WO2017176007A1 - Antibody-drug conjugate comprising modified antibody - Google Patents

Antibody-drug conjugate comprising modified antibody Download PDF

Info

Publication number
WO2017176007A1
WO2017176007A1 PCT/KR2017/003508 KR2017003508W WO2017176007A1 WO 2017176007 A1 WO2017176007 A1 WO 2017176007A1 KR 2017003508 W KR2017003508 W KR 2017003508W WO 2017176007 A1 WO2017176007 A1 WO 2017176007A1
Authority
WO
WIPO (PCT)
Prior art keywords
antibody
seq
genbank accession
drug
receptor
Prior art date
Application number
PCT/KR2017/003508
Other languages
French (fr)
Korean (ko)
Inventor
박순재
정혜신
이선배
변민수
김류련
Original Assignee
(주)알테오젠
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by (주)알테오젠 filed Critical (주)알테오젠
Priority to EP17779307.2A priority Critical patent/EP3441088B1/en
Priority to EP22191369.2A priority patent/EP4144377A1/en
Priority to CN202210306570.8A priority patent/CN114887074A/en
Priority to US16/088,804 priority patent/US11235065B2/en
Priority to CA3018691A priority patent/CA3018691C/en
Priority to ES17779307T priority patent/ES2929010T3/en
Priority to JP2018552180A priority patent/JP6708751B2/en
Priority to CN201780027885.6A priority patent/CN109069657B/en
Priority claimed from KR1020170040472A external-priority patent/KR102001821B1/en
Publication of WO2017176007A1 publication Critical patent/WO2017176007A1/en
Priority to US17/551,103 priority patent/US20220105197A1/en

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/337Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having four-membered rings, e.g. taxol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/5365Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines ortho- or peri-condensed with heterocyclic ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans

Definitions

  • the present invention relates to an antibody-drug conjugate in which a modified antibody comprising a motif of a specific structure at the terminal of the antibody is bound to a drug through a linker, and specifically to a drug linked to the heavy or light chain C-terminus.
  • a modified Antibody-Drug Conjugate (mADC) conjugate comprising a modified antibody having a significantly increased conjugation yield and a composition comprising the same.
  • Drugs used for chemotherapy often show toxicity, especially bone marrow, mucosa, and neurotoxicity. Therefore, there is a need for the development of an anticancer agent that is safer while showing strong anticancer activity and specificity to cancer cells. Anti-cancer drugs that act only on cancer cells and reduce side effects are being developed in various ways.
  • therapies using antibodies that are specifically expressed in specific diseases that is, antibodies that specifically bind to antigens
  • antibodies that specifically bind to antigens are currently being actively studied among biopharmaceuticals.
  • methods for diagnosing and treating tumors using antibodies, such as anti-cancer antibodies, which identify tumor-associated antigens specifically expressed on the surface of cancer cells, bind to them, and inhibit cell growth or induce death are now widely used. Is also a very bright technology field.
  • anticancer antibodies have very high target specificities, but the killing effect of cancer cells is often lower than that of conventional cytotoxic drugs (anticancer drugs), so they may be used in combination with cytotoxic drugs and other cell proliferation inhibitors. It is often used as a combination therapy.
  • Anticancer drugs show significantly higher cytotoxicity than anticancer antibodies, but have very high side effects compared to antibody therapeutics because of their low target specificity to cancer cells. Therefore, the combination therapy of anticancer antibody and anticancer drug shows a higher therapeutic effect than when each drug is administered separately, but has the fundamental limitation that side effects of anticancer drug are always accompanied.
  • anticancer drugs due to its very high cytotoxicity, among the anticancer drugs, drugs that can be used alone as anticancer drugs are limited to relatively low toxicity Taxol or cisplatin-based drugs. Most anticancer drugs with high cytotoxicity are virtually impossible to prescribe as single drugs because of their very high cytotoxicity.
  • an anticancer drug that cannot be used as a prescription alone to an antibody having a very high target specificity for cancer cells, the anticancer drug can be delivered only to the target cancer cells without the side effects of normal cells. Therefore, antibody-drug conjugates have been in the spotlight as a method for improving the therapeutic efficacy of anti-cancer drugs that cannot be used conventionally.
  • Antibody-drug conjugates currently on the market include Adcetris®, a treatment for Hodgkin's lymphoma, and Kadcyla®, a treatment for metastatic breast cancer. These antibody-drug conjugates are conjugated to tubulin inhibitors that inhibit the growth and division of cancer by conjugating to the tubule, an intracellular microtubule involved in cell division, thereby inhibiting cell division. Has These antibody-drug conjugates bind the drug to the lysine originally possessed by the antibody (cadcyla), or to the cysteine group that reduced the disulfide group linking the heavy chain-heavy chain or heavy-light chain that maintains the structural stability of the antibody. (Adsetless).
  • the anticancer drug conjugation method used in the first-generation antibody-drug conjugate does not control the number of conjugated drugs per antibody, and even in antibody-drug conjugates having the same number of drug conjugates, the positional conjugations of the drugs are different from each other. isomer) is generated.
  • the number of conjugates of drugs per antibody affects not only the cytotoxicity of antibody-drug conjugates but also the stability of antibody-drug conjugates and the possibility of aggregate formation.In general, the more conjugated drugs, the less stable the antibody itself and the formation of aggregates. The chances are high.
  • the number of conjugated drugs per antibody has an average value.
  • the number of drugs conjugated to the antibody may range from 1 to 8, and on average 3.5 drug conjugates.
  • the properties of the antibody-drug conjugate may vary depending on the location of the drug conjugate.
  • the drug is conjugated near the Fab or near the hinge of the Fc, there is a possibility that the stability of the antibody or antigen-antibody reactivity may be impaired due to a difference in the binding force between the antigen and the Fc ⁇ or FcRn receptor.
  • the number of conjugated drugs per antibody increases, the number of positional isomers with different conjugated positions of the drug increases proportionally, and this result may have a great influence on maintaining the consistent characteristics of the antibody-drug conjugate according to the production batch. .
  • Conjugation using non-natural amino acid is a natural amino acid that synthesizes tRNA that can introduce artificial amino acids into proteins through mutants of tRNA synthase. It is based on a technique that can introduce side-chains into proteins (Wang et al., Proc. Natl. Acad. Sci. USA, 2003, 100, 56-61). This allows the drug to be conjugated at a desired position in such a way as to perform site-specific conjugation to the residues of the artificial amino acids introduced (skeman et. Al, Bioconjugate Chem. 2014, 25, 351-361).
  • these methods require high-level tasks that require high levels of genetic engineering to regulate the translation pathway.
  • Antibodies act as carriers for the specific delivery of anticancer drugs to cancer cells, and the drug is a basic concept of an antibody-drug conjugate that needs to be stably conjugated to an antibody until it is delivered to cancer cells.
  • the necessity of producing antibody-drug conjugates using the same complex and time-consuming and expensive conjugation method is questionable.
  • the location-specific conjugation in the production of antibody-drug conjugates, and the economic conjugation method, which is excellent in economical efficiency, overcome the limitations of the existing method of conjugation of the first generation antibody-drug conjugates as well as the newly proposed location-specific conjugation method. It can be a great alternative.
  • Antibody-drug conjugates show superior in vitro and in vivo efficacy over existing antibody pharmaceuticals.
  • some clinical results for using antibody-drug conjugates as primary therapeutics failed to show significant clinical utility differences compared to the combination therapy of monoclonal antibody and chemosynthetic drugs (http: // www.roche.com/media/store/releases/med-cor-2014-12-19.htm).
  • These results can be a significant constraint on the economic utility of antibody-drug conjugates, given that antibody-drug conjugates require significantly higher therapeutic costs than synthetic drugs as well as monoclonal antibody therapeutics.
  • the above-mentioned location-specific conjugation methods have serious problems in providing economic utility that can be used as a primary therapeutic agent.
  • the present invention provides a antibody (hereinafter modified antibody) of the type having a specific binding motif to the parent antibody having a drug binding site.
  • modified antibodies modify modified antibodies comprising existing metal ion binding peptide motifs to provide modified antibodies with higher drug conjugate yields while maintaining the properties of site specific conjugation.
  • the present invention also provides an antibody-drug conjugate using the modified antibody and a method for producing an anticancer drug.
  • the present invention provides an antibody-drug conjugate wherein a modified antibody comprising a motif represented by the following structural formula (1) at the terminal of the antibody is bound to the drug via a linker:
  • M motif1 or M motif2 each independently include any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4), wherein X in SEQ ID NO: 3 or 4 includes amino acid residues other than cysteine,
  • X a And X b is a peptide consisting of 0 to 20 amino acid residues each independently selected from the group consisting of A (alanine), S (serine), G (glycine),
  • n1 and n2 are integers of 1-10, respectively.
  • the present invention also provides a composition for preventing or treating cancer comprising the antibody-drug conjugate.
  • FIG. 1 Samples conjugated to MC-vc-PAB-MMAE with FM2, FM2b (lot no: 4390f), FM2b (lot no: 5698f), FM2a, FM2L, and FM1 antibody variants of the antibody targeting the Folate receptor SDS-PAGE.
  • the drug is conjugated to a motif containing a cysteine residue introduced into the heavy chain of the antibody, and appears on the SDS-PAGE as two bands in the heavy chain near 50 kDa.
  • FIG. 1 SDS-PAGE of a sample conjugated with the antibody variants of FM2, FM2b, FM2a, FM2L, FM1 targeting the Folate receptor to br-vc-PAB-MMAE, a drug-linker conjugate linked to MMAE via bromoacetamide .
  • the drug is conjugated to a motif containing a cysteine residue introduced into the heavy chain of the antibody, and appears on the SDS-PAGE as two bands in the heavy chain near 50 kDa.
  • FIG. 3 Cell growth inhibition test using KB-cells overexpressed Folate receptor.
  • Parent antibody Fwt and modified antibody-drug conjugate FM2-D2 modified antibody-drug conjugate with DAR 2 in modified antibody-drug conjugate of modified antibody FM2 and MMAE
  • FM2b-D2 or FM2b-S-D2
  • FM2-D2 and FM2b-D2 show almost the same intracellular activity.
  • FIG. 4 Cell growth inhibition test using KB-cells overexpressed Folate receptor. Cell growth inhibition of FM2b-S-D2, FM2b-F-D2, and FM2b-Y-D2. FM2b-S-D2, FM2b-F-D2, and FM2b-Y-D2 show almost the same intracellular activity.
  • the antibody-drug conjugate must be stably bound to the antibody until the anti-cancer drug is delivered to the target cancer cell.
  • Anticancer drugs delivered to target cancer cells must be released from the antibody to induce the death of cancer cells.
  • the anti-cancer drug must be stably deficient in the antibody by using a linker and have a structure of a drug-linker having sufficient cytotoxicity to induce cancer cell death when released from cancer cells.
  • the drug should show the anti-potency of the antibody-drug conjugate and its uniformity in the manufacturing process by site-specific conjugation to the antibody.
  • the modified antibody comprising a peptide motif containing a metal ion binding motif is improved, that is, the ratio of conjugation of the drug conjugated to the antibody according to the arrangement and primary structure of the metal ion binding motif introduced into the terminal of the parent antibody. We tried to prove that this could increase significantly.
  • the present invention relates to an antibody-drug conjugate wherein a modified antibody comprising a motif represented by the following structural formula (1) at the terminal of the antibody is bound to a drug via a linker.
  • M motif1 or M motif2 each independently include any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4), wherein X in SEQ ID NO: 3 or 4 includes amino acid residues other than cysteine,
  • X a and X b are each independently a peptide consisting of 0 to 20 amino acid residues selected from the group consisting of A (alanine), S (serine) and G (glycine),
  • n1 and n2 are integers of 1-10, respectively.
  • the motif represented by Structural Formula (1) is a peptide including a CGH motif which is a metal ion binding motif, and the CGH motif has the structure of Formula 1 below.
  • M means a metal ion
  • R is an amino acid residue except cysteine, particularly alanine is preferable.
  • M motif1 or M motif2 includes ACGHA (SEQ ID NO: 1) in which alanine is located at the C-terminal and N-terminal, respectively, or AXGHA (SEQ ID NO: 3) in which a cysteine is substituted with an amino acid residue other than cysteine. do. N-terminus and C-terminus in ACGHA (SEQ ID NO: 1) or AXGHA (SEQ ID NO: 3) for M motif1 or M motif2 in the motif according to the present invention, even if the N-terminus and C-terminus are still changed. Also included are AHGCA (SEQ ID NO: 2) or AHGXA (SEQ ID NO: 4), which have been terminated.
  • M motif1 or M motif2 may each include the same sequence and may include different sequences from each other.
  • M motif 1 or M motif 2 corresponds to AXGHA or AHGXA, wherein X is serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid It may be an amino acid residue selected from the group consisting of (D), lysine (K) and phenylalanine (F).
  • the M motif2 includes AXGHA (SEQ ID NO: 3), and X may show higher drug conjugation ability than the case of including ACGHA when amino acid residues other than cysteine are included.
  • the M motif2 is AXGHA, where X is, for the amino acid residue Examples of the non-cysteine, serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and It may be an amino acid residue selected from the group consisting of phenylalanine (F).
  • X a is an amino acid residue present at the 5 'end of the motif M motif1 , which may be a peptide positioned to connect with the terminal of the antibody, and may be composed of A (alanine), S (serine), and G (glycine).
  • the amino acid residue selected from the group is a peptide consisting of 0-20.
  • the motif does not include X a and may be in a form in which M motif1 of the motif directly bonds to the antibody.
  • X a May be one or more, two or more, three or more, four or more, five or more amino acid residues of X a , for example, 2-20, 2-18, 2-16, 2-14 , 2-12, 2-10, 2-8, 2-6, 2, 3, 4, 5, 6, 7, 8, 9, 10.
  • X b is a linker for linking M motif1 and M motif2 and is a peptide consisting of 0 to 20 amino acid residues selected from the group consisting of A (alanine), S (serine) and G (glycine). If the amino acid residue of X b is 0, the motif does not include X b and may be in a form in which M motif 1 and M motif 2 are directly connected.
  • X b of the amino acid residues in one or more, two or more, three or more, four or more, may be more than five, e.g., 2-20, 2-18, 2-16, 2-14 , 2-12, 2-10, 2-8, 2-6, 2, 3, 4, 5, 6, 7, 8, 9, 10.
  • n1 and n2 represent the number of repetitions of M motif1 and M motif2 , respectively, and are an integer of 1 to 10.
  • N1 and n2 may each be 1, wherein M motif1 And M motif2 to ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3) and AHGXA (SEQ ID NO: 4) any of the sequences consisting of comprises a linker for X b without a linker or X b Can be connected.
  • [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker X b ACGHAACGHA (SEQ ID NO: 5), ACGHAAHGCA (SEQ ID NO: 6), ACGHAAXGHA (SEQ ID NO: 7), ACGHAAHGXA ( SEQ ID NO: 8), AHGCAAHGCA (SEQ ID NO: 9), AHGCAACGHA (SEQ ID NO: 10), AHGCAAXGHA (SEQ ID NO: 11), AHGCAAHGXA (SEQ ID NO: 12), AXGHAAXGHA (SEQ ID NO: 13), AXGHAACGHA (SEQ ID NO: 14), AXGHAAHGCA Number 15), AXGHAAHGXA (SEQ ID NO: 16), AHGXAAHGXA (SEQ ID NO: 17), AHGXAACGHA (SEQ ID NO: 18), AHGXAAHGCA (SEQ ID NO: 19), or AHGXAAXGHA
  • X b When X b is present, a peptide consisting of 1 to 20 amino acid residues at the 5 'end of the amino acid sequences may be further included.
  • X may be selected from the group consisting of serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and phenylalanine (F).
  • the n2 structure contains a serine at position X in SEQ ID NOs 7, 8,11 to 20, including X if there is no linker of X b , for example if X is serine ACGHAASGHA (SEQ ID NO: 21), ACGHAAHGSA (SEQ ID NO: 22), AHGCAASGHA (SEQ ID NO: 23), AHGCAAHGSA (SEQ ID NO: 24), ASGHAASGHA (SEQ ID NO: 25), ASGHAACGHA (SEQ ID NO: 26), ASGHAAHGCA (SEQ ID NO: 27), ASGHAAHGSA (SEQ ID NO: 28), AHGSAAHGSA (SEQ ID NO: 29), AHGSAACGHA (SEQ ID NO: 30), AHGSAAHGCA (SEQ ID NO: 31), or AHGSAASGHA (SEQ ID NO: 32).
  • n2 structure is ACGHAAAGHA (SEQ ID NO: 33), ACGHAAHGAA (SEQ ID NO: 34), AHGCAAAGHA (if X is alanine (A) when there is no linker of X b ) SEQ ID NO: 35), AHGCAAHGAA (SEQ ID NO: 36), AAGHAAAGHA (SEQ ID NO: 37), AAGHAACGHA (SEQ ID NO: 38), AAGHAAHGCA (SEQ ID NO: 39), AAGHAAHGAA (SEQ ID NO: 40), AHGAAAHGAA (SEQ ID NO: 41), AHGAAACGHA (SEQ ID NO: 41) Number 42), AHGAAAHGCA (SEQ ID NO: 43), or AHGAAAAGHA (SEQ ID NO: 44).
  • n2 structure is ACGHAATGHA (SEQ ID NO: 45), ACGHAAHGTA (SEQ ID NO: 46), AHGCAATGHA (if X is a threonine (T) when there is no linker of X b ) SEQ ID NO: 47), AHGCAAHGTA (SEQ ID NO: 48), ATGHAATGHA (SEQ ID NO: 49), ATGHAACGHA (SEQ ID NO: 50), ATGHAAHGCA (SEQ ID NO: 51), ATGHAAHGTA (SEQ ID NO: 52), AHGTAAHGTA (SEQ ID NO: 53), AHGTAACGHA (SEQ ID NO: 53) Number 54), AHGTAAHGCA (SEQ ID NO: 55), or AHGTAATGHA (SEQ ID NO: 56).
  • n2 structure is the absence of a linker of X b , for example, if X is tyrosine (Y), ACGHAAYGHA (SEQ ID NO: 57), ACGHAAHGYA (SEQ ID NO: 58), AHGCAAYGHA (SEQ ID NO: Number 59), AHGCAAHGYA (SEQ ID NO: 60), AYGHAAYGHA (SEQ ID NO: 61), AYGHAACGHA (SEQ ID NO: 62), AYGHAAHGCA (SEQ ID NO: 63), AYGHAAHGYA (SEQ ID NO: 64), AHGYAAHGYA (SEQ ID NO: 65), AHGYAACGHA (SEQ ID NO: 65) 66), AHGYAAHGCA (SEQ ID NO: 67), or AHGYAAYGHA (SEQ ID NO: 68).
  • Y tyrosine
  • ACGHAAYGHA SEQ ID NO: 57
  • ACGHAAHGYA SEQ ID NO: 58
  • n2 structure is the absence of a linker of X b , for example, if X is aspartic acid (D), ACGHAADGHA (SEQ ID NO: 69), ACGHAAHGDA (SEQ ID NO: 70), AHGCAADGHA ( SEQ ID NO: 71), AHGCAAHGDA (SEQ ID NO: 72), ADGHAADGHA (SEQ ID NO: 73), ADGHAACGHA (SEQ ID NO: 74), ADGHAAHGCA (SEQ ID NO: 75), ADGHAAHGDA (SEQ ID NO: 76), AHGDAAHGDA (SEQ ID NO: 77), AHGDAACGHA (SEQ ID NO: 77) Number 78), AHGDAAHGCA (SEQ ID NO: 79), or AHGDAADGHA (SEQ ID NO: 80).
  • D aspartic acid
  • ACGHAADGHA SEQ ID NO: 69
  • ACGHAAHGDA SEQ ID NO: 70
  • AHGCAADGHA SEQ ID
  • [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker of X b, for example when X is lysine (K) ACGHAAKGHA (SEQ ID NO: 81), ACGHAAHGKA (SEQ ID NO: 82), AHGCAAKGHA (SEQ ID NO: Number 83), AHGCAAHGKA (SEQ ID NO: 84), AKGHAAKGHA (SEQ ID NO: 85), AKGHAACGHA (SEQ ID NO: 86), AKGHAAHGCA (SEQ ID NO: 87), AKGHAAHGKA (SEQ ID NO: 88), AHGKAAHGKA (SEQ ID NO: 89), AHGKAACGHA (SEQ ID NO: 90), AHGKAAHGKA (SEQ ID NO: 91), or AHGKAAKGHA (SEQ ID NO: 92).
  • K is lysine
  • ACGHAAKGHA SEQ ID NO: 81
  • [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker X b
  • X is phenylalanine (F) ACGHAAFGHA (SEQ ID NO: 93), ACGHAAHGFA (SEQ ID NO: 94), AHGCAAFGHA (SEQ ID NO: Number 95), AHGCAAHGFA (SEQ ID NO: 96), AFGHAAFGHA (SEQ ID NO: 97), AFGHAACGHA (SEQ ID NO: 98), AFGHAAHGCA (SEQ ID NO: 99), AFGHAAHGFA (SEQ ID NO: 100), AHGFAAHGFA (SEQ ID NO: 101), AHGFAACGHA (SEQ ID NO: 102), AHGFAAHGFA (SEQ ID NO: 103), or AHGFAAFGHA (SEQ ID NO: 104).
  • a peptide consisting of 1 to 20 amino acid residues may be further included at the 6th position at the 5 'end in the amino acid sequences of SEQ ID NOs: 5 to 104.
  • X may be selected from the group consisting of serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and phenylalanine (F).
  • N1 and n2 may each be two or more, for example, when n1 and n2 are 2, AC MHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), and AXGHA if M motif 1 and M motif 2 each include the same amino acid sequence Either one consisting of (SEQ ID NO: 3) and AHGXA (SEQ ID NO: 4) includes a sequence in which each is repeated twice, or a different sequence such as ACGHA and AHGCA, ACGHA and AXGHA, ACGHA and AHGXA, AHGCA and ACGHA , AHGCA and AXGHA, AHGCA and AHGXA, AXGHA and ACGHA, AXGHA and AHGCA, AXGHA and AHGXA, AHGXA and ACGHA, AHGXA and AHGCA, or AHGXA and AXGHA, respectively.
  • AC MHA SEQ ID NO: 1
  • AHGCA SEQ ID NO: 2
  • AXGHA
  • M motif1 and M motif2 are each any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4). It may include the same sequence corresponding to one, or may be repeated 3-10 times each including a different sequence.
  • n1 and n2 may each be 1, and there may not be a linker of X b , wherein the motif according to the present invention is one or more sequences selected from the group consisting of SEQ ID NOs: 5 to 104, for example. It may include.
  • the motif may bind to the heavy or light chain C-terminus of the antibody, specifically the heavy chain C-terminus, thereby providing a modified antibody and a drug conjugate comprising the same, which significantly increase the drug conjugation yield.
  • the yield of the antibody-drug conjugate can be increased by increasing the yield of the drug.
  • Drugs conjugated in high yield can be specifically delivered to target cancer cells by modified antibodies, thereby increasing the therapeutic effect, and lowering the production cost of the antibody-drug conjugate therapeutics due to the high yield of conjugated antibody-drug conjugates.
  • the motif may be directly bonded in the form of a fusion through an amino bond with the parent antibody, or in a form in which the terminal functional group of the parent antibody and the terminal functional group in the motif are chemically bonded, or mediated to a linker connecting the terminal functional group and the drug in the motif.
  • Linker-mediated forms of coupling are also possible.
  • the linker may be in the form of linking a drug with a specific residue in a motif, and may have a reactive site having an electrophilic group that reacts to a nucleophilic residue (eg, cysteine) present on a motif among modified antibodies.
  • the linker may comprise, for example, reactive functional groups, amino acids and self-cutting spacers that bind to the motif.
  • the functional groups are i) maleimide groups, acetamide groups, or derivatives thereof, ii) aziridine groups, arylhalides, acryloyl groups, or derivatives thereof, iii) alkylation reactors, arylation reactors, pyridyl disulfides, thionitrobenzo Ikic acid, or a derivative thereof.
  • the linker may specifically include, for example, i) a maleimide group or a derivative thereof-valine-citurulline-para-aniline benzoic acid (PABA); Or ii) an acetamide group or derivative thereof-valine-citurulline-paraaniline benzoic acid (PABA), but is not limited thereto.
  • the binding of the moiety to the drug via the linker may be carried out using known methods such as alkylation, disulfide interchange and transthioesterification. This allows the antibody and the drug to be conjugated via the thiol group of the cysteine residue in the motif.
  • the nucleophilic reactivity of the thiol of the cysteine residue to the maleimide group is present in the protein, such as other amino acid functional groups, such as amino groups of lysine residues or Because it is about 1,000 times higher than the N-terminal amino group, it can be used to specifically bind to cysteine.
  • the modified antibody-drug conjugates via maleimide groups, derivatives thereof or acetamide groups or derivatives thereof, such as bromo acetamide groups, iodo acetamide groups can be seen that cysteine is bound to the drug by thioether linkage. .
  • the antibody may be one or more selected from the group consisting of monoclonal antibodies, bispecific antibodies, chimeric antibodies, human antibodies and humanized antibodies. Moreover, both modified antibodies, such as a bispecific antibody, antibody fragments, etc. can be used.
  • 'Antibody fragments' refer to fragments that have at least the binding function to the antigen, and are single-chain antibodies, diabodies, triabodies, tetrabodies, Fab fragments, F (ab') 2 fragments, Fd, scFv, domain antibodies, mini Body, single chain antibody (scAb), derivatives of antibody constant regions, phosphorus based on protein scaffolds and the like.
  • the antibody may be selected from the group consisting of IgA, IgD, IgE, IgG and IgM.
  • the antibody specifically includes cancer specific antigens, cell surface receptor proteins, cell surface proteins, transmembrane proteins, signaling proteins, cell survival regulators, cell proliferation regulators, molecules associated with tissue development or differentiation, lymphokines, cytokines. May have binding capacity and specificity for Cain, a molecule involved in cell cycle regulation, an angiogenesis related molecule, or an angiogenesis related molecule, for example, (1) BMPR1B (bone morphogenetic protein receptor-IB type, gene Bank grant number NM_001203);
  • MPF MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Accession No. NM — 005823
  • Napi3b (NAPI-3B, NPTIIb, SLC34A2, Solute Carrier Family 34 (Sodium Phosphate), Member 2, Type II Sodium-Dependent Phosphate Transporter 3b, GenBank Accession No. NM_006424);
  • Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, Sema Domain, 7 Thrombospondin Repeats (Type 1 and Similar Type 1), Transmembrane Domain (TM) And short cytoplasmic domain, (semaphorin) 5B, Genbank Accession No. AB040878);
  • PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genebank Accession No. AY358628);
  • ETBR endothelin type B receptor, Genbank Accession No. AY275463
  • MSG783 (10) MSG783 (RNF124, hypothetical protein FLJ20315, Genebank Accession No. NM_017763);
  • STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, Prostate Cancer Related Gene 1, Prostate Cancer Related Protein 1, Prostate 6 Transmembrane Epithelial Antigen 2, 6 Transmembrane Prostate Protein, Genebank Authorization number AF455138);
  • TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subgroup M, member 4, Genbank Accession No. NM_017636);
  • CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, teratocarcinoma-derived growth factor, Genbank accession no. NP — 003203 or NM — 003212);
  • CD21 CR2 (complementary receptor 2) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792 Genbank Accession No. M26004);
  • CD79b (CD79B, CD79 ⁇ , IGb (immunoglobulin-associated beta), B29, Genbank Accession No. NM — 000626);
  • FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchoring protein 1a), SPAP1B, SPAP1C, GenBank Accession No. NM_030764);
  • ErbB receptors selected from EGFR, HER3 and HER4
  • NCA Gene Accession No. M18728
  • PSCA Genbank Accession No. AJ297436
  • BAFF-R B cell activating factor receptor, BLyS receptor 3, BR3, NP_443177.1
  • CD22 B-cell receptor CD22-B isotype, NP-001762.1
  • CD79a, CD79A, CD79 ⁇ , and immunoglobulin-associated alpha which are covalently interacting with CD79a (Ig beta (CD79B) and forming complexes on the surface with IgM molecules, are signals involved in B cell differentiation Forwarded, Genbank approval number NP_001774.1);
  • CXCR5 (Bucket Lymphoma Receptor 1, a G protein coupled receptor activated by CXCL13 chemokines, acts on lymphocyte migration and humoral defense, participates in HIV-2 infection, and develops AIDS, lymphoma, myeloma and leukemia Considered to be related to, Genbank approval number NP_001707.1);
  • HLA-DOB beta subunit of MHC class II molecules (Ia antigen), binding to peptides and presenting in CD4 + T lymphocytes, Genbank Accession No. NP — 002111.1);
  • P2X5 purine receptor P2X ligand-gate ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, the lack of which may contribute to the pathophysiology of idiopathic detrusor instability Yes, Genbank approval number NP_002552.2);
  • CD72 B-cell differentiation antigen CD72, Lyb-2, Genbank Accession No. NP — 001773.1
  • Lymphocyte antigen 64 (RP105), a type I membrane protein of the LY64 (leucine rich repeat (LRR) family), modulates B cell activation and apoptosis, and its loss of function is attributed to systemic lupus erythematosus patients. Associated with increased disease activity, GenBank Accession No. NP_005573.1);
  • FcRH1 Fc receptor-like protein 1, a putative receptor for immunoglobulin Fc domains containing C2 Ig-like and ITAM domains, may be involved in B lymphocyte differentiation, Genbank accession number NP_443170.1) ;
  • IRTA2 associated gene deregulation by immunoglobulin macrophage receptor translocation, a putative immunoreceptor capable of acting on B cell development and lymphoma development, occurs in some B cell malignancies, Genbank approval number NP_112571.1);
  • TENB2 estimated transmembrane proteoglycan associated with EGF / Heregulin family of growth factors and follistatin, Genbank Accession No. AF179274);
  • folate receptor a family of proteins expressed by the FoLR gene, which has high binding capacity with Folic acid and carries 5-methyltetrahydrofolate into cells
  • gangliosides GD2, GD3, GM2;
  • (64) may have binding capacity to one or more targets selected from the group consisting of tenascin, but is not limited thereto.
  • an antibody that binds to a folate receptor comprising a heavy chain of SEQ ID NO: 115 and a light chain of SEQ ID NO: 116 was used as a parent antibody.
  • a metal ion binding motif to the heavy chain terminal of the parent antibody to prepare a variety of modified antibodies according to the sequence and placement of the peptide motif, the difference in the conjugate ratio of the drug according to the modified antibody was measured.
  • the modified antibody may include one or more heavy chains selected from the group consisting of SEQ ID NOs: 117 to 121.
  • an antibody that specifically binds to Her2 was used as the parent antibody.
  • a metal ion binding motif to the heavy chain terminal of the parent antibody to prepare a variety of modified antibodies according to the sequence and placement of the peptide motif, the difference in the conjugate ratio of the drug according to the modified antibody was measured.
  • the antibody-drug conjugate according to the present invention had a similarly high drug conjugation yield when the motifs were introduced into Farletuzumab and Trastuzumab. Therefore, the motif according to the present invention can be utilized as a platform technology for binding a drug to an antibody in antibody-drug conjugate preparation regardless of the type of antibody.
  • the antibody may comprise the variable region of the parent or modified antibody and CH1, CH2 and CH3 of IgG2 or IgG4.
  • the VH and VL of Farletuzumab, Trastuzumab or its modified antibodies can be used and include CH1, CH2, CH3 of IgG2 or IgG4.
  • the variable region of the Farletuzumab antibody may include a heavy chain variable region of SEQ ID NO: 122 and / or a light chain variable region of SEQ ID NO: 123.
  • the antibody may comprise a Fab of the parent antibody or a modified antibody and an Fc of IgG2 or IgG4. Specifically, it may include a form in which the Fab portion of Farletuzumab, Trastuzumab or its modified antibody and the Fc portion of IgG2 or IgG4 are fused.
  • the Fab of the Farletuzumab antibody may comprise a heavy chain variable region of SEQ ID NO: 122 and a CH1 containing sequence (SEQ ID NO: 124) and / or a light chain variable region of SEQ ID NO: 123.
  • the Trastuzumab antibody may comprise a heavy chain of SEQ ID NO: 127 and / or a light chain of SEQ ID NO: 128.
  • the drug combined with the modified antibody in the present invention can be used without limitation any drug that has a therapeutic effect of the disease, and particularly preferred is a drug for treating cancer having a proliferation inhibitory effect of tumor cells.
  • the drug may be conjugated to the cysteine group or serine group of the motif introduced at the terminal of the modified antibody.
  • Drugs that may be used in the modified antibody-drug conjugates of the present invention specifically include any compound, moiety or group that has a cytotoxic or cytostatic effect, and (i) a microtubulin inhibitor, mitosis inhibitor, topoiso Chemotherapeutic agents that can function as merase inhibitors, or DNA intercalators; (ii) protein toxins that can function enzymatically; (iii) micro RNA (miRNA), siRNA, shRNA capable of inhibiting the expression of certain oncogenes; Or (iv) radioisotopes.
  • a microtubulin inhibitor mitosis inhibitor, topoiso Chemotherapeutic agents that can function as merase inhibitors, or DNA intercalators
  • protein toxins that can function enzymatically
  • miRNA micro RNA
  • siRNA siRNA
  • shRNA capable of inhibiting the expression of certain oncogenes
  • radioisotopes radioisotopes.
  • Such drugs include, for example, maytansinoids, orstatin, aminopterin, actinomycin, bleomycin, thalisomycin, camptocecin, N8-acetyl spermidine, 1- (2 chloroethyl) -1, 2-dimethyl sulfonyl hydrazide, esperamycin, etoposide, 6-mercaptopurine, dolastatin, tricortesene, calicheamicin, taxane, methotrexate, vincristine, vinblastine, doxorubicin, melphalan, mito Mycin A, mitomycin C, chlorambucil, duocarmycin, nucleolytic enzymes, toxins derived from bacteria or plants, cisplatin, irinotecan, paclitaxel and docetaxel, but is not limited thereto.
  • the drug may react with an amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, which can react to form covalent bonds with electrophilic groups on the linker and linker reagent, And one or more nucleophilic groups selected from the group consisting of arylhydrazide groups.
  • the present invention provides a therapeutic composition
  • a therapeutic composition comprising the antibody-drug conjugate as an active ingredient.
  • the drug of the modified antibody-drug conjugate in the composition is characterized by the use of an antibody conjugate for topical delivery of a drug that kills or inhibits tumor cells in the treatment of cancer, with the drug moiety being targeted to the antibody-antigen into the tumor and into the cell. Enable accumulation
  • the present invention also provides a method of inhibiting proliferation of target cells by contacting the target cells with cancer, autoimmune, inflammatory or infectious diseases or diseases using the modified antibody-drug conjugate as an active ingredient.
  • Treatable cancers in the present invention are liver cancer, stomach cancer, breast cancer, colon cancer, bone cancer, pancreatic cancer, head or neck cancer, uterine cancer, ovarian cancer, rectal cancer, esophageal cancer, small intestine cancer, anal muscle cancer, colon cancer, fallopian tube carcinoma, endometrial carcinoma,
  • a modified antibody-drug conjugate may be contacted in KB cells, which are cancer cells amplified with in vitro folate receptors, to induce cell proliferation inhibition. Therefore, the inhibitory method using the modified antibody-drug conjugate of the present invention as an active ingredient has an effect of killing or reducing the rate of proliferation and inhibiting cells associated with the disease.
  • Expression vector cloning was performed using the pAV4 vector, which was developed and modified for the purpose of using the parent vector pSGHV0 (GenBank Accession No. AF285183) to manufacture antibodies in the industry.
  • the parent vector is overexpressed into cells when a human-derived protein is expressed using bacteria such as Escherichia coli, but in the case of a protein that is difficult to obtain as an active substance, a high concentration of the protein of interest having a physiological activity outside the cell is obtained using animal cells. It is a research vector produced for the purpose of easy purification by expression. However, since there are various limitations to use for production in industry, the vector has been improved to be used in industry in order to use the high expression amount, which is the biggest advantage of this vector for production. In addition, in the case of an antibody, a heavy chain and a light chain must simultaneously express two proteins, so a vector suitable for this purpose has been developed.
  • Example 2 Vector Preparation of Modified Antibodies Incorporating ACGHA, a Parent Antibody Having a Binding Capability to the Folate Receptor, and a Metal Ion Binding Motif
  • a parental antibody (Fwt) vector capable of binding to a folate receptor
  • the heavy chain of SEQ ID NO: 125 and the light chain coding cDNA of SEQ ID NO: 126 were respectively synthesized into codon optimized sequences to maximize expression in CHO cells.
  • This gene was cloned into XhoI / NotI and ApaI / SmaI of the pAV4 vector, respectively, to prepare a parental antibody vector (pFwt).
  • XhoI-Q5-F was used as a template to prepare FM2 (Fwt-ACGHAACGHA (SEQ ID NO: 5), FM2), a modified antibody of Fwt having two metal ion binding motifs (ACGHA), as a template.
  • PCR was performed using forward primers (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 105) and M2 reverse primer (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': SEQ ID NO: 106).
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2).
  • FM1 Fwt-GGGACGHA, pFM1
  • ACGHA metal ion binding motif
  • a forward primer 5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 '
  • reverse primers 5'-CCATGCGGCCGCTCATTTAGGCATGGCC ACAAGCA CCTC CACCACCCGGAGACAGGGAGA-3 ': SEQ ID NO: 108
  • Ez004S Site-directed mutagenesis
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM1).
  • a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': Site-directed mutagenesis (SEQ ID NO: 109) and reverse primers (5'-CCATGCGGCCGCTCATTTAGGCATGGCCACAAGCACCTCCACCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3 ': SEQ ID NO: 110) using two methods: site-directed mutagenesis (enzymeics EzChange site-directed mutagenesis kit, PCR using two metal ion methods) Glycine linkers were added between the motifs.
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2L).
  • ACGHAACGHA (SEQ ID NO: 5)
  • two metal ion binding motifs present in the FM2 modified antibody a modified antibody FM2a (Fwt-ASGHAACGHA ( SEQ ID NO: 26), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ') and a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCGA AGGCACCCG: SEQ ID NO: 112) was used to replace inner cysteine with serine using PCR.
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with the expression vector pFwt having the XhoI / NotI cleavage to prepare a modified antibody vector (pFM2a).
  • the two metal ion binding motifs present in the FM2 modified antibody, FM2b (Fwt-ACGHAASGHA (SEQ ID NO: 21)), in which only one metal ion binding motif exists, by replacing the outer cysteine with serine
  • a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': SEQ ID NO: 113)
  • a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACCC-3): Internal cysteine was replaced with serine by PCR using site-directed mutagenesis (EzChange Site-directed mutagenesis kit, Ez004S).
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2b).
  • CHO-K1 Chinese hamster ovary cells
  • DMEM Dulbecco's Modified Eagle Media
  • FBS Fetal Bovine Serum
  • antibiotics at 37 ° C, 5% CO 2 , incubator.
  • DMEM Dulbecco's Modified Eagle Media
  • FBS Fetal Bovine Serum
  • antibiotics at 37 ° C, 5% CO 2 , incubator.
  • cells were inoculated at a concentration of 5 ⁇ 10 6 / ml in a 100 mm culture dish, followed by incubation with 800 ⁇ l of DMEM without FBS and antibiotics, and 10 ⁇ g of Fwt or modified.
  • the antibody expression vector was mixed and maintained at room temperature for 1 minute, and then mixed with 20 ⁇ g of PEI (Polyethylenimine, linear, Polysciences Inc (Cat. No: 23966, MW ⁇ 25,000)) and left at room temperature for 10 to 15 minutes. .
  • PEI Polyethylenimine, linear, Polysciences Inc (Cat. No: 23966, MW ⁇ 25,000)
  • the cells cultured one day ago were washed with PBS and 6 ml of DMEM was added.
  • Expression vectors of Fwt or its modified antibodies left at room temperature for 10-15 minutes were added to this culture dish.
  • Fwt and its metal ion binding motif modified antibody was purified as follows. Specifically, in order to purify the Fwt and its metal ion binding motif-modified antibody secreted into the cell culture, the culture medium was centrifuged to remove the cells, and then only the supernatant was taken and the supernatant was equilibrated with the equilibration buffer HiTrap Protein A HP. (GE Healthcare, USA) The protein was eluted by injection into the column, washing well with equilibration buffer, and changing the pH with Glycine buffer (100 mM Glycine, pH 2.8). The solution was dialyzed with phosphate buffer, concentrated using Vivaspin20 (Sartorius, USA), and finally purified protein was obtained with high purity.
  • Glycine buffer 100 mM Glycine, pH 2.8
  • a modified antibody of MMAE and Fwt was conjugated to prepare an FMx (metal ion-binding motif variant of Fwt) -MMAE conjugate.
  • FMx metal ion-binding motif variant of Fwt
  • MMAE conjugation derivative of Auristatin known as MMAE
  • MC maleimido caproic acid
  • V valine-citurulline
  • PAB para-aniline benzoic acid
  • Auristatin is a substance with strong intracellular toxicity and IC 50 value in the cell proliferation inhibition test is known to be 200 ⁇ 300 pM. .
  • Modified antibodies such as FM1, FM2, FM2a, and FM2L show conjugation yields of about 63-66% under the same drug conjugation conditions, while FM2b shows very high drug conjugation yields of 97.5%.
  • FM2b nearly identical conjugation yields were shown in samples from two different transfection batches.
  • an antibody-drug conjugate that binds to a thiol group of cysteine through a bromoacetamide group was prepared.
  • Bromoacetamide is bound to MMAE via valine-citurulline, which is degraded by protease in cells, and para-aniline benzoic acid (PABA), a self-decomposing spacer group.
  • PABA para-aniline benzoic acid
  • MMAE is bound to the modified antibody through the coupling of bromoacetamide and thiol group. This is called br (bromo acetamide) -VC (valine-citurulline) -PAB-MMAE.
  • the yield of drug conjugated to the heavy chain between each of the modified antibodies shows that the conjugate yield of FM2b is superior to other modified antibodies.
  • FM2b showed superior conjugation yield compared to other modified antibodies.
  • a modified antibody was prepared by introducing a metal ion motif containing cysteine at the C-terminus of trastuzumab.
  • HM2 HR-ACGHAACGHA (SEQ ID NO: 5), HM2
  • ACGHA metal ion binding motifs
  • pHR a vector of the parent antibody trastuzumab was used.
  • PCR was performed using forward primers (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 111) and M2 reverse primers (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': SEQ ID NO: 112).
  • the amplified nucleotides were cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2).
  • ACGHAACGHA (SEQ ID NO: 5), two metal ion-binding motifs present in the HM2 modified antibody, a substitution antibody HM2a (HR-ASGHAACGHA ( SEQ ID NO: 26), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 111) and a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCGA AGGCAG-3CC): SEQ ID NO: 112) was used to replace inner cysteine with serine using PCR.
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2a).
  • HM2b two metal ion binding motifs present in the HM2 modified antibody, HM2b (HR-ACGHAASGHA (SEQ ID NO: 21)), a variant antibody in which only one metal ion binding motif exists by replacing the outer cysteine with serine
  • the forward antibody (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': SEQ ID NO: 109)
  • the reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACGA)
  • EzChange Site-directed mutagenesis kit, Ez004S site-directed mutagenesis
  • the amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2b).
  • CHO-K1 Chinese hamster ovary cells
  • HM2, HM2a, HM2b trastuzumab modified antibodies
  • HM2b trastuzumab modified antibodies
  • DMEM Dulbecco's Modified Eagle Media
  • FBS Fetal Bovine Serum
  • the modified antibody expressed as above was purified as follows. Specifically, in order to purify the modified antibody secreted into the cell culture, the culture medium was centrifuged to remove the cells, and then only the supernatant was taken and the supernatant was equilibrated with the equilibration buffer HiTrap Protein A HP (GE Healthcare, USA) column. The protein was eluted by changing the pH with Glycine buffer (100mM Glycine, pH 2.8) after washing with equilibration buffer. The solution was dialyzed with phosphate buffer, concentrated using Vivaspin20 (Sartorius, USA), and finally purified protein was obtained with high purity.
  • Glycine buffer 100mM Glycine, pH 2.8
  • the modified antibody of trastuzumab produced in Example 6, 7 was conjugated with MMAE to prepare a HMx (metal ion binding motif variant of Trastuzumab) -MMAE conjugate.
  • HMx metal ion binding motif variant of Trastuzumab
  • MMAE metal ion binding motif variant of Trastuzumab
  • TCEP a reducing agent per equivalent of purified antibody
  • MC-vc-PAB-MMAE 2.5 equivalents of MC-vc-PAB-MMAE are added and reacted at room temperature for 2 hours.
  • reaction was terminated by the addition of excess cysteine, and excess MC-vc-PAB-MMAE and TCEP were removed through dialysis in a centrifugal filtration filter and phosphate buffer to remove the final purified FMx-MC-vc-PAB-MMAE. Prepared.
  • the yield of drug conjugation to the heavy chain between the modified antibodies shows a very large difference.
  • the modified antibodies of HM2 and HM2a show about 55-62% conjugation yield under the same drug conjugation conditions, while HM2b shows very high drug conjugation yield of 85.5%.
  • M2b in which the rear cysteine was substituted with serine in M2 (ACGHAACGHA), which is a metal ion binding motif, showed much higher drug binding ability than the M2 sequence.
  • a modified antibody was substituted with various amino acids at this position.
  • Modified antibody which substituted another amino acid at serine site in M2b sequence
  • Modified antibody C-terminal metal ion binding motif sequence M2b or M2b-S ACGHAASGHA SEQ ID NO: 21) M2b-A ACGHAAAGHA (SEQ ID NO: 33) M2b-T ACGHAATGHA (SEQ ID NO 45) M2b-Y ACGHAAYGHA (SEQ ID NO 57) M2b-D ACGHAADGHA (SEQ ID NO: 69) M2b-K ACGHAAKGHA (SEQ ID NO: 81) M2b-F ACGHAAFGHA (SEQ ID NO: 93)
  • modified antibodies having the C-terminal sequence as shown in Table 7 above were introduced into FM2b or FM2b-S, respectively, of FM2b-A, FM2b-T, FM2b-Y, FM2b-D, FM2b-K, and FM2b-F. Modified antibodies were produced.
  • X A, T, Y, D, K, or F
  • TCEP a reducing agent per equivalent of purified antibody
  • MC-vc-PAB-MMAE 2.5 equivalents of MC-vc-PAB-MMAE are added and reacted at room temperature for 2 hours.
  • the reaction was terminated by the addition of excess cysteine and excess MC-vc-PAB-MMAE and TCEP were removed via dialysis in a centrifugal filtration filter and phosphate buffer solution to obtain the final purified FMb-X-MC-vc-PAB- MMAE was prepared.
  • the yield of conjugation to the heavy chain of each modified antibody is shown in Table 8 below.
  • Each modified antibody prepared as in the above embodiment four-drug conjugate is modified number of the bonding substance (DAR: drug-to-antibody ratio) per antibody because it is different, to compare the cytotoxicity of the drug from in vitro Is not easy. Therefore, in order to purify the modified antibody-drug conjugate with the same DAR, the modified antibody-drug conjugate was purified using hydrophobic chromatography. Phenyl column chromatography was used to purify the modified antibody-drug conjugate having a DAR of 2. The column was equilibrated with 10 mM sodium succinate, 0.5 M NaCl, pH 5.0 buffer, and then the modified antibody-drug conjugate was injected into the column.
  • DAR drug-to-antibody ratio
  • the buffer containing 30% acetonitrile was added to elute the modified antibody-drug conjugate according to DAR.
  • the eluted modified antibody-drug conjugates were exchanged for buffer using dialysis in a buffer solution of 10 mM sodium succinate, 30 mM sucrose, pH 6.0.
  • FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y which are antibody-drug conjugates having two conjugated MC-vc-PAB-MMAE drugs as in Examples 9, 10, and 11 above Produced -D2.
  • the produced antibody-drug conjugates were incubated at 25 ° C. and 50 ° C., respectively, to measure the change in the number of drug conjugates and the change in aggregation.
  • Each test sample was prepared at a concentration of 110 uL at a concentration of 1 mg / mL, each of 12.
  • Six samples of each antibody-drug conjugate were stored at 25 ° C. and the remaining six samples were kept at 0 ° C. for 1, 3, 5, 7, and 14 days to measure changes in DAR and monomer.
  • the content of DAR2 was decreased by 1.5 ⁇ 2% for each sample, and the purity of monomer was observed about 0.3 ⁇ 4%, but the difference was not large. Changes in DAR2 content and monomer purity at 50 ° C were larger than those observed at 25 ° C, but the differences between samples were not significant. Therefore, it was confirmed that the difference between the antibody variants is not very large.
  • cell growth inhibitory activity was performed using KB-cells overexpressed with the folate receptor.
  • KB-cells were diluted in DMEM / F12 medium with 10% FBS adjusted to 1 ⁇ 10 4 / well and 100 ⁇ l cell culture was added to each well of a 96-well plate. It was. The well plates were then incubated for 24 hours in an incubator set at 5% carbon dioxide and 37 ° C. to attach the cells to the plates.
  • Each test sample was diluted in medium and added to final concentrations of 6.45 nM, 3.23 nM, 1.61 nM, 0.806 nM, 0.403 nM, 0.202 nM, 0.101 nM, 0.0504 nM, 0.0252 nM and 0.0126 nM, together with the medium in the control wells. (No drug) was added.
  • CellTiter 96- AQueous One Solution reagent [MTS-based assay; MTS forms purple formazan by dehydrogenase of living cells, and proliferation is measured by the amount of purple formazan produced]
  • MTS-based assay MTS forms purple formazan by dehydrogenase of living cells, and proliferation is measured by the amount of purple formazan produced
  • Cell lysis was measured at 490 nm using an absorbance spectrometer to determine viability (%).
  • FM2-D2 modified antibody-drug conjugate having DAR 2 as MMAE drug conjugate of modified antibody FM2
  • FM2b purified after conjugation of the parent antibody Fwt with MMAE in the above example
  • -D2 modified antibody-drug conjugate having DAR 2 as MMAE drug conjugate of modified antibody FM2b
  • the antibody-drug conjugates FM2-D2 and FM2b-D2 show superior anticancer efficacy compared to the parent antibody.
  • FM2-D2 and FM2b-D2 show nearly identical cancer cell growth inhibition.
  • MC-vc-PAB-MMAE was conjugated to FM2b-S, -F, and -Y variants and purified to have the same DAR. Then, after treating the KB cells as described above, the cell growth inhibition of the drug was compared.
  • FM2b-S-D2 and FM2b-F-D2 show almost the same cell growth inhibition.
  • the respective IC 50 values were 0.25 nM for FM2b-S-D2, 0.26 nM for FM2b-F-D2, and 0.24 nM for FM2b-Y-D2.
  • the antibody-drug conjugate produced by the antibody variant according to the present invention can greatly improve the productivity of the antibody-drug conjugate by increasing the conjugation yield of the drug.
  • the drug conjugated to the terminal portion of the antibody through position-specific conjugation does not inhibit the structural stability of the parent antibody, thereby maintaining the antigen specificity and structural stability of the parent antibody originally, and the high antigen possessed by the parent antibody. Specificity allows the drug conjugated to the antibody to be specifically delivered to cancer cells.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Animal Behavior & Ethology (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Immunology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Mycology (AREA)
  • Microbiology (AREA)
  • Organic Chemistry (AREA)
  • Biochemistry (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)

Abstract

The present invention relates to: an antibody-drug conjugate in which a modified antibody, containing a motif of a specific structure at an end of the antibody, is coupled to a drug through a linker; and a composition containing the same and, specifically, to: a modified antibody-drug conjugate (mADC) comprising a modified antibody coupled to the C-terminus of heavy chain or light chain so as to have a remarkably increased drug conjugation yield; and a composition containing the same.

Description

변형항체를 포함하는 항체-약물 접합체Antibody-Drug Conjugates Including Modified Antibodies
본 발명은 특정 구조의 모티프를 항체의 말단에 포함하는 변형항체가 링커를 통해 약물에 결합된 항체-약물 접합체 및 이를 포함하는 조성물에 관한 것으로, 구체적으로는 중쇄 또는 경쇄 C-말단에 결합되어 약물 접합 수율이 현저히 증가한 변형항체를 포함하는 변형항체-약물 접합체(mADC, modified Antibody-Drug Conjugate) 접합체 및 이를 포함하는 조성물에 관한 것이다.The present invention relates to an antibody-drug conjugate in which a modified antibody comprising a motif of a specific structure at the terminal of the antibody is bound to a drug through a linker, and specifically to a drug linked to the heavy or light chain C-terminus. A modified Antibody-Drug Conjugate (mADC) conjugate comprising a modified antibody having a significantly increased conjugation yield and a composition comprising the same.
항암 치료에 이용되는 약물은 흔히 독성, 특히 골수, 점막, 신경 독성을 나타낸다. 따라서, 강한 항암작용을 하면서 더 안전하면서 암세포에 특이성을 보여주는 항암제의 개발이 요구된다. 암세포에만 특이적으로 작용하면서 부작용은 감소하는 항암제는 여러 방면에서 개발이 진행되고 있다. Drugs used for chemotherapy often show toxicity, especially bone marrow, mucosa, and neurotoxicity. Therefore, there is a need for the development of an anticancer agent that is safer while showing strong anticancer activity and specificity to cancer cells. Anti-cancer drugs that act only on cancer cells and reduce side effects are being developed in various ways.
이러한 측면에서 특정 질환에서 특이적으로 발현되는 표적 (target), 즉 항원에 특이적으로 결합하는 항체를 이용한 치료제는 바이오 의약품 중에서 현재 가장 활발하게 연구가 진행되고 있다. 특히 암 세포 표면에 특이적으로 발현되는 종양-관련 항원을 규명하고, 이에 결합하여 세포의 성장을 억제하거나 사멸을 유도하는 항체, 즉 항암 항체를 이용한 종양 진단 및 치료 방법은 현재 널리 사용되고 있으며, 앞으로의 전망도 매우 밝은 기술분야이다. In this regard, therapies using antibodies that are specifically expressed in specific diseases, that is, antibodies that specifically bind to antigens, are currently being actively studied among biopharmaceuticals. In particular, methods for diagnosing and treating tumors using antibodies, such as anti-cancer antibodies, which identify tumor-associated antigens specifically expressed on the surface of cancer cells, bind to them, and inhibit cell growth or induce death, are now widely used. Is also a very bright technology field.
하지만, 이러한 항암 항체는 표적 특이성은 매우 높지만, 암세포의 사멸효과는 기존의 세포독성 약물(항암제), 즉 항암 약물에 비해 낮은 경우가 많기 때문에, 세포독성 약물 및 기타 세포 증식 억제 약물 등과의 병용 투여요법(combination therapy)으로 사용되는 경우가 많다. 항암 약물은 항암 항체에 비해서는 월등히 높은 세포독성을 보이지만, 암세포로의 표적 특이성은 낮기 때문에 항체 치료제에 비하여 매우 높은 부작용(side effect)를 보인다. 따라서, 항암 항체와 항암 약물의 병용 투여요법은 각각의 약물을 개별 투여했을 때보다 더 높은 치료 효과를 보이지만, 항암 약물의 부작용은 항상 수반된다는 근본적인 한계를 가지고 있다. However, these anticancer antibodies have very high target specificities, but the killing effect of cancer cells is often lower than that of conventional cytotoxic drugs (anticancer drugs), so they may be used in combination with cytotoxic drugs and other cell proliferation inhibitors. It is often used as a combination therapy. Anticancer drugs show significantly higher cytotoxicity than anticancer antibodies, but have very high side effects compared to antibody therapeutics because of their low target specificity to cancer cells. Therefore, the combination therapy of anticancer antibody and anticancer drug shows a higher therapeutic effect than when each drug is administered separately, but has the fundamental limitation that side effects of anticancer drug are always accompanied.
또한, 매우 높은 세포독성 때문에 항암 약물 중에서 단독 처방으로 항암 치료제로 쓸 수 있는 약물은 상대적으로 독성이 낮은 택솔 계열이나 시스플라틴 계열의 약물로 제한적이다. 세포독성이 큰 대부분의 항암 약물의 경우는 매우 높은 세포독성 때문에 단독 약물로 처방하기는 사실상 불가능하다. 단독 처방으로 사용하기는 불가능한 항암 약물을 암세포에 대한 표적 특이성이 매우 높은 항체에 결합시킴으로써 정상 세포로의 부작용 없이 표적 암세포에만 항암 약물을 운반할 수 있다. 따라서, 항체-약물 접합체는 기존에는 사용 불가능한 항암 약물의 치료효능을 제고할 수 있는 방법으로 각광받고 있다. In addition, due to its very high cytotoxicity, among the anticancer drugs, drugs that can be used alone as anticancer drugs are limited to relatively low toxicity Taxol or cisplatin-based drugs. Most anticancer drugs with high cytotoxicity are virtually impossible to prescribe as single drugs because of their very high cytotoxicity. By binding an anticancer drug that cannot be used as a prescription alone to an antibody having a very high target specificity for cancer cells, the anticancer drug can be delivered only to the target cancer cells without the side effects of normal cells. Therefore, antibody-drug conjugates have been in the spotlight as a method for improving the therapeutic efficacy of anti-cancer drugs that cannot be used conventionally.
현재 시장에 출시된 항체-약물 접합체는 호지킨 림프종 치료제인 애드세트리스(Adcetris®)와 전이성 유방암 치료제인 캐드싸일라(Kadcyla®)가 있다. 이들 항체-약물 접합체는 세포의 분열과정에 관여하는 세포내 미세소관인 튜불린(tubuline)에 접합하여 세포의 분열을 억제함으로써 암의 성장과 분열을 저해하는 튜불린 억제제가 항체에 접합되어 있는 구조를 갖는다. 이들 항체-약물 접합체는 항체가 원래 가지고 있는 라이신에 약물을 접합하거나 (캐드싸일라), 항체의 구조적 안정성을 유지시켜주는 중쇄-중쇄, 혹은 중쇄-경쇄를 연결하는 다이설파이드기를 환원시킨 시스테인기에 약물을 접합한다 (애드세트리스). 이러한 1세대 항체-약물 접합체에 사용된 항암 약물의 접합 방식은 항체당 접합하는 약물의 개수를 조절하지 못하고, 같은 약물 접합수를 갖는 항체-약물 접합체에서도 약물의 접합 위치가 서로 다른 위치 이성질체(positional isomer)가 생성되게 된다. 항체당 약물의 접합 개수는 항체-약물 접합체의 세포독성 뿐만 아니라 항체-약물 접합체의 안정성, 응집체 형성 가능성 등에도 영향을 미치는 요소로써, 일반적으로 약물이 많이 접합할수록 항체 자체의 안정성이 떨어지고 응집체의 형성 가능성이 높아진다. Antibody-drug conjugates currently on the market include Adcetris®, a treatment for Hodgkin's lymphoma, and Kadcyla®, a treatment for metastatic breast cancer. These antibody-drug conjugates are conjugated to tubulin inhibitors that inhibit the growth and division of cancer by conjugating to the tubule, an intracellular microtubule involved in cell division, thereby inhibiting cell division. Has These antibody-drug conjugates bind the drug to the lysine originally possessed by the antibody (cadcyla), or to the cysteine group that reduced the disulfide group linking the heavy chain-heavy chain or heavy-light chain that maintains the structural stability of the antibody. (Adsetless). The anticancer drug conjugation method used in the first-generation antibody-drug conjugate does not control the number of conjugated drugs per antibody, and even in antibody-drug conjugates having the same number of drug conjugates, the positional conjugations of the drugs are different from each other. isomer) is generated. The number of conjugates of drugs per antibody affects not only the cytotoxicity of antibody-drug conjugates but also the stability of antibody-drug conjugates and the possibility of aggregate formation.In general, the more conjugated drugs, the less stable the antibody itself and the formation of aggregates. The chances are high.
또한, 1세대 항체-약물 접합체는 항체당 접합하는 약물의 개수를 조절하지 못하기 때문에, 항체당 접합하는 약물의 개수는 평균적인 수치를 가지게 된다. 예를 들어 캐싸일라의 경우에는 항체에 접합된 약물의 개수가 1개에서 8개까지의 분포를 가지게 되며, 평균적으로 3.5개의 약물 접합수를 가지게 된다. 같은 약물 접합 개수에서도 약물의 접합 위치에 따라서 항체-약물 접합체의 특성이 달라질 수 있다. Fab 부근이나 Fc의 hinge 부근에 약물이 접합하는 경우 항원이나 Fcγ나 FcRn 수용체와의 결합력에 차이가 생김으로 인하여 항체의 안정성이나 항원-항체 반응성이 저해될 가능성도 있다. 항체당 접합하는 약물의 개수가 늘어날수록 약물의 접합 위치가 서로 다른 위치 이성질체의 개수도 비례하여 늘어나며, 이러한 결과는 생산 배치에 따른 항체-약물 접합체의 일관된 특성을 유지하는 데도 큰 영향을 미칠 수 있다.In addition, since the first generation antibody-drug conjugate does not control the number of drugs conjugated per antibody, the number of conjugated drugs per antibody has an average value. For example, in the case of cascaila, the number of drugs conjugated to the antibody may range from 1 to 8, and on average 3.5 drug conjugates. Even in the same number of drug conjugates, the properties of the antibody-drug conjugate may vary depending on the location of the drug conjugate. When the drug is conjugated near the Fab or near the hinge of the Fc, there is a possibility that the stability of the antibody or antigen-antibody reactivity may be impaired due to a difference in the binding force between the antigen and the Fcγ or FcRn receptor. As the number of conjugated drugs per antibody increases, the number of positional isomers with different conjugated positions of the drug increases proportionally, and this result may have a great influence on maintaining the consistent characteristics of the antibody-drug conjugate according to the production batch. .
따라서, 최근에는 위와 같은 제1세대 항체-약물 접합체의 단점을 개선하기 위하여 위치 특이적 약물 접합을 위한 여러 가지 접합 기술들이 개발되고 있다. 제넨텍(Genetech)에서는 항체의 아미노산을 치환하여 시스테인기를 도입한 후, 도입된 시스테인기에 위치 특이적으로 약물을 접합하는 ThioMab 기술을 이용하여 약물을 접합하는 기술을 개발하였다. 이를 통해서 위치 특이적으로 약물이 접합된 항체-약물 접합체가 1세대 항체-약물 접합체의 접합 기술로 제조된 것보다 생체 내에서의 활성이 우수함을 보여주었다 (Junutula et al., Nature Biotechnology, 2008, 26, 925-932). 위치 특이적 접합은 기질 특이성이 매우 높은 단백질 효소를 이용한 약물 접합 방식으로 또한 여러 회사들에서 시도되었다. Therefore, in recent years, various conjugation techniques for site-specific drug conjugation have been developed to ameliorate the disadvantages of the first generation of antibody-drug conjugates. Genentech has developed a technique for conjugating drugs using ThioMab technology, which introduces a cysteine group by substituting an amino acid of an antibody, and then conjugates the drug in a specific position to the introduced cysteine group. This demonstrates that the site-specific drug conjugated antibody-drug conjugate has superior activity in vivo than that produced by the first-generation antibody-drug conjugate (Junutula et al., Nature Biotechnology, 2008, 26, 925-932). Site specific conjugation has also been tried by several companies in a drug conjugation manner using protein enzymes with very high substrate specificity.
포르밀글리신-생성 효소 (Formylglycine-generating enzyme)를 이용한 위치 특이적 접합 (Drake et al., Bioconjugate Chemistry, 2014, 25, 1331-1341), 글루타민 트랜스퍼라제 (glutamine transferase)를 이용한 접합 (Strop et al., Chemistry & Biology, 2013, 20, 161-167) 등의 방법을 통해 원하는 개수의 약물을 위치 선택적으로 항체에 접합할 수 있음이 보고되었다. 그러나, 이러한 단백질 효소를 이용한 접합은 37℃에서 72시간 동안 과량의 약물의 존재하에서 접합을 하거나, 또는 고농도의 접합 효소가 필요한다는 점 등의 단점이 존재한다. Site-specific conjugation using formylglycine-generating enzyme (Drake et al., Bioconjugate Chemistry, 2014, 25, 1331-1341), conjugation using glutamine transferase (Strop et al. , Chemistry & Biology, 2013, 20, 161-167) and the like have been reported that the desired number of drugs can be selectively conjugated to the antibody. However, the conjugation using such a protein enzyme has disadvantages such as conjugation in the presence of an excess of drug at 37 ° C. for 72 hours, or the need for a high concentration of conjugation enzyme.
또 다른 위치 특이적 접합 방법인 인공적 아미노산(non-natural amino acid)를 이용한 접합 방법은 tRNA 합성효소의 mutant를 통해서 인공적 아미노산을 단백질에 도입할 수 있는 tRNA를 합성해냄으로써 자연상태의 아미노산이 가지고 있지 않은 잔기(side-chain)를 단백질 내로 도입할 수 있는 기술에 기반하고 있다 (Wang et al., Proc. Natl. Acad. Sci. USA, 2003, 100, 56-61). 이를 통해서 도입된 인공적 아미노산의 잔기에 위치 특이적 접합을 하는 방식으로 원하는 위치에 약물을 접합할 수 있다 (zimmerman et. Al, Bioconjugate Chem. 2014, 25, 351-361). 그러나, 이러한 방법은 고도의 유전공학적 방법을 통해서 전사단계(translation pathway)를 조절해야 하는 고난위도의 작업이 요구된다.Conjugation using non-natural amino acid, another site-specific conjugation method, is a natural amino acid that synthesizes tRNA that can introduce artificial amino acids into proteins through mutants of tRNA synthase. It is based on a technique that can introduce side-chains into proteins (Wang et al., Proc. Natl. Acad. Sci. USA, 2003, 100, 56-61). This allows the drug to be conjugated at a desired position in such a way as to perform site-specific conjugation to the residues of the artificial amino acids introduced (zimmerman et. Al, Bioconjugate Chem. 2014, 25, 351-361). However, these methods require high-level tasks that require high levels of genetic engineering to regulate the translation pathway.
위와 같은 위치 특이적 접합은 복잡한 유전공학적 방법을 이용하여 전사시스템을 변형하거나 또는 과량의 접합 효소를 첨가해야 하는 등, 위치 특이적 접합 반응을 위한 시간과 비용의 소요가 매우 크다. 항체는 항암 약물을 암세포에 특이적으로 전달하는 운반체(carrier)의 역할을 하며, 약물은 암세포에 운반될 때까지 안정적으로 항체에 접합되어 있으면 되는 항체-약물 접합체의 기본 개념으로 볼 때, 과연 위와 같은 복잡하고 시간과 경제적 비용이 큰 접합 방법을 사용하여 항체-약물 접합체를 제조해야만 하는 필연성에 의문이 제기되지 않을 수 없다. 항체-약물 접합체를 생산하는데 있어서 위치 특이적으로 접합할 수 있으면서, 경제적 효용성에서도 우수한 접합 방식은 기존의 1세대 항체-약물 접합체의 접합 방법뿐만 아니라, 새로이 제시된 위치 특이적 접합방식의 한계를 극복할 수 있는 훌륭한 대안이 될 수 있다.Such site-specific conjugation is very time-consuming and costly for site-specific conjugation reactions, such as modification of transcriptional systems using complex genetic engineering methods or addition of excess conjugation enzymes. Antibodies act as carriers for the specific delivery of anticancer drugs to cancer cells, and the drug is a basic concept of an antibody-drug conjugate that needs to be stably conjugated to an antibody until it is delivered to cancer cells. The necessity of producing antibody-drug conjugates using the same complex and time-consuming and expensive conjugation method is questionable. The location-specific conjugation in the production of antibody-drug conjugates, and the economic conjugation method, which is excellent in economical efficiency, overcome the limitations of the existing method of conjugation of the first generation antibody-drug conjugates as well as the newly proposed location-specific conjugation method. It can be a great alternative.
항체-약물 접합체는 기존의 항체 의약품과 대비하여 월등히 우월한 in vitroin vivo 효능을 보여주고 있다. 그러나, 항체-약물 접합체를 1차 치료제로써 사용하기 위한 몇몇 임상 결과는 단클론 항체 치료제와 화학합성 약물의 복합 치료 (combination therapy)에 비하여 유의한 임상적 효용성의 차이를 보여주는데 실패하였다 (http://www.roche.com/media/store/releases/med-cor-2014-12-19.htm). 이러한 결과는 항체-약물 접합체가 합성 약물뿐 만 아니라 단클론 항체 치료제에 비하여 현저히 높은 치료비가 필요하다는 점을 고려할 때, 항체-약물 접합체의 경제적 효용성에 커다란 제약으로 작용할 수 있다. 이러한 점에서 앞서 언급한 위치 특이적 접합 방식들로는 1차 치료제로써 사용할 수 있는 경제적 효용성을 제공하는 데 있어서 심각한 문제점을 내포하고 있다고 할 수 있다. Antibody-drug conjugates show superior in vitro and in vivo efficacy over existing antibody pharmaceuticals. However, some clinical results for using antibody-drug conjugates as primary therapeutics failed to show significant clinical utility differences compared to the combination therapy of monoclonal antibody and chemosynthetic drugs (http: // www.roche.com/media/store/releases/med-cor-2014-12-19.htm). These results can be a significant constraint on the economic utility of antibody-drug conjugates, given that antibody-drug conjugates require significantly higher therapeutic costs than synthetic drugs as well as monoclonal antibody therapeutics. In this respect, the above-mentioned location-specific conjugation methods have serious problems in providing economic utility that can be used as a primary therapeutic agent.
이러한 기술적 배경하에서, 본 출원의 발명자들은 위치 특이적 접합 반응을 하는 동시에, 제시된 접합 반응보다 경제적 효용성에서 월등히 뛰어난 항체-약물 접합체에 대한 개발이 절실히 요구되고 있음을 인식하고, 이러한 문제를 해결하기 위하여, 금속이온 결합 모티프가 포함된 펩타이드 모티프를 포함하는 변형항체를 통해 위치 특이적 접합을 하는 동시에 보다 월등히 우월한 약물 접합 수율을 갖는 변형항체를 발명하였다. 이러한 발명을 통하여 위치 특이적 접합 방법에 따른 항체-약물 접합체의 우월한 생체내 항암 효과뿐만 아니라, 보다 경제적으로 제조될 수 있는 항체-약물 접합체를 제공하고자 한다.Under this technical background, the inventors of the present application recognize that there is an urgent need for the development of antibody-drug conjugates that perform site-specific conjugation reactions, and which are superior in economic efficiency over the proposed conjugation reactions, and to solve these problems. Invented by a modified antibody comprising a peptide motif containing a metal ion binding motif, a modified antibody having a position-specific conjugation and at the same time superior drug conjugation yield. Through this invention, as well as the superior in vivo anticancer effect of the antibody-drug conjugate according to the site-specific conjugation method, it is to provide an antibody-drug conjugate that can be produced more economically.
발명의 요약Summary of the Invention
상기와 같은 문제를 해결하기 위하여, 본 발명에서는 특정 구조의 모티프가 모항체에 결합되어 약물 결합 부위를 가지는 형태의 항체(이하 변형항체)를 제공한다. 이 변형항체는 기존의 금속이온 결합 펩타이드 모티프를 포함하는 변형항체를 개량하여, 위치 특이적 접합의 특성을 유지하면서 보다 더 높은 약물 접합 수율을 갖는 변형항체를 제공한다. 또한 이 변형항체를 이용한 항체-약물 접합체의 제조 및 항암 약물의 제조 방법을 제공하는 것이다. In order to solve the above problems, the present invention provides a antibody (hereinafter modified antibody) of the type having a specific binding motif to the parent antibody having a drug binding site. These modified antibodies modify modified antibodies comprising existing metal ion binding peptide motifs to provide modified antibodies with higher drug conjugate yields while maintaining the properties of site specific conjugation. The present invention also provides an antibody-drug conjugate using the modified antibody and a method for producing an anticancer drug.
상기 목적을 달성하기 위하여, 본 발명은 하기 구조식 (1)로 표시되는 모티프를 항체의 말단에 포함하는 변형항체가 링커를 통해 약물에 결합된 항체-약물 접합체를 제공한다:In order to achieve the above object, the present invention provides an antibody-drug conjugate wherein a modified antibody comprising a motif represented by the following structural formula (1) at the terminal of the antibody is bound to the drug via a linker:
구조식 (1)Structural Formula (1)
Xa-[Mmotif1]n1-Xb-[Mmotif2]n2 X a - [M motif1] n1 -X b - [M motif2] n2
상기 식에서, Mmotif1 또는 Mmotif2는 각각 독립적으로 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나를 포함하고, 상기 서열번호 3 또는 4에서 X는 시스테인 이외의 아미노산 잔기를 포함하며, Wherein M motif1 or M motif2 each independently include any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4), wherein X in SEQ ID NO: 3 or 4 includes amino acid residues other than cysteine,
Xa 및 Xb는 각각 독립적으로 A (alanine), S (serine), G (glycine)로 이루어진 군에서 선택된 아미노산 잔기가 0개 내지 20개로 구성된 펩타이드이며,X a And X b is a peptide consisting of 0 to 20 amino acid residues each independently selected from the group consisting of A (alanine), S (serine), G (glycine),
n1 및 n2는 각각 1 내지 10의 정수이다. n1 and n2 are integers of 1-10, respectively.
또한, 본 발명은 상기 항체-약물 접합체를 포함하는 암 예방 또는 치료용 조성물을 제공한다.The present invention also provides a composition for preventing or treating cancer comprising the antibody-drug conjugate.
도 1. Folate receptor를 타겟으로 하는 항체의 항체 변이체인 FM2, FM2b (lot no: 4390f), FM2b (lot no: 5698f), FM2a, FM2L, FM1를 MC-vc-PAB-MMAE에 접합한 시료의 SDS-PAGE. 약물이 항체의 중쇄에 도입한 시스테인 잔기를 포함한 모티프에 접합하여, SDS-PAGE 상에서 50kDa 부근의 중쇄에서 두 개의 밴드로 나타난다.Figure 1. Samples conjugated to MC-vc-PAB-MMAE with FM2, FM2b (lot no: 4390f), FM2b (lot no: 5698f), FM2a, FM2L, and FM1 antibody variants of the antibody targeting the Folate receptor SDS-PAGE. The drug is conjugated to a motif containing a cysteine residue introduced into the heavy chain of the antibody, and appears on the SDS-PAGE as two bands in the heavy chain near 50 kDa.
도 2. Folate receptor를 타겟으로 하는 항체의 항체 변이체인 FM2, FM2b, FM2a, FM2L, FM1를 bromoacetamide를 통해 MMAE와 연결된 약물-링커 결합체인 br-vc-PAB-MMAE에 접합한 시료의 SDS-PAGE. 약물이 항체의 중쇄에 도입한 시스테인 잔기를 포함한 모티프에 접합하여, SDS-PAGE 상에서 50kDa 부근의 중쇄에서 두 개의 밴드로 나타난다.Figure 2. SDS-PAGE of a sample conjugated with the antibody variants of FM2, FM2b, FM2a, FM2L, FM1 targeting the Folate receptor to br-vc-PAB-MMAE, a drug-linker conjugate linked to MMAE via bromoacetamide . The drug is conjugated to a motif containing a cysteine residue introduced into the heavy chain of the antibody, and appears on the SDS-PAGE as two bands in the heavy chain near 50 kDa.
도 3. Folate receptor가 과발현된 KB-세포를 이용한 세포성장 억제능 시험. 모항체인 Fwt와 변형항체-약물 접합체인 FM2-D2 (변형항체인 FM2와 MMAE의 변형항체-약물 접합체에서 DAR이 2를 갖는 변형항체-약물 접합체), FM2b-D2 (or FM2b-S-D2) (변형항체인 FM2b-S와 MMAE의 변형항체-약물 접합체에서 DAR이 2를 갖는 변형항체-약물 접합체)의 세포성장 억제능 비교. FM2-D2와 FM2b-D2는 거의 동일한 세포내 활성을 보여주고 있다.Figure 3. Cell growth inhibition test using KB-cells overexpressed Folate receptor. Parent antibody Fwt and modified antibody-drug conjugate FM2-D2 (modified antibody-drug conjugate with DAR 2 in modified antibody-drug conjugate of modified antibody FM2 and MMAE), FM2b-D2 (or FM2b-S-D2) Comparison of cell growth inhibitory ability of (modified antibody-drug conjugate having DAR 2 in modified antibody-drug conjugate of FM2b-S and MMAE modified antibody). FM2-D2 and FM2b-D2 show almost the same intracellular activity.
도 4. Folate receptor가 과발현된 KB-세포를 이용한 세포성장 억제능 시험. FM2b-S-D2, FM2b-F-D2, FM2b-Y-D2의 세포성장 억제능 비교. FM2b-S-D2, FM2b-F-D2, FM2b-Y-D2는 거의 동일한 세포내 활성을 보여주고 있다.Figure 4. Cell growth inhibition test using KB-cells overexpressed Folate receptor. Cell growth inhibition of FM2b-S-D2, FM2b-F-D2, and FM2b-Y-D2. FM2b-S-D2, FM2b-F-D2, and FM2b-Y-D2 show almost the same intracellular activity.
도 5. FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y-D2의 25℃와 50℃에서 0, 1, 3, 5, 7, 14 일간 보관한 후, 약물의 접합개수 DAR과 aggregate의 변화 측정. 25℃에서는 DAR과 monomer의 변화가 거의 발견되지 않았으나, 50℃에서는 DAR2의 함량이 감소하였고 aggregate가 시간에 따라서 형성됨이 확인되었다. 그러나 각 변이체별로 DAR의 변화와 aggregate 형성에 있어서 차이점은 관찰되지 않았다.Figure 5. FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y-D2 after storage for 0, 1, 3, 5, 7, 14 days at 25 ℃ and 50 ℃, drug Determination of the change in the number of joints DAR and aggregate. Almost no changes of DAR and monomer were observed at 25 ℃, but at 50 ℃, the content of DAR2 was decreased and aggregates formed over time. However, no differences in DAR changes and aggregate formation were observed for each variant.
발명의 상세한 설명 및 바람직한 Detailed description of the invention and preferred 구현예Embodiment
다른 식으로 정의되지 않는 한, 본 명세서에서 사용된 모든 기술적 및 과학적 용어들은 본 발명이 속하는 기술분야에서 숙련된 전문가에 의해서 통상적으로 이해되는 것과 동일한 의미를 갖는다. 일반적으로, 본 명세서에서 사용된 명명법은 본 기술분야에서 잘 알려져 있고 통상적으로 사용되는 것이다.Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. In general, the nomenclature used herein is well known and commonly used in the art.
항체-약물 접합체는 타겟 암세포로 항암 약물을 전달하기 전까지 항암 약물이 항체에 안정적으로 결합되어 있어야 한다. 타겟 암세포로 전달된 항암 약물은 항체로부터 유리되어 암세포의 사멸을 유도해야 한다. 이를 위해서는 항암 약물을 링커를 이용하여 항체에 안정적으로 결함함과 동시에 암세포에서 유리될 때는 암세포의 사멸을 유도할 충분한 세포독성을 갖는 약물-링커의 구조를 가지고 있어야 한다. 또한, 약물은 항체에 위치 특이적으로 접합함으로써 항체-약물 접합체의 항함 효능성과 제조 과정에서의 균일성을 보여주어야 한다. The antibody-drug conjugate must be stably bound to the antibody until the anti-cancer drug is delivered to the target cancer cell. Anticancer drugs delivered to target cancer cells must be released from the antibody to induce the death of cancer cells. To this end, the anti-cancer drug must be stably deficient in the antibody by using a linker and have a structure of a drug-linker having sufficient cytotoxicity to induce cancer cell death when released from cancer cells. In addition, the drug should show the anti-potency of the antibody-drug conjugate and its uniformity in the manufacturing process by site-specific conjugation to the antibody.
이를 위하여, 본 출원의 발명자들은 한국등록특허 제1541764호에서 항체의 C-말단에 금속이온 결합 모티프를 포함하는 펩타이드를 도입하여 위치 특이적으로 약물이 접합할 수 있으며, 일정한 항체 대 약물의 접합 비율을 달성할 수 있음을 보여주었다. 이를 통해서 약물이 변형항체에 위치 특이적으로 접합하는 동시에 모항체의 특성은 유지되면서도, 매우 높은 표적 특이성과 약물효과를 기대할 수 있음을 보여주었다. 그러나 합성 약물이나 단클론 항체 치료제와 비교할 때 현저히 높은 생산비는 약물의 접합 수율 향상을 통한 경제적 효용성의 확대를 절실히 필요로 하고 있다.To this end, the inventors of the present application in Korea Patent No. 1541764 introduced a peptide containing a metal ion binding motif at the C-terminus of the antibody can be drug-specifically conjugated, the constant antibody to drug conjugation ratio Showed that it can achieve. This demonstrates that the drug can be site-specifically conjugated to the modified antibody and at the same time expect high target specificity and drug effect while maintaining the parental antibody characteristics. However, the significantly higher production cost compared to synthetic drugs or monoclonal antibody therapeutics is urgently needed to expand the economic efficiency through improved drug yield.
이에, 본 발명에서는 금속이온 결합 모티프가 포함된 펩타이드 모티프를 포함하는 변형항체를 개량, 즉 모항체의 말단에 도입된 금속이온 결합 모티프의 배열과 1차 구조에 따라서 항체에 접합되는 약물의 접합 비율이 현저히 증가할 수 있음을 증명하고자 하였다.Accordingly, in the present invention, the modified antibody comprising a peptide motif containing a metal ion binding motif is improved, that is, the ratio of conjugation of the drug conjugated to the antibody according to the arrangement and primary structure of the metal ion binding motif introduced into the terminal of the parent antibody. We tried to prove that this could increase significantly.
일 관점에서, 본 발명은 하기 구조식 (1)로 표시되는 모티프를 항체의 말단에 포함하는 변형항체가 링커를 통해 약물에 결합된 항체-약물 접합체에 관한 것이다.In one aspect, the present invention relates to an antibody-drug conjugate wherein a modified antibody comprising a motif represented by the following structural formula (1) at the terminal of the antibody is bound to a drug via a linker.
구조식 (1)Structural Formula (1)
Xa-[Mmotif1]n1-Xb-[Mmotif2]n2 X a - [M motif1] n1 -X b - [M motif2] n2
상기 식에서, Mmotif1 또는 Mmotif2는 각각 독립적으로 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나를 포함하고, 상기 서열번호 3 또는 4에서 X는 시스테인 이외의 아미노산 잔기를 포함하며, Wherein M motif1 or M motif2 each independently include any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4), wherein X in SEQ ID NO: 3 or 4 includes amino acid residues other than cysteine,
Xa 및 Xb는 각각 독립적으로 A (alanine), S (serine), G (glycine)로 이루어진 군에서 선택된 아미노산 잔기가 0개 내지 20개로 구성된 펩타이드이며,X a and X b are each independently a peptide consisting of 0 to 20 amino acid residues selected from the group consisting of A (alanine), S (serine) and G (glycine),
n1 및 n2는 각각 1 내지 10의 정수이다. n1 and n2 are integers of 1-10, respectively.
상기 구조식 (1)로 표시되는 모티프는 금속이온 결합 모티프인 CGH 모티프를 포함하는 펩타이드로, CGH 모티프는 아래의 화학식 1의 구조를 가진다. The motif represented by Structural Formula (1) is a peptide including a CGH motif which is a metal ion binding motif, and the CGH motif has the structure of Formula 1 below.
Figure PCTKR2017003508-appb-C000001
Figure PCTKR2017003508-appb-C000001
상기 화학식 1에서, M은 금속이온을 의미하며, R은 시스테인을 제외한 아미노산 잔기, 특히 알라닌이 바람직하다. In Chemical Formula 1, M means a metal ion, and R is an amino acid residue except cysteine, particularly alanine is preferable.
본 발명에 따른 모티프에서 Mmotif1 또는 Mmotif2는 각각 C 말단과 N 말단에 알라닌이 위치하는 ACGHA (서열번호 1) 또는 이 중 시스테인을 시스테인 이외의 아미노산 잔기로 치환한 AXGHA (서열번호 3)를 포함한다. N-말단과 C-말단이 바뀌어도 여전히 금속 이온 결합 특성을 가지므로, 본 발명에 따른 모티프에서 Mmotif1 또는 Mmotif2에 ACGHA (서열번호 1) 또는 AXGHA (서열번호 3)에서 N-말단과 C-말단이 변경된 AHGCA (서열번호 2) 또는 AHGXA (서열번호 4)도 포함된다.In the motif according to the present invention, M motif1 or M motif2 includes ACGHA (SEQ ID NO: 1) in which alanine is located at the C-terminal and N-terminal, respectively, or AXGHA (SEQ ID NO: 3) in which a cysteine is substituted with an amino acid residue other than cysteine. do. N-terminus and C-terminus in ACGHA (SEQ ID NO: 1) or AXGHA (SEQ ID NO: 3) for M motif1 or M motif2 in the motif according to the present invention, even if the N-terminus and C-terminus are still changed. Also included are AHGCA (SEQ ID NO: 2) or AHGXA (SEQ ID NO: 4), which have been terminated.
본 발명에 따른 모티프에서 Mmotif1 또는 Mmotif2는 각각 동일 서열을 포함할 수 있고, 서로 상이한 서열을 포함할 수도 있다. In the motif according to the present invention, M motif1 or M motif2 may each include the same sequence and may include different sequences from each other.
본 발명에 따른 일 실시예에서, 상기 Mmotif1 또는 Mmotif2가 AXGHA 또는 AHGXA에 해당하는 경우, 이 때 X는 세린 (S), 알라닌 (A), 트레오닌 (T), 타이로신 (Y), 아스파르트산(D), 라이신 (K) 및 페닐알라닌 (F)으로 구성된 군에서 선택된 아미노산 잔기일 수 있다. In one embodiment according to the invention, when M motif 1 or M motif 2 corresponds to AXGHA or AHGXA, wherein X is serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid It may be an amino acid residue selected from the group consisting of (D), lysine (K) and phenylalanine (F).
본 발명에 따른 일 실시예에서, 상기 Mmotif2가 AXGHA (서열번호 3)을 포함하고, X는 시스테인 이외의 아미노산 잔기인 경우 ACGHA를 포함하는 경우에 비해 높은 약물 접합능을 보일 수 있음을 확인하였다. 상기 Mmotif2는 AXGHA, 이 때 X는 시스테인 이외의 아미노산 잔기 예를 들어, 세린 (S), 알라닌 (A), 트레오닌 (T), 타이로신 (Y), 아스파르트산(D), 라이신 (K) 및 페닐알라닌 (F)으로 구성된 군에서 선택된 아미노산 잔기일 수 있다.In one embodiment according to the present invention, it was confirmed that the M motif2 includes AXGHA (SEQ ID NO: 3), and X may show higher drug conjugation ability than the case of including ACGHA when amino acid residues other than cysteine are included. . The M motif2 is AXGHA, where X is, for the amino acid residue Examples of the non-cysteine, serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and It may be an amino acid residue selected from the group consisting of phenylalanine (F).
Xa는 모티프 Mmotif1의 5’ 말단에 존재하는 아미노산 잔기로, 경우에 따라서 항체의 말단과 연결하기 위해 위치하는 펩타이드일 수 있으며, A (alanine), S (serine), G (glycine)로 이루어진 군에서 선택된 아미노산 잔기가 0개 내지 20개로 구성된 펩타이드이다. Xa의 아미노산 잔기가 0인 경우 모티프는 Xa를 포함하지 않으며, 항체에 모티프의 Mmotif1이 직접 결합하는 형태일 수 있다. Xa의 아미노산 잔기 1개 이상, 2개 이상, 3개 이상, 4개 이상, 5개 이상일 수 있고, 예를 들어, 2-20개, 2-18개, 2-16개, 2-14개, 2-12개, 2-10개, 2-8개, 2-6개, 2개, 3개, 4개, 5개, 6개, 7개, 8개, 9개, 10개일 수 있다. X a is an amino acid residue present at the 5 'end of the motif M motif1 , which may be a peptide positioned to connect with the terminal of the antibody, and may be composed of A (alanine), S (serine), and G (glycine). The amino acid residue selected from the group is a peptide consisting of 0-20. When the amino acid residue of X a is 0, the motif does not include X a and may be in a form in which M motif1 of the motif directly bonds to the antibody. May be one or more, two or more, three or more, four or more, five or more amino acid residues of X a , for example, 2-20, 2-18, 2-16, 2-14 , 2-12, 2-10, 2-8, 2-6, 2, 3, 4, 5, 6, 7, 8, 9, 10.
Xb는 Mmotif1 및 Mmotif2를 연결하기 위한 링커로 A (alanine), S (serine), G (glycine)로 이루어진 군에서 선택된 아미노산 잔기가 0개 내지 20개로 구성된 펩타이드이다. Xb의 아미노산 잔기가 0인 경우 모티프는 Xb를 포함하지 않으며, Mmotif1 및 Mmotif2이 직접 연결되는 형태일 수 있다. Xb의 아미노산 잔기 1개 이상, 2개 이상, 3개 이상, 4개 이상, 5개 이상일 수 있고, 예를 들어, 2-20개, 2-18개, 2-16개, 2-14개, 2-12개, 2-10개, 2-8개, 2-6개, 2개, 3개, 4개, 5개, 6개, 7개, 8개, 9개, 10개일 수 있다. X b is a linker for linking M motif1 and M motif2 and is a peptide consisting of 0 to 20 amino acid residues selected from the group consisting of A (alanine), S (serine) and G (glycine). If the amino acid residue of X b is 0, the motif does not include X b and may be in a form in which M motif 1 and M motif 2 are directly connected. X b of the amino acid residues in one or more, two or more, three or more, four or more, may be more than five, e.g., 2-20, 2-18, 2-16, 2-14 , 2-12, 2-10, 2-8, 2-6, 2, 3, 4, 5, 6, 7, 8, 9, 10.
n1 및 n2는 각각 Mmotif1 및 Mmotif2의 반복 횟수를 나타내며, 1 내지 10의 정수이다. 상기 n1 및 n2는 각각 1일 수 있으며, 이 때 Mmotif1 및 Mmotif2로 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나가 Xb의 링커 없이 또는 Xb의 링커를 포함하여 연결될 수 있다. 예를 들어, [Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 ACGHAACGHA (서열번호 5), ACGHAAHGCA (서열번호 6), ACGHAAXGHA (서열번호 7), ACGHAAHGXA (서열번호 8), AHGCAAHGCA (서열번호 9), AHGCAACGHA (서열번호 10), AHGCAAXGHA (서열번호 11), AHGCAAHGXA (서열번호 12), AXGHAAXGHA (서열번호 13), AXGHAACGHA (서열번호 14), AXGHAAHGCA (서열번호 15), AXGHAAHGXA (서열번호 16), AHGXAAHGXA (서열번호 17), AHGXAACGHA (서열번호 18), AHGXAAHGCA (서열번호 19), 또는 AHGXAAXGHA (서열번호 20)일 수 있으며, 약물의 결합을 위해 C(시스테인)을 필수적으로 포함하는 모티프가 Mmotif1 및 Mmotif2로 선별될 수 있다.n1 and n2 represent the number of repetitions of M motif1 and M motif2 , respectively, and are an integer of 1 to 10. N1 and n2 may each be 1, wherein M motif1 And M motif2 to ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3) and AHGXA (SEQ ID NO: 4) any of the sequences consisting of comprises a linker for X b without a linker or X b Can be connected. For example, [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker X b ACGHAACGHA (SEQ ID NO: 5), ACGHAAHGCA (SEQ ID NO: 6), ACGHAAXGHA (SEQ ID NO: 7), ACGHAAHGXA ( SEQ ID NO: 8), AHGCAAHGCA (SEQ ID NO: 9), AHGCAACGHA (SEQ ID NO: 10), AHGCAAXGHA (SEQ ID NO: 11), AHGCAAHGXA (SEQ ID NO: 12), AXGHAAXGHA (SEQ ID NO: 13), AXGHAACGHA (SEQ ID NO: 14), AXGHAAHGCA Number 15), AXGHAAHGXA (SEQ ID NO: 16), AHGXAAHGXA (SEQ ID NO: 17), AHGXAACGHA (SEQ ID NO: 18), AHGXAAHGCA (SEQ ID NO: 19), or AHGXAAXGHA (SEQ ID NO: 20), and C ( Motifs (essentially cysteine) may be selected as M motif1 and M motif2 .
Xb가 존재하는 경우 위 아미노산 서열들에서 5’ 말단에서 6번째 위치에 아미노산 잔기가 1개 내지 20개로 구성된 펩타이드가 추가로 포함될 수 있다. 이 때, X는 세린 (S), 알라닌 (A), 트레오닌 (T), 타이로신 (Y), 아스파르트산(D), 라이신 (K) 및 페닐알라닌 (F)으로 구성된 군에서 선택될 수 있다. When X b is present, a peptide consisting of 1 to 20 amino acid residues at the 5 'end of the amino acid sequences may be further included. In this case, X may be selected from the group consisting of serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and phenylalanine (F).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 세린이면 X를 포함하는 서열번호 7, 8,11 내지 20에서 X 위치에 세린을 포함하여 ACGHAASGHA (서열번호 21), ACGHAAHGSA (서열번호 22), AHGCAASGHA (서열번호 23), AHGCAAHGSA (서열번호 24), ASGHAASGHA (서열번호 25), ASGHAACGHA (서열번호 26), ASGHAAHGCA (서열번호 27), ASGHAAHGSA (서열번호 28), AHGSAAHGSA (서열번호 29), AHGSAACGHA (서열번호 30), AHGSAAHGCA (서열번호 31), 또는 AHGSAASGHA (서열번호 32)일 수 있다. [M motif1 ] n1 -X b- [M motif2 ] The n2 structure contains a serine at position X in SEQ ID NOs 7, 8,11 to 20, including X if there is no linker of X b , for example if X is serine ACGHAASGHA (SEQ ID NO: 21), ACGHAAHGSA (SEQ ID NO: 22), AHGCAASGHA (SEQ ID NO: 23), AHGCAAHGSA (SEQ ID NO: 24), ASGHAASGHA (SEQ ID NO: 25), ASGHAACGHA (SEQ ID NO: 26), ASGHAAHGCA (SEQ ID NO: 27), ASGHAAHGSA (SEQ ID NO: 28), AHGSAAHGSA (SEQ ID NO: 29), AHGSAACGHA (SEQ ID NO: 30), AHGSAAHGCA (SEQ ID NO: 31), or AHGSAASGHA (SEQ ID NO: 32).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 알라닌 (A)이면, ACGHAAAGHA (서열번호 33), ACGHAAHGAA (서열번호 34), AHGCAAAGHA (서열번호 35), AHGCAAHGAA (서열번호 36), AAGHAAAGHA (서열번호 37), AAGHAACGHA (서열번호 38), AAGHAAHGCA (서열번호 39), AAGHAAHGAA (서열번호 40), AHGAAAHGAA (서열번호 41), AHGAAACGHA (서열번호 42), AHGAAAHGCA (서열번호 43), 또는 AHGAAAAGHA (서열번호 44)일 수 있다.[M motif1 ] n1 -X b- [M motif2 ] The n2 structure is ACGHAAAGHA (SEQ ID NO: 33), ACGHAAHGAA (SEQ ID NO: 34), AHGCAAAGHA (if X is alanine (A) when there is no linker of X b ) SEQ ID NO: 35), AHGCAAHGAA (SEQ ID NO: 36), AAGHAAAGHA (SEQ ID NO: 37), AAGHAACGHA (SEQ ID NO: 38), AAGHAAHGCA (SEQ ID NO: 39), AAGHAAHGAA (SEQ ID NO: 40), AHGAAAHGAA (SEQ ID NO: 41), AHGAAACGHA (SEQ ID NO: 41) Number 42), AHGAAAHGCA (SEQ ID NO: 43), or AHGAAAAGHA (SEQ ID NO: 44).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 트레오닌 (T)이면, ACGHAATGHA (서열번호 45), ACGHAAHGTA (서열번호 46), AHGCAATGHA (서열번호 47), AHGCAAHGTA (서열번호 48), ATGHAATGHA (서열번호 49), ATGHAACGHA (서열번호 50), ATGHAAHGCA (서열번호 51), ATGHAAHGTA (서열번호 52), AHGTAAHGTA (서열번호 53), AHGTAACGHA (서열번호 54), AHGTAAHGCA (서열번호 55), 또는 AHGTAATGHA (서열번호 56)일 수 있다.[M motif1 ] n1 -X b- [M motif2 ] The n2 structure is ACGHAATGHA (SEQ ID NO: 45), ACGHAAHGTA (SEQ ID NO: 46), AHGCAATGHA (if X is a threonine (T) when there is no linker of X b ) SEQ ID NO: 47), AHGCAAHGTA (SEQ ID NO: 48), ATGHAATGHA (SEQ ID NO: 49), ATGHAACGHA (SEQ ID NO: 50), ATGHAAHGCA (SEQ ID NO: 51), ATGHAAHGTA (SEQ ID NO: 52), AHGTAAHGTA (SEQ ID NO: 53), AHGTAACGHA (SEQ ID NO: 53) Number 54), AHGTAAHGCA (SEQ ID NO: 55), or AHGTAATGHA (SEQ ID NO: 56).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 타이로신 (Y)이면 ACGHAAYGHA (서열번호 57), ACGHAAHGYA (서열번호 58), AHGCAAYGHA (서열번호 59), AHGCAAHGYA (서열번호 60), AYGHAAYGHA (서열번호 61), AYGHAACGHA (서열번호 62), AYGHAAHGCA (서열번호 63), AYGHAAHGYA (서열번호 64), AHGYAAHGYA (서열번호 65), AHGYAACGHA (서열번호 66), AHGYAAHGCA (서열번호 67), 또는 AHGYAAYGHA (서열번호 68)일 수 있다.[M motif1 ] n1 -X b- [M motif2 ] The n2 structure is the absence of a linker of X b , for example, if X is tyrosine (Y), ACGHAAYGHA (SEQ ID NO: 57), ACGHAAHGYA (SEQ ID NO: 58), AHGCAAYGHA (SEQ ID NO: Number 59), AHGCAAHGYA (SEQ ID NO: 60), AYGHAAYGHA (SEQ ID NO: 61), AYGHAACGHA (SEQ ID NO: 62), AYGHAAHGCA (SEQ ID NO: 63), AYGHAAHGYA (SEQ ID NO: 64), AHGYAAHGYA (SEQ ID NO: 65), AHGYAACGHA (SEQ ID NO: 65) 66), AHGYAAHGCA (SEQ ID NO: 67), or AHGYAAYGHA (SEQ ID NO: 68).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 아스파르트산(D)이면 ACGHAADGHA (서열번호 69), ACGHAAHGDA (서열번호 70), AHGCAADGHA (서열번호 71), AHGCAAHGDA (서열번호 72), ADGHAADGHA (서열번호 73), ADGHAACGHA (서열번호 74), ADGHAAHGCA (서열번호 75), ADGHAAHGDA (서열번호 76), AHGDAAHGDA (서열번호 77), AHGDAACGHA (서열번호 78), AHGDAAHGCA (서열번호 79), 또는 AHGDAADGHA (서열번호 80)일 수 있다.[M motif1 ] n1 -X b- [M motif2 ] The n2 structure is the absence of a linker of X b , for example, if X is aspartic acid (D), ACGHAADGHA (SEQ ID NO: 69), ACGHAAHGDA (SEQ ID NO: 70), AHGCAADGHA ( SEQ ID NO: 71), AHGCAAHGDA (SEQ ID NO: 72), ADGHAADGHA (SEQ ID NO: 73), ADGHAACGHA (SEQ ID NO: 74), ADGHAAHGCA (SEQ ID NO: 75), ADGHAAHGDA (SEQ ID NO: 76), AHGDAAHGDA (SEQ ID NO: 77), AHGDAACGHA (SEQ ID NO: 77) Number 78), AHGDAAHGCA (SEQ ID NO: 79), or AHGDAADGHA (SEQ ID NO: 80).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 라이신 (K)이면 ACGHAAKGHA (서열번호 81), ACGHAAHGKA (서열번호 82), AHGCAAKGHA (서열번호 83), AHGCAAHGKA (서열번호 84), AKGHAAKGHA (서열번호 85), AKGHAACGHA (서열번호 86), AKGHAAHGCA (서열번호 87), AKGHAAHGKA (서열번호 88), AHGKAAHGKA (서열번호 89), AHGKAACGHA (서열번호 90), AHGKAAHGKA (서열번호 91), 또는 AHGKAAKGHA (서열번호 92)일 수 있다. [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker of X b, for example when X is lysine (K) ACGHAAKGHA (SEQ ID NO: 81), ACGHAAHGKA (SEQ ID NO: 82), AHGCAAKGHA (SEQ ID NO: Number 83), AHGCAAHGKA (SEQ ID NO: 84), AKGHAAKGHA (SEQ ID NO: 85), AKGHAACGHA (SEQ ID NO: 86), AKGHAAHGCA (SEQ ID NO: 87), AKGHAAHGKA (SEQ ID NO: 88), AHGKAAHGKA (SEQ ID NO: 89), AHGKAACGHA (SEQ ID NO: 90), AHGKAAHGKA (SEQ ID NO: 91), or AHGKAAKGHA (SEQ ID NO: 92).
[Mmotif1]n1-Xb-[Mmotif2]n2 구조는 Xb의 링커가 없는 경우 예를 들어 X가 페닐알라닌 (F)이면 ACGHAAFGHA (서열번호 93), ACGHAAHGFA (서열번호 94), AHGCAAFGHA (서열번호 95), AHGCAAHGFA (서열번호 96), AFGHAAFGHA (서열번호 97), AFGHAACGHA (서열번호 98), AFGHAAHGCA (서열번호 99), AFGHAAHGFA (서열번호 100), AHGFAAHGFA (서열번호 101), AHGFAACGHA (서열번호 102), AHGFAAHGFA (서열번호 103), 또는 AHGFAAFGHA (서열번호 104)일 수 있다. [M motif1] n1 -X b - [M motif2] n2 structure is the absence of a linker X b For example, if X is phenylalanine (F) ACGHAAFGHA (SEQ ID NO: 93), ACGHAAHGFA (SEQ ID NO: 94), AHGCAAFGHA (SEQ ID NO: Number 95), AHGCAAHGFA (SEQ ID NO: 96), AFGHAAFGHA (SEQ ID NO: 97), AFGHAACGHA (SEQ ID NO: 98), AFGHAAHGCA (SEQ ID NO: 99), AFGHAAHGFA (SEQ ID NO: 100), AHGFAAHGFA (SEQ ID NO: 101), AHGFAACGHA (SEQ ID NO: 102), AHGFAAHGFA (SEQ ID NO: 103), or AHGFAAFGHA (SEQ ID NO: 104).
Xb가 존재하는 경우 위 서열번호 5 내지 104의 아미노산 서열들에서 5’ 말단에서 6번째 위치에 아미노산 잔기가 1개 내지 20개로 구성된 펩타이드가 추가로 포함될 수 있다. 이 때, X는 세린 (S), 알라닌 (A), 트레오닌 (T), 타이로신 (Y), 아스파르트산(D), 라이신 (K) 및 페닐알라닌 (F)으로 구성된 군에서 선택될 수 있다. When X b is present, a peptide consisting of 1 to 20 amino acid residues may be further included at the 6th position at the 5 'end in the amino acid sequences of SEQ ID NOs: 5 to 104. In this case, X may be selected from the group consisting of serine (S), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) and phenylalanine (F).
상기 n1 및 n2는 각각 2 이상일 수 있으며, 예를 들어 상기 n1 및 n2가 2인 경우 Mmotif1 및 Mmotif2 각각이 동일한 아미노산 서열을 포함한다면 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나가 각각 2회씩 반복된 서열이 포함되거나, 또는 상이한 서열 예를 들어 ACGHA 및 AHGCA, ACGHA 및 AXGHA, ACGHA 및 AHGXA, AHGCA 및 ACGHA, AHGCA 및 AXGHA, AHGCA 및 AHGXA, AXGHA 및 ACGHA, AXGHA 및 AHGCA, AXGHA 및 AHGXA, AHGXA 및 ACGHA, AHGXA 및 AHGCA, 또는 AHGXA 및 AXGHA이 각각 2회씩 반복된 서열이 포함될 수 있다. 상기 n1 및 n2이 3-10인 경우는 Mmotif1 및 Mmotif2 각각이 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나에 해당하는 동일 서열을 포함하거나, 상이한 서열을 포함하여 각각 3-10회씩 반복될 수 있다.N1 and n2 may each be two or more, for example, when n1 and n2 are 2, AC MHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), and AXGHA if M motif 1 and M motif 2 each include the same amino acid sequence Either one consisting of (SEQ ID NO: 3) and AHGXA (SEQ ID NO: 4) includes a sequence in which each is repeated twice, or a different sequence such as ACGHA and AHGCA, ACGHA and AXGHA, ACGHA and AHGXA, AHGCA and ACGHA , AHGCA and AXGHA, AHGCA and AHGXA, AXGHA and ACGHA, AXGHA and AHGCA, AXGHA and AHGXA, AHGXA and ACGHA, AHGXA and AHGCA, or AHGXA and AXGHA, respectively. When n1 and n2 are 3-10, M motif1 and M motif2 are each any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4). It may include the same sequence corresponding to one, or may be repeated 3-10 times each including a different sequence.
바람직하게, 상기 n1 및 n2는 각각 1일 수 있으며, Xb의 링커가 존재하지 않을 수 있으며, 이 때 본 발명에 따른 모티프는 예를 들어 서열번호 5 내지 104로 구성된 군에서 선택된 하나 이상의 서열을 포함할 수 있다. Preferably, n1 and n2 may each be 1, and there may not be a linker of X b , wherein the motif according to the present invention is one or more sequences selected from the group consisting of SEQ ID NOs: 5 to 104, for example. It may include.
상기 모티프는 항체의 중쇄 또는 경쇄 C-말단, 구체적으로 중쇄 C-말단에 결합할 수 있으며, 이를 통해 약물 접합 수율이 현저히 증가한 변형항체 및 이를 포함하는 약물 접합체를 제공할 수 있다. 상기 약물의 접합 수율을 높임으로 해서 항체-약물 접합체의 생산 수율을 높일 수 있다. 높은 수율로 접합된 약물은 변형 항체에 의해서 타겟 암세포에 특이적으로 전달됨으로써 치료효과를 높일 수 있으며, 높은 항체-약물 접합체의 접합 수율로 인하여, 항체-약물 접합체 치료제의 생산 비용을 낮출 수 있다.The motif may bind to the heavy or light chain C-terminus of the antibody, specifically the heavy chain C-terminus, thereby providing a modified antibody and a drug conjugate comprising the same, which significantly increase the drug conjugation yield. The yield of the antibody-drug conjugate can be increased by increasing the yield of the drug. Drugs conjugated in high yield can be specifically delivered to target cancer cells by modified antibodies, thereby increasing the therapeutic effect, and lowering the production cost of the antibody-drug conjugate therapeutics due to the high yield of conjugated antibody-drug conjugates.
상기 모티프는 모항체와 아미노 결합을 통한 융합(fusion) 형태로 직접 결합되거나, 모항체의 말단 작용기와 모티프 중 말단 작용기가 화학적으로 결합하는 형태이거나, 모티프 중 말단 작용기와 약물을 연결하는 링커에 매개된(linker-mediated) 형태의 결합도 가능하다. The motif may be directly bonded in the form of a fusion through an amino bond with the parent antibody, or in a form in which the terminal functional group of the parent antibody and the terminal functional group in the motif are chemically bonded, or mediated to a linker connecting the terminal functional group and the drug in the motif. Linker-mediated forms of coupling are also possible.
상기 링커는 모티프 내의 특정 잔기와 약물을 연결하는 형태일 수 있으며, 변형항체 중 모티프 상에 존재하는 친핵성 잔기 (예를 들어, 시스테인)에 반응하는 친전자성기를 갖는 반응성 부위를 가질 수 있다. 상기 링커는 예를 들어, 모티프에 결합하는 반응성 기능기, 아미노산 및 자가 절단 스페이서를 포함할 수 있다. The linker may be in the form of linking a drug with a specific residue in a motif, and may have a reactive site having an electrophilic group that reacts to a nucleophilic residue (eg, cysteine) present on a motif among modified antibodies. The linker may comprise, for example, reactive functional groups, amino acids and self-cutting spacers that bind to the motif.
상기 기능기는 i) 말레이마이드기, 아세트아마이드기, 또는 이의 유도체, ii) 아지리딘기, 아릴할라이드, 아크릴로일기, 또는 이의 유도체, iii) 알킬화 반응기, 아릴화 반응기, 피리딜 다이설파이드, 티오니트로벤조익산, 또는 이의 유도체일 수 있다. 구체적으로, 상기 링커는 구체적 예로 i) 말레이마이드기 또는 이의 유도체-발린-시트룰린(valine-citurulline)-파라 아닐린 벤조산(para-aniline benzoic acid: PABA); 또는 ii) 아세트아마이드기 또는 이의 유도체-발린-시트룰린(valine-citurulline)-파라 아닐린 벤조산(para-aniline benzoic acid: PABA)의 형태일 수 있으나, 이에 제한되는 것은 아니다. The functional groups are i) maleimide groups, acetamide groups, or derivatives thereof, ii) aziridine groups, arylhalides, acryloyl groups, or derivatives thereof, iii) alkylation reactors, arylation reactors, pyridyl disulfides, thionitrobenzo Ikic acid, or a derivative thereof. Specifically, the linker may specifically include, for example, i) a maleimide group or a derivative thereof-valine-citurulline-para-aniline benzoic acid (PABA); Or ii) an acetamide group or derivative thereof-valine-citurulline-paraaniline benzoic acid (PABA), but is not limited thereto.
상기 링커를 통한 잔기와 약물의 결합은 공지의 방법, 예를 들어 알킬레이션, 이황화(disulfide) 상호교환 방법 및 트랜스티오에스테르화 반응법이 이용될 수 있다. 이를 통해 모티프 내 시스테인 잔기의 티올기를 통해 항체와 약물이 접합될 수 있다. The binding of the moiety to the drug via the linker may be carried out using known methods such as alkylation, disulfide interchange and transthioesterification. This allows the antibody and the drug to be conjugated via the thiol group of the cysteine residue in the motif.
하나의 구체예에서, 티올-링커 결합을 위해서 사용되는 말레이미드 기의 경우, 시스테인 잔기의 티올이 말레이미드 기에 대하여 갖는 친핵 반응성이 단백질 중에 존재하는 다른 아미노산 기능기, 예를 들어 리신 잔기의 아미노기 또는 N-말단 아미노기에 비하여 약 1,000배 더 높기 때문에 시스테인에 특이적으로 결합시키는데 활용될 수 있다. 따라서, 말레이미드기, 이의 유도체 또는 아세트아마이드기 또는 이의 유도체, 예를 들어 브로모 아세트아마이드기, 이오도 아세트아마이드기를 통한 변형항체-약물 접합체는 시스테인이 티오에테르 결합으로 약물과 결합됨을 알 수 있다.In one embodiment, for maleimide groups used for thiol-linker linkages, the nucleophilic reactivity of the thiol of the cysteine residue to the maleimide group is present in the protein, such as other amino acid functional groups, such as amino groups of lysine residues or Because it is about 1,000 times higher than the N-terminal amino group, it can be used to specifically bind to cysteine. Thus, the modified antibody-drug conjugates via maleimide groups, derivatives thereof or acetamide groups or derivatives thereof, such as bromo acetamide groups, iodo acetamide groups, can be seen that cysteine is bound to the drug by thioether linkage. .
상기 항체는 모노클로날 항체, 이중특이적 항체, 키메릭 항체, 인간 항체 및 인간화 항체로 구성된 군에서 선택된 하나 이상일 수 있다. 또한 이중특이적 항체 (bispecific antibody) 등의 변형 항체나, 항체 단편 등도 모두 사용 가능하다. '항체 단편'은 적어도 항원에 대한 결합 기능을 보유하고 있는 단편을 뜻하며, 단쇄 항체, 디아바디, 트리아바디, 테트라바디, Fab 단편, F(ab')2 단편, Fd, scFv, 도메인 항체, 미니바디, 스캡(single chain antibody, scAb), 항체 불변영역의 유도체들, 단백질 스캐폴드 (protein scaffolds)에 기초한 인공항체 등을 포함한다.The antibody may be one or more selected from the group consisting of monoclonal antibodies, bispecific antibodies, chimeric antibodies, human antibodies and humanized antibodies. Moreover, both modified antibodies, such as a bispecific antibody, antibody fragments, etc. can be used. 'Antibody fragments' refer to fragments that have at least the binding function to the antigen, and are single-chain antibodies, diabodies, triabodies, tetrabodies, Fab fragments, F (ab') 2 fragments, Fd, scFv, domain antibodies, mini Body, single chain antibody (scAb), derivatives of antibody constant regions, phosphorus based on protein scaffolds and the like.
경우에 따라서, 상기 항체는 IgA, IgD, IgE, IgG 및 IgM으로 구성된 군에서 선택될 수 있다. In some cases, the antibody may be selected from the group consisting of IgA, IgD, IgE, IgG and IgM.
상기 항체는 구체적으로, 암 특이 항원, 세포 표면 수용체 단백질, 세포 표면 단백질, 막횡단 단백질, 신호전달 단백질, 세포생존 조절인자, 세포 증식 조절인자, 조직 발달 또는 분화와 연관된 분자, 림포카인, 사이토카인, 세포 주기 조절에 관련된 분자, 혈관형성에 관련된 분자, 또는 혈관신생에 관련된 분자에 대한 결합능과 특이성을 가질 수 있으며, 예를 들어, (1) BMPR1B (골 형태형성 단백질 수용체-IB형, 진뱅크 승인 번호 NM_001203);The antibody specifically includes cancer specific antigens, cell surface receptor proteins, cell surface proteins, transmembrane proteins, signaling proteins, cell survival regulators, cell proliferation regulators, molecules associated with tissue development or differentiation, lymphokines, cytokines. May have binding capacity and specificity for Cain, a molecule involved in cell cycle regulation, an angiogenesis related molecule, or an angiogenesis related molecule, for example, (1) BMPR1B (bone morphogenetic protein receptor-IB type, gene Bank grant number NM_001203);
(2) E16 (LAT1, SLC7A5, 진뱅크 승인 번호 NM_003486);(2) E16 (LAT1, SLC7A5, GenBank Accession No. NM_003486);
(3) STEAP1 (전립선의 6회의 막횡단 상피 항원, 진뱅크 승인 번호 NM_012449);(3) STEAP1 (six transmembrane epithelial antigens of the prostate, Genbank Accession No. NM_012449);
(4) 0772P (CA125, MUC16, 진뱅크 승인 번호 AF361486);(4) 0772P (CA125, MUC16, GenBank Accession No. AF361486);
(5) MPF (MPF, MSLN, SMR, 거핵세포 강화 인자, 메소텔린, 진뱅크 승인 번호 NM_005823);(5) MPF (MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Accession No. NM — 005823);
(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, 용질 운반체 족 34 (인산나트륨), 구성원 2, 제II형 나트륨-의존성 포스페이트 수송체 3b, 진뱅크 승인 번호 NM_006424);(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, Solute Carrier Family 34 (Sodium Phosphate), Member 2, Type II Sodium-Dependent Phosphate Transporter 3b, GenBank Accession No. NM_006424);
(7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, 세마포린 5b Hlog, 세마 도메인, 7개의 트롬보스폰딘 반복체 (제1형 및 유사 제1형), 막횡단 도메인 (TM) 및 짧은 세포질 도메인, (세마포린) 5B, 진뱅크 승인 번호 AB040878);(7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, Sema Domain, 7 Thrombospondin Repeats (Type 1 and Similar Type 1), Transmembrane Domain (TM) And short cytoplasmic domain, (semaphorin) 5B, Genbank Accession No. AB040878);
(8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 유전자, 진뱅크 승인 번호 AY358628);(8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genebank Accession No. AY358628);
(9) ETBR (엔도텔린 B형 수용체, 진뱅크 승인 번호 AY275463);(9) ETBR (endothelin type B receptor, Genbank Accession No. AY275463);
(10) MSG783 (RNF124, 가상 단백질 FLJ20315, 진뱅크 승인 번호 NM_017763);(10) MSG783 (RNF124, hypothetical protein FLJ20315, Genebank Accession No. NM_017763);
(11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, 전립선암 관련 유전자 1, 전립선암 관련단백질 1, 전립선의 6회의 막횡단 상피 항원 2, 6회의 막횡단 전립선 단백질, 진뱅크 승인 번호 AF455138);(11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, Prostate Cancer Related Gene 1, Prostate Cancer Related Protein 1, Prostate 6 Transmembrane Epithelial Antigen 2, 6 Transmembrane Prostate Protein, Genebank Authorization number AF455138);
(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, 일시적 수용체 잠재적 양이온 채널, M 아족, 구성원 4, 진뱅크 승인 번호 NM_017636);(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subgroup M, member 4, Genbank Accession No. NM_017636);
(13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, 기형암종-유래 성장인자, 진뱅크 승인 번호 NP_003203 또는 NM_003212);(13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, teratocarcinoma-derived growth factor, Genbank accession no. NP — 003203 or NM — 003212);
(14) CD21 (CR2 (보체 수용체 2) 또는 C3DR (C3d/엡스타인 바르 바이러스 수용체) 또는 Hs.73792 진뱅크 승인 번호 M26004);(14) CD21 (CR2 (complementary receptor 2) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792 Genbank Accession No. M26004);
(15) CD79b (CD79B, CD79β, IGb (이뮤노글로불린-관련 베타), B29, 진뱅크 승인 번호 NM_000626);(15) CD79b (CD79B, CD79β, IGb (immunoglobulin-associated beta), B29, Genbank Accession No. NM — 000626);
(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 도메인 함유 포스파타제 고정 단백질 1a), SPAP1B, SPAP1C, 진뱅크 승인 번호 NM_030764);(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchoring protein 1a), SPAP1B, SPAP1C, GenBank Accession No. NM_030764);
(17) HER2 (진뱅크 승인 번호 M11730)17 HER2 (Genbank approval number M11730)
(18) EGFR, HER3 및 HER4로부터 선택된 ErbB 수용체(18) ErbB receptors selected from EGFR, HER3 and HER4
(19) NCA (진뱅크 승인 번호 M18728);(19) NCA (Genbank Accession No. M18728);
(20) MDP (진뱅크 승인 번호 BC017023);(20) MDP (GenBank Accession No. BC017023);
(21) IL20Rα (진뱅크 승인 번호 AF184971);(21) IL20Rα (GenBank Accession No. AF184971);
(22) 브레비칸 (진뱅크 승인 번호 AF229053);(22) Brevican (Genbank Accession No. AF229053);
(23) EphB2R (진뱅크 승인 번호 NM_004442);(23) EphB2R (GenBank Accession No. NM_004442);
(24) ASLG659 (진뱅크 승인 번호 AX092328);(24) ASLG659 (Genbank Accession No. AX092328);
(25) PSCA (진뱅크 승인 번호 AJ297436);(25) PSCA (Genbank Accession No. AJ297436);
(26) GEDA (진뱅크 승인 번호 AY260763);(26) GEDA (Genbank Accession No. AY260763);
(27) BAFF-R (B 세포 활성화 인자 수용체, BLyS 수용체 3, BR3, NP_443177.1);(27) BAFF-R (B cell activating factor receptor, BLyS receptor 3, BR3, NP_443177.1);
(28) CD22 (B-세포 수용체 CD22-B 이소형, NP-001762.1);(28) CD22 (B-cell receptor CD22-B isotype, NP-001762.1);
(29) CD79a (Ig 베타 (CD79B)와 공유적으로 상호작용하고 IgM 분자와 표면에서 복합체를 형성하는 B 세포 특이적 단백질인 CD79A, CD79α, 이뮤노글로불린-관련 알파는 B 세포 분화에 관여하는 신호를 전달함, 진뱅크 승인 번호 NP_001774.1);(29) CD79a, CD79A, CD79α, and immunoglobulin-associated alpha, which are covalently interacting with CD79a (Ig beta (CD79B) and forming complexes on the surface with IgM molecules, are signals involved in B cell differentiation Forwarded, Genbank approval number NP_001774.1);
(30) CXCR5 (CXCL13 케모킨에 의해 활성화된 G 단백질 커플링된 수용체인 버킷 림프종 수용체 1은 림프구 이동 및 체액성 방어에 작용하고 HIV-2 감염에 참여하며, AIDS, 림프종, 골수종 및 백혈병의 발병과 관련이 있다고 여겨짐, 진뱅크 승인 번호 NP_001707.1);(30) CXCR5 (Bucket Lymphoma Receptor 1, a G protein coupled receptor activated by CXCL13 chemokines, acts on lymphocyte migration and humoral defense, participates in HIV-2 infection, and develops AIDS, lymphoma, myeloma and leukemia Considered to be related to, Genbank approval number NP_001707.1);
(31) HLA-DOB (펩티드에 결합하여 CD4+ T 림프구에 제시하는, MHC 클래스 II 분자 (Ia 항원)의 베타 서브유닛, 진뱅크 승인 번호 NP_002111.1);(31) HLA-DOB (beta subunit of MHC class II molecules (Ia antigen), binding to peptides and presenting in CD4 + T lymphocytes, Genbank Accession No. NP — 002111.1);
(32) P2X5 (세포외 ATP에 의해 게이트되는 이온 채널인, 퓨린성 수용체 P2X 리간드-게이트 이온 채널 5는 시냅스 전달 및 신경발생에 관여할 수 있으며, 이의 결핍은 특발성 배뇨근 불안정의 병태생리에 기여할 수 있음, 진뱅크 승인 번호 NP_002552.2);(32) P2X5 (purine receptor P2X ligand-gate ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, the lack of which may contribute to the pathophysiology of idiopathic detrusor instability Yes, Genbank approval number NP_002552.2);
(33) CD72 (B-세포 분화 항원 CD72, Lyb-2, 진뱅크 승인 번호 NP_001773.1);(33) CD72 (B-cell differentiation antigen CD72, Lyb-2, Genbank Accession No. NP — 001773.1);
(34) LY64 (루이신 풍부 반복체 (LRR) 족의 제I형 막 단백질인, 림프구 항원 64 (RP105)는 B 세포 활성화 및 세포자멸을 조절하며, 이것의 기능 상실은 전신성 홍반성 루푸스 환자의 질병 활성 증가와 관련이 있음, 진뱅크 승인 번호 NP_005573.1);(34) Lymphocyte antigen 64 (RP105), a type I membrane protein of the LY64 (leucine rich repeat (LRR) family), modulates B cell activation and apoptosis, and its loss of function is attributed to systemic lupus erythematosus patients. Associated with increased disease activity, GenBank Accession No. NP_005573.1);
(35) FcRH1 (C2형 Ig-유사 및 ITAM 도메인을 함유하는 이뮤노글로불린 Fc 도메인에 대한 추정적 수용체인 Fc 수용체 유사 단백질 1은 B 림프구 분화에 관여할 수 있음, 진뱅크 승인 번호 NP_443170.1);(35) FcRH1 (Fc receptor-like protein 1, a putative receptor for immunoglobulin Fc domains containing C2 Ig-like and ITAM domains, may be involved in B lymphocyte differentiation, Genbank accession number NP_443170.1) ;
(36) IRTA2 (B 세포 발생 및 림프종발생에 작용할 수 있는 추정적 면역수용체인 이뮤노글로불린 거대족 수용체 전좌 관련 2, 전좌에 의한 상기 유전자 탈조절은 몇몇 B 세포 악성종양에서 일어남, 진뱅크 승인 번호 NP_112571.1); (36) IRTA2 (associated gene deregulation by immunoglobulin macrophage receptor translocation, a putative immunoreceptor capable of acting on B cell development and lymphoma development, occurs in some B cell malignancies, Genbank approval number NP_112571.1);
(37) TENB2 (성장 인자의 EGF/헤레굴린 족 및 폴리스타틴과 관련이 있는 추정적 막횡단 프로테오글리칸, 진뱅크 승인 번호 AF179274);(37) TENB2 (estimated transmembrane proteoglycan associated with EGF / Heregulin family of growth factors and follistatin, Genbank Accession No. AF179274);
(38) MAGE-C1/CT7 (고환암 과발현 단백질);(38) MAGE-C1 / CT7 (testicular cancer overexpressed protein);
(39) androgen receptor, PTEN, human kallikrein-related peptidase 3 (전립선암에서 과발현되는 단백질);(39) androgen receptor, PTEN, human kallikrein-related peptidase 3 (protein overexpressed in prostate cancer);
(40) CD20;(40) CD20;
(41) CD30;(41) CD30;
(42) CD33;(42) CD33;
(43) CD52;(43) CD52;
(44) EpCam;(44) EpCam;
(45) CEA;(45) CEA;
(46) gpA33;(46) gpA33;
(47) Mucins;(47) Mucins;
(48) TAG-72;(48) TAG-72;
(49) Carbonic anhydrase IX;(49) Carbonic anhydrase IX;
(50) PSMA;(50) PSMA;
(51) folate receptor (FOLR gene에 의해서 발현되는 단백질 패밀리. Folic acid와 높은 결합력을 가지고 있으며, 5-methyltetrahydrofolate를 세포 내로 운반한다);(51) folate receptor (a family of proteins expressed by the FoLR gene, which has high binding capacity with Folic acid and carries 5-methyltetrahydrofolate into cells);
(52) gangliosides (GD2, GD3, GM2);(52) gangliosides (GD2, GD3, GM2);
(53) 당수화물 Lewis-Y;(53) carbohydrate Lewis-Y;
(54) VEGF;(54) VEGF;
(55) VEGFR;(55) VEGFR;
(56) aVb3;(56) aVb3;
(57) a5b1;(57) a5b1;
(58) ERB3;(58) ERB3;
(59) c-MET;(59) c-MET;
(60) EphA3;(60) EphA3;
(61) TRAIL-R1, TRAIL-R2;(61) TRAIL-R1, TRAIL-R2;
(62) RANKL;(62) RANKL;
(63) FAP; 및(63) FAP; And
(64) Tenascin로 구성된 군에서 선택된 하나 이상의 타겟에 결합능을 가질 수 있으나, 이에 제한되는 것은 아니다. (64) may have binding capacity to one or more targets selected from the group consisting of tenascin, but is not limited thereto.
본 발명에 따른 일 실시예에서, 서열번호 115의 중쇄 및 서열번호 116의 경쇄를 포함하는 엽산 수용체 (folate receptor)에 결합하는 항체 (Farletuzumab)를 모항체로 이용하였다. 상기 모항체의 중쇄 말단에 금속이온 결합 모티프를 도입하여 펩타이드 모티프의 서열 및 배치에 따라 여러 가지 변형항체를 제조한 후, 변형 항체에 따른 약물의 접합 비율의 차이를 측정하였다. 상기 변형항체는 서열번호 117 내지 121로 구성된 군에서 선택된 하나 이상의 중쇄를 포함할 수 있다.In one embodiment according to the present invention, an antibody (Farletuzumab) that binds to a folate receptor comprising a heavy chain of SEQ ID NO: 115 and a light chain of SEQ ID NO: 116 was used as a parent antibody. By introducing a metal ion binding motif to the heavy chain terminal of the parent antibody to prepare a variety of modified antibodies according to the sequence and placement of the peptide motif, the difference in the conjugate ratio of the drug according to the modified antibody was measured. The modified antibody may include one or more heavy chains selected from the group consisting of SEQ ID NOs: 117 to 121.
또한, 본 발명에 따른 다른 실시예에서, Her2에 특이적으로 결합하는 항체 (Trastuzumab)를 모항체로 이용하였다. 상기 모항체의 중쇄 말단에 금속이온 결합 모티프를 도입하여 펩타이드 모티프의 서열 및 배치에 따라 여러 가지 변형항체를 제조한 후, 변형 항체에 따른 약물의 접합 비율의 차이를 측정하였다. In another embodiment according to the present invention, an antibody (Trastuzumab) that specifically binds to Her2 was used as the parent antibody. By introducing a metal ion binding motif to the heavy chain terminal of the parent antibody to prepare a variety of modified antibodies according to the sequence and placement of the peptide motif, the difference in the conjugate ratio of the drug according to the modified antibody was measured.
그 결과, 본 발명에 따른 항체-약물 접합체에서 Farletuzumab 및 Trastuzumab에 모티프가 도입되었을 때 동등한 정도의 높은 약물 접합 수율을 가짐을 확인하였다. 따라서, 본 발명에 따른 모티프는 항체의 종류와 무관하게 항체-약물 접합체 제조에서 항체에 약물을 결합하기 위한 플랫폼 기술로 활용될 수 있다. As a result, it was confirmed that the antibody-drug conjugate according to the present invention had a similarly high drug conjugation yield when the motifs were introduced into Farletuzumab and Trastuzumab. Therefore, the motif according to the present invention can be utilized as a platform technology for binding a drug to an antibody in antibody-drug conjugate preparation regardless of the type of antibody.
하나의 실시예에서, 상기 항체는 모항체 또는 변형항체의 가변영역 및 IgG2 또는 IgG4의 CH1, CH2 및 CH3를 포함할 수 있다. 예를 들어, Farletuzumab, Trastuzumab 또는 이의 변형항체의 VH와 VL을 쓰고, IgG2 또는 IgG4의 CH1, CH2, CH3를 포함할 수 있다. 예를 들어 상기 Farletuzumab 항체의 가변영역은 서열번호 122의 중쇄 가변영역 및/또는 서열번호 123의 경쇄 가변영역을 포함할 수 있다.In one embodiment, the antibody may comprise the variable region of the parent or modified antibody and CH1, CH2 and CH3 of IgG2 or IgG4. For example, the VH and VL of Farletuzumab, Trastuzumab or its modified antibodies can be used and include CH1, CH2, CH3 of IgG2 or IgG4. For example, the variable region of the Farletuzumab antibody may include a heavy chain variable region of SEQ ID NO: 122 and / or a light chain variable region of SEQ ID NO: 123.
다른 실시예에서, 상기 항체는 모항체 또는 변형항체의 Fab 및 IgG2 또는IgG4의 Fc를 포함할 수 있다. 구체적으로, Farletuzumab, Trastuzumab 또는 이의 변형항체의 Fab 부분과 IgG2 또는 IgG4의 Fc 부분을 퓨전 (fusion)한 형태를 포함할 수 있다. 예를 들어 상기 Farletuzumab 항체의 Fab는 서열번호 122의 중쇄 가변영역 및 CH1 포함 서열 (서열번호 124) 및/또는 서열번호 123의 경쇄 가변영역을 포함할 수 있다. 상기 Trastuzumab 항체는 서열번호 127의 중쇄 및/또는 서열번호 128의 경쇄를 포함할 수 있다.In another embodiment, the antibody may comprise a Fab of the parent antibody or a modified antibody and an Fc of IgG2 or IgG4. Specifically, it may include a form in which the Fab portion of Farletuzumab, Trastuzumab or its modified antibody and the Fc portion of IgG2 or IgG4 are fused. For example, the Fab of the Farletuzumab antibody may comprise a heavy chain variable region of SEQ ID NO: 122 and a CH1 containing sequence (SEQ ID NO: 124) and / or a light chain variable region of SEQ ID NO: 123. The Trastuzumab antibody may comprise a heavy chain of SEQ ID NO: 127 and / or a light chain of SEQ ID NO: 128.
본 발명에서의 변형항체와 결합되는 약물은 질병의 치료 효과가 있는 약물이면 어느 것이라도 제한없이 사용가능하며, 특히 종양 세포의 증식억제 효능이 있는 암 치료용 약물이 바람직하다. The drug combined with the modified antibody in the present invention can be used without limitation any drug that has a therapeutic effect of the disease, and particularly preferred is a drug for treating cancer having a proliferation inhibitory effect of tumor cells.
상기 약물은 변형항체 말단에 도입된 모티프의 시스테인기 또는 세린기에 접합될 수 있다.The drug may be conjugated to the cysteine group or serine group of the motif introduced at the terminal of the modified antibody.
본 발명의 변형항체-약물 접합체에 사용될 수 있는 약물은 구체적으로, 세포독성 또는 세포증식 억제 효과를 갖는 임의의 화합물, 부분 또는 기를 포함하며, (i) 마이크로투불린 억제제, 유사분열 억제제, 토포이소머라아제 억제제, 또는 DNA 인터컬레이터로서 기능할 수 있는 화학요법제; (ii) 효소적으로 기능할 수 있는 단백질 독소; (iii) 특정 암유전자(oncogene)의 발현을 억제시킬 수 있는 마이크로 RNA (miRNA), siRNA, shRNA; 또는 (iv) 방사선동위원소 등이 포함될 수 있다. Drugs that may be used in the modified antibody-drug conjugates of the present invention specifically include any compound, moiety or group that has a cytotoxic or cytostatic effect, and (i) a microtubulin inhibitor, mitosis inhibitor, topoiso Chemotherapeutic agents that can function as merase inhibitors, or DNA intercalators; (ii) protein toxins that can function enzymatically; (iii) micro RNA (miRNA), siRNA, shRNA capable of inhibiting the expression of certain oncogenes; Or (iv) radioisotopes.
이러한 약물에는 예를 들어, 마이탄시노이드, 오리스타틴, 아미노프테린, 악티노마이신, 블레오마이신, 탈리소마이신, 캄프토쎄신, N8-아세틸 스퍼미딘, 1-(2 클로로에틸)-1,2-다이메틸 술포닐 하이드라자이드, 에스퍼라마이신, 에토포사이드, 6-머캅토퓨린, 돌라스타틴, 트리코테센, 칼리케아미신, 탁산, 메토트렉세이트, 빈크리스틴, 빈블라스틴, 독소루비신, 멜팔란, 미토마이신 A, 미토마이신 C, 클로람부실, 듀오카마이신, 핵산 분해 효소, 세균이나 동식물 유래의 독소, 시스플라틴, 이리노테칸, 파클리탁셀 및 도세탁셀에서 선택된 하나 이상일 수 있으나, 이에 한정되는 것은 아니다. Such drugs include, for example, maytansinoids, orstatin, aminopterin, actinomycin, bleomycin, thalisomycin, camptocecin, N8-acetyl spermidine, 1- (2 chloroethyl) -1, 2-dimethyl sulfonyl hydrazide, esperamycin, etoposide, 6-mercaptopurine, dolastatin, tricortesene, calicheamicin, taxane, methotrexate, vincristine, vinblastine, doxorubicin, melphalan, mito Mycin A, mitomycin C, chlorambucil, duocarmycin, nucleolytic enzymes, toxins derived from bacteria or plants, cisplatin, irinotecan, paclitaxel and docetaxel, but is not limited thereto.
경우에 따라서, 상기 약물은 링커 및 링커 시약 상의 친전자성 기와 공유결합을 형성하기 위해 반응할 수 있는 아민, 티올, 히드록실, 히드라지드, 옥심, 히드라진, 티오세미카바존, 히드라진 카르복실레이트, 및 아릴히드라지드기로 구성된 군에서 선택된 하나 이상의 친핵기를 포함할 수 있다. In some cases, the drug may react with an amine, thiol, hydroxyl, hydrazide, oxime, hydrazine, thiosemicarbazone, hydrazine carboxylate, which can react to form covalent bonds with electrophilic groups on the linker and linker reagent, And one or more nucleophilic groups selected from the group consisting of arylhydrazide groups.
또 다른 관점에서, 본 발명은 상기 항체-약물 접합체를 유효성분으로 포함하는 치료용 조성물을 제공한다. 상기 조성물에서 변형항체-약물 접합체의 약물은 암 치료에 있어서 종양 세포를 사멸 또는 억제하는 약물의 국소적 전달을 위한 항체 접합체의 사용으로 약물 부분이 종양 내로 항체-항원으로 표적화된 전달 및 세포 내로의 축적을 가능하게 한다. In another aspect, the present invention provides a therapeutic composition comprising the antibody-drug conjugate as an active ingredient. The drug of the modified antibody-drug conjugate in the composition is characterized by the use of an antibody conjugate for topical delivery of a drug that kills or inhibits tumor cells in the treatment of cancer, with the drug moiety being targeted to the antibody-antigen into the tumor and into the cell. Enable accumulation
또한, 본 발명에서는 변형항체-약물 접합체를 유효성분으로 하여 암, 자가면역, 염증성 또는 감염성 질병 또는 질환이 있는 상기 표적 세포에 접촉하여 표적 세포의 증식을 억제하는 방법을 제공한다. The present invention also provides a method of inhibiting proliferation of target cells by contacting the target cells with cancer, autoimmune, inflammatory or infectious diseases or diseases using the modified antibody-drug conjugate as an active ingredient.
본 발명에서의 치료 가능한 암은 간암, 위암, 유방암, 결장암, 골암, 췌장암, 두부 또는 경부 암, 자궁암, 난소암, 직장암, 식도암, 소장암, 항문부근암, 결장암, 나팔관암종, 자궁내막암종, 자궁경부암종, 질암종, 음문암종, 호지킨병(Hodgkin 's disease), 전립선암, 방광암, 신장암, 수뇨관암, 신장세포암종, 신장골반암종 및 중추신경계 종양으로부터 선택되는 하나 이상의 것으로 이에 한정되지 않는다. 구체적인 예로 시험관내의 엽산 수용체가 증폭된 암 세포인 KB 세포에서 변형항체-약물 접합체를 접촉시켜 세포증식 억제를 유도할 수 있다. 그러므로 본 발명에서의 변형항체-약물 접합체를 유효성분으로 한 억제 방법은 상기 질병과 관련된 세포를 사멸시키거나 증식 속도를 감소시키며 억제시키는 효과를 가진다.Treatable cancers in the present invention are liver cancer, stomach cancer, breast cancer, colon cancer, bone cancer, pancreatic cancer, head or neck cancer, uterine cancer, ovarian cancer, rectal cancer, esophageal cancer, small intestine cancer, anal muscle cancer, colon cancer, fallopian tube carcinoma, endometrial carcinoma, One or more selected from cervical carcinoma, vaginal carcinoma, vulvar carcinoma, Hodgkin's disease, prostate cancer, bladder cancer, kidney cancer, ureter cancer, renal cell carcinoma, renal pelvic carcinoma and central nervous system tumor It doesn't work. As a specific example, a modified antibody-drug conjugate may be contacted in KB cells, which are cancer cells amplified with in vitro folate receptors, to induce cell proliferation inhibition. Therefore, the inhibitory method using the modified antibody-drug conjugate of the present invention as an active ingredient has an effect of killing or reducing the rate of proliferation and inhibiting cells associated with the disease.
본 발명에서 사용되는 기술용어 및 과학용어에 있어 다른 정의가 없다면, 이 발명이 속하는 기술 분야에서 통상의 지식을 가진 자가 일반적으로 이해하는 의미를 지닌다. 또한, 종래와 동일한 기술적 구성 및 작용에 대한 반복되는 설명은 생략하기로 한다.Unless otherwise defined in the technical and scientific terms used in the present invention, those having ordinary skill in the art to which the present invention pertains generally have meaning. In addition, repeated description of the same technical configuration and operation as in the prior art will be omitted.
이하, 본 발명을 구체적인 실시예에 의해 보다 상세히 설명하고자 한다. 하지만, 본 발명은 하기 실시예에 의해 한정되는 것은 아니며, 본 발명의 아이디어와 범위 내에서 여러 가지 변형 또는 수정될 수 있음은 통상의 기술자에게는 자명한 것이다.Hereinafter, the present invention will be described in more detail with reference to specific examples. However, the present invention is not limited by the following examples, and it will be apparent to those skilled in the art that various modifications or changes can be made within the idea and scope of the present invention.
실시예 1. 발현 벡터 pAV4의 제조Example 1 Preparation of Expression Vector pAV4
발현벡터 클로닝은 모벡터인 pSGHV0(GenBank Accession No. AF285183)를 이용하여 산업체에서 항체제작에 사용 가능하도록 목적에 맞게 개량시켜 개발한 pAV4 벡터를 이용하였다. 모벡터는 대장균과 같은 박테리아를 이용하여 인체 유래 단백질을 발현시킬 경우 세포 내로 과량 발현되지만, 활성을 가진 물질로 얻기 어려운 단백질의 경우에 동물세포를 이용하여 세포 외로 생리활성을 가진 관심 단백질을 고농도로 발현시켜 손쉽게 정제할 목적으로 제작된 연구용 벡터이다. 그러나, 산업체에서 생산용으로 사용하기에는 여러 가지로 제한이 있으므로 이 벡터의 가장 큰 장점인 발현량이 높은 것을 생산에 이용하기 위하여 산업체에서 사용 가능하도록 개량한 것이다. 또한, 항체의 경우 중쇄(heavy chain)와 경쇄(light chain) 두 개의 단백질을 동시에 발현시켜야 하므로 이러한 목적에 적합한 벡터를 개발하였다.Expression vector cloning was performed using the pAV4 vector, which was developed and modified for the purpose of using the parent vector pSGHV0 (GenBank Accession No. AF285183) to manufacture antibodies in the industry. The parent vector is overexpressed into cells when a human-derived protein is expressed using bacteria such as Escherichia coli, but in the case of a protein that is difficult to obtain as an active substance, a high concentration of the protein of interest having a physiological activity outside the cell is obtained using animal cells. It is a research vector produced for the purpose of easy purification by expression. However, since there are various limitations to use for production in industry, the vector has been improved to be used in industry in order to use the high expression amount, which is the biggest advantage of this vector for production. In addition, in the case of an antibody, a heavy chain and a light chain must simultaneously express two proteins, so a vector suitable for this purpose has been developed.
실시예 2. Folate receptor에 결합능이 있는 모항체 및 금속이온 결합 모티프인 ACGHA를 도입한 변형항체의 벡터 제조Example 2 Vector Preparation of Modified Antibodies Incorporating ACGHA, a Parent Antibody Having a Binding Capability to the Folate Receptor, and a Metal Ion Binding Motif
엽산 수용체에 결합능이 있는 모항체 (Fwt) 벡터를 제조하기 위하여, 서열번호 125의 중쇄와 서열번호 126의 경쇄 코딩 cDNA를 CHO 세포에서 발현이 극대화되도록 코돈 최적화된 서열로 각각 합성하였다. 이 유전자를 pAV4 벡터의 XhoI/NotI 과 ApaI/SmaI에 각각 클로닝하여 모항체 벡터(pFwt)를 제조하였다. To prepare a parental antibody (Fwt) vector capable of binding to a folate receptor, the heavy chain of SEQ ID NO: 125 and the light chain coding cDNA of SEQ ID NO: 126 were respectively synthesized into codon optimized sequences to maximize expression in CHO cells. This gene was cloned into XhoI / NotI and ApaI / SmaI of the pAV4 vector, respectively, to prepare a parental antibody vector (pFwt).
Fwt 항체 아미노산 서열Fwt Antibody Amino Acid Sequence
구분division 서열order 번호number
중쇄Heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGKEVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGK 서열번호 115SEQ ID NO: 115
경쇄Light chain DIQLTQSPSSLSASVGDRVTITCSVSSSISSNNLHWYQQKPGKAPKPWIYGTSNLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSYPYMYTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVDNALQSGNSQESVTEQDSKDSTYSLSSTLTLSKADYEKHKVYACEVTHQGLSSPVTKSFNRGECDIQLTQSPSSLSASVGDRVTITCSVSSSISSNNLHWYQQKPGKAPKPWIYGTSNLASGVPSRFSGSGSGTDYTFTISSLQPEDIATYYCQQWSSYPYMYTFGQGTKVEIKRTVAAPSVFIFPPSDEQLKSGTASVVCLLNNFYPREAKVQWKVKLSKLSKLSKVKKKVKVKDR 서열번호 116SEQ ID NO: 116
2-1. Folate receptor 결합 모항체인 Fwt의 변형항체 FM2 제조: 2-1. Preparation of Modified Antibody FM2 from Fwt, a Folate Receptor Binding Parent Antibody:
금속이온 결합 모티프(ACGHA) 2개를 갖는 Fwt의 변형 항체인 FM2 (Fwt-ACGHAACGHA (서열번호 5), FM2)을 제조하기 위해 모항체인 Fwt의 벡터(pFwt)를 주형으로 하여 XhoI-Q5-F 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3': 서열번호 105)와 M2 역방향 프라이머 (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': 서열번호 106)를 이용하여 PCR로 증폭하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pFwt와 접합하여 변형항체 벡터(pFM2)를 제조하였다.XhoI-Q5-F was used as a template to prepare FM2 (Fwt-ACGHAACGHA (SEQ ID NO: 5), FM2), a modified antibody of Fwt having two metal ion binding motifs (ACGHA), as a template. PCR was performed using forward primers (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 105) and M2 reverse primer (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': SEQ ID NO: 106). The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2).
2-2. Folate receptor 결합 변형항체 FM1 제조: 2-2. Preparation of Folate receptor-binding modified antibody FM1:
금속 이온 결합 모티프(ACGHA)를 하나만 갖는 트라스트주맵 변형 항체인 FM1 (Fwt-GGGACGHA, pFM1)를 제조하기 위해 위에서 제조한 FM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3': 서열번호 107)와 역방향 프라이머(5'-CCATGCGGCCGCTCATTTAGGCATGGCC ACAAGCA CCTC CACCACCCGGAGACAGGGAGA-3': 서열번호 108)를 사용하여 site-directed mutagenesis (엔지노믹스 EzChange Site-directed mutagenesis kit, Ez004S) 방법으로 이용하여 PCR로 증폭하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pFwt와 접합하여 변형항체 벡터(pFM1)를 제조하였다.In order to prepare FM1 (Fwt-GGGACGHA, pFM1), a trastuzimab modified antibody having only one metal ion binding motif (ACGHA), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ') was used as a template. ) And reverse primers (5'-CCATGCGGCCGCTCATTTAGGCATGGCC ACAAGCA CCTC CACCACCCGGAGACAGGGAGA-3 ': SEQ ID NO: 108) were used to amplify by PCR using the site-directed mutagenesis (Enzymex EzChange Site-directed mutagenesis kit, Ez004S) method. The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM1).
2-3. Folate receptor 결합 변형항체 FM2L 제조:2-3. Preparation of Folate receptor-binding modified antibody FM2L:
금속이온 결합 모티프(ACGHA)가 3개의 아미노산 링커로 연결된 변형 항체인 FM2L (Fwt-ACGHAGGGACGHA, pFM2L)를 제조하기 위해 위에서 제조한 변형항체 FM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3': 서열번호 109)와 역방향 프라이머(5'-CCATGCGGCCGCTCATTTAGGCATGGCCACAAGCACCTCCACCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': 서열번호 110)를 사용하여 site-directed mutagenesis (엔지노믹스 EzChange Site-directed mutagenesis kit, Ez004S) 방법으로 PCR을 이용하여 두 개의 금속 이온 결합 모티프 사이에 글라이신 링커를 첨가하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pFwt와 접합하여 변형항체 벡터(pFM2L)를 제조하였다. In order to prepare a modified antibody FM2L (Fwt-ACGHAGGGACGHA, pFM2L), in which a metal ion binding motif (ACGHA) is linked with a three amino acid linker, a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': Site-directed mutagenesis (SEQ ID NO: 109) and reverse primers (5'-CCATGCGGCCGCTCATTTAGGCATGGCCACAAGCACCTCCACCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3 ': SEQ ID NO: 110) using two methods: site-directed mutagenesis (enzymeics EzChange site-directed mutagenesis kit, PCR using two metal ion methods) Glycine linkers were added between the motifs. The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2L).
2-4. Folate receptor 결합 변형항체 FM2a 제조:2-4. Preparation of Folate receptor-binding modified antibody FM2a:
FM2 변형항체에 존재하는 2개의 금속이온 결합모티프인 ACGHAACGHA (서열번호 5)에서 안쪽의 시스테인을 세린(serine)으로 치환하여 하나의 금속이온 결합 모티프만이 존재하는 변형항체인 FM2a (Fwt-ASGHAACGHA (서열번호 26), pFM2a)를 제조하기 위해 위에서 제조한 변형항체 FM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3': 서열번호 111)와 역방향 프라이머 (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCTG AGGCACCCGGAGACAGG-3’: 서열번호 112)를 PCR을 이용하여 안쪽의 시스테인을 세린으로 치환하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pFwt와 접합하여 변형항체 벡터(pFM2a)를 제조하였다.In the ACGHAACGHA (SEQ ID NO: 5), two metal ion binding motifs present in the FM2 modified antibody, a modified antibody FM2a (Fwt-ASGHAACGHA ( SEQ ID NO: 26), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ') and a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCGA AGGCACCCG: SEQ ID NO: 112) was used to replace inner cysteine with serine using PCR. The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with the expression vector pFwt having the XhoI / NotI cleavage to prepare a modified antibody vector (pFM2a).
2-5. Folate receptor 결합 변형항체 FM2b 제조:2-5. Preparation of Folate receptor-binding modified antibody FM2b:
FM2 변형항체에 존재하는 2개의 금속이온 결합모티프인 ACGHAACGHA에서 바깥쪽의 시스테인을 세린(serine)으로 치환하여 하나의 금속이온 결합 모티프만이 존재하는 변형항체인 FM2b (Fwt-ACGHAASGHA (서열번호 21), pFM2b)를 제조하기 위해 위에서 제조한 변형항체 FM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3': 서열번호 113)와 역방향 프라이머 (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACCCGGAGACAGG-3': 서열번호 114)를 사용하여 site-directed mutagenesis (엔지노믹스 EzChange Site-directed mutagenesis kit, Ez004S) 방법으로 PCR을 이용하여 안쪽의 시스테인을 세린으로 치환하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두 개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pFwt와 접합하여 변형항체 벡터(pFM2b)를 제조하였다.In the ACGHAACGHA, the two metal ion binding motifs present in the FM2 modified antibody, FM2b (Fwt-ACGHAASGHA (SEQ ID NO: 21)), in which only one metal ion binding motif exists, by replacing the outer cysteine with serine To prepare pFM2b), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': SEQ ID NO: 113) and a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACCC-3): Internal cysteine was replaced with serine by PCR using site-directed mutagenesis (EzChange Site-directed mutagenesis kit, Ez004S). The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pFwt having an XhoI / NotI cleavage to prepare a modified antibody vector (pFM2b).
FM 항체FM antibody
구분division 서열order 번호number
FM2 중쇄FM2 heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGACGHAACGHA EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG ACGHAACGHA 서열번호 117SEQ ID NO: 117
FM2a 중쇄FM2a heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGASGHAACGHA EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG ASGHAACGHA 서열번호 118SEQ ID NO: 118
FM2b 중쇄FM2b heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGACGHAASGHA EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG ACGHAASGHA 서열번호 119SEQ ID NO: 119
FM1 중쇄FM1 heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGGGGACGHA EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG GGGACGHA 서열번호 120SEQ ID NO: 120
FM2L 중쇄FM2L heavy chain EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPGACGHAGGGACGHA EVQLVESGGGVVQPGRSLRLSCSASGFTFSGYGLSWVRQAPGKGLEWVAMISSGGSYTYYADSVKGRFAISRDNAKNTLFLQMDSLRPEDTGVYFCARHGDDPAWFAYWGQGTPVTVSSASTKGPSVFPLAPSSKSTSGGTAALGCLVKDYFPEPVTVSWNSGALTSGVHTFPAVLQSSGLYSLSSVVTVPSSSLGTQTYICNVNHKPSNTKVDKKVEPKSCDKTHTCPPCPAPELLGGPSVFLFPPKPKDTLMISRTPEVTCVVVDVSHEDPEVKFNWYVDGVEVHNAKTKPREEQYNSTYRVVSVLTVLHQDWLNGKEYKCKVSNKALPAPIEKTISKAKGQPREPQVYTLPPSRDELTKNQVSLTCLVKGFYPSDIAVEWESNGQPENNYKTTPPVLDSDGSFFLYSKLTVDKSRWQQGNVFSCSVMHEALHNHYTQKSLSLSPG ACGHAGGGACGHA 서열번호 121SEQ ID NO: 121
실시예 3. Folate receptor 결합 모항체인 Fwt와 변형항체들의 발현 및 정제Example 3 Expression and Purification of Fwt and Folate Receptor Binding Antibody
차이니즈 햄스터 난소세포(CHO-K1)를 이용하여 실시예 2에서 제조한 Fwt 및 이의 금속이온 결합 모티프 변형항체들(FM1, FM2, FM2L, FM2a, FM2b)의 단백질 발현을 확인하였다. CHO-K1은 10% FBS(Fetal Bovine Serum)와 항생제를 포함한 DMEM(Dulbecco's Modified Eagle Media)에 37℃, 5% CO2, 배양기에서 배양 하였다. Fwt 및 이의 변형항체 발현벡터를 도입하기 하루 전, 100㎜ 배양접시에 세포를 5×106/ml 농도로 접종하여 배양한 후, FBS와 항생제가 없는 800㎕의 DMEM과 10㎍의 Fwt 또는 변형항체 발현벡터를 혼합하여 상온에서 1분 동안 유지한 후, 20㎍의 PEI(Polyethylenimine, linear, Polysciences Inc (Cat. no: 23966, MW~25,000))와 혼합하여 10~15분 정도 상온에서 방치하였다. 이 때 하루 전 배양하였던 세포를 PBS로 씻어내고 새로운 배양액 6㎖의 DMEM을 첨가하였다. 10~15분 동안 상온에 방치한 Fwt 또는 그의 변형항체의 발현벡터를 이 배양접시에 첨가하였다. 다음 날 PBS로 씻어내고 FBS가 없는 IMDM(Cat. No 12200-028, Gibco, Iscove's Modified Dulbecco's Medium) 배지를 첨가하여 단백질 발현을 확인하였다.Chinese hamster ovary cells (CHO-K1) was used to confirm the protein expression of the Fwt and its metal ion binding motif modified antibodies (FM1, FM2, FM2L, FM2a, FM2b) prepared in Example 2. CHO-K1 was incubated in Dulbecco's Modified Eagle Media (DMEM) containing 10% FBS (Fetal Bovine Serum) and antibiotics at 37 ° C, 5% CO 2 , incubator. One day before the introduction of Fwt and its modified antibody expression vector, cells were inoculated at a concentration of 5 × 10 6 / ml in a 100 mm culture dish, followed by incubation with 800 μl of DMEM without FBS and antibiotics, and 10 μg of Fwt or modified. The antibody expression vector was mixed and maintained at room temperature for 1 minute, and then mixed with 20 μg of PEI (Polyethylenimine, linear, Polysciences Inc (Cat. No: 23966, MW ~ 25,000)) and left at room temperature for 10 to 15 minutes. . At this time, the cells cultured one day ago were washed with PBS and 6 ml of DMEM was added. Expression vectors of Fwt or its modified antibodies left at room temperature for 10-15 minutes were added to this culture dish. The next day, the protein expression was confirmed by washing with PBS and adding IMDM (Cat. No 12200-028, Gibco, Iscove's Modified Dulbecco's Medium) medium without FBS.
이와 같이 발현된 Fwt 및 이의 금속이온 결합 모티프 변형항체는 하기와 같이 정제하였다. 구체적으로는, 세포 배양액으로 분비된 Fwt 및 이의 금속이온 결합 모티프 변형항체를 정제하기 위하여, 배양액을 원심 분리하여 세포를 제거한 후 상층액만을 취하고 이 상층액을 평형 완충용액으로 평형화된 HiTrap Protein A HP(GE Healthcare, 미국) 컬럼에 주입하고 평형 완충용액으로 충분히 세척한 후 Glycine 완충용액(100mM Glycine, pH 2.8)으로 pH를 변화시켜 단백질을 용출시켰다. 상기 용액을 인산염 완충용액으로 투석한 후, Vivaspin20(Sartorius, 미국)을 사용하여 농축하였고, 최종적으로 고순도로 정제된 단백질을 얻었다. Thus expressed Fwt and its metal ion binding motif modified antibody was purified as follows. Specifically, in order to purify the Fwt and its metal ion binding motif-modified antibody secreted into the cell culture, the culture medium was centrifuged to remove the cells, and then only the supernatant was taken and the supernatant was equilibrated with the equilibration buffer HiTrap Protein A HP. (GE Healthcare, USA) The protein was eluted by injection into the column, washing well with equilibration buffer, and changing the pH with Glycine buffer (100 mM Glycine, pH 2.8). The solution was dialyzed with phosphate buffer, concentrated using Vivaspin20 (Sartorius, USA), and finally purified protein was obtained with high purity.
실시예 4. Fwt의 변형항체와 maleimide기와 시스테인의 반응을 통한 항체-약물 접합체 제조Example 4 Preparation of Antibody-Drug Conjugate by Reaction of Fwt Modified Antibody with Maleimide Group and Cysteine
본 발명에서는 MMAE와 Fwt의 변형항체를 접합시켜 FMx(Fwt의 금속이온 결합 모티프 변이체)-MMAE 결합체를 제조하였다. MMAE로 알려진 아우리스타틴(Auristatin)의 접합 가능성 유도체인 단일 메틸 아우리스타틴 E (monomethyl Auristatin E, 화학식 2 참조)로서, In the present invention, a modified antibody of MMAE and Fwt was conjugated to prepare an FMx (metal ion-binding motif variant of Fwt) -MMAE conjugate. As a single methyl auristatin E (see Formula 2), a conjugation derivative of Auristatin known as MMAE,
Figure PCTKR2017003508-appb-C000002
Figure PCTKR2017003508-appb-C000002
세포 내에서 단백질 분해효소(protease)에 의해서 분해되는 발린-시트룰린(valine-citurulline)과 자체 분해 스페이서 그룹인 파라 아닐린 벤조산(para-aniline benzoic acid: PABA)를 통하여 티올기에 선택적으로 결합하는 말레이마이드기에 연결되는 구조를 갖는다. 이를 통칭하여 MC(maleimido caproic acid)-VC(valine-citurulline)-PAB-MMAE라고 하며 아우리스타틴은 세포내 독성이 강한 물질로써 세포증식 억제 시험에서의 IC50 값이 200~300 pM로 알려져 있다.Maleimide groups that selectively bind to thiol groups through valine-citurulline, which is degraded by protease in cells, and para-aniline benzoic acid (PABA), a self-decomposing spacer group It has a structure to be connected. Collectively, it is called MC (maleimido caproic acid) -VC (valine-citurulline) -PAB-MMAE. Auristatin is a substance with strong intracellular toxicity and IC 50 value in the cell proliferation inhibition test is known to be 200 ~ 300 pM. .
본 발명에서는 정제된 변형항체 1당량 당 환원제인 TCEP를 3 당량 가하여 4℃에서 30분간 반응시켜서 티올기를 환원 시킨 후, MC-vc-PAB-MMAE를 3 당량 첨가하여 상온에서 2시간 가량 반응 시킨다. 반응은 과량의 시스테인을 가하여 종결시키고, 과량의 MC-vc-PAB-MMAE와 TCEP는 원심분리 여과 필터와 인산염 완충용액에서의 투석을 통하여 제거하여 최종 정제된 FMx-MC-vc-PAB-MMAE를 제조하였다.In the present invention, after adding 3 equivalents of TCEP, a reducing agent per equivalent of purified antibody, and reacting at 4 ° C. for 30 minutes to reduce thiol groups, 3 equivalents of MC-vc-PAB-MMAE are added and reacted at room temperature for 2 hours. The reaction was terminated by the addition of excess cysteine, and excess MC-vc-PAB-MMAE and TCEP were removed through dialysis in a centrifugal filtration filter and phosphate buffer to remove the final purified FMx-MC-vc-PAB-MMAE. Prepared.
각 변형항체의 중쇄로의 접합 수율은 아래의 표 3과 같다.The yield of conjugation to the heavy chain of each modified antibody is shown in Table 3 below.
Maleimide기를 통한 각 변형항체의 접합 수율Conjugation yield of each modified antibody through maleimide group
변형항체Modified antibody FM1FM1 FM2FM2 FM2aFM2a FM2bFM2b FM2LFM2L
접합수율Junction yield 62.7%62.7% 64.1%64.1% 64.5%64.5% 97.5%97.5% 66.3%66.3%
위의 표 3에서 볼 수 있는 바와 같이, 각 변형항체간에 중쇄로의 약물 접합 수율은 매우 큰 차이를 보여주고 있다. FM1, FM2, FM2a, FM2L 등의 변형 항체들은 같은 약물 접합 조건에서 약 63~66%의 접합 수율을 보임에 반하여, FM2b의 경우는 97.5%의 매우 높은 약물 접합 수율을 보여주고 있다. FM2b의 경우, 서로 다른 2개의 발현 배치(transient transfection batch)에서 나온 시료에서 거의 유사한 접합 수율을 보여주고 있다.As can be seen in Table 3 above, the yield of drug conjugation to the heavy chain between the modified antibodies shows a very large difference. Modified antibodies such as FM1, FM2, FM2a, and FM2L show conjugation yields of about 63-66% under the same drug conjugation conditions, while FM2b shows very high drug conjugation yields of 97.5%. In the case of FM2b, nearly identical conjugation yields were shown in samples from two different transfection batches.
실시예 5. Fwt의 변형항체와 브로모아세트아마이드(bromoacetamide)기와 시스테인의 반응을 통한 항체-약물 접합체 제조Example 5 Preparation of Antibody-Drug Conjugate by Reaction of Fwt Modified Antibody with Bromoacetamide Group
본 실시예에서는 브로모아세트아마이드기를 통하여 시스테인의 티올기에 결합하는 항체-약물 접합체를 제조하였다. 브로모아세트아마이드는 세포 내에서 단백질 분해효소(protease)에 의해서 분해되는 발린-시트룰린(valine-citurulline)과 자체 분해 스페이서 그룹인 파라 아닐린 벤조산(para-aniline benzoic acid: PABA)를 통하여 MMAE와 결합된 구조를 가지고, bromoacetamide와 티올기와의 결합을 통하여 MMAE를 변형항체에 결합시킨다. 이를 br(bromo acetamide)-VC(valine-citurulline)-PAB-MMAE라고 명명한다.In this example, an antibody-drug conjugate that binds to a thiol group of cysteine through a bromoacetamide group was prepared. Bromoacetamide is bound to MMAE via valine-citurulline, which is degraded by protease in cells, and para-aniline benzoic acid (PABA), a self-decomposing spacer group. With the structure, MMAE is bound to the modified antibody through the coupling of bromoacetamide and thiol group. This is called br (bromo acetamide) -VC (valine-citurulline) -PAB-MMAE.
본 발명에서는 정제된 변형항체 1당량 당 환원제인 TCEP를 3 당량 가하여 4℃에서 30분간 반응시켜서 티올기를 환원 시킨 후, br-vc-PAB-MMAE를 3 당량 첨가하여 37℃에서 2시간 반응 시킨다. 반응은 과량의 시스테인을 가하여 종결시키고, 과량의 br-vc-PAB-MMAE와 TCEP는 원심분리 여과 필터와 인산염 완충용액에서의 투석을 통하여 제거하여 최종 정제된 FMx-acetamide-vc-PAB-MMAE를 제조하였다.In the present invention, after adding 3 equivalents of TCEP, a reducing agent per equivalent of purified antibody, and reacting at 4 ° C. for 30 minutes to reduce thiol groups, 3 equivalents of br-vc-PAB-MMAE are added and reacted at 37 ° C. for 2 hours. The reaction was terminated by the addition of excess cysteine, and excess br-vc-PAB-MMAE and TCEP were removed via dialysis in a centrifugal filtration filter and phosphate buffer to remove the final purified FMx-acetamide-vc-PAB-MMAE. Prepared.
각 변형항체의 중쇄로의 접합 수율은 아래의 표 4와 같다.The yield of conjugation to the heavy chain of each modified antibody is shown in Table 4 below.
Bromoacetamide기를 통한 각 변형항체의 접합 수율Conjugation yield of each modified antibody through bromoacetamide group
변형항체Modified antibody FM1FM1 FM2FM2 FM2aFM2a FM2bFM2b FM2LFM2L
접합수율Junction yield 42%42% 44%44% 38%38% 73%73% 49%49%
위의 표 4에서 볼 수 있는 바와 같이, 각 변형항체간에 중쇄로의 약물 접합 수율은 FM2b가 여타 다른 변형항체에 비하여 월등히 우월한 접합 수율을 보여주고 있다. Iodoacetamide를 통한 접합 반응에서도 FM2b는 다른 변형항체에 비하여 월등히 우월한 접합 수율을 보여준다.As can be seen in Table 4, the yield of drug conjugated to the heavy chain between each of the modified antibodies shows that the conjugate yield of FM2b is superior to other modified antibodies. In the conjugation reaction via iodoacetamide, FM2b showed superior conjugation yield compared to other modified antibodies.
Iodoacetamide기를 통한 각 변형항체의 접합 수율Conjugation yield of each modified antibody through iodoacetamide group
변형항체Modified antibody FM1FM1 FM2FM2 FM2aFM2a FM2bFM2b FM2LFM2L
접합수율Junction yield 51%51% 53%53% 44%44% 80%80% 55%55%
실시예 6. Trastuzumab 변형항체의 생산Example 6 Production of Trastuzumab Modified Antibodies
실시예 2에서 사용한 방법과 같이, trastuzumab의 C-말단에 시스테인을 포함하는 금속이온 모티프를 도입한 변형항체를 제작하였다. As in the method used in Example 2, a modified antibody was prepared by introducing a metal ion motif containing cysteine at the C-terminus of trastuzumab.
6-1. Trastuzumab의 변형항체 HM2 제조: 6-1. Preparation of Modified Antibody HM2 from Trastuzumab:
금속이온 결합 모티프(ACGHA) 2개를 갖는 trastuzumab의 변형 항체인 HM2 (HR-ACGHAACGHA(서열번호 5), HM2)을 제조하기 위해 모항체인 trastuzumab의 벡터(pHR)를 주형으로 하여 XhoI-Q5-F 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3': 서열번호 111)와 M2 역방향 프라이머 (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': 서열번호 112)를 이용하여 PCR로 증폭하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pHR와 접합하여 변형항체 벡터(pHM2)를 제조하였다.To prepare HM2 (HR-ACGHAACGHA (SEQ ID NO: 5), HM2), a modified antibody of trastuzumab having two metal ion binding motifs (ACGHA), as a template, a vector (pHR) of the parent antibody trastuzumab was used. PCR was performed using forward primers (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 111) and M2 reverse primers (5'-CCATGCGGCCGCTCATTTAGGCATGGCCA CAAGCAGCATGGCCACAGGCACCCGGAGACAGGGAGAGGC-3': SEQ ID NO: 112). The amplified nucleotides were cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2).
6-2. Trastuzumab의 변형항체 HM2a 제조:6-2. Preparation of the modified antibody HM2a of Trastuzumab:
HM2 변형항체에 존재하는 2개의 금속이온 결합모티프인 ACGHAACGHA (서열번호 5)에서 안쪽의 시스테인을 세린(serine)으로 치환하여 하나의 금속이온 결합 모티프만이 존재하는 변형항체인 HM2a (HR-ASGHAACGHA(서열번호 26), pHM2a)를 제조하기 위해 위에서 제조한 변형항체 HM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3': 서열번호 111)와 역방향 프라이머 (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCTG AGGCACCCGGAGACAGG-3': 서열번호 112)를 PCR을 이용하여 안쪽의 시스테인을 세린으로 치환하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pHR와 접합하여 변형항체 벡터(pHM2a)를 제조하였다.In the ACGHAACGHA (SEQ ID NO: 5), two metal ion-binding motifs present in the HM2 modified antibody, a substitution antibody HM2a (HR-ASGHAACGHA ( SEQ ID NO: 26), a forward primer (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGT ATCATCC-3 ': SEQ ID NO: 111) and a reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCACAAGCAGCATGGCCGA AGGCAG-3CC): SEQ ID NO: 112) was used to replace inner cysteine with serine using PCR. The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2a).
6-3. Trastuzumab의 변형항체 HM2b 제조:6-3. Preparation of the modified antibody HM2b of Trastuzumab:
HM2 변형항체에 존재하는 2개의 금속이온 결합모티프인 ACGHAACGHA에서 바깥쪽의 시스테인을 세린(serine)으로 치환하여 하나의 금속이온 결합 모티프만이 존재하는 변형항체인 HM2b (HR-ACGHAASGHA(서열번호 21), pHM2b)를 제조하기 위해 위에서 제조한 변형항체 HM2를 주형으로 하여 정방향 프라이머 (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3': 서열번호 109)와 역방향 프라이머 (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACCCGGAGACAGG-3': 서열번호 114)를 사용하여 site-directed mutagenesis (엔지노믹스 EzChange Site-directed mutagenesis kit, Ez004S) 방법으로 PCR을 이용하여 안쪽의 시스테인을 세린으로 치환하였다. 상기 증폭된 뉴클레오티드를 말단에 존재하는 두 개의 제한효소인 XhoI과 NotI으로 절단하고, XhoI/NotI 절단부를 가지고 있는 발현 벡터 pHR와 접합하여 변형항체 벡터(pHM2b)를 제조하였다.In the ACGHAACGHA, two metal ion binding motifs present in the HM2 modified antibody, HM2b (HR-ACGHAASGHA (SEQ ID NO: 21)), a variant antibody in which only one metal ion binding motif exists by replacing the outer cysteine with serine To prepare the pHM2b), the forward antibody (5'-GCTCCTCGAGGCCACCATGGGATGGAGCTGTATCATCC-3 ': SEQ ID NO: 109) and the reverse primer (5'-CAGATTGCGGCCGCTCATTAGGCATGGCCTGAAGCAGCATGGCCACA GGCACGA) Internal cysteine was replaced with serine by PCR using site-directed mutagenesis (EzChange Site-directed mutagenesis kit, Ez004S). The amplified nucleotide was cleaved with two restriction enzymes, XhoI and NotI, present at the ends, and conjugated with an expression vector pHR having an XhoI / NotI cleavage to prepare a modified antibody vector (pHM2b).
실시예 7. Trastuzumab 변형항체들의 발현 및 정제Example 7 Expression and Purification of Trastuzumab Modified Antibodies
차이니즈 햄스터 난소세포(CHO-K1)를 이용하여 실시예 6에서 제조한 trastuzumab 변형항체들(HM2, HM2a, HM2b)의 단백질 발현을 확인하였다. CHO-K1은 10% FBS(Fetal Bovine Serum)와 항생제를 포함한 DMEM(Dulbecco's Modified Eagle Media)에 37℃, 5% CO2, 배양기에서 배양 하였다. 변형항체 발현벡터를 도입하기 하루 전, 100㎜ 배양접시에 세포를 5×106/ml 농도로 접종하여 배양한 후, FBS와 항생제가 없는 800㎕의 DMEM과 10㎍의 변형항체 발현벡터를 혼합하여 상온에서 1분 동안 유지한 후, 20㎍의 PEI(Polyethylenimine, linear, Polysciences Inc (Cat. no: 23966, MW~25,000))와 혼합하여 10~15분 정도 상온에서 방치하였다. 이 때 하루 전 배양하였던 세포를 PBS로 씻어내고 새로운 배양액 6㎖의 DMEM을 첨가하였다. 10~15분 동안 상온에 방치한 변형항체의 발현벡터를 이 배양접시에 첨가하였다. 다음 날 PBS로 씻어내고 FBS가 없는 IMDM(Cat. No 12200-028, Gibco, Iscove's Modified Dulbecco's Medium) 배지를 첨가하여 단백질 발현을 확인하였다.Chinese hamster ovary cells (CHO-K1) was used to confirm the protein expression of the trastuzumab modified antibodies (HM2, HM2a, HM2b) prepared in Example 6. CHO-K1 was incubated in Dulbecco's Modified Eagle Media (DMEM) containing 10% FBS (Fetal Bovine Serum) and antibiotics at 37 ° C, 5% CO 2 , incubator. One day before the introduction of the modified antibody expression vector, cells were inoculated at a concentration of 5 × 10 6 / ml in a 100 mm culture dish, followed by incubation with 800 μl of DMEM without antibiotics and 10 μg of the modified antibody expression vector. After maintaining for 1 minute at room temperature, and mixed with 20 ㎍ PEI (Polyethylenimine, linear, Polysciences Inc (Cat. No: 23966, MW ~ 25,000)) and left at room temperature for about 10-15 minutes. At this time, the cells cultured one day ago were washed with PBS and 6 ml of DMEM was added. Expression vectors of the modified antibodies left at room temperature for 10-15 minutes were added to the culture dish. The next day, the protein expression was confirmed by washing with PBS and adding IMDM (Cat. No 12200-028, Gibco, Iscove's Modified Dulbecco's Medium) medium without FBS.
이와 같이 발현된 변형항체는 하기와 같이 정제하였다. 구체적으로는, 세포 배양액으로 분비된 변형항체를 정제하기 위하여, 배양액을 원심 분리하여 세포를 제거한 후 상층액만을 취하고 이 상층액을 평형 완충용액으로 평형화된 HiTrap Protein A HP(GE Healthcare, 미국) 컬럼에 주입하고 평형 완충용액으로 충분히 세척한 후 Glycine 완충용액(100mM Glycine, pH 2.8)으로 pH를 변화시켜 단백질을 용출시켰다. 상기 용액을 인산염 완충용액으로 투석한 후, Vivaspin20(Sartorius, 미국)을 사용하여 농축하였고, 최종적으로 고순도로 정제된 단백질을 얻었다. The modified antibody expressed as above was purified as follows. Specifically, in order to purify the modified antibody secreted into the cell culture, the culture medium was centrifuged to remove the cells, and then only the supernatant was taken and the supernatant was equilibrated with the equilibration buffer HiTrap Protein A HP (GE Healthcare, USA) column. The protein was eluted by changing the pH with Glycine buffer (100mM Glycine, pH 2.8) after washing with equilibration buffer. The solution was dialyzed with phosphate buffer, concentrated using Vivaspin20 (Sartorius, USA), and finally purified protein was obtained with high purity.
실시예 8. Trastuzumab의 변형항체와 MC-vc-PAB-MMAE의 접합을 통한 항체-약물 접합체 제조Example 8 Preparation of Antibody-Drug Conjugate by Conjugation of Modified Antibody of Trastuzumab with MC-vc-PAB-MMAE
본 발명에서는 MMAE와 실시예 6,7에서 생산한 trastuzumab의 변형항체를 접합시켜 HMx(Trastuzumab의 금속이온 결합 모티프 변이체)-MMAE 결합체를 제조하였다. 본 발명에서는 정제된 변형항체 1당량 당 환원제인 TCEP를 3 당량 가하여 4℃에서 30분간 반응시켜서 티올기를 환원 시킨 후, MC-vc-PAB-MMAE를 2.5 당량 첨가하여 상온에서 2시간 가량 반응 시킨다. 반응은 과량의 시스테인을 가하여 종결시키고, 과량의 MC-vc-PAB-MMAE와 TCEP는 원심분리 여과 필터와 인산염 완충용액에서의 투석을 통하여 제거하여 최종 정제된 FMx-MC-vc-PAB-MMAE를 제조하였다.In the present invention, the modified antibody of trastuzumab produced in Example 6, 7 was conjugated with MMAE to prepare a HMx (metal ion binding motif variant of Trastuzumab) -MMAE conjugate. In the present invention, after adding 3 equivalents of TCEP, a reducing agent per equivalent of purified antibody, and reacting at 4 ° C. for 30 minutes to reduce thiol groups, 2.5 equivalents of MC-vc-PAB-MMAE are added and reacted at room temperature for 2 hours. The reaction was terminated by the addition of excess cysteine, and excess MC-vc-PAB-MMAE and TCEP were removed through dialysis in a centrifugal filtration filter and phosphate buffer to remove the final purified FMx-MC-vc-PAB-MMAE. Prepared.
각 변형항체의 중쇄로의 접합 수율은 아래의 표 6과 같다.The yield of conjugation to the heavy chain of each modified antibody is shown in Table 6 below.
Maleimide기를 통한 각 변형항체의 접합 수율Conjugation yield of each modified antibody through maleimide group
변형항체Modified antibody HM2HM2 HM2aHM2a HM2bHM2b
접합수율Junction yield 55.5%55.5% 62.4%62.4% 85.5%85.5%
위의 표 6에서 볼 수 있는 바와 같이, 각 변형항체간에 중쇄로의 약물 접합 수율은 매우 큰 차이를 보여주고 있다. HM2와 HM2a의 변형 항체들은 같은 약물 접합 조건에서 약 55~62%의 접합 수율을 보임에 반하여, HM2b의 경우는 85.5%의 매우 높은 약물 접합 수율을 보여주고 있다. 이러한 결과는 M2b의 sequence가 Farletuzumab 뿐만 아니라, 다른 항체에 도입되었을 때도 동일하게 높은 접합수율을 갖는 다는 것을 보여준다.As can be seen in Table 6 above, the yield of drug conjugation to the heavy chain between the modified antibodies shows a very large difference. The modified antibodies of HM2 and HM2a show about 55-62% conjugation yield under the same drug conjugation conditions, while HM2b shows very high drug conjugation yield of 85.5%. These results show that the sequence of M2b has the same high conjugation yield when introduced into other antibodies as well as Farletuzumab.
실시예 9. M2b(ACGHAASGHA: 서열번호 21) 서열에서 Serine을 치환한 변형항체의 생산Example 9 Production of Modified Antibody Substituted with Serine in M2b (ACGHAASGHA: SEQ ID NO: 21) Sequence
앞서의 실시예에서 금속이온 결합 모티프인 M2(ACGHAACGHA)에서 뒤쪽 cysteine을 serine으로 치환한 M2b(ACGHAASGHA)가 M2 서열에 비하여 월등히 높은 약물 접합능을 보임을 확인할 수 있었다. 이러한 serine 치환자리가 다른 아미노산의 치환에서도 동일한 효과를 보이는지 확인하기 위하여 이 위치에 여러가지 아미노산을 치환한 변형항체를 생산하였다. In the previous example, it was confirmed that M2b (ACGHAASGHA) in which the rear cysteine was substituted with serine in M2 (ACGHAACGHA), which is a metal ion binding motif, showed much higher drug binding ability than the M2 sequence. In order to confirm that the serine substitution sites showed the same effect in the substitution of other amino acids, a modified antibody was substituted with various amino acids at this position.
M2b 서열에서 serine 자리에 다른 아미노산을 치환한 변형항체Modified antibody which substituted another amino acid at serine site in M2b sequence
변형항체Modified antibody C-말단의 금속이온 결합 모티프 서열C-terminal metal ion binding motif sequence
M2b 또는 M2b-SM2b or M2b-S ACGHAASGHA (서열번호 21)ACGHAASGHA (SEQ ID NO: 21)
M2b-AM2b-A ACGHAAAGHA (서열번호 33)ACGHAAAGHA (SEQ ID NO: 33)
M2b-TM2b-T ACGHAATGHA (서열번호 45)ACGHAATGHA (SEQ ID NO 45)
M2b-YM2b-Y ACGHAAYGHA (서열번호 57)ACGHAAYGHA (SEQ ID NO 57)
M2b-DM2b-D ACGHAADGHA (서열번호 69)ACGHAADGHA (SEQ ID NO: 69)
M2b-KM2b-K ACGHAAKGHA (서열번호 81)ACGHAAKGHA (SEQ ID NO: 81)
M2b-FM2b-F ACGHAAFGHA (서열번호 93)ACGHAAFGHA (SEQ ID NO: 93)
위의 표 7과 같은 C-말단의 서열을 갖는 변형항체를 FM2b 또는 FM2b-S에 도입하여, 각각 FM2b-A, FM2b-T, FM2b-Y, FM2b-D, FM2b-K, FM2b-F의 변형항체를 생산하였다.The modified antibodies having the C-terminal sequence as shown in Table 7 above were introduced into FM2b or FM2b-S, respectively, of FM2b-A, FM2b-T, FM2b-Y, FM2b-D, FM2b-K, and FM2b-F. Modified antibodies were produced.
실시예 10. FM2b 변형항체와 MC-vc-PAB-MMAE의 접합에 의한 항체-약물 접합체 생산Example 10 Antibody-Drug Conjugate Production by Conjugation of FM2b Modified Antibody to MC-vc-PAB-MMAE
본 발명에서는 MMAE와 실시예 9와 같이 생산한 FM2b-X (X = A, T, Y, D, K, 혹은 F) 변형항체를 접합시켜서 FM2b-X-MMAE 결합체를 제조하였다. 본 발명에서는 정제된 변형항체 1당량 당 환원제인 TCEP를 3 당량 가하여 4℃에서 30분간 반응시켜서 티올기를 환원 시킨 후, MC-vc-PAB-MMAE를 2.5 당량 첨가하여 상온에서 2시간 가량 반응 시킨다. 반응은 과량의 시스테인을 가하여 종결시키고, 과량의 MC-vc-PAB-MMAE와 TCEP는 원심분리 여과 필터와 인산염 완충용액에서의 투석을 통하여 제거하여 최종 정제된 FMb-X-MC-vc-PAB-MMAE를 제조하였다. 각 변형항체의 중쇄로의 접합 수율은 아래의 표 8과 같다.In the present invention, FM2b-X-MMAE conjugate was prepared by conjugating MMAE with FM2b-X (X = A, T, Y, D, K, or F) modified antibodies produced as in Example 9. In the present invention, after adding 3 equivalents of TCEP, a reducing agent per equivalent of purified antibody, and reacting at 4 ° C. for 30 minutes to reduce thiol groups, 2.5 equivalents of MC-vc-PAB-MMAE are added and reacted at room temperature for 2 hours. The reaction was terminated by the addition of excess cysteine and excess MC-vc-PAB-MMAE and TCEP were removed via dialysis in a centrifugal filtration filter and phosphate buffer solution to obtain the final purified FMb-X-MC-vc-PAB- MMAE was prepared. The yield of conjugation to the heavy chain of each modified antibody is shown in Table 8 below.
FM2b-X 변형항체와 MC-vc-PAB-MMAE 접합시 DAR2 수율DAR2 yield when conjugated with FM2b-X modified antibody and MC-vc-PAB-MMAE
변형항체Modified antibody FM2bFM2b FM2b-AFM2b-A FM2b-TFM2b-T FM2b-YFM2b-Y FM2b-DFM2b-D FM2b-KFM2b-K FM2b-FFM2b-F
접합수율Junction yield 72.8%72.8% 62.5%62.5% 69%69% 74.5%74.5% 58.9%58.9% 75.3%75.3% 72.9%72.9%
위의 표 8에서 볼 수 있는 바와 같이, M2b 서열인 ACGHAASGHA의 뒤쪽 serine 자리에 다른 아미노산으로 치환하여도 FM2b와 거의 유사한 접합 결과가 나옴을 알 수 있다.As can be seen in Table 8 above, substitution of other amino acids in the rear serine site of the ACGHAASGHA, which is the M2b sequence, shows a similar result to that of FM2b.
실시예 11. DAR2인 항체-약물 접합체 정제Example 11. Purification of Antibody-Drug Conjugates That Are DAR2
위의 실시예 4에서와 같이 제조된 각 변형항체-약물 접합체는 변형항체당 접합된 약물의 개수(DAR: drug-to-antibody ratio)가 상이하기 때문에, in vitro에서 약물의 세포독성을 비교하기가 용이하지 않다. 따라서, 같은 DAR을 갖도록 변형항체-약물 접합체를 정제하기 위하여 소수성 크로마토그래피를 이용하여 변형항체-약물 접합체를 정제하였다. 페닐(phenyl) 컬럼 크로마토 그래피를 이용하여 DAR이 2를 갖는 변형항체-약물 접합체를 정제하였다. 컬럼을 10mM sodium succinate, 0.5M NaCl, pH 5.0 완충용액으로 평형화 시킨 후, 변형항체-약물 접합체를 컬럼에 주입하였다. 동일한 완충용액으로 컬럼을 세척한 후, 30%의 아세토나이트릴(acetonitrile)이 포함된 상기 완충용액을 가하여 DAR에 따라서 변형항체-약물 접합체를 용출하였다. 용출된 변형항체-약물 접합체는 10mM sodium succinate, 30mM sucrose, pH 6.0의 완충용액에서 투석(dialysis)을 이용하여 완충용액을 교환하였다.Each modified antibody prepared as in the above embodiment four-drug conjugate is modified number of the bonding substance (DAR: drug-to-antibody ratio) per antibody because it is different, to compare the cytotoxicity of the drug from in vitro Is not easy. Therefore, in order to purify the modified antibody-drug conjugate with the same DAR, the modified antibody-drug conjugate was purified using hydrophobic chromatography. Phenyl column chromatography was used to purify the modified antibody-drug conjugate having a DAR of 2. The column was equilibrated with 10 mM sodium succinate, 0.5 M NaCl, pH 5.0 buffer, and then the modified antibody-drug conjugate was injected into the column. After washing the column with the same buffer, the buffer containing 30% acetonitrile was added to elute the modified antibody-drug conjugate according to DAR. The eluted modified antibody-drug conjugates were exchanged for buffer using dialysis in a buffer solution of 10 mM sodium succinate, 30 mM sucrose, pH 6.0.
실시예 12. 시험관 내 항체-약물 접합체의 안정성 시험Example 12. Stability Testing of In Vitro Antibody-Drug Conjugates
위의 실시예 9, 10, 11과 같이 MC-vc-PAB-MMAE 약물이 2개 접합된 항체-약물 접합체인 FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y-D2를 생산하였다. 생산된 항체-약물 접합체를 각각, 25℃와 50℃의 온도 조건에서 incubation하여, 약물 접합 개수의 변화와 aggregation의 변화를 측정하였다.FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y, which are antibody-drug conjugates having two conjugated MC-vc-PAB-MMAE drugs as in Examples 9, 10, and 11 above Produced -D2. The produced antibody-drug conjugates were incubated at 25 ° C. and 50 ° C., respectively, to measure the change in the number of drug conjugates and the change in aggregation.
각 시험 시료는 1mg/mL의 농도로 110uL의 농도로, 각각 12개씩을 준비하였다. 각 항체-약물 접합체의 시료 중 6개의 시료는 25℃, 나머지 6개의 시료는 60℃ 조건에서 0, 1, 3, 5, 7, 14 일간 보관하여 DAR과 monomer의 변화를 측정하였다. 25℃에서 DAR2의 함량은 각 시료별로 1.5~2% 정도의 감소가 관찰되었고, monomer의 순도는 0.3~4% 정도의 감소가 관찰되었으나, 각 시료별 차이는 그리 크지 않았다. 50℃에서의 DAR2의 함량과 monomer 순도의 변화는 25℃에서 관찰된 값보다는 크게 관찰되었으나, 시료별 차이는 그리 크지 않았다. 따라서 각 항체 변이체 사이의 차이는 그리 크지 않음을 확인할 수 있었다.Each test sample was prepared at a concentration of 110 uL at a concentration of 1 mg / mL, each of 12. Six samples of each antibody-drug conjugate were stored at 25 ° C. and the remaining six samples were kept at 0 ° C. for 1, 3, 5, 7, and 14 days to measure changes in DAR and monomer. At 25 ° C, the content of DAR2 was decreased by 1.5 ~ 2% for each sample, and the purity of monomer was observed about 0.3 ~ 4%, but the difference was not large. Changes in DAR2 content and monomer purity at 50 ° C were larger than those observed at 25 ° C, but the differences between samples were not significant. Therefore, it was confirmed that the difference between the antibody variants is not very large.
FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y-D2의 25℃와 50℃의 보관조건에서 0, 1, 3, 5, 7, 15일차의 모노머의 함량Contents of monomers on Days 0, 1, 3, 5, 7, and 15 at 25 ° C and 50 ° C storage conditions of FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, and FM2b-Y-D2
보관온도Storage temperature 시료sample Day 0Day 0 Day 1 Day 1 Day 3 Day 3 Day 5 Day 5 Day 7Day 7 Day 15 Day 15
25℃25 ℃ FM2b-S-D2FM2b-S-D2 97.697.6 97.697.6 97.297.2 96.496.4 95.695.6 95.195.1
FM2b-F-D2FM2b-F-D2 9797 97.897.8 97.297.2 96.696.6 96.196.1 9595
FM2b-K-D2FM2b-K-D2 97.697.6 97.697.6 97.397.3 96.696.6 96.196.1 96.396.3
FM2b-Y-D2FM2b-Y-D2 96.796.7 96.696.6 9797 95.895.8 95.495.4 94.994.9
50℃50 ℃ FM2b-S-D2FM2b-S-D2 97.697.6 93.993.9 88.888.8 82.882.8 79.179.1 68.968.9
FM2b-F-D2FM2b-F-D2 9797 93.793.7 86.986.9 80.980.9 76.976.9 63.563.5
FM2b-K-D2FM2b-K-D2 97.697.6 93.993.9 87.787.7 81.881.8 77.877.8 66.866.8
FM2b-Y-D2FM2b-Y-D2 96.796.7 93.993.9 88.488.4 82.782.7 77.977.9 66.666.6
FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, FM2b-Y-D2의 25℃와 50℃의 보관조건에서 0, 1, 3, 5, 7, 15일차의 DAR 2의 함량Contents of DAR 2 on Days 0, 1, 3, 5, 7, and 15 at 25 ° C and 50 ° C storage conditions of FM2b-S-D2, FM2b-F-D2, FM2b-K-D2, and FM2b-Y-D2
보관온도Storage temperature DAYDAY Day 0Day 0 Day 1 Day 1 Day 3 Day 3 Day 5 Day 5 Day 7Day 7 Day 15 Day 15
25℃25 ℃ FM2b-S-D2FM2b-S-D2 98.998.9 98.698.6 98.698.6 98.298.2 98.498.4 98.198.1
FM2b-F-D2FM2b-F-D2 98.298.2 98.498.4 98.498.4 98.298.2 98.398.3 97.997.9
FM2b-K-D2FM2b-K-D2 97.997.9 97.497.4 97.597.5 97.397.3 97.397.3 9797
FM2b-Y-D2FM2b-Y-D2 97.697.6 97.697.6 97.497.4 96.696.6 95.595.5 93.293.2
50℃50 ℃ FM2b-S-D2FM2b-S-D2 98.998.9 97.197.1 93.693.6 91.891.8 90.790.7 87.987.9
FM2b-F-D2FM2b-F-D2 98.298.2 96.596.5 93.293.2 89.489.4 88.388.3 85.185.1
FM2b-K-D2FM2b-K-D2 97.997.9 96.296.2 94.694.6 91.891.8 90.890.8 88.988.9
FM2b-Y-D2FM2b-Y-D2 97.697.6 95.895.8 91.391.3 89.489.4 87.687.6 84.484.4
실시예 13. 시험관 내 세포증식억제능 시험Example 13. In Vitro Cell Proliferation Inhibition Test
변형항체-약물 접합체의 시험관내 세포 증식억제능을 비교하기 위하여 folate receptor가 과발현된 KB-세포를 이용하여 세포성장억제능 시험을 수행하였다. KB-세포는 10% FBS가 첨가된 DMEM/F12 배지에 희석하여 1×104개/웰(well)이 되도록 조정한 후 100㎕ 세포 배양물을 96-웰(well) 플레이트의 각 웰에 가하였다. 이후 웰 플레이트를 5% 이산화탄소 및 37℃로 설정된 배양기에서 24시간 동안 배양하여 세포를 플레이트에 부착시켰다. 각 시험 시료를 배지에 희석한 후 최종 농도 6.45nM, 3.23nM, 1.61nM, 0.806nM, 0.403nM, 0.202nM, 0.101nM, 0.0504nM, 0.0252nM 및 0.0126nM이 되게 첨가하였으며, 함께 대조군 웰에는 배지만(약물 없음) 첨가하였다. 5일 동안 배양 후에, 20㎕/웰로 CellTiter 96- AQueous One Solution 시약 [MTS-기초 검정; 살아있는 세포의 디하이드로게나제(dehydrogenase)에 의해 MTS가 보라색 포마잔(formazan)을 형성하며, 생성된 보라색 포마잔의 양에 의해 증식 측정]을 첨가한 후 37℃로 설정된 배양기에서 2시간 동안 배양 하였다. 세포 용균을 흡광분석기로 O.D. 490nm에서 측정하여 viability(%)를 측정하였다. In order to compare the in vitro cell proliferation inhibitory activity of the modified antibody-drug conjugates, cell growth inhibitory activity was performed using KB-cells overexpressed with the folate receptor. KB-cells were diluted in DMEM / F12 medium with 10% FBS adjusted to 1 × 10 4 / well and 100 μl cell culture was added to each well of a 96-well plate. It was. The well plates were then incubated for 24 hours in an incubator set at 5% carbon dioxide and 37 ° C. to attach the cells to the plates. Each test sample was diluted in medium and added to final concentrations of 6.45 nM, 3.23 nM, 1.61 nM, 0.806 nM, 0.403 nM, 0.202 nM, 0.101 nM, 0.0504 nM, 0.0252 nM and 0.0126 nM, together with the medium in the control wells. (No drug) was added. After incubation for 5 days, CellTiter 96- AQueous One Solution reagent [MTS-based assay; MTS forms purple formazan by dehydrogenase of living cells, and proliferation is measured by the amount of purple formazan produced], followed by incubation for 2 hours in an incubator set at 37 ° C. It was. Cell lysis was measured at 490 nm using an absorbance spectrometer to determine viability (%).
13-1. FM2-D2와 FM2b-D2 (or FM2b-S-D2)의 세포성장억제능 비교 시험:13-1. Comparison of cell growth inhibitory activity between FM2-D2 and FM2b-D2 (or FM2b-S-D2):
모항체인 Fwt와 상기 예에서 MMAE를 접합 시킨 후, 동일한 DAR을 갖도록 정제한 변형항체-약물 접합체인 FM2-D2 (변형항체인 FM2의 MMAE 약물접합체로써 DAR 2를 갖는 변형항체-약물 접합체)와 FM2b-D2 (변형항체인 FM2b의 MMAE 약물접합체로써 DAR 2를 갖는 변형항체-약물 접합체)를 위와 같이 KB cell에 처리한 후, 약물의 세포성장 억제능을 비교하였다.FM2-D2 (modified antibody-drug conjugate having DAR 2 as MMAE drug conjugate of modified antibody FM2) and FM2b purified after conjugation of the parent antibody Fwt with MMAE in the above example After treating -D2 (modified antibody-drug conjugate having DAR 2 as MMAE drug conjugate of modified antibody FM2b) to KB cells as described above, the cell growth inhibition of the drug was compared.
도 3에서 볼 수 있는 바와 같이, 모항체에 비하여 항체-약물 접합체인 FM2-D2와 FM2b-D2는 월등히 우월한 항암 효능을 보여주고 있다. FM2-D2와 FM2b-D2는 거의 동일한 암세포 성장 억제능을 보여주고 있다. 이와 같은 결과는 변형항체인 FM2와 FM2b 사이에는 변형항체-약물 접합체로써 같은 DAR을 가지고 있을 때, 암세포 성장 억제능의 활성 차이가 없다는 것을 보여주고 있다. As can be seen in Figure 3, the antibody-drug conjugates FM2-D2 and FM2b-D2 show superior anticancer efficacy compared to the parent antibody. FM2-D2 and FM2b-D2 show nearly identical cancer cell growth inhibition. These results show that there is no difference in activity of inhibiting cancer cell growth when the same antibody DAR is used as a modified antibody-drug conjugate between the modified antibodies FM2 and FM2b.
13-2. FM2-D2와 FM2b-D2 (or FM2b-S-D2)의 세포성장억제능 비교 시험:13-2. Comparison of cell growth inhibitory activity between FM2-D2 and FM2b-D2 (or FM2b-S-D2):
FM2b-S와 다른 변이체에 기반한 항체-약물 접합체간의 세포성장 억제능을 비교하기 위하여, FM2b-S, -F, -Y 변이체에 MC-vc-PAB-MMAE를 접합 시킨 후, 동일한 DAR을 갖도록 정제한 후, 위와 같이 KB cell에 처리한 후, 약물의 세포성장 억제능을 비교하였다.To compare cell growth inhibition between FM2b-S and other variants-based antibody-drug conjugates, MC-vc-PAB-MMAE was conjugated to FM2b-S, -F, and -Y variants and purified to have the same DAR. Then, after treating the KB cells as described above, the cell growth inhibition of the drug was compared.
도 4에서 볼 수 있는 바와 같이, FM2b-S-D2와 FM2b-F-D2, FM2b-Y-D2는 거의 동일한 세포성장 억제능을 보여주고 있다. 각각의 IC50값은 FM2b-S-D2는 0.25nM, FM2b-F-D2는 0.26nM, FM2b-Y-D2는 0.24nM이 측정되었다. 이와 같은 결과는 FM2b-S의 serine 대신에 각각 phenylalanine, tyrosine으로 치환한 항체 변이체에 기반한 항체-약물 접합체 사이에는 암세포 성장 억제능의 활성 차이가 없다는 것을 보여주고 있다.As can be seen in Figure 4, FM2b-S-D2 and FM2b-F-D2, FM2b-Y-D2 shows almost the same cell growth inhibition. The respective IC 50 values were 0.25 nM for FM2b-S-D2, 0.26 nM for FM2b-F-D2, and 0.24 nM for FM2b-Y-D2. These results show that there is no difference in the activity of inhibiting cancer cell growth between antibody-drug conjugates based on antibody variants substituted with phenylalanine and tyrosine instead of the serine of FM2b-S.
이와 같은 결과에서 볼 때, M2b 서열을 도입한 항체를 이용하여 항체-약물 접합체로 제조되었을 때, 다른 변형 항체와 비교하여 월등히 우월한 약물의 접합 수율을 가지고 있으면서 항체-약물 접합체로써의 항암 활성에서는 차이가 없다는 점을 보여주고 있다. 또한, M2b서열인 ACGHA-ASGHA에서 serine자리에 다른 아미노산이 치환되어도 약물의 접합 수율이나 안정성, 항암 활성에도 차이가 없음을 확인할 수 있었다. 따라서, FM2b나 HM2b에 사용된 바와 같은 모티프인 ACGHA-AXGHA(X는 시스테인을 제외한 다른 아미노산)를 항체의 중쇄 말단에 도입할 때, 여타 다른 변형항체에 비하여 월등히 우월한 접합 수율을 가짐으로써, 보다 효율적이며 경제적인 항체-약물 접합체를 생산할 수 있음을 의미한다.From these results, when the antibody-drug conjugate was prepared by using the antibody to which the M2b sequence was introduced, there was a difference in the anti-cancer activity as the antibody-drug conjugate while having a superior conjugated yield of drugs compared to other modified antibodies. Shows that there is no. In addition, even if the amino acid is substituted in the serine site in the ACGHA-ASGHA M2b sequence, it was confirmed that there is no difference in the drug yield, stability, and anticancer activity. Therefore, when introducing ACGHA-AXGHA (X is an amino acid other than cysteine), a motif as used in FM2b or HM2b, the conjugate yield is superior to other modified antibodies, thereby providing a more efficient conjugation yield. And economic antibody-drug conjugates.
본 발명에 따른 항체 변이체에 의해 생산되는 항체-약물 접합체는 약물의 접합 수율을 높임으로써 항체-약물 접합체의 생산성을 크게 향상시킬 수 있다. 또한, 위치 특이적 접합을 통해서 항체의 말단 부분에 접합한 약물은 모항체의 구조적 안정성을 저해하지 않으므로, 모항체가 원래 가지고 있는 항원 특이성과 구조적 안정성을 유지할 수 있으며, 모항체가 가지고 있는 높은 항원 특이성으로 인해서 항체에 접합된 약물을 암세포에 특이적으로 운반할 수 있다. The antibody-drug conjugate produced by the antibody variant according to the present invention can greatly improve the productivity of the antibody-drug conjugate by increasing the conjugation yield of the drug. In addition, the drug conjugated to the terminal portion of the antibody through position-specific conjugation does not inhibit the structural stability of the parent antibody, thereby maintaining the antigen specificity and structural stability of the parent antibody originally, and the high antigen possessed by the parent antibody. Specificity allows the drug conjugated to the antibody to be specifically delivered to cancer cells.
이상으로 본 발명의 내용의 특정한 부분을 상세히 기술하였는바, 당업계의 통상의 지식을 가진 자에게 있어서, 이러한 구체적 기술은 단지 바람직한 실시양태일 뿐이며, 이에 의해 본 발명의 범위가 제한되는 것이 아닌 점은 명백할 것이다. 따라서, 본 발명의 실질적인 범위는 첨부된 청구항들과 그것들의 등가물에 의하여 정의된다고 할 것이다.As described above in detail a specific part of the content of the present invention, for those skilled in the art, such a specific description is only a preferred embodiment, which is not limited by the scope of the present invention Will be obvious. Thus, the substantial scope of the present invention will be defined by the appended claims and their equivalents.
전자파일 첨부하였음.Electronic file attached.

Claims (14)

  1. 하기 구조식 (1)로 표시되는 모티프를 항체의 말단에 포함하는 변형항체가 링커를 통해 약물에 결합된 항체-약물 접합체:An antibody-drug conjugate wherein a modified antibody comprising a motif represented by the following structural formula (1) at the terminal of the antibody is bound to a drug via a linker:
    구조식 (1)Structural Formula (1)
    Xa-[Mmotif1]n1-Xb-[Mmotif2]n2 X a - [M motif1] n1 -X b - [M motif2] n2
    상기 식에서, Mmotif1 또는 Mmotif2는 각각 독립적으로 ACGHA (서열번호 1), AHGCA (서열번호 2), AXGHA (서열번호 3) 및 AHGXA (서열번호 4)로 구성된 서열 중 어느 하나를 포함하고, 상기 서열번호 3 또는 4에서 X는 시스테인 이외의 아미노산 잔기를 포함하며, Wherein M motif1 or M motif2 each independently include any one of a sequence consisting of ACGHA (SEQ ID NO: 1), AHGCA (SEQ ID NO: 2), AXGHA (SEQ ID NO: 3), and AHGXA (SEQ ID NO: 4), wherein X in SEQ ID NO: 3 or 4 includes amino acid residues other than cysteine,
    Xa 및 Xb는 각각 독립적으로 A (alanine), S (serine), G (glycine)로 이루어진 군에서 선택된 아미노산 잔기가 0개 내지 20개로 구성된 펩타이드이며,X a And X b is a peptide consisting of 0 to 20 amino acid residues each independently selected from the group consisting of A (alanine), S (serine), G (glycine),
    n1 및 n2는 각각 1 내지 10의 정수이다. n1 and n2 are integers of 1-10, respectively.
  2. 제1항에 있어서, 상기 Mmotif2는 AXGHA (서열번호 3)을 포함하고, X는 시스테인 이외의 아미노산 잔기 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1 wherein the M motif2 comprises AXGHA (SEQ ID NO: 3) and X is an amino acid residue other than cysteine.
  3. 제1항에 있어서, 상기 Mmotif1 또는 Mmotif2의 X는 세린 (S), 글리신 (G), 알라닌 (A), 트레오닌 (T), 타이로신 (Y), 아스파르트산(D), 라이신 (K) 및 페닐알라닌 (F)으로 구성된 군에서 선택된 아미노산 잔기인 것을 특징으로 하는 항체-약물 접합체.The method of claim 1, wherein X of M motif 1 or M motif 2 is serine (S), glycine (G), alanine (A), threonine (T), tyrosine (Y), aspartic acid (D), lysine (K) And phenylalanine (F). An antibody-drug conjugate, characterized in that the amino acid residue is selected from the group consisting of.
  4. 제1항에 있어서, 상기 구조식 (1)로 표시되는 모티프는 서열번호 5 내지 104로 구성된 군에서 선택된 하나 이상의 서열을 포함하는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1, wherein the motif represented by the structural formula (1) comprises at least one sequence selected from the group consisting of SEQ ID NOs: 5 to 104.
  5. 제1항에 있어서, 상기 모티프는 항체의 중쇄 C-말단에 도입되는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1, wherein the motif is introduced at the heavy chain C-terminus of the antibody.
  6. 제1항에 있어서, 상기 링커는 모티프에 결합하는 반응성 기능기, 아미노산 및 자가 절단 스페이서를 포함하는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1 wherein the linker comprises a reactive functional group that binds to a motif, an amino acid, and a self cleaving spacer.
  7. 제1항에 있어서, 상기 약물은 마이탄시노이드, 오리스타틴, 아미노프테린, 악티노마이신, 블레오마이신, 탈리소마이신, 캄프토쎄신, N8-아세틸 스퍼미딘, 1-(2 클로로에틸)-1,2-다이메틸 술포닐 하이드라자이드, 에스퍼라마이신, 에토포사이드, 6-머캅토퓨린, 돌라스타틴, 트리코테센, 칼리케아미신, 탁산, 메토트렉세이트, 빈크리스틴, 빈블라스틴, 독소루비신, 멜팔란, 미토마이신 A, 미토마이신 C, 클로람부실, 듀오카마이신, 핵산 분해 효소, 세균이나 동식물 유래의 독소, 시스플라틴, 이리노테칸, 파클리탁셀 및 도세탁셀에서 선택된 하나 이상인 것을 특징으로 하는 항체-약물 접합체.The method of claim 1, wherein the drug is maytansinoid, oristatin, aminopterin, actinomycin, bleomycin, thalisomycin, camptothecin, N8-acetyl spermidine, 1- (2 chloroethyl)- 1,2-dimethyl sulfonyl hydrazide, esperamycin, etoposide, 6-mercaptopurine, dolastatin, tricortesene, calicheamicin, taxane, methotrexate, vincristine, vinblastine, doxorubicin, melphalan Antibody-drug conjugate, characterized in that at least one selected from, mitomycin A, mitomycin C, chlorambucil, duocarmycin, nucleases, toxins derived from bacteria or plants, cisplatin, irinotecan, paclitaxel and docetaxel.
  8. 제1항에 있어서, 상기 항체는 모노클로날 항체, 이중특이적 항체, 키메릭 항체, 인간 항체 및 인간화 항체로 구성된 군에서 선택된 하나 이상인 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1, wherein the antibody is at least one selected from the group consisting of monoclonal antibodies, bispecific antibodies, chimeric antibodies, human antibodies and humanized antibodies.
  9. 제1항에 있어서, 상기 항체는 IgA, IgD, IgE, IgG 및 IgM으로 구성된 군에서 선택되는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1 wherein the antibody is selected from the group consisting of IgA, IgD, IgE, IgG and IgM.
  10. 제1항에 있어서, 상기 항체는 암 특이 항원, 세포 표면 수용체 단백질, 세포 표면 단백질, 막횡단 단백질, 신호전달 단백질, 세포생존 조절인자, 세포 증식 조절인자, 조직 발달 또는 분화와 연관된 분자, 림포카인, 사이토카인, 세포 주기 조절에 관련된 분자, 혈관형성에 관련된 분자, 또는 혈관신생에 관련된 분자에 대한 결합능과 특이성을 가지는 것을 특징으로 하는 항체-약물 접합체.The method of claim 1, wherein the antibody is a cancer specific antigen, cell surface receptor protein, cell surface protein, transmembrane protein, signaling protein, cell survival regulator, cell proliferation regulator, molecule associated with tissue development or differentiation, lymphokah An antibody-drug conjugate, having binding capacity and specificity for phosphorus, cytokines, molecules involved in cell cycle regulation, molecules involved in angiogenesis, or molecules involved in angiogenesis.
  11. 제1항에 있어서, 상기 항체는,The method of claim 1, wherein the antibody,
    (1) BMPR1B (골 형태형성 단백질 수용체-IB형, 진뱅크 승인 번호 NM_001203);(1) BMPR1B (bone morphogenic protein receptor-IB type, Genbank Accession No. NM_001203);
    (2) E16 (LAT1, SLC7A5, 진뱅크 승인 번호 NM_003486);(2) E16 (LAT1, SLC7A5, GenBank Accession No. NM_003486);
    (3) STEAP1 (전립선의 6회의 막횡단 상피 항원, 진뱅크 승인 번호 NM_012449);(3) STEAP1 (six transmembrane epithelial antigens of the prostate, Genbank Accession No. NM_012449);
    (4) 0772P (CA125, MUC16, 진뱅크 승인 번호 AF361486);(4) 0772P (CA125, MUC16, GenBank Accession No. AF361486);
    (5) MPF (MPF, MSLN, SMR, 거핵세포 강화 인자, 메소텔린, 진뱅크 승인 번호 NM_005823);(5) MPF (MPF, MSLN, SMR, megakaryocyte enhancing factor, mesothelin, Genbank Accession No. NM — 005823);
    (6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, 용질 운반체 족 34 (인산나트륨), 구성원 2, 제II형 나트륨-의존성 포스페이트 수송체 3b, 진뱅크 승인 번호 NM_006424);(6) Napi3b (NAPI-3B, NPTIIb, SLC34A2, Solute Carrier Family 34 (Sodium Phosphate), Member 2, Type II Sodium-Dependent Phosphate Transporter 3b, GenBank Accession No. NM_006424);
    (7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, 세마포린 5b Hlog, 세마 도메인, 7개의 트롬보스폰딘 반복체 (제1형 및 유사 제1형), 막횡단 도메인 (TM) 및 짧은 세포질 도메인, (세마포린) 5B, 진뱅크 승인 번호 AB040878);(7) Sema 5b (FLJ10372, KIAA1445, Mm.42015, SEMA5B, SEMAG, Semaphorin 5b Hlog, Sema Domain, 7 Thrombospondin Repeats (Type 1 and Similar Type 1), Transmembrane Domain (TM) And short cytoplasmic domain, (semaphorin) 5B, Genbank Accession No. AB040878);
    (8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 유전자, 진뱅크 승인 번호 AY358628);(8) PSCA hlg (2700050C12Rik, C530008O16Rik, RIKEN cDNA 2700050C12, RIKEN cDNA 2700050C12 gene, Genebank Accession No. AY358628);
    (9) ETBR (엔도텔린 B형 수용체, 진뱅크 승인 번호 AY275463);(9) ETBR (endothelin type B receptor, Genbank Accession No. AY275463);
    (10) MSG783 (RNF124, 가상 단백질 FLJ20315, 진뱅크 승인 번호 NM_017763);(10) MSG783 (RNF124, hypothetical protein FLJ20315, Genebank Accession No. NM_017763);
    (11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, 전립선암 관련 유전자 1, 전립선암 관련단백질 1, 전립선의 6회의 막횡단 상피 항원 2, 6회의 막횡단 전립선 단백질, 진뱅크 승인 번호 AF455138);(11) STEAP2 (HGNC_8639, IPCA-1, PCANAP1, STAMP1, STEAP2, STMP, Prostate Cancer Related Gene 1, Prostate Cancer Related Protein 1, Prostate 6 Transmembrane Epithelial Antigen 2, 6 Transmembrane Prostate Protein, Genebank Authorization number AF455138);
    (12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, 일시적 수용체 잠재적 양이온 채널, M 아족, 구성원 4, 진뱅크 승인 번호 NM_017636);(12) TrpM4 (BR22450, FLJ20041, TRPM4, TRPM4B, transient receptor potential cation channel, subgroup M, member 4, Genbank Accession No. NM_017636);
    (13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, 기형암종-유래 성장인자, 진뱅크 승인 번호 NP_003203 또는 NM_003212);(13) CRIPTO (CR, CR1, CRGF, CRIPTO, TDGF1, teratocarcinoma-derived growth factor, Genbank accession no. NP — 003203 or NM — 003212);
    (14) CD21 (CR2 (보체 수용체 2) 또는 C3DR (C3d/엡스타인 바르 바이러스 수용체) 또는 Hs.73792 진뱅크 승인 번호 M26004);(14) CD21 (CR2 (complementary receptor 2) or C3DR (C3d / Epstein Barr virus receptor) or Hs.73792 Genbank Accession No. M26004);
    (15) CD79b (CD79B, CD79β, IGb (이뮤노글로불린-관련 베타), B29, 진뱅크 승인 번호 NM_000626);(15) CD79b (CD79B, CD79β, IGb (immunoglobulin-associated beta), B29, Genbank Accession No. NM — 000626);
    (16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 도메인 함유 포스파타제 고정 단백질 1a), SPAP1B, SPAP1C, 진뱅크 승인 번호 NM_030764);(16) FcRH2 (IFGP4, IRTA4, SPAP1A (SH2 domain containing phosphatase anchoring protein 1a), SPAP1B, SPAP1C, GenBank Accession No. NM_030764);
    (17) HER2 (진뱅크 승인 번호 M11730)17 HER2 (Genbank approval number M11730)
    (18) EGFR, HER3 및 HER4로부터 선택된 ErbB 수용체(18) ErbB receptors selected from EGFR, HER3 and HER4
    (19) NCA (진뱅크 승인 번호 M18728);(19) NCA (Genbank Accession No. M18728);
    (20) MDP (진뱅크 승인 번호 BC017023);(20) MDP (GenBank Accession No. BC017023);
    (21) IL20Rα (진뱅크 승인 번호 AF184971);(21) IL20Rα (GenBank Accession No. AF184971);
    (22) 브레비칸 (진뱅크 승인 번호 AF229053);(22) Brevican (Genbank Accession No. AF229053);
    (23) EphB2R (진뱅크 승인 번호 NM_004442);(23) EphB2R (GenBank Accession No. NM_004442);
    (24) ASLG659 (진뱅크 승인 번호 AX092328);(24) ASLG659 (Genbank Accession No. AX092328);
    (25) PSCA (진뱅크 승인 번호 AJ297436);(25) PSCA (Genbank Accession No. AJ297436);
    (26) GEDA (진뱅크 승인 번호 AY260763);(26) GEDA (Genbank Accession No. AY260763);
    (27) BAFF-R (B 세포 활성화 인자 수용체, BLyS 수용체 3, BR3, NP_443177.1);(27) BAFF-R (B cell activating factor receptor, BLyS receptor 3, BR3, NP_443177.1);
    (28) CD22 (B-세포 수용체 CD22-B 이소형, NP-001762.1);(28) CD22 (B-cell receptor CD22-B isotype, NP-001762.1);
    (29) CD79a (Ig 베타 (CD79B)와 공유적으로 상호작용하고 IgM 분자와 표면에서 복합체를 형성하는 B 세포 특이적 단백질인 CD79A, CD79α, 이뮤노글로불린-관련 알파는 B 세포 분화에 관여하는 신호를 전달함, 진뱅크 승인 번호 NP_001774.1);(29) CD79a, CD79A, CD79α, and immunoglobulin-associated alpha, which are covalently interacting with CD79a (Ig beta (CD79B) and forming complexes on the surface with IgM molecules, are signals involved in B cell differentiation Forwarded, Genbank approval number NP_001774.1);
    (30) CXCR5 (CXCL13 케모킨에 의해 활성화된 G 단백질 커플링된 수용체인 버킷 림프종 수용체 1은 림프구 이동 및 체액성 방어에 작용하고 HIV-2 감염에 참여하며, AIDS, 림프종, 골수종 및 백혈병의 발병과 관련이 있다고 여겨짐, 진뱅크 승인 번호 NP_001707.1);(30) CXCR5 (Bucket Lymphoma Receptor 1, a G protein coupled receptor activated by CXCL13 chemokines, acts on lymphocyte migration and humoral defense, participates in HIV-2 infection, and develops AIDS, lymphoma, myeloma and leukemia Considered to be related to, Genbank approval number NP_001707.1);
    (31) HLA-DOB (펩티드에 결합하여 CD4+ T 림프구에 제시하는, MHC 클래스 II 분자 (Ia 항원)의 베타 서브유닛, 진뱅크 승인 번호 NP_002111.1);(31) HLA-DOB (beta subunit of MHC class II molecules (Ia antigen), binding to peptides and presenting in CD4 + T lymphocytes, Genbank Accession No. NP — 002111.1);
    (32) P2X5 (세포외 ATP에 의해 게이트되는 이온 채널인, 퓨린성 수용체 P2X 리간드-게이트 이온 채널 5는 시냅스 전달 및 신경발생에 관여할 수 있으며, 이의 결핍은 특발성 배뇨근 불안정의 병태생리에 기여할 수 있음, 진뱅크 승인 번호 NP_002552.2);(32) P2X5 (purine receptor P2X ligand-gate ion channel 5, an ion channel gated by extracellular ATP, may be involved in synaptic transmission and neurogenesis, the lack of which may contribute to the pathophysiology of idiopathic detrusor instability Yes, Genbank approval number NP_002552.2);
    (33) CD72 (B-세포 분화 항원 CD72, Lyb-2, 진뱅크 승인 번호 NP_001773.1);(33) CD72 (B-cell differentiation antigen CD72, Lyb-2, Genbank Accession No. NP — 001773.1);
    (34) LY64 (루이신 풍부 반복체 (LRR) 족의 제I형 막 단백질인, 림프구 항원 64 (RP105)는 B 세포 활성화 및 세포자멸을 조절하며, 이것의 기능 상실은 전신성 홍반성 루푸스 환자의 질병 활성 증가와 관련이 있음, 진뱅크 승인 번호 NP_005573.1);(34) Lymphocyte antigen 64 (RP105), a type I membrane protein of the LY64 (leucine rich repeat (LRR) family), modulates B cell activation and apoptosis, and its loss of function is attributed to systemic lupus erythematosus patients. Associated with increased disease activity, GenBank Accession No. NP_005573.1);
    (35) FcRH1 (C2형 Ig-유사 및 ITAM 도메인을 함유하는 이뮤노글로불린 Fc 도메인에 대한 추정적 수용체인 Fc 수용체 유사 단백질 1은 B 림프구 분화에 관여할 수 있음, 진뱅크 승인 번호 NP_443170.1);(35) FcRH1 (Fc receptor-like protein 1, a putative receptor for immunoglobulin Fc domains containing C2 Ig-like and ITAM domains, may be involved in B lymphocyte differentiation, Genbank accession number NP_443170.1) ;
    (36) IRTA2 (B 세포 발생 및 림프종발생에 작용할 수 있는 추정적 면역수용체인 이뮤노글로불린 거대족 수용체 전좌 관련 2, 전좌에 의한 상기 유전자 탈조절은 몇몇 B 세포 악성종양에서 일어남, 진뱅크 승인 번호 NP_112571.1); (36) IRTA2 (associated gene deregulation by immunoglobulin macrophage receptor translocation, a putative immunoreceptor capable of acting on B cell development and lymphoma development, occurs in some B cell malignancies, Genbank approval number NP_112571.1);
    (37) TENB2 (성장 인자의 EGF/헤레굴린 족 및 폴리스타틴과 관련이 있는 추정적 막횡단 프로테오글리칸, 진뱅크 승인 번호 AF179274);(37) TENB2 (estimated transmembrane proteoglycan associated with EGF / Heregulin family of growth factors and follistatin, Genbank Accession No. AF179274);
    (38) MAGE-C1/CT7 (고환암 과발현 단백질);(38) MAGE-C1 / CT7 (testicular cancer overexpressed protein);
    (39) androgen receptor, PTEN, human kallikrein-related peptidase 3 (전립선암에서 과발현되는 단백질);(39) androgen receptor, PTEN, human kallikrein-related peptidase 3 (protein overexpressed in prostate cancer);
    (40) CD20;(40) CD20;
    (41) CD30;(41) CD30;
    (42) CD33;(42) CD33;
    (43) CD52;(43) CD52;
    (44) EpCam;(44) EpCam;
    (45) CEA;(45) CEA;
    (46) gpA33;(46) gpA33;
    (47) Mucins;(47) Mucins;
    (48) TAG-72;(48) TAG-72;
    (49) Carbonic anhydrase IX;(49) Carbonic anhydrase IX;
    (50) PSMA;(50) PSMA;
    (51) folate receptor (FOLR gene에 의해서 발현되는 단백질 패밀리. Folic acid와 높은 결합력을 가지고 있으며, 5-methyltetrahydrofolate를 세포 내로 운반한다);(51) folate receptor (a family of proteins expressed by the FoLR gene, which has high binding capacity with Folic acid and carries 5-methyltetrahydrofolate into cells);
    (52) gangliosides (GD2, GD3, GM2);(52) gangliosides (GD2, GD3, GM2);
    (53) 당수화물 Lewis-Y;(53) carbohydrate Lewis-Y;
    (54) VEGF;(54) VEGF;
    (55) VEGFR;(55) VEGFR;
    (56) aVb3;(56) aVb3;
    (57) a5b1;(57) a5b1;
    (58) ERB3;(58) ERB3;
    (59) c-MET;(59) c-MET;
    (60) EphA3;(60) EphA3;
    (61) TRAIL-R1, TRAIL-R2;(61) TRAIL-R1, TRAIL-R2;
    (62) RANKL;(62) RANKL;
    (63) FAP; 및(63) FAP; And
    (64) Tenascin로 구성된 군에서 선택된 하나 이상의 타겟에 결합능을 가지는 것을 특징으로 하는 항체-약물 접합체.(64) An antibody-drug conjugate, having an ability to bind to one or more targets selected from the group consisting of Tenascin.
  12. 제1항에 있어서, 상기 항체는 가변영역 및 IgG2 또는 IgG4의 CH1, CH2 및 CH3를 포함, 또는 Fab 및 IgG2 또는 IgG4의 Fc를 포함하는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1 wherein the antibody comprises a variable region and CH1, CH2 and CH3 of IgG2 or IgG4, or Fab and Fc of IgG2 or IgG4.
  13. 제1항에 있어서, 상기 약물은 모티프의 시스테인 또는 X에 접합되는 것을 특징으로 하는 항체-약물 접합체.The antibody-drug conjugate of claim 1 wherein the drug is conjugated to the cysteine or X of the motif.
  14. 제1항 내지 제13항 중 어느 한 항에 따른 항체-약물 접합체를 포함하는 암 예방 또는 치료용 조성물.14. A composition for preventing or treating cancer, comprising the antibody-drug conjugate according to any one of claims 1 to 13.
PCT/KR2017/003508 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody WO2017176007A1 (en)

Priority Applications (9)

Application Number Priority Date Filing Date Title
EP17779307.2A EP3441088B1 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody
EP22191369.2A EP4144377A1 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody
CN202210306570.8A CN114887074A (en) 2016-04-06 2017-03-30 Antibody-drug conjugates comprising modified antibodies
US16/088,804 US11235065B2 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody
CA3018691A CA3018691C (en) 2016-04-06 2017-03-30 Antibody-drug conjugates comprising antibodies modified with metal ion-binding motifs
ES17779307T ES2929010T3 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody
JP2018552180A JP6708751B2 (en) 2016-04-06 2017-03-30 Antibody-drug conjugates including modified antibodies
CN201780027885.6A CN109069657B (en) 2016-04-06 2017-03-30 Antibody-drug conjugates comprising modified antibodies
US17/551,103 US20220105197A1 (en) 2016-04-06 2021-12-14 Antibody-drug conjugate comprising modified antibody

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
KR10-2016-0042269 2016-04-06
KR20160042269 2016-04-06
KR1020170040472A KR102001821B1 (en) 2016-04-06 2017-03-30 Antibody-Drug Conjugate Comprising Antibody Variant
KR10-2017-0040472 2017-03-30

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US16/088,804 A-371-Of-International US11235065B2 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody
US17/551,103 Division US20220105197A1 (en) 2016-04-06 2021-12-14 Antibody-drug conjugate comprising modified antibody

Publications (1)

Publication Number Publication Date
WO2017176007A1 true WO2017176007A1 (en) 2017-10-12

Family

ID=60000602

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/KR2017/003508 WO2017176007A1 (en) 2016-04-06 2017-03-30 Antibody-drug conjugate comprising modified antibody

Country Status (1)

Country Link
WO (1) WO2017176007A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210009685A1 (en) * 2018-03-14 2021-01-14 Alteogen, Inc. Antibody specifically binding to folr1 and uses thereof
JP2022528318A (en) * 2019-03-25 2022-06-10 イミュンワーク インク. Complex polypeptides with metal binding motifs and molecular constructs comprising them

Citations (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070054682A (en) * 2004-09-23 2007-05-29 제넨테크, 인크. Cystein engineered antibodies and conjugates
WO2009026274A1 (en) * 2007-08-22 2009-02-26 Medarex, Inc. Site-specific attachment of drugs or other agents to engineered antibodies with c-terminal extensions
KR20130097669A (en) * 2012-02-24 2013-09-03 (주)알테오젠 Antibody variant with cysteine residues, antibody-drug conjugate comprising the antibody variant, and method of manufacturing thereof
US20140303084A1 (en) * 2011-06-10 2014-10-09 Biogen Idec Ma Inc. Pro-Coagulant Compounds and Methods of Use Thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20070054682A (en) * 2004-09-23 2007-05-29 제넨테크, 인크. Cystein engineered antibodies and conjugates
WO2009026274A1 (en) * 2007-08-22 2009-02-26 Medarex, Inc. Site-specific attachment of drugs or other agents to engineered antibodies with c-terminal extensions
US20140303084A1 (en) * 2011-06-10 2014-10-09 Biogen Idec Ma Inc. Pro-Coagulant Compounds and Methods of Use Thereof
KR20130097669A (en) * 2012-02-24 2013-09-03 (주)알테오젠 Antibody variant with cysteine residues, antibody-drug conjugate comprising the antibody variant, and method of manufacturing thereof
KR101541764B1 (en) 2012-02-24 2015-08-05 (주)알테오젠 Antibody variant with cysteine residues, antibody-drug conjugate comprising the antibody variant, and method of manufacturing thereof

Non-Patent Citations (22)

* Cited by examiner, † Cited by third party
Title
"Genbank", Database accession no. AB040878
"Genbank", Database accession no. AF184971
"Genbank", Database accession no. AF229053
"Genbank", Database accession no. AF361486
"Genbank", Database accession no. AJ297436
"Genbank", Database accession no. AX092328
"Genbank", Database accession no. AY260763
"Genbank", Database accession no. BC017023
"Genbank", Database accession no. M11730
"Genbank", Database accession no. M18728
"Genbank", Database accession no. M26004
"Genbank", Database accession no. NM 017636
"Genbank", Database accession no. NM 030764
"Genbank", Database accession no. NM_000626
"Genbank", Database accession no. NM_004442
"Genbank", Database accession no. NM_005823
DRAKE ET AL., BIOCONJUGATE CHEMISTRY, vol. 25, 2014, pages 1331 - 1341
GRENACS, A. ET AL.: "Copper (II) and Nickel (II) Binding Sites of Peptide Containing Adjacent Histidyl Residues", JOURNAL OF INORGANIC BIOCHEMISTRY, vol. 151, 2015, pages 87 - 93, XP029344785 *
JUNUTULA ET AL., NATURE BIOTECHNOLOGY, vol. 26, 2008, pages 925 - 932
STROP ET AL., CHEMISTRY & BIOLOGY, vol. 20, 2013, pages 161 - 167
WANG ET AL., PROC. NATL. ACAD. SCI. USA, vol. 100, 2003, pages 56 - 61
ZIMMERMAN, BIOCONJUGATE CHEM., vol. 25, 2014, pages 351 - 361

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20210009685A1 (en) * 2018-03-14 2021-01-14 Alteogen, Inc. Antibody specifically binding to folr1 and uses thereof
US11866492B2 (en) * 2018-03-14 2024-01-09 Alteogen, Inc. Antibody specifically binding to FOLR1 and uses thereof
JP2022528318A (en) * 2019-03-25 2022-06-10 イミュンワーク インク. Complex polypeptides with metal binding motifs and molecular constructs comprising them
JP7395200B2 (en) 2019-03-25 2023-12-11 イミュンワーク インク. Composite polypeptide having a metal-binding motif and a molecular construct comprising the same

Similar Documents

Publication Publication Date Title
WO2013125891A1 (en) Modified antibody in which motif comprising cysteine residue is bound, modified antibody-drug conjugate comprising the modified antibody, and production method for same
KR102076219B1 (en) Tridentate connexon and use thereof
EP3302570B1 (en) Trimaleimide linkers and uses thereof
KR102604938B1 (en) Tetramaleimide linker and its uses
WO2017176007A1 (en) Antibody-drug conjugate comprising modified antibody
US11235065B2 (en) Antibody-drug conjugate comprising modified antibody
WO2019132579A2 (en) Immunotoxin comprising ribonuclease fused to cytotransmab
WO2021101349A1 (en) Antibody that binds to ror1 and b7-h3, antibody-drug conjugate containing same, and use thereof
WO2022211508A1 (en) Antibody-drug conjugate including antibody against human cldn18.2, and use thereof
WO2017073981A1 (en) Antibody-drug conjugate and preparation method therefor
WO2023080695A1 (en) Resistin-specific antibody and use thereof
WO2024025343A1 (en) Anti-ror1 antibody and use thereof
WO2024043643A1 (en) Il2 variant and protein complex comprising same
WO2022245186A1 (en) Antibody-drug conjugate that binds to ror1 and b7-h3, and use thereof

Legal Events

Date Code Title Description
ENP Entry into the national phase

Ref document number: 3018691

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2018552180

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2017779307

Country of ref document: EP

ENP Entry into the national phase

Ref document number: 2017779307

Country of ref document: EP

Effective date: 20181106

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

Ref document number: 17779307

Country of ref document: EP

Kind code of ref document: A1