WO2024117781A1 - Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof - Google Patents

Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof Download PDF

Info

Publication number
WO2024117781A1
WO2024117781A1 PCT/KR2023/019470 KR2023019470W WO2024117781A1 WO 2024117781 A1 WO2024117781 A1 WO 2024117781A1 KR 2023019470 W KR2023019470 W KR 2023019470W WO 2024117781 A1 WO2024117781 A1 WO 2024117781A1
Authority
WO
WIPO (PCT)
Prior art keywords
cells
seq
fusion protein
domain
cancer
Prior art date
Application number
PCT/KR2023/019470
Other languages
French (fr)
Korean (ko)
Inventor
황도원
김신일
기영욱
박성진
Original Assignee
(주) 테라베스트
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by (주) 테라베스트 filed Critical (주) 테라베스트
Publication of WO2024117781A1 publication Critical patent/WO2024117781A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/705Receptors; Cell surface antigens; Cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N5/00Undifferentiated human, animal or plant cells, e.g. cell lines; Tissues; Cultivation or maintenance thereof; Culture media therefor
    • C12N5/06Animal cells or tissues; Human cells or tissues

Definitions

  • the present invention relates to a fusion protein containing the CD137 extracellular domain or a fragment thereof, immune cells expressing the fusion protein, and uses thereof.
  • T cells expressing CD3 are known as effector cells that can eliminate tumor cells.
  • Immune checkpoints are co-signaling molecules that play a central role in T cell activation and regulate the signal of the T cell receptor (TCR) to be suppressed or elevated (Immunological reviews, 2017, 276:52- 65).
  • NK cells natural killer cells
  • the activity of NK cells is regulated by a balance of various activating and inhibitory receptor signaling. It is known that the anticancer activity of NK cells is also achieved by distinguishing cancer cells through various immune receptors present on their surface.
  • NK cells can eliminate not only cancer cells but also cancer stem cells, so they are in the spotlight as a source of development for treatments that can not only suppress the occurrence, proliferation, and metastasis of cancer, but also reduce the recurrence of cancer after cure.
  • 4-1BB is one of the TNF-receptor superfamily (TNFRSF), a costimulatory molecule that is expressed upon activation of immune cells, both innate and adaptive immune cells, and regulates the activation of various immune cells.
  • TNFRSF TNF-receptor superfamily
  • the 4-1BB agonist enhances immune cell proliferation, survival, cytokine secretion, and CD8+ T cell lytic activity. This action has shown anticancer effects in several mouse tumor models and is attracting attention as a target for anticancer drugs. However, to this day, new methods that can enhance the anticancer activity of immune cells against cancer cells through 4-1BB activation are being studied.
  • the present inventors studied a new method that can enhance the anti-cancer activity of immune cells against cancer cells, and as a result, a fusion protein containing the extracellular domain and intracellular signaling domain of CD137 (or 4-1BB) was introduced into immune cells.
  • a fusion protein containing the extracellular domain and intracellular signaling domain of CD137 (or 4-1BB) was introduced into immune cells.
  • cytotoxicity against cancer cells increased compared to normal immune cells.
  • INF- ⁇ interferon gamma
  • one aspect of the present invention provides a fusion protein comprising the following (i) to (iii): (i) the extracellular domain of CD137 (4-1BB) or a fragment thereof; (ii) Transmembrane domain; and (iii) an intracellular signaling domain.
  • Another aspect of the present invention provides a polynucleotide encoding the fusion protein, a vector and a virus containing the polynucleotide.
  • Another aspect of the present invention provides immune cells expressing the fusion protein.
  • Another aspect of the present invention includes the steps of i) preparing the virus; and ii) processing the virus into immune cells.
  • Another aspect of the present invention is a pharmaceutical composition for preventing or treating cancer comprising the immune cells as an active ingredient; and pharmaceutical compositions and kits for cancer treatment comprising the immune cells and bispecific antibodies.
  • Another aspect of the invention provides CD137 extracellular domain variants or fragments thereof.
  • cBBR T cells overexpressing the 4-1BB/CD3 ⁇ fusion protein (cBBR) or a variant thereof specifically killed cancer cells overexpressing 4-1BBL In the case of T cells overexpressing cBBR containing the 4-1BB extracellular domain variant, it was confirmed that the binding affinity and cytotoxicity to 4-1BBL were reduced compared to cBBR T cells containing the native 4-1BB extracellular domain. did.
  • cBBR T cells overexpressing cBBR containing the above mutant were stimulated with Urelumab, their activity significantly increased compared to normal T cells.
  • anti-BCMA/anti-4-1BB bispecific antibodies it showed a significantly superior killing effect on BCMA-expressing cancer cells compared to normal T cells. Therefore, T cells overexpressing cBBR can be used as an anticancer agent.
  • Figure 1a is a schematic diagram of a nucleic acid cassette encoding a fusion protein combining 4-1BB and CD3 ⁇ intracellular domains.
  • SP represents the signal peptide
  • CRD represents the cysteine-rich domain
  • TM represents the transmembrane domain
  • ICD represents the intracellular domain of 4-1BB.
  • CD3 ⁇ represents the intracellular domain of CD3 ⁇ .
  • Figure 1b is a schematic diagram of a nucleic acid cassette encoding a fusion protein combining 4-1BB and CD3 ⁇ intracellular domains. Specifically, the domain structure of a fusion protein expressing only CRD1 and CDR1/CRD2 among CRD1, CRD2, CRD3, and CRD4 of the CD137 extracellular domain is schematized.
  • Figure 2 is a diagram showing that a fusion protein combining 4-1BB and CD3 ⁇ intracellular domains is stably expressed in transformed NK92MI cells.
  • Figure 3 shows the level of expression of 4-1BB after treating 1.0 x 10 6 NK92MI cells with Urelumab at different concentrations.
  • Figure 4 shows the expression level of 4-1BB quantified through FACS analysis after treatment with urelumab at different concentrations.
  • Figure 5 shows the measurement of the expression level of INF- ⁇ secreted by transformed NK92MI cells and normal NK cells cultured in the presence of urelumab. At this time, the group in which NK92MI cells and normal NK cells were cultured together with K562 cells was used as the control group.
  • Figure 6 shows a schedule for an experiment in which PBMC-derived NK cells were cultured, transfected with CD137 mRNA, and then co-cultured with Urelumab.
  • Figure 7 shows the expression level of 4-1BB over time using flow cytometry.
  • Figure 8 shows the amount of IFN- ⁇ secreted through ELISA.
  • Figure 9 shows that, despite the presence of only CRD1 and CRD1/CRD2 of the CD137 extracellular domain, it was confirmed through flow cytometry that Urelumab and the 4B4-1 antibody bind well.
  • FIG 10 is a diagram showing a schematic diagram of T (CER T, cBBR T) cells overexpressing chimeric engager receptor (cBBR) as an embodiment of the present invention.
  • Figure 11 is a graph showing the results of confirmation of cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R) overexpressed in Jurkat T cells and 4-1BBL overexpressed in Nalm6 cells through flow cytometry.
  • cBBR Chimeric 4-1BB receptor
  • variants I64R, V71R
  • Figure 12 is a graph showing the results of confirming the expression of cBBR or its variants (I64R, V71R) overexpressed in blood-derived T cells through flow cytometry.
  • Figure 13 is a graph showing the results of confirming the binding rate of cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R) overexpressed in Jurkat T cells and 4-1BBL overexpressed in Nalm6 cells through flow cytometry.
  • cBBR Chimeric 4-1BB receptor
  • variants I64R, V71R
  • Figure 14 shows the killing ability of T cells (cBBR T cells) overexpressing cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R, I64R/V71R) against cancer cells overexpressing 4-1BBL by CFSE fluorescence staining and This is a graph showing the results measured by flow cytometry.
  • Figure 15 is a graph showing the level of activation of cBBR T cells overexpressing cBBR variants (I64R, V71R, I64R/V71R) by urelumab, confirmed by flow cytometry after staining with anti-CD8a antibody and anti-CD107a antibody. am.
  • Figure 16 is a graph showing the results of confirming the degree of activation of cBBR T cells overexpressing cBBR variants (I64R, V71R, I64R/V71R) by urelumab by measuring the concentration of IFN- ⁇ .
  • Figure 17 shows cBBR T cells or Ramos cells overexpressing cBBR variants (I64R, V71R) treated with a bispecific antibody (anti-BCMA/anti-4-1BB), and then cBBR T cells or Ramos cells and the bispecific antibody.
  • BiTE anti-BCMA/anti-4-1BB bispecific antibody
  • 19F2 anti-BCMA antibody
  • 4B4-1 anti-4-1BB antibody
  • Figure 18 is a graph showing the results of confirming the cancer cell killing ability of cBBR T cells by co-culturing cBBR T cells overexpressing cBBR variants (I64R, V71R) with cancer cells expressing dual specific antibodies and BCMA.
  • Figure 19 confirms that tumor suppression occurred when transformed T cells and AB-101 antibody were administered.
  • One aspect of the present invention provides a fusion protein comprising (i) to (iii) below:
  • CD137 is a membrane protein belonging to the tumor necrosis factor receptor family.
  • CD137 is referred to as tumor necrosis factor receptor superfamily member 9 or 4-1BB.
  • CD137 is a tumor necrosis factor receptor and mainly acts as a co-stimulatory molecule in T cells.
  • the CD137 extracellular domain contains four cysteine-rich pseudo repeat (CRD) domains.
  • the CD137 protein may preferably be human CD137 protein, but is not limited thereto.
  • CD137 extracellular domain fragment refers to a fragment in which part of the N-terminus and/or C-terminus of the native CD137 extracellular domain protein is truncated, and is identical to or similar to the native CD137 extracellular domain 4- This may indicate 1BBL binding force.
  • wild type includes all proteins found in nature or nucleic acids encoding them and can be used interchangeably with wild type.
  • the CD137 extracellular domain or fragment thereof includes CRD1 comprising the amino acid sequence of SEQ ID NO: 2; CRD2 comprising the amino acid sequence of SEQ ID NO: 3; CRD3 comprising the amino acid sequence of SEQ ID NO: 6; CRD4 comprising the amino acid sequence of SEQ ID NO: 7; And it may include any one selected from the group consisting of combinations thereof.
  • the CD137 extracellular domain or fragment thereof may include CRD1. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD2. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2 and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2 and CRD4.
  • the CD137 extracellular domain or fragment thereof may include CRD3 and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD3, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2, CRD3, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, CRD3, and CRD4.
  • the CD137 extracellular domain or fragment thereof may include or consist of the amino acid sequence of CRD1.
  • the CD137 extracellular domain or fragment thereof may include or consist of the amino acid sequences of CRD1 and CRD2.
  • the extracellular domain of CD137 or a fragment thereof may include or consist of the amino acid sequences of CRD1, CRD2, CRD3, and CRD4.
  • the CD137 extracellular domain or fragment thereof may include the amino acid sequence of SEQ ID NO: 15.
  • the protein consists of a sequence in which one or several amino acids are added, deleted, or substituted. It can be.
  • the CD137 extracellular domain has about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100% amino acid identity with the amino acid sequence of SEQ ID NO: 15. It may contain or consist of sequences.
  • the CD137 extracellular domain or fragment thereof may further include mutations.
  • mutant refers to a form in which some amino acids of the above-described CD137 extracellular domain or fragment thereof are substituted. That is, the variant of the CD137 extracellular domain or fragment thereof may have a different sequence from the native CD137 extracellular domain or fragment thereof. In the present invention, the mutant may have reduced binding ability to 4-1BBL (CD137L) compared to native CD137.
  • 4-1BBL CD137L
  • the binding ability with 4-1BBL can be measured using methods known to those skilled in the art.
  • the mutation may be included in any one selected from the group consisting of CRD1, CRD2, CRD3, CRD4, and combinations thereof.
  • the mutation may be included in CRD1. In one embodiment, the mutation may be included in CRD2. In one embodiment, the mutation may be included in CRD3. In one embodiment, the mutation may be included in CRD4. In one embodiment, the mutation may be included in CRD1 and CRD2. In one embodiment, the mutation may be included in CRD1 and CRD3. In one embodiment, the mutation may be included in CRD1 and CRD4. In one embodiment, the mutation may be included in CRD2 and CRD3. In one embodiment, the mutation may be included in CRD2 and CRD4. In one embodiment, the mutation may be included in CRD3 and CRD4. In one embodiment, the mutation may be included in CRD1, CRD2, and CRD3.
  • the mutation may be included in CRD1, CRD2, and CRD4. In one embodiment, the mutation may be included in CRD1, CRD3, and CRD4. In one embodiment, the mutation may be included in CRD2, CRD3, and CRD4. In one embodiment, the mutation may be included in CRD1, CRD2, CRD3, and CRD4.
  • the mutation may be included in CRD2 and/or CRD3.
  • the mutation of CRD2 is any one amino acid selected from the group consisting of P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26, and combinations thereof in the amino acid sequence of SEQ ID NO: 3. It may have been replaced. Additionally, the mutation of CRD3 may be a substitution of any one amino acid selected from the group consisting of S14, M15, C16, and combinations thereof in the amino acid sequence of SEQ ID NO: 6.
  • amino acids introduced by the substitution include glycine, alanine, valine, leucine, isoleucine, serine, threonine, cysteine, Methionine, aspartic acid, glutamic acid, asparagine, glutamine, lysine, arginine, phenylalanine, tyrosine, tryptophan ( It may be any one selected from the group consisting of tryptophan, histidine, and proline.
  • the mutation of CRD2 may be a substitution of any one amino acid selected from the group consisting of I18R, V25R, and combinations thereof in the amino acid sequence of SEQ ID NO: 3.
  • the mutation in CRD2 may be one in which isoleucine (I), the 18th amino acid of SEQ ID NO: 3, is substituted with arginine (R) (I18R).
  • a mutation in CRD2 may be a substitution of valine (V), the 25th amino acid of SEQ ID NO: 3, with arginine (R) (V25R).
  • the mutation in CRD2 may be one in which isoleucine (I), the 18th amino acid of SEQ ID NO: 3, and valine (V), the 25th amino acid, are each substituted with arginine (R) (I18R, V25R).
  • CRD2 containing the mutation may include or be composed of any one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 26.
  • the CD137 extracellular domain containing the above mutation may include any one amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 27.
  • the binding ability of the CD137 extracellular domain to 4-1BBL may be reduced compared to the native type.
  • transmembrane domain used in the present invention refers to a portion of the protein structure located in the cell membrane that connects the extracellular domain and the intracellular signaling domain and penetrates the cell membrane.
  • the fusion protein according to the present invention can be fixed to the cell membrane through the transmembrane domain.
  • the transmembrane domain includes T cell receptor (TCR), CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD4, CD5, CD8 ⁇ , CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, It may be derived from any one molecule selected from the group consisting of CD137, CD152, and CD154. Specifically, the transmembrane domain may be derived from CD137.
  • the transmembrane domain may be derived from CD137. Specifically, the transmembrane domain may include the amino acid sequence of SEQ ID NO: 8.
  • the protein may be composed of a sequence in which one or several amino acids are added, deleted, or substituted. there is.
  • the transmembrane domain derived from CD137 may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 8.
  • the (i) CD137 extracellular domain and (ii) transmembrane domain may be connected through a spacer.
  • the spacer refers to a linker and may be a protein or peptide. Additionally, it may be composed of about 1 to about 1,000 amino acids, and may include about 10 to about 300 amino acids, about 15 to about 100 amino acids, about 15 to about 60 amino acids, or about 15 to about 45 amino acids. Additionally, the linker may be a fragment of a protein in the human body, such as an Fc region.
  • the linker consists of CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD4, CD5, CD8 ⁇ , CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD152 and CD154. It may be derived from any one molecule selected from the group.
  • the spacer may be derived from CD137. Specifically, the spacer may include or be composed of the amino acid sequence of SEQ ID NO: 11.
  • intracellular signaling domain refers to the case where an extracellular binding domain present on the cell surface binds to a biological target molecule (e.g., binding of CD137 extracellular domain and 4-1BBL). ), refers to a site that transmits signals into the cell to induce reactions such as cell activation, release of cytotoxic factors, cytokine production, and cell proliferation.
  • the intracellular signaling domain may include a co-stimulatory signaling domain and/or a primary signaling domain.
  • co-stimulatory domain refers to the intracellular signaling domain of a co-stimulatory molecule.
  • the “co-stimulatory molecule” is a cell surface molecule other than the Fc receptor that provides a secondary signal that induces efficient activation and function of immune cells when the extracellular binding site binds to the target molecule.
  • the costimulatory domain may be derived from any one molecule selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and CD137. .
  • the costimulatory domain may be derived from CD137.
  • the co-stimulatory domain may include or consist of the amino acid sequence of SEQ ID NO: 9.
  • primary signaling domain refers to a protein that constitutes the CD3 complex of T cells and refers to a signaling domain that regulates the primary activation of T cells in a stimulatory or inhibitory manner.
  • the primary signaling domain that stimulates activation of the T cell may contain a signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM (Immunoreceptor tyrosine-based activation motif).
  • ITAM containing the primary signaling domain may be derived from any one molecule selected from the group consisting of TCR ⁇ , FcR ⁇ , FcR ⁇ , CD3 ⁇ , CD3 ⁇ , CD3 ⁇ , CD22, CD79a, CD79b and CD66d.
  • the primary signaling domain may be derived from CD3 ⁇ .
  • CD3 ⁇ used herein refers to a protein that constitutes the CD3 complex.
  • the CD3 complex is a protein complex and T cell co-receptor involved in activating both cytotoxic T cells and T helper cells.
  • the CD3 complex contains the CD3 ⁇ chain, CD3 ⁇ chain, CD3 ⁇ chain, CD3 ⁇ chain, CD3 ⁇ chain, and CD3 ⁇ .
  • the primary signaling domain may include or consist of the amino acid sequence of SEQ ID NO: 10.
  • CD28 is one of the proteins expressed on T cells that provide costimulatory signals necessary for T cell activation and survival. At this time, the intracellular domain of CD28 may have the amino acid sequence of SEQ ID NO: 34.
  • OX40 refers to tumor necrosis factor receptor superfamily, member 4, also known as CD134. At this time, the intracellular domain of OX40 may have the amino acid sequence of SEQ ID NO: 35.
  • each of the domains consists of a sequence in which one or several amino acids are added, deleted, or substituted. It can be.
  • the co-stimulatory domain may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 9.
  • the primary signaling domain may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 10. there is.
  • the intracellular signaling domain of the present invention may be an appropriate combination of the costimulatory domain and the primary signaling domain.
  • the intracellular signaling domain may include CD137 and CD3 ⁇ .
  • the co-stimulatory domain and the primary signaling domain may be connected through a linker.
  • the fusion protein of the present invention may contain the following structural formula (I).
  • N' is the N terminus of each polypeptide
  • the C' is the C terminus of each polypeptide
  • the TM is a transmembrane domain
  • B and C are intracellular signaling domains, each independently a co-stimulatory domain and a primary signaling domain;
  • L1, L2 and L3 are each peptide linkers
  • n, m and l are each independently 0 or 1.
  • extracellular domain or fragment thereof, transmembrane domain, and intracellular signaling domain of CD137 are the same as described above.
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-CD3z (SEQ ID NO: 10)-C'
  • N'-CRD1 SEQ ID NO: 2
  • CRD4 SEQ ID NO: 7
  • Linker SEQ ID NO: 11
  • TM SEQ ID NO: 8
  • CD137 intracellular domain SEQ ID NO: 18
  • CD3z SEQ ID NO: 10
  • N'-CRD4 SEQ ID NO: 7
  • Linker SEQ ID NO: 11
  • T SEQ ID NO: 8
  • CD137 intracellular domain SEQ ID NO: 18
  • CD3z SEQ ID NO: 10
  • N'-CRD1 SEQ ID NO: 2
  • Linker SEQ ID NO: 11
  • T SEQ ID NO: 8
  • CD137 intracellular domain SEQ ID NO: 18
  • CD28 SEQ ID NO: 34
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34) - C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34)-C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-CD28 (SEQ ID NO: 34)-C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34) - C'
  • N'-CRD4 (SEQ ID NO: 7)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-CD28 (SEQ ID NO: 34)-C'
  • N'-CRD1 (SEQ ID NO: 2)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35) - C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35)-C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
  • N'-CRD1 (SEQ ID NO: 2) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35) - C'
  • N'-CRD4 (SEQ ID NO: 7)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
  • N' refers to the N terminus of the fusion protein
  • C' refers to the C terminus of the fusion protein
  • Another aspect of the present invention provides a polynucleotide encoding the fusion protein.
  • the fusion protein is the same as described above.
  • the polynucleotide encoding the fusion protein may include or be composed of any one nucleotide selected from the group consisting of SEQ ID NO: 21 to SEQ ID NO: 23 and SEQ ID NO: 28.
  • one or more bases may be mutated by substitution, deletion, insertion, or a combination thereof.
  • a synthesis method well known in the art for example, a method described in the literature (Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988), can be used. and triester, phosphite, phosphoramidite and H-phosphate methods, PCR and other autoprimer methods, and oligonucleotide synthesis methods on solid supports.
  • the polynucleotide is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, and at least about the base sequences of SEQ ID NO: 21 to SEQ ID NO: 23 and SEQ ID NO: 28, respectively. 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about It may include a nucleic acid sequence having an identity of 97%, at least about 98%, at least about 99%, or at least about 100%.
  • the polynucleotide may additionally include a nucleic acid encoding a signal sequence or leader sequence.
  • signal sequence used herein refers to a signal peptide that directs secretion of a target protein.
  • the signal peptide is cleaved after translation in the host cell.
  • the signal sequence is an amino acid sequence that initiates the movement of proteins through the ER (Endoplasmic reticulum) membrane.
  • the signal sequence has well-known characteristics in the art, and usually contains 16 to 30 amino acid residues, but may contain more or fewer amino acid residues.
  • a typical signal peptide consists of three regions: a basic N-terminal region, a central hydrophobic region, and a more polar C-terminal region.
  • the central hydrophobic region contains 4 to 12 hydrophobic residues that anchor the signal sequence throughout the membrane lipid bilayer while the immature polypeptide moves.
  • the signal sequence is cleaved within the lumen of the ER by a cellular enzyme commonly known as signal peptidase.
  • the signal sequence may be a secretion signal sequence of tPa (Tissue Plasminogen Activation), HSV gDs (Signal sequence of Herpes simplex virus glycoprotein D), or growth hormone.
  • tPa tissue Plasminogen Activation
  • HSV gDs Synignal sequence of Herpes simplex virus glycoprotein D
  • growth hormone a secretion signal sequence used in higher eukaryotic cells, including mammals.
  • the signal sequence can be used as a wild-type signal sequence, or by replacing it with a codon that is frequently expressed in host cells.
  • the signal sequence may be the signal sequence of CD137.
  • the signal sequence may include or consist of the amino acid sequence of SEQ ID NO: 1.
  • the vector may be plasmid DNA, phage DNA, etc., commercially developed plasmids (e.g., pUC18, pBAD, pIDTSAMRT-AMP, etc.), E. coli-derived plasmids (e.g., pYG601BR322, pBR325, pUC118, pUC119, etc.) , Bacillus subtilis-derived plasmids (e.g., pUB110, pTP5, etc.), yeast-derived plasmids (e.g., YEp13, YEp24, YCp50, etc.), phage DNA (Charon4A, Charon21A, EMBL3, EMBL4, ⁇ gt10, ⁇ gt11, ⁇ ZAP, etc.), It may be an animal virus vector (e.g., retrovirus, adenovirus, vaccinia virus, etc.) or an insect virus vector (baculovirus, etc.
  • N' refers to the N terminus of the fusion protein
  • C' refers to the C terminus of the fusion protein.
  • CRD1, CRD2, CRD3, and CRD4 may include mutations. At this time, the mutation is as described above.
  • N' refers to the N terminus of the fusion protein
  • C' refers to the C terminus of the fusion protein
  • intracellular domain of CD137 the intracellular domain of CD3 ⁇ , the intracellular domain of CD28, and the intracellular domain of OX40 are the same as described above.
  • the immune cells may be natural killer cells, T cells, or macrophages.
  • the immune cell expressing the fusion protein is a cell transformed by introducing the polynucleotide, and can be transformed using a vector loaded with the polynucleotide or a virus containing the polynucleotide.
  • the polynucleotide, vector and virus are the same as described above.
  • NK cell naturally killer cell
  • cytotoxic lymphocyte that constitutes a major component of the innate immune system
  • LGL large granular lymphocyte
  • CLP lymphoid progenitor
  • T cell refers to a type of lymphocyte that hosts antigen-specific adaptive immunity, including immature T cells that have not come into contact with an antigen and effector T cells that have matured after contact with an antigen (helper T cells, cytotoxic T cells). cells, natural killer T cells) and memory T cells.
  • T cells Activation of T cells occurs when a na ⁇ ve T cell antigen receptor (TCR) binds to the MHC:antigen complex and activates the CD3 complexes of T cells: CD3 gamma ( ⁇ ), delta ( ⁇ ), epsilon ( ⁇ ), and zeta ( ⁇ ) is induced by phosphorylating the ITAM of the chain.
  • the ITAM phosphorylation induces phosphorylation of ZAP-70 protein, LAT protein, and SLP-76 protein, thereby inducing a signal response that activates transcription factors such as NF ⁇ B, AP-1, and NFAT.
  • Transcription factors such as NF ⁇ B, AP-1, and NFAT induce T cell division and differentiation by promoting the expression of interleukin-2 (IL-2).
  • IL-2 interleukin-2
  • macrophage is also referred to as a phagocytic cell and is a major cell responsible for innate immunity.
  • the macrophages may exist as settlers in tissues, such as Kupffer cells, Langerhans cells, and microglial cells, or may exist in the form of monocytes in the blood. At this time, the monocytes may differentiate into dendritic cells or macrophages. Macrophages control inflammatory responses through phagocytosis and secretion of cytokines, which absorb and digest foreign proteins such as cell tissue, foreign substances, microorganisms, and cancer cells. In addition, after removing the antigen, it acts as an antigen-presenting cell that delivers the antigen to lymphocytes, thereby inducing an immune response.
  • the immune cells can be obtained from an individual or from various tissues. Additionally, the immune cells include unmodified immune cells obtained from an individual and may include immature immune cells as well as mature immune cells.
  • the immune cells can be obtained by separating them from tissues, hematopoietic cells, hematopoietic stem cells, or hematopoietic progenitor cells.
  • the immune cells may include, but are not limited to, hematopoietic cells, hematopoietic stems or precursors from placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, etc.
  • the immune cells may be autologous or foreign cells.
  • the immune cells may be derived from mammals such as rats, mice, livestock, and humans.
  • the immune cells may be obtained from an individual seeking treatment.
  • the immune cells can be directly isolated from the blood of the individual and used, or they can be used by differentiating immature undifferentiated (or immature) immune cells or stem cells obtained from the individual.
  • the immune cells may be differentiated from iPSCs (Induced pluripotent stem cells).
  • Methods for introducing the vector or virus into immune cells can be methods known in the art, such as transient transfection, microinjection, transduction, cell fusion, and calcium phosphate precipitation.
  • Method, Liposome-mediated transfection, DEAE Dextran-mediated transfection, Polybrene-mediated transfection, electroporation can be introduced into cells by a gene gun or other known methods for introducing nucleic acids into cells (Wu et al., J. Bio. Chem., 267:963-967, 1992; Wu and Wu, J. Bio. Chem., 263:14621-14624, 1988). However, it is not limited to this.
  • Transduced or transfected immune cells can be propagated ex vivo following vector or viral infection.
  • the transfected immune cells are allowed to proliferate for at least about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, It can be cultured for 13, 14, 15, 16, 17, 18, 19 or 20 days, preferably for 13 to 15 days.
  • Methods for confirming whether the vector has been successfully introduced into the immune cells include, for example, molecular biological assays (e.g., Southern and Northern blotting, RT-PCR and PCR) well known to those skilled in the art; It may include detection of the presence or absence of a particular peptide by biochemical assays (e.g., immunological methods (e.g., ELISAs, Western blots, flow cytometry)).
  • biochemical assays e.g., immunological methods (e.g., ELISAs, Western blots, flow cytometry)).
  • Another aspect of the present invention includes the steps of i) preparing a virus containing the polynucleotide; and ii) processing the virus into immune cells. It provides a method for producing immune cells expressing the fusion protein.
  • the polynucleotide, virus containing the polynucleotide, immune cell, and fusion protein are the same as described above.
  • Methods for producing the virus may use methods known in the art. Additionally, immune cells transfected with the virus can be proliferated by further culturing after infection.
  • culture used in the present invention refers to growing cells in appropriately controlled environmental conditions, and the culture process of the present invention can be carried out according to appropriate media and culture conditions known in the art. This culture process can be easily adjusted by a person skilled in the art depending on the cells selected.
  • the medium refers to a known medium used for culturing cells, and includes all known media for cell culture or modified media thereof.
  • immune cells transfected with the above virus persist for approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12 days after infection.
  • the production of immune cells expressing the fusion protein can be confirmed by confirming the expression of the fusion protein through methods such as molecular biological assays and biochemical assays well known to those skilled in the art.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer, comprising immune cells expressing the fusion protein as an active ingredient.
  • the pharmaceutical composition may further include an anti-CD137 antibody or a bispecific antibody.
  • the fusion protein and immune cells are the same as described above.
  • the above cancers include stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, leukemia, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, and salivary gland cancer.
  • lymphoma, kidney cancer, melanoma multiple myeloma, brain cancer, osteosarcoma, glioblastoma, IgG-opsonized tumor, lymphoma, neuroma, mesothelioma, and esophageal cancer.
  • the term “antibody” refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen.
  • the heavy and light chains of immunoglobulins may each include a constant region and a variable region.
  • the light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs).
  • CDR complementarity determining regions
  • FRs framework regions
  • the antibody may refer to any one selected from IgG, IgE, IgM, IgD, and IgA, and may be a subclass of IgG, such as IgG1, IgG2, IgG3, IgG4, IgA1, or IgA2. Additionally, the antibody may be an agonistic antibody or an antagonistic antibody.
  • the antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, and Fab fragments. , F(ab') fragment, Fv(sdFv) linked by a disulfide bond, and any of the above epitope binding fragments, and is not limited thereto, as long as it specifically binds to the cancer antigen.
  • the term “dual specific antibody” refers to a substance that binds to at least one target or antigen.
  • the bispecific antibody includes a first domain that specifically binds to a tumor antigen; and a second domain that specifically binds to the extracellular domain of CD137.
  • the bispecific antibody may be a fragment of an anti-CD137 antibody, for example, scFv, bound to an antibody that specifically binds to an antigen.
  • the scFv may be bound to the C terminus of an antibody that specifically binds to an antigen, and at this time, the antibody and scFv may be bound through a peptide linker.
  • the peptide linker may be (G4S)n.
  • the linker of the peptide may be (G4S) 4 .
  • the bispecific antibody may be an engager.
  • the bispecific antibody may be in the form of BiTE (Bi-specific T-cell engager).
  • the first domain may be an antibody or scFv that specifically binds to a cancer antigen
  • the second domain may be an scFv that specifically binds to CD137.
  • the first domain and the second domain may be linked through a peptide linker.
  • the cancer antigen is alpha folate receptor, 5T4, ⁇ v ⁇ 6 integrin, BCMA, B7-H3, B7-H6, CAIX, CD16, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a.
  • EGFR EGFR family including ErbB2 (HER2), EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, fetal AchR, FR ⁇ , GD2, GD3, glypican -3(GPC3), HLA-A1+MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ESO-1, HLA-A2+NY-ESO-1, HLA-A3+NY- ESO-1, IL-11R ⁇ , IL-13R ⁇ 2, Lambda, Lewis-Y, Kappa, Mesothelin, Muc1, Muc16, NCAM, NKG2D Ligand, NY-ESO-1, PRAME, PSCA, PSMA, ROR1, SSX, Survi It may be any one selected from the group consisting of bin, TAG72, TEMs, VEGFR2, and W
  • the extracellular domain binding site of CD137 which is the second domain of the bispecific antibody, can serve as an agonistic antibody. Therefore, the bispecific antibody can activate CD137 by specifically binding to the extracellular domain of CD137.
  • the bispecific antibody specifically binds to the cancer antigen overexpressed on the tumor surface and specifically binds to the CD137 extracellular domain of the fusion protein of the present invention, thereby inhibiting tumor cells and the fusion protein of the present invention. It can be used as a cell engager to bind the expressed immune cells.
  • the bispecific antibody By using the bispecific antibody as a cell binder, it can target cancer and exhibit safer and superior anticancer activity by activating immune cells specifically for cancer cells.
  • T cells expressing the fusion protein of the present invention are treated in combination with anti-BCMA/anti-4-1BB bispecific antibodies, it is confirmed that cytotoxicity increases compared to wild type T cells. (Figure 17).
  • prevention refers to all actions that suppress or delay the onset of a disease by administering the pharmaceutical composition.
  • treatment refers to any action that improves or beneficially changes the symptoms of a disease by administering the pharmaceutical composition.
  • immune cells expressing the fusion protein may be included in any amount (effective amount) depending on the use, formulation, purpose of formulation, etc., as long as they can show activity.
  • the pharmaceutical composition of the present invention can be used to kill about 1 ⁇ 10 2 cells to about 1 ⁇ 10 16 cells, about 1 ⁇ 10 2 cells to about 1 ⁇ 10 15 cells, about 1 ⁇ 10 2 cells to about 1 ⁇ 10 14 cells, or about 1 cell. It may include immune cells expressing the fusion protein of ⁇ 10 2 cells to about 1 ⁇ 10 13 cells.
  • the pharmaceutical composition may include about 2 ⁇ 10 2 to about 2 ⁇ 10 11 cells of immune cells expressing the fusion protein.
  • effective amount refers to the amount of active ingredient that can induce an anti-aging effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
  • Compositions of the present invention can be used unfrozen or frozen for later use. If freezing is required, use standard cryopreservation agents (e.g. DMSO, glycerol, polyvinylpyrrolidone, polyethylene glycol, albumin, dextran, sucrose, ethylene glycol, id erythritol, D-ribitol, D-mannitol, D- Sorbitol, i-inositol, D-lactose, choline chloride, and Epilife ® cell freezing medium (Cascade biologics) can be added to the cells before freezing.
  • cryopreservation agents e.g. DMSO, glycerol, polyvinylpyrrolidone, polyethylene glycol, albumin, dextran, sucrose, ethylene glycol, id erythritol, D-ribitol, D-mannitol, D- Sorbitol, i-inositol, D-
  • composition of the present invention may be formulated to further include a pharmaceutically acceptable carrier.
  • the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not significantly irritate living organisms and does not inhibit the biological activity and properties of the administered ingredient.
  • the pharmaceutical composition of the present invention can be applied as a cell therapy agent, and pharmaceutically acceptable carriers that can be included as a cell therapy agent include buffers, preservatives, analgesics, solubilizers, isotonic agents, stabilizers, bases, excipients, lubricants, etc. If it is known in the industry, it can be used without restrictions.
  • the pharmaceutical composition of the present invention can be prepared according to commonly used techniques in the form of various dosage forms.
  • the pharmaceutical composition of the present invention can be administered in a pharmaceutically effective amount, and can be administered through any route as long as it can induce movement to the diseased area.
  • administration means introducing a predetermined substance into an individual by an appropriate method.
  • loading the pharmaceutical composition of the present invention into a vehicle equipped with a means to direct it to the lesion may be considered.
  • it may be parenteral administration.
  • parenteral administration may include subcutaneous, intradermal, intramuscular, instillation, intravenous, intraarterial, intraarticular, and intracerebrospinal fluid administration.
  • the preferred administration method and formulation of the pharmaceutical composition of the present invention is injection.
  • Injections include aqueous solvents such as physiological saline solution, Ringer's solution, Hank's solution, or sterilized aqueous solution, vegetable oils such as olive oil, higher fatty acid esters such as ethyl oleic acid, and non-aqueous solvents such as ethanol, benzyl alcohol, propylene glycol, polyethylene glycol, or glycerin.
  • aqueous solvents such as physiological saline solution, Ringer's solution, Hank's solution, or sterilized aqueous solution
  • vegetable oils such as olive oil
  • higher fatty acid esters such as ethyl oleic acid
  • non-aqueous solvents such as ethanol, benzyl alcohol, propylene glycol, polyethylene glycol, or glycerin.
  • impermeable agents known in the art suitable for the barrier to pass through can be used, and as a stabilizer to prevent deterioration, ascorbic acid, sodium bisulfite, BHA, tocopherol, EDTA It may additionally contain pharmaceutical carriers such as emulsifiers, buffers for pH adjustment, and preservatives to inhibit microbial growth such as phenylmercuric nitrate, thimerosal, benzalkonium chloride, phenol, cresol, and benzyl alcohol.
  • pharmaceutical carriers such as emulsifiers, buffers for pH adjustment, and preservatives to inhibit microbial growth such as phenylmercuric nitrate, thimerosal, benzalkonium chloride, phenol, cresol, and benzyl alcohol.
  • the term “pharmaceutically effective amount” refers to an amount that is sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment and does not cause side effects, and the effective dose level is the patient's Factors including gender, age, weight, health status, type of disease, severity, activity of drug, sensitivity to drug, method of administration, time of administration, route of administration and excretion rate, treatment period, drugs combined or used simultaneously, and other factors. It can be readily determined by a person skilled in the art according to factors well known in the medical field.
  • the preferred dosage of the pharmaceutical composition of the present invention is about 1 ⁇ 10 cells/kg to about 1 ⁇ 10 12 cells/kg per day, based on cells, depending on the patient's condition, weight, gender, age, patient's severity, and route of administration. It may be about 1 ⁇ 10 cells/kg to about 1 ⁇ 10 11 cells/kg or about 1 ⁇ 10 cells/kg to about 1 ⁇ 10 10 cells/kg.
  • the dosage may be prescribed in various ways depending on factors such as formulation method, administration method, patient's age, weight, gender, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity, and those skilled in the art will Taking these factors into consideration, the dosage can be adjusted appropriately.
  • the number of administrations can be once or twice or more within the range of clinically acceptable side effects.
  • the pharmaceutical composition of the present invention may be administered 1 to 3 times/day, 1 to 3 times/week, or 1 to 10 times/month.
  • the pharmaceutical composition of the present invention can be used 1 to 9 times/month, 1 to 8 times/month, 1 to 7 times/month, 1 to 6 times/month, 1 to 5 times/month, 1 It may be administered once to four times/month, once to three times/month, or once to twice per month.
  • the administration site it can be administered at one or two or more locations.
  • the dosage per kg or per individual is the same as that for humans, or, for example, the above-mentioned administration is based on the volume ratio (e.g., average value) of the organs (e.g., heart, etc.) between the target animal and human.
  • the converted dose can be administered.
  • the term "individual” used herein refers to a subject who has or may develop a disease to which the composition of the present invention can be applied (prescribed), and may be a mammal such as a rat, mouse, or livestock, including humans. . Preferably, it may be a human, but is not limited thereto.
  • the pharmaceutical composition of the present invention may additionally contain known substances that exhibit a preventive or therapeutic effect on the above diseases.
  • the pharmaceutical composition of the present invention may further include a substance that increases the activity of immune cells expressing the fusion protein.
  • the pharmaceutical composition of the present invention can be administered in combination with conventionally known substances for preventing or treating the above diseases.
  • the immune cells expressing the fusion protein, substances showing a preventive or therapeutic effect for the disease, and/or substances that increase the activity of the immune cells can be administered simultaneously or sequentially.
  • Another aspect of the present invention provides the use of immune cells expressing the fusion protein or a pharmaceutical composition containing the same for preventing or treating cancer.
  • Another aspect of the present invention provides a method for preventing or treating cancer, comprising administering to an individual an immune cell expressing the fusion protein or a pharmaceutical composition containing the same.
  • Immune cells expressing the fusion protein, pharmaceutical composition, cancer, prevention, treatment, subject, and administration are the same as described above.
  • Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising an immune cell expressing the fusion protein and a dual-specific antibody as active ingredients.
  • kits for preventing or treating cancer including an immune cell expressing the fusion protein and a dual-specific antibody.
  • Immune cells and dual-specific antibodies expressing the fusion protein contained in the pharmaceutical composition and kit may be included in one formulation and may be administered in combination. At this time, the immune cells expressing the fusion protein and the dual-specific antibody may be administered simultaneously or sequentially.
  • Immune cells expressing the fusion protein, bispecific antibodies, cancer, prevention and treatment are the same as described above.
  • CD137 extracellular domain variants thereof or fragments thereof
  • CD137 extracellular domain variants or fragments thereof may include a mutation in any one selected from the group consisting of CRD1, CRD2, CRD3, CRD4, and combinations thereof.
  • the variant may include mutations in CRD2 and/or CRD3.
  • the mutations of CRD2 and CRD3 are the same as described above.
  • the CD137 extracellular domain variant or fragment thereof may include or consist of any one amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 27.
  • Antibody information used in the following experimental examples is shown in Table 1 below.
  • Fluorescently labeled antibodies of each FITC, PE, APC, BV421, BV605, and BV711 wavelength were used to stain NK cells. Specifically, 1.0 x 10 5 NK cells were harvested and washed once with Flow Cytometry Staining Buffer (eBioscience, 00-4222-26). Reaction was performed in a refrigerator at 4°C, shielded from light for 30 minutes, and analyzed using flow cytometry.
  • the transgene containing the 4-1BB and CD3 ⁇ domains was inserted into the pVAX1 vector using EcoRI and XhoI.
  • the vector was converted into a linear template strand using XbaI (NEB, R0145S) for mRNA production.
  • in vitro transcription was performed using the mMESSAGE mMACHINE TM T7 ULTRA Transcription Kit (Invitrogen, AM1345).
  • BBz Specific amino acid sequences and nucleic acid sequences are shown in Table 2.
  • Example 1.2 Isolation and proliferation of PB-NK cells
  • PBMC Peripheral Blood Mononuclear Cell
  • ficoll-Paque PLUS Cytiva, 17144002
  • Miltenyi Biotec's MACS Magnetically activated cell sorting
  • NK cells were electroporated using the Neon ® Transfection system (ThermoFisher) and MaxCyte's GTx.
  • NK cells were suspended at 5.0x10 6 cells/100 ⁇ L with Buffer T (Invitrogen, MPK10096). 2.5 ⁇ g of mRNA was added to the cell suspension, and electroporation was performed at 1850 V, 20 ms, 1 purse. GTx followed the manufacturer's instructions, and electroporation was performed with the code set to 'Expanded NK-5'.
  • a cell line expressing 4-1BB was isolated using anti-PE MicroBeads from Miltenyi Biotec (MiltenyiBiotec, 130-048-801).
  • Signal sequence (SEQ ID NO: 1), extracellular domain of CD137 (4-1BB) (SEQ ID NO: 15) or a variant thereof (SEQ ID NO: 16, 17 or 27), linker (SEQ ID NO: 11), transmembrane domain (SEQ ID NO: 8 ), a polynucleotide (SEQ ID NO: 12, 13, 14) encoding a fusion protein (SEQ ID NO: 18, 19, 20 or 31) containing a co-stimulatory domain (SEQ ID NO: 9) and a primary signaling domain (SEQ ID NO: 10) or 30) was synthesized and loaded into the pCDH-EF1-MCS-IRES-Puro vector (SBI, CD532A-2). The amino acid sequences of the fusion proteins are shown in Tables 3 to 6.
  • the fusion protein according to the present invention was named “cBBR (Chimeric 4-1BB receptor)”.
  • Fusion protein domain amino acid sequence sequence number cBBR (SEQ ID NO: 18) Signal peptide MGNSCYNIVATLLVLNFERTRS One CRD1 LQDPCSNCPAGTFCDNNRNQICS 2 CRD2 PCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAEC 3 CRD3 DCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 6 CRD4 DCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 7 linker PSPADLSPGASSVTPPAPAREPGHSPQ 11 Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV 8 co-stimulatory domain KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 9 CD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEI
  • Fusion protein domain amino acid sequence sequence number cBBR_I64R (SEQ ID NO: 19) Signal peptide MGNSCYNIVATLLVLNFERTRS One CRD1 LQDPCSNCPAGTFCDNNRNQICS 2 CRD2 mutant PCPPNSFSSAGGQRTCD R CRQCKGVFRTRKECSSTSNAEC 4 CRD3 DCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 6 CRD4 DCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 7 linker PSPADLSPGASSVTPPAPAREPGHSPQ 11 Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV 8 co-stimulatory domain KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 9 CD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQK
  • Fusion protein domain amino acid sequence sequence number cBBR_V71R (SEQ ID NO: 20) Signal peptide MGNSCYNIVATLLVLNFERTRS One CRD1 LQDPCSNCPAGTFCDNNRNQICS 2 CRD2 mutant PCPPNSFSSAGGQRTCDICRQCKG R FRTRKECSSTSNAEC 5 CRD3 DCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 6 CRD4 DCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 7 linker PSPADLSPGASSVTPPAPAREPGHSPQ 11 Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV 8 co-stimulatory domain KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 9 CD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQK
  • Fusion protein domain amino acid sequence sequence number cBBR_ I64R/V71R (SEQ ID NO: 31) Signal peptide MGNSCYNIVATLLVLNFERTRS One CRD1 LQDPCSNCPAGTFCDNNRNQICS 2 CRD2 mutant PCPPNSFSSAGGQRTCD R CRQCKG R FRTRKECSSTSNAEC 26 CRD3 DCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 6 CRD4 DCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 7 linker PSPADLSPGASSVTPPAPAREPGHSPQ 11 Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV 8 co-stimulatory domain KRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 9 CD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEG
  • the 4-1BBL expression vector was constructed by synthesizing the polynucleotide of SEQ ID NO: 25 and loading it into the pCDH-EF1-MCS-IRES-Puro vector (SBI, CD532A-2).
  • pCDH-EF1-MCS-IRES-Puro vector loaded with polynucleotides encoding VSV-G, Rev, and cBBR or variants thereof (SEQ ID NO: 12, 13, 14 or 30) or a polynucleotide encoding 4-1BBL The pCDH-EF1-MCS-IRES-Puro vector loaded with SEQ ID NO: 25) was transduced into Lenti-X 293T cells (Clontech, 632180) using Lipofectamine 2000 (Thermo Fisher, 11668019).
  • each supernatant was harvested, mixed with Lenti-X Concentrator (Clontech, 631232) in a 1:3 ratio, and stored in the refrigerator for more than 1 hour.
  • Lenti-X Concentrator (Clontech, 631232) in a 1:3 ratio, and stored in the refrigerator for more than 1 hour.
  • Each of the above mixed solutions was centrifuged at 1,500g at 4°C, and the pellet was collected and used as virus particles capable of expressing cBBR or a variant thereof and virus particles expressing the 4-1BBL gene, respectively.
  • K562 cells and Nalm6 cells were purchased from the European collection of authenticated cell cultures (ECACC), and Jurkat T cells and Ramos cells were purchased from the Korean Cell Line Bank.
  • ECACC European collection of authenticated cell cultures
  • Various cBBR Jurkat T cells and 4-1BBL Nalm6 cells were transduced using the lentivirus prepared in Example 2.2 above. All cells were cultured using RPMI-1640 (Hyclone, SH30027.01) medium containing 10% FBS (Gibco, 10100147).
  • PBMC Peripheral Blood Mononuclear Cell
  • Ficoll Paque PLUS Cytiva, 17144002
  • T cells in PBMC were isolated using CD4+ T cell isolation kit, human (Miltenyi biotec, 130-096-533) and CD8+ T cell isolation kit, human (Miltenyi biotec, 130-096-495).
  • T cells isolated as above were activated by adding T Cell TransAct, human (Miltenyi biotec, 130-111-160).
  • Activated T cells were cultured using RPMI-1640 medium containing 10% FBS and 300 U/mL Proleukin IL-2 (NOVARTIS).
  • cBBR T cells The day after inducing activation, cBBR or its variants (I64R, V71R) were transduced into T cells using the lentivirus prepared in Example 2.2, and cultured for up to 14 days.
  • the cells prepared as above are hereinafter referred to as “cBBR T cells.”
  • Lymphoblast patient-derived NK cells NK92MI were supplemented with 12.5% FBS (fetal bovine serum, Gibco, 10100147), 12.5% horse serum (Gibco, 16050122), and 1% penicillin/streptomycin ( Gibco, 15140-122), alpha MEM (Cytiva, containing 0.2mM inositol (DAEJUNG, 5008-4105), 0.1mM 2-mercaptoethanol (Gibco, 21985023), and 0.02mM folic acid (Sigma Aldrich, F8758).
  • SH30265.01 was cultured in a CO 2 incubator using medium.
  • cBBR NK92MI cells 4-1BB expressing NK92MI cells.
  • a cell line was developed to determine whether NK92MI cells expressing 4-1BB are activated by the monoclonal antibody urelumab (Figure 2). Titration evaluation was performed to confirm the appropriate concentration of urelumab ( Figures 3 and 4). Then, in order to confirm the IFN- ⁇ produced by NK cells, the concentration of IFN- ⁇ was measured in the NK cell culture medium using the Human IFN-gamma Quantikine ELISA kit (R&D systems, DIF50C).
  • each cBBR Jurkat T cell, 4-1BBL Nalm6 cell, and cBBR T cell (1.0 ⁇ 10 5 cells) were washed once with Flow Cytometry Staining Buffer (eBioscience, 00-4222-26), and then incubated with each antibody. was treated and reacted at 4°C, shielded from light for 30 minutes. After the reaction was completed, the stained cells were analyzed using flow cytometry. At this time, the antibodies used are shown in Table 7.
  • cBBR Jurkat T cells were stained with 1 ⁇ M CellTracker Deep Red Dye (Invitrogen, C34565), and 4-1BBL Nalm6 cell line was stained with 5 ⁇ M CellTrace CFSE Cell Proliferation Kit (Invitrogen, C34554).
  • the two types of stained cells were co-cultured for 30 minutes and then fixed by adding 4% paraformaldehyde solution (Biosesang, PC2031-100-00).
  • the fixed cells were analyzed using flow cytometry (% of CFSE+ 7-AAD+ cells).
  • cBBR T cells and 4-1BBL Nalm6 cells were co-cultured to measure the cytotoxicity of cBBR T cells against cancer cells.
  • cBBR T cells were toxicity of cBBR T cells using the Cytotoxicity Assay Kit (Abcam, ab270780). 4-1BBL Nalm6 cells were stained with the CFSE reagent included in the kit, mixed with cBBR T cells at a ratio of 0.5:1, 1:1, and 2:1, respectively, and co-cultured for 24 hours. At this time, Nalm6, in which 4-1BBL was not transduced, was used as a control. After co-culture, the cells were stained with 7-AAD and dead cells were confirmed through flow cytometry (% of CFSE+ 7-AAD+ cells).
  • cBBR T cells (including mutants) exhibited cytotoxicity against 4-1BBL Nalm6 cells due to the binding of the 4-1BB extracellular domain and 4-1BBL.
  • cytotoxicity was reduced in cBBR T cells (I64R, V71R, I64R/V71R) in which some amino acids of cBBR were substituted (mutants). The above results are believed to be due to a decrease in binding force between the CD137 (4-1BB) extracellular domain of the cBBR variant and 4-1BBL.
  • a 96-well plate was coated with urelumab (MedChemExpress, HY-P99055) (1 ⁇ g/mL, 10 ⁇ g/mL), and then cBBR T cells (I64R, V71R, I64R/V71R) prepared by the method of Example 2 above were added. were inoculated at a concentration of 5 ⁇ 10 5 cell/mL and reacted for 24 hours. After the reaction was completed, the cells were stained with anti-CD8a antibody (APC) and anti-CD107a antibody (PE), respectively, and analyzed using flow cytometry. As a result, as shown in Figure 15, the binding affinity of cBBR T cells with amino acid substitutions (mutants) to 4-1BBL decreased. However, it was confirmed that the activity caused by Urelumab, an anti-CD137 antibody (agonist), was not reduced.
  • urelumab MedChemExpress, HY-P99055
  • cBBR T cells (I64R, V71R, I64R/V71R) produced by the method of Example 2 were treated with urelumab at a concentration of 1 ⁇ g/mL or 10 ⁇ g/mL, and then incubated for 24 hours to induce activation. After the reaction, the concentration of IFN- ⁇ secreted from activated cBBR T cells into the culture medium was measured using the Human IFN-gamma Quantikine ELISA Kit (R&D systems, SIF50C). As a result, as shown in Figure 16, the binding affinity of cBBR T cells with amino acid substitutions (mutants) to 4-1BBL decreased. However, it was confirmed that the activity caused by Urelumab, an anti-CD137 antibody (agonist), was not reduced.
  • the bispecific antibody used in the present invention was produced by Genescript Probio.
  • the amino acid sequence of the bispecific antibody is shown in Table 8 below.
  • the bispecific antibody is an anti-BCMA antibody, and an scFv that binds to 4-1BB is bound to the C terminus.
  • each cBBR T cell Control, I64R, V71R
  • Ramos cell 1.0 ⁇ 10 5 cells
  • Flow cytometry staining buffer eBioscience, 00-4222-26
  • the bispecific antibody prepared by the method was treated and reacted at 4°C for 30 minutes, shielded from light. After the reaction was completed, the stained cells were analyzed using flow cytometry.
  • cBBR T cells In order to confirm the killing ability of cBBR T cells whose activation was induced by a bispecific antibody (BiTE) against cancer cells, cBBR T cells (I64R, V71R) were treated with a bispecific antibody, and then cancer cells were killed using the method of Experimental Example 3. Cytotoxicity was measured. At this time, the bispecific antibody was treated at a concentration of 1 or 10 ⁇ g/mL for 24 hours.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Immunology (AREA)
  • Medicinal Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Biochemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Zoology (AREA)
  • Cell Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Biophysics (AREA)
  • Biomedical Technology (AREA)
  • Microbiology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Wood Science & Technology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Biotechnology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Toxicology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • General Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Epidemiology (AREA)
  • Peptides Or Proteins (AREA)

Abstract

cBBR T cells in which a 4-1BB/CD3ζ (cBBR) fusion protein or a variant thereof is overexpressed in T cells specifically kill cancer cells in which 4-1BBL is overexpressed. Compared to cBBR T cells comprising a native form 4-1BB extracellular domain, the T cells, in which cBBR is overexpressed, comprising a 4-1BB extracellular domain variant have lower binding to 4-1BBL and cytotoxicity. If stimulated by Urelumab, the cBBR T cells, in which cBBR is overexpressed, comprising the variant have remarkably better activity than normal T cells. Furthermore, if reacted with an anti-BCMA/ anti-4-1BB bispecific antibody, the T cells show remarkably better killing activity, on cancer cells in which BCMA is expressed, than normal T cells. Therefore, T cells in which the cBBR fusion protein is overexpressed can be used as an anticancer agent. In addition, since it has been identified that NK cells in which a cBBR fusion protein is overexpressed are effectively activated by an anti-4-1BB antibody, it has been identified that the NK cells in which a cBBR fusion protein is overexpressed can be used as an anticancer agent.

Description

CD137 세포외 도메인을 포함하는 융합단백질, 이를 발현하는 면역세포 및 이의 용도Fusion protein containing the CD137 extracellular domain, immune cells expressing the same, and uses thereof
본 발명은 CD137 세포외 도메인 또는 이의 단편을 포함하는 융합단백질, 상기 융합단백질을 발현하는 면역세포 및 이의 용도에 관한 것이다.The present invention relates to a fusion protein containing the CD137 extracellular domain or a fragment thereof, immune cells expressing the fusion protein, and uses thereof.
우리의 면역 시스템은 자가 항원에 대한 내성을 유지하면서 침입 병원체를 효과적으로 제거할 수 있도록 미세 조율되고 있다. 이 중 CD3을 발현하고 있는 T 세포는 종양세포를 제거할 수 있는 효과기 세포(Effector cell)로 알려져 있다. 면역관문분자(Immune checkpoint)들은 T 세포 활성에 중추적인 역할을 하는 공동신호분자로서 T 세포 수용체(T cell receptor, TCR)의 신호를 억제하거나 상승되도록 조절한다(Immunological reviews, 2017, 276:52-65).Our immune system is being fine-tuned to effectively eliminate invading pathogens while maintaining tolerance to self-antigens. Among these, T cells expressing CD3 are known as effector cells that can eliminate tumor cells. Immune checkpoints are co-signaling molecules that play a central role in T cell activation and regulate the signal of the T cell receptor (TCR) to be suppressed or elevated (Immunological reviews, 2017, 276:52- 65).
또한, 자연살해세포(natural killer cell, 이하 NK 세포)는 암세포를 제거하여 항암 활성을 나타내는 것으로 알려져 있다. NK 세포의 활성은 다양한 활성화 및 억제 수용체 신호전달 균형에 의하여 조절된다. NK 세포의 항암 활성 또한 그 표면에 존재하는 다양한 면역수용체를 통해 암세포를 구분하여 이루어지는 것으로 알려져 있다. NK 세포는 암세포 외에도 암 줄기세포(cancer stem cell)를 제거할 수 있어 암의 발생, 증식, 전이를 억제할 뿐 아니라 완치 후 암의 재발을 저감시킬 수 있는 치료제의 개발원으로 각광받고 있다.In addition, natural killer cells (NK cells) are known to exhibit anticancer activity by eliminating cancer cells. The activity of NK cells is regulated by a balance of various activating and inhibitory receptor signaling. It is known that the anticancer activity of NK cells is also achieved by distinguishing cancer cells through various immune receptors present on their surface. NK cells can eliminate not only cancer cells but also cancer stem cells, so they are in the spotlight as a source of development for treatments that can not only suppress the occurrence, proliferation, and metastasis of cancer, but also reduce the recurrence of cancer after cure.
한편, 4-1BB는 TNF-수용체 슈퍼 패밀리(TNF-receptor superfamily, TNFRSF) 중 하나로서, 선천성 및 적응성 면역 세포 모두의 면역 세포의 활성화에 따라 발현되어 다양한 면역 세포의 활성화를 조절하는 공자극 분자이다. 4-1BB 작용제(agonist)는 면역 세포 증식, 생존, 사이토카인의 분비 및 CD8+ T 세포 용해 활성을 증진시킨다. 이러한 작용은 여러 마우스 종양 모델에서 항암 효과를 나타내어 항암제의 타겟으로 주목받고 있다. 하지만, 현재까지도 4-1BB 활성화를 통해 면역세포의 암 세포에 대한 항암활성을 증진시킬 수 있는 새로운 방법이 연구되고 있는 실정이다.Meanwhile, 4-1BB is one of the TNF-receptor superfamily (TNFRSF), a costimulatory molecule that is expressed upon activation of immune cells, both innate and adaptive immune cells, and regulates the activation of various immune cells. . The 4-1BB agonist enhances immune cell proliferation, survival, cytokine secretion, and CD8+ T cell lytic activity. This action has shown anticancer effects in several mouse tumor models and is attracting attention as a target for anticancer drugs. However, to this day, new methods that can enhance the anticancer activity of immune cells against cancer cells through 4-1BB activation are being studied.
이에 본 발명자들은 면역세포의 암 세포에 대한 항암활성을 증진시킬 수 있는 새로운 방법을 연구한 결과, CD137(또는 4-1BB)의 세포외 도메인 및 세포내 신호전달 도메인을 포함하는 융합단백질을 면역세포에 과발현 시킨 경우, 일반 면역세포에 비해 암세포에 대한 세포 독성이 증가한 것을 확인하였다. 특히, CD137(또는 4-1BB)의 세포외 도메인을 NK 세포에 과발현시킬 경우, 우렐루맙으로 자극을 줄 경우 일반 NK 세포에 비해 INF-γ(interferon gamma)를 5배 많이 방출하는 것을 확인함으로써 본 발명을 완성하였다.Accordingly, the present inventors studied a new method that can enhance the anti-cancer activity of immune cells against cancer cells, and as a result, a fusion protein containing the extracellular domain and intracellular signaling domain of CD137 (or 4-1BB) was introduced into immune cells. When overexpressed, it was confirmed that cytotoxicity against cancer cells increased compared to normal immune cells. In particular, it was confirmed that when the extracellular domain of CD137 (or 4-1BB) is overexpressed in NK cells and stimulated with urelumab, INF-γ (interferon gamma) is released 5 times more than normal NK cells. The invention was completed.
상기 과제를 해결하기 위하여, 본 발명의 일 측면은 하기 (i) 내지 (iii)을 포함하는 융합단백질을 제공한다: (i) CD137(4-1BB)의 세포외 도메인 또는 이의 단편; (ii) 막관통 도메인(Transmembrane domain); 및 (iii) 세포내 신호전달 도메인.In order to solve the above problem, one aspect of the present invention provides a fusion protein comprising the following (i) to (iii): (i) the extracellular domain of CD137 (4-1BB) or a fragment thereof; (ii) Transmembrane domain; and (iii) an intracellular signaling domain.
본 발명의 다른 측면은 상기 융합단백질을 암호화하는 폴리뉴클레오티드, 상기 폴리뉴클레오티드를 포함하는 벡터 및 바이러스를 제공한다.Another aspect of the present invention provides a polynucleotide encoding the fusion protein, a vector and a virus containing the polynucleotide.
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포를 제공한다.Another aspect of the present invention provides immune cells expressing the fusion protein.
본 발명의 또 다른 측면은 i) 상기 바이러스를 제조하는 단계; 및 ii) 상기 바이러스를 면역세포에 처리하는 단계;를 포함하는 상기 융합단백질이 발현된 면역세포의 생산 방법을 제공한다.Another aspect of the present invention includes the steps of i) preparing the virus; and ii) processing the virus into immune cells.
본 발명의 또 다른 측면은 상기 면역세포를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물; 및 상기 면역세포 및 이중 특이 항체를 포함하는 암 치료용 약학 조성물 및 키트를 제공한다.Another aspect of the present invention is a pharmaceutical composition for preventing or treating cancer comprising the immune cells as an active ingredient; and pharmaceutical compositions and kits for cancer treatment comprising the immune cells and bispecific antibodies.
본 발명의 또 다른 측면은 CD137 세포외 도메인 변이체 또는 이의 단편을 제공한다.Another aspect of the invention provides CD137 extracellular domain variants or fragments thereof.
4-1BB/CD3ζ 융합단백질(cBBR) 또는 이의 변이체를 T 세포에 과발현시킨 cBBR T 세포는 4-1BBL이 과발현된 암 세포를 특이적으로 사멸시켰다. 4-1BB 세포외 도메인 변이체를 포함하는 cBBR이 과발현된 T 세포의 경우, 천연형 4-1BB 세포외 도메인을 포함하는 cBBR T 세포와 비교하여 4-1BBL에 대한 결합력 및 세포 독성이 감소하는 것을 확인하였다. 상기 변이체를 포함하는 cBBR이 과발현된 cBBR T 세포는 우렐루맙(Urelumab)으로 자극할 경우, 일반 T 세포와 비교하여 활성이 월등히 증가하였다. 나아가, 항-BCMA/항-4-1BB 이중 특이 항체와 함께 반응시켰을 경우, BCMA가 발현된 암 세포에 대하여 일반 T 세포와 비교하여 월등히 우수한 살상 효과를 나타내었다. 따라서, 상기 cBBR이 과발현된 T 세포는 항암제로서 사용될 수 있다.cBBR T cells overexpressing the 4-1BB/CD3ζ fusion protein (cBBR) or a variant thereof specifically killed cancer cells overexpressing 4-1BBL. In the case of T cells overexpressing cBBR containing the 4-1BB extracellular domain variant, it was confirmed that the binding affinity and cytotoxicity to 4-1BBL were reduced compared to cBBR T cells containing the native 4-1BB extracellular domain. did. When cBBR T cells overexpressing cBBR containing the above mutant were stimulated with Urelumab, their activity significantly increased compared to normal T cells. Furthermore, when reacted with anti-BCMA/anti-4-1BB bispecific antibodies, it showed a significantly superior killing effect on BCMA-expressing cancer cells compared to normal T cells. Therefore, T cells overexpressing cBBR can be used as an anticancer agent.
도 1a는 4-1BB 및 CD3ζ 세포내도메인이 결합된 융합단백질을 코딩하는 핵산 카세트를 도식화한 것이다. 이때, SP는 신호 펩티드(Signal peptide), CRD는 시스테인-풍부 도메인(cystein-rich domain), TM은 막관통 도메인(transmembrane domain), ICD는 4-1BB의 세포내도메인(intracellular domain)을 나타낸다. 또한, CD3ζ는 CD3ζ의 세포내도메인을 나타낸다.Figure 1a is a schematic diagram of a nucleic acid cassette encoding a fusion protein combining 4-1BB and CD3ζ intracellular domains. At this time, SP represents the signal peptide, CRD represents the cysteine-rich domain, TM represents the transmembrane domain, and ICD represents the intracellular domain of 4-1BB. Additionally, CD3ζ represents the intracellular domain of CD3ζ.
도 1b는 4-1BB 및 CD3ζ 세포내도메인이 결합된 융합단백질을 코딩하는 핵산 카세트를 도식화한 것이다. 구체적으로, CD137 세포외 도메인의 CRD1, CRD2, CRD3 및 CRD4 중 CRD1 및 CDR1/CRD2 만을 발현시킨 융합단백질의 도메인 구조를 도식화한 것이다.Figure 1b is a schematic diagram of a nucleic acid cassette encoding a fusion protein combining 4-1BB and CD3ζ intracellular domains. Specifically, the domain structure of a fusion protein expressing only CRD1 and CDR1/CRD2 among CRD1, CRD2, CRD3, and CRD4 of the CD137 extracellular domain is schematized.
도 2는 형질전환된 NK92MI의 세포에서 4-1BB 및 CD3ζ 세포내도메인이 결합된 융합단백질이 안정적으로 발현됨을 보여주는 도면이다.Figure 2 is a diagram showing that a fusion protein combining 4-1BB and CD3ζ intracellular domains is stably expressed in transformed NK92MI cells.
도 3은 1.0 x 106 NK92MI 세포에 우렐루맙(Urelumab)을 농도별로 처리한 후 4-1BB의 발현 정도를 확인한 것이다.Figure 3 shows the level of expression of 4-1BB after treating 1.0 x 10 6 NK92MI cells with Urelumab at different concentrations.
도 4는 우렐루맙을 농도별로 처리한 후 4-1BB의 발현 정도를 FACS 분석을 통해 정량한 것이다.Figure 4 shows the expression level of 4-1BB quantified through FACS analysis after treatment with urelumab at different concentrations.
도 5는 우렐루맙 존재하에 배양된 형질전환된 NK92MI의 세포 및 일반 NK 세포에서 분비하는 INF-γ의 발현량을 측정한 것이다. 이때, NK92MI 세포 및 일반 NK 세포를 K562 세포와 함께 배양한 그룹을 대조군으로 하였다. Figure 5 shows the measurement of the expression level of INF-γ secreted by transformed NK92MI cells and normal NK cells cultured in the presence of urelumab. At this time, the group in which NK92MI cells and normal NK cells were cultured together with K562 cells was used as the control group.
도 6은 PBMC 유래 NK세포를 배양하여 CD137 mRNA를 Tranfection한 이후 Urelumab과 공동배양하는 실험에 대한 스케줄을 나타낸 것이다.Figure 6 shows a schedule for an experiment in which PBMC-derived NK cells were cultured, transfected with CD137 mRNA, and then co-cultured with Urelumab.
도 7은 유세포 분석기를 통해 시간에 따른 4-1BB의 발현 정도를 확인한 것이다.Figure 7 shows the expression level of 4-1BB over time using flow cytometry.
도 8은 ELISA를 통해 분비되는 IFN-γ의 양을 측정한 것이다.Figure 8 shows the amount of IFN-γ secreted through ELISA.
도 9는 CD137 세포외 도메인의 CRD1 및 CRD1/CRD2 만 있음에도 불구하고, 유세포분석기를 통해 Urelumab과 4B4-1 항체가 잘 결합한다는 것을 확인한 것이다.Figure 9 shows that, despite the presence of only CRD1 and CRD1/CRD2 of the CD137 extracellular domain, it was confirmed through flow cytometry that Urelumab and the 4B4-1 antibody bind well.
도 10은 본 발명의 일 구체예로 키메라 인게이저 수용체(cBBR)가 과발현된 T(CER T, cBBR T) 세포의 모식도를 나타낸 도면이다.Figure 10 is a diagram showing a schematic diagram of T (CER T, cBBR T) cells overexpressing chimeric engager receptor (cBBR) as an embodiment of the present invention.
도 11은 Jurkat T 세포에 과발현된 cBBR(Chimeric 4-1BB receptor) 또는 이의 변이체(I64R, V71R) 및 Nalm6 세포에 과발현된 4-1BBL을 유세포 분석을 통해 확인한 결과를 나타낸 그래프이다.Figure 11 is a graph showing the results of confirmation of cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R) overexpressed in Jurkat T cells and 4-1BBL overexpressed in Nalm6 cells through flow cytometry.
도 12는 혈액 유래 T 세포에 과발현된 cBBR 또는 이의 변이체(I64R, V71R)의 발현을 유세포 분석을 통해 확인한 결과를 나타낸 그래프이다.Figure 12 is a graph showing the results of confirming the expression of cBBR or its variants (I64R, V71R) overexpressed in blood-derived T cells through flow cytometry.
도 13은 Jurkat T 세포에 과발현된 cBBR(Chimeric 4-1BB receptor) 또는 이의 변이체(I64R, V71R) 및 Nalm6 세포에 과발현된 4-1BBL의 결합율을 유세포 분석을 통해 확인한 결과를 나타낸 그래프이다.Figure 13 is a graph showing the results of confirming the binding rate of cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R) overexpressed in Jurkat T cells and 4-1BBL overexpressed in Nalm6 cells through flow cytometry.
도 14는 cBBR(Chimeric 4-1BB receptor) 또는 이의 변이체(I64R, V71R, I64R/V71R)가 과발현된 T 세포(cBBR T 세포)의 4-1BBL이 과발현된 암세포에 대한 살상능을 CFSE 형광 염색 및 유세포 분석으로 측정한 결과를 나타낸 그래프이다.Figure 14 shows the killing ability of T cells (cBBR T cells) overexpressing cBBR (Chimeric 4-1BB receptor) or its variants (I64R, V71R, I64R/V71R) against cancer cells overexpressing 4-1BBL by CFSE fluorescence staining and This is a graph showing the results measured by flow cytometry.
도 15는 우렐루맙에 의한 cBBR의 변이체(I64R, V71R, I64R/V71R)가 과발현된 cBBR T 세포의 활성화 정도를 항-CD8a 항체 및 항-CD107a 항체로 염색하여 유세포 분석을 통해 확인한 결과를 나타낸 그래프이다.Figure 15 is a graph showing the level of activation of cBBR T cells overexpressing cBBR variants (I64R, V71R, I64R/V71R) by urelumab, confirmed by flow cytometry after staining with anti-CD8a antibody and anti-CD107a antibody. am.
도 16은 우렐루맙에 의한 cBBR의 변이체(I64R, V71R, I64R/V71R)가 과발현된 cBBR T 세포의 활성화 정도를 IFN-γ 농도 측정을 통해 확인한 결과를 나타낸 그래프이다.Figure 16 is a graph showing the results of confirming the degree of activation of cBBR T cells overexpressing cBBR variants (I64R, V71R, I64R/V71R) by urelumab by measuring the concentration of IFN-γ.
도 17은 cBBR의 변이체(I64R, V71R)가 과발현된 cBBR T 세포 또는 Ramos 세포에 이중 특이 항체(항-BCMA/항-4-1BB)를 처리한 뒤, cBBR T 세포 또는 Ramos 세포 및 이중 특이 항체의 결합 정도를 유세포 분석을 통해 확인한 결과를 나타낸 그래프이다. BiTE:항-BCMA/항-4-1BB 이중 특이 항체, 19F2:항-BCMA 항체, 4B4-1:항-4-1BB 항체Figure 17 shows cBBR T cells or Ramos cells overexpressing cBBR variants (I64R, V71R) treated with a bispecific antibody (anti-BCMA/anti-4-1BB), and then cBBR T cells or Ramos cells and the bispecific antibody. This is a graph showing the results of confirming the degree of binding through flow cytometry. BiTE: anti-BCMA/anti-4-1BB bispecific antibody, 19F2: anti-BCMA antibody, 4B4-1: anti-4-1BB antibody
도 18은 cBBR의 변이체(I64R, V71R)가 과발현된 cBBR T 세포를 이중 특이 항체 및 BCMA가 발현된 암 세포와 공배양하여 cBBR T 세포의 암 세포 살상능을 확인한 결과를 나타낸 그래프이다.Figure 18 is a graph showing the results of confirming the cancer cell killing ability of cBBR T cells by co-culturing cBBR T cells overexpressing cBBR variants (I64R, V71R) with cancer cells expressing dual specific antibodies and BCMA.
도 19는 형질전환된 T 세포 및 AB-101 항체를 투여한 경우 종양억제가 이루어짐을 확인하였다.Figure 19 confirms that tumor suppression occurred when transformed T cells and AB-101 antibody were administered.
CD137 세포외 도메인을 포함하는 융합단백질Fusion protein containing CD137 extracellular domain
본 발명의 일 측면은 하기 (i) 내지 (iii)을 포함하는 융합단백질을 제공한다:One aspect of the present invention provides a fusion protein comprising (i) to (iii) below:
(i) CD137(4-1BB) 세포외 도메인 또는 이의 단편; (i) CD137 (4-1BB) extracellular domain or fragment thereof;
(ii) 막관통 도메인(Transmembrane domain); 및(ii) Transmembrane domain; and
(iii) 세포내 신호전달 도메인.(iii) Intracellular signaling domain.
본 명세서에서 사용하는 용어, "CD137"은 종양 괴사 인자 수용체 패밀리에 속하는 막 단백질이다. CD137은 종양 괴사 인자 수용체 슈퍼패밀리 구성원 9 또는 4-1BB로 지칭된다. 구체적으로, CD137은 종양 괴사 인자(Tumor necrosis factor) 수용체로서, T 세포에서 주로 공자극 분자(Co-stimulatory molecule)로서 작용한다. 상기 CD137 세포외 도메인은 4개의 CRD(Cysteine-rich pseudo repeat) 도메인을 포함하고 있다. 상기 CD137 단백질은 바람직하게는 인간 CD137 단백질일 수 있으나, 이에 제한되지 않는다. As used herein, “CD137” is a membrane protein belonging to the tumor necrosis factor receptor family. CD137 is referred to as tumor necrosis factor receptor superfamily member 9 or 4-1BB. Specifically, CD137 is a tumor necrosis factor receptor and mainly acts as a co-stimulatory molecule in T cells. The CD137 extracellular domain contains four cysteine-rich pseudo repeat (CRD) domains. The CD137 protein may preferably be human CD137 protein, but is not limited thereto.
상기 "CD137 세포외 도메인 단편"은 천연형 CD137 세포외 도메인 단백질의 N 말단 및/또는 C 말단의 일부가 결실된(truncated) 단편을 의미하는 것으로, 천연형 CD137 세포외 도메인과 동일하거나 유사한 4-1BBL 결합력을 나타내는 것일 수 있다. 상기 "천연형(Wild type)"은 자연에서 발견되는 단백질 또는 이를 암호화하는 핵산을 모두 포함하는 것으로 야생형과 혼용하여 기재할 수 있다.The "CD137 extracellular domain fragment" refers to a fragment in which part of the N-terminus and/or C-terminus of the native CD137 extracellular domain protein is truncated, and is identical to or similar to the native CD137 extracellular domain 4- This may indicate 1BBL binding force. The “wild type” includes all proteins found in nature or nucleic acids encoding them and can be used interchangeably with wild type.
본 발명에서 상기 CD137 세포외 도메인 또는 이의 단편은 서열번호 2의 아미노산 서열을 포함하는 CRD1; 서열번호 3의 아미노산 서열을 포함하는 CRD2; 서열번호 6의 아미노산 서열을 포함하는 CRD3; 서열번호 7의 아미노산 서열을 포함하는 CRD4; 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나를 포함하는 것일 수 있다. In the present invention, the CD137 extracellular domain or fragment thereof includes CRD1 comprising the amino acid sequence of SEQ ID NO: 2; CRD2 comprising the amino acid sequence of SEQ ID NO: 3; CRD3 comprising the amino acid sequence of SEQ ID NO: 6; CRD4 comprising the amino acid sequence of SEQ ID NO: 7; And it may include any one selected from the group consisting of combinations thereof.
일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD2를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD3을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1 및 CRD2를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1 및 CRD3을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1 및 CRD4을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD2 및 CRD3을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD2 및 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD3 및 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1, CRD2 및 CRD3을 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1, CRD2 및 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1, CRD3 및 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD2, CRD3 및 CRD4를 포함하는 것일 수 있다. 일 구체예로, CD137 세포외 도메인 또는 이의 단편은 CRD1, CRD2, CRD3 및 CRD4를 포함하는 것일 수 있다. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD2. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1 and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2 and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2 and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD3 and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, and CRD3. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD3, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD2, CRD3, and CRD4. In one embodiment, the CD137 extracellular domain or fragment thereof may include CRD1, CRD2, CRD3, and CRD4.
바람직하게, 상기 CD137 세포외 도메인 또는 이의 단편은 CRD1의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다. 바람직하게, 상기 CD137 세포외 도메인 또는 이의 단편은 CRD1 및 CRD2의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다. 바람직하게 상기 CD137의 세포외 도메인 또는 이의 단편은 CRD1, CRD2, CRD3 및 CRD4의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.Preferably, the CD137 extracellular domain or fragment thereof may include or consist of the amino acid sequence of CRD1. Preferably, the CD137 extracellular domain or fragment thereof may include or consist of the amino acid sequences of CRD1 and CRD2. Preferably, the extracellular domain of CD137 or a fragment thereof may include or consist of the amino acid sequences of CRD1, CRD2, CRD3, and CRD4.
본 발명의 일 실시예에서 상기 CD137 세포외 도메인 또는 이의 단편은 서열번호 15의 아미노산 서열을 포함하는 것일 수 있다.In one embodiment of the present invention, the CD137 extracellular domain or fragment thereof may include the amino acid sequence of SEQ ID NO: 15.
또한, CD137의 세포외 도메인 또는 이의 단편과 동일한 활성을 갖거나 염색체 상의 상기 CD137 세포외 도메인을 암호화하는 유전자 위치가 동일한 이상, 상기 단백질의 하나 또는 몇 개의 아미노산이 첨가, 결실, 치환되는 서열로 구성될 수 있다. CD137 세포외 도메인은 서열번호 15의 아미노산 서열과 약 80%, 약 85% , 약 90%, 약 95%, 약 96%, 약 97%, 약 98%, 약 99% 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다. In addition, if it has the same activity as the extracellular domain of CD137 or a fragment thereof, or the location of the gene encoding the extracellular domain of CD137 on the chromosome is the same, the protein consists of a sequence in which one or several amino acids are added, deleted, or substituted. It can be. The CD137 extracellular domain has about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, about 99%, or 100% amino acid identity with the amino acid sequence of SEQ ID NO: 15. It may contain or consist of sequences.
상기 CD137 세포외 도메인 또는 이의 단편은 변이를 추가로 포함할 수 있다. The CD137 extracellular domain or fragment thereof may further include mutations.
본 명세서에서 상기 "변이"는 상술한 CD137 세포외 도메인 또는 이의 단편의 아미노산 일부가 치환된 형태를 의미한다. 즉, CD137 세포외 도메인 또는 이의 단편의 변이체는 천연형의 CD137 세포외 도메인 또는 이의 단편과 다른 서열을 가지는 것일 수 있다. 본 발명에서 상기 변이체는 천연형의 CD137과 비교하여 4-1BBL(CD137L)에 대한 결합능이 저하된 것일 수 있다. 여기서, 4-1BBL과의 결합능은 당업자에게 알려진 방법을 통해 측정할 수 있다.As used herein, “mutation” refers to a form in which some amino acids of the above-described CD137 extracellular domain or fragment thereof are substituted. That is, the variant of the CD137 extracellular domain or fragment thereof may have a different sequence from the native CD137 extracellular domain or fragment thereof. In the present invention, the mutant may have reduced binding ability to 4-1BBL (CD137L) compared to native CD137. Here, the binding ability with 4-1BBL can be measured using methods known to those skilled in the art.
상기 변이는 CRD1, CRD2, CRD3, CRD4 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나에 포함될 수 있다. The mutation may be included in any one selected from the group consisting of CRD1, CRD2, CRD3, CRD4, and combinations thereof.
일 구체예로, 상기 변이는 CRD1에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD2에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD3에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1 및 CRD2에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1 및 CRD3에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD2 및 CRD3에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD2 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD3 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1, CRD2 및 CRD3에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1, CRD2 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1, CRD3 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD2, CRD3 및 CRD4에 포함될 수 있다. 일 구체예로, 상기 변이는 CRD1, CRD2, CRD3 및 CRD4에 포함될 수 있다.In one embodiment, the mutation may be included in CRD1. In one embodiment, the mutation may be included in CRD2. In one embodiment, the mutation may be included in CRD3. In one embodiment, the mutation may be included in CRD4. In one embodiment, the mutation may be included in CRD1 and CRD2. In one embodiment, the mutation may be included in CRD1 and CRD3. In one embodiment, the mutation may be included in CRD1 and CRD4. In one embodiment, the mutation may be included in CRD2 and CRD3. In one embodiment, the mutation may be included in CRD2 and CRD4. In one embodiment, the mutation may be included in CRD3 and CRD4. In one embodiment, the mutation may be included in CRD1, CRD2, and CRD3. In one embodiment, the mutation may be included in CRD1, CRD2, and CRD4. In one embodiment, the mutation may be included in CRD1, CRD3, and CRD4. In one embodiment, the mutation may be included in CRD2, CRD3, and CRD4. In one embodiment, the mutation may be included in CRD1, CRD2, CRD3, and CRD4.
구체적으로, 상기 변이는 CRD2 및/또는 CRD3에 포함된 것일 수 있다.Specifically, the mutation may be included in CRD2 and/or CRD3.
이때, 상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산이 치환된 것일 수 있다. 또한, 상기 CRD3의 변이는 서열번호 6의 아미노산 서열에서 S14, M15, C16 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나의 아미노산이 치환된 것일 수 있다.At this time, the mutation of CRD2 is any one amino acid selected from the group consisting of P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26, and combinations thereof in the amino acid sequence of SEQ ID NO: 3. It may have been replaced. Additionally, the mutation of CRD3 may be a substitution of any one amino acid selected from the group consisting of S14, M15, C16, and combinations thereof in the amino acid sequence of SEQ ID NO: 6.
상기 치환에 의해 도입되는 "아미노산"은 글라이신(glycine), 알라닌(alanine), 발린(valine), 류신(leucine), 이소류신(isoleucine), 세린(serine), 트레오닌(threonine), 시스테인(cysteine), 메티오닌(methionine), 아스파르트산(aspartic acid), 글루탐산(glutamic acid), 아스파라긴(asparagine), 글루타민(glutamine), 리신(lysine), 아르기닌(arginine), 페닐알라닌(phenylalanine), 티로신(tyrosine), 트립토판(tryptophan), 히스티딘(histidine) 및 프롤린(proline)으로 이루어진 군으로부터 선택되는 어느 하나일 수 있다.The "amino acids" introduced by the substitution include glycine, alanine, valine, leucine, isoleucine, serine, threonine, cysteine, Methionine, aspartic acid, glutamic acid, asparagine, glutamine, lysine, arginine, phenylalanine, tyrosine, tryptophan ( It may be any one selected from the group consisting of tryptophan, histidine, and proline.
보다 더 구체적으로, 상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 I18R, V25R 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산으로 치환된 것일 수 있다.More specifically, the mutation of CRD2 may be a substitution of any one amino acid selected from the group consisting of I18R, V25R, and combinations thereof in the amino acid sequence of SEQ ID NO: 3.
일 구체예로 상기 CRD2의 변이는 서열번호 3의 18번째 아미노산인 이소루신(I)이 아르기닌(R)으로 치환(I18R)된 것일 수 있다. 일 구체예로 CRD2의 변이는 서열번호 3의 25번째 아미노산인 발린(V)이 아르기닌(R)으로 치환(V25R)된 것일 수 있다. 일 구체예로 상기 CRD2의 변이는 서열번호 3의 18번째 아미노산인 이소루신(I) 및 25번째 아미노산인 발린(V)이 각각 아르기닌(R)으로 치환(I18R, V25R)된 것일 수 있다.In one embodiment, the mutation in CRD2 may be one in which isoleucine (I), the 18th amino acid of SEQ ID NO: 3, is substituted with arginine (R) (I18R). As an example, a mutation in CRD2 may be a substitution of valine (V), the 25th amino acid of SEQ ID NO: 3, with arginine (R) (V25R). In one embodiment, the mutation in CRD2 may be one in which isoleucine (I), the 18th amino acid of SEQ ID NO: 3, and valine (V), the 25th amino acid, are each substituted with arginine (R) (I18R, V25R).
일 구체예로, 상기 변이를 포함하는 CRD2은 서열번호 4, 서열번호 5 및 서열번호 26으로 이루어진 군에서 선택되는 어느 하나의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다. In one embodiment, CRD2 containing the mutation may include or be composed of any one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 26.
또한, 본 발명의 일 실시예로 상기 변이를 포함하는 CD137 세포외 도메인은 서열번호 16, 서열번호 17 및 서열번호 27로 이루어진 군에서 선택되는 어느 하나의 아미노산 서열을 포함하는 것일 수 있다.Additionally, in one embodiment of the present invention, the CD137 extracellular domain containing the above mutation may include any one amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 27.
본 발명에서 상기 CD137 세포외 도메인 또는 이의 단편이 변이를 포함하는 경우, 상기 CD137 세포외 도메인은 천연형과 비교하여 4-1BBL(CD137L)에 대한 결합능이 저하되는 것일 수 있다. In the present invention, when the CD137 extracellular domain or a fragment thereof contains a mutation, the binding ability of the CD137 extracellular domain to 4-1BBL (CD137L) may be reduced compared to the native type.
본 발명에서 사용하는 용어, "막관통 도메인(Transmembrane domain)"은 세포막에 위치하는 단백질의 구조 중에서 세포외 도메인과 세포내 신호전달 도메인을 연결하며 세포막을 관통하고 있는 부위를 의미한다. 본 발명에 의한 융합단백질은 상기 막관통 도메인을 통하여 세포막에 고정될 수 있다. 상기 막관통 도메인은 T 세포 수용체(TCR), CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD152 및, CD154로 구성된 군으로부터 선택되는 어느 하나의 분자로부터 유래되는 것일 수 있다. 구체적으로, 상기 막관통 도메인은 CD137로부터 유래된 것일 수 있다.The term “transmembrane domain” used in the present invention refers to a portion of the protein structure located in the cell membrane that connects the extracellular domain and the intracellular signaling domain and penetrates the cell membrane. The fusion protein according to the present invention can be fixed to the cell membrane through the transmembrane domain. The transmembrane domain includes T cell receptor (TCR), CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, It may be derived from any one molecule selected from the group consisting of CD137, CD152, and CD154. Specifically, the transmembrane domain may be derived from CD137.
본 발명의 일 실시예에서 상기 막관통 도메인은 CD137로부터 유래된 것일 수 있다. 구체적으로 상기 막관통 도메인은 서열번호 8의 아미노산 서열을 포함하는 것일 수 있다.In one embodiment of the present invention, the transmembrane domain may be derived from CD137. Specifically, the transmembrane domain may include the amino acid sequence of SEQ ID NO: 8.
또한, 상기 막관통 도메인과 동일한 활성을 갖거나 염색체 상의 상기 CD137 유래의 막관통 도메인을 암호화하는 유전자 위치가 동일한 이상, 상기 단백질의 하나 또는 몇 개의 아미노산이 첨가, 결실, 치환되는 서열로 구성될 수 있다. CD137 유래의 막관통 도메인은 서열번호 8의 아미노산 서열과 약 95%, 약 96%, 약 97%, 약 98%, 약 99% 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다. In addition, as long as it has the same activity as the transmembrane domain or the location of the gene encoding the transmembrane domain derived from CD137 on the chromosome is the same, the protein may be composed of a sequence in which one or several amino acids are added, deleted, or substituted. there is. The transmembrane domain derived from CD137 may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 8.
본 발명에서 상기 (i) CD137 세포외 도메인 및 (ii) 막관통 도메인은 스페이서를 통하여 연결될 수 있다. 상기 스페이서는 링커를 의미하며, 단백질 또는 펩타이드일 수 있다. 또한, 약 1 내지 약 1,000개의 아미노산으로 구성될 수 있으며, 약 10 내지 약 300개, 약 15 내지 약 100개, 약 15 내지 약 60개 또는 약 15 내지 약 45의 아미노산을 포함하는 것일 수 있다. 또한, 상기 링커는 Fc 영역과 같은 인체 내 단백질의 단편일 수 있다. 또한, 상기 링커는 CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD152 및 CD154로 이루어진 군에서 선택되는 어느 하나의 분자에서 유래되는 것일 수 있다.In the present invention, the (i) CD137 extracellular domain and (ii) transmembrane domain may be connected through a spacer. The spacer refers to a linker and may be a protein or peptide. Additionally, it may be composed of about 1 to about 1,000 amino acids, and may include about 10 to about 300 amino acids, about 15 to about 100 amino acids, about 15 to about 60 amino acids, or about 15 to about 45 amino acids. Additionally, the linker may be a fragment of a protein in the human body, such as an Fc region. In addition, the linker consists of CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD152 and CD154. It may be derived from any one molecule selected from the group.
본 발명의 일 구체예로 상기 스페이서는 CD137에서 유래한 것일 수 있다. 구체적으로 상기 스페이서는 서열번호 11의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.In one embodiment of the present invention, the spacer may be derived from CD137. Specifically, the spacer may include or be composed of the amino acid sequence of SEQ ID NO: 11.
본 발명에서 사용하는 용어, "세포내 신호전달 도메인(Intracellular signaling domain)"은 세포 표면에 존재하는 세포외 결합 도메인이 생물학적 표적 분자와 결합하는 경우(예, CD137 세포외 도메인 및 4-1BBL의 결합), 세포의 활성화, 세포독성 인자 방출, 사이토카인 생산, 세포 증식 등의 반응을 유도하기 위하여 세포 내부로 신호를 전달하는 부위를 의미한다. As used in the present invention, the term "intracellular signaling domain" refers to the case where an extracellular binding domain present on the cell surface binds to a biological target molecule (e.g., binding of CD137 extracellular domain and 4-1BBL). ), refers to a site that transmits signals into the cell to induce reactions such as cell activation, release of cytotoxic factors, cytokine production, and cell proliferation.
본 발명에서 상기 세포내 신호전달 도메인은 공자극 도메인(Co-stimulatory signaling domain) 및/또는 1차 신호전달 도메인(Primary signaling domain)을 포함할 수 있다. In the present invention, the intracellular signaling domain may include a co-stimulatory signaling domain and/or a primary signaling domain.
본 명세서에서 사용하는 용어, "공자극 도메인"은 공자극 분자의 세포내 신호전달 도메인을 의미한다. 상기 "공자극 분자"는 세포외 결합 부위가 표적 분자와 결합 시 면역 세포의 효율적 활성화 및 기능을 유도하는 2차 신호를 제공하는 Fc 수용체 이외의 세포 표면 분자이다. 상기 공자극 도메인은 OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1(CD11a/CD18), ICOS(CD278) 및 CD137로 이루어진 군에서 선택되는 어느 하나의 분자로부터 유래된 것일 수 있다. As used herein, the term “co-stimulatory domain” refers to the intracellular signaling domain of a co-stimulatory molecule. The “co-stimulatory molecule” is a cell surface molecule other than the Fc receptor that provides a secondary signal that induces efficient activation and function of immune cells when the extracellular binding site binds to the target molecule. The costimulatory domain may be derived from any one molecule selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and CD137. .
일 구체예로 상기 공자극 도메인은 CD137로부터 유래된 것일 수 있다. 구체적으로, 상기 공자극 도메인은 서열번호 9의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.In one embodiment, the costimulatory domain may be derived from CD137. Specifically, the co-stimulatory domain may include or consist of the amino acid sequence of SEQ ID NO: 9.
본 명세서에서 사용하는 용어, "1차 신호전달 도메인"은 T 세포의 CD3 복합체를 구성하는 단백질로, T 세포의 1차 활성화를 자극 또는 억제 방식으로 조절하는 신호전달 도메인을 의미한다. 상기 T 세포의 활성화를 자극하는 1차 신호전달 도메인은 면역수용체 티로신-기반 활성화 모티브 또는 ITAM(Immunoreceptor tyrosine-based activation motif)으로 공지되어 있는 신호전달 모티브를 함유할 수 있다. 1차 신호전달 도메인을 함유하는 ITAM은 TCRζ, FcRγ, FcRβ, CD3γ, CD3δ, CD3ε, CD3ζ, CD22, CD79a, CD79b 및 CD66d로 이루어진 군에서 선택되는 어느 하나의 분자로부터 유래된 것일 수 있다. As used herein, the term “primary signaling domain” refers to a protein that constitutes the CD3 complex of T cells and refers to a signaling domain that regulates the primary activation of T cells in a stimulatory or inhibitory manner. The primary signaling domain that stimulates activation of the T cell may contain a signaling motif known as an immunoreceptor tyrosine-based activation motif or ITAM (Immunoreceptor tyrosine-based activation motif). ITAM containing the primary signaling domain may be derived from any one molecule selected from the group consisting of TCRζ, FcRγ, FcRβ, CD3γ, CD3δ, CD3ε, CD3ζ, CD22, CD79a, CD79b and CD66d.
본 발명의 일 구체예로, 상기 1차 신호전달 도메인은 CD3ζ로부터 유래한 것일 수 있다. 본 명세서에서 사용한 용어, "CD3ζ"는 CD3 복합체를 구성하는 단백질이다. CD3 복합체는 세포독성 T 세포와 T 헬퍼 세포 모두를 활성화하는 데 관여하는 단백질 복합체이자 T 세포 공동 수용체이다. CD3 복합체는 CD3α 사슬, CD3β 사슬, CD3γ 사슬, CD3δ 사슬, CD3ε 사슬 및 CD3ζ를 포함하고 있다. 구체적으로 상기 1차 신호전달 도메인은 서열번호 10의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.In one embodiment of the present invention, the primary signaling domain may be derived from CD3ζ. The term “CD3ζ” used herein refers to a protein that constitutes the CD3 complex. The CD3 complex is a protein complex and T cell co-receptor involved in activating both cytotoxic T cells and T helper cells. The CD3 complex contains the CD3α chain, CD3β chain, CD3γ chain, CD3δ chain, CD3ε chain, and CD3ζ. Specifically, the primary signaling domain may include or consist of the amino acid sequence of SEQ ID NO: 10.
본 명세서에서 사용한 용어, "CD28"은 T 세포 활성화 및 생존에 필요한 공동 자극 신호를 제공하는 T 세포에서 발현되는 단백질 중 하나이다. 이때, 상기 CD28의 세포내도메인은 서열번호 34의 아미노산 서열을 가질 수 있다.As used herein, “CD28” is one of the proteins expressed on T cells that provide costimulatory signals necessary for T cell activation and survival. At this time, the intracellular domain of CD28 may have the amino acid sequence of SEQ ID NO: 34.
본 명세서에서 사용한 용어, "OX40"는 종양 괴사 인자 수용체 슈퍼패밀리, 구성원 4로서, CD134로도 알려져 있다. 이때, 상기 OX40의 세포내도메인은 서열번호 35의 아미노산 서열을 가질 수 있다.As used herein, “OX40” refers to tumor necrosis factor receptor superfamily, member 4, also known as CD134. At this time, the intracellular domain of OX40 may have the amino acid sequence of SEQ ID NO: 35.
또한, 상기 각각의 공자극 도메인 및 1차 신호전달 도메인과 동일한 활성을 갖거나 염색체 상의 암호화하는 유전자 위치가 동일한 이상, 상기 각각의 도메인은 하나 또는 몇 개의 아미노산이 첨가, 결실, 치환되는 서열로 구성될 수 있다. 구체적으로 상기 공자극 도메인은 서열번호 9의 아미노산 서열과 약 95%, 약 96%, 약 97%, 약 98%, 약 99% 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다. 구체적으로 상기 1차 신호전달 도메인은 서열번호 10의 아미노산 서열과 약 95%, 약 96%, 약 97%, 약 98%, 약 99% 또는 100% 동일성을 갖는 아미노산 서열을 포함하거나 이들로 이루어질 수 있다.In addition, as long as it has the same activity as each of the co-stimulatory domains and the primary signaling domain or has the same coding gene location on the chromosome, each of the domains consists of a sequence in which one or several amino acids are added, deleted, or substituted. It can be. Specifically, the co-stimulatory domain may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 9. Specifically, the primary signaling domain may include or consist of an amino acid sequence having about 95%, about 96%, about 97%, about 98%, about 99%, or 100% identity with the amino acid sequence of SEQ ID NO: 10. there is.
본 발명의 세포내 신호전달 도메인은 상기 공자극 도메인 및 1차 신호전달 도메인이 적절히 조합된 것일 수 있다. 일 구체예로서, 세포내 신호전달 도메인은 CD137 및 CD3ζ를 포함할 수 있다. 이때, 상기 공자극 도메인 및 1차 신호전달 도메인은 링커를 통해 연결될 수 있다. The intracellular signaling domain of the present invention may be an appropriate combination of the costimulatory domain and the primary signaling domain. In one embodiment, the intracellular signaling domain may include CD137 and CD3ζ. At this time, the co-stimulatory domain and the primary signaling domain may be connected through a linker.
융합단백질의 구조Structure of fusion protein
구체적으로, 본 발명의 융합단백질은 하기 구조식 (I)을 포함하는 것일 수 있다.Specifically, the fusion protein of the present invention may contain the following structural formula (I).
<구조식 (I)><Structural Formula (I)>
N'-A-(L1)n-TM-(L2)m-B-(L3)l-C-C' (I)N'-A-(L1)n-TM-(L2)m-B-(L3)l-C-C' (I)
이때, 상기 구조식 (I)에 있어서,At this time, in the structural formula (I),
상기 N'은 각 폴리펩타이드의 N 말단이고,Wherein N' is the N terminus of each polypeptide,
상기 C'은 각 폴리펩타이드의 C 말단이며,The C' is the C terminus of each polypeptide,
상기 - 은 결합을 의미하고,where - means bonding,
상기 A는 CD137의 세포외 도메인 또는 이의 단편이며;wherein A is the extracellular domain of CD137 or a fragment thereof;
상기 TM은 막관통 도메인이고;The TM is a transmembrane domain;
상기 B 및 C는 세포내 신호전달 도메인으로, 각각 독립적으로 공자극 도메인 및 1차 신호전달 도메인이며; B and C are intracellular signaling domains, each independently a co-stimulatory domain and a primary signaling domain;
상기 L1, L2 및 L3은 각각 펩타이드 링커이며,Wherein L1, L2 and L3 are each peptide linkers,
상기 n, m 및 l은 각각 독립적으로 0 또는 1이다.The n, m and l are each independently 0 or 1.
이때, 상기 CD137의 세포외 도메인 또는 이의 단편, 막관통 도메인 및 세포내 신호전달 도메인을 상술한 바와 동일하다.At this time, the extracellular domain or fragment thereof, transmembrane domain, and intracellular signaling domain of CD137 are the same as described above.
상기 L1은 약 1 내지 약 100개의 아미노산으로 구성될 수 있으며, 약 10 내지 약 80개, 약 15 내지 약 70개, 약 15 내지 약 60개 또는 약 15 내지 약 45개의 아미노산을 포함할 수 있다. 상기 L2 및 L3은 각각 약 1 내지 약 30개의 아미노산을 포함할 수 있다. 구체적으로, 상기 L2 및 L3은 각각 약 5 내지 약 25개, 약 10 내지 약 20개 또는 약 15개의 아미노산을 포함할 수 있다. 보다 구체적으로, 상기 L2 및 L3은 각각 약 2 내지 약 30개, 약 2 내지 약 28개, 약 2 내지 약 26개, 약 2 내지 약 24개, 약 2 내지 약 22개, 약 2 내지 약 20개, 약 2 내지 약 18개, 약 2 내지 약 16개, 약 2 내지 약 14개, 약 2 내지 약 12개 또는 약 2 내지 약 10개 아미노산을 포함할 수 있다. 일 구체예로, 상기 L1은 CD137 유래의 펩타이드일 수 있으며, 서열번호 11의 아미노산을 포함하거나 이들로 이루어진 것일 수 있다.The L1 may be composed of about 1 to about 100 amino acids, and may include about 10 to about 80 amino acids, about 15 to about 70 amino acids, about 15 to about 60 amino acids, or about 15 to about 45 amino acids. The L2 and L3 may each include about 1 to about 30 amino acids. Specifically, L2 and L3 may each include about 5 to about 25 amino acids, about 10 to about 20 amino acids, or about 15 amino acids. More specifically, the L2 and L3 are each about 2 to about 30, about 2 to about 28, about 2 to about 26, about 2 to about 24, about 2 to about 22, and about 2 to about 20. , about 2 to about 18 amino acids, about 2 to about 16 amino acids, about 2 to about 14 amino acids, about 2 to about 12 amino acids, or about 2 to about 10 amino acids. In one embodiment, the L1 may be a peptide derived from CD137, and may include or consist of the amino acid of SEQ ID NO: 11.
융합단백질의 구체예Specific examples of fusion proteins
CD137 세포내도메인 및 CD3z 포함Includes CD137 intracellular domain and CD3z
1) N'-CRD1(서열번호 2)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'1) N'-CRD1 (SEQ ID NO: 2)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-CD3z (SEQ ID NO: 10)-C'
2) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'2) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD3z (SEQ ID NO: 10) - C'
3) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'3) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD3z (SEQ ID NO: 10)-C'
4) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'4) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-CD3z (SEQ ID NO: 10)-C'
5) N'-CRD1(서열번호 2)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'5) N'-CRD1 (SEQ ID NO: 2) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD3z (SEQ ID NO: 10) - C'
6) N'-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD3z(서열번호 10)-C'6) N'-CRD4 (SEQ ID NO: 7)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-CD3z (SEQ ID NO: 10)-C'
CD137 세포내도메인 및 CD28 포함Includes CD137 intracellular domain and CD28
7) N'-CRD1(서열번호 2)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'7) N'-CRD1 (SEQ ID NO: 2)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-CD28 (SEQ ID NO: 34)-C'
8) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'8) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34) - C'
9) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'9) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34)-C'
10) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'10) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-CD28 (SEQ ID NO: 34)-C'
11) N'-CRD1(서열번호 2)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'11) N'-CRD1 (SEQ ID NO: 2) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - CD28 (SEQ ID NO: 34) - C'
12) N'-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-CD28(서열번호 34)-C'12) N'-CRD4 (SEQ ID NO: 7)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-CD28 (SEQ ID NO: 34)-C'
CD137 세포내도메인 및 OX40 포함Includes CD137 intracellular domain and OX40
13) N'-CRD1(서열번호 2)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'13) N'-CRD1 (SEQ ID NO: 2)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
14) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'14) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35) - C'
15) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'15) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35)-C'
16) N'-CRD1(서열번호 2)-CRD2(서열번호 3)-CRD3(서열번호 6)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'16) N'-CRD1 (SEQ ID NO: 2) - CRD2 (SEQ ID NO: 3) - CRD3 (SEQ ID NO: 6) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular Domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
17) N'-CRD1(서열번호 2)-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'17) N'-CRD1 (SEQ ID NO: 2) - CRD4 (SEQ ID NO: 7) - Linker (SEQ ID NO: 11) - TM (SEQ ID NO: 8) - CD137 intracellular domain (SEQ ID NO: 18) - OX40 (SEQ ID NO: 35) - C'
18) N'-CRD4(서열번호 7)-링커(서열번호 11)-TM(서열번호 8)-CD137 세포내도메인(서열번호 18)-OX40(서열번호 35)-C'18) N'-CRD4 (SEQ ID NO: 7)-Linker (SEQ ID NO: 11)-TM (SEQ ID NO: 8)-CD137 intracellular domain (SEQ ID NO: 18)-OX40 (SEQ ID NO: 35)-C'
이때, N'은 융합단백질의 N 말단을 의미하며, C'은 융합단백질의 C 말단을 의미한다.At this time, N' refers to the N terminus of the fusion protein, and C' refers to the C terminus of the fusion protein.
융합단백질을 암호화하는 폴리뉴클레오티드Polynucleotide encoding fusion protein
본 발명의 다른 측면은 상기 융합단백질을 암호화하는 폴리뉴클레오티드를 제공한다. 상기 융합단백질을 상술한 바와 동일하다. 구체적으로 상기 융합단백질을 암호화하는 폴리뉴클레오티드는 서열번호 21 내지 서열번호 23 및 서열번호 28로 이루어진 군에서 선택되는 어느 하나의 뉴클레오티드를 포함하거나 이들로 이루어진 것일 수 있다.Another aspect of the present invention provides a polynucleotide encoding the fusion protein. The fusion protein is the same as described above. Specifically, the polynucleotide encoding the fusion protein may include or be composed of any one nucleotide selected from the group consisting of SEQ ID NO: 21 to SEQ ID NO: 23 and SEQ ID NO: 28.
또한, 상기 폴리뉴클레오티드는 동일한 폴리펩타이드를 암호화한다면, 하나 이상의 염기가 치환, 결실, 삽입 또는 이들의 조합에 의해 변이될 수 있다. 폴리뉴클레오티드 서열을 화학적으로 합성하여 제조하는 경우, 당업계에 널리 공지된 합성법, 예를 들어 문헌(Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988)에 기술된 방법을 이용할 수 있으며, 트리에스테르, 포스파이트, 포스포르아미다이트 및 H-포스페이트 방법, PCR 및 기타 오토프라이머 방법, 고체 지지체상의 올리고뉴클레오티드 합성법 등을 들 수 있다.Additionally, if the polynucleotide encodes the same polypeptide, one or more bases may be mutated by substitution, deletion, insertion, or a combination thereof. When preparing a polynucleotide sequence by chemical synthesis, a synthesis method well known in the art, for example, a method described in the literature (Engels and Uhlmann, Angew Chem IntEd Engl., 37:73-127, 1988), can be used. and triester, phosphite, phosphoramidite and H-phosphate methods, PCR and other autoprimer methods, and oligonucleotide synthesis methods on solid supports.
구체적으로 상기 폴리뉴클레오티드는 각각의 서열번호 21 내지 서열번호 23 및 서열번호 28의 염기서열과 적어도 약 70%, 적어도 약 75%, 적어도 약 80%, 적어도 약 85%, 적어도 약 86%, 적어도 약 87%, 적어도 약 88%, 적어도 약 89%, 적어도 약 90%, 적어도 약 91%, 적어도 약 92%, 적어도 약 93%, 적어도 약 94%, 적어도 약 95%, 적어도 약 96%, 적어도 약 97%, 적어도 약 98%, 적어도 약 99% 또는 적어도 약 100%의 동일성을 가지는 핵산서열을 포함할 수 있다. Specifically, the polynucleotide is at least about 70%, at least about 75%, at least about 80%, at least about 85%, at least about 86%, and at least about the base sequences of SEQ ID NO: 21 to SEQ ID NO: 23 and SEQ ID NO: 28, respectively. 87%, at least about 88%, at least about 89%, at least about 90%, at least about 91%, at least about 92%, at least about 93%, at least about 94%, at least about 95%, at least about 96%, at least about It may include a nucleic acid sequence having an identity of 97%, at least about 98%, at least about 99%, or at least about 100%.
상기 폴리뉴클레오티드는 신호서열(Signal sequence) 또는 리더 서열(Leader sequence)을 코딩하는 핵산을 추가적으로 포함할 수 있다. 본 명세서에서 사용한 용어 "신호서열"은 목적 단백질의 분비를 지시하는 신호펩타이드를 의미한다. 상기 신호펩타이드는 숙주 세포에서 번역된 후에 절단된다. 구체적으로, 상기 신호서열은 ER(Endoplasmic reticulum) 막을 관통하는 단백질의 이동을 개시하는 아미노산 서열이다. The polynucleotide may additionally include a nucleic acid encoding a signal sequence or leader sequence. The term “signal sequence” used herein refers to a signal peptide that directs secretion of a target protein. The signal peptide is cleaved after translation in the host cell. Specifically, the signal sequence is an amino acid sequence that initiates the movement of proteins through the ER (Endoplasmic reticulum) membrane.
상기 신호서열은 당업계에 그 특징이 잘 알려져 있으며, 통상 16 내지 30개의 아미노산 잔기를 포함하나, 그보다 더 많거나 적은 아미노산 잔기를 포함할 수 있다. 통상적인 신호 펩타이드는 기본 N 말단 영역, 중심의 소수성 영역, 및 보다 극성인(polar) C 말단 영역의 세 영역으로 구성된다. 중심 소수성 영역은 미성숙 폴리펩타이드가 이동하는 동안 막지질 이중층을 통하여 신호서열을 고정시키는 4 내지 12개의 소수성 잔기를 포함한다. 개시 이후에, 신호서열은 흔히 신호 펩티다아제(Signal peptidase)로 알려진 세포 효소에 의하여 ER의 루멘(lumen) 내에서 절단된다. 이때, 상기 신호서열은 tPa(Tissue Plasminogen Activation), HSV gDs(Signal sequence of Herpes simplex virus glycoprotein D), 또는 성장 호르몬(Growth hormone)의 분비 신호서열일 수 있다. 바람직하게, 포유동물 등을 포함하는 고등 진핵 세포에서 사용되는 분비 신호서열을 사용할 수 있다. 또한, 상기 신호서열은 야생형 신호서열을 사용하거나, 숙주세포에서 발현 빈도가 높은 코돈으로 치환하여 사용할 수 있다. 일 구체예로 상기 신호서열은 CD137의 신호서열일 수 있다. 본 발명의 일 실시예에서 상기 신호서열은 서열번호 1의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.The signal sequence has well-known characteristics in the art, and usually contains 16 to 30 amino acid residues, but may contain more or fewer amino acid residues. A typical signal peptide consists of three regions: a basic N-terminal region, a central hydrophobic region, and a more polar C-terminal region. The central hydrophobic region contains 4 to 12 hydrophobic residues that anchor the signal sequence throughout the membrane lipid bilayer while the immature polypeptide moves. After initiation, the signal sequence is cleaved within the lumen of the ER by a cellular enzyme commonly known as signal peptidase. At this time, the signal sequence may be a secretion signal sequence of tPa (Tissue Plasminogen Activation), HSV gDs (Signal sequence of Herpes simplex virus glycoprotein D), or growth hormone. Preferably, a secretion signal sequence used in higher eukaryotic cells, including mammals, can be used. In addition, the signal sequence can be used as a wild-type signal sequence, or by replacing it with a codon that is frequently expressed in host cells. In one specific example, the signal sequence may be the signal sequence of CD137. In one embodiment of the present invention, the signal sequence may include or consist of the amino acid sequence of SEQ ID NO: 1.
폴리뉴클레오티드가 적재된 벡터Vector loaded with polynucleotide
본 발명의 또 다른 측면은 상기 폴리뉴클레오티드가 적재된 벡터를 제공한다. 상기 벡터는 구조식 (I)의 융합단백질을 암호화하는 폴리뉴클레오티드를 포함하거나 이들로 이루어진 것일 수 있다. 상기 융합단백질을 상술한 바와 동일하다.Another aspect of the present invention provides a vector loaded with the polynucleotide. The vector may contain or consist of a polynucleotide encoding a fusion protein of structural formula (I). The fusion protein is the same as described above.
본 명세서에서 사용하는 용어, "벡터"는 숙주세포에 도입되어 숙주세포 유전체 내로 재조합 및 삽입될 수 있다. 또는 상기 벡터는 에피좀으로서 자발적으로 복제될 수 있는 뉴클레오티드 서열을 포함하는 핵산 수단으로 이해된다. 상기 벡터는 선형 핵산, 플라스미드, 파지미드, 코스미드, RNA 벡터, 바이러스 벡터, 미니-염색체 및 이의 유사체들을 포함한다. 바이러스 벡터의 예로는 레트로바이러스, 아데노바이러스 및 아데노-관련 바이러스를 포함하나 이에 제한되지 않는다. As used herein, the term “vector” can be introduced into a host cell and recombined and inserted into the host cell genome. Alternatively, the vector is understood as a nucleic acid vehicle containing a nucleotide sequence capable of spontaneous replication as an episome. The vectors include linear nucleic acids, plasmids, phagemids, cosmids, RNA vectors, viral vectors, mini-chromosomes and analogs thereof. Examples of viral vectors include, but are not limited to, retroviruses, adenoviruses, and adeno-associated viruses.
구체적으로, 상기 벡터는 플라스미드 DNA, 파아지 DNA 등이 될 수 있고, 상업적으로 개발된 플라스미드(예, pUC18, pBAD, pIDTSAMRT-AMP 등), 대장균 유래 플라스미드(예, pYG601BR322, pBR325, pUC118, pUC119 등), 바실러스 서브틸리스 유래 플라스미드(예, pUB110, pTP5 등), 효모-유래 플라스미드(예, YEp13, YEp24, YCp50 등), 파아지 DNA(Charon4A, Charon21A, EMBL3, EMBL4, λgt10, λgt11, λZAP 등), 동물 바이러스 벡터(예, 레트로바이러스(retrovirus), 아데노바이러스(adenovirus), 백시니아 바이러스(Vaccinia virus) 등), 곤충 바이러스 벡터(배큘로바이러스(baculovirus) 등)이 될 수 있다. 상기 벡터는 숙주 세포에 따라서 단백질의 발현량과 수식 등이 다르게 나타나므로, 목적에 가장 적합한 숙주세포를 선택하여 사용함이 바람직하다.Specifically, the vector may be plasmid DNA, phage DNA, etc., commercially developed plasmids (e.g., pUC18, pBAD, pIDTSAMRT-AMP, etc.), E. coli-derived plasmids (e.g., pYG601BR322, pBR325, pUC118, pUC119, etc.) , Bacillus subtilis-derived plasmids (e.g., pUB110, pTP5, etc.), yeast-derived plasmids (e.g., YEp13, YEp24, YCp50, etc.), phage DNA (Charon4A, Charon21A, EMBL3, EMBL4, λgt10, λgt11, λZAP, etc.), It may be an animal virus vector (e.g., retrovirus, adenovirus, vaccinia virus, etc.) or an insect virus vector (baculovirus, etc.). Since the expression level and modification of the protein of the vector varies depending on the host cell, it is desirable to select and use the host cell most suitable for the purpose.
바람직하게는, 상기 벡터는 바이러스 벡터일 수 있으며, 상기 바이러스 벡터는 레트로바이러스, 렌티바이러스, 아데노바이러스, 아데노-연관 바이러스, 헤르페스바이러스, 폭스바이러스, 바큘로바이러스, 유두종바이러스 및 파보바이러스로부터 유래된 것일 수 있다. 본 발명의 일 실시예에서 상기 벡터는 렌티바이러스 벡터일 수 있다. Preferably, the vector may be a viral vector, and the viral vector is derived from a retrovirus, lentivirus, adenovirus, adeno-associated virus, herpesvirus, poxvirus, baculovirus, papillomavirus, and parvovirus. You can. In one embodiment of the present invention, the vector may be a lentiviral vector.
본 명세서에서 사용하는 용어, 목적 단백질의 "유전자 발현" 또는 "발현"은, DNA 서열의 전사, mRNA 전사체의 번역 및 융합단백질 생산물 또는 이의 단편의 분비를 의미하는 것으로 이해된다. 유용한 발현 벡터는 RcCMV(Invitrogen, Carlsbad) 또는 이의 변이체일 수 있다. 상기 발현 벡터는 포유류 세포에서 목적 유전자의 연속적인 전사를 촉진하기 위한 인간 CMV(cytomegalovirus) 프로모터, 및 전사 후 RNA의 안정상태 수준을 높이기 위한 우태 성장 인자(Bovine growth hormone) 폴리아데닐레이션 신호서열을 포함할 수 있다.As used herein, the term “gene expression” or “expression” of a protein of interest is understood to mean transcription of a DNA sequence, translation of an mRNA transcript, and secretion of a fusion protein product or fragment thereof. A useful expression vector may be RcCMV (Invitrogen, Carlsbad) or variants thereof. The expression vector contains a human CMV (cytomegalovirus) promoter to promote continuous transcription of the target gene in mammalian cells, and a bovine growth hormone polyadenylation signal sequence to increase the steady-state level of RNA after transcription. can do.
폴리뉴클레오티드가 포함된 바이러스Viruses containing polynucleotides
본 발명의 또 다른 측면은 상기 폴리뉴클레오티드가 포함된 바이러스를 제공한다. 이때, 상기 폴리뉴클레오티드는 상술한 바와 동일하다. 상기 바이러스는 레트로바이러스, 렌티바이러스, 아데노바이러스, 아데노-연관바이러스, 헤르페스바이러스, 폭스바이러스, 바큘로바이러스, 유두종바이러스 또는 파보바이러스일 수 있다.Another aspect of the present invention provides a virus containing the polynucleotide. At this time, the polynucleotide is the same as described above. The virus may be a retrovirus, lentivirus, adenovirus, adeno-associated virus, herpesvirus, poxvirus, baculovirus, papillomavirus or parvovirus.
융합단백질이 발현된 면역세포Immune cells expressing fusion protein
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포를 제공한다. 상기 융합단백질은 상술한 바와 동일하다.Another aspect of the present invention provides immune cells expressing the fusion protein. The fusion protein is the same as described above.
이때, 상기 CD137의 세포외 도메인은 CRD1(cystein-rich pseudo repeat 1), CRD2, CRD3, CRD4 및 이의 조합으로 이루어진 군에서 선택되는 적어도 어느 하나의 도메인을 포함하는 것일 수 있다. 이때, 면역세포에 과발현되는 CD137의 세포외 도메인은 하기의 구조중 어느 하나일 수 있다:At this time, the extracellular domain of CD137 may include at least one domain selected from the group consisting of CRD1 (cysteine-rich pseudo repeat 1), CRD2, CRD3, CRD4, and combinations thereof. At this time, the extracellular domain of CD137 overexpressed in immune cells may have any of the following structures:
i) N'-CRD1-C'; ii) N'-CRD1-CRD2-C'; iii) N'-CRD1-CRD2-CRD3-C'; 또는 iv) N'-CRD1-CRD2-CRD3-CRD4-C'.i) N'-CRD1-C'; ii) N'-CRD1-CRD2-C'; iii) N'-CRD1-CRD2-CRD3-C'; or iv) N'-CRD1-CRD2-CRD3-CRD4-C'.
이때, N'은 융합단백질의 N 말단을 의미하며, C'은 융합단백질의 C 말단을 의미한다. 또한, 이때, 상기 CRD1, CRD2, CRD3 및 CRD4는 변이를 포함할 수 있다. 이때, 상기 변이는 상술한 바와 같다.At this time, N' refers to the N terminus of the fusion protein, and C' refers to the C terminus of the fusion protein. Also, at this time, CRD1, CRD2, CRD3, and CRD4 may include mutations. At this time, the mutation is as described above.
또한, 면역세포에 과발현된 상기 CD137의 세포외 도메인은 CD137의 세포내도메인(intracellular domain) 또는 이의 단편; CD3ζ의 세포내도메인 또는 이의 단편; CD28의 세포내도메인 또는 이의 단편; 및 OX40 세포내도메인 또는 이의 단편;으로 이루어진 그룹 중에서 선택되는 적어도 어느 하나의 세포내도메인을 더 포함하는 것일 수 있다. 이때, 면역세포에 과발현된 상기 CD137의 세포내도메인의 일 구체예는 하기와 같다:In addition, the extracellular domain of CD137 overexpressed in immune cells may include the intracellular domain of CD137 or a fragment thereof; The intracellular domain of CD3ζ or a fragment thereof; The intracellular domain of CD28 or a fragment thereof; and OX40 intracellular domain or fragment thereof; it may further include at least one intracellular domain selected from the group consisting of. At this time, a specific example of the intracellular domain of CD137 overexpressed in immune cells is as follows:
i) N'-CD137의 세포내도메인 또는 이의 단편-C';i) N'-Intracellular domain of CD137 or fragment-C' thereof;
ii) N'-CD3ζ의 세포내도메인 또는 이의 단편-C';ii) the intracellular domain of N'-CD3ζ or its fragment -C';
iii) N'-CD28의 세포내도메인 또는 이의 단편-C';iii) N'-intracellular domain of CD28 or fragment-C' thereof;
iv) N'-OX40 세포내도메인 또는 이의 단편-C'';iv) N'-OX40 intracellular domain or fragment thereof-C'';
v) N'-CD137의 세포내도메인 또는 이의 단편-CD3ζ의 세포내도메인 또는 이의 단편-C';v) N'-intracellular domain of CD137 or fragment thereof-intracellular domain of CD3ζ or fragment thereof-C';
vi) N'-CD137의 세포내도메인 또는 이의 단편-CD28의 세포내도메인 또는 이의 단편-C''vi) N'-Intracellular domain of CD137 or fragment thereof-Intracellular domain of CD28 or fragment thereof-C''
vii) N'-CD137의 세포내도메인 또는 이의 단편-OX40 세포내도메인 또는 이의 단편-C';vii) N'-CD137 intracellular domain or fragment thereof-OX40 intracellular domain or fragment thereof-C';
viii) N'-CD3ζ의 세포내도메인 또는 이의 단편-CD28의 세포내도메인 또는 이의 단편-C';viii) N'-intracellular domain of CD3ζ or fragment thereof-intracellular domain of CD28 or fragment thereof-C';
ix) N'-CD3ζ의 세포내도메인 또는 이의 단편-OX40 세포내도메인 또는 이의 단편-C'; 또는ix) N'-CD3ζ intracellular domain or fragment thereof-OX40 intracellular domain or fragment thereof-C'; or
x) N'-CD28의 세포내도메인 또는 이의 단편-OX40 세포내도메인 또는 이의 단편-C'.x) N'-CD28 intracellular domain or fragment thereof-OX40 intracellular domain or fragment thereof-C'.
이때, N'은 융합단백질의 N 말단을 의미하며, C'은 융합단백질의 C 말단을 의미한다.At this time, N' refers to the N terminus of the fusion protein, and C' refers to the C terminus of the fusion protein.
또한, CD137의 세포내도메인, CD3ζ의 세포내도메인, CD28의 세포내도메인 및 OX40 세포내도메인은 상술한 바와 같다.In addition, the intracellular domain of CD137, the intracellular domain of CD3ζ, the intracellular domain of CD28, and the intracellular domain of OX40 are the same as described above.
상기 면역세포는 자연살해세포, T 세포 또는 대식세포일 수 있다. 상기 융합단백질이 발현된 면역세포는 상기 폴리뉴클레오티드가 도입되어 형질전환된 세포로, 상기 폴리뉴클레오티드가 적재된 벡터 또는 상기 폴리뉴클레오티드를 포함하는 바이러스를 이용하여 형질전환될 수 있다. 상기 폴리뉴클레오티드, 벡터 및 바이러스는 상술한 바와 동일하다.The immune cells may be natural killer cells, T cells, or macrophages. The immune cell expressing the fusion protein is a cell transformed by introducing the polynucleotide, and can be transformed using a vector loaded with the polynucleotide or a virus containing the polynucleotide. The polynucleotide, vector and virus are the same as described above.
본 명세서에서 사용하는 용어, "자연살해세포(Natural killer cell, NK cell)"는 선천성 면역계의 주요 성분을 구성하는 세포독성 림프구로서, 대형과립 림프구(Large granular lymphocyte, LGL)로 정의되고 림프계 전구세포(Common lymphoid progenitor, CLP) 생성 B 및 T 세포로부터 분화된 제3의 세포를 구성한다. 면역반응의 일익을 담당하는 림프구계 세포로, 정상인 혈액 내에 약 10~15% 가량 존재하며, 비자기(Non-self)와 반응할 때 높은 살해능을 가진다. 각종 바이러스에 감염된 세포나 세균 침투, 혹은 비정상 세포의 생성에 있어, 자연살해세포는 비특이적으로 즉각적으로 반응하여 이물질을 제거할 수 있다. 본 명세서에서 자연살해세포는 NK 세포라고도 한다.As used herein, the term "natural killer cell (NK cell)" refers to a cytotoxic lymphocyte that constitutes a major component of the innate immune system, and is defined as a large granular lymphocyte (LGL) and a lymphoid progenitor cell. (Common lymphoid progenitor, CLP) constitutes a third cell differentiated from B and T cells. It is a lymphocyte cell that plays a part in the immune response, exists in approximately 10-15% of normal blood, and has a high killing ability when reacting with non-self. In the case of cells infected with various viruses, bacterial invasion, or the creation of abnormal cells, natural killer cells can react immediately and non-specifically to remove foreign substances. In this specification, natural killer cells are also referred to as NK cells.
본 명세서에서 사용하는 용어, "T 세포"는 항원 특이적인 적응 면역을 주관하는 림프구의 하나로서, 항원과 접촉하지 않은 미성숙 T 세포 및 항원 접촉 후 성숙된 효과 T 세포(보조 T 세포, 세포독성 T 세포, 자연살상 T 세포) 및 기억 T 세포로 분류된다.As used herein, the term “T cell” refers to a type of lymphocyte that hosts antigen-specific adaptive immunity, including immature T cells that have not come into contact with an antigen and effector T cells that have matured after contact with an antigen (helper T cells, cytotoxic T cells). cells, natural killer T cells) and memory T cells.
T 세포의 활성화는 미접촉 T 세포 항원 수용체(T cell antigen receptor, TCR)가 MHC:항원 복합체와 결합하여 T 세포의 CD3 복합체인 CD3 감마(γ), 델타(δ), 엡실론(ε) 및 제타(ζ) 사슬의 ITAM을 인산화시킴으로써 유도된다. 상기 ITAM 인산화는 ZAP-70 단백질, LAT 단백질 및 SLP-76 단백질의 인산화를 유도하여 NFκB, AP-1 및 NFAT 등의 전사 인자를 활성화하는 신호 반응을 유도한다. 상기 NFκB, AP-1 및 NFAT와 같은 전사 인자들은 인터루킨-2(IL-2)의 발현을 촉진함으로써 T 세포가 분열 및 분화를 유도하게 된다. Activation of T cells occurs when a naïve T cell antigen receptor (TCR) binds to the MHC:antigen complex and activates the CD3 complexes of T cells: CD3 gamma (γ), delta (δ), epsilon (ε), and zeta ( ζ) is induced by phosphorylating the ITAM of the chain. The ITAM phosphorylation induces phosphorylation of ZAP-70 protein, LAT protein, and SLP-76 protein, thereby inducing a signal response that activates transcription factors such as NFκB, AP-1, and NFAT. Transcription factors such as NFκB, AP-1, and NFAT induce T cell division and differentiation by promoting the expression of interleukin-2 (IL-2).
본 명세서에서 사용하는 용어, "대식세포(macrophage)"는 탐식세포라고도 일컬어지며, 선천 면역을 담당하는 주요한 세포이다. 상기 대식세포는 쿠퍼 세포, 랑게르한스 세포, 미세아교세포 등과 같이 조직 내에 정착성으로 존재하기도 하고, 혈액 내에서 단핵구의 형태로 존재하기도 한다. 이때, 상기 단핵구는 수지상 세포나 대식세포로 분화할 수 있다. 대식세포는 세포 조직이나 이물질, 미생물, 암세포 등 외래 단백질을 흡수하고 소화시키는 식세포 작용 및 사이토카인 등의 분비를 통해 염증 반응을 조절한다. 또한, 항원을 제거한 뒤, 림프구에 항원을 전달하는 항원제시세포로서 역할을 수행하여 면역반응을 유도한다.As used herein, the term “macrophage” is also referred to as a phagocytic cell and is a major cell responsible for innate immunity. The macrophages may exist as settlers in tissues, such as Kupffer cells, Langerhans cells, and microglial cells, or may exist in the form of monocytes in the blood. At this time, the monocytes may differentiate into dendritic cells or macrophages. Macrophages control inflammatory responses through phagocytosis and secretion of cytokines, which absorb and digest foreign proteins such as cell tissue, foreign substances, microorganisms, and cancer cells. In addition, after removing the antigen, it acts as an antigen-presenting cell that delivers the antigen to lymphocytes, thereby inducing an immune response.
본 발명에서 상기 면역세포는 개체로부터 수득할 수 있으며, 다양한 조직으로부터 수득할 수 있다. 또한, 상기 면역세포는 개체로부터 수득한 변형이 없는 면역세포를 포함하고, 성숙한 면역세포 뿐 아니라, 미성숙한 면역세포를 포함할 수 있다. 상기 면역세포는 조직, 조혈세포, 조혈 줄기세포 또는 조혈 전구세포에서 분리하여 수득할 수 있다. 구체적으로, 상기 면역세포는 예를 들어 태반 조직, 태반 관류액, 제대혈, 태반혈, 말초혈, 비장, 간 등으로부터의 조혈세포, 조혈 줄기 또는 전구체를 포함할 수 있으나, 이에 제한되지 않는다. 상기 면역세포는 자가 또는 타가에서 수득한 세포일 수 있다. 또한, 상기 면역세포는 쥐, 생쥐, 가축, 인간 등의 포유동물로부터 유래인 것일 수 있다. 바람직하게는 면역세포는 치료를 받고자 하는 개체에서 수득한 것일 수 있다. 이때, 상기 면역세포는 개체의 혈액에서 직접 분리하여 사용하거나, 개체에서 수득한 미성숙 미분화(또는 미성숙) 면역세포 또는 줄기세포를 분화시켜 사용할 수 있다. 뿐만 아니라, 상기 면역세포는 iPSCs(Induced pluripotent stem cells)로부터 분화된 것일 수 있다.In the present invention, the immune cells can be obtained from an individual or from various tissues. Additionally, the immune cells include unmodified immune cells obtained from an individual and may include immature immune cells as well as mature immune cells. The immune cells can be obtained by separating them from tissues, hematopoietic cells, hematopoietic stem cells, or hematopoietic progenitor cells. Specifically, the immune cells may include, but are not limited to, hematopoietic cells, hematopoietic stems or precursors from placental tissue, placental perfusate, umbilical cord blood, placental blood, peripheral blood, spleen, liver, etc. The immune cells may be autologous or foreign cells. Additionally, the immune cells may be derived from mammals such as rats, mice, livestock, and humans. Preferably, the immune cells may be obtained from an individual seeking treatment. At this time, the immune cells can be directly isolated from the blood of the individual and used, or they can be used by differentiating immature undifferentiated (or immature) immune cells or stem cells obtained from the individual. In addition, the immune cells may be differentiated from iPSCs (Induced pluripotent stem cells).
상기 벡터 또는 바이러스를 면역세포 내로 도입하는 방법은 당해 분야에서 공지된 방법을 이용할 수 있으며, 예를 들면, 일시적 형질감염(Transient transfection), 미세주사, 형질도입(transduction), 세포융합, 칼슘 포스페이트 침전법, 리포좀 매개된 형질감염(Liposome-mediated transfection), DEAE 덱스트란-매개된 형질감염(DEAE Dextran-mediated transfection), 폴리브렌-매개된 형질감염(Polybrene-mediated transfection), 전기침공법(electroporation), 유전자 총(Gene gun) 또는 세포 내로 핵산을 유입시키기 위한 다른 공지의 방법에 의해 세포 내로 도입할 수 있다(Wu et al., J. Bio. Chem., 267:963-967, 1992; Wu and Wu, J. Bio. Chem., 263:14621-14624, 1988). 하지만, 이에 제한하는 것은 아니다.Methods for introducing the vector or virus into immune cells can be methods known in the art, such as transient transfection, microinjection, transduction, cell fusion, and calcium phosphate precipitation. Method, Liposome-mediated transfection, DEAE Dextran-mediated transfection, Polybrene-mediated transfection, electroporation , can be introduced into cells by a gene gun or other known methods for introducing nucleic acids into cells (Wu et al., J. Bio. Chem., 267:963-967, 1992; Wu and Wu, J. Bio. Chem., 263:14621-14624, 1988). However, it is not limited to this.
형질도입된 또는 형질감염된 면역세포는 벡터 또는 바이러스 감염 후, 탈체에서 증식될 수 있다. 일 구체예에서 형질감염된 면역세포는 증식을 위하여 최소 약 1일, 2일, 3일, 4일, 5일, 6일, 7일, 8일, 9일, 10일, 11일, 12일, 13일, 14일, 15일, 16일, 17일, 18일, 19일 또는 20일 동안 배양될 수 있으며, 바람직하게는 13일 내지 15일 동안 배양될 수 있다. Transduced or transfected immune cells can be propagated ex vivo following vector or viral infection. In one embodiment, the transfected immune cells are allowed to proliferate for at least about 1 day, 2 days, 3 days, 4 days, 5 days, 6 days, 7 days, 8 days, 9 days, 10 days, 11 days, 12 days, It can be cultured for 13, 14, 15, 16, 17, 18, 19 or 20 days, preferably for 13 to 15 days.
상기 면역세포에서 벡터가 잘 도입되었는지 확인하는 방법은 예를 들어, 당업자에게 널리 공지된 분자 생물학적 검정(예를 들면, 서던 및 노던 블로팅, RT-PCR 및 PCR); 생화학적 검정(예를 들면, 면역학적 방법(가령, ELISAs, 웨스턴 블롯, 유세포 분석))에 의한 특정 펩타이드의 존재 또는 부재의 검출을 포함할 수 있다.Methods for confirming whether the vector has been successfully introduced into the immune cells include, for example, molecular biological assays (e.g., Southern and Northern blotting, RT-PCR and PCR) well known to those skilled in the art; It may include detection of the presence or absence of a particular peptide by biochemical assays (e.g., immunological methods (e.g., ELISAs, Western blots, flow cytometry)).
융합단백질이 발현된 면역세포를 생산하는 방법Method for producing immune cells expressing fusion proteins
본 발명의 또 다른 측면은 i) 상기 폴리뉴클레오티드가 포함된 바이러스를 제조하는 단계; 및 ii) 상기 바이러스를 면역세포에 처리하는 단계를 포함하는 상기 융합단백질이 발현된 면역세포의 생산 방법을 제공한다. 이때, 상기 폴리뉴클레오티드, 폴리뉴클레오티드가 포함된 바이러스, 면역세포 및 융합단백질은 상술한 바와 동일하다.Another aspect of the present invention includes the steps of i) preparing a virus containing the polynucleotide; and ii) processing the virus into immune cells. It provides a method for producing immune cells expressing the fusion protein. At this time, the polynucleotide, virus containing the polynucleotide, immune cell, and fusion protein are the same as described above.
상기 바이러스를 제조하는 방법은 당해 분야에서 공지된 방법을 이용할 수 있다. 또한, 상기 바이러스로 형질감염된 면역세포는 감염 후, 추가로 배양하여 증식될 수 있다. Methods for producing the virus may use methods known in the art. Additionally, immune cells transfected with the virus can be proliferated by further culturing after infection.
본 발명에서 사용하는 용어, "배양"은 세포를 적당히 조절된 환경 조건에서 생육시키는 것을 의미하며, 본 발명의 배양과정은 당업계에 알려진 적당한 배지와 배양 조건에 따라 이루어질 수 있다. 이러한 배양 과정은 선택되는 세포에 따라 당업자가 용이하게 조정하여 사용할 수 있다. 상기 배지는 세포의 배양 시 사용되는 공지된 배지를 의미하며, 공지의 세포 배양용 배지 또는 이들의 변형된 배지를 모두 포함한다.The term "culture" used in the present invention refers to growing cells in appropriately controlled environmental conditions, and the culture process of the present invention can be carried out according to appropriate media and culture conditions known in the art. This culture process can be easily adjusted by a person skilled in the art depending on the cells selected. The medium refers to a known medium used for culturing cells, and includes all known media for cell culture or modified media thereof.
구체적으로 상기 바이러스로 형질감염된 면역세포는 감염 후 추가로 약 1일, 2일, 3일, 4일, 5일, 6일, 7일, 8일, 9일, 10일, 11일, 12일, 13일, 14일, 15일, 16일, 17일, 18일, 19일 또는 20일 동안 배양될 수 있으며, 바람직하게는 13일 내지 15일 동안 배양될 수 있다. Specifically, immune cells transfected with the above virus persist for approximately 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, and 12 days after infection. , may be cultured for 13 days, 14 days, 15 days, 16 days, 17 days, 18 days, 19 days or 20 days, preferably for 13 to 15 days.
또한, 상기 융합단백질이 발현된 면역세포의 생산 여부는 당업자에게 널리 공지된 분자 생물학적 검정, 생화학적 검정 등의 방법을 통하여 상기 융합단백질의 발현 여부를 확인함으로써 확인할 수 있다.In addition, the production of immune cells expressing the fusion protein can be confirmed by confirming the expression of the fusion protein through methods such as molecular biological assays and biochemical assays well known to those skilled in the art.
융합단백질이 발현된 면역세포를 포함하는 약학 조성물Pharmaceutical composition containing immune cells expressing fusion protein
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다. 이때, 상기 약학 조성물은 항-CD137 항체 또는 이중 특이 항체를 추가로 포함할 수 있다. 상기 융합단백질 및 면역세포는 상술한 바와 동일하다.Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer, comprising immune cells expressing the fusion protein as an active ingredient. At this time, the pharmaceutical composition may further include an anti-CD137 antibody or a bispecific antibody. The fusion protein and immune cells are the same as described above.
또한, 상기 암은 위암, 간암, 폐암, 대장암, 유방암, 전립선암, 난소암, 췌장암, 자궁경부암, 갑상선암, 후두암, 백혈병, 급성 골수성 백혈병, 뇌종양, 신경모세포종, 망막 모세포종, 두경부암, 침샘암, 림프종, 신장암, 흑색종, 다발 골수종, 뇌암, 골육종, 교모세포종, IgG-opsonized tumor, 림프종, 신경종, 중피종, 및 식도암으로 이루어진 군에서 선택되는 어느 하나일 수 있다.In addition, the above cancers include stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, leukemia, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, and salivary gland cancer. , lymphoma, kidney cancer, melanoma, multiple myeloma, brain cancer, osteosarcoma, glioblastoma, IgG-opsonized tumor, lymphoma, neuroma, mesothelioma, and esophageal cancer.
본 명세서에서 사용하는 용어, "항체"는 특정 항원과 면역학적으로 반응하는 면역글로불린 분자로, 항원을 특이적으로 인식하는 단백질 분자를 의미한다. 면역글로불린의 중쇄 및 경쇄는 각각 불변 영역(Constant region) 및 가변 영역(Variable region)을 포함할 수 있다. 면역글로불린의 경쇄 및 중쇄 가변 영역은, 상보성 결정 영역(Complementarity determining region, CDR)이라 불리는 3개의 다변가능한 영역 및 4개의 구조 영역(Framework region, FR)을 포함한다. 상기 CDR은 주로 항원의 에피토프에 결합하는 항원 결합 부위이다. As used herein, the term “antibody” refers to an immunoglobulin molecule that reacts immunologically with a specific antigen, and refers to a protein molecule that specifically recognizes the antigen. The heavy and light chains of immunoglobulins may each include a constant region and a variable region. The light and heavy chain variable regions of immunoglobulins include three variable regions called complementarity determining regions (CDRs) and four framework regions (FRs). The CDR is an antigen binding site that mainly binds to the epitope of the antigen.
상기 항체는 IgG, IgE, IgM, IgD 및 IgA에서 선택되는 어느 하나를 의미할 수 있으며, IgG의 하위 부류인 IgG1, IgG2, IgG3, IgG4, IgA1 또는 IgA2일 수 있다. 또한, 항체는 작용 항체 또는 길항 항체일 수 있다. 또한, 상기 항체는 전체(whole) 항체, 단일클론항체, 다클론항체, 단일 도메인 항체, 단쇄 항체, 다중특이적 항체, 인간 항체, 인간화 항체, 키메라 항체, 인트라바디(intrabody), scFv, Fab 단편, F(ab') 단편, 이황화 결합으로 연결한 Fv(sdFv) 및 상기 중 임의의 에피토프(epitope) 결합 단편을 포함할 수 있으며, 암 항원에 특이적으로 결합하는 한, 이에 제한되지 않는다. The antibody may refer to any one selected from IgG, IgE, IgM, IgD, and IgA, and may be a subclass of IgG, such as IgG1, IgG2, IgG3, IgG4, IgA1, or IgA2. Additionally, the antibody may be an agonistic antibody or an antagonistic antibody. In addition, the antibodies include whole antibodies, monoclonal antibodies, polyclonal antibodies, single domain antibodies, single chain antibodies, multispecific antibodies, human antibodies, humanized antibodies, chimeric antibodies, intrabodies, scFvs, and Fab fragments. , F(ab') fragment, Fv(sdFv) linked by a disulfide bond, and any of the above epitope binding fragments, and is not limited thereto, as long as it specifically binds to the cancer antigen.
본 명세서에서 사용하는 용어, "이중 특이 항체"는 적어도 하나 이상의 표적 또는 항원에 결합하는 물질을 의미한다. 본 발명에서 상기 이중 특이 항체는 암 항원(Tumor antigen)에 특이적으로 결합하는 제1 도메인; 및 CD137의 세포외 도메인에 특이적으로 결합하는 제2 도메인을 포함할 수 있다. As used herein, the term “dual specific antibody” refers to a substance that binds to at least one target or antigen. In the present invention, the bispecific antibody includes a first domain that specifically binds to a tumor antigen; and a second domain that specifically binds to the extracellular domain of CD137.
상기 이중 특이 항체의 일 구체예는 항원에 특이적으로 결합하는 항체에 항-CD137 항체의 단편, 예를 들어 scFv가 결합된 형태일 수 있다. 이때, 상기 scFv는 항원에 특이적으로 결합하는 항체의 C 말단에 결합될 수 있으며, 이때, 항체와 scFv는 펩타이드 링커를 통해 결합될 수 있다. 이때, 상기 펩타이드 링커는 (G4S)n일 수 있다. 또한, 이때 상기 펩타이드는 링커는 (G4S)4 일 수 있다.One specific example of the bispecific antibody may be a fragment of an anti-CD137 antibody, for example, scFv, bound to an antibody that specifically binds to an antigen. At this time, the scFv may be bound to the C terminus of an antibody that specifically binds to an antigen, and at this time, the antibody and scFv may be bound through a peptide linker. At this time, the peptide linker may be (G4S)n. Also, at this time, the linker of the peptide may be (G4S) 4 .
상기 이중 특이 항체의 또 다른 구체예는 인게이져(engager) 일 수 있다. 구체적으로, 상기 이중 특이 항체는 BiTE(Bi-specific T-cell engager) 형태일 수 있다. 구체적으로, 제1 도메인은 암 항원에 특이적으로 결합하는 항체 또는 scFv일 수 있으며, 제2 도메인은 CD137에 특이적으로 결합하는 scFv일 수 있다. 또한, 제1 도메인과 제2 도메인은 펩타이드 링커를 통해 결합되어 있을 수 있다.Another specific example of the bispecific antibody may be an engager. Specifically, the bispecific antibody may be in the form of BiTE (Bi-specific T-cell engager). Specifically, the first domain may be an antibody or scFv that specifically binds to a cancer antigen, and the second domain may be an scFv that specifically binds to CD137. Additionally, the first domain and the second domain may be linked through a peptide linker.
이때, 상기 암 항원은 알파 엽산 수용체, 5T4, αvβ6 인테그린, BCMA, B7-H3, B7-H6, CAIX, CD16, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a, CD79b, CD123, CD138, CD171, CEA, CSPG4, EGFR, ErbB2(HER2)를 포함하는 EGFR 패밀리, EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, 태아 AchR, FRα, GD2, GD3, 글리피칸-3(GPC3), HLA-A1+MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ESO-1, HLA-A2+NY-ESO-1, HLA-A3+NY-ESO-1, IL-11Rα, IL-13Rα2, 람다, 루이스-Y, 카파, 메소텔린, Muc1, Muc16, NCAM, NKG2D 리간드, NY-ESO-1, PRAME, PSCA, PSMA, ROR1, SSX, 서바이빈, TAG72, TEMs, VEGFR2 및 WT-1로 이루어진 군으로부터 선택되는 어느 하나인 것일 수 있다. At this time, the cancer antigen is alpha folate receptor, 5T4, αvβ6 integrin, BCMA, B7-H3, B7-H6, CAIX, CD16, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a. , CD79b, CD123, CD138, CD171, CEA, CSPG4, EGFR, EGFR family including ErbB2 (HER2), EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, fetal AchR, FRα, GD2, GD3, glypican -3(GPC3), HLA-A1+MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ESO-1, HLA-A2+NY-ESO-1, HLA-A3+NY- ESO-1, IL-11Rα, IL-13Rα2, Lambda, Lewis-Y, Kappa, Mesothelin, Muc1, Muc16, NCAM, NKG2D Ligand, NY-ESO-1, PRAME, PSCA, PSMA, ROR1, SSX, Survi It may be any one selected from the group consisting of bin, TAG72, TEMs, VEGFR2, and WT-1.
또한, 상기 이중 특이 항체의 제2 도메인인 CD137의 세포외 도메인 결합 부위는 작용 항체로서의 역할을 할 수 있다. 따라서, 상기 이중 특이 항체는 CD137의 세포외 도메인에 특이적으로 결합하여 CD137를 활성화할 수 있다.Additionally, the extracellular domain binding site of CD137, which is the second domain of the bispecific antibody, can serve as an agonistic antibody. Therefore, the bispecific antibody can activate CD137 by specifically binding to the extracellular domain of CD137.
따라서, 본 발명에서 상기 이중 특이 항체는 종양 표면에 과발현된 암 항원에 특이적으로 결합하고, 본 발명의 상기 융합단백질의 CD137 세포외 도메인에 특이적으로 결합함으로써, 종양 세포 및 본 발명의 융합단백질이 발현된 면역세포를 결합시키는 세포 결합자(Cell engager)로서 이용될 수 있다. 상기 이중 특이 항체를 세포 결합자로 이용함으로써, 암을 표적화 하고, 암 세포 특이적으로 면역세포를 활성화시킴으로써 보다 안전하고 우수한 항암 활성을 나타낼 수 있다. Therefore, in the present invention, the bispecific antibody specifically binds to the cancer antigen overexpressed on the tumor surface and specifically binds to the CD137 extracellular domain of the fusion protein of the present invention, thereby inhibiting tumor cells and the fusion protein of the present invention. It can be used as a cell engager to bind the expressed immune cells. By using the bispecific antibody as a cell binder, it can target cancer and exhibit safer and superior anticancer activity by activating immune cells specifically for cancer cells.
본 발명의 일 실시예에서, 본 발명의 융합단백질이 발현된 T 세포와 항-BCMA/항-4-1BB 이중 특이 항체를 병용처리한 경우, 야생형 T 세포와 비교하여 세포독성이 증가하는 것을 확인할 수 있었다(도 17).In one embodiment of the present invention, when T cells expressing the fusion protein of the present invention are treated in combination with anti-BCMA/anti-4-1BB bispecific antibodies, it is confirmed that cytotoxicity increases compared to wild type T cells. (Figure 17).
본 명세서에서 사용하는 용어, "예방"은 상기 약학적 조성물의 투여에 의해 질환의 발생을 억제하거나 그의 발병을 지연시키는 모든 행위를 말한다. 상기 용어 "치료"는 상기 약학적 조성물의 투여에 의해 질환의 증세가 호전되거나 이롭게 변경하는 모든 행위를 말한다.As used herein, the term “prevention” refers to all actions that suppress or delay the onset of a disease by administering the pharmaceutical composition. The term “treatment” refers to any action that improves or beneficially changes the symptoms of a disease by administering the pharmaceutical composition.
본 발명의 약학 조성물에서 상기 융합단백질이 발현된 면역세포는 활성을 나타낼 수 있는 한, 용도, 제형, 배합 목적 등에 따라 임의의 양(유효량)으로 포함될 수 있다. 본 발명의 약학 조성물은 약 1×102 세포 내지 약 1×1016 세포, 약 1×102 세포 내지 약 1×1015 세포, 약 1×102 세포 내지 약 1×1014 세포 또는 약 1×102 세포 내지 약 1×1013 세포의 융합단백질이 발현된 면역세포를 포함할 수 있다. 바람직하게, 상기 약학 조성물은 약 2×102 세포 내지 약 2×1011 세포의 융합단백질이 발현된 면역세포를 포함할 수 있다.In the pharmaceutical composition of the present invention, immune cells expressing the fusion protein may be included in any amount (effective amount) depending on the use, formulation, purpose of formulation, etc., as long as they can show activity. The pharmaceutical composition of the present invention can be used to kill about 1×10 2 cells to about 1×10 16 cells, about 1×10 2 cells to about 1×10 15 cells, about 1×10 2 cells to about 1×10 14 cells, or about 1 cell. It may include immune cells expressing the fusion protein of ×10 2 cells to about 1 × 10 13 cells. Preferably, the pharmaceutical composition may include about 2×10 2 to about 2×10 11 cells of immune cells expressing the fusion protein.
여기서 "유효량"은 항노화 효과를 유도할 수 있는 유효성분의 양을 의미한다. 이러한 유효량은 당업자의 통상의 능력 범위 내에서 실험적으로 결정될 수 있다.Here, “effective amount” refers to the amount of active ingredient that can induce an anti-aging effect. Such effective amounts can be determined experimentally within the scope of the ordinary ability of those skilled in the art.
본 발명의 조성물은 동결되지 않은 채 사용되거나, 차후 사용을 위해 동결될 수 있다. 동결되어야 할 경우, 표준 냉동보존제(예를 들어 DMSO, 글리세롤, 폴리비닐피롤리돈, 폴리에틸렌 글리콜, 알부민, 덱스트란, 수크로스, 에틸렌 글리콜, i-d 에리스리톨, D-리비톨, D-만니톨, D-솔비톨, i-이노시톨, D-락토스, 콜린클로라이드, 에피라이프(Epilife®) 세포동결배지(Cascade biologics)가 동결 전 세포에 첨가될 수 있다.Compositions of the present invention can be used unfrozen or frozen for later use. If freezing is required, use standard cryopreservation agents (e.g. DMSO, glycerol, polyvinylpyrrolidone, polyethylene glycol, albumin, dextran, sucrose, ethylene glycol, id erythritol, D-ribitol, D-mannitol, D- Sorbitol, i-inositol, D-lactose, choline chloride, and Epilife ® cell freezing medium (Cascade biologics) can be added to the cells before freezing.
본 발명의 약학 조성물은 약학적으로 허용가능한 담체를 추가로 포함하여 제제화될 수 있다.The pharmaceutical composition of the present invention may be formulated to further include a pharmaceutically acceptable carrier.
본 명세서에서 사용하는 용어, "약학적으로 허용가능한 담체"란 생물체를 상당히 자극하지 않고 투여 성분의 생물학적 활성 및 특성을 저해하지 않는 담체 또는 희석제를 말한다. 본 발명의 약학 조성물은 세포치료제로 적용될 수 있으며, 세포 치료제로서 포함할 수 있는 약학적으로 허용 가능한 담체는 완충제, 보존제, 무통화제, 가용화제, 등장제, 안정화제, 기제, 부형제, 윤활제 등 당업계에 공지된 것이라면 제한 없이 사용할 수 있다. 본 발명의 약학 조성물은 각종 제형의 형태로 통용되는 기법에 따라 제조될 수 있다. As used herein, the term “pharmaceutically acceptable carrier” refers to a carrier or diluent that does not significantly irritate living organisms and does not inhibit the biological activity and properties of the administered ingredient. The pharmaceutical composition of the present invention can be applied as a cell therapy agent, and pharmaceutically acceptable carriers that can be included as a cell therapy agent include buffers, preservatives, analgesics, solubilizers, isotonic agents, stabilizers, bases, excipients, lubricants, etc. If it is known in the industry, it can be used without restrictions. The pharmaceutical composition of the present invention can be prepared according to commonly used techniques in the form of various dosage forms.
본 발명의 약학적 조성물은 약학적으로 유효한 양으로 투여할 수 있으며, 질병 부위로 이동을 유도할 수 있다면 어떠한 경로를 통해서든지 투여 가능하다. 여기서, "투여"란 적절한 방법으로 개체에게 소정의 물질을 도입하는 것을 의미한다. 경우에 따라서는 본 발명의 약학 조성물을 병변으로 향하게 하는 수단을 구비한 비히클에 로딩하는 방안을 고려할 수도 있다. 바람직하게 비경구 투여일 수 있다. 이때 비경구 투여로는 피하, 피내, 근육내, 점적, 정맥 내, 동맥 내, 관절 내 및 뇌척수액 내 투여 등을 포함할 수 있다.The pharmaceutical composition of the present invention can be administered in a pharmaceutically effective amount, and can be administered through any route as long as it can induce movement to the diseased area. Here, “administration” means introducing a predetermined substance into an individual by an appropriate method. In some cases, loading the pharmaceutical composition of the present invention into a vehicle equipped with a means to direct it to the lesion may be considered. Preferably, it may be parenteral administration. At this time, parenteral administration may include subcutaneous, intradermal, intramuscular, instillation, intravenous, intraarterial, intraarticular, and intracerebrospinal fluid administration.
본 발명의 약학 조성물의 바람직한 투여방식 및 제제는 주사제이다. 주사제는 생리식염액, 링겔액, Hank 용액 또는 멸균된 수용액 등의 수성용제, 올리브 오일 등의 식물유, 에틸올레인산 등의 고급 지방산 에스테르 및 에탄올, 벤질알코올, 프로필렌글리콜, 폴리에틸렌글리콜 또는 글리세린 등의 비수성용제 등을 이용하여 제조할 수 있고, 점막 투과를 위해, 통과할 배리어에 적합한 당업계에 공지된 비침투성제가 사용될 수 있으며, 변질방지를 위한 안정화제로 아스코르빈산, 아황산수소나트륨, BHA, 토코페롤, EDTA 등과, 유화제, pH 조절을 위한 완충제, 질산페닐수은, 치메로살, 염화벤잘코늄, 페놀, 크레솔, 벤질알코올 등의 미생물 발육을 저지하기 위한 보존제 등의 약학적 담체를 추가적으로 포함할 수 있다. 약제학적 조성물의 제제화와 관련하여서는 당업계에 공지되어 있으며, 구체적으로 문헌[Remington's Pharmaceutical Sciences(19th ed., 1995)] 등을 참조할 수 있다. 상기 문헌은 본 명세서의 일부로서 간주된다. The preferred administration method and formulation of the pharmaceutical composition of the present invention is injection. Injections include aqueous solvents such as physiological saline solution, Ringer's solution, Hank's solution, or sterilized aqueous solution, vegetable oils such as olive oil, higher fatty acid esters such as ethyl oleic acid, and non-aqueous solvents such as ethanol, benzyl alcohol, propylene glycol, polyethylene glycol, or glycerin. It can be manufactured using, etc., and for mucosal penetration, impermeable agents known in the art suitable for the barrier to pass through can be used, and as a stabilizer to prevent deterioration, ascorbic acid, sodium bisulfite, BHA, tocopherol, EDTA It may additionally contain pharmaceutical carriers such as emulsifiers, buffers for pH adjustment, and preservatives to inhibit microbial growth such as phenylmercuric nitrate, thimerosal, benzalkonium chloride, phenol, cresol, and benzyl alcohol. Regarding the formulation of pharmaceutical compositions, it is known in the art, and specifically, references can be made to the literature [Remington's Pharmaceutical Sciences (19th ed., 1995)]. The above documents are considered part of this specification.
본 명세서에서 사용하는 용어, "약학적으로 유효한 양"이란 의학적 치료에 적용 가능한 합리적인 수혜/위험 비율로 질환을 치료하기에 충분하며 부작용을 일으키지 않을 정도의 양을 의미하며, 유효 용량 수준은 환자의 성별, 연령, 체중, 건강상태, 질병의 종류, 중증도, 약물의 활성, 약물에 대한 민감도, 투여 방법, 투여 시간, 투여 경로 및 배출 비율, 치료 기간, 배합 또는 동시에 사용되는 약물을 포함한 요소 및 기타 의학 분야에 잘 알려진 요소에 따라 당업자에 의해 용이하게 결정될 수 있다.As used herein, the term “pharmaceutically effective amount” refers to an amount that is sufficient to treat a disease with a reasonable benefit/risk ratio applicable to medical treatment and does not cause side effects, and the effective dose level is the patient's Factors including gender, age, weight, health status, type of disease, severity, activity of drug, sensitivity to drug, method of administration, time of administration, route of administration and excretion rate, treatment period, drugs combined or used simultaneously, and other factors. It can be readily determined by a person skilled in the art according to factors well known in the medical field.
본 발명의 약학 조성물의 바람직한 투여량은 환자의 상태, 체중, 성별, 연령, 환자의 중증도, 투여 경로에 따라 세포 기준으로 1일 약 1×10 세포/kg 내지 약 1×1012 세포/kg, 약 1×10 세포/kg 내지 약 1×1011 세포/kg 또는 약 1×10 세포/kg 내지 약 1×1010 세포/kg일 수 있다. 다만, 투여량은 제제화 방법, 투여 방식, 환자의 연령, 체중, 성별, 병적 상태, 음식, 투여 시간, 투여 경로, 배설 속도 및 반응 감응성과 같은 요인들에 의해 다양하게 처방될 수 있고, 당업자라면 이러한 요인들을 고려하여 투여량을 적절히 조절할 수 있다. 투여 횟수는 1회 또는 임상적으로 용인 가능한 부작용의 범위 내에서 2회 이상이 가능하다. 본 발명의 약학 조성물은 1회 내지 3회/일, 1회 내지 3회/주 또는 1회 내지 10회/월로 투여될 수 있다. 예를 들어, 본 발명 약학 조성물은 1회 내지 9회/월, 1회 내지 8회/월, 1회 내지 7회/월, 1회 내지 6회/월, 1회 내지 5회/월, 1회 내지 4회/월, 1회 내지 3회/월 또는 1회 내지 2회/월 투여될 수 있다. 투여 부위에 대해서도 1개소 또는 2개소 이상에 투여할 수 있다. 인간 이외의 동물에 대해서도, kg당 또는 개체당 인간과 동일한 투여량으로 하거나, 또는 예를 들면 목적의 동물과 인간과의 기관(심장 등)의 용적비(예를 들면, 평균값) 등으로 상기의 투여량을 환산한 양을 투여할 수 있다. 이러한 투여량은 어떠한 측면으로든 본 발명의 범위를 제한하는 것으로 해석되어서는 아니 된다. The preferred dosage of the pharmaceutical composition of the present invention is about 1×10 cells/kg to about 1×10 12 cells/kg per day, based on cells, depending on the patient's condition, weight, gender, age, patient's severity, and route of administration. It may be about 1×10 cells/kg to about 1×10 11 cells/kg or about 1×10 cells/kg to about 1×10 10 cells/kg. However, the dosage may be prescribed in various ways depending on factors such as formulation method, administration method, patient's age, weight, gender, pathological condition, food, administration time, administration route, excretion rate, and reaction sensitivity, and those skilled in the art will Taking these factors into consideration, the dosage can be adjusted appropriately. The number of administrations can be once or twice or more within the range of clinically acceptable side effects. The pharmaceutical composition of the present invention may be administered 1 to 3 times/day, 1 to 3 times/week, or 1 to 10 times/month. For example, the pharmaceutical composition of the present invention can be used 1 to 9 times/month, 1 to 8 times/month, 1 to 7 times/month, 1 to 6 times/month, 1 to 5 times/month, 1 It may be administered once to four times/month, once to three times/month, or once to twice per month. Regarding the administration site, it can be administered at one or two or more locations. For animals other than humans, the dosage per kg or per individual is the same as that for humans, or, for example, the above-mentioned administration is based on the volume ratio (e.g., average value) of the organs (e.g., heart, etc.) between the target animal and human. The converted dose can be administered. These dosages should not be construed as limiting the scope of the invention in any respect.
본 명세서에서 사용한 용어, "개체"는 본 발명의 조성물이 적용(처방)될 수 있는 질환이 발병하였거나 발병할 수 있는 대상을 의미하며, 인간을 포함한 쥐, 생쥐, 가축 등의 포유동물일 수 있다. 바람직하게는, 인간일 수 있으나 이에 제한되지 않는다.The term "individual" used herein refers to a subject who has or may develop a disease to which the composition of the present invention can be applied (prescribed), and may be a mammal such as a rat, mouse, or livestock, including humans. . Preferably, it may be a human, but is not limited thereto.
본 발명 약학 조성물은 필요에 따라, 본 발명 약학 조성물은 상기 질환에 대하여 예방 또는 치료 효과를 나타내는 공지된 물질을 추가적으로 포함할 수 있다.If necessary, the pharmaceutical composition of the present invention may additionally contain known substances that exhibit a preventive or therapeutic effect on the above diseases.
본 발명 약학 조성물은 상기 융합단백질이 발현된 면역세포의 활성을 증가시키는 물질을 더 포함할 수 있다. 본 발명 약학 조성물은 종래에 알려져 있는 상기 질환의 예방 또는 치료 물질과 병용하여 투여될 수 있다. 이때, 상기 융합단백질이 발현된 면역세포, 및 상기 질환의 예방 또는 치료 효과를 나타내는 물질 및/또는 상기 면역세포의 활성을 증가시키는 물질은 동시 또는 순차적으로 투여할 수 있다.The pharmaceutical composition of the present invention may further include a substance that increases the activity of immune cells expressing the fusion protein. The pharmaceutical composition of the present invention can be administered in combination with conventionally known substances for preventing or treating the above diseases. At this time, the immune cells expressing the fusion protein, substances showing a preventive or therapeutic effect for the disease, and/or substances that increase the activity of the immune cells can be administered simultaneously or sequentially.
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포 또는 이를 포함하는 약학 조성물의 암 예방 또는 치료용 용도를 제공한다. Another aspect of the present invention provides the use of immune cells expressing the fusion protein or a pharmaceutical composition containing the same for preventing or treating cancer.
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포 또는 이를 포함하는 약학 조성물을 개체에 투여하는 단계를 포함하는 암 예방 또는 치료용 방법을 제공한다. 상기 융합단백질이 발현된 면역세포, 약학 조성물, 암, 예방, 치료, 개체 및 투여는 상술한 바와 동일하다.Another aspect of the present invention provides a method for preventing or treating cancer, comprising administering to an individual an immune cell expressing the fusion protein or a pharmaceutical composition containing the same. Immune cells expressing the fusion protein, pharmaceutical composition, cancer, prevention, treatment, subject, and administration are the same as described above.
융합단백질이 발현된 면역세포 및 이중 특이 항체의 병용Combination of immune cells expressing fusion proteins and dual-specific antibodies
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포 및 이중 특이 항체를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물을 제공한다.Another aspect of the present invention provides a pharmaceutical composition for preventing or treating cancer comprising an immune cell expressing the fusion protein and a dual-specific antibody as active ingredients.
본 발명의 또 다른 측면은 상기 융합단백질이 발현된 면역세포 및 이중 특이 항체를 포함하는 암 예방 또는 치료용 키트를 제공한다.Another aspect of the present invention provides a kit for preventing or treating cancer, including an immune cell expressing the fusion protein and a dual-specific antibody.
상기 약학 조성물 및 키트에 포함된 상기 융합단백질이 발현된 면역세포 및 이중 특이 항체는 하나의 제형에 포함될 수 있고, 병용 투여될 수도 있다. 이때, 상기 융합단백질이 발현된 면역세포 및 이중 특이 항체는 동시에 투여되거나, 순차적으로 투여될 수 있다. Immune cells and dual-specific antibodies expressing the fusion protein contained in the pharmaceutical composition and kit may be included in one formulation and may be administered in combination. At this time, the immune cells expressing the fusion protein and the dual-specific antibody may be administered simultaneously or sequentially.
상기 융합단백질이 발현된 면역세포, 이중 특이 항체, 암, 예방 및 치료는 상술한 바와 동일하다.Immune cells expressing the fusion protein, bispecific antibodies, cancer, prevention and treatment are the same as described above.
CD137 세포외 도메인, 이의 변이체 또는 이들의 단편CD137 extracellular domain, variants thereof or fragments thereof
본 발명의 또 다른 측면은 CD137 세포외 도메인 변이체 또는 이의 단편을 제공한다. 이때, 상기 변이체는 CRD1, CRD2, CRD3, CRD4 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나에 변이를 포함할 수 있다. Another aspect of the invention provides CD137 extracellular domain variants or fragments thereof. At this time, the variant may include a mutation in any one selected from the group consisting of CRD1, CRD2, CRD3, CRD4, and combinations thereof.
구체적으로, 상기 변이체는 CRD2 및/또는 CRD3에 변이를 포함하는 것일 수 있다. 상기 CRD2 및 CRD3의 변이는 상술한 바와 동일하다. Specifically, the variant may include mutations in CRD2 and/or CRD3. The mutations of CRD2 and CRD3 are the same as described above.
보다 구체적으로 CD137 세포외 도메인 변이체 또는 이의 단편은 서열번호 16, 서열번호 17 및 서열번호 27로 이루어진 군에서 선택되는 어느 하나의 아미노산 서열을 포함하거나 이들로 이루어진 것일 수 있다.More specifically, the CD137 extracellular domain variant or fragment thereof may include or consist of any one amino acid sequence selected from the group consisting of SEQ ID NO: 16, SEQ ID NO: 17, and SEQ ID NO: 27.
이하, 본 발명의 이해를 돕기 위하여 바람직한 실시예를 제시한다. 그러나 하기의 실시예는 본 발명을 보다 쉽게 이해하기 위하여 제공되는 것일 뿐, 하기 실시예에 의해 본 발명의 내용이 한정되는 것은 아니다.Below, preferred embodiments are presented to aid understanding of the present invention. However, the following examples are provided only to make the present invention easier to understand, and the content of the present invention is not limited by the following examples.
준비예 1. 사용 항체 정보Preparation example 1. Antibody information used
하기 실험예에 사용된 항체 정보는 하기 표 1에 나타내었다.Antibody information used in the following experimental examples is shown in Table 1 below.
AntibodiesAntibodies SourceSource Cat No.Cat No.
CD3 Monoclonal Antibody (UCHT1), FITCCD3 Monoclonal Antibody (UCHT1), FITC eBiosciencebioscience 11-0038-4211-0038-42
CD56 (NCAM) Monoclonal Antibody, PECD56 (NCAM) Monoclonal Antibody, PE eBiosciencebioscience 12-0567-4212-0567-42
CD56 (NCAM) Monoclonal Antibody, FITCCD56 (NCAM) Monoclonal Antibody, FITC eBiosciencebioscience 11-0566-4211-0566-42
CD16 Monoclonal Antibody, APCCD16 Monoclonal Antibody, APC eBiosciencebioscience 17-0168-4217-0168-42
Brilliant Violet 605™ anti-human CD226 (DNAM-1) AntibodyBrilliant Violet 605™ anti-human CD226 (DNAM-1) Antibody BiolegendBiolegend 338324338324
PE anti-human CD335 (NKp46) AntibodyPE anti-human CD335 (NKp46) Antibody BiolegendBiolegend 331908331908
APC anti-human CD336 (NKp44) AntibodyAPC anti-human CD336 (NKp44) Antibody BiolegendBiolegend 325110325110
Brilliant Violet 421™ anti-human CD314 (NKG2D) AntibodyBrilliant Violet 421™ anti-human CD314 (NKG2D) Antibody BiolegendBiolegend 320822320822
BV711 Mouse Anti-Human CD337 (NKp30)BV711 Mouse Anti-Human CD337 (NKp30) BD BioscienceBD Bioscience 563383563383
UrelumabUrelumab MedChemExpressMedChemExpress HY-99055HY-99055
Biotin Antibody, PE, REAfinityBiotin Antibody, PE, REAfinity MiltenyiBiotecMiltenyiBiotec 130-110-951130-110-951
PE Mouse Anti-Human CD137PE Mouse Anti-Human CD137 BD PharmingenBD Pharmingen 555956555956
실험 방법 1. 유세포 측정기를 이용한 세포 분석 Experimental Method 1. Cell analysis using flow cytometry
각각의 FITC, PE, APC, BV421, BV605 그리고 BV711 파장의 형광 표지 항체는 NK 세포를 염색하는데 사용되었다. 구체적으로, 1.0 x 105 NK 세포는 수확하여 Flow Cytometry Staining Buffer(eBioscience, 00-4222-26)로 한 차례 세척하였다. 30분 동안 차광하여 4℃ 냉장고에서 반응하고, Flow Cytometry를 이용하여 분석하였다. Fluorescently labeled antibodies of each FITC, PE, APC, BV421, BV605, and BV711 wavelength were used to stain NK cells. Specifically, 1.0 x 10 5 NK cells were harvested and washed once with Flow Cytometry Staining Buffer (eBioscience, 00-4222-26). Reaction was performed in a refrigerator at 4°C, shielded from light for 30 minutes, and analyzed using flow cytometry.
실시예 1. 형질전환된 PB-NK 세포의 제조Example 1. Preparation of transformed PB-NK cells
실시예 1.1. 유전자 컨스트럭션 제조Example 1.1. Genetic construction manufacturing
4-1BB와 CD3ζ 도메인을 포함하고 있는 이식 유전자를 EcoRI과 XhoI을 이용하여 pVAX1 벡터에 삽입하였다. 해당 벡터는 mRNA 생산을 위해 XbaI(NEB, R0145S)을 이용하여 선형 주형 가닥이 되도록 하였다. 이후, mMESSAGE mMACHINETM T7 ULTRA Transcription Kit(Invitrogen, AM1345)를 이용하여 in vitro transcription을 진행하였다. 자세한 내용은 제조사의 지시서를 참고하였다. 구체적인 융합단백질의 구조는 도 1a 및 도 1b에 도시하였다. 또한, CD137의 세포외 도메인 및 CD3ζ의 세포내도메인을 포함한 융합단백질을 통칭하여 BBz라 명명하였다. 구체적인 아미노산 서열 및 핵산 서열을 표 2에 나타내었다.The transgene containing the 4-1BB and CD3ζ domains was inserted into the pVAX1 vector using EcoRI and XhoI. The vector was converted into a linear template strand using XbaI (NEB, R0145S) for mRNA production. Afterwards, in vitro transcription was performed using the mMESSAGE mMACHINE TM T7 ULTRA Transcription Kit (Invitrogen, AM1345). For detailed information, refer to the manufacturer's instructions. The structure of the specific fusion protein is shown in Figures 1A and 1B. In addition, the fusion protein containing the extracellular domain of CD137 and the intracellular domain of CD3ζ was collectively named BBz. Specific amino acid sequences and nucleic acid sequences are shown in Table 2.
서열번호sequence number 명칭designation 서열order
1One 신호서열(SP)Signal sequence (SP) MGNSCYNIVATLLLVLNFERTRS MGNSCYNIVATLLVLNFERTRS
22 CRD1 CRD1 LQDPCSNCPAGTFCDNNRNQICLQDPCSNCPAGTFCDNNRNQIC
3 3 CRD2CRD2 SPCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAECSPCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAEC
66 CRD3 CRD3 DC TPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK
77 CRD4 CRD4 DCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG
1616 4-1BB 세포외 도메인4-1BB extracellular domain LQDPCSNCPAGTFCDNNRNQICSPCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAECDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGLQDPCSNCPAGTFCDNNRNQICSPCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAECDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG
1111 linker linker PSPADLSPGASSVTPPAPAREPGHSPQPSPADLSPGASSVTPPAPAREPGHSPQ
88 막관통도메인(TM)Transmembrane domain (TM) IISFFLALTSTALLFLLFFLTLRFSVV IISFFLALTSTALLFLLFFLTLRFSVV
99 CD137의 세포내도메인(ICD)Intracellular domain (ICD) of CD137 KRGRKKLLYIFK QPFMRPVQTTQEEDGCSCRFPEEEEGGCELKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL
1010 CD3z의 세포내도메인(ICD)Intracellular domain (ICD) of CD3z RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR
1818 BBz의 세포내도메인(ICD)Intracellular domain (ICD) of BBz MGNSCYNIVATLLLVLNFERTRSLQDPCSNCPAGTFCDNNRNQICSPCPPNSFSSAGGQITCDRCRQCKGVFRTRKECSSTSNAECDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCGPSPADLSPGASSVTPPAPAREPGHSPQIISFFLALTSTALLFLLFFLTLRFSVVKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPRMGNSCYNIVATLLVLNFERTRSLQDPCSNCPAGTFCDNNRNQICSPCPPNSFSSAGGQITCDRCRQCKGVFRTRTRKECSSTSNAECDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCGPSPADLSPGASSVTPPAPAREPGHSPQIISFFLALTSTALLFLLFFLTLRFSVKRGR KKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCELRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR
3030 nucleotide coding amino acid of BBznucleotide coding amino acid of BBz atgggcaaca gctgctacaa catcgtggcc acactgctgc tggtgctgaa cttcgagaga accagaagcc tgcaggaccc ctgcagcaat tgtcctgccg gcaccttctg cgacaacaac cggaatcaga tctgcagccc ctgtcctcct aacagcttta gctctgccgg cggacagaga acctgcgaca tctgcagaca gtgcaagggc cggttccgga ccagaaaaga gtgcagcagc acctccaacg ccgagtgcga ttgcacacct ggcttccatt gtctcggagc cggctgttct atgtgcgagc aggattgcaa gcagggccaa gagctgacca agaagggctg caaggattgc tgcttcggca cctttaacga ccagaagcgg ggcatctgtc ggccctggac aaattgctct ctggacggca agagcgtgct ggtcaacggc accaaagaac gggatgtcgt gtgcggacct tctccagccg atttgtctcc tggcgcctct agcgttacac ctcctgctcc agctagagag cctggccact ctcctcagat catctcattc tttctggccc tgacctctac agccctgctg tttctgctgt tcttcctgac actgcggttc agcgtggtca agagaggccg gaagaagctg ctgtacatct tcaagcagcc tttcatgcgg cccgtgcaga ccacacaaga ggaagatggc tgctcctgca gattccccga ggaagaagaa ggcggctgcg aacttagagt gaagttcagc aggagcgcag acgcccccgc gtaccagcag ggccagaacc agctctataa cgagctcaat ctaggacgaa gagaggagta cgatgttttg gacaagagac gtggccggga ccctgagatg gggggaaagc cgagaaggaa gaaccctcag gaaggcctgt acaatgaact gcagaaagat aagatggcgg aggcctacag tgagattggg atgaaaggcg agcgccggag gggcaagggg cacgatggcc tttaccaggg tctcagtaca gccaccaagg acacctacga cgcccttcac atgcaggccc tgccccctcg ctaaatgggcaaca gctgctacaa catcgtggcc acactgctgc tggtgctgaa cttcgagaga accagaagcc tgcaggaccc ctgcagcaat tgtcctgccg gcaccttctg cgacaacaac cggaatcaga tctgcagccc ctgtcctcct aacagcttta gctctgccgg cggacagaga acctgcgaca t ctgcagaca gtgcaagggc cggttccgga ccagaaaaga gtgcagcagc acctccaacg ccgagtgcga ttgcacacct ggcttccatt gtctcggagc cggctgttct atgtgcgagc aggattgcaa gcagggccaa gagctgacca agaagggctg caaggattgc tgcttcggca cc agaagcgg ggcatctgtc ggccctggac aaattgctct ctggacggca agagcgtgct ggtcaacggc accaaagaac gggatgtcgt gtgcggacct tctccagccg atttgtctcc tggcgcctct agcgttacac ctcctgctcc agctagagag cctggccact ctcctcagat catctcattc tttctggccc tgacctctac agccctgctg tttctgctgt tcttcctgac actgcggttc agcgtggtca agagaggccg gaagaagctg ctgtacatct tcaagttcatg cgg cccgtgcaga ccacacaaga ggaagatggc tgctcctgca gattccccga ggaagaagaa ggcggctgcg aacttagagt gaagttcagc aggagcgcag acgcccccgc gtaccagcag ggccagaacc agctctataa cgagctcaat ctaggacgaa gagaggagta cgatgttttg gacaagagac g tggccggga ccctgagatg gggggaaagc cgagaaggaa gaaccctcag gaaggcctgt acaatgaact gcagaaagat aagatggcgg aggcctacag tgagattggg atgaaaggcg agcgccggag gggcaagggg cacgatggcc tttaccaggg tctcagtaca gccaccaagg acacctacga cgcccttcac atgcaggccc tgccccctcg ctaa
3434 CD28의 세포내도메인(ICD)Intracellular domain (ICD) of CD28 RSKRSRLLHSD YMNMTPRRPGPTRKHYQPYAPPRDFAAYRSRSKRSRLLHSDYMNMTPRRPGPTRKHYQPYAPPRDFAAYRS
3535 OX40의 세포내도메인(ICD)Intracellular domain (ICD) of OX40 ALYLLRRDQRLPPDAHKPPGGGSFRTPIQEEQADAHSTLAKI ALYLLRRDQRLPPDAHKPPGGGSFRTPIQEEQADAHSTLAKI
실시예 1.2. PB-NK 세포의 분리 및 증식Example 1.2. Isolation and proliferation of PB-NK cells
테라베스트내 SOP(Standard operating procedure)를 참고하여 공정을 진행하였다. 구체적으로, PBMC(Peripheral Blood Mononuclear Cell)는 ficoll-Paque PLUS(Cytiva, 17144002)를 이용하여 분리하였다. 순수한 NK 세포를 얻기 위해, Miltenyi Biotec사의 MACS(Magnetically activated cell sorting) 시스템을 이용하였다. 분리한 NK 세포는 X-ray 조사한 GE-K562 세포와 1:2 비율로 공배양하였다. 0일에는 ALyS505N-0(CSTI, 1020P10)에 4% human AB serum(GeminiBio, 100-512), 1% L-Glutamine(ThermoFisher, 25030149), 500 IU/mL IL-2(Hileukin), 3 ng/mL IL-12(BD Bioscience, 554613), 50 ng/mL IL-15(R&D systems, 247ILB-500CF), 30 ng/mL IL-18(MBL, B003-2)이 포함되어 있는 배지를 사용하여 NK 세포를 활성화하였다. 이후 IL-2 외의 사이토카인이 제거된 배지에서 NK 세포를 배양하였다.The process was carried out by referring to Therabest's SOP (Standard Operating Procedure). Specifically, PBMC (Peripheral Blood Mononuclear Cell) was isolated using ficoll-Paque PLUS (Cytiva, 17144002). To obtain pure NK cells, Miltenyi Biotec's MACS (Magnetically activated cell sorting) system was used. The isolated NK cells were co-cultured with X-ray irradiated GE-K562 cells in a 1:2 ratio. On day 0, ALyS505N-0 (CSTI, 1020P10), 4% human AB serum (GeminiBio, 100-512), 1% L-Glutamine (ThermoFisher, 25030149), 500 IU/mL IL-2 (Hileukin), 3 ng/ NK using medium containing mL IL-12 (BD Bioscience, 554613), 50 ng/mL IL-15 (R&D systems, 247ILB-500CF), and 30 ng/mL IL-18 (MBL, B003-2). Cells were activated. Afterwards, NK cells were cultured in medium from which cytokines other than IL-2 were removed.
실시예 1.3. PB-NK 세포의 형질전환Example 1.3. Transformation of PB-NK cells
NK 세포는 Neon® Transfection 시스템(ThermoFisher)과 MaxCyte의 GTx를 이용하여 일렉트로포레이션(electroporation)을 진행하였다. 먼저, Buffer T(Invitrogen, MPK10096)로 NK 세포를 5.0x106 cells/100 μL로 현탁하였다. 세포현탁액에 mRNA를 2.5 μg 넣어주고, 1850 V, 20 ms, 1 purse로 하여 electroporation 하였다. GTx는 제조사의 지시사항에 따르며, 코드는‘Expanded NK-5’로 설정하여 일렉트로포레이션을 진행하였다. 그 후, Miltenyi Biotec사의 anti-PE MicroBeads(MiltenyiBiotec, 130-048-801)를 이용하여 4-1BB를 발현하는 세포주를 분리하였다. NK cells were electroporated using the Neon ® Transfection system (ThermoFisher) and MaxCyte's GTx. First, NK cells were suspended at 5.0x10 6 cells/100 μL with Buffer T (Invitrogen, MPK10096). 2.5 μg of mRNA was added to the cell suspension, and electroporation was performed at 1850 V, 20 ms, 1 purse. GTx followed the manufacturer's instructions, and electroporation was performed with the code set to 'Expanded NK-5'. Afterwards, a cell line expressing 4-1BB was isolated using anti-PE MicroBeads from Miltenyi Biotec (MiltenyiBiotec, 130-048-801).
실시예 2. cBBR 면역세포 제조Example 2. Preparation of cBBR immune cells
실시예 2.1. 렌티바이러스 벡터 제조Example 2.1. Lentiviral vector preparation
실시예 2.1.1 유전자 컨스트럭션 제조Example 2.1.1 Genetic Construction Preparation
신호서열(서열번호 1), CD137(4-1BB)의 세포외 도메인(서열번호 15) 또는 이의 변이체(서열번호 16, 17 또는 27), 링커(서열번호 11), 막관통 도메인(서열번호 8), 공자극 도메인(서열번호 9) 및 1차 신호전달 도메인(서열번호 10)을 포함하는 융합단백질(서열번호 18, 19, 20 또는 31)을 암호화하는 폴리뉴클레오티드(서열번호 12, 13, 14 또는 30)를 합성하여 pCDH-EF1-MCS-IRES-Puro 벡터(SBI, CD532A-2)에 적재하였다. 상기 융합단백질의 아미노산 서열을 표 3 내지 표 6에 나타내었다. 본 발명에 의한 상기 융합단백질을 "cBBR(Chimeric 4-1BB receptor)"로 명명하였다.Signal sequence (SEQ ID NO: 1), extracellular domain of CD137 (4-1BB) (SEQ ID NO: 15) or a variant thereof (SEQ ID NO: 16, 17 or 27), linker (SEQ ID NO: 11), transmembrane domain (SEQ ID NO: 8 ), a polynucleotide (SEQ ID NO: 12, 13, 14) encoding a fusion protein (SEQ ID NO: 18, 19, 20 or 31) containing a co-stimulatory domain (SEQ ID NO: 9) and a primary signaling domain (SEQ ID NO: 10) or 30) was synthesized and loaded into the pCDH-EF1-MCS-IRES-Puro vector (SBI, CD532A-2). The amino acid sequences of the fusion proteins are shown in Tables 3 to 6. The fusion protein according to the present invention was named “cBBR (Chimeric 4-1BB receptor)”.
융합단백질Fusion protein 도메인domain 아미노산 서열amino acid sequence 서열번호 sequence number
cBBR
(서열번호 18)
cBBR
(SEQ ID NO: 18)
Signal peptide Signal peptide MGNSCYNIVATLLLVLNFERTRSMGNSCYNIVATLLVLNFERTRS 1 One
CRD1CRD1
LQDPCSNCPAGTFCDNNRNQICSLQDPCSNCPAGTFCDNNRNQICS 2 2
CRD2CRD2
PCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAECPCPPNSFSSAGGQRTCDICRQCKGVFRTRKECSSTSNAEC 33
CRD3 CRD3 DC TPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 66
CRD4 CRD4 DCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 77
linker linker PSPADLSPGASSVTPPAPAREPGHSPQPSPADLSPGASSVTPPAPAREPGHSPQ 1111
Transmembrane (TM)Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV IISFFLALTSTALLFLLFFLTLRFSVV 88
co-stimulatory domain co-stimulatory domain KRGRKKLLYIFK QPFMRPVQTTQEEDGCSCRFPEEEEGGCELKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 99
CD3z signal domainCD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR 1010
융합단백질Fusion protein 도메인domain 아미노산 서열amino acid sequence 서열번호 sequence number
cBBR_I64R
(서열번호 19)
cBBR_I64R
(SEQ ID NO: 19)
Signal peptide Signal peptide MGNSCYNIVATLLLVLNFERTRSMGNSCYNIVATLLVLNFERTRS 1 One
CRD1CRD1
LQDPCSNCPAGTFCDNNRNQICSLQDPCSNCPAGTFCDNNRNQICS 22
CRD2 mutantCRD2 mutant PCPPNSFSSAGGQRTCD R CRQCKGVFRTRKECSSTSNAEC PCPPNSFSSAGGQRTCD R CRQCKGVFRTRKECSSTSNAEC 44
CRD3 CRD3 DC TPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 66
CRD4 CRD4 DCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 77
linker linker PSPADLSPGASSVTPPAPAREPGHSPQPSPADLSPGASSVTPPAPAREPGHSPQ 1111
Transmembrane (TM)Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV IISFFLALTSTALLFLLFFLTLRFSVV 88
co-stimulatory domain co-stimulatory domain KRGRKKLLYIFK QPFMRPVQTTQEEDGCSCRFPEEEEGGCELKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 99
CD3z signal domainCD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR 1010
융합단백질Fusion protein 도메인domain 아미노산 서열amino acid sequence 서열번호 sequence number
cBBR_V71R
(서열번호 20)
cBBR_V71R
(SEQ ID NO: 20)
Signal peptide Signal peptide MGNSCYNIVATLLLVLNFERTRSMGNSCYNIVATLLVLNFERTRS 1 One
CRD1CRD1
LQDPCSNCPAGTFCDNNRNQICSLQDPCSNCPAGTFCDNNRNQICS 22
CRD2 mutantCRD2 mutant PCPPNSFSSAGGQRTCDICRQCKG R FRTRKECSSTSNAEC PCPPNSFSSAGGQRTCDICRQCKG R FRTRKECSSTSNAEC 55
CRD3 CRD3 DC TPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 66
CRD4 CRD4 DCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 77
linker linker PSPADLSPGASSVTPPAPAREPGHSPQPSPADLSPGASSVTPPAPAREPGHSPQ 1111
Transmembrane (TM)Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV IISFFLALTSTALLFLLFFLTLRFSVV 88
co-stimulatory domain co-stimulatory domain KRGRKKLLYIFK QPFMRPVQTTQEEDGCSCRFPEEEEGGCELKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 99
CD3z signal domainCD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR 1010
융합단백질Fusion protein 도메인domain 아미노산 서열amino acid sequence 서열번호 sequence number
cBBR_
I64R/V71R
(서열번호 31)
cBBR_
I64R/V71R
(SEQ ID NO: 31)
Signal peptide Signal peptide MGNSCYNIVATLLLVLNFERTRSMGNSCYNIVATLLVLNFERTRS 1 One
CRD1CRD1
LQDPCSNCPAGTFCDNNRNQICSLQDPCSNCPAGTFCDNNRNQICS 22
CRD2 mutantCRD2 mutant PCPPNSFSSAGGQRTCD R CRQCKG R FRTRKECSSTSNAECPCPPNSFSSAGGQRTCD R CRQCKG R FRTRKECSSTSNAEC 2626
CRD3 CRD3 DC TPGFHCLGAGCSMCEQDCKQGQELTKKGCKDCTPGFHCLGAGCSMCEQDCKQGQELTKKGCK 66
CRD4 CRD4 DCCFGTFNDQK RGICRPWTNCSLDGKSVLVNGTKERDVVCGDCCFGTFNDQKRGICRPWTNCSLDGKSVLVNGTKERDVVCG 77
linker linker PSPADLSPGASSVTPPAPAREPGHSPQPSPADLSPGASSVTPPAPAREPGHSPQ 1111
Transmembrane (TM)Transmembrane (TM) IISFFLALTSTALLFLLFFLTLRFSVV IISFFLALTSTALLFLLFFLTLRFSVV 88
co-stimulatory domain co-stimulatory domain KRGRKKLLYIFK QPFMRPVQTTQEEDGCSCRFPEEEEGGCELKRGRKKLLYIFKQPFMRPVQTTQEEDGCSCRFPEEEEGGCEL 99
CD3z signal domainCD3z signal domain RVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRR GKGHDGLYQGLSTATKDTYDALHMQALPPRRVKFSRSADAPAYQQGQNQLYNELNLGRREEYDVLDKRRGRDPEMGGKPRRKNPQEGLYNELQKDKMAEAYSEIGMKGERRRGKGHDGLYQGLSTATKDTYDALHMQALPPR 1010
4-1BBL 발현 벡터는 서열번호 25의 폴리뉴클레오티드를 합성하여 pCDH-EF1-MCS-IRES-Puro 벡터(SBI, CD532A-2)에 적재하여 제작하였다.The 4-1BBL expression vector was constructed by synthesizing the polynucleotide of SEQ ID NO: 25 and loading it into the pCDH-EF1-MCS-IRES-Puro vector (SBI, CD532A-2).
실시예 2.2. 렌티바이러스 생산Example 2.2. Lentivirus production
VSV-G, Rev, 및 cBBR 또는 이의 변이체를 암호화하는 폴리뉴클레오티드(서열번호 12, 13, 14 또는 30)가 적재된 pCDH-EF1-MCS-IRES-Puro 벡터 또는 4-1BBL을 암호화하는 폴리뉴클레오티드(서열번호 25)가 적재된 pCDH-EF1-MCS-IRES-Puro 벡터를 Lipofectamine 2000(Thermo Fisher, 11668019)을 이용하여 Lenti-X 293T 세포(Clontech, 632180)에 각각 형질도입하였다. 형질도입 후 72시간 뒤, 각각의 상층액을 수확하여 Lenti-X Concentrator(Clontech, 631232)와 1:3 비율로 혼합하여 1시간 이상 냉장 보관하였다. 상기 각각의 혼합액을 4℃에서 1,500g로 원심분리한 뒤, 펠렛을 수거하여 각각 cBBR 또는 이의 변이체를 발현시킬 수 있는 바이러스 입자 및 4-1BBL 유전자를 발현하는 바이러스 입자로 사용하였다. pCDH-EF1-MCS-IRES-Puro vector loaded with polynucleotides encoding VSV-G, Rev, and cBBR or variants thereof (SEQ ID NO: 12, 13, 14 or 30) or a polynucleotide encoding 4-1BBL ( The pCDH-EF1-MCS-IRES-Puro vector loaded with SEQ ID NO: 25) was transduced into Lenti-X 293T cells (Clontech, 632180) using Lipofectamine 2000 (Thermo Fisher, 11668019). 72 hours after transfection, each supernatant was harvested, mixed with Lenti-X Concentrator (Clontech, 631232) in a 1:3 ratio, and stored in the refrigerator for more than 1 hour. Each of the above mixed solutions was centrifuged at 1,500g at 4°C, and the pellet was collected and used as virus particles capable of expressing cBBR or a variant thereof and virus particles expressing the 4-1BBL gene, respectively.
실시예 2.3. cBBR T 세포 제작Example 2.3. cBBR T cell production
실시예 2.3.1. cBBR이 발현되는 세포주 제작Example 2.3.1. Production of cell lines expressing cBBR
K562 세포 및 Nalm6 세포는 ECACC(European collection of authenticated cell cultures)에서 구매하였고, Jurkat T 세포 및 Ramos 세포는 한국세포주은행에서 구매하였다. 각종 cBBR Jurkat T 세포 및 4-1BBL Nalm6 세포는 상기 실시예 2.2의 방법으로 제조된 렌티바이러스를 이용하여 형질도입하였다. 모든 세포는 10% FBS(Gibco, 10100147)가 포함되어 있는 RPMI-1640(Hyclone, SH30027.01) 배지를 사용하여 배양하였다.K562 cells and Nalm6 cells were purchased from the European collection of authenticated cell cultures (ECACC), and Jurkat T cells and Ramos cells were purchased from the Korean Cell Line Bank. Various cBBR Jurkat T cells and 4-1BBL Nalm6 cells were transduced using the lentivirus prepared in Example 2.2 above. All cells were cultured using RPMI-1640 (Hyclone, SH30027.01) medium containing 10% FBS (Gibco, 10100147).
실시예 2.3.2. cBBR이 발현되는 혈액 유래 T 세포 제작Example 2.3.2. Production of blood-derived T cells expressing cBBR
먼저 혈액 내 T 세포를 분리하여 위하여, Ficoll Paque PLUS(Cytiva, 17144002)를 이용하여 혈액 내 PBMC(Peripheral Blood Mononuclear Cell)를 분리하였다. PBMC 내 T 세포는 CD4+ T cell isolation kit, human(Miltenyi biotec, 130-096-533) 및 CD8+ T cell isolation kit, human(Miltenyi biotec, 130-096-495)를 이용하여 분리하였다. 상기와 같이 분리된 T 세포에 T Cell TransAct, human(Miltenyi biotec, 130-111-160)을 첨가하여 활성화시켰다. 활성화된 T 세포는 10% FBS 및 300 U/mL Proleukin IL-2(NOVARTIS)가 포함된 RPMI-1640 배지를 이용하여 배양하였다. 활성화를 유도한 다음 날, 상기 실시예 2.2의 방법으로 제조된 렌티바이러스를 이용하여 T 세포에 cBBR 또는 이의 변이체(I64R, V71R)를 형질도입 하고, 14일까지 배양하였다. 상기와 같이 제작된 세포를 이하 "cBBR T 세포"로 기재하였다. First, to isolate T cells in the blood, PBMC (Peripheral Blood Mononuclear Cell) in the blood was isolated using Ficoll Paque PLUS (Cytiva, 17144002). T cells in PBMC were isolated using CD4+ T cell isolation kit, human (Miltenyi biotec, 130-096-533) and CD8+ T cell isolation kit, human (Miltenyi biotec, 130-096-495). T cells isolated as above were activated by adding T Cell TransAct, human (Miltenyi biotec, 130-111-160). Activated T cells were cultured using RPMI-1640 medium containing 10% FBS and 300 U/mL Proleukin IL-2 (NOVARTIS). The day after inducing activation, cBBR or its variants (I64R, V71R) were transduced into T cells using the lentivirus prepared in Example 2.2, and cultured for up to 14 days. The cells prepared as above are hereinafter referred to as “cBBR T cells.”
실시예 2.4. cBBR NK 세포 제작Example 2.4. cBBR NK cell production
실시예 2.4.1. NK 세포의 준비Example 2.4.1. Preparation of NK cells
Lymphoblast의 환자로부터 유래한 NK 세포 NK92MI(ATCC, CRL-2408)는 12.5% FBS(fetal bovine serum, Gibco, 10100147), 12.5% 말혈청(horse serum, Gibco, 16050122), 1% 페니실린/스트렙토마이신(Gibco, 15140-122), 0.2 mM 이노시톨(DAEJUNG,5008-4105), 0.1 mM 2-머캅토에탄올(Gibco, 21985023), 및 0.02 mM 엽산(Sigma Aldrich, F8758)가 포함되어 있는 alpha MEM(Cytiva, SH30265.01) 배지를 이용하여 CO2 배양기에서 배양하였다. Lymphoblast patient-derived NK cells NK92MI (ATCC, CRL-2408) were supplemented with 12.5% FBS (fetal bovine serum, Gibco, 10100147), 12.5% horse serum (Gibco, 16050122), and 1% penicillin/streptomycin ( Gibco, 15140-122), alpha MEM (Cytiva, containing 0.2mM inositol (DAEJUNG, 5008-4105), 0.1mM 2-mercaptoethanol (Gibco, 21985023), and 0.02mM folic acid (Sigma Aldrich, F8758). SH30265.01) was cultured in a CO 2 incubator using medium.
실시예 2.4.2. cBBR이 발현되는 NK 세포주 제작Example 2.4.2. Construction of NK cell lines expressing cBBR
실시예 2.3.의 방법과 동일하게 실시예 2.4.1.에서 수득한 NK 세포를 이용하여 cBBR이 발현되는 NK 세포주를 제작하였다. 상기와 같이 제작된 세포를 이하 "cBBR NK92MI 세포" 또는 "4-1BB 발현 NK92MI 세포"로 기재하였다. An NK cell line expressing cBBR was created using the NK cells obtained in Example 2.4.1 in the same manner as in Example 2.3. The cells prepared as above are hereinafter referred to as “cBBR NK92MI cells” or “4-1BB expressing NK92MI cells.”
실험예 1. 형질전환된 NK 세포의 활성 변화 확인Experimental Example 1. Confirmation of changes in activity of transformed NK cells
실험예 1.1. 우렐루맙에 의한 BBz를 발현하는 NK 세포의 활성 변화 확인Experimental Example 1.1. Confirmation of changes in the activity of NK cells expressing BBz by urelumab
4-1BB를 발현하고 있는 NK92MI 세포가 단클론항체인 우렐루맙에 의해 활성화 되는지 알아보기 위해 세포주를 개발하였다(도 2). 적절한 우렐루맙의 농도를 확인하기 위해 적정(titration) 평가를 수행하였다(도 3 및 도 4). 그 후, NK 세포에서 생성된 IFN-γ를 확인하기 위해, Human IFN-gamma Quantikine ELISA kit(R&D systems, DIF50C)를 이용하여 NK 세포배양액에서 IFN-γ의 농도를 측정하였다. 그 결과, NT NK92MI와 다르게 BBzNK92MI에서 우렐루맙(MedChemExpress, HY-P99055)을 10 μg/mL을 처리하면 IFN-γ양이 9배 이상 증가하는 것을 확인하였다(도 5).A cell line was developed to determine whether NK92MI cells expressing 4-1BB are activated by the monoclonal antibody urelumab (Figure 2). Titration evaluation was performed to confirm the appropriate concentration of urelumab (Figures 3 and 4). Then, in order to confirm the IFN-γ produced by NK cells, the concentration of IFN-γ was measured in the NK cell culture medium using the Human IFN-gamma Quantikine ELISA kit (R&D systems, DIF50C). As a result, unlike NT NK92MI, it was confirmed that when 10 μg/mL of urelumab (MedChemExpress, HY-P99055) was treated in BBzNK92MI, the amount of IFN-γ increased by more than 9-fold (Figure 5).
실험예 1.2. 우렐루맙에 의한 PB-NK 세포의 활성 변화 확인Experimental Example 1.2. Confirmation of changes in PB-NK cell activity caused by urelumab
세포주와 마찬가지로 peripheral blood(PB) NK 세포에서 동일한 결과를 보이는지, 4-1BB의 세포내도메인(ICD)이 없는 구조가 더 강력한 효능을 보이는지 알아보기 위해 클로닝을 진행하였다(도 1a). IVT(In vitro transcription)을 하여 생산한 mRNA를 이용하여 PB-NK에 일렉트로포레이션(electroporation, 이하 EP라 함)을 하였다. EP 직후 우렐루맙이 코팅된 플레이트에서 PB-NK를 24시간 배양한 후, 생성된 IFN-γ을 확인하였다. BBz와 tBBzNK세포에서 모두 약 3~4배 이상 IFN-γ가 증가하였다. 그러나, 우렐루맙을 soluble로 처리한 실험군에서는 변화가 없는 것을 확인하였다(도 8).As with cell lines, cloning was performed to determine whether the same results were observed in peripheral blood (PB) NK cells and whether the structure without the intracellular domain (ICD) of 4-1BB showed stronger efficacy (Figure 1a). Electroporation (hereinafter referred to as EP) was performed on PB-NK using mRNA produced through IVT ( in vitro transcription). Immediately after EP, PB-NK was cultured on a plate coated with urelumab for 24 hours, and the produced IFN-γ was confirmed. In both BBz and tBBzNK cells, IFN-γ increased by approximately 3 to 4 times. However, it was confirmed that there was no change in the experimental group treated with soluble urelumab (Figure 8).
이러한 결과로부터 우렐루맙 단독에 의해서는 PB-NK를 활성화시키는 효과가 낮았으나, 우렐루맙의 한쪽이 고정될 경우에, 우렐루맙에 의한 PB-NK 활성화 효과가 커지는 것을 확인하였다. 즉, 우렐루맙과 같은 agonist 4-1BB antibody가 CD137에 결합한다고 하여 활성되는 것이 아니라, immunological synapse가 형성되어야만 우렐루맙이 CD137이 과별현된 NK를 활성화시킬 수 있다는 것을 알 수 있다. 또한, 흥미롭게도 BBz는 4시간 이후 점차 발현이 감소하고 있음에도 불구하고, tBBzNK 세포에 비해 1.3배 많은 IFN-γ을 발현하고 있었다.From these results, it was confirmed that the effect of urelumab alone on activating PB-NK was low, but that when one side of urelumab was fixed, the effect of urelumab on activating PB-NK increased. In other words, it can be seen that agonist 4-1BB antibody, such as urelumab, is not activated by binding to CD137, but that urelumab can activate NK with overexpressed CD137 only when immunological synapse is formed. Also, interestingly, BBz expressed 1.3 times more IFN-γ than tBBzNK cells, although the expression gradually decreased after 4 hours.
실험예 1.3. 4-1BB 발현 세포의 분리를 위한 도메인 매핑Experimental Example 1.3. Domain mapping for isolation of 4-1BB expressing cells
iPSC의 유전자 재조합 효율은 상대적으로 낮은 편이다. 이러한 단점을 보완하기 위해서는 농축은 필수 과정이다. MiltenyiBiotec사의 GMP 4B4-1 클론의 항체와 결합하는 4-1BB의 최소 도메인을 찾기 위해 클로닝을 진행하였다. 4B4-1(PE Mouse Anti-Human CD137 (BD Biosciences, 555956))은 4-1BB의 CRD(Cystein-Rich Domain)2, 우렐루맙은 CRD1에 결합한다고 보고되어 있어, 이를 근거로 클로닝을 진행하였다(도 1b). 하지만 참고 문헌과 다르게, 12BBz 뿐만 아니라 1BBz K562가 4B4-1과 결합하는 것을 확인하였다(도 9).The genetic recombination efficiency of iPSCs is relatively low. To compensate for these shortcomings, enrichment is an essential process. Cloning was performed to find the minimal domain of 4-1BB that binds to the antibody of MiltenyiBiotec's GMP 4B4-1 clone. 4B4-1 (PE Mouse Anti-Human CD137 (BD Biosciences, 555956)) is reported to bind to CRD (Cystein-Rich Domain) 2 of 4-1BB, and urelumumab is reported to bind to CRD1, so cloning was performed based on this ( Figure 1b). However, unlike the reference, it was confirmed that not only 12BBz but also 1BBz K562 binds to 4B4-1 (FIG. 9).
실험예 2. 유세포 측정기를 이용한 형질도입 세포 분석Experimental Example 2. Analysis of transduced cells using flow cytometry
상기 실시예 2의 방법으로 제작된 cBBR 또는 이의 변이체, 또는 4-1BBL을 발현하는 세포주, 및 cBBR 또는 이의 변이체를 발현하는 혈액 유래의 T 세포(cBBR T 세포)의 세포 표면 단백질 발현을 FITC, PE 및 APC 파장의 형광 표지된 항체를 이용하여 확인하였다.Cell surface protein expression of the cell line expressing cBBR or a variant thereof, or 4-1BBL, produced by the method of Example 2, and blood-derived T cells (cBBR T cells) expressing cBBR or a variant thereof was measured using FITC, PE. and was confirmed using a fluorescently labeled antibody of APC wavelength.
구체적으로, 각각의 cBBR Jurkat T 세포, 4-1BBL Nalm6 세포 및 cBBR T 세포(1.0×105 세포)를 Flow Cytometry Staining Buffer(eBioscience, 00-4222-26)로 1회 세척한 뒤, 각각의 항체를 처리하여 30분 동안 차광하여 4℃에서 반응시켰다. 반응이 끝난 뒤, 염색된 세포는 유세포 측정기를 이용하여 분석하였다. 이때, 사용한 항체는 표 7에 나타내었다.Specifically, each cBBR Jurkat T cell, 4-1BBL Nalm6 cell, and cBBR T cell (1.0×10 5 cells) were washed once with Flow Cytometry Staining Buffer (eBioscience, 00-4222-26), and then incubated with each antibody. was treated and reacted at 4°C, shielded from light for 30 minutes. After the reaction was completed, the stained cells were analyzed using flow cytometry. At this time, the antibodies used are shown in Table 7.
항체antibody 제조사manufacturing company 카탈로그 번호catalog number
CD3 Monoclonal Antibody (OKT3), FITCCD3 Monoclonal Antibody (OKT3), FITC eBiosciencebioscience 11-0037-4211-0037-42
CD8a Monoclonal Antibody (SK1), APCCD8a Monoclonal Antibody (SK1), APC eBiosciencebioscience 17-0087-4217-0087-42
PE Mouse Anti-Human CD137PE Mouse Anti-Human CD137 BD BiosciencesBD Biosciences 555956555956
PE Mouse Anti-Human CD137 LigandPE Mouse Anti-Human CD137 Ligand BD BiosciencesBD Biosciences 559446559446
Goat Anti-Human IgG H&L (DyLight®488)Goat Anti-Human IgG H&L (DyLight®488) AbcamAbcam ab96907ab96907
PE anti-human CD269 (BCMA)PE anti-human CD269 (BCMA) BiolegendBiolegend 357504357504
PE Mouse Anti-Human CD107aPE Mouse Anti-Human CD107a BD BiosciencesBD Biosciences 555801555801
그 결과, 도 11 및 도 12에 나타낸 바와 같이, cBBR Jurkat T 세포 및 cBBR T 세포에서 아미노산 치환과 관계없이 cBBR의 대조군(cBBR을 과발현시키지 않은 세포)과 비교하여 높은 CD137 발현을 관찰할 수 있었다. 또한, 4-1BBL를 과발현시킨 Nalm6 세포(4-1BBL Nalm6 세포)의 경우에도 4-1BBL를 과발현시키지 않은 세포와 비교하여 4-1BBL의 발현율이 높은 것을 확인할 수 있었다.As a result, as shown in Figures 11 and 12, high CD137 expression was observed in cBBR Jurkat T cells and cBBR T cells compared to the cBBR control group (cells that did not overexpress cBBR) regardless of amino acid substitution. In addition, in the case of Nalm6 cells that overexpressed 4-1BBL (4-1BBL Nalm6 cells), it was confirmed that the expression rate of 4-1BBL was high compared to cells that did not overexpress 4-1BBL.
실험예 3. cBBR 및 4-1BBL의 결합능 확인Experimental Example 3. Confirmation of binding ability of cBBR and 4-1BBL
상기 실시예 2의 방법으로 제작된 cBBR JurKat T 세포 및 4-1BBL Nalm6 세포주를 이용하여 cBBR 또는 이의 변이체, 및 4-1BBL의 결합능을 확인하였다.The binding ability of cBBR or its variant, and 4-1BBL was confirmed using cBBR JurKat T cells and 4-1BBL Nalm6 cell line produced by the method of Example 2.
구체적으로, cBBR Jurkat T 세포는 1 μM CellTracker Deep Red Dye(Invitrogen, C34565)로 염색하였고, 4-1BBL Nalm6 세포주는 5 μM CellTrace CFSE Cell Proliferation Kit(Invitrogen, C34554)로 염색하였다. 상기 염색된 두 종류의 세포를 30분 동안 공배양한 뒤, 4% 파라포름알데히드 용액(바이오세상, PC2031-100-00)을 첨가하여 고정하였다. 상기 고정된 세포는 유세포 측정기를 이용하여 분석하였다(% of CFSE+ 7-AAD+ 세포).Specifically, cBBR Jurkat T cells were stained with 1 μM CellTracker Deep Red Dye (Invitrogen, C34565), and 4-1BBL Nalm6 cell line was stained with 5 μM CellTrace CFSE Cell Proliferation Kit (Invitrogen, C34554). The two types of stained cells were co-cultured for 30 minutes and then fixed by adding 4% paraformaldehyde solution (Biosesang, PC2031-100-00). The fixed cells were analyzed using flow cytometry (% of CFSE+ 7-AAD+ cells).
그 결과, 도 13에 나타낸 바와 같이, T 세포에 과발현시킨 cBBR에 아미노산 치환(변이체)에 의하여 Nalm6 세포에 과발현된 4-1BBL과 결합력이 감소하는 것을 확인하였다.As a result, as shown in Figure 13, it was confirmed that the binding affinity of cBBR overexpressed in T cells to 4-1BBL overexpressed in Nalm6 cells was reduced due to amino acid substitution (mutant).
실험예 4. cBBR T 세포의 암세포 살상능 확인Experimental Example 4. Confirmation of cancer cell killing ability of cBBR T cells
상기 실시예 2의 방법으로 제작된 cBBR T 세포의 암세포에 대한 살상능을 확인하여 위하여, cBBR T 세포 및 4-1BBL Nalm6 세포를 공배양하여 암세포에 대한 cBBR T 세포의 세포 독성을 측정하였다.To confirm the killing ability of cBBR T cells produced by the method of Example 2 against cancer cells, cBBR T cells and 4-1BBL Nalm6 cells were co-cultured to measure the cytotoxicity of cBBR T cells against cancer cells.
구체적으로, cBBR T 세포의 독성은 Cytotoxicity Assay Kit(Abcam, ab270780)을 이용하여 진행하였다. 4-1BBL Nalm6 세포는 상기 키트 내 포함된 CFSE 시약으로 염색하고, cBBR T 세포와 각각 0.5:1, 1:1 및 2:1의 비율로 혼합하여 24시간 동안 공배양하였다. 이때, 4-1BBL이 형질도입되지 않은 Nalm6을 대조군으로 사용하였다. 공배양 후, 상기 세포를 7-AAD로 염색하여 유세포 분석을 통해 사멸 세포를 확인하였다(% of CFSE+ 7-AAD+ 세포).Specifically, the toxicity of cBBR T cells was performed using the Cytotoxicity Assay Kit (Abcam, ab270780). 4-1BBL Nalm6 cells were stained with the CFSE reagent included in the kit, mixed with cBBR T cells at a ratio of 0.5:1, 1:1, and 2:1, respectively, and co-cultured for 24 hours. At this time, Nalm6, in which 4-1BBL was not transduced, was used as a control. After co-culture, the cells were stained with 7-AAD and dead cells were confirmed through flow cytometry (% of CFSE+ 7-AAD+ cells).
그 결과, 도 14에 나타낸 바와 같이, cBBR T 세포(변이체 포함)는 4-1BBL Nalm6 세포에 대하여 4-1BB 세포외 도메인 및 4-1BBL의 결합으로 인하여 세포 독성을 나타내었다. 특히, cBBR의 일부 아미노산이 치환(변이체)된 cBBR T 세포(I64R, V71R, I64R/V71R)에서는 세포 독성이 감소하였다. 상기 결과는 cBBR 변이체의 CD137(4-1BB) 세포외 도메인과 4-1BBL 간의 결합력의 감소한 것에서 기인한 것으로 사료된다.As a result, as shown in Figure 14, cBBR T cells (including mutants) exhibited cytotoxicity against 4-1BBL Nalm6 cells due to the binding of the 4-1BB extracellular domain and 4-1BBL. In particular, cytotoxicity was reduced in cBBR T cells (I64R, V71R, I64R/V71R) in which some amino acids of cBBR were substituted (mutants). The above results are believed to be due to a decrease in binding force between the CD137 (4-1BB) extracellular domain of the cBBR variant and 4-1BBL.
실험예 5. 우렐루맙에 의한 cBBR T 세포의 활성화 확인Experimental Example 5. Confirmation of activation of cBBR T cells by urelumab
실험예 5.1. 유세포 분석을 통한 우렐루맙에에 의한 cBBR T 세포 활성화 확인Experimental Example 5.1. Confirmation of cBBR T cell activation by urelumab through flow cytometry
96웰 플레이트를 우렐루맙(MedChemExpress, HY-P99055)(1 ㎍/mL, 10 ㎍/mL)으로 코팅한 뒤, 상기 실시예 2의 방법으로 제작된 cBBR T 세포(I64R, V71R, I64R/V71R)를 5×105 cell/mL의 농도로 접종한 뒤 24시간 동안 반응시켰다. 반응이 끝난 뒤, 상기 세포를 항-CD8a 항체(APC) 및 항-CD107a 항체(PE)로 각각 염색하고, 유세포 측정기를 이용하여 분석하였다. 그 결과, 도 15에 나타낸 바와 같이, 아미노산이 치환(변이체)된 cBBR T 세포의 4-1BBL에 대한 결합력은 감소하였다. 하지만, 항-CD137 항체(아고니스트)인 우렐루맙(Urelumab)에 의한 활성은 감소하지 않은 것을 확인할 수 있었다.A 96-well plate was coated with urelumab (MedChemExpress, HY-P99055) (1 μg/mL, 10 μg/mL), and then cBBR T cells (I64R, V71R, I64R/V71R) prepared by the method of Example 2 above were added. were inoculated at a concentration of 5×10 5 cell/mL and reacted for 24 hours. After the reaction was completed, the cells were stained with anti-CD8a antibody (APC) and anti-CD107a antibody (PE), respectively, and analyzed using flow cytometry. As a result, as shown in Figure 15, the binding affinity of cBBR T cells with amino acid substitutions (mutants) to 4-1BBL decreased. However, it was confirmed that the activity caused by Urelumab, an anti-CD137 antibody (agonist), was not reduced.
실험예 5.2. 우렐루맙에 의한 cBBR T 세포의 IFN-γ 분비능 확인Experimental Example 5.2. Confirmation of the IFN-γ secretion ability of cBBR T cells by urelumab
실시예 2의 방법으로 제작된 cBBR T 세포(I64R, V71R, I64R/V71R)에 1 ㎍/mL 또는 10 ㎍/mL의 농도가 되도록 우렐루맙을 처리한 뒤, 24시간 반응시켜 활성화를 유도하였다. 반응 후, 활성화된 cBBR T 세포로부터 배양배지 내로 분비된 IFN-γ의 농도Human IFN-gamma Quantikine ELISA Kit(R&D systems, SIF50C)를 이용하여 측정하였다. 그 결과, 도 16에 나타낸 바와 같이, 아미노산이 치환(변이체)된 cBBR T 세포의 4-1BBL에 대한 결합력은 감소하였다. 하지만, 항-CD137 항체(아고니스트)인 우렐루맙(Urelumab)에 의한 활성은 감소하지 않은 것을 확인할 수 있었다.cBBR T cells (I64R, V71R, I64R/V71R) produced by the method of Example 2 were treated with urelumab at a concentration of 1 μg/mL or 10 μg/mL, and then incubated for 24 hours to induce activation. After the reaction, the concentration of IFN-γ secreted from activated cBBR T cells into the culture medium was measured using the Human IFN-gamma Quantikine ELISA Kit (R&D systems, SIF50C). As a result, as shown in Figure 16, the binding affinity of cBBR T cells with amino acid substitutions (mutants) to 4-1BBL decreased. However, it was confirmed that the activity caused by Urelumab, an anti-CD137 antibody (agonist), was not reduced.
실시예 3. 이중 특이 항체 제작Example 3. Production of bispecific antibody
본 발명에 사용된 이중 특이 항체는 Genescript Probio사에 의뢰하여 제작하였다. 상기 이중 특이 항체의 아미노산 서열을 하기 표 8에 나타내었다. 이때, 상기 이중 특이 항체는 항-BCMA 항체이며, C 말단에 4-1BB에 결합하는 scFv가 결합된 형태이다.The bispecific antibody used in the present invention was produced by Genescript Probio. The amino acid sequence of the bispecific antibody is shown in Table 8 below. At this time, the bispecific antibody is an anti-BCMA antibody, and an scFv that binds to 4-1BB is bound to the C terminus.
명칭designation 아미노산 서열amino acid sequence 서열번호 sequence number
중쇄영역Heavy chain region MGWSCIILFLVATATGVHSEVQLLESGGGLVQPGGSLRLSCAASGFTFSDYGLSWVRQAPGKGLEWVSLIDSSGSSTFYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKEHGLFDSWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKGSGSGSGSGSGSGSGSGSQSVLTQPPSASGTPGQRVTISCSGSSSNIGNNYVTWYQQLPGTAPKLLIYADSHRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCATWDYSLSGYVFGCGTKLTVLGGGGSGGGGSGGGGSGGGGSEVQLLESGGGLVQPGGSLRLSCAASGFTFSSYDMSWVRQAPGKCLEWVSWISYSGGSIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDAQRNSMREFDYWGQGTLVTVSSMGWSCIILFLVATATGVHSEVQLLESGGGLVQPGGSLRLSCAASGFTFSDYGLSWVRQAPGKGLEWVSLIDSSGSSTFYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCAKEHGLFDSWGQGTLVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSS SLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKT KPREEQYASTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSREEMTKNQVSLTCLVKGFYPSDIAVEWESNGQP ENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKGSGSGSGSGSGSGSGSQSVLTQPPSASGTPGQRVTISCSG SSSNIGNNYVTWYQQLPGTAPKLLIYADSHRPSGVPDRFSGSKSGTSASLAISGLRSEDEADYYCATWDYSLSGYVFGCGTKLTVLGGGGSGGGGSGGGGS GGSEVQLLESGGGLVQPGGSLRLSCAASGFTFSSYDMSWVRQAPGKCLEWVSWISYSGGSIYYADSVKGRFTISRDNSKNTLYLQMNSLRAEDTAVYYCARDAQRNSMREFDYWGQGTLVTVSS 3232
경쇄영역light chain region MGWSCIILFLVATATGVHSEIVLTQSPGTLSLSPGERATLSCKASQDIDDAINWYQQKPGQAPRLLIYDASLRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQSLRTPITFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECMGWSCIILFLVATATGVHSEIVLTQSPGTLSLSPGERATLSCKASQDIDDAINWYQQKPGQAPRLLIYDASLRATGIPDRFSGSGSGTDFTLTISRLEPEDFAVYYCQQSLRTPITFGQGTKLEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKA DYEKHKVYACEVTHQGLSSPVTKSFNRGEC 3333
실험예 6. 이중 특이 항체에 의한 cBBR T 세포의 활성Experimental Example 6. Activation of cBBR T cells by bispecific antibodies
실험예 6.1. cBBR T 세포의 cBBR 및 이중 특이 항체의 결합능 확인Experimental Example 6.1. Confirmation of binding ability of cBBR T cells with cBBR and bispecific antibodies
상기 실시예 2의 방법으로 제작된 cBBR T 세포(I64R, V71R) 및 실시예 3의 방법으로 제작된 이중 특이 항체(BiTE)의 결합능을 확인하였다. The binding ability of cBBR T cells (I64R, V71R) produced by the method of Example 2 and the bispecific antibody (BiTE) produced by the method of Example 3 was confirmed.
구체적으로, 각각의 cBBR T 세포(Control, I64R, V71R), Ramos 세포(1.0×105 세포)를 Flow cytometry staining buffer(eBioscience, 00-4222-26)로 1회 세척한 뒤, 상기 실시예 3의 방법으로 제작한 이중 특이 항체를 처리하고 4℃에서 30분 동안 차광하여 반응시켰다. 반응이 끝난 뒤, 염색된 세포는 유세포 측정기를 이용하여 분석하였다.Specifically, each cBBR T cell (Control, I64R, V71R) and Ramos cell (1.0 × 10 5 cells) were washed once with Flow cytometry staining buffer (eBioscience, 00-4222-26), and then washed in Example 3 above. The bispecific antibody prepared by the method was treated and reacted at 4°C for 30 minutes, shielded from light. After the reaction was completed, the stained cells were analyzed using flow cytometry.
그 결과, 도 17에 나타낸 바와 같이, 이중 특이 항체의 CD137 결합 부위(항-CD137 Fab 부위) 및 cBBR T 세포의 CD137 세포외 도메인이 결합하는 것을 확인할 수 있었다. 또한, 이중 특이 항체의 BCMA 결합 부위(항-BCMA Fab 부위)와 Ramos 세포의 BCMA 단백질이 결합하는 것을 확인할 수 있다.As a result, as shown in Figure 17, it was confirmed that the CD137 binding site (anti-CD137 Fab site) of the bispecific antibody and the CD137 extracellular domain of cBBR T cells bound. In addition, it can be confirmed that the BCMA binding site (anti-BCMA Fab site) of the bispecific antibody binds to the BCMA protein of Ramos cells.
실험예 6.2. 이중 헝체에 의한 cBBR T 세포의 암세포 살상능 확인Experimental Example 6.2. Confirmation of cancer cell killing ability of cBBR T cells by double binding
이중 특이 항체(BiTE)에 의해 활성화가 유도된 cBBR T 세포의 암세포에 대한 살상능을 확인하기 위하여, cBBR T 세포(I64R, V71R)에 이중 특이 항체를 처리한 뒤, 실험예 3의 방법으로 암세포에 대한 세포 독성을 측정하였다. 이때, 이중 특이 항체는 1 또는 10 ㎍/mL의 농도로 24시간 처리하였다.In order to confirm the killing ability of cBBR T cells whose activation was induced by a bispecific antibody (BiTE) against cancer cells, cBBR T cells (I64R, V71R) were treated with a bispecific antibody, and then cancer cells were killed using the method of Experimental Example 3. Cytotoxicity was measured. At this time, the bispecific antibody was treated at a concentration of 1 or 10 μg/mL for 24 hours.
그 결과, 도 18에 나타낸 바와 같이, 아미노산이 치환(변이체)된 cBBR T 세포(I64R, V71R)와 Ramos 세포를 공배양한 후 항-BCMA/항-4-1BB 이중 특이 항체를 처리한 경우, 이중 특이 항체의 처리 농도에 따라 세포 독성이 증가하였다. 상기 결과를 통하여, 이중 특이 항체에 의해 cBBR T 세포(Effector cell)와 Ramos 세포(Target cell) 사이에 면역학적 시냅스가 형성된다는 것을 확인할 수 있었다.As a result, as shown in Figure 18, when cBBR T cells (I64R, V71R) with amino acid substitutions (mutants) and Ramos cells were co-cultured and then treated with anti-BCMA/anti-4-1BB bispecific antibody, Cytotoxicity increased depending on the treatment concentration of the bispecific antibody. Through the above results, it was confirmed that an immunological synapse was formed between cBBR T cells (Effector cells) and Ramos cells (Target cells) by the bispecific antibody.
실험예 7. 형질전환된 T 세포의 활성 확인Experimental Example 7. Confirmation of activity of transformed T cells
형질전환된 T 세포를 투여하기 하루 전에 NSG 마우스에 3x106개의 Ramos cell(ATCC, CRL-1596)을 복강내 투여하였다(IP Injection). 형질전환된 T 세포를 투여하기 전에, 루시페린(Perkin elmer, cat.122799) 3 mg을 복강내 투여한 후 IVIS® 형광장비를 이용하여 Ramos cell의 형광값을 측정하였다. 그 후, 1x107개의 cBBR T cell과 AB-101 이중항체 200 ug을 복강내 투여하였다. 형질전환된 T 세포 투여 후 4일 후에, 루시페린 3 mg을 복강내 투여한 후 IVIS® 형광장비를 이용하여 Ramos cell의 형광값을 측정하였다. CleVue 분석프로그램을 사용하여 측정된 형광값을 분석하였다. 그 결과를 도 19에 나타내었다.One day before administering the transfected T cells, 3x10 6 Ramos cells (ATCC, CRL-1596) were intraperitoneally administered to NSG mice (IP Injection). Before administering the transformed T cells, 3 mg of luciferin (Perkin elmer, cat.122799) was administered intraperitoneally and the fluorescence value of Ramos cells was measured using IVIS ® fluorescence equipment. Afterwards, 1x10 7 cBBR T cells and 200 ug of AB-101 bispecific antibody were administered intraperitoneally. Four days after administration of the transformed T cells, 3 mg of luciferin was administered intraperitoneally and the fluorescence value of Ramos cells was measured using an IVIS ® fluorescence equipment. The measured fluorescence values were analyzed using the CleVue analysis program. The results are shown in Figure 19.

Claims (41)

  1. 하기 (i) 내지 (iii)을 포함하는 융합단백질:Fusion protein comprising the following (i) to (iii):
    (i) CD137(4-1BB) 세포외 도메인 또는 이의 단편; (i) CD137 (4-1BB) extracellular domain or fragment thereof;
    (ii) 막관통 도메인(Transmembrane domain); 및(ii) Transmembrane domain; and
    (iii) 1차 신호전달 도메인.(iii) Primary signaling domain.
  2. 제1항에 있어서,According to paragraph 1,
    상기 CD137 세포외 도메인 또는 이의 단편은 The CD137 extracellular domain or fragment thereof is
    서열번호 2의 아미노산 서열을 포함하는 CRD1(Cystein-rich pseudo repeat 1); 서열번호 3의 아미노산 서열을 포함하는 CRD2; 서열번호 6의 아미노산 서열을 포함하는 CRD3; 서열번호 7의 아미노산 서열을 포함하는 CRD4; 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나를 포함하는 것인, 융합단백질.CRD1 (Cystein-rich pseudo repeat 1) containing the amino acid sequence of SEQ ID NO: 2; CRD2 comprising the amino acid sequence of SEQ ID NO: 3; CRD3 comprising the amino acid sequence of SEQ ID NO: 6; CRD4 comprising the amino acid sequence of SEQ ID NO: 7; And a fusion protein comprising any one selected from the group consisting of combinations thereof.
  3. 제2항에 있어서,According to paragraph 2,
    상기 CD137 세포외 도메인은 CRD1, CRD2, CRD3 및 CRD4를 포함하는 것인, 융합단백질.The CD137 extracellular domain includes CRD1, CRD2, CRD3, and CRD4.
  4. 제2항에 있어서,According to paragraph 2,
    상기 CD137 세포외 도메인은 변이를 추가로 포함하는 것인, 융합단백질.A fusion protein in which the CD137 extracellular domain further includes mutations.
  5. 제4항에 있어서,According to clause 4,
    상기 변이는 CRD2, CRD3 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나에 포함되는 것인, 융합단백질.The fusion protein is one in which the mutation is selected from the group consisting of CRD2, CRD3, and combinations thereof.
  6. 제5항에 있어서,According to clause 5,
    상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산이 치환된 것인, 융합단백질.The mutation of CRD2 is a substitution of any one amino acid selected from the group consisting of P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26, and combinations thereof in the amino acid sequence of SEQ ID NO: 3. It's a fusion protein.
  7. 제5항에 있어서,According to clause 5,
    상기 CRD3의 변이는 서열번호 6의 아미노산 서열에서 S14, M15, C16 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나의 아미노산이 치환된 것인, 융합단백질.The mutation of CRD3 is a fusion protein in which any one amino acid selected from the group consisting of S14, M15, C16, and combinations thereof is substituted in the amino acid sequence of SEQ ID NO: 6.
  8. 제6항에 있어서,According to clause 6,
    상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 I18R, V25R 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산으로 치환된 것인, 융합단백질.The mutation of CRD2 is a fusion protein in which any one amino acid selected from the group consisting of I18R, V25R, and combinations thereof is substituted in the amino acid sequence of SEQ ID NO: 3.
  9. 제8항에 있어서,According to clause 8,
    상기 CRD2의 변이는 서열번호 4, 서열번호 5 및 서열번호 26으로 이루어진 군에서 선택되는 어느 하나의 아미노산 서열을 포함하는 것인, 융합단백질.The mutation of CRD2 is a fusion protein comprising any one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 26.
  10. 제1항에 있어서, According to paragraph 1,
    상기 막관통 도메인이 T 세포 수용체(TCR), CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, CD137, CD152 및 CD154로 이루어진 군에서 선택되는 어느 하나인 것인, 융합단백질.The transmembrane domain is T cell receptor (TCR), CD3δ, CD3ε, CD3γ, CD3ζ, CD4, CD5, CD8α, CD9, CD16, CD22, CD27, CD28, CD33, CD37, CD45, CD64, CD80, CD86, CD134, A fusion protein selected from the group consisting of CD137, CD152, and CD154.
  11. 제10항에 있어서,According to clause 10,
    상기 막관통 도메인이 CD137로부터 유래된 것인, 융합단백질.A fusion protein in which the transmembrane domain is derived from CD137.
  12. 제11항에 있어서,According to clause 11,
    상기 막관통 도메인이 서열번호 8의 아미노산 서열을 포함하는 것인, 융합단백질.A fusion protein in which the transmembrane domain includes the amino acid sequence of SEQ ID NO: 8.
  13. 제1항에 있어서,According to paragraph 1,
    상기 세포내 신호전달 도메인이 공자극 도메인 및 활성화 도메인을 포함하는 것인, 융합단백질.A fusion protein, wherein the intracellular signaling domain includes a co-stimulatory domain and an activation domain.
  14. 제13항에 있어서, According to clause 13,
    상시 공자극 도메인이 OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1(CD11a/CD18), ICOS(CD278) 및 CD137로 이루어진 군에서 선택되는 어느 하나인 것인, 융합단백질.A fusion protein, wherein the constant costimulatory domain is any one selected from the group consisting of OX40, CD2, CD27, CD28, CDS, ICAM-1, LFA-1 (CD11a/CD18), ICOS (CD278), and CD137.
  15. 제14항에 있어서, According to clause 14,
    상기 공자극 도메인이 CD137로부터 유래된 것인, 융합단백질.A fusion protein in which the costimulatory domain is derived from CD137.
  16. 제15항에 있어서, According to clause 15,
    상기 공자극 도메인이 서열번호 9로 표시되는 아미노산 서열로 이루어진 것인, 융합단백질.A fusion protein in which the co-stimulatory domain consists of the amino acid sequence represented by SEQ ID NO: 9.
  17. 제13항에 있어서, According to clause 13,
    상기 1차 신호전달 도메인이 FcRγ, FcRβ, CD3γ, CD3δ, CD3ε, CD3ζ, CD22, CD79a, CD79b 및 CD66d로 이루어진 군에서 선택되는 어느 하나인 것인, 융합단백질. A fusion protein, wherein the primary signaling domain is any one selected from the group consisting of FcRγ, FcRβ, CD3γ, CD3δ, CD3ε, CD3ζ, CD22, CD79a, CD79b and CD66d.
  18. 제17항에 있어서, According to clause 17,
    상기 1차 신호전달 도메인이 CD3ζ로부터 유래된 것인, 융합단백질.A fusion protein in which the primary signaling domain is derived from CD3ζ.
  19. 제18항에 있어서,According to clause 18,
    상기 1차 신호전달 도메인이 서열번호 10의 아미노산 서열을 포함하는 것인, 융합단백질.A fusion protein wherein the primary signaling domain includes the amino acid sequence of SEQ ID NO: 10.
  20. 제1항에 있어서,According to paragraph 1,
    (i) CD137 세포외 도메인 또는 이의 단편; 및 (ii) 막관통 도메인 사이에는 링커를 추가로 포함하는 것인, 융합단백질. (i) CD137 extracellular domain or fragment thereof; and (ii) a fusion protein further comprising a linker between the transmembrane domains.
  21. 제1항에 있어서,According to paragraph 1,
    상기 융합단백질은 하기 구조식 (I)을 포함하는 것인, 융합단백질:The fusion protein contains the following structural formula (I):
    N'-A-(L1)n-TM-(L2)m-B-(L3)l-C-C' (I)N'-A-(L1)n-TM-(L2)m-B-(L3)l-C-C' (I)
    이때, 상기 구조식 (I)에 있어서,At this time, in the structural formula (I),
    상기 N'은 각 폴리펩타이드의 N 말단이고,Wherein N' is the N terminus of each polypeptide,
    상기 C'은 각 폴리펩타이드의 C 말단이며,The C' is the C terminus of each polypeptide,
    상기 - 은 결합을 의미하고,where - means bonding,
    상기 A는 CD137 세포외 도메인 또는 이의 단편이며;wherein A is the CD137 extracellular domain or fragment thereof;
    상기 TM은 막관통 도메인이고;The TM is a transmembrane domain;
    상기 B 및 C는 세포내 신호전달 도메인으로, 각각 독립적으로 공자극 도메인 및 1차 신호전달 도메인이며; B and C are intracellular signaling domains, each independently a co-stimulatory domain and a primary signaling domain;
    상기 L1, L2 및 L3은 각각 펩타이드 링커이며,Wherein L1, L2 and L3 are each peptide linkers,
    상기 n, m 및 l은 각각 독립적으로 0 또는 1이다.The n, m and l are each independently 0 or 1.
  22. 제1항 내지 제21항 중 어느 한 항의 융합단백질을 암호화하는 폴리뉴클레오티드.A polynucleotide encoding the fusion protein of any one of claims 1 to 21.
  23. 제22항의 폴리뉴클레오티드가 적재된 벡터.A vector loaded with the polynucleotide of claim 22.
  24. 제22항의 폴리뉴클레오티드가 포함된 바이러스.A virus containing the polynucleotide of claim 22.
  25. 제1항의 융합단백질이 발현된 면역세포.Immune cells expressing the fusion protein of claim 1.
  26. 제25항에 있어서,According to clause 25,
    상기 면역세포는 자연살해세포, T 세포 또는 대식세포인 것인, 면역세포.The immune cells are natural killer cells, T cells, or macrophages.
  27. 제25항에 있어서,According to clause 25,
    상기 융합단백질이 발현된 세포는 제22항의 폴리뉴클레오티드가 도입되어 형질전환된 것인, 면역세포.An immune cell in which the cell expressing the fusion protein is transformed by introducing the polynucleotide of claim 22.
  28. 제25항의 면역세포를 유효성분으로 포함하는 암 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer comprising the immune cell of claim 25 as an active ingredient.
  29. 제28항에 있어서,According to clause 28,
    상기 약학 조성물은 이중 특이 항체를 추가로 포함하는 것인, 암 예방 또는 치료용 약학 조성물.A pharmaceutical composition for preventing or treating cancer, wherein the pharmaceutical composition further comprises a bispecific antibody.
  30. 제29항에 있어서,According to clause 29,
    상기 이중 특이 항체는 암 항원에 특이적으로 결합하는 제1 도메인; 및 CD137에 특이적으로 결합하는 제2 도메인을 포함하는 것인, 암 예방 또는 치료용 약학 조성물.The bispecific antibody includes a first domain that specifically binds to a cancer antigen; And a pharmaceutical composition for preventing or treating cancer, comprising a second domain that specifically binds to CD137.
  31. 제30항에 있어서,According to clause 30,
    상기 암 항원은 알파 엽산 수용체, 5T4, αvβ6 인테그린, BCMA, B7-H3, B7-H6, CAIX, CD16, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a, CD79b, CD123, CD138, CD171, CEA, CSPG4, EGFR, ErbB2(HER2)를 포함하는 EGFR 패밀리, EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, 태아 AchR, FRα, GD2, GD3, 글리피칸-3(GPC3), HLA-A1+MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ESO-1, HLA-A2+NY-ESO-1, HLA-A3+NY-ESO-1, IL-11Rα, IL-13Rα2, 람다, 루이스-Y, 카파, 메소텔린, Muc1, Muc16, NCAM, NKG2D 리간드, NY-ESO-1, PRAME, PSCA, PSMA, ROR1, SSX, 서바이빈, TAG72, TEMs, VEGFR2 및 WT-1로 이루어진 군으로부터 선택되는 어느 하나인 것인, 암 예방 또는 치료용 약학 조성물.The cancer antigens include alpha folate receptor, 5T4, αvβ6 integrin, BCMA, B7-H3, B7-H6, CAIX, CD16, CD19, CD20, CD22, CD30, CD33, CD44, CD44v6, CD44v7/8, CD70, CD79a, CD79b. , CD123, CD138, CD171, CEA, CSPG4, EGFR, EGFR family including ErbB2 (HER2), EGFRvIII, EGP2, EGP40, EPCAM, EphA2, EpCAM, FAP, fetal AchR, FRα, GD2, GD3, glypican-3 (GPC3), HLA-A1+MAGE1, HLA-A2+MAGE1, HLA-A3+MAGE1, HLA-A1+NY-ESO-1, HLA-A2+NY-ESO-1, HLA-A3+NY-ESO- 1, IL-11Rα, IL-13Rα2, lambda, Lewis-Y, kappa, mesothelin, Muc1, Muc16, NCAM, NKG2D ligand, NY-ESO-1, PRAME, PSCA, PSMA, ROR1, SSX, survivin, A pharmaceutical composition for preventing or treating cancer, which is any one selected from the group consisting of TAG72, TEMs, VEGFR2 and WT-1.
  32. 제28항에 있어서,According to clause 28,
    상기 암은 위암, 간암, 폐암, 대장암, 유방암, 전립선암, 난소암, 췌장암, 자궁경부암, 갑상선암, 후두암, 백혈병, 급성 골수성 백혈병, 뇌종양, 신경모세포종, 망막 모세포종, 두경부암, 침샘암, 림프종, 신장암, 흑색종, 다발 골수종, 뇌암, 골육종, 교모세포종, IgG-opsonized tumor, 림프종, 신경종, 중피종, 및 식도암으로 이루어진 군에서 선택되는 어느 하나인 것인, 암 예방 또는 치료용 약학 조성물.The above cancers include stomach cancer, liver cancer, lung cancer, colon cancer, breast cancer, prostate cancer, ovarian cancer, pancreas cancer, cervical cancer, thyroid cancer, laryngeal cancer, leukemia, acute myeloid leukemia, brain tumor, neuroblastoma, retinoblastoma, head and neck cancer, salivary gland cancer, and lymphoma. , a pharmaceutical composition for preventing or treating cancer, which is any one selected from the group consisting of kidney cancer, melanoma, multiple myeloma, brain cancer, osteosarcoma, glioblastoma, IgG-opsonized tumor, lymphoma, neuroma, mesothelioma, and esophageal cancer.
  33. 제25항의 면역세포 및 제29항의 이중 특이 항체를 포함하는 암 치료용 키트.A kit for cancer treatment comprising the immune cell of item 25 and the dual specific antibody of item 29.
  34. i) 제24항의 바이러스를 제조하는 단계; 및i) preparing the virus of paragraph 24; and
    ii) 상기 바이러스를 면역세포에 처리하는 단계;를 포함하는 제1항의 융합단백질이 발현된 면역세포의 생산 방법.ii) Processing the virus into immune cells; A method of producing immune cells expressing the fusion protein of claim 1, comprising:
  35. CD137 세포외 도메인 변이체 또는 이의 단편.CD137 extracellular domain variant or fragment thereof.
  36. 제35항에 있어서,According to clause 35,
    상기 변이체는 CRD1, CRD2, CRD3, CRD4 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나에 변이를 포함하는 것인, 변이체 또는 이의 단편.The variant is a variant or fragment thereof that includes a mutation in any one selected from the group consisting of CRD1, CRD2, CRD3, CRD4, and combinations thereof.
  37. 제36항에 있어서,According to clause 36,
    상기 변이체는 CRD2, CRD3 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나에 변이를 포함하는 것인, 변이체 또는 이의 단편.The variant is a variant or fragment thereof that includes a mutation in any one selected from the group consisting of CRD2, CRD3, and combinations thereof.
  38. 제37항에 있어서,According to clause 37,
    상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산이 치환된 것인, 변이체 또는 이의 단편.The mutation of CRD2 is a substitution of any one amino acid selected from the group consisting of P3, S6, Q13, T15, C16, D17, I18, Q21, K23, V25, F26, and combinations thereof in the amino acid sequence of SEQ ID NO: 3. A variant or fragment thereof.
  39. 제37항에 있어서,According to clause 37,
    상기 CRD3의 변이는 서열번호 6의 아미노산 서열에서 S14, M15, C16 및 이들의 조합으로 이루어진 군에서 선택되는 어느 하나의 아미노산이 치환된 것인, 변이체 또는 이의 단편.The mutation of CRD3 is a variant or fragment thereof in which any one amino acid selected from the group consisting of S14, M15, C16, and combinations thereof is substituted in the amino acid sequence of SEQ ID NO: 6.
  40. 제38항에 있어서,According to clause 38,
    상기 CRD2의 변이는 서열번호 3의 아미노산 서열에서 I18R, V25R 및 이들의 조합으로 이루어진 군으로부터 선택되는 어느 하나의 아미노산으로 치환된 것인, 변이체 또는 이의 단편.The mutation of CRD2 is a variant or fragment thereof in which any one amino acid selected from the group consisting of I18R, V25R, and a combination thereof is substituted in the amino acid sequence of SEQ ID NO: 3.
  41. 제40항에 있어서,According to clause 40,
    상기 CRD2의 변이는 서열번호 4, 서열번호 5 및 서열번호 26으로 이루어진 군에서 선택되는 어느 하나의 아미노산 서열을 포함하는 것인, 변이체 또는 이의 단편.The CRD2 mutation is a variant or fragment thereof comprising any one amino acid sequence selected from the group consisting of SEQ ID NO: 4, SEQ ID NO: 5, and SEQ ID NO: 26.
PCT/KR2023/019470 2022-11-29 2023-11-29 Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof WO2024117781A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2022-0162933 2022-11-29
KR20220162933 2022-11-29
KR20230115156 2023-08-31
KR10-2023-0115156 2023-08-31

Publications (1)

Publication Number Publication Date
WO2024117781A1 true WO2024117781A1 (en) 2024-06-06

Family

ID=91324715

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2023/019470 WO2024117781A1 (en) 2022-11-29 2023-11-29 Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof

Country Status (1)

Country Link
WO (1) WO2024117781A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20200009190A1 (en) * 2017-03-17 2020-01-09 Fred Hutchinson Cancer Research Center Immunomodulatory fusion proteins and uses thereof
EP3636320A1 (en) * 2018-10-09 2020-04-15 Numab Therapeutics AG Antibodies targeting cd137 and methods of use thereof
KR102426765B1 (en) * 2016-04-22 2022-07-29 엘리게이터 바이오사이언스 에이비 Novel bispecific polypeptide for CD137

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR102426765B1 (en) * 2016-04-22 2022-07-29 엘리게이터 바이오사이언스 에이비 Novel bispecific polypeptide for CD137
US20200009190A1 (en) * 2017-03-17 2020-01-09 Fred Hutchinson Cancer Research Center Immunomodulatory fusion proteins and uses thereof
EP3636320A1 (en) * 2018-10-09 2020-04-15 Numab Therapeutics AG Antibodies targeting cd137 and methods of use thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
CHIN S MICHAEL; KIMBERLIN CHRISTOPHER R; ROE-ZURZ ZYGY; ZHANG PAMELA; XU ALLISON; LIAO-CHAN SINDY; SEN DEBASISH; NAGER ANDREW R; O: "Structure of the 4-1BB/4-1BBL complex and distinct binding and functional properties of utomilumab and urelumab", NATURE COMMUNICATIONS, NATURE PUBLISHING GROUP, UK, vol. 9, 1 January 2018 (2018-01-01), UK, pages Article No.: 4679 - 13, XP009521036, ISSN: 2041-1723, DOI: 10.1038/s41467-018-07136-7 *
ESKIOCAK UGUR, GUZMAN WILSON, WOLF BENJAMIN, CUMMINGS CHRISTINE, MILLING LAUREN, WU HSIN-JUNG, OPHIR MICHAEL, LAMBDEN CONNER, BAKH: "Differentiated agonistic antibody targeting CD137 eradicates large tumors without hepatotoxicity", JCI INSIGHT, vol. 5, no. 5, 12 March 2020 (2020-03-12), XP093060188, DOI: 10.1172/jci.insight.133647 *

Similar Documents

Publication Publication Date Title
WO2020060122A1 (en) Fusion protein comprising il-2 protein and cd80 protein, and use thereof
WO2018124766A2 (en) Chimeric antigen receptor and natural killer cells expressing same
WO2015133817A1 (en) Monoclonal antibody specifically recognizing b-cell lymphoma cells and use thereof
WO2019182425A1 (en) Genetically modified nk cell line having novel chimeric antigen receptor-encoding gene transduced therein, and use thereof
WO2018217064A2 (en) Method for culturing natural killer cell, using transformed t cell
WO2017023138A1 (en) Chimeric antigen receptor, and t cells in which chimeric antigen receptor is expressed
WO2021256724A1 (en) Chimeric antigen receptor targeting bcma and use thereof
WO2021162521A1 (en) Immune cells overexpressing cell signaling regulatory factor introduced from outside and use thereof
WO2018030806A1 (en) Cytokine fused to immunoglobulin fc heterodimer and pharmaceutical composition comprising same
WO2014025198A2 (en) Lfa3 mutant, fusion protein in which target-specific polypeptides are connected to the mutant or lfa3 cd2 binding region, and use thereof
WO2020032784A1 (en) Chimeric antigen receptor binding to hla-dr, and car-t cell
WO2019112347A2 (en) Antibody or antigen binding fragment thereof for specifically recognizing b cell malignancy, chimeric antigen receptor comprising same, and use thereof
WO2020153605A1 (en) Mesothelin-specific chimeric antigen receptor and t cells expressing same
WO2014025199A2 (en) Staphylococcal enterotoxin-derived superantigen mutant, fusion protein in which target-specific polypeptides are connected to the mutant and use thereof
WO2021235696A1 (en) Cd22-specific antibody and use thereof
WO2020101361A1 (en) Method for culturing cord blood-derived natural killer cells using transformed t-cells
WO2021107689A1 (en) Pharmaceutical composition for treatment of cancer, comprising immune checkpoint inhibitor and fusion protein including il-2 protein and cd80 protein
WO2022025638A1 (en) Chimeric antigen receptor (car) t cell stabilizing immune synapse
WO2023277361A1 (en) Mesothelin-specific antibodies and use thereof
WO2021153979A1 (en) Fusion protein comprising anti-taa antibody, anti-pd-l1 antibody, and il-2, and uses thereof
WO2024117781A1 (en) Fusion protein comprising cd137 extracellular domain, immune cells expressing same, and use thereof
WO2020022782A1 (en) Composition for preventing or treating immune-related diseases
WO2021235697A1 (en) Cd22-specific antibody and use thereof
WO2021107635A1 (en) Composition for anticancer treatment, comprising nk cells and fusion protein which comprises il-2 protein and cd80 protein
WO2022145739A1 (en) Humanized antibody specific for cd22 and chimeric antigen receptor using the same