[go: up one dir, main page]

WO2024248286A1 - Pharmaceutical composition for treating cancer comprising polypeptide derived from mycobacterium tuberculosis - Google Patents

Pharmaceutical composition for treating cancer comprising polypeptide derived from mycobacterium tuberculosis Download PDF

Info

Publication number
WO2024248286A1
WO2024248286A1 PCT/KR2024/003333 KR2024003333W WO2024248286A1 WO 2024248286 A1 WO2024248286 A1 WO 2024248286A1 KR 2024003333 W KR2024003333 W KR 2024003333W WO 2024248286 A1 WO2024248286 A1 WO 2024248286A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
polypeptide
cells
variant
asc
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
PCT/KR2024/003333
Other languages
French (fr)
Korean (ko)
Inventor
양철수
김효근
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Industry University Cooperation Foundation IUCF HYU
Original Assignee
Industry University Cooperation Foundation IUCF HYU
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
Priority claimed from KR1020230103350A external-priority patent/KR20240173318A/en
Application filed by Industry University Cooperation Foundation IUCF HYU filed Critical Industry University Cooperation Foundation IUCF HYU
Publication of WO2024248286A1 publication Critical patent/WO2024248286A1/en
Anticipated expiration legal-status Critical
Pending legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • 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
    • C07K14/195Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria
    • C07K14/35Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from bacteria from Mycobacteriaceae (F)

Definitions

  • the present invention relates to a tuberculosis-derived polypeptide that specifically binds to ASC, and a pharmaceutical composition for treating cancer comprising the same.
  • 1st generation anticancer drugs operate on the principle of killing cancer cells or cancer tissues by administering substances that exhibit cytotoxicity or substances that act on biosignals, and metabolic inhibitors, alkylating agents, platinizing agents, antibiotics, and hormonal agents are used, but there is a problem that various side effects occur because they can also act on normal tissues.
  • Targeted anticancer drugs were developed to minimize side effects by acting specifically on cancer cells, but there were problems with limited targets and the occurrence of drug resistance. Accordingly, immunotherapy drugs that remove cancer cells by improving innate immune function, and metabolic anticancer drugs that remove cancer cells by blocking the supply of nutrients to cancer cells, etc. have been proposed.
  • ASC or PYCARD protein has been found in aggregate form in human leukemia cells treated with chemotherapy. It has been reported that ASC is methylated in many tumor patients, which inhibits the role of ASC as a cell death and tumor suppressor. Consequently, ASC has been identified as a protein important for the formation of NLRP3 inflammasome complex, which mediates the secretion of inflammatory cytokines IL-1 ⁇ and IL-18.
  • NLRP3 inflammasome signaling is controlled by various factors such as genetic polymorphisms and mutations that can affect gene expression and ultimately lead to activation.
  • Such effects have been found in patients with inflammatory diseases (erma D, Saerndahl E, Andersson H, Eriksson P, Fredrikson M, Joensson JI, et al.
  • the Q705K polymorphism in NLRP3 is a gain-of-function alteration leading to excessive interleukin-1 ⁇ and L-18 roduction.
  • LoS ne. 2012 :e34977.; Touitou I, Lesage S, McDermott M, Cuisset L, Hoffman H, Dode C, et al.
  • NLRP3 is overexpressed in HNSCC, LSCC, and squamous cell carcinoma tissues compared with normal tissues and is often associated with poor prognosis and pathology
  • Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma.
  • the present invention aims to provide a polypeptide capable of treating cancer by activating NLRP3 inflammasome.
  • the present invention provides a polypeptide comprising a sequence represented by SEQ ID NO: 1 or a variant thereof, which specifically binds to ASC (Apoptosis-associated Speck-like protein containing a CARD).
  • the polypeptide consists of the sequence represented by SEQ ID NO: 1, and in another embodiment of the present invention, the variant may comprise conservative substitutions at positions other than the 2nd, 7th, 10th, and 13th amino acids of SEQ ID NO: 1, and in yet another embodiment of the present invention, the polypeptide comprises the sequence represented by SEQ ID NO: 2, and in yet another embodiment of the present invention, the conservative substitutions are at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8V, G9A, G11A, A12G, and G14A, or at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8L, G9A, G11A, A12G, and
  • the present invention provides an ASC-specific binding molecule comprising the above-mentioned polypeptide or a variant thereof.
  • the present invention provides a pharmaceutical composition for treating cancer, comprising the above-described polypeptide or variant.
  • the cancer may be inflammatory colon cancer, colorectal cancer, melanoma, or hepatocellular carcinoma.
  • the present invention provides a nucleic acid molecule expressing the above polypeptide or variant.
  • the present invention provides a pharmaceutical composition for treating cancer, comprising the nucleic acid molecule described above.
  • the present invention provides a recombinant vector comprising the nucleic acid molecule described above.
  • the present invention provides a host cell comprising the nucleic acid molecule described above.
  • the polypeptide according to the present invention specifically binds to the ASC protein, thereby inducing multimerization of ASC, thereby activating the NLRP3 inflammasome. Therefore, the polypeptide according to the present invention, or a variant thereof, or a nucleic acid molecule expressing the same, can be used as an anticancer agent.
  • Figure 1a shows the results of confirming that the Rv0747/PE_PGRS10 protein interacts with ASC
  • Figure 1b shows the results of confirming the interacting domain of the Rv0747/PE_PGRS10 protein and ASC
  • Figure 1c shows the results of confirming the sequence of the Rv0747/PE_PGRS10 protein interacting with ASC
  • Figure 1d shows the sequence of a peptide consisting of 14 amino acids interacting with ASC.
  • FIG. 2a shows the results confirming that the binding of NLRP3 and ASC increases by treating with the peptide according to the present invention
  • FIG. 2b shows the results confirming that the secretion of IL-1 ⁇ and IL-18 increases by treating with the peptide according to the present invention
  • FIG. 2c shows the results confirming that the secretion of caspse-1 increases by treating with the peptide according to the present invention
  • FIG. 2d shows the results confirming that the oligomerization of ASC increases by treating with the peptide according to the present invention
  • E shows the results confirming that the ratio of intracellular oligomerized ASC increases by treating with the peptide according to the present invention.
  • Figure 3 shows the results of confirming amino acids involved in the binding of a peptide and ASC according to one embodiment of the present invention.
  • NLRP3 inflammasome is a multiprotein complex that plays a central role in regulating the innate immune system and inflammatory signaling.
  • PAMPs pathogen-associated molecular patterns
  • DAMPs damage-associated molecular patterns
  • NLRP3 oligomerizes and activates caspase-1, which initiates the processing and release of the proinflammatory cytokines IL-1 ⁇ and IL-18.
  • the NLRP3 inflammasome has anti-tumor activity. It has been reported that the NLRP3 inflammasome is required for dendritic cell-mediated priming of IFN- ⁇ -producing T lymphocytes against tumor cells (Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1 ⁇ -dependent adaptive immunity against tumors. Nat Med. (2009) 15:1170-8.).
  • the NLRP3 inflammasome appears to act as a negative regulator of tumorigenesis during colitis-associated cancer (Allen IC, TeKippe EM, Woodford R-MT, Uronis JM, Holl EK, Rogers AB, et al.
  • the NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. (2010) 207:1045-56.), suggesting a critical role for the NLRP3 inflammasome in regulating intestinal homeostasis and consequently in protection against colitis (Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti TD.
  • NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity. (2010) 32:379-91). Furthermore, NLRP3 inflammasome deficiency has been shown to increase tumorigenesis in colorectal cancer. In addition, NLRP3 inflammasome-mediated IL-18 production has been reported to suppress colorectal cancer metastatic growth in the liver (Dupaul-Chicoine J, Arabzadeh A, Dagenais M, Douglas T, Champagne C, Morizot A, et al. The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity. (2015) 43:751-63.).
  • NLRP3/IL-18-mediated downregulation of IL-22BP provides a protective role against intestinal tissue damage during peak inflammation (Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. (2012) 491:259-63.).
  • NLRP3 in the TME has been shown to attenuate antitumor immune responses to cancer vaccines by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells (MDSCs) and suppressing T cell responses (Van Deventer HW, Burgents JE, Wu QP, Woodford RMT, Brickey WJ, Allen IC, et al.
  • the inflammasome component Nlrp3 impairs antitumor vaccine by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells. Cancer Res. (2010) 70:10161-9.).
  • the present invention provides a peptide derived from Mycobacterium tuberculosis that can be used as an anticancer agent by specifically binding to ASC (Apoptosis-associated Speck-like protein containing a CARD) and activating NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome. More specifically, the peptide according to the present invention is derived from Rv0747/PE_PGRS10 (SEQ ID NO: 2) of Mycobacterium tuberculosis. Wild-type Rv0747/PE_PGRS10 consists of 801 amino acids.
  • the present invention provides a peptide having a TLTGRPLIGNGANG sequence (SEQ ID NO: 1) consisting of the 401st to 414th amino acids of Rv0747/PE_PGRS10, which specifically binds to ASC.
  • the DNA sequence encoding the full-length Rv0747/PE_PGRS10 is represented by SEQ ID NO: 4, and the DNA sequence encoding the full-length Rv0747/PE_PGRS10 is represented by SEQ ID NO: 2.
  • ASC (NP_037390), also named PYCARD, consists of an N-terminal PYD (PYRIN-PAAD-DAPIN) domain and a C-terminal CARD (caspase-recruitment domain) domain.
  • the PYD and CARD domains are members of the six-helix bundle death domain fold superfamily, which mediates the assembly of large signaling complexes in inflammatory and apoptotic signaling pathways through caspase activation. In normal cells, this protein is located in the cytoplasm, but in cells undergoing apoptosis, it forms globular aggregates near the perinuclear region.
  • the ASC protein consists of 195 amino acids.
  • ASC is multimerized or polymerized, which induces activation of NLRP3 inflammasome and specifically increases the secretion of IL-1 ⁇ and/or IL-18 and caspase-1, thereby inducing the death of cancer cells.
  • fragment means a part of the amino acid sequence, i.e. a sequence which represents an amino acid sequence that is shortened at the N-terminus and/or the C-terminus.
  • a fragment shortened at the C-terminus can be obtained, for example, by translating a truncated open reading frame lacking the 3'-end of the open reading frame.
  • a fragment shortened at the N-terminus can be obtained, for example, by translating a truncated open reading frame lacking the 5'-end of the open reading frame, as long as the truncated open reading frame comprises an initiation codon which serves to initiate translation.
  • a fragment of an amino acid sequence comprises, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% of the amino acid residues from the amino acid sequence.
  • the fragment of the amino acid sequence preferably comprises at least 6, in particular at least 8, at least 12, at least 13, at least 14, at least 15, at least 20, at least 30, at least 50 or at least 100 consecutive amino acids from the amino acid sequence.
  • a “variant”, “variant”, “variant protein”, or “variant polypeptide” of a polypeptide or protein refers to any analog, fragment, derivative, or mutant derived from the polypeptide or protein and which retains at least one biological property of the polypeptide or protein.
  • Different variants of the polypeptide or protein may exist in nature. These variants may be allelic mutations characterized by differences in the nucleotide sequence of the structural gene encoding the protein, or may include differential splicing or post-translational modifications.
  • One skilled in the art can generate variants having one or more amino acid substitutions, deletions, additions, or replacements.
  • variants may include, inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids, (b) variants in which one or more amino acids are added to the polypeptide or protein, (c) variants in which one or more of the amino acids comprises a substitution group, and (d) variants in which the polypeptide or protein is fused to another polypeptide, such as serum albumin.
  • Conservative variants also refer to amino acid sequences having sequence changes that do not adversely affect the biological function of the protein.
  • a substitution, insertion, or deletion is described as adversely affecting the protein if the altered sequence disrupts or destroys the biological function associated with the protein.
  • the overall charge, structure, or hydrophobic-hydrophilicity of a protein can be altered without adversely affecting the biological activity.
  • the amino acid sequence can be altered to render the peptide more hydrophobic or more hydrophilic without adversely affecting the biological activity of the protein.
  • parent polypeptide As used herein, the terms "parent polypeptide”, “parent protein”, “precursor polypeptide” or “precursor protein” refer to an unmodified polypeptide that is later modified to form a variant.
  • the parent polypeptide can be a wild-type polypeptide, or a variant or engineered version of a wild-type polypeptide.
  • wild type or WT or “native” refers to an amino acid sequence found in nature, including allelic variants.
  • a wild type protein or polypeptide has an amino acid sequence that has not been intentionally modified.
  • a “variant" of an amino acid sequence includes amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants.
  • the term “variant” includes all splice variants, post-translationally modified variants, conformations, isoforms and species homologs, particularly those naturally expressed by a cell.
  • the term “variant” particularly includes fragments of an amino acid sequence.
  • Amino acid insertion variants comprise the insertion of one or more amino acids into a particular amino acid sequence.
  • amino acid sequence variants having insertions one or more amino acid residues are inserted at a specific site in the amino acid sequence, although random insertions using appropriate screening of the resulting products are also possible.
  • Amino acid addition variants comprise amino- and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50 or more amino acids.
  • Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as the removal of 1, 2, 3, 5, 10, 20, 30, 50 or more amino acids. The deletion can be at any position in the protein.
  • Amino acid deletion variants comprising deletions at the N-terminus and/or C-terminus of a protein are also referred to as N-terminal and/or C-terminal truncation variants.
  • Amino acid substitution variants are characterized by the deletion of one or more residues in the sequence and the insertion of another residue in its place. Preference is given to modifications located in an amino acid sequence that is not conserved between homologous proteins or peptides and/or to replacement of an amino acid with another amino acid having similar properties.
  • polypeptide according to the present invention may include conservative substitutions at positions other than the 2nd, 7th, 10th, and 13th amino acids of SEQ ID NO: 1.
  • a variant encompassed by the present invention is a protein comprising an amino acid sequence having 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence represented by SEQ ID NO: 1 or 2, which maintains functional identity with a protein comprised of the amino acid sequence represented by SEQ ID NO: 1 or 2.
  • the amino acid changes in the peptide and protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids.
  • a conservative amino acid change involves substitution of one of a family of amino acids with a similar side chain.
  • Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar amino acids (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and non-charged polar amino acids (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine). Phenylalanine, tryptophan, and tyrosine are sometimes grouped together as aromatic amino acids.
  • a conservative amino acid substitution comprises a substitution within the following groups:
  • valine isoleucine, leucine
  • Phenylalanine, tyrosine Phenylalanine, tyrosine.
  • a variant in SEQ ID NO: 1 is replaced with serine (T1S); threonine at position 3 is replaced with serine (T3S); glycine at position 4 is replaced with alanine (G4A); arginine at position 5 is replaced with lysine (R5K); isoleucine at position 8 is replaced with valine (I8V) or leucine (I8L); glycine at position 9 is replaced with alanine (G9A); glycine at position 11 is replaced with alanine (G11A); alanine at position 12 is replaced with glycine (A12G); or glycine at position 14 is replaced with alanine (G14A).
  • acidic amino acid residue preferably relates to glutamic acid (glutamate, Glu) or aspartic acid (aspartate, Asp), particularly glutamic acid.
  • basic amino acid residue preferably relates to lysine (Lys) or arginine (Arg), particularly lysine.
  • the degree of similarity, preferably identity, between a given amino acid sequence and an amino acid sequence which is a variant of said given amino acid sequence will be at least about 60%, 65%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%.
  • the degree of similarity or identity is preferably provided for an amino acid region which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or about 100% of the full length of the reference amino acid sequence.
  • the degree of similarity or identity is preferably provided for at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180 or about 200 amino acids, preferably contiguous amino acids.
  • the degree of similarity or identity is provided for the full length of the reference amino acid sequence. Alignments for determining sequence similarity, preferably sequence identity, can be performed using tools known in the art, preferably using best sequence alignment, e.g. Align, with standard settings, preferably EMBOSS::needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5.
  • Sequence similarity refers to the percentage of amino acids that are identical or represent conservative amino acid substitutions.
  • Sequence identity refers to the percentage of amino acids that are identical between the sequences.
  • % identity is intended to refer to the percentage of amino acid residues that are identical between the two sequences being compared, obtained by best alignment, and this percentage is purely statistical, the differences between the two sequences being distributed randomly and over their entire length.
  • Sequence comparisons between two amino acid sequences are typically performed by optimally aligning those sequences and then comparing them, said comparison being performed by segments or "comparison windows" in order to identify and compare local regions of sequence similarity.
  • Optimal alignment of sequences for comparison may be performed manually, or by means of the local homology algorithm of Smith and Waterman, 1981, Ads App. Math. 2, 482, or by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. Mol. Biol.
  • % Identity is calculated by determining the number of identical positions between the two sequences being compared, dividing this number by the number of positions compared, and then multiplying the result by 100 to obtain the % identity between the two sequences.
  • Homologous amino acid sequences according to the present specification exhibit at least 40%, particularly at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, and preferably at least 95%, at least 98% or at least 99% identity with respect to amino acid residues.
  • amino acid sequence variants described herein can be readily prepared by those skilled in the art, for example, by recombinant DNA manipulation. Manipulation of DNA sequences to produce peptides or proteins having substitutions, additions, insertions or deletions is described in detail, for example, in Sambrook et al. (1989). In addition, the peptides and amino acid variants described herein can be readily prepared with the aid of known peptide synthesis techniques, for example, by solid phase synthesis and similar methods.
  • the fragment or variant of an amino acid sequence is preferably a "functional fragment” or "functional variant".
  • the term "functional fragment” or “functional variant” of an amino acid sequence relates to any fragment or variant that exhibits one or more functional properties identical or similar to the amino acid sequence from which it is derived, i.e. is a functional equivalent.
  • the term “functional fragment” or “functional variant,” as used herein, refers to a variant molecule or sequence that comprises an amino acid sequence in which one or more amino acids have been altered, particularly compared to the amino acid sequence of the parent molecule or sequence, and which is still capable of performing one or more of the functions of the parent molecule or sequence, e.g., binding to a target molecule or contributing to binding to a target molecule.
  • the alteration in the amino acid sequence of the parent molecule or sequence does not significantly affect or alter the binding characteristics of the molecule or sequence.
  • the binding of the functional fragment or functional variant may be reduced but still significantly present, e.g., the binding of the functional variant may be at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of that of the parent molecule or sequence. However, in other embodiments, the binding of the functional fragment or functional variant may be increased relative to the parent molecule or sequence.
  • amino acid sequence "derived from" a specified amino acid sequence (peptide, protein or polypeptide) means the origin of the first amino acid sequence.
  • an amino acid sequence derived from a particular amino acid sequence has an amino acid sequence that is identical, essentially identical or homologous to that particular sequence or a fragment thereof.
  • An amino acid sequence derived from a particular amino acid sequence may be a variant of that particular sequence or a fragment thereof.
  • Rv0747/PE_PGRS10 suitable for use herein may be modified in sequence from a naturally occurring or native sequence derived therefrom, but which retains the desired activity of the native sequence.
  • isolated means altered or removed from its natural state.
  • a nucleic acid or peptide naturally occurring in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from its natural coexisting materials is “isolated.”
  • An isolated nucleic acid or protein may exist in a substantially purified form, or may exist in a non-natural environment, such as, for example, a host cell.
  • recombinant means “created through genetic manipulation.”
  • a “recombinant subject,” such as a recombinant cell, in the context of the present invention does not occur naturally.
  • naturally occurring means that a subject can be found in nature.
  • a peptide or nucleic acid is naturally occurring if it is present in an organism (including a virus), can be isolated from a source in nature, and has not been intentionally modified by humans in a laboratory.
  • the term “genetic modification” includes transfection of a cell with a nucleic acid.
  • the term “transfection” relates to the introduction of a nucleic acid, particularly RNA, into a cell.
  • the term “transfection” also includes the introduction of a nucleic acid into a cell or the uptake of a nucleic acid by such a cell.
  • RNA can be transfected into a cell and cause it to transiently express its encoded protein. Since the nucleic acid introduced during the transfection process is generally not integrated into the nuclear genome, the foreign nucleic acid will be diluted or degraded through mitosis.
  • Cells capable of episomal amplification of the nucleic acid significantly reduce the dilution rate. If the transfected nucleic acid is in fact retained in the cell and its progenitor cells, stable transfection must occur. Such stable transfection can be achieved by using a virus-based system or a transposon-based system for transfection.
  • cells genetically modified to express a receptor polypeptide and/or an antigen receptor are stably transfected with a nucleic acid encoding a receptor polypeptide and/or a nucleic acid encoding an antigen receptor, whereas typically, a nucleic acid encoding a ligand polypeptide and/or a nucleic acid encoding an antigen are transiently transfected into the cell.
  • nucleic acid refers to a polymer of the phosphate esters of ribonucleosides (adenosine, guanosine, uridine or cytidine; "RNA molecule”) or deoxyribonucleosides (deoxyadenosine, deoxyguanosine, deoxythymine or deoxycytidine; "DNA molecule”), either in single-stranded form or as a double-stranded helix or any of their phosphate ester analogues such as phosphorothioates and thioesters.
  • Double-stranded DNA-DNA, DNA-RNA and RNA-RNA helices are possible.
  • nucleic acid molecule and particularly DNA or RNA molecule, refer only to the primary and secondary structures of said molecules and are not limited to any particular tertiary configuration.
  • the term includes, among others, linear or circular DNA molecules (e.g., restriction fragments), plasmids, supercoiled DNA, and double-stranded DNA found as chromosomes.
  • the sequence may be described herein according to the general convention of presenting the sequence only in the 5' to 3' direction along the non-transcribed DNA strand (i.e., the strand having the sequence matching the mRNA).
  • a "recombinant DNA molecule” is a DNA molecule that has undergone molecular biological manipulation.
  • the DNA includes, but is not limited to, cDNA, genomic DNA, plasmid DNA, synthetic DNA, and semi-synthetic DNA.
  • the nucleic acid may be contained within a vector.
  • vector includes any vector known to those skilled in the art, including plasmid vectors, cosmid vectors, phage vectors such as lambda phage, viral vectors such as retrovirus, adenovirus or baculovirus vectors, or artificial chromosome vectors such as bacterial artificial chromosomes (BAC), yeast artificial chromosomes (YAC) or P1 artificial chromosomes (PAC).
  • BAC bacterial artificial chromosomes
  • YAC yeast artificial chromosomes
  • PAC P1 artificial chromosomes
  • Such vectors include expression as well as cloning vectors.
  • Expression vectors include plasmids as well as viral vectors, and generally contain the desired coding sequence and appropriate DNA sequences necessary for expression of the coding sequence operably linked in a particular host organism (e.g., bacteria, yeast, plants, insects, or mammals) or in vitro expression system.
  • Cloning vectors are generally used to manipulate and amplify a particular desired DNA fragment and may lack functional sequences necessary for expression of the desired DNA fragment.
  • Viral vectors have been used for a wide range of gene transfer applications in living animals as well as cells.
  • Viral vectors that may be used include, but are not limited to, adenovirus, retrovirus, vaccinia virus, poxvirus, adeno-associated virus, herpes simplex virus, lentivirus, baculovirus, sendai virus, measles virus, simian virus 40, and Epstein-Barr virus vectors.
  • Non-viral vectors include plasmids, lipoplexes (cationic liposome-DNA complexes), polyplexes (cationic polymer-DNA complexes), and protein-DNA complexes.
  • the vector may include one or more regulatory regions and/or selectable markers that are useful for selecting, measuring, and monitoring the outcome of the nucleic acid delivery (e.g., delivery to tissues, persistence of expression, etc.).
  • expression vector refers to a vector, plasmid or vehicle designed to transform a host cell by expressing an inserted nucleic acid sequence.
  • the cloned gene i.e., the inserted nucleic acid sequence, is generally placed under the control of regulatory elements such as a promoter, a minimal promoter, an enhancer, and the like.
  • regulatory elements such as a promoter, a minimal promoter, an enhancer, and the like.
  • the initiation regulatory regions or promoters useful for directing expression of the nucleic acid in a desired host cell are numerous and are well known to those skilled in the art.
  • Virtually any promoter capable of directing expression of these genes includes, but is not limited to, viral promoters, bacterial promoters, animal promoters, mammalian promoters, synthetic promoters, constitutive promoters, tissue-specific promoters, pathogenesis or disease-related promoters, developmental specific promoters, inducible promoters, light regulated promoters; SV40 early (SV40) promoter region, promoter contained in the 3' long terminal repeat (LTR) of Rous sarcoma virus (RSV), E1A or major late promoter (MLP) of adenovirus (Ad), immediate early promoter of human cytomegalovirus (HCMV), herpes simplex virus (HSV) thymidine kinase (TK) promoter, baculovirus IE1 promoter, elongation factor 1 alpha (EF1) promoter, glyceraldehyde-3-phosphate dehydrogenase (GSPDH) promoter, phosphoglycer
  • Vectors can be transfected by methods known in the art, such as injection, transfection, electroporation, microinjection, transduction, cell fusion, lipofection, calcium phosphate precipitation (Graham, F.L. et al., Virology, 52:456(1973); and Chen and Okayama, Mol. Cell. Biol. 7:2745-2752(1987)), liposome-mediated transfection (Wong, T.K. et al., Gene, 10:87(1980); Nicolau and Sene, Biochim. Biophys.
  • the polynucleotides according to the present invention can be introduced in vivo by lipofection. Over the past several decades, there has been an increasing use of liposomes for in vitro nucleic acid encapsulation and transfection. Synthetic cationic lipids, which are designed to limit the difficulties and risks encountered with liposome-mediated transfection, can be used to prepare liposomes for in vivo gene transfection (Felgner et al., Proc. Natl. Acad. Sci. USA. 84:7413 (1987); Mackey et al., Proc. Natl. Acad. Sci. USA 85:8027 (1988); and Ulmer et al., Science 259:1745 (1993)).
  • cationic lipids can facilitate encapsulation of negatively charged nucleic acids, and can also facilitate fusion with negatively charged cell membranes (Felgner et al., Science 337:387 (1989)).
  • Particularly useful lipid compounds and compositions for the delivery of nucleic acids are described in WO95/18863, WO96/17823, and U.S. 5,459,127.
  • the use of lipofection to introduce exogenous genes into specific tissues in vivo has several practical advantages. Targeting of molecules to specific cells by liposomes presents one area of advantage. Direct transfection into specific cell types would clearly be particularly desirable in tissues with cellular heterogeneity, such as the pancreas, liver, kidney, and brain.
  • Lipids can be chemically linked to other molecules for targeting purposes (Mackey et al. 1988).
  • Targeted peptides such as hormones or neurotransmitters, and proteins, such as antibodies, or non-peptide molecules can be chemically bound to the liposomes.
  • cationic oligopeptides e.g., WO95/21931
  • peptides derived from DNA binding proteins e.g., WO96/25508
  • cationic polymers e.g., WO95/21931
  • nucleic acids described herein may be recombinant and/or isolated molecules.
  • transcription refers to the process by which the genetic code in a DNA sequence is transcribed into RNA.
  • the RNA can then be translated into peptides or proteins.
  • Encoding refers to the inherent property of a particular sequence of nucleotides in a polynucleotide, such as a gene, cDNA or mRNA, to serve as a template for the synthesis of other polymers and macromolecules in a biological process, having a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties derived therefrom.
  • a gene encodes a protein if the transcription and translation of the mRNA corresponding to that gene produces a protein in a cell or other biological system.
  • Both the coding strand which is identical to the mRNA sequence and is usually provided as a sequence listing, and the non-coding strand, which is used as a template for transcription of the gene or cDNA, may be said to encode a protein or other product of that gene or cDNA.
  • the present invention provides a host cell comprising a vector of the present invention.
  • the host cell includes prokaryotic (e.g., bacterial) and eukaryotic (e.g., fungal, yeast, animal, insect, plant) cells and may be any cell suitable for expression of a polypeptide according to the present invention.
  • Suitable prokaryotic host cells include, but are not limited to, E. coli (e.g., strains DH5, HB101, JM109 or W3110), Bacillus, Streptomyces, Salmonella, Serratia and Pseudomonas species.
  • Suitable eukaryotic host cells include, but are not limited to, COS, CHO, HepG-2, CV-1, LLCMK2, 3T3, HeLa, RPMI8226, 293, BHK-21, Sf9, Saccharomyces, Pichia, Hansenula, Kluyveromyces, Aspergillus or Trichoderma species.
  • endogenous means any substance derived from or produced within an organism, cell, tissue, or system.
  • exogenous means any substance originating or produced outside an organism, cell, tissue, or system.
  • the term “expression” refers to the biological production of a product encoded by a coding sequence, and can be defined as the transcription and/or translation of a particular nucleotide sequence.
  • a DNA sequence comprising a coding sequence is transcribed to form messenger RNA (mRNA).
  • mRNA messenger RNA
  • the messenger RNA is then translated to form a polypeptide product having the relevant biological activity.
  • the expression process may also include additional processing steps for the RNA transcript product (e.g., splicing to remove introns), and/or post-translational processing of the polypeptide product.
  • the peptides, proteins, polypeptides, RNAs, RNA particles and additional agents, e.g., immune checkpoint inhibitors, described herein can be administered as pharmaceutical compositions or medicaments for therapeutic or prophylactic treatment and can be administered in the form of any suitable pharmaceutical composition, which can include a pharmaceutically acceptable carrier and optionally can include one or more adjuvants, stabilizers, and the like.
  • the pharmaceutical composition is for use in therapeutic or prophylactic treatment, e.g., treating or preventing an antigen-associated disease, such as cancer diseases, as described herein.
  • composition relates to a formulation comprising a therapeutically effective substance, preferably together with a pharmaceutically acceptable carrier, diluent and/or excipient.
  • the pharmaceutical composition is useful for reducing the severity of, preventing or treating a disease or disorder by administering the pharmaceutical composition to a subject.
  • Pharmaceutical compositions are also known in the art as pharmaceutical dosage forms.
  • a pharmaceutical composition comprises a peptide, a protein, a polypeptide, an RNA, an RNA particle, an immune effector cell and/or an additional substance as described herein.
  • compositions of the present disclosure may include one or more adjuvants or may be administered together with one or more adjuvants.
  • adjuvant refers to a compound that prolongs, enhances, or accelerates an immune response.
  • adjuvants include heterogeneous compounds such as oil emulsions (e.g., Freund's adjuvant), mineral compounds (e.g., alum), bacterial products (e.g., pertussis toxin), or immune-stimulating complexes.
  • adjuvants include, but are not limited to, cytokines such as LPS, GP96, CpG oligodeoxynucleotides, growth factors and monokines, lymphokines, interleukins, chemokines.
  • the cytokines can be IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL12, IFN ⁇ , IFN ⁇ , GM-CSF, LT-a.
  • adjuvants are aluminum hydroxide, Freund's adjuvant or oils such as Montanide®ISA51.
  • suitable adjuvants for use herein include lipopeptides such as Pam3Cys.
  • composition according to the present specification is generally applied as a “pharmaceutically effective amount” and as a “pharmaceutically acceptable formulation”.
  • pharmaceutically acceptable refers to the non-toxicity of a substance that does not interact with the action of the active ingredient of a pharmaceutical composition.
  • the term "pharmaceutically effective amount” or “therapeutically effective amount” means an amount that, alone or in combination with additional administration, achieves the desired response or desired effect.
  • the desired response preferably means inhibition of the progression of the disease. This includes slowing the progression of the disease, and in particular, stopping or reversing the progression of the disease.
  • the desired response may also be delaying the onset or preventing the onset of the disease or condition.
  • the effective amount of the compositions described herein will be determined based on the individual characteristics of the patient, including the condition being treated, the severity of the disease, age, physical condition, height and weight, the duration of the treatment, the type of concomitant therapy (if any), the specific route of administration, and similar factors. Accordingly, the dosage of the compositions described herein can be determined based on these various characteristics. If the patient does not respond sufficiently with the first administration, a higher dose (or a higher dose achieved effectively by another, more local route of administration) may be used.
  • compositions of the present disclosure may include salts, buffers, preservatives, and optionally other therapeutic agents.
  • the pharmaceutical compositions of the present disclosure include one or more pharmaceutically acceptable carriers, diluents, and/or excipients.
  • Preservatives suitable for use in the pharmaceutical compositions of the present disclosure include, but are not limited to, benzalkonium chloride, chlorobutanol, parabens and thimerosal.
  • excipient refers to a material that may be present in the pharmaceutical compositions of the present disclosure but is not an active ingredient.
  • excipients include, but are not limited to, carriers, binders, diluents, lubricants, thickeners, surfactants, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, or coloring agents.
  • diluting and/or thinning substance means a diluting and/or thinning substance.
  • the term “diluent” includes any one or more of a fluid, a liquid or solid suspension and/or a mixed medium. Examples of suitable diluents include ethanol, glycerol, and water.
  • carrier refers to an ingredient which may be natural, synthetic, organic or inorganic, which is combined with an active ingredient to facilitate, enhance or enable administration of the pharmaceutical composition.
  • a carrier can be one or more compatible solid or liquid fillers, diluents or encapsulating materials suitable for administration to a subject. Suitable carriers include, but are not limited to, sterile water, Ringer's, Ringer's lactate, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes, and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers.
  • the pharmaceutical compositions herein comprise isotonic saline.
  • compositions may be selected depending on the intended route of administration and standard pharmaceutical practice.
  • the pharmaceutical compositions described herein can be administered intravenously, intraarterially, subcutaneously, intradermally, or intramuscularly.
  • the pharmaceutical compositions are formulated for topical administration or systemic administration.
  • Systemic administration can include enteral administration involving absorption through the gastrointestinal tract or parenteral administration.
  • parenteral administration means administration by any means other than via the gastrointestinal tract, such as intravenous injection.
  • the pharmaceutical compositions are formulated for systemic administration.
  • systemic administration is by intravenous administration.
  • the polypeptides described herein or variants thereof, or nucleic acid molecules encoding them are administered systemically.
  • co-administration means administering multiple compounds or compositions (e.g., immune effector cells, a polypeptide described herein or a variant thereof or a nucleic acid molecule encoding the same, and optionally an RNA encoding a vaccine antigen) to the same patient.
  • the multiple compounds or compositions can be administered simultaneously, essentially simultaneously, or sequentially.
  • the materials, compositions and methods described herein can be used to treat a subject having a disease, for example, a disease characterized by the presence of cells that express the antigen.
  • a particularly preferred disease is cancer.
  • the antigen is derived from a virus
  • the materials, compositions and methods can be useful for treating a viral disease caused by the virus.
  • the antigen is a tumor antigen
  • the materials, compositions and methods can be useful for treating a cancer disease, wherein cancer cells express the tumor antigen.
  • the materials, compositions, and methods described herein may be used in the therapeutic or prophylactic treatment of a variety of diseases, wherein support of immune effector cells and/or activation of immune effector cells as described herein is beneficial to a patient, such as cancer and infectious diseases.
  • the materials, compositions, and methods described herein are useful in the prophylactic and/or therapeutic treatment of diseases associated with an antigen.
  • disease refers to an abnormal condition affecting the body of an individual.
  • a disease is often interpreted as a medical condition associated with specific symptoms and signs.
  • a disease may be caused by an agent originating from an external source, such as an infectious disease, or may be caused by an internal dysfunction, such as an autoimmune disease.
  • "disease” is used in a broader sense to refer to any condition that, when contacted by an individual, causes pain, dysfunction, distress, social problems, or death or similar problems to the individual suffering from the disease. In a broader sense, it sometimes includes injuries, disabilities, disorders, syndromes, infections, isolated symptoms, deviant behaviors, and structural and functional atypical changes, which in different contexts and for different purposes may be considered distinct categories. Disease generally affects an individual not only physically but also emotionally, as living with various diseases can change the individual's outlook on life and personality.
  • treatment means the care and attention of a subject for the purpose of combating a condition, such as a disease or disorder.
  • the term is intended to encompass the full range of treatments for a given condition from which a subject is suffering, such as the administration of a therapeutically effective compound to alleviate symptoms or complications, delay the progression of the disease, disorder or condition, relieve or alleviate symptoms and complications, and/or cure or eliminate the disease, disorder or condition, as well as prevent the condition, wherein prevention will be understood as the care and attention of a subject for the purpose of combating the disease, condition or disorder, and includes the administration of an active compound to prevent the onset of symptoms or complications.
  • terapéutica treatment means any treatment that improves the health status of a subject and/or prolongs (increases) the life span of a subject. Such treatment may be elimination of a disease in a subject, arrest or slowing of the progression of a disease in a subject, inhibition or slowing of the progression of a disease in a subject, reduction in the frequency or severity of symptoms in a subject, and/or reduction in relapse in a subject currently suffering from or previously suffering from a disease.
  • prophylactic treatment means any treatment intended to prevent a disease from occurring in a subject.
  • prophylactic treatment or “prophylactic treatment” are used interchangeably herein.
  • the terms “individual” and “subject” are used interchangeably herein. These terms refer to a human or other mammal (e.g., a mouse, rat, rabbit, dog, cat, cow, pig, sheep, horse, or primate) that may be susceptible to or may be susceptible to a disease or disorder (e.g., cancer), but may or may not have the disease or disorder. In many embodiments, the subject is a human. Unless otherwise specified, the terms “individual” and “subject” do not imply a particular age, and thus encompass adults, elderly people, children, and newborns. In embodiments herein, an "individual” or “individual” is a "patient.”
  • patient means an individual or entity in need of treatment, specifically an individual or entity suffering from a disease.
  • the goal is to provide an immune response to cells having a disease, such as cancer cells expressing an antigen, and to treat a disease, such as a cancer disease, involving cells expressing an antigen, such as a tumor antigen.
  • immune response refers to a coordinated body response to an antigen or cells expressing an antigen, and includes a cellular immune response and/or a humoral immune response.
  • cell-mediated immunity refers to a cellular response directed against cells characterized by the expression of an antigen, particularly by presentation of the antigen with class I or class II MHC.
  • the cellular response relates to cells called T cells or T lymphocytes, which act either as “helper” or “killer” cells.
  • Helper T cells also called CD4+ T cells
  • killer cells also called cytotoxic T cells, cytolytic T cells, CD8 + T cells, or CTLs kill diseased cells, such as cancer cells, thereby preventing the development of more diseased cells.
  • inducing [or inducing] an immune response may mean that there is no immune response to a particular antigen prior to induction, or it may mean that an immune response to a particular antigen exists at a baseline level prior to induction and is enhanced following induction.
  • inducing [or inducing] an immune response includes “potentiating [or enhancing] an immune response.”
  • immunotherapy refers to the treatment of a disease or condition by induction or enhancement of an immune response.
  • immunotherapy includes antigen immunization or antigen vaccination.
  • professional antigen-presenting cell is an antigen-presenting cell that constitutively expresses major histocompatibility complex class II (MHC class II) molecules required for interaction with na ⁇ ve T cells. If a T cell interacts with a complex of MHC class II molecules on the membrane of the antigen-presenting cell, the antigen-presenting cell produces co-stimulatory molecules that induce activation of the T cell.
  • Professional antigen-presenting cells include dendritic cells and macrophages.
  • disease involving an antigen means any disease involving an antigen, for example a disease characterized by the presence of an antigen.
  • the disease involving an antigen may be an infectious disease or a cancer disease or a simple cancer.
  • the antigen may be a disease-associated antigen, such as a tumor-associated antigen, a viral antigen or a bacterial antigen.
  • the disease involving an antigen is a disease involving cells expressing the antigen, preferably on the surface of the cells.
  • infectious disease refers to any disease that can be transmitted from subject to subject or from organism to organism and is caused by a microbial agent (e.g., a cold). Infectious diseases are known in the art and include, for example, viral, bacterial or parasitic diseases caused by viruses, bacteria and parasites, respectively.
  • an infectious disease can be, for example, hepatitis, sexually transmitted diseases (e.g., chlamydia or gonorrhea), tuberculosis, HIV/acquired immunodeficiency syndrome (AIDS), diphtheria, hepatitis B, hepatitis C, cholera, severe acute respiratory syndrome (SARS), avian influenza and influenza.
  • cancer disease refers to or means a pathological condition that is typically characterized by uncontrolled cell proliferation in an organism.
  • cancers include, but are not limited to, carcinomas, lymphomas, blastomas, sarcomas, and leukemias.
  • cancers include bone cancer, blood cancer, lung cancer, liver cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal area, stomach cancer, colon cancer, breast cancer, prostate cancer, uterine cancer, carcinomas of the sexual and reproductive organs, Hodgkin's disease, esophageal cancer, small intestinal cancer, endocrine cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, neoplasms of the central nervous system (CNS), neuroectodermal cancers, spinal tumors, gliomas, meningiomas, and pituitary adenomas.
  • cancer also includes cancer metastasis.
  • Monoclonal antibodies may induce cell death via antibody-dependent cell-mediated cytotoxicity (ADCC), or may bind to complement proteins to induce direct cytotoxicity, known as complement-dependent cytotoxicity (CDC).
  • ADCC antibody-dependent cell-mediated cytotoxicity
  • CDC complement-dependent cytotoxicity
  • anti-cancer antibodies and potential antibody targets (in parentheses) include Abagovomab (CA-125), Abciximab (CD41), Adecatumumab (atumumab) (EpCAM), Afutuzumab (CD20), Alacizumab pegol (VEGFR2), Altumomab pentetate (CEA), Amatuximab (MORAb-009), Anatumomab mafenatox (TAG-72), Apolizumab (HLA-DR), Arcitumomab (CEA), Atezolizumab (Atezolizumab) (PD-L1), Bavituximab (phosphat
  • Wild-type C57BL/6 mice were supplied by Samtako Bio Korea (Osan, Korea).
  • Primary bone marrow-derived macrophages (BMDMs) were collected from mice and cultured in DMEM with M-CSF (R&D Systems, 416-ML) for 3–5 days.
  • HEK293T cells ATCC-11268; American Type Culture Collection
  • DMEM Gibco
  • FBS FBS
  • nonessential amino acids amino acids
  • sodium pyruvate sodium pyruvate
  • streptomycin 100 ⁇ g/mL
  • penicillin 100 IU/mL
  • LPS Escherichia coli O111:B4, tlrl-eblps
  • ATP adenosine 5'-triphosphate
  • nigericin adenosine 5'-triphosphate
  • DSS dextran sulfate sodium
  • Actin (I-19) ASC (N-15-R), IL-18 (H-173-Y), caspase-1 p10 (M-20), HA (12CA5), Flag (D-8), GST (B-14), His (H-3), and V5 (C-9) were purchased from Santa Cruz Biotechnology.
  • IL-1 ⁇ and NLRP3 were from R&D system and Adipogen, respectively.
  • Wild-type NLRP3 (Int. J. Mol. Sci. 2020, 21, 8437) is 1036 amino acids long, ⁇ PYD was 93-1036 in wtNLRP3, ⁇ NACHT was 1-219 and 537-1036 in wtNLRP3, and ⁇ LRR was 1-741 in wtNLRP3. Plasmids for expressing full-length (FL) NLRP3 and mutants ( ⁇ PYD, ⁇ NACHT, and ⁇ LRR) were inserted into the pcDNA3 vector.
  • Plasmids encoding different regions of Rv0747 (1-801, 1-93, 94-200, 201-300, 301-400, 401-500, 501-600, 601-700, 701-800) were amplified by PCR from FL Rv0747 cDNA and cloned downstream into a pEBG derivative encoding an N-terminal GST epitope tag flanked by BamHI and NotI sites.
  • Plasmids encoding different regions of ASC (1-195, 1-91, 107-195) were generated by PCR amplification from FL ASC cDNA and cloned downstream into a pEF derivative encoding a C-terminal Flag tag between the BamHI and NotI sites.
  • transient proteins encoded in the plasmids were produced in mammalian cells using the pEBG-GST mammalian fusion vector and the pEF-IRES-Puro expression vector. All constructs were verified to be 100% identical to the original sequence using an ABI PRISM 377 automated DNA sequencer.
  • NLRP3 or ASC peptides conjugated with 9R were provided by Peptron (Daejeon, Republic of Korea), manufactured and purified in acetate form to avoid undesirable intracellular reactions. Endotoxin levels were measured by Limulus amebocyte lysate test (Charles River Endosafe ®Endochrome-KTMUSA), and the concentrations of peptides used in the experiments were less than 3-5 pg/mL.
  • GST pull-down, western blot, and co-immunoprecipitation assays were performed with 293T and BMDM cells.
  • 293T cells were harvested and lysed in NP40 buffer containing a protease inhibitor cocktail (Roche, Basal, CH). After centrifugation, the supernatant was pretreated with protein A/G beads for 2 h at 4°C, and the pretreated lysate was combined with a 50% slurry of glutathione-conjugated Sepharose beads (Amersham Biosciences, Amersham, UK) and incubated for 4 h at 4°C. The precipitate was rinsed thoroughly with lysis buffer.
  • the proteins bound to the glutathione beads were eluted by boiling in sodium dodecyl sulfate (SDS) loading buffer for 5 min.
  • SDS sodium dodecyl sulfate
  • cells were harvested and lysed in NP40 buffer containing a protease inhibitor cocktail (Roche, Basal, CH).
  • NP40 buffer containing a protease inhibitor cocktail (Roche, Basal, CH).
  • Whole-cell lysates were pretreated with protein A/G agarose beads for 1 h at 4°C and then immunoprecipitated with the treated antibodies.
  • 1 mL of cell lysate was treated with 1–4 ⁇ g of antibody for 8–12 h at 4°C.
  • ASC is composed of a PYD domain that binds to NLRP3 and a CARD domain that binds the effector protein caspase-1.
  • GST-tagged Rv0747 GST-Rv0747
  • V5-tagged ASC WT PYD domain of ASC
  • CARD domain of ASC
  • Rv0747-derived peptide Rv0747 401 TLTGRPLIGNGANG 414
  • RRRRRRRRR cell-penetrating peptide R9
  • Flag-NLRP3 and V5-ASC were transfected into 293T cells, and the binding between NLRP3 and ASC was observed after treatment with the peptide in a concentration-dependent manner. As a result, the binding between NLRP3 and ASC was increased in a concentration-dependent manner of Rv0747 401 TLTGRPLIGNGANG 414 .
  • BMDMs bone marrow-derived macrophages

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Organic Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Biophysics (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Biochemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Immunology (AREA)
  • Epidemiology (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)

Abstract

The present invention relates to a polypeptide derived from Mycobacterium tuberculosis, or a variant thereof, and a pharmaceutical composition for treating cancer, the composition comprising same. The pharmaceutical composition according to the present invention specifically binds to ASC to induce multimerization of the ASC, thereby activating NLRP3 inflammasome, and thus can be applied as an anticancer agent.

Description

결핵균 유래 폴리펩타이드를 포함하는 암 치료용 약학 조성물Pharmaceutical composition for treating cancer comprising a polypeptide derived from Mycobacterium tuberculosis

본 발명은 ASC에 특이적으로 결합하는 결핵균 유래의 폴리펩타이드, 및 이를 포함하는 암 치료용 약학 조성물에 관한 것이다.The present invention relates to a tuberculosis-derived polypeptide that specifically binds to ASC, and a pharmaceutical composition for treating cancer comprising the same.

인간에서 200가지 이상의 암이 발견된 것으로 알려져 있다. 암으로 인해 전 세계적으로 매년 수백만명이 사망하며, 인구 노령화에 의해 암 발생률도 증가하는 추세이다. 항암제는 기술의 발전에 따라 그 형태가 변화되어 왔으며, 흔히 화학 항암제를 1세대 항암제로, 표적 항암제를 2세대 항암제로, 면역 항암제를 3세대 항암제로, 대사 항암제를 4세대 항암제 구분한다. 1세대 항암제는 세포 독성을 나타내는 물질 또는 생체 신호에 작용하는 물질을 투여함으로써 암 세포 또는 암 조직을 사멸시키는 원리로 작동하며, 대사저해제, 알킬화제, 백금화제, 항생제, 호르몬 요법제 등이 사용되는데, 정상 조직에도 작용할 수 있기 때문에 다양한 부작용이 발생되는 문제가 존재한다. 표적 항암제는 암세포에만 특이적으로 작용하게 함으로써 부작용을 최소화하고자 개발되었으나, 표적이 제한적이고, 치료제 내성이 발생되는 문제가 존재하였다. 이에 따라, 타고난 면역 작용을 개선시킴으로써 암세포를 제거하는 면역 항암제, 및 암세포에 영양 공급을 차단하는 등으로 암세포를 제거하는 대사 항암제 등이 제안되었다. It is known that more than 200 types of cancer have been discovered in humans. Millions of people die from cancer every year worldwide, and the incidence of cancer is increasing due to the aging population. The forms of anticancer drugs have changed with the advancement of technology, and are commonly classified as 1st generation anticancer drugs, 2nd generation anticancer drugs, 3rd generation anticancer drugs, and 4th generation anticancer drugs. 1st generation anticancer drugs operate on the principle of killing cancer cells or cancer tissues by administering substances that exhibit cytotoxicity or substances that act on biosignals, and metabolic inhibitors, alkylating agents, platinizing agents, antibiotics, and hormonal agents are used, but there is a problem that various side effects occur because they can also act on normal tissues. Targeted anticancer drugs were developed to minimize side effects by acting specifically on cancer cells, but there were problems with limited targets and the occurrence of drug resistance. Accordingly, immunotherapy drugs that remove cancer cells by improving innate immune function, and metabolic anticancer drugs that remove cancer cells by blocking the supply of nutrients to cancer cells, etc. have been proposed.

ASC 또는 PYCARD 단백질은 화학 항암제가 처리된 인간 백혈병 세포에서 응집체 형태로 발견되었다. 많은 종양 환자에서 ASC가 메틸레이션되어 세포 사멸 및 종양 억제자로서의 ASC의 역할이 억제되는 것으로 알려졌다. 결과적으로, ASC는 염증성 사이토카인인 IL-1β 및 IL-18의 분비를 매개하는 NLRP3 인플라마좀 복합체 형성에 중요한 단백질인 것으로 확인된 바 있다. ASC or PYCARD protein has been found in aggregate form in human leukemia cells treated with chemotherapy. It has been reported that ASC is methylated in many tumor patients, which inhibits the role of ASC as a cell death and tumor suppressor. Consequently, ASC has been identified as a protein important for the formation of NLRP3 inflammasome complex, which mediates the secretion of inflammatory cytokines IL-1β and IL-18.

한편, 인간의 NLRP3 인플라마좀 신호는 유전자 발현에 영향을 미치며 궁극적으로 활성화로 이어질 수 있는 유전적 다형성 및 돌연변이와 같은 다양한 요인에 의해 제어된다. 이러한 효과는 염증성 질환 환자에서 발견되었다(erma D, Saerndahl E, Andersson H, Eriksson P, Fredrikson M, Joensson JI, et al. The Q705K polymorphism in NLRP3 is a gain-of-function alteration leading to excessive interleukin-1β and L-18 roduction. LoS ne. 2012) :e34977.; Touitou I, Lesage S, McDermott M, Cuisset L, Hoffman H, Dode C, et al. Infevers: an evolving mutation database for auto-inflammatory syndromes. Hum Mutat. (2004) 24:194-8.; Levy R, Geerard L, Kuemmerle-Deschner J, Lachmann HJ, Konee-Paut I, Cantarini L, et al. Phenotypic and genotypic characteristics of cryopyrin-associated periodic syndrome: a series of 136 patients from the Eurofever Registry. Ann Rheum Dis. (2015) 74:2043-9.; Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. (2001) 29:301-5.). 유사하게, NLRP3 인플라마좀과 관련된 유전적 다형성도 암과 관련이 있다. 예를 들어, NLRP3 유전자의 SNP(single-nucleotide polymorphism), Q705K(rs35829419)는 침윤성 결장직장암 환자의 생존율 저하와 관련이 있고(Ungerbaeck J, Belenki D, Jawad ul-Hassan A, Fredrikson M, Franseen K, Elander N, et al. Genetic variation and alterations of genes involved in NFκB/TNFAIP3- nd LRP3-inflammasome ignaling ffect usceptibility nd utcome f olorectal ancer. arcinogenesis. 2012) 3:2126-34), 스웨덴 남성의 산발성 전이성 흑색종의 위험 대립 유전자로 추정되었으며(Verma D, Bivik C, Farahani E, Synnerstad I, Fredrikson M, Enerbaeck C, et al. Inflammasome polymorphisms confer susceptibility to sporadic malignant melanoma. Pigment Cell Melanoma Res. (2012) 25:506-13), 췌장암 환자에서도 높은 빈도로 발생하였다 (Miskiewicz A, Szparecki G, Durlik M, Rydzewska G, Ziobrowski I, Goerska R. The Q705K and F359L single-nucleotide polymorphisms of NOD-like receptor signaling pathway: association with chronic pancreatitis, pancreatic cancer, and periodontitis. Arch Immunol Ther Exp (Warsz). (2015) 63:485-94). 또한 NLRP3 다형성(rs10754558 및 rs4612666)을 가진 사람들은 Helicobacter pylori에 감염되었을 때 위암에 더 취약한 것으로 나타났다(Castano-Rodrlguez N, Kaakoush NO, Goh KL, Fock KM, Mitchell HM. The NOD-like receptor signalling pathway in Helicobacter pylori infection and related gastric cancer: a case-control study and gene expression analyses. PLoS One. (2014) 9:e98899). 혈액 악성 종양에서 IL-1β및 IL-18에만 국한된 다형성은 AML 및 만성 골수성 백혈병(CML)의 임상 및 병태생리학적 특성과 관련이 있는 것으로 나타났다(Zhang A, Yu J, Yan S, Zhao X, Chen C, Zhou Y, et al. The genetic polymorphism and expression profiles of NLRP3 inflammasome in patients with chronic myeloid leukemia. Hum Immunol. (2018) 79:57-62; Wang H, Hua M, Wang S, Yu J, Chen C, Zhao X, et al. Genetic polymorphisms of IL-18 rs1946518 and IL-1βrs16944 are associated with prognosis and survival of acute myeloid leukemia. Inflamm Res. (2017) 66:249-58). 또한, 유전자 발현 프로파일링을 활용한 연구에서는 여러 암에서 NLRP3 인플라마좀의 상향 조절을 보고하였다. 예를 들어, NLRP3는 정상 조직에 비해 HNSCC, LSCC 및 편평 세포 암종 조직에서 과발현되며 종종 불량한 예후 및 병리와 관련이 있고(Chen L, Huang CF, Li YC, Deng WW, Mao L, Wu L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cell Mol Life Sci. (2018) 75:2045-58; Xue Y, Du H-D, Tang D, Zhang D, Zhou J, Zhai C-W, et al. Correlation between the NLRP3 inflammasome and the prognosis of patients with LSCC. Front Oncol. (2019) 9:588; Wu CS, Chang KP, OuYang CN, Kao HK, Hsueh C, Chen LC, et al. ASC contributes to metastasis of oral cavity squamous cell carcinoma. Oncotarget. (2016) 7:50074-85), 방광암에서는 NLRP3 인플라마좀의 높은 발현도 발견되어 진단을 위한 잠재적인 바이오마커가 될 수 있음을 확인하였다(Poli G, Brancorsini S, Cochetti G, Barillaro F, Egidi MG, Mearini E. Expression of inflammasome-related genes in bladder cancer and their association with cytokeratin 20 messenger RNA. Urol Oncol Semin Orig Investig. (2015) 33:505.e1-e7). Meanwhile, human NLRP3 inflammasome signaling is controlled by various factors such as genetic polymorphisms and mutations that can affect gene expression and ultimately lead to activation. Such effects have been found in patients with inflammatory diseases (erma D, Saerndahl E, Andersson H, Eriksson P, Fredrikson M, Joensson JI, et al. The Q705K polymorphism in NLRP3 is a gain-of-function alteration leading to excessive interleukin-1β and L-18 roduction. LoS ne. 2012) :e34977.; Touitou I, Lesage S, McDermott M, Cuisset L, Hoffman H, Dode C, et al. Infevers: an evolving mutation database for auto-inflammatory syndromes. Hum Mutat. (2004) 24:194-8.; Levy R, Geerard L, Kuemmerle-Deschner J, Lachmann HJ, Konee-Paut I, Cantarini L, et al. Phenotypic and genotypic characteristics of cryopyrin-associated periodic syndrome: a series of 136 patients from the Eurofever Registry. Ann Rheum Dis. (2015) 74:2043-9.; Hoffman HM, Mueller JL, Broide DH, Wanderer AA, Kolodner RD. Mutation of a new gene encoding a putative pyrin-like protein causes familial cold autoinflammatory syndrome and Muckle-Wells syndrome. Nat Genet. (2001) 29:301-5.). Similarly, genetic polymorphisms involving the NLRP3 inflammasome have also been associated with cancer. For example, a single-nucleotide polymorphism (SNP) in the NLRP3 gene, Q705K (rs35829419), was associated with poor survival in patients with invasive colorectal cancer (Ungerbaeck J, Belenki D, Jawad ul-Hassan A, Fredrikson M, Franseen K, Elander N, et al. Genetic variation and alterations of genes involved in NFκB/TNFAIP3- and LRP3-inflammasome ignaling effect usceptibility nd utcome f olorectal ancer. arcinogenesis. 2012) 3:2126-34) and was suggested to be a risk allele for sporadic metastatic melanoma in Swedish men (Verma D, Bivik C, Farahani E, Synnerstad I, Fredrikson M, Enerbaeck C, et al. Inflammasome polymorphisms confer susceptibility to sporadic malignant melanoma. Pigment Cell Melanoma Res. (2012) 25:506-13), and also occurred at a high frequency in patients with pancreatic cancer (Miskiewicz A, Szparecki G, Durlik M, Rydzewska G, Ziobrowski I, Goerska R. The Q705K and F359L single-nucleotide polymorphisms of NOD-like receptor signaling pathway: association with chronic pancreatitis, pancreatic cancer, and periodontitis. Arch Immunol Ther Exp (Warsz). (2015) 63:485-94). Additionally, individuals with NLRP3 polymorphisms (rs10754558 and rs4612666) were found to be more susceptible to gastric cancer when infected with Helicobacter pylori (Castano-Rodrlguez N, Kaakoush NO, Goh KL, Fock KM, Mitchell HM. The NOD-like receptor signalling pathway in Helicobacter pylori infection and related gastric cancer: a case-control study and gene expression analyses. PLoS One. (2014) 9:e98899). In hematological malignancies, polymorphisms limited to IL-1β and IL-18 have been associated with the clinical and pathophysiological characteristics of AML and chronic myeloid leukemia (CML) (Zhang A, Yu J, Yan S, Zhao X, Chen C, Zhou Y, et al. The genetic polymorphism and expression profiles of NLRP3 inflammasome in patients with chronic myeloid leukemia. Hum Immunol. (2018) 79:57-62; Wang H, Hua M, Wang S, Yu J, Chen C, Zhao X, et al. Genetic polymorphisms of IL-18 rs1946518 and IL-1βrs16944 are associated with prognosis and survival of acute myeloid leukemia. Inflamm Res. (2017) 66:249-58). In addition, studies utilizing gene expression profiling have reported the upregulation of NLRP3 inflammasome in several cancers. For example, NLRP3 is overexpressed in HNSCC, LSCC, and squamous cell carcinoma tissues compared with normal tissues and is often associated with poor prognosis and pathology (Chen L, Huang CF, Li YC, Deng WW, Mao L, Wu L, et al. Blockage of the NLRP3 inflammasome by MCC950 improves anti-tumor immune responses in head and neck squamous cell carcinoma. Cell Mol Life Sci. (2018) 75:2045-58; Xue Y, Du H-D, Tang D, Zhang D, Zhou J, Zhai C-W, et al. Correlation between the NLRP3 inflammasome and the prognosis of patients with LSCC. Front Oncol. (2019) 9:588; Wu CS, Chang KP, OuYang CN, Kao HK, Hsueh C, Chen LC, et al. ASC contributes to metastasis of oral cavity squamous cell carcinoma. Oncotarget. (2016) 7:50074-85), and high expression of NLRP3 inflammasome was also found in bladder cancer, confirming that it could be a potential biomarker for diagnosis (Poli G, Brancorsini S, Cochetti G, Barillaro F, Egidi MG, Mearini E. Expression of inflammasome-related genes in bladder cancer and their association with cytokeratin 20 messenger RNA. Urol Oncol Semin Orig Investig. (2015) 33:505.e1-e7).

본 발명은 NLRP3 인플라마좀을 활성화시킴으로써 암을 치료할 수 있는 폴리펩타이드를 제공하는 것을 목적으로 한다. The present invention aims to provide a polypeptide capable of treating cancer by activating NLRP3 inflammasome.

본 발명은 ASC (Apoptosis-associated Speck-like protein containing a CARD)에 특이적으로 결합하는, 서열번호 1로 표시되는 서열을 포함하는 폴리펩타이드 또는 이의 변이체를 제공한다. 본 발명의 일 실시예에서, 상기 폴리펩타이드는 서열번호 1로 표시되는 서열로 구성되고, 본 발명의 다른 실시예에서, 상기 변이체는 서열번호 1의 2번째, 7번째, 10번째, 및 13번째 아미노산을 제외한 위치에서 보존적 치환을 포함할 수 있으며, 본 발명의 또 다른 실시예에서, 상기 폴리펩타이드는 서열번호 2로 표시되는 서열을 포함하고, 본 발명의 또 다른 실시예에서, 상기 보존적 치환은 T1S, T3S, G4A, R5K, I8V, G9A, G11A, A12G, 및 G14A로 구성된 군으로부터 선택된 적어도 하나, 또는 T1S, T3S, G4A, R5K, I8L, G9A, G11A, A12G, 및 G14A로 구성된 군으로부터 선택된 적어도 하나이다. The present invention provides a polypeptide comprising a sequence represented by SEQ ID NO: 1 or a variant thereof, which specifically binds to ASC (Apoptosis-associated Speck-like protein containing a CARD). In one embodiment of the present invention, the polypeptide consists of the sequence represented by SEQ ID NO: 1, and in another embodiment of the present invention, the variant may comprise conservative substitutions at positions other than the 2nd, 7th, 10th, and 13th amino acids of SEQ ID NO: 1, and in yet another embodiment of the present invention, the polypeptide comprises the sequence represented by SEQ ID NO: 2, and in yet another embodiment of the present invention, the conservative substitutions are at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8V, G9A, G11A, A12G, and G14A, or at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8L, G9A, G11A, A12G, and G14A.

본 발명에서는 상기의 폴리펩타이드 또는 이의 변이체를 포함하는, ASC 특이적 결합 분자를 제공한다. The present invention provides an ASC-specific binding molecule comprising the above-mentioned polypeptide or a variant thereof.

본 발명에서 상기의 폴리펩타이드 또는 변이체를 포함하는, 암 치료용 약학 조성물을 제공한다. 본 발명의 일 실시예에서, 상기 암은 염증성 대장암, 결장직장암, 흑색종, 또는 간세포암일 수 있다.The present invention provides a pharmaceutical composition for treating cancer, comprising the above-described polypeptide or variant. In one embodiment of the present invention, the cancer may be inflammatory colon cancer, colorectal cancer, melanoma, or hepatocellular carcinoma.

본 발명에서 상기의 폴리펩타이드 또는 변이체를 발현하는 핵산 분자를 제공한다. The present invention provides a nucleic acid molecule expressing the above polypeptide or variant.

본 발명에서는 상기의 핵산 분자를 포함하는, 암 치료용 약학 조성물을 제공한다.The present invention provides a pharmaceutical composition for treating cancer, comprising the nucleic acid molecule described above.

본 발명에서는 상기의 핵산 분자를 포함하는, 재조합 벡터를 제공한다. The present invention provides a recombinant vector comprising the nucleic acid molecule described above.

본 발명에서는 상기의 핵산 분자를 포함하는, 숙주 세포를 제공한다.The present invention provides a host cell comprising the nucleic acid molecule described above.

본 발명에 따른 폴리펩타이드는 ASC 단백질에 특이적으로 결함함으로써 ASC의 다량체화를 유도하고, 이를 통해 NLRP3 인플라마좀을 활성화한다. 따라서 본 발명에 따른 폴리펩타이드, 또는 이의 변이체, 또는 이를 발현하는 핵산 분자는 항암제로서 이용 가능하다. The polypeptide according to the present invention specifically binds to the ASC protein, thereby inducing multimerization of ASC, thereby activating the NLRP3 inflammasome. Therefore, the polypeptide according to the present invention, or a variant thereof, or a nucleic acid molecule expressing the same, can be used as an anticancer agent.

도 1a는 Rv0747/PE_PGRS10 단백질이 ASC와 상호작용함을 확인한 결과이고, 도 1b는 Rv0747/PE_PGRS10 단백질과 ASC의 상호작용하는 도메인을 확인한 결과, 도 1c는 ASC와 상호작용하는 Rv0747/PE_PGRS10 단백질의 서열을 확인한 결과, 및 도 1d는 ASC와 상호작용하는 14개의 아미노산으로 구성된 펩타이드의 서열을 나타낸 것이다. Figure 1a shows the results of confirming that the Rv0747/PE_PGRS10 protein interacts with ASC, Figure 1b shows the results of confirming the interacting domain of the Rv0747/PE_PGRS10 protein and ASC, Figure 1c shows the results of confirming the sequence of the Rv0747/PE_PGRS10 protein interacting with ASC, and Figure 1d shows the sequence of a peptide consisting of 14 amino acids interacting with ASC.

도 2a는 본 발명에 따른 펩타이드를 처리함으로써 NLRP3와 ASC의 결합이 증가함을 확인한 결과, 도 2b는 본 발명에 따른 펩타이드를 처리함으로써 IL-1β 및 IL-18의 분비를 증가시킴을 확인한 결과, 도 2c는 본 발명에 따른 펩타이드를 처리함으로써 caspse-1 분비를 증가시킴을 확인한 결과, 도 2d는 본 발명에 따른 펩타이드를 처리함으로써 ASC의 올리고머화가 증가된 결과, 및 E는 본 발명에 따른 펩타이드를 처리함으로써 세포 내 올리고머화된 ASC의 비율이 증가함을 확인한 결과이다. FIG. 2a shows the results confirming that the binding of NLRP3 and ASC increases by treating with the peptide according to the present invention, FIG. 2b shows the results confirming that the secretion of IL-1β and IL-18 increases by treating with the peptide according to the present invention, FIG. 2c shows the results confirming that the secretion of caspse-1 increases by treating with the peptide according to the present invention, FIG. 2d shows the results confirming that the oligomerization of ASC increases by treating with the peptide according to the present invention, and E shows the results confirming that the ratio of intracellular oligomerized ASC increases by treating with the peptide according to the present invention.

도 3은 본 발명의 일 실시예에 따른 펩타이드와 ASC의 결합에 관여하는 아미노산을 확인한 결과이다. Figure 3 shows the results of confirming amino acids involved in the binding of a peptide and ASC according to one embodiment of the present invention.

이하, 첨부된 도면을 참조하여 본 발명의 구현예로 본 발명을 상세히 설명하기로 한다. 다만, 하기 구현예는 본 발명에 대한 예시로 제시되는 것으로, 당업자에게 주지 저명한 기술 또는 구성에 대한 구체적인 설명이 본 발명의 요지를 불필요하게 흐릴 수 있다고 판단되는 경우에는 그 상세한 설명을 생략할 수 있고, 이에 의해 본 발명이 제한되지는 않는다. 본 발명은 후술하는 특허청구범위의 기재 및 그로부터 해석되는 균등 범주 내에서 다양한 변형 및 응용이 가능하다. Hereinafter, the present invention will be described in detail with reference to the attached drawings as embodiments of the present invention. However, the following embodiments are presented as examples of the present invention, and if it is judged that a detailed description of a technology or configuration well known to those skilled in the art may unnecessarily obscure the gist of the present invention, the detailed description thereof may be omitted, and the present invention is not limited thereby. The present invention is capable of various modifications and applications within the scope of the following claims and equivalents interpreted therefrom.

또한, 본 명세서에서 사용되는 용어(terminology)들은 본 발명의 바람직한 실시예를 적절히 표현하기 위해 사용된 용어들로서, 이는 사용자, 운용자의 의도 또는 본 발명이 속하는 분야의 관례 등에 따라 달라질 수 있다. 따라서, 본 용어들에 대한 정의는 본 명세서 전반에 걸친 내용을 토대로 내려져야 할 것이다. 명세서 전체에서, 어떤 부분이 어떤 구성요소를 "포함"한다고 할 때, 이는 특별히 반대되는 기재가 없는 한 다른 구성요소를 제외하는 것이 아니라 다른 구성 요소를 더 포함할 수 있는 것을 의미한다.In addition, the terminology used in this specification is a terminology used to appropriately express the preferred embodiment of the present invention, and this may vary depending on the intention of the user or operator, or the custom of the field to which the present invention belongs. Therefore, the definition of these terms should be determined based on the contents throughout this specification. Throughout the specification, when a part is said to "include" a certain component, this does not mean that other components are excluded, but rather that other components can be included, unless specifically stated otherwise.

본 발명에서 사용되는 모든 기술용어는, 달리 정의되지 않는 이상, 본 발명의 관련 분야에서 통상의 당업자가 일반적으로 이해하는 바와 같은 의미로 사용된다. 또한 본 명세서에는 바람직한 방법이나 시료가 기재되나, 이와 유사하거나 동등한 것들도 본 발명의 범주에 포함된다. 본 명세서에 참고문헌으로 기재되는 모든 간행물의 내용은 본 발명에 통합된다.All technical terms used in the present invention, unless otherwise defined, are used with the same meaning as commonly understood by those skilled in the art in the relevant field of the present invention. In addition, although preferred methods or samples are described in this specification, similar or equivalent ones are also included in the scope of the present invention. The contents of all publications mentioned as references in this specification are incorporated into the present invention.

NLRP3 인플라마좀(inflammasome)은 선천적 면역 체계와 염증 신호를 조절하는 데에 중추적인 역할을 하는 다중 단백질 복합체이다. PAMP (pathogen-associated molecular pattern) 및 DAMP (damage-associated molecular pattern)에 의해 활성화되면, NLRP3는 올리고머화되어 전 염증성 사이토카인인 IL-1β 및 IL-18의 처리 및 방출을 시작시키는 caspase-1을 활성화시킨다. The NLRP3 inflammasome is a multiprotein complex that plays a central role in regulating the innate immune system and inflammatory signaling. Upon activation by pathogen-associated molecular patterns (PAMPs) and damage-associated molecular patterns (DAMPs), NLRP3 oligomerizes and activates caspase-1, which initiates the processing and release of the proinflammatory cytokines IL-1β and IL-18.

NLRP3 인플라마좀은 항-종양 활성을 갖는다. NLRP3 인플라마좀이 종양 세포에 대한 IFN-γ생성 T 림프구의 수지상 세포 매개 프라이밍에 필요하다고 보고하였다 (Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β-dependent adaptive immunity against tumors. Nat Med. (2009) 15:1170-8.). NLRP3 inflammasome은 대장염 관련 암에서 종양 형성의 음성 조절자 역할을 하는 것으로 보이며(Allen IC, TeKippe EM, Woodford R-MT, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. (2010) 207:1045-56.), 이는 장의 항상성 조절 및 결과적으로 대장염에 대한 보호에서 NLRP3 inflammasome이 중요한 역할을 한다는 사실이 보고되었다 (Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti TD. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity. (2010) 32:379-91). 또한, NLRP3 인플라마좀이 결핍된 경우, 결장직장암에서 종양을 증가시키는 것으로 나타났다. 또한, NLRP3 inflammasome 매개 IL-18 생성이 간에서 결장직장암 전이 성장을 억제한다고 보고된 바 있다 (Dupaul-Chicoine J, Arabzadeh A, Dagenais M, Douglas T, Champagne C, Morizot A, et al. The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity. (2015) 43:751-63.). NLRP3/IL-18 매개 IL-22BP의 하향 조절은 염증 피크 동안 장 조직 손상에 대한 보호 역할을 제공한다 (Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. (2012) 491:259-63.). 흑색종에서 TME의 NLRP3는 골수 유래 억제 세포(MDSC)의 이동을 지원하여 T 세포 반응을 억제함으로써 암 백신에 대한 항종양 면역 반응을 약화시키는 것으로 나타났다(Van Deventer HW, Burgents JE, Wu QP, Woodford RMT, Brickey WJ, Allen IC, et al. The inflammasome component Nlrp3 impairs antitumor vaccine by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells. Cancer Res. (2010) 70:10161-9.).The NLRP3 inflammasome has anti-tumor activity. It has been reported that the NLRP3 inflammasome is required for dendritic cell-mediated priming of IFN-γ-producing T lymphocytes against tumor cells (Ghiringhelli F, Apetoh L, Tesniere A, Aymeric L, Ma Y, Ortiz C, et al. Activation of the NLRP3 inflammasome in dendritic cells induces IL-1β-dependent adaptive immunity against tumors. Nat Med. (2009) 15:1170-8.). The NLRP3 inflammasome appears to act as a negative regulator of tumorigenesis during colitis-associated cancer (Allen IC, TeKippe EM, Woodford R-MT, Uronis JM, Holl EK, Rogers AB, et al. The NLRP3 inflammasome functions as a negative regulator of tumorigenesis during colitis-associated cancer. J Exp Med. (2010) 207:1045-56.), suggesting a critical role for the NLRP3 inflammasome in regulating intestinal homeostasis and consequently in protection against colitis (Zaki MH, Boyd KL, Vogel P, Kastan MB, Lamkanfi M, Kanneganti TD. The NLRP3 inflammasome protects against loss of epithelial integrity and mortality during experimental colitis. Immunity. (2010) 32:379-91). Furthermore, NLRP3 inflammasome deficiency has been shown to increase tumorigenesis in colorectal cancer. In addition, NLRP3 inflammasome-mediated IL-18 production has been reported to suppress colorectal cancer metastatic growth in the liver (Dupaul-Chicoine J, Arabzadeh A, Dagenais M, Douglas T, Champagne C, Morizot A, et al. The Nlrp3 inflammasome suppresses colorectal cancer metastatic growth in the liver by promoting natural killer cell tumoricidal activity. Immunity. (2015) 43:751-63.). NLRP3/IL-18-mediated downregulation of IL-22BP provides a protective role against intestinal tissue damage during peak inflammation (Huber S, Gagliani N, Zenewicz LA, Huber FJ, Bosurgi L, Hu B, et al. IL-22BP is regulated by the inflammasome and modulates tumorigenesis in the intestine. Nature. (2012) 491:259-63.). In melanoma, NLRP3 in the TME has been shown to attenuate antitumor immune responses to cancer vaccines by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells (MDSCs) and suppressing T cell responses (Van Deventer HW, Burgents JE, Wu QP, Woodford RMT, Brickey WJ, Allen IC, et al. The inflammasome component Nlrp3 impairs antitumor vaccine by enhancing the accumulation of tumor-associated myeloid-derived suppressor cells. Cancer Res. (2010) 70:10161-9.).

본 발명은 ASC(Apoptosis-associated Speck-like protein containing a CARD)에 특이적으로 결합하며, NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) 인플라마좀(inflammasome)을 활성화시킴으로써 항암제로서 사용 가능한 결핵균 (Mycobacterium tuberculosis) 유래의 펩타이드를 제공한다. 더욱 구체적으로, 본 발명에 따른 펩타이드는 결핵균의 Rv0747/PE_PGRS10 (서열번호 2)에서 유래된 것이다. 야생형 Rv0747/PE_PGRS10은 801개의 아미노산으로 구성되어 있다. 본 발명에서는 ASC에 특이적으로 결합하는, Rv0747/PE_PGRS10의 401번째 아미노산부터 414번째 아미노산으로 구성된, TLTGRPLIGNGANG 서열(서열번호 1)의 펩타이드를 제공한다. 전장 Rv0747/PE_PGRS10를 인코딩하는 DNA 서열은 서열번호 4로, 서열번호 2를 인코딩하는 DNA 서열은 서열번호 3으로 표시한다. The present invention provides a peptide derived from Mycobacterium tuberculosis that can be used as an anticancer agent by specifically binding to ASC (Apoptosis-associated Speck-like protein containing a CARD) and activating NLRP3 (NOD-, LRR- and pyrin domain-containing protein 3) inflammasome. More specifically, the peptide according to the present invention is derived from Rv0747/PE_PGRS10 (SEQ ID NO: 2) of Mycobacterium tuberculosis. Wild-type Rv0747/PE_PGRS10 consists of 801 amino acids. The present invention provides a peptide having a TLTGRPLIGNGANG sequence (SEQ ID NO: 1) consisting of the 401st to 414th amino acids of Rv0747/PE_PGRS10, which specifically binds to ASC. The DNA sequence encoding the full-length Rv0747/PE_PGRS10 is represented by SEQ ID NO: 4, and the DNA sequence encoding the full-length Rv0747/PE_PGRS10 is represented by SEQ ID NO: 2.

ASC(NP_037390)는 PYCARD로도 명명되며, N-말단의 PYD (PYRIN-PAAD-DAPIN) 도메인 및 C-말단의 CARD (caspase-recruitment domain) 도메인으로 구성된다. PYD 및 CARD 도메인은 caspase의 활성화를 통해 염증 및 세포사멸 신호 경로에서 거대한 신호 전달 복합체의 조립을 매개하는 6-나선 번들 사멸 도메인 접힘 수퍼패밀리의 구성원이다. 정상 세포에서 이 단백질은 세포질에 위치되어 있으나, 세포 사멸을 겪는 세포에서는 핵 주변 근처에서 공 모양의 응집체를 형성한다. ASC 단백질은 195개의 아미노산으로 구성되어 있다. ASC (NP_037390), also named PYCARD, consists of an N-terminal PYD (PYRIN-PAAD-DAPIN) domain and a C-terminal CARD (caspase-recruitment domain) domain. The PYD and CARD domains are members of the six-helix bundle death domain fold superfamily, which mediates the assembly of large signaling complexes in inflammatory and apoptotic signaling pathways through caspase activation. In normal cells, this protein is located in the cytoplasm, but in cells undergoing apoptosis, it forms globular aggregates near the perinuclear region. The ASC protein consists of 195 amino acids.

본 발명에 따른 상기의 펩타이드가 ASC에 결합되면, ASC는 다량체화 또는 폴리머화되고, 이는 NLRP3 인플라마좀의 활성화를 유도하고, IL-1β 및/또는 IL-18, 및 caspase-1의 분비를 특이적으로 증가시킴으로써 암 세포의 사멸을 유도한다. When the above peptide according to the present invention binds to ASC, ASC is multimerized or polymerized, which induces activation of NLRP3 inflammasome and specifically increases the secretion of IL-1β and/or IL-18 and caspase-1, thereby inducing the death of cancer cells.

아미노산 서열 (펩타이드 또는 단백질)과 관련하여, "단편"은 아미노산 서열의 일부를 의미하며, 즉 N-말단 및/또는 C-말단에서 짧아진 아미노산 서열을 나타내는 서열을 의미한다. C-말단에서 짧아진 단편 (N-말단 단편)은, 예를 들어, 오픈 리딩 프레임의 3'-말단이 결핍된 절단된 오픈 리딩 프레임을 번역함으로써, 수득 가능하다. N-말단에서 짧아진 단편 (C-말단 단편)은, 절단된 오픈 리딩 프레임이 번역을 개시하는 역할을 하는 개시 코돈을 포함하는 한, 예를 들어, 오픈 리딩 프레임의 5'-말단이 결핍된 절단된 오픈 리딩 프레임을 번역함으로써, 수득 가능하다. 아미노산 서열의 단편은, 예를 들어, 아미노산 서열로부터 적어도 50%, 적어도 60%, 적어도 70%, 적어도 80%, 적어도 90%의 아미노산 잔기들을 포함한다. 아미노산 서열의 단편은 바람직하게는 아미노산 서열로부터 적어도 6개, 특히 적어도 8개, 적어도 12개, 적어도 13개, 적어도 14개, 적어도 15개, 적어도 20개, 적어도 30개, 적어도 50개 또는 적어도 100개의 연속적인 아미노산을 포함한다.In relation to an amino acid sequence (peptide or protein), "fragment" means a part of the amino acid sequence, i.e. a sequence which represents an amino acid sequence that is shortened at the N-terminus and/or the C-terminus. A fragment shortened at the C-terminus (N-terminal fragment) can be obtained, for example, by translating a truncated open reading frame lacking the 3'-end of the open reading frame. A fragment shortened at the N-terminus (C-terminal fragment) can be obtained, for example, by translating a truncated open reading frame lacking the 5'-end of the open reading frame, as long as the truncated open reading frame comprises an initiation codon which serves to initiate translation. A fragment of an amino acid sequence comprises, for example, at least 50%, at least 60%, at least 70%, at least 80%, at least 90% of the amino acid residues from the amino acid sequence. The fragment of the amino acid sequence preferably comprises at least 6, in particular at least 8, at least 12, at least 13, at least 14, at least 15, at least 20, at least 30, at least 50 or at least 100 consecutive amino acids from the amino acid sequence.

폴리펩타이드 또는 단백질의 "변형체(variant)", "변이체" 또는 "변이체 단백질" 또는 "변이체 폴리펩타이드"는 폴리펩타이드 또는 단백질로부터 유래하고 상기 폴리펩타이드 또는 단백질의 적어도 하나의 생물학적 특성을 유지하고 있는 임의의 유사체, 단편, 유도체 또는 돌연변이를 지칭한다. 상기 폴리펩타이드 또는 단백질의 상이한 변형체들이 자연 내에 존재할 수 있다. 이들 변형체들은 단백질을 암호화하는 구조 유전자의 뉴클레오타이드 서열이 다른 것에 특징이 있는 대립 유전자의 변이일 수 있거나, 차별적 스플라이싱 또는 번역 후 수식(modification)을 포함할 수 있다. 숙련된 당업자는 하나 또는 복수의 아미노산 치환, 결손, 부가 또는 대체를 갖는 변이체를 생성할 수 있다. 이들 변이체들은 그중에서도: (a) 하나 이상의 아미노산 잔기들이 보존적 또는 비보존적 아미노산들로 치환된 변이체, (b) 하나 이상의 아미노산이 폴리펩타이드 또는 단백질에 부가된 변이체, (c) 아미노산 중 하나 이상이 치환기를 포함하는 변이체, 및 (d) 폴리펩타이드 또는 단백질이 혈청 알부민과 같은 또 다른 폴리펩타이드와 융합된 변이체를 포함할 수 있다. A "variant", "variant", "variant protein", or "variant polypeptide" of a polypeptide or protein refers to any analog, fragment, derivative, or mutant derived from the polypeptide or protein and which retains at least one biological property of the polypeptide or protein. Different variants of the polypeptide or protein may exist in nature. These variants may be allelic mutations characterized by differences in the nucleotide sequence of the structural gene encoding the protein, or may include differential splicing or post-translational modifications. One skilled in the art can generate variants having one or more amino acid substitutions, deletions, additions, or replacements. These variants may include, inter alia: (a) variants in which one or more amino acid residues are substituted with conservative or non-conservative amino acids, (b) variants in which one or more amino acids are added to the polypeptide or protein, (c) variants in which one or more of the amino acids comprises a substitution group, and (d) variants in which the polypeptide or protein is fused to another polypeptide, such as serum albumin.

또한 보존적 변이체는 단백질의 생물학적 기능에 불리한 영향을 미치지 않는 서열 변경을 가진 아미노산 서열을 의미한다. 변경된 서열이 단백질과 관련된 생물학적 기능을 방해하거나 파괴하는 경우, 치환, 삽입 또는 결실이 단백질에 불리한 영향을 미친다고 기재된다. 예를 들어, 단백질의 총 전하, 구조 또는 소수성-친수성은 생물학적 활성에 불리한 영향을 미치지 않으면서 변경될 수 있다. 따라서, 아미노산 서열은 예를 들어, 단백질의 생물학적 활성에 불리한 영향을 미치지 않으면서 펩타이드가 보다 높은 소수성 또는 친수성을 나타내도록 변경될 수 있다. 유전적(억제, 결손, 돌연변이 등), 화학적 및 효소적 기술들을 포함하는, 이러한 변형체를 수득하는 기술들이 본 기술분야에서 통상의 기술을 가진 자에게 알려져 있다.Conservative variants also refer to amino acid sequences having sequence changes that do not adversely affect the biological function of the protein. A substitution, insertion, or deletion is described as adversely affecting the protein if the altered sequence disrupts or destroys the biological function associated with the protein. For example, the overall charge, structure, or hydrophobic-hydrophilicity of a protein can be altered without adversely affecting the biological activity. Thus, for example, the amino acid sequence can be altered to render the peptide more hydrophobic or more hydrophilic without adversely affecting the biological activity of the protein. Techniques for obtaining such variants, including genetic (suppression, deletion, mutation, etc.), chemical, and enzymatic techniques, are known to those skilled in the art.

본원에서 사용된 것과 같은 "모 폴리펩타이드", "모 단백질", "전구체 폴리펩타이드" 또는 "전구체 단백질"은 향후 변형되어 변이체를 만드는, 변형되지 않은 폴리펩타이드를 의미한다. 모 폴리펩타이드는 야생형 폴리펩타이드, 또는 야생형 폴리펩타이드의 변이체 또는 조작된 버전일 수 있다.As used herein, the terms "parent polypeptide", "parent protein", "precursor polypeptide" or "precursor protein" refer to an unmodified polypeptide that is later modified to form a variant. The parent polypeptide can be a wild-type polypeptide, or a variant or engineered version of a wild-type polypeptide.

본원에서 "야생형" 또는 "WT" 또는 "천연 (native)"은 대립유전자 변형을 비롯하여, 자연계에서 발견되는 아미노산 서열을 의미한다. 야생형 단백질 또는 폴리펩타이드는 의도적으로 변형되지 않은 아미노산 서열을 가진다.As used herein, "wild type" or "WT" or "native" refers to an amino acid sequence found in nature, including allelic variants. A wild type protein or polypeptide has an amino acid sequence that has not been intentionally modified.

본 명세서의 목적에서, 아미노산 서열 (펩타이드, 단백질 또는 폴리펩타이드)의 "변이체"는 아미노산 삽입 변이체, 아미노산 부가 변이체, 아미노산 결손 변이체 및/또는 아미노산 치환 변이체를 포함한다. 용어 "변이체"는 모든 스플라이스 변이체, 번역 후 변형된 변이체, 입체구조 (conformations), 이소형 및 종 상동체 (species homolog), 특히 세포에 의해 천연적으로 발현되는 것을 포함한다. 용어 “변이체”는, 특히, 아미노산 서열의 단편을 포함한다. For the purposes of this specification, a "variant" of an amino acid sequence (peptide, protein or polypeptide) includes amino acid insertion variants, amino acid addition variants, amino acid deletion variants and/or amino acid substitution variants. The term "variant" includes all splice variants, post-translationally modified variants, conformations, isoforms and species homologs, particularly those naturally expressed by a cell. The term "variant" particularly includes fragments of an amino acid sequence.

아미노산 삽입 변이체는 특정 아미노산 서열에 하나 또는 2 이상의 아미노산의 삽입을 포함한다. 삽입을 갖는 아미노산 서열 변이체의 경우, 수득되는 산물에 대한 적절한 스크리닝을 이용한 무작위 삽입도 가능하지만, 아미노산 서열에서 특정 부위에 하나 이상의 아미노산 잔기가 삽입된다. 아미노산 부가 변이체는, 예를 들어 아미노산 1개, 2개, 3개, 5개, 10개, 20개, 30개, 50개 또는 그 초과와 같은 하나 이상의 아미노산의 아미노- 및/또는 카르복시-말단 융합을 포함한다. 아미노산 결손 변이체는 1개, 2개, 3개, 5개, 10개, 20개, 30개, 50개 또는 그 이상의 아미노산의 제거와 같이, 서열로부터 하나 이상의 아미노산의 제거를 특징으로 한다. 결손은 단백질의 임의의 위치일 수 있다. 단백질의 N-말단 및/또는 C-말단에 결손을 포함하는 아미노산 결손 변이체는 N-말단 및/또는 C-말단 절단 변이체 (truncation variant)로도 지칭된다. 아미노산 치환 변이체는 서열에서 하나 이상의 잔기를 제거하고 그 위치에 다른 잔기를 삽입하는 것을 특징으로 한다. 상동적인 단백질들 또는 펩타이드들 간에 보존되지 않은 아미노산 서열에서 변형이 위치하는 것 및/또는 아미노산을 유사한 특성의 다른 아미노산으로 치환하는 것이 선호된다. Amino acid insertion variants comprise the insertion of one or more amino acids into a particular amino acid sequence. For amino acid sequence variants having insertions, one or more amino acid residues are inserted at a specific site in the amino acid sequence, although random insertions using appropriate screening of the resulting products are also possible. Amino acid addition variants comprise amino- and/or carboxy-terminal fusions of one or more amino acids, such as 1, 2, 3, 5, 10, 20, 30, 50 or more amino acids. Amino acid deletion variants are characterized by the removal of one or more amino acids from the sequence, such as the removal of 1, 2, 3, 5, 10, 20, 30, 50 or more amino acids. The deletion can be at any position in the protein. Amino acid deletion variants comprising deletions at the N-terminus and/or C-terminus of a protein are also referred to as N-terminal and/or C-terminal truncation variants. Amino acid substitution variants are characterized by the deletion of one or more residues in the sequence and the insertion of another residue in its place. Preference is given to modifications located in an amino acid sequence that is not conserved between homologous proteins or peptides and/or to replacement of an amino acid with another amino acid having similar properties.

특히 본 발명에 따른 폴리펩타이드에서 서열번호 1의 2번째, 7번째, 10번째, 및 13번째 아미노산을 제외한 위치에서 보존적 치환을 포함할 수 있다. 예를 들어, 본 발명에 포함되는 변이체는 서열번호 1 또는 2로 표시되는 아미노산 서열과 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 또는 99% 동일성을 갖는 아미노산 서열을 포함하는 단백질이며, 이는 서열번호 1 또는 2로 표시되는 아미노산 서열로 구성되는 단백질과 기능적 동일성을 유지한다. In particular, the polypeptide according to the present invention may include conservative substitutions at positions other than the 2nd, 7th, 10th, and 13th amino acids of SEQ ID NO: 1. For example, a variant encompassed by the present invention is a protein comprising an amino acid sequence having 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% identity to the amino acid sequence represented by SEQ ID NO: 1 or 2, which maintains functional identity with a protein comprised of the amino acid sequence represented by SEQ ID NO: 1 or 2.

바람직하게는, 펩타이드 및 단백질 변이체에서 아미노산 변화는 보존적 아미노산 변화, 즉 유사하게 하전되거나 비-하전된 아미노산의 치환이다. 보존적 아미노산 변화는 측쇄와 비슷한 아미노산 계열 중 하나의 치환과 관련된다. 자연 발생 아미노산은 일반적으로 4가지 계열로 나뉜다: 산성 (아스파테이트, 글루타메이트), 염기성 (라이신, 아르기닌, 히스티딘), 비-극성 아미노산 (알라닌, 발린, 루신, 이소루신, 프롤린, 페닐알라닌, 메티오닌, 트립토판), 및 비-하전된 극성 아미노산 (글리신, 아스파라긴, 글루타민, 시스테인, 세린, 트레오닌, 티로신). 페닐알라닌, 트립토판 및 티로신은 때때로 방향족 아미노산으로 같이 분류된다. 일 구현예에서, 보존적인 아미노산 치환은 다음과 같은 군 내에서 치환을 포함한다:Preferably, the amino acid changes in the peptide and protein variants are conservative amino acid changes, i.e., substitutions of similarly charged or uncharged amino acids. A conservative amino acid change involves substitution of one of a family of amino acids with a similar side chain. Naturally occurring amino acids are generally divided into four families: acidic (aspartate, glutamate), basic (lysine, arginine, histidine), non-polar amino acids (alanine, valine, leucine, isoleucine, proline, phenylalanine, methionine, tryptophan), and non-charged polar amino acids (glycine, asparagine, glutamine, cysteine, serine, threonine, tyrosine). Phenylalanine, tryptophan, and tyrosine are sometimes grouped together as aromatic amino acids. In one embodiment, a conservative amino acid substitution comprises a substitution within the following groups:

글리신, 알라닌;Glycine, Alanine;

발린, 이소루신, 루신;valine, isoleucine, leucine;

아스파르테이트, 글루타메이트;aspartate, glutamate;

아스파라긴, 글루타민;Asparagine, glutamine;

세린, 트레오닌;Serine, threonine;

라이신, 아르기닌; 및Lysine, arginine; and

페닐알라닌, 티로신.Phenylalanine, tyrosine.

본 발명의 일 실시예에서, 서열번호 1에서 1번 위치의 트레오닌이 세린으로 치환(T1S)되고/거나; 3번 위치의 트레오닌이 세린으로 치환(T3S)되고/거나; 4번 위치의 글리신이 알라닌으로 치환(G4A)되고/거나; 5번 위치의 아르기닌이 라이신으로 치환(R5K)되고/거나; 8번 위치의 이소루신이 발린(I8V) 또는 루신(I8L)으로 치환되고/거나; 9번 위치의 글리신이 알라닌으로 치환(G9A)되고/거나; 11번 위치의 글리신이 알라닌으로 치환(G11A)되고/거나; 12번 위치의 알라닌이 글리신으로 치환(A12G)되고 거나; 14번 위치의 글리신이 알라닌으로 치환(G14A)된, 보존적 치환을 포함하는 변이체를 제공한다. In one embodiment of the present invention, a variant is provided that includes conservative substitutions, wherein threonine at position 1 in SEQ ID NO: 1 is replaced with serine (T1S); threonine at position 3 is replaced with serine (T3S); glycine at position 4 is replaced with alanine (G4A); arginine at position 5 is replaced with lysine (R5K); isoleucine at position 8 is replaced with valine (I8V) or leucine (I8L); glycine at position 9 is replaced with alanine (G9A); glycine at position 11 is replaced with alanine (G11A); alanine at position 12 is replaced with glycine (A12G); or glycine at position 14 is replaced with alanine (G14A).

용어 “산성 아미노산 잔기”는 바람직하게 글루탐산 (글루타메이트, Glu) 또는 아스파르트산 (아스파르테이트, Asp), 특히 글루탐산과 관련된다. 용어 “염기성 아미노산 잔기”는 라이신 (Lys) 또는 아르기닌 (Arg), 특히 라이신과 관련된다. The term “acidic amino acid residue” preferably relates to glutamic acid (glutamate, Glu) or aspartic acid (aspartate, Asp), particularly glutamic acid. The term “basic amino acid residue” preferably relates to lysine (Lys) or arginine (Arg), particularly lysine.

바람직하게는, 주어진 아미노산 서열과, 상기 주어진 아미노산 서열의 변이체인 아미노산 서열 간의 유사성, 바람직하게는 동일성 정도는 적어도 약 60%, 65%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 또는 99%일 것이다. 유사성 또는 동일성 정도는 바람직하게는 참조 아미노산 서열의 전장의 적어도 약 10%, 적어도 약 20%, 적어도 약 30%, 적어도 약 40%, 적어도 약 50%, 적어도 약 60%, 적어도 약 70%, 적어도 약 80%, 적어도 약 90% 또는 약 100%인 아미노산 영역에 대해 제공된다. 예를 들어, 참조 아미노산 서열이 200 아미노산으로 구성된다면, 유사성 또는 동일성 정도는 바람직하게는 적어도 약 20개, 적어도 약 40개, 적어도 약 60개, 적어도 약 80개, 적어도 약 100개, 적어도 약 120개, 적어도 약 140개, 적어도 약 160개, 적어도 약 180개 또는 약 200개 아미노산, 바람직하게는 연속적인 아미노산들에 대해 제공된다. 바람직한 구현예에서, 유사성 또는 동일성 정도는 참조 아미노산 서열의 전장에 대해 제공된다. 서열 유사성, 바람직하게는 서열 동일성을 결정하기 위한 정렬은 당해 기술 분야에 공지된 도구를 사용해, 바람직하게는 최상의 서열 정렬, 예를 들어, Align을 사용해, 표준 설정 조건으로, 바람직하게는 EMBOSS::needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5 하에 수행할 수 있다.Preferably, the degree of similarity, preferably identity, between a given amino acid sequence and an amino acid sequence which is a variant of said given amino acid sequence will be at least about 60%, 65%, 70%, 80%, 81%, 82%, 83%, 84%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99%. The degree of similarity or identity is preferably provided for an amino acid region which is at least about 10%, at least about 20%, at least about 30%, at least about 40%, at least about 50%, at least about 60%, at least about 70%, at least about 80%, at least about 90% or about 100% of the full length of the reference amino acid sequence. For example, if the reference amino acid sequence consists of 200 amino acids, the degree of similarity or identity is preferably provided for at least about 20, at least about 40, at least about 60, at least about 80, at least about 100, at least about 120, at least about 140, at least about 160, at least about 180 or about 200 amino acids, preferably contiguous amino acids. In a preferred embodiment, the degree of similarity or identity is provided for the full length of the reference amino acid sequence. Alignments for determining sequence similarity, preferably sequence identity, can be performed using tools known in the art, preferably using best sequence alignment, e.g. Align, with standard settings, preferably EMBOSS::needle, Matrix: Blosum62, Gap Open 10.0, Gap Extend 0.5.

"서열 유사성"은 동일하거나 보존적 아미노산 치환을 나타내는 아미노산의 백분율을 나타낸다. 2개의 아미노산 서열 간의 "서열 동일성"은 서열 간에 동일한 아미노산의 백분율을 나타낸다."Sequence similarity" refers to the percentage of amino acids that are identical or represent conservative amino acid substitutions. "Sequence identity" between two amino acid sequences refers to the percentage of amino acids that are identical between the sequences.

용어 "동일성 %"는 최상으로 정렬하여 수득되는, 비교되는 2개의 서열 간에 동일한 아미노산 잔기의 백분율을 나타내는 것으로 의도되며, 이 백분율은 순전히 통계적이며, 두 서열 간의 차이는 무작위로, 그리고 전장에 걸쳐 분포된다. 2개의 아미노산 서열 간의 서열 비교는 통상적으로 이들 서열을 최적으로 정렬한 후 비교함으로써 수행되며, 상기 비교는 서열 유사성의 국소 영역을 식별 및 비교하기 위해 세그먼트 또는 "비교 창"에 의해 수행된다. 비교하기 위한 서열의 최적 정렬은 수동 외에도 Smith and Waterman, 1981, Ads App. Math. 2, 482의 국소 상동성 알고리즘 수단을 이용하거나, Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443의 국소 상동성 알고리즘의 수단을 이용하거나, Pearson and Lipman, 1988, Proc. Natl Acad. Sci. USA 85, 2444의 유사성 검색 방법의 수단을 이용하거나, 또는 이들 알고리즘을 사용하는 컴퓨터 프로그램 (위스콘신 제네틱스 소프트웨어 패키지의 GAP, BESTFIT, FASTA, BLAST P, BLAST N 및 TFASTA, Genetics Computer Group, 575 Science Drive, Madison, Wis.)을 이용하여, 생성할 수 있다.The term "% identity" is intended to refer to the percentage of amino acid residues that are identical between the two sequences being compared, obtained by best alignment, and this percentage is purely statistical, the differences between the two sequences being distributed randomly and over their entire length. Sequence comparisons between two amino acid sequences are typically performed by optimally aligning those sequences and then comparing them, said comparison being performed by segments or "comparison windows" in order to identify and compare local regions of sequence similarity. Optimal alignment of sequences for comparison may be performed manually, or by means of the local homology algorithm of Smith and Waterman, 1981, Ads App. Math. 2, 482, or by means of the local homology algorithm of Neddleman and Wunsch, 1970, J. Mol. Biol. 48, 443, or by means of the local homology algorithm of Pearson and Lipman, 1988, Proc. Natl Acad. Sci. By means of the similarity search methods of USA 85, 2444, or by using computer programs utilizing these algorithms (GAP, BESTFIT, FASTA, BLAST P, BLAST N, and TFASTA of the Wisconsin Genetics Software Package, Genetics Computer Group, 575 Science Drive, Madison, Wis.).

동일성 %는 2개의 서열 간의 동일성 %를 구하기 위해 비교되는 2개의 서열 간에 동일한 위치의 수를 결정한 다음 이 숫자를 비교한 위치의 수로 나눈 후 수득된 결과에 100을 곱하여 계산한다.% Identity is calculated by determining the number of identical positions between the two sequences being compared, dividing this number by the number of positions compared, and then multiplying the result by 100 to obtain the % identity between the two sequences.

상동성 아미노산 서열은 본 명세서에 따르면 아미노산 잔기들에 대해 적어도 40%, 특히 적어도 50%, 적어도 60%, 적어도 70%, 적어도 80%, 적어도 90%, 및 바람직하게는 적어도 95%, 적어도 98% 또는 적어도 99% 동일성을 나타낸다.Homologous amino acid sequences according to the present specification exhibit at least 40%, particularly at least 50%, at least 60%, at least 70%, at least 80%, at least 90%, and preferably at least 95%, at least 98% or at least 99% identity with respect to amino acid residues.

본원에 기술된 아미노산 서열 변이체는 당해 기술 분야의 당업자에 의해, 예를 들어, 재조합 DNA 조작에 의해 쉽게 제조될 수 있다. 치환, 부가, 삽입 또는 결손을 가진 펩타이드 또는 단백질을 제조하기 위한 DNA 서열의 조작은 예를 들어 Sambrook et al. (1989)에 상세하게 기술되어 있다. 또한, 본원에 기술된 펩타이드 및 아미노산 변이체는, 예를 들어 고상 합성 및 유사 방법과 같은, 공지된 펩타이드 합성 기법의 도움을 받아 쉽게 제조할 수 있다.The amino acid sequence variants described herein can be readily prepared by those skilled in the art, for example, by recombinant DNA manipulation. Manipulation of DNA sequences to produce peptides or proteins having substitutions, additions, insertions or deletions is described in detail, for example, in Sambrook et al. (1989). In addition, the peptides and amino acid variants described herein can be readily prepared with the aid of known peptide synthesis techniques, for example, by solid phase synthesis and similar methods.

일 구현예에서, 아미노산 서열 (펩타이드 또는 단백질)의 단편 또는 변이체는 바람직하게는 "기능성 단편" 또는 "기능성 변이체"이다. 아미노산 서열의 "기능성 단편" 또는 "기능성 변이체"라는 용어는 이것이 유래되는 아미노산 서열과 동일한 또는 비슷한 하나 이상의 기능적 특성을 나타내는 임의의 단편 또는 변이체에 관한 것이고, 즉 기능적 등가체이다. In one embodiment, the fragment or variant of an amino acid sequence (peptide or protein) is preferably a "functional fragment" or "functional variant". The term "functional fragment" or "functional variant" of an amino acid sequence relates to any fragment or variant that exhibits one or more functional properties identical or similar to the amino acid sequence from which it is derived, i.e. is a functional equivalent.

용어 “기능성 단편” 또는 “기능성 변이체”는, 본원에서 사용된 바와 같이, 특히 모 분자 또는 서열의 아미노산 서열과 비교하여 하나 이상의 아미노산이 변경된 아미노산 서열을 포함하고 모 분자 또는 서열의 하나 이상의 기능들, 예를 들어, 표적 분자에 결합 또는 표적 분자에 결합에 기여를 여전히 수행할 수 있는 변이체 분자 또는 서열을 의미한다. 일 구현예에서, 모 분자 또는 서열의 아미노산 서열에서 변형은 분자 또는 서열의 결합 특징에 유의미하게 영향을 미치거나 변경하지 않는다. 다른 구현예에서, 기능성 단편 또는 기능성 변이체의 결합은 감소될 수 있지만 여전히 유의미하게 존재하고, 예를 들어, 기능성 변이체의 결합은 모 분자 또는 서열의 적어도 50%, 적어도 60%, 적어도 70%, 적어도 80%, 또는 적어도 90%일 수 있다. 그러나, 다른 구현예에서, 기능성 단편 또는 기능성 변이체의 결합은 모 분자 또는 서열에 비하여 증가될 수 있다. The term “functional fragment” or “functional variant,” as used herein, refers to a variant molecule or sequence that comprises an amino acid sequence in which one or more amino acids have been altered, particularly compared to the amino acid sequence of the parent molecule or sequence, and which is still capable of performing one or more of the functions of the parent molecule or sequence, e.g., binding to a target molecule or contributing to binding to a target molecule. In one embodiment, the alteration in the amino acid sequence of the parent molecule or sequence does not significantly affect or alter the binding characteristics of the molecule or sequence. In other embodiments, the binding of the functional fragment or functional variant may be reduced but still significantly present, e.g., the binding of the functional variant may be at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of that of the parent molecule or sequence. However, in other embodiments, the binding of the functional fragment or functional variant may be increased relative to the parent molecule or sequence.

지정된 아미노산 서열 (펩타이드, 단백질 또는 폴리펩타이드)"로부터 유래되는" 아미노산 서열 (펩타이드, 단백질 또는 폴리펩타이드)은 제1 아미노산 서열의 기원을 의미한다. 바람직하게는, 특정 아미노산 서열로부터 유래된 아미노산 서열은 이 특정 서열 또는 이의 단편과 동일한, 본질적으로 동일한 또는 상동성인 아미노산 서열을 가진다. 특정 아미노산 서열로부터 유래된 아미노산 서열은 이 특정 서열 또는 이의 단편의 변이체일 수 있다. 예를 들어, 당해 기술 분야에서 통상의 지식을 가진 자는 본원에서 사용하기에 적합한 Rv0747/PE_PGRS10이 이들로부터 유래된 자연적으로 발생되거나 천연의 서열로부터 서열에서는 변형되지만 천연 서열의 요망되는 활성은 유지되도록 변경될 수 있음을 이해할 것이다. An amino acid sequence (peptide, protein or polypeptide) "derived from" a specified amino acid sequence (peptide, protein or polypeptide) means the origin of the first amino acid sequence. Preferably, an amino acid sequence derived from a particular amino acid sequence has an amino acid sequence that is identical, essentially identical or homologous to that particular sequence or a fragment thereof. An amino acid sequence derived from a particular amino acid sequence may be a variant of that particular sequence or a fragment thereof. For example, a person skilled in the art will appreciate that Rv0747/PE_PGRS10 suitable for use herein may be modified in sequence from a naturally occurring or native sequence derived therefrom, but which retains the desired activity of the native sequence.

“분리된”은 자연적 상태로부터 변경되거나 제거된 것을 의미한다. 예를 들어, 살아있는 동물에서 자연적으로 존재하는 핵산 또는 펩타이드는 “분리된”이 아니지만, 이의 자연 상태의 공존하는 물질로부터 부분적으로 또는 완전히 분리된 동일한 핵산 또는 펩타이드는 “분리된”이다. 분리된 핵산 또는 단백질은 실질적으로 정제된 형태로 존재할 수 있거나, 예를 들어, 숙주 세포와 같은 비-자연적 환경에 존재할 수 있다. “Isolated” means altered or removed from its natural state. For example, a nucleic acid or peptide naturally occurring in a living animal is not “isolated,” but the same nucleic acid or peptide partially or completely separated from its natural coexisting materials is “isolated.” An isolated nucleic acid or protein may exist in a substantially purified form, or may exist in a non-natural environment, such as, for example, a host cell.

본 발명의 맥락에서 용어 “재조합”은 “유전적 조작을 통해 만들어진”을 의미한다. 바람직하게, 본 발명의 맥락에서 재조합 세포와 같은 “재조합 대상”은 자연적으로 발생되지 않는다. In the context of the present invention, the term “recombinant” means “created through genetic manipulation.” Preferably, a “recombinant subject,” such as a recombinant cell, in the context of the present invention does not occur naturally.

본원에서 사용된 바와 같이 용어 “자연적으로 발생한”은 대상이 자연계에서 발견될 수 있다는 사실을 의미한다. 예를 들어, 유기체 (바이러스 포함) 내에 존재하고 자연계 내 공급원으로부터 분리될 수 있고 실험실에서 인간에 의해 의도적으로 변형되지 않은 펩타이드 또는 핵산은 자연적으로 발생한 것이다. As used herein, the term “naturally occurring” means that a subject can be found in nature. For example, a peptide or nucleic acid is naturally occurring if it is present in an organism (including a virus), can be isolated from a source in nature, and has not been intentionally modified by humans in a laboratory.

용어 “유전적 변형”은 핵산으로 세포를 형질감염시키는 것을 포함한다. 용어 “형질감염”은 핵산, 특히 RNA의 세포로의 도입에 관한 것이다. 본 발명의 모든 목적을 위하여, 용어 “형질감염”은 또한 핵산의 세포에의 도입 또는 이러한 세포에 의해 핵산의 섭취를 포함한다. 형질감염의 일부 적용의 경우, 만약 형질감염된 유전 물질이 일시적으로만 발현된다면 충분하다. RNA는 세포에 형질감염되어 이의 암호화된 단백질을 일시적으로 발현할 수 있다. 형질감염 과정에서 도입된 핵산은 일반적으로 핵 유전체에 삽입되지 않기 때문에, 외래 핵산은 유사 분열을 통해 희석되거나 분해될 것이다. 핵산의 에피좀성 증폭이 가능한 세포는 희석 비율을 현저하게 감소시킨다. 만약 형질감염된 핵산이 사실상 세포 및 이의 자세포에 잔존한다면, 안정한 형질감염이 반드시 일어나야 한다. 이러한 안정한 형질감염은 형질감염을 위한 바이러스-기반 시스템 또는 트랜스포존-기반 시스템을 사용함으로써 달성할 수 있다. 일반적으로, 유전적으로 변형되어 수용체 폴리펩타이드 및/또는 항원 수용체를 발현하는 세포는 수용체 폴리펩타이드를 엔코딩하는 핵산 및/또는 항원 수용체를 엔코딩하는 핵산으로 안정적으로 형질감염되는 반면, 일반적으로 리간드 폴리펩타이드를 엔코딩하는 핵산 및/또는 항원을 엔코딩하는 핵산은 일시적으로 세포 내로 형질감염 된다.The term “genetic modification” includes transfection of a cell with a nucleic acid. The term “transfection” relates to the introduction of a nucleic acid, particularly RNA, into a cell. For all purposes of the present invention, the term “transfection” also includes the introduction of a nucleic acid into a cell or the uptake of a nucleic acid by such a cell. For some applications of transfection, it is sufficient if the transfected genetic material is only transiently expressed. RNA can be transfected into a cell and cause it to transiently express its encoded protein. Since the nucleic acid introduced during the transfection process is generally not integrated into the nuclear genome, the foreign nucleic acid will be diluted or degraded through mitosis. Cells capable of episomal amplification of the nucleic acid significantly reduce the dilution rate. If the transfected nucleic acid is in fact retained in the cell and its progenitor cells, stable transfection must occur. Such stable transfection can be achieved by using a virus-based system or a transposon-based system for transfection. Typically, cells genetically modified to express a receptor polypeptide and/or an antigen receptor are stably transfected with a nucleic acid encoding a receptor polypeptide and/or a nucleic acid encoding an antigen receptor, whereas typically, a nucleic acid encoding a ligand polypeptide and/or a nucleic acid encoding an antigen are transiently transfected into the cell.

"핵산", "핵산 분자", "올리고뉴클레오타이드" 및 "폴리뉴클레오타이드"는 상호교환적으로 사용되고, 단일 가닥 형태나 이중 가닥 나선(helix)으로의 리보뉴클레오사이드(아데노신, 구아노신, 우리딘 또는 시티딘; "RNA 분자") 또는 데옥시리보뉴클레오사이드(데옥시아데노신, 데옥시구아노신, 데옥시티민 또는 데옥시시티딘; "DNA 분자")의 인산 에스테르의 중합체 형태 또는 포스포로티오에이트(phosphorothioate) 및 티오에스테르와 같은 이의 임의의 인산 에스테르 유사체를 지칭한다. 이중 나선 DNA-DNA, DNA-RNA 및 RNA-RNA 나선이 가능하다. 용어 핵산 분자, 및 특히 DNA 또는 RNA 분자는 상기 분자의 일차 및 이차 구조만을 지칭하며 어느 특정 삼차 형태에 제한하지는 않는다. 따라서, 이 용어는 그 중에서도 선형 또는 환형 DNA 분자(예컨대, 제한효소 단편), 플라스미드, 초나선형(supercoiled) DNA 및 염색체로 발견되는 이중 가닥 DNA를 포함한다. 특정 이중 가닥 DNA 분자의 구조를 논할 때, 서열을 전사되지 않은 DNA 가닥(즉, mRNA에 일치하는 서열을 갖는 가닥)을 따라 5'에서 3' 방향으로만 제시하는 일반적인 규약에 따라 본 명세서에 서열이 기술될 수 있다. "재조합 DNA 분자"는 분자 생물학적 조작을 거친 DNA 분자이다. DNA는 cDNA, 게놈 DNA, 플라스미드 DNA, 합성 DNA 및 반합성 DNA를 포함하나, 이에 제한되지 않는다.The terms "nucleic acid", "nucleic acid molecule", "oligonucleotide" and "polynucleotide" are used interchangeably and refer to a polymer of the phosphate esters of ribonucleosides (adenosine, guanosine, uridine or cytidine; "RNA molecule") or deoxyribonucleosides (deoxyadenosine, deoxyguanosine, deoxythymine or deoxycytidine; "DNA molecule"), either in single-stranded form or as a double-stranded helix or any of their phosphate ester analogues such as phosphorothioates and thioesters. Double-stranded DNA-DNA, DNA-RNA and RNA-RNA helices are possible. The terms nucleic acid molecule, and particularly DNA or RNA molecule, refer only to the primary and secondary structures of said molecules and are not limited to any particular tertiary configuration. Thus, the term includes, among others, linear or circular DNA molecules (e.g., restriction fragments), plasmids, supercoiled DNA, and double-stranded DNA found as chromosomes. When discussing the structure of a particular double-stranded DNA molecule, the sequence may be described herein according to the general convention of presenting the sequence only in the 5' to 3' direction along the non-transcribed DNA strand (i.e., the strand having the sequence matching the mRNA). A "recombinant DNA molecule" is a DNA molecule that has undergone molecular biological manipulation. The DNA includes, but is not limited to, cDNA, genomic DNA, plasmid DNA, synthetic DNA, and semi-synthetic DNA.

핵산은 벡터 내에 포함될 수 있다. 용어 “벡터”는 본원에서 사용된 바와 같이, 플라스미드 벡터, 코스미드 벡터, 람다 파아지와 같은 파아지 벡터, 레트로바이러스, 아데노바이러스 또는 배큘로바이러스 벡터와 같은 바이러스 벡터, 또는 박테리아 인공 염색체 (BAC), 효모 인공 염색체 (YAC) 또는 P1 인공 염색체 (PAC)와 같은 인공적 염색체 벡터를 포함하는, 당업자에게 공지된 임의의 벡터를 포함한다. 상기 벡터는 발현뿐 아니라 클로닝 벡터를 포함한다. 발현 벡터는 플라스미드뿐 아니라 바이러스 벡터를 포함하고, 일반적으로 특정 숙주 유기체 (예를 들어, 박테리아, 효모, 식물, 곤충, 또는 포유류) 또는 in vitro 발현 시스템에서 작동적으로 연결된 암호 서열의 발현에 필요한 요망되는 암호 서열 및 적당한 DNA 서열을 포함한다. 클로닝 벡터는 일반적으로 특정 요망되는 DNA 단편을 조작하고 증폭하기 위해 사용되고 요망되는 DNA 단편의 발현에 필요한 기능성 서열이 없을 수 있다.The nucleic acid may be contained within a vector. The term “vector,” as used herein, includes any vector known to those skilled in the art, including plasmid vectors, cosmid vectors, phage vectors such as lambda phage, viral vectors such as retrovirus, adenovirus or baculovirus vectors, or artificial chromosome vectors such as bacterial artificial chromosomes (BAC), yeast artificial chromosomes (YAC) or P1 artificial chromosomes (PAC). Such vectors include expression as well as cloning vectors. Expression vectors include plasmids as well as viral vectors, and generally contain the desired coding sequence and appropriate DNA sequences necessary for expression of the coding sequence operably linked in a particular host organism (e.g., bacteria, yeast, plants, insects, or mammals) or in vitro expression system. Cloning vectors are generally used to manipulate and amplify a particular desired DNA fragment and may lack functional sequences necessary for expression of the desired DNA fragment.

바이러스 벡터는 살아있는 동물 대상뿐만 아니라 세포에서의 광범위한 유전자 전달 적용 분야에 사용되어 왔다. 사용될 수 있는 바이러스 벡터들은 아데노바이러스, 레트로바이러스, 백시나(vaccinia) 바이러스, 수두바이러스(poxvirus), 아데노 관련 바이러스, 단순포진바이러스(herpes simplex virus), 렌티바이러스 (lentivirus), 배큘로바이러스(baculovirus), 센다이 바이러스(sendai virus), 홍역 바이러스(measles virus), 시미안 바이러스(simian virus) 40 및 엡스타인-바(Epstein-Barr) 바이러스 벡터를 포함하나, 이에 제한되지 않는다. 비-바이러스성 벡터는 플라스미드, 리포플렉스(lipoplex)(양이온 리포좀-DNA 복합체), 폴리플렉스(polyplex)(양이온 중합체-DNA 복합체) 및 단백질-DNA 복합체를 포함한다. 핵산 이외에, 벡터는 핵산 전달 결과(조직으로의 전달, 발현 지속성 등)를 선별, 측정 및 추적관찰하는데 유용한 하나 이상의 조절 영역 및/또는 선별 마커를 포함할 수 있다. Viral vectors have been used for a wide range of gene transfer applications in living animals as well as cells. Viral vectors that may be used include, but are not limited to, adenovirus, retrovirus, vaccinia virus, poxvirus, adeno-associated virus, herpes simplex virus, lentivirus, baculovirus, sendai virus, measles virus, simian virus 40, and Epstein-Barr virus vectors. Non-viral vectors include plasmids, lipoplexes (cationic liposome-DNA complexes), polyplexes (cationic polymer-DNA complexes), and protein-DNA complexes. In addition to the nucleic acid, the vector may include one or more regulatory regions and/or selectable markers that are useful for selecting, measuring, and monitoring the outcome of the nucleic acid delivery (e.g., delivery to tissues, persistence of expression, etc.).

용어 "발현 벡터"는 삽입된 핵산 서열을 발현한 후 숙주를 형질전환시킬 수 있도록 고안된 벡터, 플라스미드 또는 운반체를 지칭한다. 클론된 유전자, 즉, 삽입된 핵산 서열은 일반적으로 프로모터, 최소 프로모터, 인핸서 등과 같은 조절 요소의 통제 하에 놓여진다. 원하는 숙주 세포에서의 핵산의 발현을 유도하는데 유용한 개시 조절 영역 또는 프로모터들은 무수히 많으며 본 기술분야의 숙련자들에게 잘 알려져 있다. 실질적으로 이들 유전자들의 발현을 유도할 수 있는 임의의 프로모터는 바이러스 프로모터, 박테리아 프로모터, 동물 프로모터, 포유동물 프로모터, 합성 프로모터, 구성 프로모터, 조직 특이적 프로모터, 발병(pathogenesis) 또는 질병 관련 프로모터, 발생 특이적 프로모터(developmental specific promoter), 유도성 프로모터(inducible promoter), 약하게 조절된 프로모터(light regulated promoter)를 포함하나, 이에 제한되지는 않으며; SV40 초기(SV40) 프로모터 영역, 라우스 육종 바이러스(Rous sarcoma virus, RSV)의 3' 긴 말단 반복(LTR)에 함유된 프로모터, 아데노바이러스(Ad)의 E1A 또는 주요 후기 프로모터(major late promoter, MLP), 인간 사이토메갈로바이러스 (human cytomegalovirus, HCMV) 즉시 초기 프로모터(immediate early promoter), 단순 포진 바이러스(HSV) 티미딘 키나아제(TK) 프로모터, 배큘로바이러스 IE1 프로모터, 신장 인자(elongation factor) 1 알파(EF1) 프로모터, 글리세르알데하이드-3-포스페이트 탈수소효소(GSPDH) 프로모터, 포스포글리세레이트 키나아제(PGK) 프로모터, 유비퀴틴 C(Ubc) 프로모터, 알부민 프로모터, 마우스 메탈로티오네인(metallothionein)-L 프로모터 및 전사 조절 영역의 조절 서열, 유비쿼터스 프로모터(HPRT, 비멘틴(vimentin), β액틴, 튜불린 등), 중간필라멘트(intermediate filament)(데스민(desmin), 신경미세섬유, 케라틴, GFAP 등), 치료 유전자의 프로모터(MDR, CFTR 또는 인자 VIII 형태 등의), 발병 또는 질병 관련 프로모터, 및 췌장 선방세포(pancreatic acinar cell) 에서 활성인 엘라스타제(elastase) I 유전자 조절 영역과 같은, 조직 특이성을 나타내며 형질전환 동물에서 이용되어 온 프로모터; 췌장 베타 세포에서 활성인 인슐린 유전자 조절 영역, 림프계(lymphoid) 세포에서 활성인 면역글로불린 유전자 조절 영역, 고환, 유방, 림프계 및 대식 세포에서 활성인 마우스 유방암 바이러스 조절 영역; 간에서 활성인 알부민 유전자, Apo AI 및 Apo AII 조절 영역, 간에서 활성인 알파-태아단백질(fetoprotein) 유전자 조절 영역, 간에서 활성인 알파1-안티트립신 유전자 조절 영역, 골수 세포에서 활성인 베타-글로빈 유전자 조절 영역, 뇌에서의 희소돌기아교세포(oligodendrocyte cell)에서 활성인 마이엘린 염기성 단백질(myelin basic protein) 조절 영역, 골격근에서 활성인 미오신 경쇄-2 유전자 조절 영역 및 시상하부에서 활성인 생식선 자극호르몬 방출 호르몬 (gonadotropic releasing hormone), 피루브산 키나아제 프로모터, 빌린(villin) 프로모터, 지방산 결합 장내 단백질의 프로모터, 평활근 세포 β액틴의 프로모터 등을 포함하나, 이에 제한되지 않는다.The term "expression vector" refers to a vector, plasmid or vehicle designed to transform a host cell by expressing an inserted nucleic acid sequence. The cloned gene, i.e., the inserted nucleic acid sequence, is generally placed under the control of regulatory elements such as a promoter, a minimal promoter, an enhancer, and the like. The initiation regulatory regions or promoters useful for directing expression of the nucleic acid in a desired host cell are numerous and are well known to those skilled in the art. Virtually any promoter capable of directing expression of these genes includes, but is not limited to, viral promoters, bacterial promoters, animal promoters, mammalian promoters, synthetic promoters, constitutive promoters, tissue-specific promoters, pathogenesis or disease-related promoters, developmental specific promoters, inducible promoters, light regulated promoters; SV40 early (SV40) promoter region, promoter contained in the 3' long terminal repeat (LTR) of Rous sarcoma virus (RSV), E1A or major late promoter (MLP) of adenovirus (Ad), immediate early promoter of human cytomegalovirus (HCMV), herpes simplex virus (HSV) thymidine kinase (TK) promoter, baculovirus IE1 promoter, elongation factor 1 alpha (EF1) promoter, glyceraldehyde-3-phosphate dehydrogenase (GSPDH) promoter, phosphoglycerate kinase (PGK) promoter, ubiquitin C (Ubc) promoter, albumin promoter, mouse metallothionein-L Regulatory sequences of promoters and transcription control regions, ubiquitous promoters (such as HPRT, vimentin, β-actin, tubulin), intermediate filaments (such as desmin, neurofilament, keratin, GFAP), promoters of therapeutic genes (such as MDR, CFTR or factor VIII form), pathogenic or disease-related promoters, and promoters that exhibit tissue specificity and have been used in transgenic animals, such as the elastase I gene control region active in pancreatic acinar cells; the insulin gene control region active in pancreatic beta cells, the immunoglobulin gene control region active in lymphoid cells, the mouse mammary carcinoma virus control region active in testicular, breast, lymphoid and macrophage cells; Including, but not limited to, albumin gene active in liver, Apo AI and Apo AII regulatory regions, alpha-fetoprotein gene regulatory region active in liver, alpha1-antitrypsin gene regulatory region active in liver, beta-globin gene regulatory region active in bone marrow cells, myelin basic protein regulatory region active in oligodendrocyte cells in brain, myosin light chain-2 gene regulatory region active in skeletal muscle, and gonadotropic releasing hormone, pyruvate kinase promoter, villin promoter, promoter of fatty acid-binding intestinal protein, promoter of smooth muscle cell β-actin, etc.

벡터는 본 기술분야에서 알려진 방법, 예컨대, 주사, 형질감염, 전기천공법, 미세주사(microinjection), 형질도입, 세포 융합, 리포펙션(lipofection), 칼슘 포스페이트 침전법 (Graham, F.L. et al., Virology, 52:456(1973); 및 Chen과 Okayama, Mol. Cell. Biol. 7:2745-2752(1987)), 리포좀-매개 형질감염법 (Wong, T.K. et al., Gene, 10:87(1980); Nicolau 및 Sene, Biochim. Biophys. Acta, 721:185-190(1982); 및 Nicolau et al., Methods Enzymol., 149:157-176(1987)), DEAE-덱스트란 처리법 (Gopal, Mol. Cell Biol., 5:1188-1190(1985)), 유전자 밤바드먼트 (Yang et al., Proc. Natl. Acad. Sci., 87:9568-9572(1990)) 방법, 유전자 총의 사용 또는 DNA 벡터 전달체(DNA vector transporter)에 의하여, 원하는 숙주 세포로 도입될 수 있다(e.g., Wu et al., J. Biol. Chem. 267:963 (1992); Wu et al., J. Biol. Chem. 263:14621 (1988); and Hartmut et al., Canadian Patent Application No. 2,012,311, 참조).Vectors can be transfected by methods known in the art, such as injection, transfection, electroporation, microinjection, transduction, cell fusion, lipofection, calcium phosphate precipitation (Graham, F.L. et al., Virology, 52:456(1973); and Chen and Okayama, Mol. Cell. Biol. 7:2745-2752(1987)), liposome-mediated transfection (Wong, T.K. et al., Gene, 10:87(1980); Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190(1982); and Nicolau et al., Methods Enzymol., 149:157-176(1987)), DEAE-dextran treatment (Gopal, Mol. Cell Biol., 5:1188-1190 (1985)), gene bombardment (Yang et al., Proc. Natl. Acad. Sci., 87:9568-9572 (1990)), use of a gene gun, or DNA vector transporters (e.g., Wu et al., J. Biol. Chem. 267:963 (1992); Wu et al., J. Biol. Chem. 263:14621 (1988); and Hartmut et al., Canadian Patent Application No. 2,012,311, see).

본 발명에 따른 폴리뉴클레오타이드는 리포펙션에 의해 생체 내에서 도입될 수 있다. 과거 수십년 동안, 시험관 내(in vitro) 핵산의 캡슐화 및 형질감염을 위해 리포좀의 사용이 증가해왔다. 리포좀-매개 형질감염이 맞닥뜨린 어려움과 위험을 제한하고자 고안된 합성 양이온성 지질(synthetic cationic lipid)이 유전자의 생체 내 (in vivo) 형질감염을 위한 리포좀을 제조하는데 사용될 수 있다(Felgner et al., Proc. Natl. Acad. Sci. USA. 84:7413 (1987); Mackey et al., Proc. Natl. Acad. Sci. USA 85:8027 (1988); 및 Ulmer et al., Science 259:1745 (1993)). 양이온성 지질의 사용은 음성 전하를 띠는 핵산의 캡슐화를 촉진할 수 있고, 또한 음성 전하를 띠는 세포막과의 융합을 촉진할 수 있다(Felgner et al., Science 337:387 (1989)). 핵산의 전달을 위한 특히 유용한 지질 화합물 및 조성물이 WO95/18863, WO96/17823 및 U.S. 5,459,127에 기술되어 있다. 외인성(exogenous) 유전자를 생체 내(in vivo) 특정 조직 내로 도입하기 위한 리포펙션의 사용은 몇 가지 실용적인 장점이 있다. 리포좀의 특정 세포로의 분자 표적화는 한가지 영역의 이점을 제시한다. 특정 세포형으로의 직접적인 형질감염이, 췌장, 간, 신장 및 뇌와 같이 세포 이질성을 갖는 조직에 특히 바람직할 것임이 분명하다. 지질은 표적화를 위해 다른 분자들과 화학적으로 결합할 수 있다(Mackey et al. 1988). 표적화된 펩타이드, 예컨대, 호르몬 또는 신경전달물질, 및 항체와 같은 단백질, 또는 비-펩타이드성 분자가 화학적으로 리포좀에 결합될 수 있다.The polynucleotides according to the present invention can be introduced in vivo by lipofection. Over the past several decades, there has been an increasing use of liposomes for in vitro nucleic acid encapsulation and transfection. Synthetic cationic lipids, which are designed to limit the difficulties and risks encountered with liposome-mediated transfection, can be used to prepare liposomes for in vivo gene transfection (Felgner et al., Proc. Natl. Acad. Sci. USA. 84:7413 (1987); Mackey et al., Proc. Natl. Acad. Sci. USA 85:8027 (1988); and Ulmer et al., Science 259:1745 (1993)). The use of cationic lipids can facilitate encapsulation of negatively charged nucleic acids, and can also facilitate fusion with negatively charged cell membranes (Felgner et al., Science 337:387 (1989)). Particularly useful lipid compounds and compositions for the delivery of nucleic acids are described in WO95/18863, WO96/17823, and U.S. 5,459,127. The use of lipofection to introduce exogenous genes into specific tissues in vivo has several practical advantages. Targeting of molecules to specific cells by liposomes presents one area of advantage. Direct transfection into specific cell types would clearly be particularly desirable in tissues with cellular heterogeneity, such as the pancreas, liver, kidney, and brain. Lipids can be chemically linked to other molecules for targeting purposes (Mackey et al. 1988). Targeted peptides, such as hormones or neurotransmitters, and proteins, such as antibodies, or non-peptide molecules can be chemically bound to the liposomes.

양이온성 올리고펩타이드(예컨대, WO95/21931), DNA 결합 단백질로부터 유래된 펩타이드(예컨대, WO96/25508), 또는 양이온성 중합체(예컨대, WO95/21931)와 같은, 다른 분자들이 또한 생체 내 핵산의 형질감염을 용이하게 하는데 유용하다.Other molecules, such as cationic oligopeptides (e.g., WO95/21931), peptides derived from DNA binding proteins (e.g., WO96/25508), or cationic polymers (e.g., WO95/21931), are also useful for facilitating transfection of nucleic acids in vivo.

생체 내(in vivo) 벡터를 순수한(naked) DNA 플라스미드로서 도입하는 것 또한 가능하다(미국 특허번호 제5,693,622호, 제5,589,466호 및 제5,580,859호 참조). 수용체-매개 DNA 전달법이 사용될 수도 있다(Curiel et al., Hum. Gene Ther. 3:147 (1992); and Wu et al., J. Biol. Chem. 262:4429 (1987)).It is also possible to introduce vectors in vivo as naked DNA plasmids (see U.S. Pat. Nos. 5,693,622, 5,589,466, and 5,580,859). Receptor-mediated DNA delivery may also be used (Curiel et al., Hum. Gene Ther. 3:147 (1992); and Wu et al., J. Biol. Chem. 262:4429 (1987)).

본원에 기술된 핵산은 재조합 및/또는 분리된 분자일 수 있다. The nucleic acids described herein may be recombinant and/or isolated molecules.

본 명세서의 맥락에서, 용어 "전사"는 DNA 서열에서 유전적 암호가 RNA로 전사되는 과정을 지칭한다. 이후 RNA는 펩타이드 또는 단백질로 번역될 수 있다.In the context of this specification, the term "transcription" refers to the process by which the genetic code in a DNA sequence is transcribed into RNA. The RNA can then be translated into peptides or proteins.

“엔코딩”은 뉴클레오티드의 정의된 서열 (즉, rRNA, tRNA 및 mRNA) 또는 아미노산의 정의된 서열 및 이로부터 유래된 생물학적 특성을 가지는, 생물학적 과정에서 다른 폴리머 및 거대분자의 합성을 위한 주형으로서의 역할을 하도록 하는, 유전자, cDNA 또는 mRNA와 같은, 폴리뉴클레오티드에서 뉴클레오티드의 특정 서열의 내재적 성질을 의미한다. 따라서, 만약 이 유전에 상응하는 mRNA의 전사 및 번역이 세포 또는 다른 생물학적 시스템에서 단백질을 생산한다면, 유전자는 단백질을 엔코딩한다. mRNA 서열과 동일하고 일반적으로 서열 목록으로 제공되는 암호화 가닥 및 유전자 또는 cDNA의 전사를 위해 주형으로 사용되는 비-암호화 가닥 모두는 단백질 또는 이 유전자 또는 cDNA의 다른 산물을 엔코딩하는 것으로 언급될 수 있다. “Encoding” refers to the inherent property of a particular sequence of nucleotides in a polynucleotide, such as a gene, cDNA or mRNA, to serve as a template for the synthesis of other polymers and macromolecules in a biological process, having a defined sequence of nucleotides (i.e., rRNA, tRNA and mRNA) or a defined sequence of amino acids and the biological properties derived therefrom. Thus, a gene encodes a protein if the transcription and translation of the mRNA corresponding to that gene produces a protein in a cell or other biological system. Both the coding strand, which is identical to the mRNA sequence and is usually provided as a sequence listing, and the non-coding strand, which is used as a template for transcription of the gene or cDNA, may be said to encode a protein or other product of that gene or cDNA.

본 발명은 본 발명의 벡터를 포함하는 숙주 세포를 제공한다. 숙주 세포는 원핵(예를 들어, 박테리아) 및 진핵 (예를 들어, 곰팡이, 효모, 동물, 곤충, 식물) 세포를 포함하며 본 발명에 따른 폴리펩타이드의 발현에 적절한 임의의 세포일 수 있다. 적절한 원핵 숙주 세포는 이.콜라이(예를 들어, DH5, HB101, JM109 또는 W3110 계통), 바실루스(Bacillus), 스트렙토마이시스(Streptomyces), 살모넬라(Salmonella), 세라티아(Serratia) 및 슈도모나스(Pseudomonas) 종을 포함하나 이에 한정되지 않는다. 적절한 진핵 숙주 세포는 COS, CHO, HepG-2, CV-1, LLCMK2, 3T3, HeLa, RPMI8226, 293, BHK-21, Sf9, 사카로마이세스, 피치아(Pichia), 한세눌라(Hansenula), 클루이베로마이세스(Kluyveromyces), 아스페르길루스(Aspergillus) 또는 트리코데르마(Trichoderma) 종을 포함하나 이에 한정되지 않는다.The present invention provides a host cell comprising a vector of the present invention. The host cell includes prokaryotic (e.g., bacterial) and eukaryotic (e.g., fungal, yeast, animal, insect, plant) cells and may be any cell suitable for expression of a polypeptide according to the present invention. Suitable prokaryotic host cells include, but are not limited to, E. coli (e.g., strains DH5, HB101, JM109 or W3110), Bacillus, Streptomyces, Salmonella, Serratia and Pseudomonas species. Suitable eukaryotic host cells include, but are not limited to, COS, CHO, HepG-2, CV-1, LLCMK2, 3T3, HeLa, RPMI8226, 293, BHK-21, Sf9, Saccharomyces, Pichia, Hansenula, Kluyveromyces, Aspergillus or Trichoderma species.

본원에서 사용된 것과 같이 “내인성”은 유기체, 세포, 조직, 또는 시스템 내부에서 유래되거나 생산된 임의의 물질을 의미한다. As used herein, “endogenous” means any substance derived from or produced within an organism, cell, tissue, or system.

본원에서 사용된 것과 같이 “외인성”은 유기체, 세포, 조직, 또는 시스템 외부에서 유래되거나 생산된 임의의 물질을 의미한다.As used herein, “exogenous” means any substance originating or produced outside an organism, cell, tissue, or system.

본원에서 사용된 것과 같이 용어 “발현”은 코딩 서열에 의해 코딩된 생성물의 생물학적 생산을 지칭하는 것으로, 특정 뉴클레오티드 서열의 전사 및/또는 번역으로 정의할 수 있다. 대부분의 경우에서, 코딩 서열을 포함하는 DNA 서열은 전사되어 메신저-RNA(mRNA)를 형성한다. 이어서, 상기 메신저 RNA는 번역되어 관련 생물학적 활성을 갖는 폴리펩타이드 생성물을 형성한다. 또한, 발현 과정은 RNA 전사 생성물에 대한 추가 가공 단계(예를 들면, 인트론을 제거하기 위한 스플라이싱), 및/또는 폴리펩타이드 생성물의 번역-후 가공을 포함할 수 있다.As used herein, the term “expression” refers to the biological production of a product encoded by a coding sequence, and can be defined as the transcription and/or translation of a particular nucleotide sequence. In most cases, a DNA sequence comprising a coding sequence is transcribed to form messenger RNA (mRNA). The messenger RNA is then translated to form a polypeptide product having the relevant biological activity. The expression process may also include additional processing steps for the RNA transcript product (e.g., splicing to remove introns), and/or post-translational processing of the polypeptide product.

본원에서 사용된 것과 같이, 용어 “연결된”, “융합된” 또는 “융합”은 상호교환적으로 사용된다. 이들 용어는 2 이상의 요소 또는 구성성분 또는 도메인들이 함께 연결되는 것을 의미한다.As used herein, the terms “linked,” “fused,” or “fusion” are used interchangeably. These terms mean that two or more elements or components or domains are joined together.

본원에 기술된 펩타이드, 단백질, 폴리펩타이드, RNA, RNA 입자 및 추가적인 물질, 예를 들어, 면역 체크포인트 저해제는 치료학적 또는 예방학적 치료를 위한 약학 조성물 또는 약제로 투여될 수 있으며, 약학적으로 허용되는 담체를 포함할 수 있고, 선택적으로 하나 이상의 보강제, 안정화제 등을 포함할 수 있는 임의의 적합한 약학적 조성물의 형태로 투여될 수 있다. 일 구현예에서, 약학 조성물은 치료학적 또는 예방학적 치료, 예를 들어 본원에 기술된 것들과 같은 암 질환과 같은 항원이 연관된 질환을 치료 또는 예방하는 데에 사용하기 위한 것이다. The peptides, proteins, polypeptides, RNAs, RNA particles and additional agents, e.g., immune checkpoint inhibitors, described herein can be administered as pharmaceutical compositions or medicaments for therapeutic or prophylactic treatment and can be administered in the form of any suitable pharmaceutical composition, which can include a pharmaceutically acceptable carrier and optionally can include one or more adjuvants, stabilizers, and the like. In one embodiment, the pharmaceutical composition is for use in therapeutic or prophylactic treatment, e.g., treating or preventing an antigen-associated disease, such as cancer diseases, as described herein.

용어 "약학 조성물"은 치료학적으로 유효한 물질을, 바람직하게는 약제학적으로 허용가능한 담체, 희석제 및/또는 부형제와 함께, 포함하는 제형에 관한 것이다. 상기 약학 조성물은, 상기 약학 조성물을 개체에 투여함으로써 질환 또는 장애의 중증도를 낮추거나, 예방하거나 또는 치료하는 데에 유용하다. 약학 조성물은 또한 약학적 제형으로서 당해 기술 분야에 공지되어 있다. 본 발명의 맥락에서, 약학 조성물은 본원에 기술된 바와 같이 펩타이드, 단백질, 폴리펩타이드, RNA, RNA 입자, 면역 효과기 세포 및/또는 추가적인 물질을 포함한다.The term "pharmaceutical composition" relates to a formulation comprising a therapeutically effective substance, preferably together with a pharmaceutically acceptable carrier, diluent and/or excipient. The pharmaceutical composition is useful for reducing the severity of, preventing or treating a disease or disorder by administering the pharmaceutical composition to a subject. Pharmaceutical compositions are also known in the art as pharmaceutical dosage forms. In the context of the present invention, a pharmaceutical composition comprises a peptide, a protein, a polypeptide, an RNA, an RNA particle, an immune effector cell and/or an additional substance as described herein.

본 명세서의 약학 조성물은 하나 이상의 보강제를 포함할 수 있거나, 하나 이상의 보강제와 함께 투여될 수 있다. 용어 "보강제"는 면역 반응을 연장, 강화 또는 가속화하는 화합물을 지칭한다. 보강제는 오일 에멀젼 (예, 프로인드 보강제), 미네랄 화합물 (예, 알럼), 박테리아 생산물 (예, 백일해균 독소) 또는 면역-자극 복합체와 같은 이종적인 화합물 군을 포함한다. 보강제의 예로는, 비-제한적으로, LPS, GP96, CpG 올리고데옥시뉴클레오티드, 성장인자 및 모노카인, 림포카인, 인터루킨, 케모카인과 같은 사이토카인을 포함한다. 사이토카인은 IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL12, IFNα, IFNγ, GM-CSF, LT-a일 수 있다. 추가적으로 알려진 보강제는 알루미늄 하이드록사이드, 프로인드 보강제 또는 Montanide®ISA51과 같은 오일이다. 본 명세서에서 사용하기 위한 적합한 다른 보강제는 Pam3Cys과 같은 리포펩타이드를 포함한다.The pharmaceutical compositions of the present disclosure may include one or more adjuvants or may be administered together with one or more adjuvants. The term "adjuvant" refers to a compound that prolongs, enhances, or accelerates an immune response. Adjuvants include heterogeneous compounds such as oil emulsions (e.g., Freund's adjuvant), mineral compounds (e.g., alum), bacterial products (e.g., pertussis toxin), or immune-stimulating complexes. Examples of adjuvants include, but are not limited to, cytokines such as LPS, GP96, CpG oligodeoxynucleotides, growth factors and monokines, lymphokines, interleukins, chemokines. The cytokines can be IL1, IL2, IL3, IL4, IL5, IL6, IL7, IL8, IL9, IL10, IL12, IFNα, IFNγ, GM-CSF, LT-a. Additionally known adjuvants are aluminum hydroxide, Freund's adjuvant or oils such as Montanide®ISA51. Other suitable adjuvants for use herein include lipopeptides such as Pam3Cys.

본 명세서에 따른 약학 조성물은 일반적으로 "약제학적 유효량"으로, "약제학적으로 허용가능한 제제"로 적용된다.The pharmaceutical composition according to the present specification is generally applied as a “pharmaceutically effective amount” and as a “pharmaceutically acceptable formulation”.

용어 "약제학적으로 허용가능한"은 약학 조성물의 활성 성분의 작용과 상호작용하지 않는 물질의 무독성을 의미한다.The term "pharmaceutically acceptable" refers to the non-toxicity of a substance that does not interact with the action of the active ingredient of a pharmaceutical composition.

용어 "약제학적 유효량" 또는 "치료학적 유효량"은 단독으로 또는 추가적인 투여와 더불어 원하는 반응 또는 원하는 효과를 달성하는 양을 의미한다. 특정 질환을 치료하는 경우에, 원하는 반응은 바람직하게는 질환의 진행 저해를 의미하다. 이는 질환의 진척 속도를 늦추는 것을 포함하며, 특히 질환의 진척을 중단시키거나 또는 역전시키는 것을 포함한다. 질환의 치료에서 원하는 반응은 또한 상기 질환 또는 상기 병태의 개시 지연 또는 개시 예방일 수 있다. 본원에 기술된 조성물의 유효량은 치료할 병태, 질환의 중등도, 나이, 신체 상태, 키 및 체중을 포함하는 환자의 개별 특성, 치료 기간, (존재하는 경우) 동반되는 요법의 유형, 구체적인 투여 경로 및 유사 인자에 따라 결정될 것이다. 따라서, 본원에 기술된 조성믈의 투여 용량은 이러한 다양한 특성에 따라 결정될 수 있다. 환자에서 반응이 1차 투여로 충분하지 않을 경우, 이 보다 고 용량 (또는 효과적으로는 다른, 보다 국지적인 투여 경로에 의해 달성되는 더 높은 용량)이 사용될 수 있다.The term "pharmaceutically effective amount" or "therapeutically effective amount" means an amount that, alone or in combination with additional administration, achieves the desired response or desired effect. In the case of treating a particular disease, the desired response preferably means inhibition of the progression of the disease. This includes slowing the progression of the disease, and in particular, stopping or reversing the progression of the disease. In the treatment of a disease, the desired response may also be delaying the onset or preventing the onset of the disease or condition. The effective amount of the compositions described herein will be determined based on the individual characteristics of the patient, including the condition being treated, the severity of the disease, age, physical condition, height and weight, the duration of the treatment, the type of concomitant therapy (if any), the specific route of administration, and similar factors. Accordingly, the dosage of the compositions described herein can be determined based on these various characteristics. If the patient does not respond sufficiently with the first administration, a higher dose (or a higher dose achieved effectively by another, more local route of administration) may be used.

본 명세서의 약학적 조성물은 염, 완충제, 보존제, 그리고 선택적으로 다른 치료학적 물질을 포함할 수 있다. 일 구현예에서, 본 명세서의 약학적 조성물은 하나 이상의 약학적으로 허용되는 담체, 희석제 및/또는 부형제를 포함한다.The pharmaceutical compositions of the present disclosure may include salts, buffers, preservatives, and optionally other therapeutic agents. In one embodiment, the pharmaceutical compositions of the present disclosure include one or more pharmaceutically acceptable carriers, diluents, and/or excipients.

본 명세서의 약학 조성물에 사용하기 적합한 보존제는, 비-제한적으로, 벤즈알코늄 클로라이드, 클로로부탄올, 파라벤 및 티메로살을 포함한다.Preservatives suitable for use in the pharmaceutical compositions of the present disclosure include, but are not limited to, benzalkonium chloride, chlorobutanol, parabens and thimerosal.

본원에서 사용되는 바와 같이, 용어 "부형제"는 본 명세서의 약학 조성물에 존재할 수 있지만 활성 성분이 아닌 물질을 지칭한다. 부형제에 대한 예로는, 비-제한적으로, 담체, 결합제, 희석제, 윤활제, 증점제, 계면활성제, 보존제, 안정화제, 유화제, 완충제, 착향제 또는 착색제를 포함한다.As used herein, the term "excipient" refers to a material that may be present in the pharmaceutical compositions of the present disclosure but is not an active ingredient. Examples of excipients include, but are not limited to, carriers, binders, diluents, lubricants, thickeners, surfactants, preservatives, stabilizers, emulsifiers, buffers, flavoring agents, or coloring agents.

용어 "희석제"는 희석 및/또는 묽게 하는 물질을 의미한다. 또한, 용어 "희석제"는 유체, 액체 또는 고체 현탁물 및/또는 혼합 매질 중 어느 하나 이상을 포함한다. 적합한 희석제의 예는 에탄올, 글리세롤 및 물을 포함한다.The term "diluent" means a diluting and/or thinning substance. Furthermore, the term "diluent" includes any one or more of a fluid, a liquid or solid suspension and/or a mixed medium. Examples of suitable diluents include ethanol, glycerol, and water.

용어 "담체"는 약학 조성물의 투여를 쉽게 만들거나, 강화하거나 또는 투여를 수행할 수 있게 하기 위해 활성 성분과 조합되는 천연, 합성, 유기, 무기일 수 있는 성분을 지칭한다. 본원에서 사용된 바와 같이, 담체는, 개체에 투여하기 적합한, 하나 이상의 혼용가능한 고체 또는 액체 충전제, 희석제 또는 캡슐화 물질일 수 있다. 적합한 담체로는, 비-제한적으로, 멸균수, 링거, 링거 락테이트, 멸균 소듐 클로라이드 용액, 등장성 식염수, 폴리알킬렌 글리콜, 수소화 나프탈렌, 및, 특히, 생체적합성 락티드 폴리머, 락티드/글리콜라이드 코폴리머 또는 폴리옥시에틸렌/폴리옥시-프로필렌 코폴리머를 포함한다. 일 구현예에서, 본 명세서의 약학 조성물은 등장성 식염수를 포함한다.The term "carrier" refers to an ingredient which may be natural, synthetic, organic or inorganic, which is combined with an active ingredient to facilitate, enhance or enable administration of the pharmaceutical composition. As used herein, a carrier can be one or more compatible solid or liquid fillers, diluents or encapsulating materials suitable for administration to a subject. Suitable carriers include, but are not limited to, sterile water, Ringer's, Ringer's lactate, sterile sodium chloride solution, isotonic saline, polyalkylene glycols, hydrogenated naphthalenes, and, in particular, biocompatible lactide polymers, lactide/glycolide copolymers or polyoxyethylene/polyoxy-propylene copolymers. In one embodiment, the pharmaceutical compositions herein comprise isotonic saline.

치료학적 용도를 위한 약학적으로 허용되는 담체, 부형제 또는 희석제는 약학 분야에 잘 알려져 있으며, 예를 들어 Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit. 1985)에 기술되어 있다.Pharmaceutically acceptable carriers, excipients or diluents for therapeutic use are well known in the art of pharmacy and are described, for example, in Remington's Pharmaceutical Sciences, Mack Publishing Co. (A. R Gennaro edit. 1985).

약학적 담체, 부형제 또는 희석제는 의도한 투여 경로 및 표준 약학 실무에 따라 선택될 수 있다.Pharmaceutical carriers, excipients or diluents may be selected depending on the intended route of administration and standard pharmaceutical practice.

일 구현예에서, 본원에 기술된 약학 조성물은 정맥내, 동맥내, 피하, 진피내 또는 근육내로 투여될 수 있다. 특정 구현예에서, 약학 조성물은 국소 투여 또는 전신 투여를 위해 제형화된다. 전신 투여는 위장관을 통한 흡수를 수반하는 장 투여 또는 비경구 투여를 포함할 수 있다. 본원에 사용된 바와 같이, "비경구 투여"는 정맥내 주사와 같이, 위장관을 통해 이루어지는 것 이외의 다른 임의의 방식으로 투여하는 것을 의미한다. 바람직한 구현예에서, 약학 조성물은 전신 투여용으로 제형화된다. 다른 바람직한 구현예에서, 전신 투여는 정맥내 투여에 의한 것이다. 본 발명의 모든 측면의 일 구현예에서, 본원에서 설명된 폴리펩타이드 또는 이의 변이체, 또는 이를 엔코딩하는 핵산 분자는 전신으로 투여된다. In one embodiment, the pharmaceutical compositions described herein can be administered intravenously, intraarterially, subcutaneously, intradermally, or intramuscularly. In certain embodiments, the pharmaceutical compositions are formulated for topical administration or systemic administration. Systemic administration can include enteral administration involving absorption through the gastrointestinal tract or parenteral administration. As used herein, "parenteral administration" means administration by any means other than via the gastrointestinal tract, such as intravenous injection. In a preferred embodiment, the pharmaceutical compositions are formulated for systemic administration. In another preferred embodiment, systemic administration is by intravenous administration. In one embodiment of all aspects of the invention, the polypeptides described herein or variants thereof, or nucleic acid molecules encoding them, are administered systemically.

본원에서 사용된 바와 같이, 용어 "공동-투여"는 여러가지 화합물 또는 조성물 (예를 들어, 면역 효과기 세포, 본원에서 설명된 폴리펩타이드 또는 이의 변이체 또는 이를 엔코딩하는 핵산 분자, 및 선택적으로 백신 항원을 엔코딩하는 RNA)을 동일한 환자에게 투여하는 것을 의미한다. 여러가지 화합물 또는 조성물은 동시에, 본질적으로 동시에, 또는 순차적으로 투여될 수 있다.As used herein, the term "co-administration" means administering multiple compounds or compositions (e.g., immune effector cells, a polypeptide described herein or a variant thereof or a nucleic acid molecule encoding the same, and optionally an RNA encoding a vaccine antigen) to the same patient. The multiple compounds or compositions can be administered simultaneously, essentially simultaneously, or sequentially.

본원에 기술된 물질, 조성물 및 방법은 질환, 예를 들어, 항원을 발현하는 질환에 걸린 세포의 존재를 특징으로 하는 질환을 가진 개체를 치료하는 데에 사용될 수 있다. 특히 바람직한 질환은 암 질환이다. 예를 들어, 만약 항원이 바이러스에서 유래되었다면, 물질, 조성물 및 방법은 상기 바이러스에 의해 야기되는 바이러스 질환의 치료에 유용할 수 있다. 만약 항원이 종양 항원이라면, 물질, 조성물 및 방법은 암 질환의 치료에 유용하고, 여기서 암 세포는 상기 종양 항원을 발현한다. The materials, compositions and methods described herein can be used to treat a subject having a disease, for example, a disease characterized by the presence of cells that express the antigen. A particularly preferred disease is cancer. For example, if the antigen is derived from a virus, the materials, compositions and methods can be useful for treating a viral disease caused by the virus. If the antigen is a tumor antigen, the materials, compositions and methods can be useful for treating a cancer disease, wherein cancer cells express the tumor antigen.

본원에 기술된 물질, 조성물 및 방법은 다양한 질환의 치료학적 또는 예방학적 치료에 사용될 수 있고, 여기서 본원에 기술된 바와 같이 면역 효과기 세포의 지원 및/또는 면역 효과기 세포의 활성은 암 및 감염성 질환과 같은 환자에 유리하다. 일 구현예에서, 본원에 기술된 물질, 조성물 및 방법은 항원과 연관된 질환의 예방학적 및/또는 치료학적 치료에 유용하다. The materials, compositions, and methods described herein may be used in the therapeutic or prophylactic treatment of a variety of diseases, wherein support of immune effector cells and/or activation of immune effector cells as described herein is beneficial to a patient, such as cancer and infectious diseases. In one embodiment, the materials, compositions, and methods described herein are useful in the prophylactic and/or therapeutic treatment of diseases associated with an antigen.

용어 "질환"은 개체의 신체에 영향을 미치는 비정상적인 병태를 지칭한다. 질환은 종종 특정 증상 및 신호와 관련있는 의학적인 상태로서 해석된다. 질환은 감염성 질환과 같은 외부 원인으로부터 기원한 인자에 의해 유발될 수 있거나, 또는 자가면역 질환과 같은 내부 기능부전에 의해 유발될 수 있다. 인간에서, "질환"은 보다 넒은 의미에서 개체와 접촉 시 질병이 병을 앓고 있는 개체에게 통증, 기능부전, 괴로움, 사회적 문제 또는 사망 또는 비슷한 문제를 유발하는 임의의 병태를 지칭하기 위해 사용된다. 보다 넓은 의미에서, 이는 때때로 상해, 불능, 장애, 증후군, 감염, 단독 증상, 일탈 행위 및 구조적 및 기능적인 비정형성 변형을 포함하며, 다른 맥락 및 다른 목적에서, 이는 구별할 수 있는 범주로 간주될 수 있다. 질환은, 여러가지 질환에 걸려 생활하면 개체의 삶에 대한 관점과 성격을 바꿀 수 있으므로, 일반적으로 개체에게 신체적으로뿐 아니라 감정적으로 영향을 미친다.The term "disease" refers to an abnormal condition affecting the body of an individual. A disease is often interpreted as a medical condition associated with specific symptoms and signs. A disease may be caused by an agent originating from an external source, such as an infectious disease, or may be caused by an internal dysfunction, such as an autoimmune disease. In humans, "disease" is used in a broader sense to refer to any condition that, when contacted by an individual, causes pain, dysfunction, distress, social problems, or death or similar problems to the individual suffering from the disease. In a broader sense, it sometimes includes injuries, disabilities, disorders, syndromes, infections, isolated symptoms, deviant behaviors, and structural and functional atypical changes, which in different contexts and for different purposes may be considered distinct categories. Disease generally affects an individual not only physically but also emotionally, as living with various diseases can change the individual's outlook on life and personality.

본 맥락에서, 용어 "치료", "치료하는" 또는 "치료학적 개입"은 질환 또는 장애와 같은 병태를 퇴치할 목적으로 개체를 관리 및 돌보는 것을 의미한다. 이 용어는 증상 또는 합병증을 완화하고/거나, 질환, 장애 또는 병태의 진행을 지연하고/거나, 증상 및 합병증을 완화 또는 경감하고/거나, 질환, 장애 또는 병태를 치유 또는 제거하는 것뿐 아니라 병태를 예방하기 위해, 치료학적으로 효과적인 화합물의 투여와 같이, 개체가 고통받는 소정의 병태에 대한 전 범위 치료를 포함하는 것으로 의도되며, 여기서 예방은 질환, 병태 또는 장애를 퇴치하기 위한 목적으로 개체를 관리 및 돌보는 것으로서 이해될 것이고, 증상 또는 합병증의 개시를 방지하기 위한 활성 화합물의 투여를 포함한다.In this context, the terms "treatment", "treating" or "therapeutic intervention" mean the care and attention of a subject for the purpose of combating a condition, such as a disease or disorder. The term is intended to encompass the full range of treatments for a given condition from which a subject is suffering, such as the administration of a therapeutically effective compound to alleviate symptoms or complications, delay the progression of the disease, disorder or condition, relieve or alleviate symptoms and complications, and/or cure or eliminate the disease, disorder or condition, as well as prevent the condition, wherein prevention will be understood as the care and attention of a subject for the purpose of combating the disease, condition or disorder, and includes the administration of an active compound to prevent the onset of symptoms or complications.

용어 "치료학적 치료"는 개체의 건강 상태를 개선하고/거나 개체의 수명을 연장(증가)하는 임의의 치료를 의미한다. 상기 치료는 개체에서 질환의 소거, 개체에서 질환 진행의 정지 또는 서행, 개체에서 질환 진행의 저해 또는 서행, 개체에서 증상의 빈도 또는 중증도 감소, 및/또는 질환을 현재 앓고 있거나 예전에 걸린 적 있는 개체에서 재발 감소일 수 있다.The term "therapeutic treatment" means any treatment that improves the health status of a subject and/or prolongs (increases) the life span of a subject. Such treatment may be elimination of a disease in a subject, arrest or slowing of the progression of a disease in a subject, inhibition or slowing of the progression of a disease in a subject, reduction in the frequency or severity of symptoms in a subject, and/or reduction in relapse in a subject currently suffering from or previously suffering from a disease.

용어 "예방학적 치료" 또는 "예방적 치료"는 개체에서 질환이 발생되는 것을 방지하기 위해 의도되는 임의의 치료를 의미한다. 용어 "예방학적 치료" 또는 "예방적 치료"는 본원에서 상호 호환적으로 사용된다.The term "prophylactic treatment" or "prophylactic treatment" means any treatment intended to prevent a disease from occurring in a subject. The terms "prophylactic treatment" or "prophylactic treatment" are used interchangeably herein.

용어 "개인" 및 "개체"는 본원에서 상호 호환적으로 사용된다. 이들 용어는, 질환 또는 장애 (예를 들어, 암)에 걸릴 수 있거나 또는 취약할 수 있는 인간 또는 기타 포유류 (예, 마우스, 랫, 토끼, 개, 고양이, 소, 돼지, 양, 말 또는 영장류)를 지칭하지만, 질환 또는 장애에 걸렸을 수 있거나 또는 걸리지 않았을 수 있다. 다수 구현예들에서, 개체는 인간이다. 달리 명시되지 않은 한, 용어 "개인" 및 "개체"는 특정 연령을 의미하지 않으며, 따라서 성인, 노인, 어린이 및 신생아를 포괄한다. 본 명세서의 구현예에서, "개체" 또는 "개인"은 "환자"이다.The terms "individual" and "subject" are used interchangeably herein. These terms refer to a human or other mammal (e.g., a mouse, rat, rabbit, dog, cat, cow, pig, sheep, horse, or primate) that may be susceptible to or may be susceptible to a disease or disorder (e.g., cancer), but may or may not have the disease or disorder. In many embodiments, the subject is a human. Unless otherwise specified, the terms "individual" and "subject" do not imply a particular age, and thus encompass adults, elderly people, children, and newborns. In embodiments herein, an "individual" or "individual" is a "patient."

용어 "환자"는 치료가 필요한 개인 또는 개체, 구체적으로 질환에 걸린 개인 또는 개체를 의미한다.The term "patient" means an individual or entity in need of treatment, specifically an individual or entity suffering from a disease.

본 명세서의 일 구현예에서, 목표는 종양 항원을 발현하는 암 세포와 같은 항원을 발현하는 질환에 걸린 세포에 대해 면역 반응을 제공하는 것 및 종양 항원과 같은 항원을 발현하는 세포를 수반하는 암 질환과 같은 질환을 치료하는 것이다.In one embodiment of the present disclosure, the goal is to provide an immune response to cells having a disease, such as cancer cells expressing an antigen, and to treat a disease, such as a cancer disease, involving cells expressing an antigen, such as a tumor antigen.

본원에서 사용된 바와 같이, "면역 반응"은 항원 또는 항원을 발현하는 세포에 대한 일체화된 신체 반응을 지칭하며, 세포성 면역 반응 및/또는 체액성 면역 반응을 지칭한다. As used herein, “immune response” refers to a coordinated body response to an antigen or cells expressing an antigen, and includes a cellular immune response and/or a humoral immune response.

“세포-매개 면역”, “세포성 면역”, “세포성 면역 반응”, 또는 유사 용어는 항원의 발현을 특징으로 하는, 특히 클래스 I 또는 클래스 II MHC로 항원을 제시하는 것을 특징으로 하는 세포에 대한 세포성 반응을 포함하는 것을 의미한다. 세포성 반응은 “헬퍼” 또는 “살상” 중 하나로서 작동하는 T 세포 또는 T 림프구로 불리는 세포에 관한 것이다. 헬퍼 T 세포 (CD4+ T 세포로도 지칭됨)는 면역 반응을 조절함으로써 중추적인 역할을 하고, 살상 세포 (세포독성 T 세포, 세포용해성 T 세포, CD8+ T 세포 또는 CTL로도 지칭됨)는 암 세포와 같은 질환에 걸린 세포를 죽여 더 많은 질환에 걸린 세포의 발생을 예방한다. The terms “cell-mediated immunity”, “cellular immunity”, “cellular immune response”, or similar terms refer to a cellular response directed against cells characterized by the expression of an antigen, particularly by presentation of the antigen with class I or class II MHC. The cellular response relates to cells called T cells or T lymphocytes, which act either as “helper” or “killer” cells. Helper T cells (also called CD4+ T cells) play a central role by regulating the immune response, while killer cells (also called cytotoxic T cells, cytolytic T cells, CD8 + T cells, or CTLs) kill diseased cells, such as cancer cells, thereby preventing the development of more diseased cells.

본 명세서는 보호적, 예방적, 예방학적 및/또는 치료학적일 수 있는 면역 반응을 고려한다. 본원에서 사용된 바와 같이, "면역 반응을 유도하다 [또는 유도하는]"는 유도하기 전 특정 항원에 대한 면역 반응이 없다는 것을 의미할 수 있거나, 또는 유도하기 전 특정 항원에 대한 면역 반응이 기저 수준으로 존재하고 유도 후 강화되는 것을 의미할 수 있다. 따라서, "면역 반응을 유도하다 [또는 유도하는]"는 "면역 반응을 강화하다 [또는 강화하는]"를 포함한다.The present disclosure contemplates immune responses that may be protective, prophylactic, prophylactic, and/or therapeutic. As used herein, "inducing [or inducing] an immune response" may mean that there is no immune response to a particular antigen prior to induction, or it may mean that an immune response to a particular antigen exists at a baseline level prior to induction and is enhanced following induction. Thus, "inducing [or inducing] an immune response" includes "potentiating [or enhancing] an immune response."

용어 "면역요법"은 면역 반응의 유도 또는 강화에 의한 질환 또는 병태의 치료를 의미한다. 용어 "면역요법"은 항원 면역화 또는 항원 백신 접종을 포함한다.The term "immunotherapy" refers to the treatment of a disease or condition by induction or enhancement of an immune response. The term "immunotherapy" includes antigen immunization or antigen vaccination.

용어 "면역화" 또는 "백신 접종"은 예를 들어, 치료학적 또는 예방학적 이유로, 면역 반응을 유도하기 위한 목적으로 항원을 개체에 투여하는 과정을 의미한다.The term "immunization" or "vaccination" refers to the process of administering an antigen to an individual for the purpose of inducing an immune response, for example, for therapeutic or prophylactic reasons.

용어 "대식 세포"는 단핵구의 분화에 의해 만들어진 식세포의 하위군을 지칭한다. 염증, 면역 사이토카인 또는 미생물 산물에 의해 활성화된 대식 세포는 비특이적으로 탐식 작용을 수행하여, 수소분해성 및 산화성 공격에 의해 대식 세포 내 외인성 병원체를 사멸시켜 병원체를 분해한다. 분해된 단백질로부터 유래된 펩타이드는 대식 세포의 세포 표면 상에 제시되고, 이는 T 세포에 의해 인지될 수 있으며, B 세포 표면 상의 항체와 직접 상호작용하여, T 및 B 세포를 활성화하고 면역 반응을 추가적으로 자극할 수 있다. 대식 세포는 항원 제시 세포 클래스에 속한다. 일 구현예에서, 대식 세포는 비장 대식 세포이다.The term "macrophage" refers to a subset of phagocytes produced by differentiation of monocytes. Macrophages activated by inflammation, immune cytokines, or microbial products perform nonspecific phagocytosis, degrading pathogens by killing exogenous pathogens within the macrophage by hydrolytic and oxidative attack. Peptides derived from degraded proteins are presented on the cell surface of the macrophage, which can be recognized by T cells and can directly interact with antibodies on the surface of B cells, thereby activating T and B cells and further stimulating an immune response. Macrophages belong to the class of antigen-presenting cells. In one embodiment, the macrophages are splenic macrophages.

용어 "수지상 세포" (DC)는 항원 제시 세포 클래스에 속하는 식세포의 다른 하위종을 지칭한다. 일 구현예에서, 수지상 세포는 조혈 골수 전구 세포로부터 유래된다. 이들 전구 세포는 먼저 미성숙 수지상 세포로 변환된다. 이들 미성숙 세포는 높은 탐식 활성과 낮은 T 세포 활성화 가능성을 특징으로 한다. 미성숙 수지상 세포는 바이러스 및 박테리아와 같은 병원체에 대해 주변 환경을 계속적으로 샘플링한다. 일단 이들이 제시가능한 항원과 접촉하게 되면, 그 세포는 성숙한 수지상 세포로 활성화되어, 비장 또는 림프절로의 이동하기 시작한다. 미성숙 수지상 세포는 병원체를 탐식하여, 이의 단백질을 작은 조각으로 분해하고, 성숙화되면 이들 조각들을 MHC 분자를 이용해 세포 표면에 제시한다. 동시에, 이들 세포는 CD80, CD86 및 CD40과 같은 T 세포 활성화에서 공동-수용체로서 작용하는 세포-표면 수용체를 상향 조절하여, 이의 T 세포 활성화 능력을 크게 강화한다. 이들은 또한 수지상 세포가 혈류를 통해 비장으로 이동하거나 또는 림프 시스템을 통해 림프절로 이동하게 유도하는 주화성 수용체인 CCR7을 상향 조절한다. 여기서, 이들은 항원-제시 세포로 작용하며, 비-항원 특이적인 공동-자극 신호와 동시에, 헬퍼 T 세포 및 살상 T 세포뿐 아니라 항원 제시에 의해 B 세포를 활성화한다. 따라서, 수지상 세포는 T 세포- 또는 B 세포-관련 면역 반응을 적극적으로 유도할 수 있다. 일 구현예에서, 수지상 세포는 비장의 수지상 세포이다.The term "dendritic cell" (DC) refers to another subset of phagocytes belonging to the antigen-presenting cell class. In one embodiment, dendritic cells are derived from hematopoietic bone marrow progenitor cells. These progenitor cells are first transformed into immature dendritic cells. These immature cells are characterized by high phagocytic activity and low T cell activation potential. Immature dendritic cells continuously sample their surrounding environment for pathogens such as viruses and bacteria. Once they come into contact with a presentable antigen, the cells are activated into mature dendritic cells and begin to migrate to the spleen or lymph nodes. Immature dendritic cells engulf pathogens, degrade their proteins into small fragments, and upon maturation present these fragments on the cell surface using MHC molecules. At the same time, these cells upregulate cell-surface receptors that act as co-receptors in T cell activation, such as CD80, CD86, and CD40, which greatly enhance their T cell activation potential. They also upregulate CCR7, a chemotactic receptor that causes dendritic cells to migrate to the spleen via the bloodstream or to lymph nodes via the lymphatic system. Here, they act as antigen-presenting cells, activating B cells by antigen presentation as well as helper T cells and killer T cells, in conjunction with non-antigen-specific co-stimulatory signals. Thus, dendritic cells can actively induce T cell- or B cell-related immune responses. In one embodiment, the dendritic cells are splenic dendritic cells.

용어 "항원 제시 세포" (APC)는 세포의 표면 상에 (또는 에) 적어도 하나의 항원 또는 항원 단편을 나열, 획득 및/또는 제시할 수 있는 다양한 세포이다. 항원-제시 세포는 전문적 항원 제시 세포 및 비-전문적 항원 제시 세포로 구분될 수 있다.The term "antigen presenting cell" (APC) refers to a variety of cells capable of displaying, acquiring, and/or presenting at least one antigen or antigen fragment on (or at) their surface. Antigen-presenting cells can be divided into professional antigen-presenting cells and non-professional antigen-presenting cells.

용어 "전문적 항원 제시 세포"는 나이브 T 세포와의 상호작용에 필요한 주 조직적합성 복합체 클래스 II (MHC 클래스 II) 분자를 구성적으로 발현하는 항원 제시 세포이다. 만약 T 세포가 항원 제시 세포의 막 상에 MHC 클래스 II 분자 복합체와 상호작용하면, 항원 제시 세포는 T 세포의 활성화를 유도하는 공동-자극성 분자를 생산한다. 전문적 항원 제시 세포는 수지상 세포 및 대식 세포를 포함한다.The term "professional antigen-presenting cell" is an antigen-presenting cell that constitutively expresses major histocompatibility complex class II (MHC class II) molecules required for interaction with naïve T cells. If a T cell interacts with a complex of MHC class II molecules on the membrane of the antigen-presenting cell, the antigen-presenting cell produces co-stimulatory molecules that induce activation of the T cell. Professional antigen-presenting cells include dendritic cells and macrophages.

용어 "비-전문적 항원 제시 세포"는 인터페론-감마와 같은 특정 사이토카인에 의한 자극시 MHC 클래스 II 분자를 구성적으로 발현하지 않는 항원 제시 세포를 지칭한다. 예를 들어, 비-전문적 항원 제시 세포는 섬유모세포, 흉선 상피 세포, 갑상선 상피 세포, 신경교 세포, 췌장 베타 세포 또는 혈관 내피 세포를 포함한다.The term "non-professional antigen-presenting cell" refers to an antigen-presenting cell that does not constitutively express MHC class II molecules upon stimulation by specific cytokines, such as interferon-gamma. For example, non-professional antigen-presenting cells include fibroblasts, thymic epithelial cells, thyroid epithelial cells, glial cells, pancreatic beta cells, or vascular endothelial cells.

"항원 가공 처리"는 항원을 처리 산물로 분해하는 것을 의미하며, 처리 산물은 상기 항원의 단편 (예, 단백질의 펩타이드로의 분해) 및 특정 T 세포에 대한 항원 제시 세포와 같이 세포에 의해 제시하기 위한 MHC 분자와 하나 이상의 이들 단편의 (예, 결합을 통한) 조합이다."Antigen processing" means the breakdown of an antigen into processing products, which are fragments of said antigen (e.g., breakdown of a protein into peptides) and combinations of one or more of these fragments with an MHC molecule (e.g., via binding) for presentation by a cell, such as an antigen presenting cell, to a particular T cell.

용어 "항원을 수반하는 질환"은 항원이 관련된 임의의 질환, 예를 들어 항원의 존재를 특징으로 하는 질환을 의미한다. 항원을 수반한 질환은 감염성 질환 또는 암 질환 또는 단순 암일 수 있다. 전술한 바와 같이, 항원은 종양-관련 항원, 바이러스 항원 또는 박테리아 항원과 같은 질환-관련 항원일 수 있다. 일 구현예에서, 항원을 수반하는 질환은, 바람직하게는 세포의 표면에, 항원을 발현하는 세포를 수반하는 질환이다. The term "disease involving an antigen" means any disease involving an antigen, for example a disease characterized by the presence of an antigen. The disease involving an antigen may be an infectious disease or a cancer disease or a simple cancer. As mentioned above, the antigen may be a disease-associated antigen, such as a tumor-associated antigen, a viral antigen or a bacterial antigen. In one embodiment, the disease involving an antigen is a disease involving cells expressing the antigen, preferably on the surface of the cells.

용어 "감염성 질환"은 개체에서 개체로 또는 유기체에서 유기체로 전파될 수 있으며, 미생물 물질 (예, 감기)에 의해 유발되는 임의의 질환을 지칭한다. 감염성 질환은 당해 기술 분야에 공지되어 있으며, 예를 들어 바이러스, 박테리아 및 기생충에 의해 각각 유발되는 바이러스성 질환, 박테리아성 질환 또는 기생충성 질환을 포함한다. 이런 점에서, 감염성 질환은, 예를 들어, 간염, 성적으로 전파되는 질환 (예, 클라미디아 또는 임질), 결핵, HIV/후천성 면역결핍 증후군 (AIDS), 디프테리아, B형 간염, C형 간염, 콜레라, 중증 급성 호흡 증후군 (SARS), 조류 독감 및 인플루엔자일 수 있다.The term "infectious disease" refers to any disease that can be transmitted from subject to subject or from organism to organism and is caused by a microbial agent (e.g., a cold). Infectious diseases are known in the art and include, for example, viral, bacterial or parasitic diseases caused by viruses, bacteria and parasites, respectively. In this regard, an infectious disease can be, for example, hepatitis, sexually transmitted diseases (e.g., chlamydia or gonorrhea), tuberculosis, HIV/acquired immunodeficiency syndrome (AIDS), diphtheria, hepatitis B, hepatitis C, cholera, severe acute respiratory syndrome (SARS), avian influenza and influenza.

용어 "암 질환" 또는 "암"은 개체에서 전형적으로 통제되지 않은 세포 증식을 특징으로 하는 병리학적 병태를 지칭하거나 또는 이를 의미한다. 암의 예로는, 비-제한적으로, 암종, 림프종, 모세포종, 육종 및 백혈병을 포함한다. 보다 상세하게는, 이러한 암의 예로는 골암, 혈액암, 폐암, 간암, 췌장암, 피부암, 두경부암, 피부 또는 안내 흑색종, 자궁암, 난소암, 직장암, 항문 부위의 암, 위암, 대장암, 유방암, 전립선암, 자궁암, 성 및 생식 기관의 암종, 호지킨 질환, 식도암, 소장암, 내분비계 암, 갑상선암, 부갑상선암, 부신암, 연조직 육종, 방광암, 신장암, 신장 세포 암종, 신우 암종, 중추 신경계 (CNS)의 신생물, 신경외배엽 암, 척추 종양, 신경교종, 수막종 및 뇌하수체 선종을 포함한다. 본 명세서에 따를 때, 용어 "암"은 암 전이를 또한 포함한다.The term "cancer disease" or "cancer" refers to or means a pathological condition that is typically characterized by uncontrolled cell proliferation in an organism. Examples of cancers include, but are not limited to, carcinomas, lymphomas, blastomas, sarcomas, and leukemias. More specifically, examples of such cancers include bone cancer, blood cancer, lung cancer, liver cancer, pancreatic cancer, skin cancer, head and neck cancer, cutaneous or intraocular melanoma, uterine cancer, ovarian cancer, rectal cancer, cancer of the anal area, stomach cancer, colon cancer, breast cancer, prostate cancer, uterine cancer, carcinomas of the sexual and reproductive organs, Hodgkin's disease, esophageal cancer, small intestinal cancer, endocrine cancer, thyroid cancer, parathyroid cancer, adrenal cancer, soft tissue sarcoma, bladder cancer, kidney cancer, renal cell carcinoma, renal pelvic carcinoma, neoplasms of the central nervous system (CNS), neuroectodermal cancers, spinal tumors, gliomas, meningiomas, and pituitary adenomas. As used herein, the term “cancer” also includes cancer metastasis.

암 치료에서 병용 전략 (combination strategy)은, 단일요법 방식의 효과 보다 더 높은 것으로 간주될 수 있는, 달성되는 상승 효과로 인해 적합할 수 있다. 일 구현예에서, 약학 조성물은 면역요법제와 함께 투여된다. 본원에서 사용된 바와 같이, "면역요법제"는 특이적인 면역 반응 및/또는 면역 효과기 기능(들)을 활성화하는데 관여할 수 있는 임의 물질을 의미한다. 본 명세서는 면역요법제로서 항체의 사용을 고려한다. 이론으로 결부시키고자 하는 것은 아니지만, 항체는, 세포자살 유도, 신호 전달 경로의 구성성분의 차단 또는 종양 세포의 증식 저해를 포함하는, 다양한 기전을 통해 암 세포에 대해 치료학적 효과를 달성할 수 있다. 특정 구현예에서, 항체는 단일클론 항체이다. 단일클론 항체는 항체-의존적인 세포 매개 세포독성 (ADCC)을 통해 세포 사멸을 유도할 수 있거나, 보체 단백질에 결합하여, 보체 의존적인 세포독성 (CDC)으로 알려진 직접적인 세포 독성을 유도할 수 있다. 본 발명과 조합하여 사용될 수 있는 항-암 항체 및 잠재적인 항체 표적(괄호 안)에 대한 비-제한적인 예로는, 아바고보맵 (Abagovomab) (CA-125), 압시시맵 (Abciximab) (CD41), 아데카투무맵 (Adecatumumab)(atumumab) (EpCAM), 아푸투주맵 (Afutuzumab) (CD20), 알라시주맵 (Alacizumab pegol) (VEGFR2), 알투모맵 펜테테이트 (Altumomab pentetate) (CEA), 아마투시맵 (Amatuximab) (MORAb-009), 아나투모맵 마페나톡스 (Anatumomab mafenatox) (TAG-72), 아폴리주맵 (Apolizumab) (HLA-DR), 아르시투모맵 (Arcitumomab) (CEA), 아테졸리주맵 (Atezolizumab) (PD-L1), 바비투시맵 (Bavituximab) (포스파티딜세린), 벡투모맵 (Bectumomab) (CD22), 벨리무맵 (Belimumab) (BAFF), 베바시주맵 (Bevacizumab) (VEGF-A), 비바투주맵 메르탄신 (Bivatuzumab mertansine)(CD44 v6), 빌리나투모맵 (Blinatumomab) (CD19), 브렌투시맵 베도틴 (Brentuximab vedotin) (CD30 TNFRSF8), 칸투주맵 메르탄신 (Cantuzumab mertansin) (mucin CanAg), 칸투주맵 라브탄신 (Cantuzumab ravtansine) (MUC1), 카프로맵 펜데티드 (Capromab pendetide) (전립선 암종 세포), 카를루맵 (Carlumab) (CNT0888), 카투맥소맵 (Catumaxomab) (EpCAM, CD3), 세투시맵 (Cetuximab) (EGFR), 시타투주맵 보가톡스 (Citatuzumab bogatox) (EpCAM), 시수투무맵 (Cixutumumab) (IGF-1 수용체), 클라우디시맵 (Claudiximab) (Claudin), 클리바투주맵 테트라세탄 (Clivatuzumab tetraxetan) (MUC1), 코나투무맵 (Conatumumab) (TRAIL-R2), 다세투주맵 (Dacetuzumab) (CD40), 달로투주맵 (Dalotuzumab) (인슐린-유사 성장인자 I 수용체), 데노수맵 (Denosumab) (RANKL), 데투모맵 (Detumomab) (B-림프종 세포), 드로지투맵 (Drozitumab) (DR5), 에크로멕시맵 (Ecromeximab) (GD3 강글리오시드), 에드레콜로맵 (Edrecolomab) (EpCAM), 엘로투주맵 (Elotuzumab) (SLAMF7), 에나바투주맵 (Enavatuzumab) (PDL192), 엔시투시맵 (Ensituximab) (NPC-1C), 에프라투주맵 (Epratuzumab) (CD22), 에르투막소맵 (Ertumaxomab) (HER2/neu, CD3), 에타라시주맵 (Etaracizumab) (인테그린 αγβ3), 파를레투주맵 (Farletuzumab)(폴레이트 수용체 1), FBTA05 (CD20), 피클라투주맵 (Ficlatuzumab) (SCH 900105), 피기투무맵 (Figitumumab) (IGF-1 수용체), 플란보투맵 (Flanvotumab) (당단백질 75), 프레솔리무맵 (Fresolimumab) (TGF-β), 갈리시맵 (Galiximab) (CD80), 가니투맵 (Ganitumab) (IGF-I), 겜투주맵 오조가미신 (Gemtuzumab ozogamicin) (CD33), 게보키주맵 (Gevokizumab) (IL1β), 기렌투시맵 (Girentuximab) (카보닉 안하이드라제 9 (CA-IX)), 글렘바투무맵 베도틴 (Glembatumumab vedotin) (GPNMB), 이브리투모맵 티우세탄 (Ibritumomab tiuxetan) (CD20), 이크루쿠맵 (Icrucumab) (VEGFR-1), 이고보마 (Igovoma) (CA-125), 인다투시맵 라브탄신 (Indatuximab ravtansine) (SDC1), 인테투무맵 (Intetumumab) (CD51), 이노투주맵 오조가미신 (Inotuzumab ozogamicin) (CD22), 이필리무맵 (Ipilimumab) (CD 152), 이라투무맵 (Iratumumab) (CD30), 라베투주맵 (Labetuzumab) (CEA), 렉사투무맵 (Lexatumumab) (TRAIL-R2), 리비비루맵 (Libivirumab) (B형 간염 표면 항원), 린투주맵 (Lintuzumab) (CD33), 로르보투주맵 메르탄신 (Lorvotuzumab mertansine) (CD56), 루카투무맵 (Lucatumumab) (CD40), 루밀리시맵 (Lumiliximab) (CD23), 마파투무맵 (Mapatumumab) (TRAIL-R1), 마투주맵 (Matuzumab) (EGFR), 메폴리주맵 (Mepolizumab) (IL5), 밀라투주맵 (Milatuzumab) (CD74), 미투모맵 (Mitumomab) (GD3 강글리오시드), 모가물리주맵 (Mogamulizumab) (CCR4), 모세투모맵 파수도톡스 (Moxetumomab pasudotox) (CD22), 나콜로맵 타페나톡스 (Nacolomab tafenatox) (C242 항원), 나프투모맵 에스타페나톡스 (Naptumomab estafenatox) (5T4), 나마투맵 (Namatumab) (RON), 넥시투무맵 (Necitumumab) (EGFR), 니모투주맵 (Nimotuzumab) (EGFR), 니볼루맵 (Nivolumab) (IgG4), 요파투무맵 (Ofatumumab) (CD20), 올라라투맵 (Olaratumab) (PDGF-R a), 오나르투주맵 (Onartuzumab) (인간 스캐터 인자 수용체 키나제 (human scatter factor receptor kinase)), 오포르투주맵 모나톡스 (Oportuzumab monatox) (EpCAM), 오레고보맵 (Oregovomab) (CA-125), 옥셀루맵 (Oxelumab) (OX-40), 파니투무맵 (Panitumumab) (EGFR), 파트리투맵 (Patritumab) (HER3), 펨투모마 (Pemtumoma) (MUC1), 페르투주마 (Pertuzuma) (HER2/neu), 핀투모맵 (Pintumomab) (선암종 항원), 프리투무맵 (Pritumumab) (비멘틴 (vimentin)), 라코투모맵 (Racotumomab) (N-글리콜릴뉴라민산), 라드레투맵 (Radretumab) (파이브로넥틴 엑스트라 도메인-B), 라피비루맵 (Rafivirumab) (광견병 바이러스 당단백질), 라무시루맵 (Ramucirumab) (VEGFR2), 릴로투무맵 (Rilotumumab) (HGF), 리투시맵 (Rituximab) (CD20), 로바투무맵 (Robatumumab) (IGF-1 수용체), 살말리주맵 (Samalizumab) (CD200), 시브로투주맵 (Sibrotuzumab) (FAP), 실투시맵 (Siltuximab) (IL6), 타발루맵 (Tabalumab) (BAFF), 타카투주맵 테트라세탄 (Tacatuzumab tetraxetan)(α-페토프로테인), 타플리투모맵 파프톡스 (Taplitumomab paptox) (CD 19), 테나투모맵 (Tenatumomab) (테나신 C), 테프로투무맵 (Teprotumumab) (CD221), 티실리무맵 (Ticilimumab) (CTLA- 4), 티가투주맵 (Tigatuzumab) (TRAIL-R2), TNX-650 (IL13), 토시투모맵 (Tositumomab) (CD20), 트라스투주맵 (Trastuzumab) (HER2/neu), TRBS07 (GD2), 트레멜리무맵 (Tremelimumab) (CTLA-4), 투코투주맵 셀모루킨 (Tucotuzumab celmoleukin) (EpCAM), 우블리투시맵 (Ublituximab) (MS4A1), 우레루맵 (Urelumab) (4-1 BB), 볼로시시맵 (Volociximab) (인테그린 α5β1), 보투무맵 (Votumumab) (종양 항원 CTAA 16.88), 잘루투무맵 (Zalutumumab) (EGFR) 및 자놀리무맵 (Zanolimumab) (CD4)을 포함한다.In cancer treatment, combination strategies may be appropriate due to the synergistic effects achieved, which may be considered to be greater than the efficacy of monotherapy. In one embodiment, the pharmaceutical composition is administered in combination with an immunotherapeutic agent. As used herein, "immunotherapeutic agent" means any substance capable of activating a specific immune response and/or immune effector function(s). The present disclosure contemplates the use of antibodies as immunotherapeutic agents. While not wishing to be bound by theory, it is believed that antibodies may achieve therapeutic effects against cancer cells through a variety of mechanisms, including inducing apoptosis, blocking components of signal transduction pathways, or inhibiting proliferation of tumor cells. In certain embodiments, the antibody is a monoclonal antibody. Monoclonal antibodies may induce cell death via antibody-dependent cell-mediated cytotoxicity (ADCC), or may bind to complement proteins to induce direct cytotoxicity, known as complement-dependent cytotoxicity (CDC). Non-limiting examples of anti-cancer antibodies and potential antibody targets (in parentheses) that may be used in combination with the present invention include Abagovomab (CA-125), Abciximab (CD41), Adecatumumab (atumumab) (EpCAM), Afutuzumab (CD20), Alacizumab pegol (VEGFR2), Altumomab pentetate (CEA), Amatuximab (MORAb-009), Anatumomab mafenatox (TAG-72), Apolizumab (HLA-DR), Arcitumomab (CEA), Atezolizumab (Atezolizumab) (PD-L1), Bavituximab (phosphatidylserine), Bectumomab (CD22), Belimumab (BAFF), Bevacizumab (VEGF-A), Bivatuzumab mertansine (CD44 v6), Blinatumomab (CD19), Brentuximab vedotin (CD30 TNFRSF8), Cantuzumab mertansin (mucin CanAg), Cantuzumab ravtansine (MUC1), Capromab pendetide (prostate cancer cells), Carlumab (CNT0888), Catumaxomab (EpCAM, CD3), Cetuximab (EGFR), Ciatuzumab bogatox (EpCAM), Cisutumumab (IGF-1 receptor), Claudiximab (Claudin), Clivatuzumab tetraxetan (MUC1), Conatumumab (TRAIL-R2), Dacetuzumab (CD40), Dalotuzumab (Insulin-like growth factor I receptor), Denosumab (RANKL), Detumomab (B-lymphoma cells), Drozitumab (DR5), Ecromeximab (GD3 ganglioside), Edrecolomab (EpCAM), Elotuzumab (SLAMF7), Enavatuzumab (PDL192), Ensituximab (NPC-1C), Epratuzumab (CD22), Ertumaxomab (HER2/neu, CD3), Etaracizumab (integrin αγβ3), Farletuzumab (folate receptor 1), FBTA05 (CD20), Ficlatuzumab (SCH 900105), Figitumumab (IGF-1 receptor), Flavoutumab (glycoprotein 75), Fresolimumab (TGF-β), Galiximab (CD80), Ganitumab (IGF-I), Gemtuzumab ozogamicin (CD33), Gevokizumab (IL1β), Girentuximab (carbonic anhydrase 9 (CA-IX)), Glembatumumab vedotin (GPNMB), Ibritumomab tiuxetan (CD20), Icrucumab (VEGFR-1), Igovoma (CA-125), Indatusimab labtansine (Indatuximab ravtansine) (SDC1), Intetumumab (CD51), Inotuzumab ozogamicin (CD22), Ipilimumab (CD 152), Iratumumab (CD30), Labetuzumab (CEA), Lexatumumab (TRAIL-R2), Libivirumab (hepatitis B surface antigen), Lintuzumab (CD33), Lorvotuzumab mertansine (CD56), Lucatumumab (CD40), Lumiliximab (CD23), Mapatumumab (Mapatumumab) (TRAIL-R1), Matuzumab (EGFR), Mepolizumab (IL5), Milatuzumab (CD74), Mitumomab (GD3 ganglioside), Mogamulizumab (CCR4), Moxetumomab pasudotox (CD22), Nacolomab tafenatox (C242 antigen), Naptumomab estafenatox (5T4), Namatumab (RON), Necitumumab (EGFR), Nimotuzumab (EGFR), Nivolumab (IgG4), Ofatumumab (CD20), Olaratumab (PDGF-R a), Onartuzumab (human scatter factor receptor kinase), Oportuzumab monatox (EpCAM), Oregovomab (CA-125), Oxelumab (OX-40), Panitumumab (EGFR), Patritumab (HER3), Pemtumoma (MUC1), Pertuzuma (HER2/neu), Pintumomab (adenocarcinoma antigen), Pritumumab (vimentin), Racotumomab (N-glycolylneuraminic acid), Radretumab (fibronectin extra domain-B), Rafivirumab (rabies virus glycoprotein), Ramucirumab (VEGFR2), Rilotumumab (HGF), Rituximab (CD20), Robatumumab (IGF-1 receptor), Salmalizumab (CD200), Sibrotuzumab (FAP), Siltuximab (IL6), Tabalumab (BAFF), Tacatuzumab tetraxetan (α-fetoprotein), Taplitumomab paptox (CD 19), Tenatumomab (Tenacin C), Teprotumumab (CD221), Ticilimumab (CTLA-4), Tigatuzumab (TRAIL-R2), TNX-650 (IL13), Tositumomab (CD20), Trastuzumab (HER2/neu), TRBS07 (GD2), Tremelimumab (CTLA-4), Tucotuzumab celmoleukin (EpCAM), Ublituximab (MS4A1), Urelumab (4-1 BB), Volociximab (integrin α5β1), Votumumab (Votumumab) (tumor antigen CTAA 16.88), Zalutumumab (Zalutumumab) (EGFR) and Zanolimumab (Zanolimumab) (CD4).

본원에 참조된 문헌 및 연구에 대한 언급은 임의의 전술한 내용이 선행 기술 분야와 관련 있다는 인정으로서 의도되진 않는다. 이들 문헌의 내용에 대한 모든 언급은 출원인에게 이용가능한 정보를 기반으로 하며, 이들 문헌의 내용에 대한 정확성에 관한 어떠한 인정으로도 간주되지 않는다.The reference to the literature and studies referred to herein is not intended as an admission that any of the aforementioned matters is relevant to the prior art. Any reference to the contents of these documents is based on information available to the applicant and is not to be construed as any admission as to the accuracy of the contents of these documents.

후술한 내용은 당해 기술 분야의 당업자가 다양한 구현예들을 만들고 이용할 수 있게 하기 위해 제공된다. 구체적인 장치, 기법 및 이용에 관한 설명은 단지 예로서 제공된다. 본원에 기술된 예에 대한 다양한 변형들이 당해 기술 분야의 당업자들에게 용이하게 자명할 것이며, 본원에 정의된 일반적인 원리는 다양한 구현예들의 사상 및 범위로부터 이탈하지 않으면서 다른 예 및 활용에 적용될 수 있다. 따라서, 다양한 구현예들은 본원에 기술되고 보여진 예들로 한정하고자 하는 것은 아니며, 청구항과 일치하는 범위에 부합되는 것으로 의도된다.The following description is provided to enable those skilled in the art to make and use various embodiments. The descriptions of specific devices, techniques, and applications are provided by way of example only. Various modifications to the embodiments described herein will be readily apparent to those skilled in the art, and the general principles defined herein may be applied to other embodiments and applications without departing from the spirit and scope of the various embodiments. Accordingly, the various embodiments are not intended to be limited to the embodiments described and shown herein, but are intended to be consistent with the scope consistent with the claims.

실시예Example

1. 재료 및 방법1. Materials and Methods

1.1 쥐와 조직 배양1.1 Mice and Tissue Culture

야생형 C57BL/6 마우스는 Samtako Bio Korea (오산, 한국)에서 공급하였다. 1차 골수 유래 대식세포(BMDM)는 생쥐에서 채취하여 M-CSF(R&D Systems, 416-ML)의 존재 하에 DMEM에서 3-5일 동안 배양하였다. HEK293T 세포(ATCC-11268; American Type Culture Collection)는 10% FBS(Gibco), 비필수 아미노산, 피루브산 나트륨, 스트렙토마이신(100μg/mL) 및 페니실린(100IU/mL)을 포함하는 DMEM(Gibco)에서 배양하였다.Wild-type C57BL/6 mice were supplied by Samtako Bio Korea (Osan, Korea). Primary bone marrow-derived macrophages (BMDMs) were collected from mice and cultured in DMEM with M-CSF (R&D Systems, 416-ML) for 3–5 days. HEK293T cells (ATCC-11268; American Type Culture Collection) were cultured in DMEM (Gibco) containing 10% FBS (Gibco), nonessential amino acids, sodium pyruvate, streptomycin (100 μg/mL), and penicillin (100 IU/mL).

1.2 시약 및 항체1.2 Reagents and Antibodies

LPS(Escherichia coli O111:B4, tlrl-eblps), Adenosine 5'-triphosphate(ATP, tlrl-atpl), Nigericin 및 DSS(Dextran Sulfate Sodium)는 Invivogen에서 구입하였다. Actin (I-19), ASC (N-15-R), IL-18 (H-173-Y), caspase-1 p10 (M-20), HA (12CA5), Flag (D-8), GST(B-14), His (H-3) 및 V5 (C-9)는 Santa Cruz Biotechnology에서 구입하였다. IL-1β와 NLRP3(AG-20B-0014)는 각각 R&D system과 Adipogen의 제품이었다.LPS ( Escherichia coli O111:B4, tlrl-eblps), adenosine 5'-triphosphate (ATP, tlrl-atpl), nigericin, and dextran sulfate sodium (DSS) were purchased from Invivogen. Actin (I-19), ASC (N-15-R), IL-18 (H-173-Y), caspase-1 p10 (M-20), HA (12CA5), Flag (D-8), GST (B-14), His (H-3), and V5 (C-9) were purchased from Santa Cruz Biotechnology. IL-1β and NLRP3 (AG-20B-0014) were from R&D system and Adipogen, respectively.

1.3 플라스미드 구성1.3 Plasmid construction

GST-Rv0747, Flag-NLRP3, V5-ASC 및 HA-Ub플라스미드는 Addgene에서 구입하였다. 야생형 NLRP3 (Int. J. Mol. Sci. 2020, 21, 8437)는 1036개의 아미노산 길이이며, ΔPYD는 wtNLRP3 중 93-1036, ΔNACHT는 wtNLPR3 중 1-219, 및 537-1036, ΔLRR은 wtNLRP3 중 1-741을 사용하여, 전체 길이(FL) NLRP3는 및 돌연변이(ΔPYD, ΔNACHT, 및 ΔLRR)를 발현하기 위한 플라스미드를 pcDNA3 벡터에 삽입하여 사용하였다. Rv0747 (1-801, 1-93, 94-200, 201-300, 301-400, 401-500, 501-600, 601-700, 701-800)의 다른 영역을 암호화하는 플라스미드는 FL Rv0747 cDNA로부터 PCR을 통해 증폭하고 BamHI과 NotI 부분에서 측면에 N-말단 GST 에피토프 태그를 암호화하는 pEBG 파생물에 하위 복제하였다. ASC(1-195, 1-91, 107-195)의 다른 영역을 다른 영역을 암호화하는 플라스미드는 FL ASC cDNA에서 PCR 증폭을 통해 생성하였고 BamHI과 NotI 부분 사이에 C-말단 Flag 태그를 암호화하는 pEF 파생물에 하위 복제하였다. pEBG-GST 포유류 융합 벡터와 pEF-IRES-Puro 발현 벡터를 사용하여 포유류 세포에서 플라스미드에 암호화된 모든 일시적인 단백질들을 생성하였다. 모든 구조는 ABI PRISM 377 자동 DNA 염기서열 분석기를 사용하여 원래 염기서열과 100% 동일함을 보장했다.GST-Rv0747, Flag-NLRP3, V5-ASC, and HA-Ub plasmids were purchased from Addgene. Wild-type NLRP3 (Int. J. Mol. Sci. 2020, 21, 8437) is 1036 amino acids long, ΔPYD was 93-1036 in wtNLRP3, ΔNACHT was 1-219 and 537-1036 in wtNLRP3, and ΔLRR was 1-741 in wtNLRP3. Plasmids for expressing full-length (FL) NLRP3 and mutants (ΔPYD, ΔNACHT, and ΔLRR) were inserted into the pcDNA3 vector. Plasmids encoding different regions of Rv0747 (1-801, 1-93, 94-200, 201-300, 301-400, 401-500, 501-600, 601-700, 701-800) were amplified by PCR from FL Rv0747 cDNA and cloned downstream into a pEBG derivative encoding an N-terminal GST epitope tag flanked by BamHI and NotI sites. Plasmids encoding different regions of ASC (1-195, 1-91, 107-195) were generated by PCR amplification from FL ASC cDNA and cloned downstream into a pEF derivative encoding a C-terminal Flag tag between the BamHI and NotI sites. All transient proteins encoded in the plasmids were produced in mammalian cells using the pEBG-GST mammalian fusion vector and the pEF-IRES-Puro expression vector. All constructs were verified to be 100% identical to the original sequence using an ABI PRISM 377 automated DNA sequencer.

1.4 펩타이드1.4 Peptides

9R와 결합된 NLRP3 또는 ASC 펩타이드는 Peptron(대전, 대한민국)에서 세포 내 바람직하지 않은 반응을 피하기 위해 초산염 형태로 제조 및 정제하여 제공받았다. 내독소 수치는 Limulus amebocyte lysate test (Charles River Endosafe ®Endochrome-K™USA)으로 측정하였으며 실험에서 사용된 펩타이드의 농도는 3-5pg/mL 미만이었다.NLRP3 or ASC peptides conjugated with 9R were provided by Peptron (Daejeon, Republic of Korea), manufactured and purified in acetate form to avoid undesirable intracellular reactions. Endotoxin levels were measured by Limulus amebocyte lysate test (Charles River Endosafe ®Endochrome-K™USA), and the concentrations of peptides used in the experiments were less than 3-5 pg/mL.

1.5 GST 풀다운, 면역 블롯 및 면역 침강 분석1.5 GST pulldown, immunoblot and immunoprecipitation assays

293T와 BMDM 세포로 GST 풀다운, 웨스턴 블롯, 공동 면역 침강 분석을 실시하였다. GST 풀다운을 하기 위해 293T 세포를 모으고 단백질 분해 효소 억제제 칵테일(Roche, Basal, CH)가 추가된 NP40 완충액으로 용해시켰다. 원심분리 후, 상층액은 단백질 A/G 비즈를 사용하여 4℃에서 2시간 동안 전처리하였고, 전처리된 용해액은 glutathione이 결합된 Sepharose 비즈(Amersham Biosciences, Amersham, UK)의 슬러리 50%와 결합하여 4℃에서 4시간동안 결합 반응을 배양하였다. 침전물은 용해 완충액으로 완전히 헹구었다. Glutathione 비즈에 결합된 단백질은 sodium dodecyl sulfate (SDS) 로딩 완충액에서 5분동안 끓임으로써 용출하였다. 면역침강 분석을 하기 위해 세포를 채취하여 단백질 분해 효소 억제제 칵테일(Roche, Basal, CH)가 추가된 NP40 완충액으로 용해시켰다. 전세포 용해제는 단백질 A/G agarose 비즈로 4℃에서 1시간 동안 전처리한 후 처리된 항체들로 면역 침전시켰다. 일반적으로 1mL의 세포 용해액은 4℃에서 8~12시간 동안 1-4μg의 항체로 처리하였다. 단백질 A/G agarose 비즈로 6시간 동안 배양 후, 면역침전물은 용해 완충액으로 충분히 세척하고 SDS 로딩 완충액으로 5분간 끓여서 용출하였다. 폴리펩타이드는 SDS-폴리아크릴아마이드 겔 전기영동에 의해 분리된 후 면역블롯팅 (IB) (Bio-Rad, Hercules, CA, USA)을 위해 PVDF 막으로 전달시켰다. 면역 검출을 하기 위해서는 특정 항체가 필요하고 표적에 결합된 항체를 시각화 하기 위해서 Chemiluminescence (ECL; Millipore, MA, USA)를 사용하였다. 그리고 이를 검출하기 위해 Vilber chemiluminescence analyzer를 사용하였다(Fusion SL3; Vilber Lourmat, Coleegien, France).GST pull-down, western blot, and co-immunoprecipitation assays were performed with 293T and BMDM cells. For GST pull-down, 293T cells were harvested and lysed in NP40 buffer containing a protease inhibitor cocktail (Roche, Basal, CH). After centrifugation, the supernatant was pretreated with protein A/G beads for 2 h at 4°C, and the pretreated lysate was combined with a 50% slurry of glutathione-conjugated Sepharose beads (Amersham Biosciences, Amersham, UK) and incubated for 4 h at 4°C. The precipitate was rinsed thoroughly with lysis buffer. The proteins bound to the glutathione beads were eluted by boiling in sodium dodecyl sulfate (SDS) loading buffer for 5 min. For immunoprecipitation assays, cells were harvested and lysed in NP40 buffer containing a protease inhibitor cocktail (Roche, Basal, CH). Whole-cell lysates were pretreated with protein A/G agarose beads for 1 h at 4°C and then immunoprecipitated with the treated antibodies. Typically, 1 mL of cell lysate was treated with 1–4 μg of antibody for 8–12 h at 4°C. After incubation with protein A/G agarose beads for 6 h, the immunoprecipitates were thoroughly washed with lysis buffer and eluted by boiling with SDS loading buffer for 5 min. Polypeptides were separated by SDS-polyacrylamide gel electrophoresis and transferred to a PVDF membrane for immunoblotting (IB) (Bio-Rad, Hercules, CA, USA). Specific antibodies were required for immunodetection, and chemiluminescence (ECL; Millipore, MA, USA) was used to visualize antibodies bound to the target. And a Vilber chemiluminescence analyzer was used to detect them (Fusion SL3; Vilber Lourmat, Coleegien, France).

1.6 효소면역측정법1.6 Enzyme-linked immunosorbent assay

BD OptEIA ELISA 시스템(BD Pharmingen)을 사용하여 세포배양 상등액과 마우스 혈청에서 TNF-α, IL-6, IL-1β및 IL-18 수준을 측정하였다. 모든 검사는 제조업체가 지시한 대로 수행하였다.TNF-α, IL-6, IL-1β, and IL-18 levels were measured in cell culture supernatants and mouse serum using the BD OptEIA ELISA system (BD Pharmingen). All assays were performed according to the manufacturer's instructions.

1.7 NLRP3, ASC의 결합 파트너와의 상호작용 운동 분석1.7 Kinetic analysis of NLRP3 and ASC binding partners

NLRP3- ASC의 상호작용은 플루오르막스-4 분광불화도계 (HORIBA Scientific)를 사용하여 모니터링 하였으며 앞에서 설명한 바와 같이 수행하였다. 제조사의 지침에 따라 NLRP3에 BODIPY FL Iodoacetamide (ThermoFisher Scientific)라는 레이블이 붙었다. NLRP3라벨은 350nm에서 들뜨고 512nm에서 차단 필터를 통해 검출하였다. NLRP3는 운동 분석을 위해 레이블이 지정되지 않은 NLRP3 또는 ASC로 지정되었다. 사용된 자극 파장과 방출 파장은 각각 498mm와 518nm였다. 획득한 데이터는 Grafit 프로그램을 사용하여 확인하였으며, 모든 형광 측정은 30 mM Tris, pH 7.4, 150 mM NaCl 및 1 mM dithiothryitol에서 25 ℃에서 수행하였다.NLRP3-ASC interaction was monitored using a Fluormax-4 spectrofluorimeter (HORIBA Scientific) as described previously. NLRP3 was labeled with BODIPY FL Iodoacetamide (ThermoFisher Scientific) according to the manufacturer's instructions. The NLRP3 label was excited at 350 nm and detected through a cutoff filter at 512 nm. NLRP3 was designated as unlabeled NLRP3 or ASC for kinetic analysis. The excitation and emission wavelengths used were 498 and 518 nm, respectively. The acquired data were viewed using the Grafit program, and all fluorescence measurements were performed at 25 °C in 30 mM Tris, pH 7.4, 150 mM NaCl, and 1 mM dithiothryitol.

2. 결과2. Results

2.1 ASC의 PYD 도메인과 Rv0747 401TLTGRPLIGNGANG414은 서로 상호 작용한다.2.1 The PYD domain of ASC and Rv0747 401 TLTGRPLIGNGANG 414 interact with each other.

NLRP3 인플라마좀 복합체 형성을 위해 필요한 어댑터 단백질인 ASC와 상호 작용하는 결핵균의 항원 단백질을 찾기 위해, 재조합 ASC(rASC-His)를 결핵균 H37Rv 용해물과 공동 면역침천을 하고 His-아가로스 구슬로 정제하였다. 정제된 rASC-His 복합체를 은 염색법과 액체 크로마토그래피 질량분석법으로 확인하였고 흥미롭게도, 결핵균의 Rv0747/PE_PGRS10 단백질이 ASC 단백질과 결합하였다(도 1a). ASC는 NLRP3와 결합하는 PYD 도메인과 이펙터 단백질인 caspase-1과 결합하는 CARD 도메인으로 구성되어 있다. ASC 단백질과 Rv0747단백질이 서로 상호 작용하는 도메인을 확인하기 위해, 293T세포에 GST로 표지된 Rv0747(GST-Rv0747)과 V5로 표지된 ASC WT, ASC의 PYD도메인, 그리고 ASC의 CARD 도메인을 형질감염하고 GST 풀다운 분석을 수행하였다. V5 항체로 면역 검출하였을 때, V5-ASC WT과 PYD 도메인은 검출되었지만, CARD 도메인은 검출되지 않은 것을 확인할 수 있다(도 1b). 또, GST-Rv0747 전체 도메인 (1-801), PE 도메인(1-93), 그리고 약 100개의 아미노산씩 나눈 도메인들과 V5-ASC를 사용하였고 Rv0747의 전체 도메인과 401-500번 펩타이드가 ASC 단백질과 상호 작용하는 것을 확인하였다 (도 1c). PredictProtein 도구를 사용하여 Rv0747의 401-500번 펩타이드 중에서 401번부터 414번의 14개 아미노산 서열이 ASC 단백질과의 상호 작용에서 중요할 것이라는 것을 확인하였다(도 1d).To find the antigenic protein of M. tuberculosis that interacts with ASC, an adaptor protein required for NLRP3 inflammasome complex formation, recombinant ASC (rASC-His) was co-immunoprecipitated with M. tuberculosis H37Rv lysate and purified with His-agarose beads. The purified rASC-His complex was identified by silver staining and liquid chromatography-mass spectrometry. Interestingly, the Rv0747/PE_PGRS10 protein of M. tuberculosis bound to the ASC protein (Fig. 1a). ASC is composed of a PYD domain that binds to NLRP3 and a CARD domain that binds the effector protein caspase-1. To identify the domain in which ASC protein and Rv0747 protein interact with each other, GST-tagged Rv0747 (GST-Rv0747) and V5-tagged ASC WT, PYD domain of ASC, and CARD domain of ASC were transfected into 293T cells, and GST pull-down assay was performed. When immunodetected with V5 antibody, it was confirmed that V5-ASC WT and PYD domain were detected, but CARD domain was not detected (Fig. 1b). In addition, GST-Rv0747 entire domain (1-801), PE domain (1-93), and domains divided into about 100 amino acids each, and V5-ASC were used, and it was confirmed that the entire domain of Rv0747 and peptide 401-500 interacted with ASC protein (Fig. 1c). Using the PredictProtein tool, we identified that a 14 amino acid sequence from amino acids 401 to 414 among peptides 401-500 of Rv0747 would be important for the interaction with ASC protein (Fig. 1d).

2.2 Rv0747 401TLTGRPLIGNGANG414는 NLRP3 인플라마좀 활성화를 증가시킨다.2.2 Rv0747 401 TLTGRPLIGNGANG 414 increases NLRP3 inflammasome activation.

ASC와 Rv0747간의 상호 작용의 역할을 확인하기 위해, 세포 관통 펩타이드 R9(RRRRRRRRR)와 결합한 ASC와 상호 작용하는 Rv0747유래 펩타이드(Rv0747 401TLTGRPLIGNGANG414)를 제작하였다. 293T 세포에 Flag-NLRP3와 V5-ASC를 형질감염하고 펩타이드를 농도 의존적으로 처리한 후 NLRP3와 ASC간의 결합을 관찰하였다. 그 결과 Rv0747 401TLTGRPLIGNGANG414의 농도에 비례하여 NLRP3와 ASC간의 결합은 증가되었다. 또한, 골수 유래 대식세포(BMDMs)에 LPS와 ATP로 자극하여 NLRP3 인플라마좀을 활성화시키고 Rv0747 401TLTGRPLIGNGANG414를 처리하였을 때, NLRP3와 ASC의 결합이 증가하는 것을 확인하였다(도 2a). 다음, LPS에의해 자극된 BMDMs에 여러 인플라마좀 활성제와 Rv0747 401TLTGRPLIGNGANG414를 처리한 후 IL-1β/18를 효소면역측정법과 면역블롯을 통해 확인하였다. Rv0747 401TLTGRPLIGNGANG414는 NLRP3 인플라마좀 활성화에 의한 IL-1β/18 분비만을 특이적으로 증가시켰고, 면역 블롯 결과에서도 NLRP3 인플라마좀 활성에서만 IL-1β/18과 인플라마좀의 이펙터 단백질인 caspase-1의 분비를 증가시켰다 (도 2b 및 도 2c). NLRP3 인플라마좀이 활성화하면 ASC 단량체가 여러 개로 이루어진 올리고머화를 하며 ASC 스펙이라고 불리는 마이크로미터 크기의 핵주위 구조물을 형성한다. LPS로 자극받은 BMDMs에서 Rv0747 401TLTGRPLIGNGANG414를 농도 의존적으로 처리하였을 때, 농도에 비례하여 ASC의 올리고머화가 증가하였고, 또한 Rv0747 401TLTGRPLIGNGANG414의 농도가 증가함에 따라 세포 내부의 ASC 다량체의 비율도 증가하는 것을 확인할 수 있었다 (도 2d). 종합하면, 이 결과들은 ASC와 Rv0747 401TLTGRPLIGNGANG414의 상호작용이 NLRP3와 ASC의 결합을 증가시켜 NLRP3 인플라마좀을 특이적으로 활성화한다는 것을 나타낸다.To confirm the role of the interaction between ASC and Rv0747, an Rv0747-derived peptide (Rv0747 401 TLTGRPLIGNGANG 414 ) that interacts with ASC conjugated with the cell-penetrating peptide R9 (RRRRRRRRR) was constructed. Flag-NLRP3 and V5-ASC were transfected into 293T cells, and the binding between NLRP3 and ASC was observed after treatment with the peptide in a concentration-dependent manner. As a result, the binding between NLRP3 and ASC was increased in a concentration-dependent manner of Rv0747 401 TLTGRPLIGNGANG 414 . In addition, when bone marrow-derived macrophages (BMDMs) were stimulated with LPS and ATP to activate the NLRP3 inflammasome and treated with Rv0747 401 TLTGRPLIGNGANG 414 , the binding of NLRP3 to ASC was confirmed to increase (Fig. 2a). Next, after treating BMDMs stimulated by LPS with various inflammasome activators and Rv0747 401 TLTGRPLIGNGANG 414 , IL-1β/18 was confirmed through enzyme-linked immunosorbent assay and immunoblot. Rv0747 401 TLTGRPLIGNGANG 414 specifically increased only IL-1β/18 secretion induced by NLRP3 inflammasome activation, and immunoblot results showed that only NLRP3 inflammasome activation increased the secretion of IL-1β/18 and caspase-1, an effector protein of inflammasome (Fig. 2b and Fig. 2c). When NLRP3 inflammasome is activated, ASC monomers oligomerize into multiple units to form micrometer-sized perinuclear structures called ASC specks. When Rv0747 401 TLTGRPLIGNGANG 414 was dose-dependently treated in LPS-stimulated BMDMs, ASC oligomerization increased in a concentration-dependent manner, and it was confirmed that the proportion of intracellular ASC multimers also increased as the concentration of Rv0747 401 TLTGRPLIGNGANG 414 increased (Fig. 2d). In summary, these results indicate that the interaction of ASC and Rv0747 401 TLTGRPLIGNGANG 414 increases the binding of NLRP3 to ASC, thereby specifically activating the NLRP3 inflammasome.

2.3 Rv0747 401TLTGRPLIGNGANG414의 서열에서 T와 N 아미노산이 ASC와의 결합에 영향을 미친다.2.3 In the sequence of Rv0747 401 TLTGRPLIGNGANG 414 , the T and N amino acids affect binding to ASC.

Rv0747 401TLTGRPLIGNGANG414에서 어떤 아미노산의 점 돌연변이가 ASC와의 상호작용에 영향을 미칠 수 있는지 확인하고자 하였다. Rv0747 401TLTGRPLIGNGANG414의 아미노산 서열 중 일정한 간격에서 대칭적으로 두 개씩 존재하는 아미노산을 각각 402번 L을 F로, 407번 L을 F로, 410번 N을 T로, 그리고 413번 N을 T로 점 돌연변이 시켰다. 그 결과 V5-ASC와 점 돌연변이를 시킨 GST-Rv0747가 서로 결합하지 못하는 것을 관찰하였고 (도 3), 이는 각각의 아미노산이 ASC와의 결합에 중요한 역할을 할 수도 있다는 것을 나타낸다.We sought to determine which amino acid point mutations in Rv0747 401 TLTGRPLIGNGANG 414 could affect the interaction with ASC. In the amino acid sequence of Rv0747 401 TLTGRPLIGNGANG 414 , two amino acids that exist symmetrically at a certain interval were point-mutated: L at position 402 to F, L at position 407 to F, N at position 410 to T, and N at position 413 to T. As a result, we observed that V5-ASC and the point-mutated GST-Rv0747 failed to bind to each other (Fig. 3), indicating that each amino acid may play an important role in the binding to ASC.

예를 들어, 청구항 구성 목적을 위해, 이하 기재되는 청구항은 어떤 식으로든 이의 문자 그대로의 언어보다 좁게 해석되어선 안 되고, 따라서 명세서로부터의 예시적 구현예가 청구항으로 읽혀서는 안 된다. 따라서, 본 발명은 예시로서 기재되었고, 청구항의 범위에 대한 제한이 아님이 이해되어야 한다. 따라서, 본 발명은 하기 청구항에 의해서만 제한된다. 본 출원에 인용된 모든 간행물, 발행된 특허, 특허 출원, 서적 및 저널 논문은 이들의 전체내용이 참조로서 본원에 각각 포함된다.For example, for purposes of claim construction, the claims set forth below should not be construed narrowly in any way beyond the literal language thereof, and thus exemplary embodiments from the specification should not be read as claims. Accordingly, it should be understood that the invention has been described by way of example, and not as a limitation on the scope of the claims. Accordingly, the invention is limited only by the claims below. All publications, issued patents, patent applications, books, and journal articles cited in this application are hereby incorporated by reference in their entirety.

SEQ ID NO: 1SEQ ID NO: 1

TLTGRPLIGN GANGTLTGRPLIGN GANG

SEQ ID NO: 2SEQ ID NO: 2

MSWVMVSPEL VVAAAADLAG IGSAISSANA AAAVNTTGLL TAGADEVSTA IAALFGAQGQ AYQAASAQAA AFYAQFVQAL SAGGGAYAAA EAAAVSPLLA PINAQFVAAT GRPLIGNGAN GAPGTGANGG PGGWLIGNGG AGGSGAPGAG AGGNGGAGGL FGSGGAGGAS TDVAGGAGGA GGAGGNAGML FGAAGVGGVG GFSNGGATGG AGGAGGAGGL FGAGRERGSG GSGNLTGGAG GAGGNAGTLA TGDGGAGGTG GASRSGGFGG AGGAGGDAGM FFGSGGSGGA GGISKSVGDS AAGGAGGAPG LIGNGGNGGN GGASTGGGDG GPGGAGGTGV LIGNGGNGGS GGTGATLGKA GIGGTGGVLL GLDGFTAPAS TSPLHTLQQD VINMVNDPFQ TLTGRPLIGN GANGTPGTGA DGGAGGWLFG NGGNGGQGTI GGVNGGAGGA GGAGGILFGT GGTGGSGGPG ATGLGGIGGA GGAALLFGSG GAGGSGGAGA VGGNGGAGGN AGALLGAAGA GGAGGAGAVG GNGGAGGNGG LFANGGAGGP GGFGSPAGAG GIGGAGGNGG LFGAGGTGGA GGGSTLAGGA GGAGGNGGLF GAGGTGGAGS HSTAAGVSGG AGGAGGDAGL LSLGASGGAG GSGGSSLTAA GVVGGIGGAG GLLFGSGGAG GSGGFSNSGN GGAGGAGGDA GLLVGSGGAG GAGASATGAA TGGDGGAGGK SGAFGLGGDG GAGGATGLSG AFHIGGKGGV GGSAVLIGNG GNGGNGGNSG NAGKSGGAPG PSGAGGAGGL LLGENGLNGL MMSWVMVSPEL VVAAAADLAG IGSAISSANA AAAVNTTGLL TAGADEVSTA IAALFGAQGQ AYQAASAQAA AFYAQFVQAL SAGGGAYAAA EAAAVSPLLA PINAQFVAAT GRPLIGNGAN GAPGTGANGG PGGWLIGNGG AGGSGAPGAG AGGNGGAGGL FGSGGAGGAS TDVAGGAGGA GGAGGNAGML FGAAGVGGVG GFSNGGATGG AGGAGGAGGL FGAGRERGSG GSGNLTGGAG GAGGNAGTLA TGDGGAGGTG GASRSGGFGG AGGAGGDAGM FFGSGGSGGA GGISKSVGDS AAGGAGGAPG LIGNGGNGGN GGASTGGGDG GPGGAGGTGV LIGNGGNGGS GGTGATLGKA GIGGTGGVLL GLDGFTAPAS TSPLHTLQQD VINMVNDPFQ TLTGRPLIGN GANGTPGTGA DGGAGGWLFG NGGNGGQGTI GGVNGGAGGA GGAGGILFGT GGTGGSGGPG ATGLGGIGGA GGAALLFGSG GAGGSGGAGA VGGNGGAGGN AGALLGAAGA GGAGGAGAVG GNGGAGGNGG LFANGGAGGP GGFGSPAGAG GIGGAGGNGG LFGAGGTGGA GGGSTLAGGA GGAGGNGGLF GAGGTGGAGS HSTAAGVSGG AGGAGGDAGL LSLGASGGAG GSGGSSLTAA GVVGGIGGAG GLLFGSGGAG GSGGFSNSGN GGAGGAGGDA GLLVGSGGAG GAGASATGAA TGGDGGAGGK SGAFGLGGDG GAGGATGLSG AFHIGGKGGV GGSAVLIGNG GNGGNGGNSG NAGKSGGAGPG PSGAGGAGGL LLGENGLNGL M

SEQ ID NO: 3SEQ ID NO: 3

acgctcaccg ggcgtccgct gatcggcaac ggcgccaacg gcacgctcaccg ggcgtccgct gatcggcaac ggcgccaacg gc

SEQ ID NO: 4SEQ ID NO: 4

atgtcatggg tgatggtttc gccggagctg gtggtggcgg cggcagcgga tttggcgggg atcgggtcgg cgattagctc ggctaatgcg gcggcggccg tcaacacgac gggattgttg accgcgggtg ccgatgaggt gtcgacagcg attgcggcgt tgttcggtgc ccaaggccag gcctaccagg cggcgagcgc acaggcggcg gcgttttacg cccagttcgt gcaggccctg agcgccggcg gaggcgcgta tgcggccgcc gaggccgccg ccgtgtcgcc gctgctggcc ccgatcaacg cgcaattcgt ggcggccacc gggcgcccgc tgatcggcaa cggcgccaac ggcgcccccg ggaccggagc caacggcggg cccggcgggt ggttgatcgg caacggcggc gccggcgggt ctggcgcccc cggcgctggg gccggcggta acggcggggc cggcgggctg ttcggcagcg gcggggccgg cggggcctcc accgacgtcg ccggcggggc cggtggggcc ggcggggccg gcggaaacgc cggcatgctg ttcggcgccg ccggggtcgg cggcgtcggc ggattctcga acggcggtgc caccggcggg gcaggcgggg ccggcggggc gggcgggctg tttggcgccg gaagggaacg cggcagcggc gggtcgggca acctcactgg cggggccggc ggggccggcg gcaacgccgg gacactcgcc actggtgatg gcggggccgg cgggaccggc ggcgctagtc gcagcggcgg attcggcggg gccggcggag ccggcggcga cgccggcatg ttcttcggct ccggcggctc cggcggcgcc ggcggcatta gtaaaagcgt cggggacagc gccgccggcg gggccggcgg ggcccccggg ctgatcggca acggcggcaa cggcggcaac ggcggcgcga gcaccggcgg cggggacggt gggcccggcg gggccggcgg caccggcgtg ttgatcggca acggcggcaa cggcggcagc ggcgggaccg gcgcgaccct gggcaaggcc ggcatcggcg gtaccggggg ggtgctgttg ggcctggacg gctttacggc ccccgccagc acctcgcccc tgcacaccct gcagcaggac gtgatcaata tggtgaacga ccccttccag acgctcaccg ggcgtccgct gatcggcaac ggcgccaacg gcactccggg gaccggggct gacggcggag ccggcggctg gttgttcggc aacggcggaa acggcgggca gggaacgatc ggcggcgtca acggcggggc cggcggggcc ggcggggccg gcgggatctt gttcggcacc ggcggcaccg ggggcagcgg cgggcccggc gccaccggcc tcggcgggat tggcggggcc ggcggagccg ccttgctctt cggctccggc ggggccggcg gaagcggtgg tgccggcgcg gtcggtggca atggcggggc cggcggcaac gccggtgcgc tcttgggcgc cgccggggcc ggcggggccg gtggtgccgg cgcggtcggt ggcaatggcg gggccggcgg taacggcggg ctgttcgcca acgggggagc cggcgggccc ggtgggtttg gcagccccgc tggggctggc gggatcggcg gggcaggtgg gaacggcggg ctgttcggcg ccggcgggac cggcggggcc ggcgggggaa gcaccctcgc cggcggcgcc ggcggggcgg gcggcaacgg cgggctgttc ggcgccggcg gcaccggcgg cgccggcagc catagcaccg ccgccggagt ttccggaggg gccggcgggg ccggcggcga cgccggcttg ctctccctcg gcgcctccgg cggggccggc ggcagcggcg gttccagcct gaccgccgcc ggcgtggtcg gcggcatcgg cggcgccgga ggcttgctct tcggctccgg cggcgccggc gggagcggcg ggttcagcaa ctctggcaac ggcggcgccg gcggggccgg cggcgacgcg ggtttgctcg tcggctccgg cggggccggc ggggccggcg cctccgccac cggcgccgcc accggcgggg acggcggggc cggcggcaag tccggagcgt tcggtctcgg aggtgacggc ggcgccggcg gcgccaccgg tttgtccggt gctttccaca tcggcggcaa gggcggcgtc ggcggcagcg ccgtgctgat cggcaacggc ggcaacggcg gcaacggcgg taacagcggt aacgccggga aatccggggg tgcacccggc cccagcggcg ccggcggcgc cggcgggctg ctgctcggtg agaacgggct gaacggcttg atgtagatgtcatggg tgatggtttc gccggagctg gtggtggcgg cggcagcgga tttggcgggg atcgggtcgg cgattagctc ggctaatgcg gcggcggccg tcaacacgac gggattgttg accgcgggtg ccgatgaggt gtcgacagcg attgcggcgt tgttcggtgc ccaaggccag gcctaccagg cggcgagcgc acaggcggcg gcgttttacg cccagttcgt gcaggccctg agcgccggcg gaggcgcgta tgcggccgcc gaggccgccg ccgtgtcgcc gctgctggcc ccgatcaacg cgcaattcgt ggcggccacc gggcgcccgc tgatcggcaa cggcgccaac ggcgcccccg ggaccggagc caacggcggg cccggcgggt ggttgatcgg caacggcggc gccggcgggt ctggcgcccc cggcgctggg gccggcggta acggcggggc cggcgggctg ttcggcagcg gcggggccgg cggggcctcc accgacgtcg ccggcggggc cggtggggcc ggcggggccg gcggaaacgc cggcatgctg ttcggcgccg ccggggtcgg cggcgtcggc ggattctcga acggcggtgc caccggcggg gcaggcgggg ccggcggggc gggcgggctg tttggcgccg gaaggggaacg cggcagcggc gggtcgggca acctcactgg cggggccggc ggggccggcg gcaacgccgg gacactcgcc actggtgatg gcggggccgg cgggaccggc ggcgctagtc gcagcggcgg attcggcggg gccggcggag ccggcggcga cgccggcatg ttcttcggct ccggcggctc cggcggcgcc ggcggcatta gtaaaagcgt cggggacagc gccgccggcg gggccggcgg ggcccccggg ctgatcggca acggcggcaa cggcggcaac ggcggcgcga gcaccggcgg cggggacggt gggcccggcg gggccggcgg caccggcgtg ttgatcggca acggcggcaa cggcggcagc ggcgggaccg gcgcgaccct gggcaaggcc ggcatcggcg gtaccggggg ggtgctgttg ggcctggacg gctttacggc ccccgccagc acctcgcccc tgcacaccct gcagcaggac gtgatcaata tggtgaacga ccccttccag acgctcaccg ggcgtccgct gatcggcaac ggcgccaacg gcactccggg gaccggggct gacggcggag ccggcggctg gttgttcggc aacggcggaa acggcgggca gggaacgatc ggcggcgtca acggcggggc cggcggggcc ggcggggccg gcgggatctt gttcggcacc ggcggcaccg ggggcagcgg cgggcccggc gccaccggcc tcggcgggat tggcggggcc ggcggagccg ccttgctctt cggctccggc ggggccggcg gaagcggtgg tgccggcgcg gtcggtggca atggcggggc cggcggcaac gccggtgcgc tcttgggcgc cgccggggcc ggcggggccg gtggtgccgg cgcggtcggt ggcaatggcg gggccggcgg taacggcggg ctgttcgcca acgggggagc cggcgggccc ggtgggtttg gcagccccgc tggggctggc gggatcggcg gggcaggtgg gaacggcggg ctgttcggcg ccggcgggac cggcggggcc ggcgggggaa gcaccctcgc cggcggcgcc ggcggggcgg gcggcaacgg cgggctgttc ggcgccggcg gcaccggcgg cgccggcagc catagcaccg ccgccggagt ttccggaggg gccggcgggg ccggcggcga cgccggcttg ctctccctcg gcgcctccgg cggggccggc ggcagcggcg gttccagcct gaccgccgcc ggcgtggtcg gcggcatcgg cggcgccgga ggcttgctct tcggctccgg cggcgccggc gggagcggcg ggttcagcaa ctctggcaac ggcggcgccg gcggggccgg cggcgacgcg ggtttgctcg tcggctccgg cggggccggc ggggccggcg cctccgccac cggcgccgcc accggcgggg acggcggggc cggcggcaag tccggagcgt tcggtctcgg aggtgacggc ggcgccggcg gcgccaccgg tttgtccggt gctttccaca tcggcggcaa gggcggcgtc ggcggcagcg ccgtgctgat cggcaacggc ggcaacggcg gcaacggcgg taacagcggt aacgccggga aatccggggg tgcacccggc cccagcggcg ccggcggcgc cggcgggctg ctgctcggtg agaacgggct gaacggcttg atgtag

Claims (15)

ASC (Apoptosis-associated Speck-like protein containing a CARD)에 특이적으로 결합하는, 서열번호 1로 표시되는 서열을 포함하는 폴리펩타이드 또는 이의 변이체. A polypeptide comprising a sequence represented by SEQ ID NO: 1 or a variant thereof, which specifically binds to ASC (Apoptosis-associated Speck-like protein containing a CARD). 제1항에 있어서, 상기 폴리펩타이드는 서열번호 1로 표시되는 서열로 구성되는, 폴리펩타이드 또는 이의 변이체. In the first paragraph, the polypeptide is a polypeptide or a variant thereof, which is composed of a sequence represented by SEQ ID NO: 1. 제1항에 있어서, 상기 폴리펩타이드는 서열번호 2로 표시되는 서열을 포함하는, 폴리펩타이드 또는 이의 변이체. In claim 1, the polypeptide is a polypeptide or a variant thereof comprising a sequence represented by SEQ ID NO: 2. 제1항에 있어서, 상기 변이체는 서열번호 1의 2번째, 7번째, 10번째, 및 13번째 아미노산을 제외한 위치에서 보존적 치환을 포함하는, 폴리펩타이드 또는 이의 변이체.A polypeptide or a variant thereof, in claim 1, wherein the variant comprises a conservative substitution at positions other than the 2nd, 7th, 10th, and 13th amino acids of SEQ ID NO: 1. 제4항에 있어서, 상기 보존적 치환은 T1S, T3S, G4A, R5K, I8V, G9A, G11A, A12G, 및 G14A로 구성된 군으로부터 선택된 적어도 하나, 또는 T1S, T3S, G4A, R5K, I8L, G9A, G11A, A12G, 및 G14A로 구성된 군으로부터 선택된 적어도 하나인, 폴리펩타이드 또는 이의 변이체.A polypeptide or a variant thereof, in claim 4, wherein the conservative substitution is at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8V, G9A, G11A, A12G, and G14A, or at least one selected from the group consisting of T1S, T3S, G4A, R5K, I8L, G9A, G11A, A12G, and G14A. 제1항 내지 제5항 중 어느 한 항의 폴리펩타이드 또는 이의 변이체를 포함하는, ASC 특이적 결합 분자. An ASC-specific binding molecule comprising a polypeptide according to any one of claims 1 to 5 or a variant thereof. 제1항 내지 제5항 중 어느 한 항의 폴리펩타이드 또는 변이체를 포함하는, 암 치료용 약학 조성물.A pharmaceutical composition for treating cancer, comprising a polypeptide or variant of any one of claims 1 to 5. 제7항에 있어서, 상기 암은 염증성 대장암, 결장직장암, 흑색종, 또는 간세포암인, 암 치료용 약학 조성물. A pharmaceutical composition for treating cancer, wherein the cancer in claim 7 is inflammatory colon cancer, colorectal cancer, melanoma, or hepatocellular cancer. 제1항 내지 제5항 중 어느 한 항의 폴리펩타이드 또는 변이체를 발현하는 핵산 분자. A nucleic acid molecule expressing a polypeptide or variant of any one of claims 1 to 5. 제9항의 핵산 분자를 포함하는, 암 치료용 약학 조성물.A pharmaceutical composition for treating cancer, comprising a nucleic acid molecule of claim 9. 제10항에 있어서, 상기 암은 염증성 대장암, 결장직장암, 흑색종, 또는 간세포암인, 암 치료용 약학 조성물. A pharmaceutical composition for treating cancer, wherein the cancer in claim 10 is inflammatory colon cancer, colorectal cancer, melanoma, or hepatocellular cancer. 제9항의 핵산 분자를 포함하는, 재조합 벡터. A recombinant vector comprising the nucleic acid molecule of claim 9. 제9항의 핵산 분자를 포함하는, 숙주 세포. A host cell comprising a nucleic acid molecule of claim 9. 제1항 내지 제5항 중 어느 한 항의 폴리펩타이드 또는 변이체를 치료가 필요한 환자에게 투여하는 단계를 포함하는, 암 치료 방법.A method for treating cancer, comprising administering to a patient in need of treatment a polypeptide or variant of any one of claims 1 to 5. 제9항의 핵산 분자를 치료가 필요한 환자에게 투여하는 단계를 포함하는, 암 치료 방법.A method for treating cancer, comprising the step of administering a nucleic acid molecule of claim 9 to a patient in need of treatment.
PCT/KR2024/003333 2023-06-01 2024-03-18 Pharmaceutical composition for treating cancer comprising polypeptide derived from mycobacterium tuberculosis Pending WO2024248286A1 (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2023-0070608 2023-06-01
KR20230070608 2023-06-01
KR1020230103350A KR20240173318A (en) 2023-06-01 2023-08-08 Pharmaceutical composition for treating cancer comprising polypeptide derived from Mycobacterium tuberculosis
KR10-2023-0103350 2023-08-08

Publications (1)

Publication Number Publication Date
WO2024248286A1 true WO2024248286A1 (en) 2024-12-05

Family

ID=93658235

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2024/003333 Pending WO2024248286A1 (en) 2023-06-01 2024-03-18 Pharmaceutical composition for treating cancer comprising polypeptide derived from mycobacterium tuberculosis

Country Status (1)

Country Link
WO (1) WO2024248286A1 (en)

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101178776B1 (en) * 2004-02-06 2012-09-07 카운슬 오브 사이언티픽 앤드 인더스트리얼 리서치 A computer apparatus for identifying adhesin and adhesin-like proteins of therapeutic potential
KR20220061482A (en) * 2020-11-06 2022-05-13 한국과학기술원 Novel polypeptide binding with oligomerization domain of Inflammasome Adaptor protein Apoptosis-Associated Speck-like protein containing a CARD(ASC) and Uses thereof
US20220339169A1 (en) * 2018-07-03 2022-10-27 Novartis Ag Methods of treating or selecting a treatment for a subject resistant to tnf inhibitor using a nlrp3 antagonist

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR101178776B1 (en) * 2004-02-06 2012-09-07 카운슬 오브 사이언티픽 앤드 인더스트리얼 리서치 A computer apparatus for identifying adhesin and adhesin-like proteins of therapeutic potential
US20220339169A1 (en) * 2018-07-03 2022-10-27 Novartis Ag Methods of treating or selecting a treatment for a subject resistant to tnf inhibitor using a nlrp3 antagonist
KR20220061482A (en) * 2020-11-06 2022-05-13 한국과학기술원 Novel polypeptide binding with oligomerization domain of Inflammasome Adaptor protein Apoptosis-Associated Speck-like protein containing a CARD(ASC) and Uses thereof

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
HAMARSHEH SHAIMA'A, ZEISER ROBERT: "NLRP3 Inflammasome Activation in Cancer: A Double-Edged Sword", FRONTIERS IN IMMUNOLOGY, vol. 11, 8 July 2020 (2020-07-08), Lausanne, CH , pages 1 - 11, XP093245438, ISSN: 1664-3224, DOI: 10.3389/fimmu.2020.01444 *
SU JIA-QI, TIAN XI, XU WEN-HAO, ANWAIER AIHETAIMUJIANG, YE SHI-QI, ZHU SHU-XUAN, WANG YUE, GU JUN, SHI GUO-HAI, QU YUAN-YUAN, ZHAN: "The Inflammasomes Adaptor Protein PYCARD Is a Potential Pyroptosis Biomarker Related to Immune Response and Prognosis in Clear Cell Renal Cell Carcinoma", CANCERS, vol. 14, no. 20, 1 January 2022 (2022-01-01), CH , pages 1 - 21, XP093245442, ISSN: 2072-6694, DOI: 10.3390/cancers14204992 *

Similar Documents

Publication Publication Date Title
US12410224B2 (en) IL-15-based fusions to IL-7 and IL-21
AU2019311233B2 (en) IL2 agonists
US7030212B1 (en) Tumor antigen based on products of the tumor suppressor gene WT1
JP7505984B2 (en) Treatment with RNA encoding cytokines
CA2456196A1 (en) Compositions and methods for modulation of immune responses
CN113795272B (en) Therapies involving CAR-engineered T cells and cytokines
JP2025131724A (en) Treatments including interleukin 2 (IL2) and interferon (IFN)
US20220356223A1 (en) IL2 Agonists
WO2024248286A1 (en) Pharmaceutical composition for treating cancer comprising polypeptide derived from mycobacterium tuberculosis
KR20240173318A (en) Pharmaceutical composition for treating cancer comprising polypeptide derived from Mycobacterium tuberculosis
WO2024248287A1 (en) Pharmaceutical composition for treating inflammatory diseases comprising polypeptide derived from mycobacterium tuberculosis
US20250346900A1 (en) Pharmaceutical composition for treating cancer comprising foxm1 mutant or foxm1 shrna
KR20240081504A (en) Pharmaceutical composition for treating cancer comprising FoxM1 mutants
Hildegund Vaccines to Breast Cancer Based on p53 Mutants.
AU2002324581A1 (en) Compositions and methods for modulation of immune responses

Legal Events

Date Code Title Description
121 Ep: the epo has been informed by wipo that ep was designated in this application

Ref document number: 24815665

Country of ref document: EP

Kind code of ref document: A1