in M2 macrophages. CD18-positive cells in tumor tissues were shown to significantly decrease in the TB511 and combination treatment groups (Figs. 128 and 129). In addition, it was shown that TB511 reduced M2 macrophages using the Kim127 antibody that binds to activated CD18 (Figs. 130 to 133). Through the above results, we confirmed that TB511 selectively targets activated CD18 of M2 macrophages in tumor tissues and induces a decrease in these cells.
13-12. TB511 및 PADCEV의 병용투여에 따른 종양 조직에서 CD8 T 세포의 증가13-12. Increase in CD8 T cells in tumor tissues following combined administration of TB511 and PADCEV
종양 미세환경 내 M2 대식세포는 면역억제 역할을 하여 CD8 T 세포의 유입을 억제한다고 보고된 바 있다(doi: 10.1186/s12964-023-01424-6). M2 대식세포를 표적으로 하는 TB511이 종양 미세환경에서 CD8 T 세포의 유입을 증가시키는지 확인하기 위해, 종양 조직을 면역형광 염색으로 분석하였다. 분석결과, 종양 조직에서 CD8 T 세포는 PBS 그룹에 비해 TB511 및 병용 치료 그룹에서 유의적으로 증가하였고, PADCEV 그룹에서는 유의미한 변화를 나타내지 않았다(도 132 및 도 133). 상기 결과를 통해, TB511이 M2 대식세포의 감소를 유도하여 CD8 T 세포의 유입을 촉진하는 것을 확인하였다. 다만, PADCEV는 종양 세포에 발현된 nectin-4를 표적으로 하여 종양 세포를 직접 타겟으로 하므로, 종양 미세환경에서 대식세포 및 CD8 T 세포와 같은 면역 세포에는 유의적인 변화를 나타내지 않았다.It has been reported that M2 macrophages in the tumor microenvironment play an immunosuppressive role and inhibit the influx of CD8 T cells (doi: 10.1186/s12964-023-01424-6). To confirm whether TB511 targeting M2 macrophages increases the influx of CD8 T cells in the tumor microenvironment, tumor tissues were analyzed by immunofluorescence staining. As a result of the analysis, CD8 T cells in the tumor tissues significantly increased in the TB511 and combination treatment groups compared to the PBS group, and no significant change was observed in the PADCEV group (Figs. 132 and 133). Through the above results, it was confirmed that TB511 induces a decrease in M2 macrophages and promotes the influx of CD8 T cells. However, since PADCEV directly targets tumor cells by targeting nectin-4 expressed on tumor cells, it did not show significant changes in immune cells such as macrophages and CD8 T cells in the tumor microenvironment.
13-13. 정상 조직에서 활성화된 CD18을 발현하는 대식세포 및 CD8 T 세포와 TB511의 연관성13-13. Association of TB511 with activated CD18-expressing macrophages and CD8 T cells in normal tissues
정상 조직에서 활성화된 CD18을 발현하는 대식세포와 CD8 T 세포와의 TB511의 연관성은 도 20에서 나타낸 바와 같이, TB511이 활성화된 CD18을 발현하는 대식세포가 감소하는 것으로 나타났다. TB511이 정상 조직의 대식세포와 CD8 T 세포에 미치는 영향을 조사하기 위해, 비장, 간, 폐, 뇌 조직에서 단일 세포를 얻어 Kim127 항체를 사용하여 활성화된 형태의 CD18을 염색하고, 유세포분석(flow cytometry)을 통해 분석하였다. TB511, PADCEV, 병용 치료 그룹은 PBS 그룹에 비해 Kim127 양성 세포인 대식세포와 CD8 T 세포에 영향을 미치지 않는 것으로 나타났다(도 134 내지 141). 상기 결과를 통해, TB511이 종양 조직에서 활성화된 CD18을 발현하는 M2 대식세포를 특이적으로 표적할 수 있음을 확인하였다.The association of TB511 with activated CD18-expressing macrophages and CD8 T cells in normal tissues was shown in Fig. 20, where TB511 decreased activated CD18-expressing macrophages. To investigate the effect of TB511 on macrophages and CD8 T cells in normal tissues, single cells were obtained from spleen, liver, lung, and brain tissues, stained for activated CD18 using Kim127 antibody, and analyzed by flow cytometry. TB511, PADCEV, and combination treatment groups showed no effect on Kim127-positive macrophages and CD8 T cells compared to the PBS group (Figs. 134 to 141). These results confirmed that TB511 could specifically target activated CD18-expressing M2 macrophages in tumor tissues.
13-14. TB511 및 PADCEV의 병용투여에 따른 종양 미세환경 내 면역 세포 조절13-14. Regulation of immune cells in the tumor microenvironment by combined administration of TB511 and PADCEV
TB511 및 PADCEV, 병용투여 후 췌장암의 종양 미세환경에서 면역 세포의 변화를 조사하기 위해, 종양 조직의 유전자 분석을 공간 전사체학(spatial transcriptomics)을 사용하여 수행했다. 도 142 내지 145에 PBS, TB511, PADCEV, 병용 치료 그룹의 세포 분포를 나타내었다. PBS 그룹에서는 상피 세포, 내피 세포, 기질 세포, 섬유아세포, 신경 세포들이 종양 세포 근처에 고르게 분포하고 있는 반면, TB511, PADCEV, 병용 치료 그룹에서는 세포 군집이 각각 4개 또는 3개로 감소한 것으로 나타났다(도 142 내지 145). To investigate the changes in immune cells in the tumor microenvironment of pancreatic cancer after combined administration of TB511 and PADCEV, genetic analysis of tumor tissues was performed using spatial transcriptomics. Figures 142 to 145 show the cell distribution of the PBS, TB511, PADCEV, and combination treatment groups. In the PBS group, epithelial cells, endothelial cells, stromal cells, fibroblasts, and neural cells were evenly distributed near the tumor cells, whereas in the TB511, PADCEV, and combination treatment groups, the cell clusters were reduced to 4 or 3, respectively (Figures 142 to 145).
상기 세포 군집들은 UMAP을 사용하여 분석하였다. 분석결과, PBS, TB511, PADCEV 및 병용투여 그룹에서 면역 세포의 비율은 각각 18%, 23%, 17%, 26%로 나타났다. 즉, TB511가 처리된 그룹들에서 다른 그룹 대비 높은 면역 세포의 비율을 나타내는 것을 확인할 수 있다(도 146 내지 153). 상기 면역 세포들을 분석한 결과, 췌장암 세포인 상피 세포의 비율은 TB511, PADCEV 및 병용투여 그룹에서 감소하였으며, TB511 및 병용투여 그룹에서는 M1 대식세포가 증가하고, M2 대식세포는 감소하는 것으로 나타났으며, CD8 T 세포와 NK 세포는 증가하는 것으로 나타났다. Treg 세포는 어떤 약물 그룹에서도 나타나지 않았다(도 154 및 155). 상기 결과를 통해, TB511 및 PADCEV가 종양 세포의 감소를 유도했으며, TB511은 M2 대식세포를 특이적으로 표적하여 미세환경 내 면역 세포의 변화를 유도하였음을 확인하였다. 다만, PADCEV의 경우, 췌장암 세포를 직접 표적하기 때문에 면역 세포의 유도나 변화를 나타내지 않았다.The above cell populations were analyzed using UMAP. As a result of the analysis, the proportions of immune cells in the PBS, TB511, PADCEV, and combination administration groups were 18%, 23%, 17%, and 26%, respectively. That is, it can be confirmed that the groups treated with TB511 showed a higher proportion of immune cells compared to the other groups (Figs. 146 to 153). As a result of analyzing the above immune cells, the proportion of epithelial cells, which are pancreatic cancer cells, decreased in the TB511, PADCEV, and combination administration groups, and M1 macrophages increased, M2 macrophages decreased, and CD8 T cells and NK cells increased in the TB511 and combination administration groups. Treg cells did not appear in any drug group (Figs. 154 and 155). Through the above results, it was confirmed that TB511 and PADCEV induced a decrease in tumor cells, and TB511 specifically targeted M2 macrophages to induce changes in immune cells within the microenvironment. However, in the case of PADCEV, since it directly targets pancreatic cancer cells, it did not show any induction or changes in immune cells.
13-15. TB511의 종양 미세환경 내 CD8 T 세포 활성화에 대한 역할13-15. Role of TB511 in CD8 T cell activation in the tumor microenvironment
TB511이 M2 대식세포를 표적하여 CD8 T 세포의 활성화를 유도하는지 확인하기 위해, 우리는 CD8 T 세포에서 exhausted marker인 PDCD1, CTLA4, FOXP3, LAG3의 유전자 발현 수준을 분석하고, 활성화 마커인 GZMB와 IFNG의 유전자 발현 수준을 분석했다. exhausted T 세포의 유전자를 발현하는 CD8 T 세포(갈색 군집 세포)는 흰색 화살표로 표시하였고, 활성화된 T 세포 유전자를 발현하는 세포는 빨간색 화살표로 표시하였다. 분석결과, TB511 및 병용 치료 그룹에서 PBS 및 PADCEV 그룹에 비해 활성화된 T 세포의 수가 증가한 것으로 나타났다(도 156 및 도 157). 또한, 각 유전자의 발현 수준을 분석한 결과, PDCD1(즉, PD-1 유전자)에는 어떤 그룹에서도 유의미한 변화가 없었지만, 병용투여 그룹에서 exhausted marker 유전자 CTLA4, FOXP3, LAG3는 유의적으로 감소하는 것으로 나타났다. , Granzyme B와 interferon gamma와 같은 염증성 사이토카인의 발현은 TB511 또는 병용투여 그룹에서 유의적으로 증가하였다. 반면, PADCEV 그룹의 경우, TB511 대비 CD8 T 세포 활성화 유도 효과가 낮게 나타났다(도 158 및 159). 상기 결과를 통해, TB511이 췌장암 종양 조직 내 M2 대식세포를 표적하여 이를 감소시킴으로써 CD8 T 세포의 침윤과 활성화를 유도하는 것을 확인하였으며, PADCEV는 종양 세포의 nectin-4를 직접 표적하여 종양 성장을 감소시키지만, 종양 미세환경 내 면역 세포의 조절에는 기여하지 않는 것을 확인하였다.To confirm whether TB511 targets M2 macrophages and induces CD8 T cell activation, we analyzed the gene expression levels of exhausted markers PDCD1, CTLA4, FOXP3, LAG3 in CD8 T cells, and the gene expression levels of activation markers GZMB and IFNG. CD8 T cells expressing exhausted T cell genes (brown cluster cells) are indicated by white arrows, and cells expressing activated T cell genes are indicated by red arrows. The analysis results showed that the number of activated T cells increased in the TB511 and combination treatment groups compared to the PBS and PADCEV groups (Figs. 156 and 157). In addition, the analysis of the expression levels of each gene showed that there was no significant change in PDCD1 (i.e., PD-1 gene) in any group, but the exhausted marker genes CTLA4, FOXP3, LAG3 were significantly decreased in the combination treatment group. , the expression of inflammatory cytokines such as Granzyme B and interferon gamma significantly increased in the TB511 or combination group. On the other hand, in the PADCEV group, the effect of inducing CD8 T cell activation was lower than that of TB511 (Figs. 158 and 159). Through the above results, it was confirmed that TB511 induces the infiltration and activation of CD8 T cells by targeting and reducing M2 macrophages in pancreatic cancer tumor tissues, and PADCEV directly targets nectin-4 of tumor cells to reduce tumor growth, but does not contribute to the regulation of immune cells in the tumor microenvironment.
상기 실시예들을 종합할때, TB511 및 PADCEV를 병용투여하는 경우 각각을 단독으로 투여하는 경우 대비 종양 크기 및 증식을 감소시키고, M1 대식세포, CD8 T 세포, NK세포의 비율을 증가시키며, M2 대식세포의 비율을 감소시키는 것으로 나타났는바, TB511 및 PADCEV 병용투여에 따른 상승효과가 있음을 확인하였다.In summary of the above examples, when TB511 and PADCEV were administered together, compared to when each was administered alone, tumor size and proliferation were reduced, the proportions of M1 macrophages, CD8 T cells, and NK cells were increased, and the proportion of M2 macrophages was reduced, confirming the synergistic effect of combined administration of TB511 and PADCEV.
상기의 인간화된 마우스 모델로부터 확인한 결과들을 통해, 다양한 고형암에서 TB511 단일요법 및 항암제들과 조합요법의 우수한 효과를 확인하였다. Through the results confirmed from the above humanized mouse model, the excellent efficacy of TB511 monotherapy and combination therapy with anticancer drugs was confirmed in various solid cancers.
Claims (22)
Applications Claiming Priority (2)
| Application Number | Priority Date | Filing Date | Title |
|---|---|---|---|
| KR10-2023-0179601 | 2023-12-12 | ||
| KR20230179601 | 2023-12-12 |
Publications (1)
| Publication Number | Publication Date |
|---|---|
| WO2025127849A1 true WO2025127849A1 (en) | 2025-06-19 |
Family
ID=96058174
Family Applications (1)
| Application Number | Title | Priority Date | Filing Date |
|---|---|---|---|
| PCT/KR2024/096873 Pending WO2025127849A1 (en) | 2023-12-12 | 2024-12-12 | Anticancer composition for co-administration with anticancer agent, comprising novel peptide as active ingredient |
Country Status (2)
| Country | Link |
|---|---|
| KR (1) | KR20250091126A (en) |
| WO (1) | WO2025127849A1 (en) |
Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20190021765A (en) * | 2017-08-23 | 2019-03-06 | 경희대학교 산학협력단 | Composition for removing M2 type tumor-associated macrophage including melittin |
| KR20190110822A (en) * | 2018-03-21 | 2019-10-01 | 백승배 | A Composition for Preventing and Treating Melanoma Comprising Bee Venom Peptide and Anti-Cancer Agent |
| KR20190127609A (en) * | 2018-05-04 | 2019-11-13 | 경희대학교 산학협력단 | Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide |
-
2024
- 2024-12-12 KR KR1020240185127A patent/KR20250091126A/en active Pending
- 2024-12-12 WO PCT/KR2024/096873 patent/WO2025127849A1/en active Pending
Patent Citations (3)
| Publication number | Priority date | Publication date | Assignee | Title |
|---|---|---|---|---|
| KR20190021765A (en) * | 2017-08-23 | 2019-03-06 | 경희대학교 산학협력단 | Composition for removing M2 type tumor-associated macrophage including melittin |
| KR20190110822A (en) * | 2018-03-21 | 2019-10-01 | 백승배 | A Composition for Preventing and Treating Melanoma Comprising Bee Venom Peptide and Anti-Cancer Agent |
| KR20190127609A (en) * | 2018-05-04 | 2019-11-13 | 경희대학교 산학협력단 | Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide |
Non-Patent Citations (2)
| Title |
|---|
| KIM SOYOUNG, CHOI ILSEOB, HAN IK-HWAN, BAE HYUNSU: "Enhanced Therapeutic Effect of Optimized Melittin-dKLA, a Peptide Agent Targeting M2-like Tumor-Associated Macrophages in Triple-Negative Breast Cancer", INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES, vol. 23, no. 15751, 12 December 2022 (2022-12-12), XP093097987, DOI: 10.3390/ijms232415751 * |
| LEE CHANJU, JEONG HYUNJU, BAE YOUNGHYEON, SHIN KYUNGMOON, KANG SINWOO, KIM HWIKYUNG, OH JAYOUNG, BAE HYUNSU: "Targeting of M2-like tumor-associated macrophages with a melittin-based pro-apoptotic peptide", JOURNAL FOR IMMUNOTHERAPY OF CANCER, vol. 7, no. 1, 1 December 2019 (2019-12-01), XP055907984, DOI: 10.1186/s40425-019-0610-4 * |
Also Published As
| Publication number | Publication date |
|---|---|
| KR20250091126A (en) | 2025-06-20 |
Similar Documents
| Publication | Publication Date | Title |
|---|---|---|
| WO2020060122A1 (en) | Fusion protein comprising il-2 protein and cd80 protein, and use thereof | |
| AU2018252193A1 (en) | Immunomodulating polynucleotides, antibody conjugates thereof, and methods of their use | |
| WO2018217058A1 (en) | Anti-human interleukin-2 antibodies and uses thereof | |
| CN105641707B (en) | Conjugates of Cytotoxin Molecules and Cell Binding Receptor Molecules | |
| CN104822656A (en) | Self-stabilizing linker conjugates | |
| WO2015133817A1 (en) | Monoclonal antibody specifically recognizing b-cell lymphoma cells and use thereof | |
| WO2022124866A1 (en) | Anti-pd-1 antibody and uses thereof | |
| WO2019050362A2 (en) | Antibodies to human DLK1 and uses thereof | |
| WO2017023138A1 (en) | Chimeric antigen receptor, and t cells in which chimeric antigen receptor is expressed | |
| WO2024025343A1 (en) | Anti-ror1 antibody and use thereof | |
| WO2018174544A2 (en) | Antibody binding specifically to muc1 and use thereof | |
| WO2014025199A2 (en) | Staphylococcal enterotoxin-derived superantigen mutant, fusion protein in which target-specific polypeptides are connected to the mutant and use thereof | |
| WO2021107689A1 (en) | Pharmaceutical composition for treatment of cancer, comprising immune checkpoint inhibitor and fusion protein including il-2 protein and cd80 protein | |
| WO2022149837A1 (en) | Anti-fgfr3 antibody and use thereof | |
| WO2009107971A2 (en) | Polypeptide specifically coupled to phosphatidylserine and use thereof | |
| WO2021187922A1 (en) | Pharmaceutical composition for cancer treatment comprising fusion protein including il-2 protein and cd80 protein and anticancer drug | |
| WO2021086159A1 (en) | Protein fused with molecule capable of binding to immune checkpoint molecule and use of same | |
| WO2024155098A1 (en) | Regulatory t cell surface antigen and antibody that specifically binds thereto | |
| WO2025127849A1 (en) | Anticancer composition for co-administration with anticancer agent, comprising novel peptide as active ingredient | |
| WO2016003158A2 (en) | New compound for inhibiting binding between dx2 protein and p14/arf protein, and pharmaceutical composition for treating or preventing cancer disease containing same as effective ingredient | |
| WO2020209645A1 (en) | Antibody to programmed cell death protein ligand-1 (pd-l1), and use thereof | |
| WO2021201654A1 (en) | Pharmaceutical composition for preventing or treating mucositis induced by radiotherapy, chemotherapy, or combination thereof, comprising glp-2 derivatives or long-acting conjugate of same | |
| WO2024117781A1 (en) | Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof | |
| WO2023068894A1 (en) | Polymer compound for surface modification to enhance anticancer immune function of natural killer cells | |
| WO2023027561A1 (en) | Bispecific molecule specifically binding to b7-h3 and tgfβ and uses thereof |
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: 24904414 Country of ref document: EP Kind code of ref document: A1 |