WO2022218288A1 - 一种抗体药物偶联物的制备方法 - Google Patents

一种抗体药物偶联物的制备方法 Download PDF

Info

Publication number
WO2022218288A1
WO2022218288A1 PCT/CN2022/086254 CN2022086254W WO2022218288A1 WO 2022218288 A1 WO2022218288 A1 WO 2022218288A1 CN 2022086254 W CN2022086254 W CN 2022086254W WO 2022218288 A1 WO2022218288 A1 WO 2022218288A1
Authority
WO
WIPO (PCT)
Prior art keywords
group
antibody
substituted
drug
unsubstituted
Prior art date
Application number
PCT/CN2022/086254
Other languages
English (en)
French (fr)
Inventor
王文元
王召印
张琳
Original Assignee
中国科学院上海有机化学研究所
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 中国科学院上海有机化学研究所 filed Critical 中国科学院上海有机化学研究所
Publication of WO2022218288A1 publication Critical patent/WO2022218288A1/zh

Links

Images

Classifications

    • 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
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K39/395Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum
    • A61K39/39533Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals
    • A61K39/39558Antibodies; Immunoglobulins; Immune serum, e.g. antilymphocytic serum against materials from animals against tumor tissues, cells, antigens
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • 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
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K49/00Preparations for testing in vivo
    • A61K49/001Preparation for luminescence or biological staining
    • A61K49/0013Luminescence
    • A61K49/0017Fluorescence in vivo
    • A61K49/005Fluorescence in vivo characterised by the carrier molecule carrying the fluorescent agent
    • A61K49/0058Antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/32Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against translation products of oncogenes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2317/00Immunoglobulins specific features
    • C07K2317/50Immunoglobulins specific features characterized by immunoglobulin fragments
    • C07K2317/55Fab or Fab'

Definitions

  • the invention belongs to the field of biomedicine, in particular to a preparation method of a bifunctional antibody drug conjugate.
  • Tumor immunotherapy is a systemic treatment method. Its principle is to enhance the function of the immune system in the patient's body through external conditional assistance, so as to achieve the purpose of killing tumor cells and relieving the disease. However, it is often less effective for tumors that develop immune tolerance. Due to the current single use of any therapy has certain limitations.
  • ADCs Antibody drug conjugates
  • ADCs are complex drugs formed by covalently coupling a small molecule anticancer drug with antitumor toxicity and a monoclonal antibody with target specificity using a linker.
  • This immunoconjugate combines the potent antitumor activity of small molecule drugs (300-1000Da, IC 50 below nmol) with the high selectivity, stability and good pharmacokinetic properties of monoclonal antibodies. After selecting a suitable target, it is necessary to prepare antibodies with good affinity for the target.
  • the molecular weight of the antibody 150KD makes it difficult for ADCs to penetrate the capillary endothelial layer and the extracellular space, resulting in a much smaller amount of the antibody entering the solid tumor site than the administered dose. Therefore, early ADCs are mainly aimed at hematological tumors. In order to enable ADCs to be used in the treatment of solid tumors, it is currently trying to remove some unnecessary fragments and reduce the molecular weight of ADCs on the premise of ensuring the ability of antibodies to specifically bind to antigens, and then couple with high-efficiency small molecular compounds to achieve for the treatment of solid tumors. So far, with the deepening of clinical research, ADCs will gradually appear drug resistance in long-term use.
  • the field of tumor targeted therapy needs to provide new therapeutic drugs for reducing drug resistance and improving drug efficacy and preparation methods thereof.
  • the purpose of the present invention is to provide an antitumor drug with reduced drug resistance and improved efficacy and a preparation method thereof.
  • a first aspect of the present invention provides a method for preparing an antibody drug conjugate, comprising the steps of:
  • the mutant antibody when the mutant antibody reacts with the drug molecule A with a hydroxylamine group or modified with a hydroxylamine group, the reaction temperature T1 is 37 ⁇ 10° C. and the pH is 3-5; and
  • the reaction temperature T2 is 4 ⁇ 10° C. and the pH is 5.1-6.5.
  • the antibody is selected from the group consisting of full-length antibodies, antigen-binding fragments, bispecific antibodies or nanobodies.
  • the antibody is selected from the following group: the antibody is selected from the antibodies targeting the following targets: CCR4, CD3, CD19, CD20, CD30, CD22, EGFR, EpCAM, HER2, PD-1, PD-L1, VEGF, VEGFA, VEGFR2, C5 protein, CD2, IL-5, PCSK9, RSVF, platelet glycoprotein IIb/IIIa, RANK, DLL3, EDAR, CLL1, BMPR1B, E16, STEAP1, 0772P, MPF, NaPi2b, Sema 5b, PSCA hlg, ETBR, MSG783, STEAP2, TrpM4, CRIPTO, CD21, CD79b, FcRH2, B7-H4, HER2, NCA, MDP, IL20R ⁇ , Brevican, EphB2R, ASLG659, PSCA, GEDA, BAFF-R, CD22, CD79a, CXCR5, HLA-DOB, P2X5, CD
  • the antibody is selected from antibodies targeting the following targets:
  • the hydrocarbon acyl aryl group has the formula
  • the ring A is selected from the following group: substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted including 1-3 5-10-membered heteroaryls selected from O, N, S;
  • R is selected from the group consisting of substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C2-C4 alkenyl, substituted or unsubstituted C2-C4 alkynyl,
  • the hydrocarbon acyl aryl group has the formula Wherein, R is C1-C4 alkyl, preferably, R is methyl, ethyl or isopropyl.
  • the mutant antibody is obtained by introducing (substituting or inserting) a natural amino acid or an unnatural amino acid capable of generating a free sulfhydryl group (-SH) and a hydrocarbyl aryl group into a wild-type antibody, respectively.
  • the mutant antibody comprises an amino acid of formula (I), thereby providing a sulfhydryl group:
  • R1 is selected from the group consisting of C1-C4 alkyl substituted with mercapto, C2-C4 alkenyl substituted with mercapto, and C2-C4 alkynyl substituted with mercapto.
  • the R1 is -CH 2 -SH or CH 2 CH 2 -SH.
  • the amino acid that generates the free thiol group is cysteine.
  • the mutant antibody comprises an amino acid of formula (II), thereby providing the alkanoyl aryl group:
  • Said A is selected from the following group: substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted including 1-3 5-10-membered heteroaryl groups selected from O, N, S;
  • R is selected from the group consisting of substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C2-C4 alkenyl, substituted or unsubstituted C2-C4 alkynyl;
  • the A is a phenyl group
  • R is a C1-C4 alkyl group, preferably, R is a methyl group, an ethyl group or an isopropyl group.
  • the amino acid generating the hydrocarbon acyl aryl group is selected from the group consisting of p-acetylphenylalanine and p-propionylphenylalanine.
  • the mutant antibody is obtained by introducing (substituting or inserting) cysteine and p-acetylphenylalanine into two sites in the wild-type antibody, respectively.
  • the mutant antibody is an antigen-binding fragment (Fab) targeting HER2, and the antigen-binding fragment includes a heavy chain and a light chain;
  • Fab antigen-binding fragment
  • amino acid sequence of the heavy chain is SEQ ID No: 1;
  • amino acid sequence of the light chain is SEQ ID No:2.
  • the reaction in step (b) is carried out by a "one-pot method". That is, after the previous reaction is completed, another drug molecule is directly added to the reaction solution of the previous reaction, the reaction conditions are adjusted, and the reaction is carried out.
  • the method further includes a post-processing step of separation and purification, preferably, the separation and purification can be performed by a method selected from the group consisting of extraction method, chromatography method (such as molecular sieve chromatography), electrophoresis, or a combination thereof.
  • a post-processing step of separation and purification preferably, the separation and purification can be performed by a method selected from the group consisting of extraction method, chromatography method (such as molecular sieve chromatography), electrophoresis, or a combination thereof.
  • the reaction solvent contains an aqueous solution of a pH buffer system, such as sodium acetate buffer.
  • a pH buffer system such as sodium acetate buffer.
  • reaction of the antibody with the drug molecule A with a hydroxylamine (-O-NH 2 ) group or modified with a hydroxylamine group includes one or more of the following features:
  • pH is 3.5-4.8, such as 3.8, 4.0, 4.2 or 4.5;
  • the time of described reaction is 1-36h, preferably, 6-24h, such as 12h, 16 or 18h; and/or
  • the dosage ratio of the antibody to the drug molecule A is 1:1-50 equivalents, preferably 1:5-30 equivalents, such as 1:10 equivalents, 1:15 equivalents, 1:20 equivalents or 1:20 equivalents 25 equiv.
  • reaction of the antibody with a drug molecule B with a maleimide group or modified with a maleimide group includes one or more of the following features:
  • pH is 5.2-6.2, such as 5.5, 5.8 or 6.0;
  • the time of described reaction is 1-36h, preferably, 6-24h, such as 12h, 16 or 18h; and/or
  • the dosage ratio of the antibody to the drug molecule B is 1:1-50 equivalents, preferably 1:2-30 equivalents, such as 1:5 equivalents, 1:10 equivalents, 1:15 equivalents, or 1 : 20 equiv.
  • the drug molecule A and the drug molecule B are independently selected from the following group:
  • Tracers such as fluorescein, radioactive-labeled compounds, etc.; compounds with definite functions such as cytotoxins, anti-tumor drugs, antibiotics, etc., and molecules with definite functions such as nucleic acids.
  • the drug molecule A and drug molecule B can be independently selected from drugs targeting (but not limited to) the following targets: FoxO1, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK1, BRSK2 , BTK, CAMKK1, CAMKK ⁇ , CAMKK ⁇ , Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, cyclin D, pl6INK4A, cdc25A, BMI1, SCF, Akt, CHK1/2, C1 ⁇ , CK1 ⁇ , C2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A3, Smad2, Smad3 , Smad4, Smad7, p53, p21Cipl, PAX, Fyn, CAS, C
  • the drug molecule is independently a small molecule drug.
  • the drug molecule is independently an antitumor drug.
  • the drug molecule A and the drug molecule B are drugs that act through two different mechanisms.
  • the drug molecule A and the drug molecule B are independently selected from the following group:
  • PARP1/2 inhibitors chemotherapy drugs that induce DNA damage in cancer cells, DNA alkylating drugs, DNA or RNA synthesis inhibitors, EGFR, ALK or FGFR tyrosine receptor kinase inhibitors, KRAS, MEK or ERK tumors signaling pathway inhibitors.
  • the drug molecule A and drug molecule B are independently selected from the group consisting of olaparib, lucaparib, niraparib, methotrexate, capecitabine, gemcitabine, Deoxyfluridine, pemetrexed disodium, pazopanib, imatinib, erlotinib, lapatinib, gefitinib, vandetanib, herceptin, paclitaxel, vinca Rebine, Docetaxel, Doxorubicin, Hydroxycamptothecin, Mitomycin, Epirubicin, Pirarubicin, Bleomycin, Letrozole, Tamoxifen, Fulvestrant tran, triptorelin, flutamide, leuprolide, anastrozole, ifosfamide, busulfan, cyclophosphamide, carmustine, nimustine, semustine, nitrogen mustard, Melphalan, Tuco Ning, Carboplatin, Cisplatin
  • the second aspect of the present invention provides a mutant antibody, the mutant antibody comprises two reactive groups, wherein one of the reactive groups is a free sulfhydryl group (-SH), and the other is a hydrocarbon acyl aryl group
  • the antibody is selected from the group consisting of full-length antibodies, antigen-binding fragments, bispecific antibodies or nanobodies.
  • the mutant antibody is obtained by replacing two sites in the wild-type antibody with natural amino acids or unnatural amino acids capable of generating free sulfhydryl (-SH) and alkanoyl aryl groups, respectively.
  • the amino acid is selected from the group consisting of L-amino acid, D-amino acid, or a combination thereof.
  • the mutant antibody has the formula:
  • the ab represents an antibody
  • A-(CO)-R has the formula
  • the ring A is selected from the following group: substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted including 1-3 5-10-membered heteroaryls selected from O, N, S;
  • R is selected from the group consisting of substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C2-C4 alkenyl, substituted or unsubstituted C2-C4 alkynyl,
  • the hydrocarbon acyl aryl group has the formula Wherein, R is C1-C4 alkyl, preferably, R is methyl, ethyl or isopropyl.
  • the mutant antibody is obtained by replacing two sites with cysteine and p-acetylphenylalanine respectively in the wild-type antibody.
  • the mutant antibody is prepared by genetic engineering.
  • the mutant antibody is an antigen-binding fragment targeting HER2, and the mutant antigen-binding fragment includes a heavy chain and a light chain; wherein,
  • amino acid sequence of the heavy chain is SEQ ID No: 1;
  • amino acid sequence of the light chain is SEQ ID No:2.
  • the third aspect of the present invention provides the use of the mutant antibody according to the second aspect of the present invention for preparing antibody drug conjugates and/or antibody fluorescent coupling reagents.
  • the antibody-drug conjugate and/or the antibody-fluorescent conjugated reagent is targeted to tumor cells.
  • the antibody-drug conjugate and/or the antibody-fluorescent conjugation reagent is a bifunctional drug conjugate obtained by coupling the antigen-binding fragment with two different drug molecules.
  • the fourth aspect of the present invention provides an antibody-drug conjugate prepared by the preparation method described in the first aspect of the present invention, or a pharmaceutically acceptable salt thereof.
  • a fifth aspect of the present invention provides a pharmaceutical composition comprising:
  • the antibody drug conjugate according to the fourth aspect of the present invention or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier.
  • the sixth aspect of the present invention provides the use of the antibody-drug conjugate according to the fourth aspect of the present invention, or a pharmaceutically acceptable salt thereof, or a composition comprising the same, for preparing a medicine, the medicine
  • the medicine For one or more purposes selected from the group consisting of:
  • A1) Prevent and/or inhibit tumor cell proliferation
  • the tumor or tumor cell is a tumor or tumor cell with high expression of HER2.
  • the tumor or tumor cell is selected from the group consisting of breast cancer, lung cancer, pancreatic cancer, ovarian cancer, gastric cancer, fibrosarcoma, bladder cancer, ovarian cancer, adenocarcinoma, colon cancer, bone cancer, brain cancer Carcinoma, neurocytoma, head and neck cancer, rectal cancer, colon cancer, familial adenomatous polyposis cancer, hereditary nonpolyposis colorectal cancer, esophageal cancer, lip cancer, laryngeal cancer, hypopharyngeal cancer, tongue cancer, salivary gland cancer , adenocarcinoma, medullary thyroid cancer, papillary thyroid cancer, kidney cancer, renal parenchyma cancer, cervical cancer, endometrial cancer, endometrial cancer, choriocarcinoma, prostate cancer, testicular cancer, urinary cancer, melanoma, Acute lymphocytic leukemia, chronic lymphocytic leukemia, acute myeloid leuk
  • FIG. 1 is a schematic structural diagram of a bifunctional mutant antibody of the present invention.
  • FIG. 2 is a schematic structural diagram of a double-conjugated antibody-drug conjugate of the present invention
  • FIG 3 shows the one-pot method to obtain dual-mechanism tumor therapy antibody conjugated drug reaction scheme 1:
  • Figure 4 is a one-pot method to obtain dual-mechanism tumor therapy antibody conjugated drug reaction scheme 2;
  • Figure 5 shows that the molecular weight of the purified anti-HER2 DualFab (anti-HER2 dual-functional Fab) was confirmed by protein mass spectrometry to meet the theoretical value.
  • Figure 6 shows the specificity of the purified anti-HER2 DualFab obtained by immunofluorescence verification.
  • Figure 7 shows the cell viability test to verify the specificity of the purified anti-HER2 DualFab.
  • Figure 8 shows an example of a double coupling reaction procedure.
  • Figure 9 shows the SDS-PAGE test results and grayscale analysis of the reaction solution of the double coupling reaction.
  • the double-coupling experimental group was compared with the control group to which only Alexa 488 with hydroxylamine group was added and the control group to which only Alexa 568 with maleimide was added. , confirming the completion of the double coupling, and performing grayscale analysis according to the results of the fluorescence imager, indicating that the ratio of the two-step coupling reaches 1:1.
  • FIG 10 shows the results of immunofluorescence staining of Alexa 488 Alexa 568 DualFab.
  • Fig. 11 is the fluorescence photographing result of Alexa 488 Alexa 568 DualFab processing SKBR3 0h, 3h, 6h, 12h, 18h and 24h.
  • FIG. 12 is the reaction formula of Example 3.
  • a mutant antibody with both sulfhydryl and acyl reactive groups is constructed on a conventional wild-type antibody, and the modified mutant antibody can be coupled through a "one-pot method" through a maleimide linker and a hydroxylamine linker. drug molecule.
  • the antibody-drug conjugate obtained by the reaction has multiple targeted inhibitory effects on target cells (such as tumor cells), so that multi-mechanism targeted therapy can be realized.
  • the present invention has been completed on this basis.
  • the term “about” means that the value may vary by no more than 1% from the recited value.
  • the expression “about 100” includes all values between 99 and 101 and (eg, 99.1, 99.2, 99.3, 99.4, etc.).
  • the terms "containing” or “including (including)” can be open, semi-closed, and closed. In other words, the term also includes “consisting essentially of,” or “consisting of.”
  • room temperature or "normal temperature” refers to a temperature of 4-40°C, preferably, 25 ⁇ 5°C.
  • alkyl refers to a straight or branched chain alkyl group containing several carbon atoms, wherein "C1-C4 alkyl” means one having 1-4 Linear or branched chain alkyl groups of carbon atoms, including alkyl groups of 1, 2, 3 or 4 carbon atoms, preferably alkyl groups such as C1-C2, C1-C3.
  • Typical "alkyl” includes, but is not limited to, methyl, ethyl, propyl, isopropyl, n-butyl, tert-butyl, isobutyl.
  • the alkyl group can also be an alkylene group, such as an alkylene group of 1, 2, 3 or 4 carbon atoms.
  • alkenyl refers to a straight or branched chain hydrocarbon having at least 2 carbon atoms and at least one double bond.
  • Alkenyl groups can include any number of carbon atoms, wherein “C2-C4 alkenyl” refers to straight or branched chain hydrocarbons having 2-4 carbon atoms and at least one double bond, such as C2 , C2 - C 3. C 2 -C 4 .
  • An alkenyl group can have any suitable number of double bonds, including but not limited to 1 or 2.
  • alkenyl groups include, but are not limited to, vinyl (vinyl group)), propenyl, isopropenyl, 1-butenyl, 2-butenyl, isobutenyl, butadienyl.
  • the alkenyl group can also be an alkenylene group, such as an alkenylene group of 2, 3 or 4 carbon atoms.
  • alkynyl refers to a straight or branched chain hydrocarbon having at least 2 carbon atoms and at least one triple bond.
  • Alkynyl groups can include any number of carbon atoms
  • C2-C4alkynyl refers to straight or branched chain hydrocarbons having 2-4 carbon atoms and at least one triple bond, such as C2, C2 - C3, C 2 -C 4 .
  • Examples of alkynyl groups include, but are not limited to, ethynyl, propynyl, 1-butynyl, 2-butynyl, isobutynyl, sec-butynyl, butadiynyl.
  • the alkynyl group can also be an alkynylene group, such as an alkynylene group of 2, 3 or 4 carbon atoms.
  • aryl refers to an aromatic cyclic hydrocarbon compound group, wherein "C6-C10 aryl” refers to an aromatic cyclic hydrocarbon compound group containing 6, 7, 8, 9 or 10 ring carbon atoms , having 1-2 rings, especially monocyclic and bicyclic groups such as phenyl, biphenyl or naphthyl. Where there are two or more aromatic rings (bicyclic rings, etc.), the aromatic rings of the aryl group can be linked by a single bond (eg, biphenyl). "Substituted aryl” means that one or more positions in the aryl group are substituted, especially 1-3 substituents, which may be substituted at any position.
  • heteroaryl refers to a heteroaromatic system containing 1-3 atoms selected from N, O, S, wherein, "5-10 membered heteroaryl” refers to containing 1-3 atoms selected from 5-10 membered heteroaromatic system from N, O, S atoms.
  • Heteroaryl is preferably a 5- to 10-membered ring, more preferably 5- or 6-membered, heteroaryl includes but is not limited to pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadi azolyl, isothiazolyl, furanyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazinyl, triazolyl and tetrazolyl, etc.
  • Heteroaryl may be substituted or unsubstituted, and when substituted, the substituent may be on a C atom or a heteroatom.
  • the term "plurality” refers to two or more, such as 2, 3, 4, 5 or 6.
  • halogen or "halogen atom” refers to F, Cl, Br, and I.
  • substitution means that one or more H on each group is independently replaced by a group selected from the group consisting of halogen, C1-C4 alkyl, C1-C4 alkoxy, C2-C4 alkene group, C2-C4alkynyl, -NO 2 , -CN or -OH.
  • wild-type antibody refers to the antibody before the reactive group of the present invention is introduced.
  • the antibody can be any desired antibody, such as full-length antibodies, antigen-binding fragments, bispecific antibodies, nanobodies and other functional proteins.
  • the introduction of the two reactive groups is known to those skilled in the art without destroying (or within acceptable levels) the ability of the antibody to bind to the target or easily achievable.
  • reactive groups can be introduced by insertion, substitution, modification and other methods.
  • the free sulfhydryl group and the hydrocarbyl aryl group can be located on the same or different peptide chains in the antibody molecule, eg, the sulfhydryl group is located on the light chain and the hydrocarbyl aryl group is located on the heavy chain.
  • the mutant antibody is obtained by introducing a natural or unnatural amino acid capable of generating free sulfhydryl (-SH) and alkanoyl aryl groups, respectively, into a wild-type antibody.
  • the "introduction” may be obtained by substitution, insertion or modification.
  • a free sulfhydryl group can be introduced by substituting one cysteine for an existing amino acid in the wild-type antibody molecule; amino acid, thereby introducing an acetylphenyl group.
  • the replaced site is a position away from the domain that binds to the target.
  • the above mutant antibodies can be constructed, expressed and obtained by common genetic engineering methods, or synthesized by chemical methods.
  • methods for constructing and expressing the mutant antibody by genetic engineering are known.
  • the antibody may be selected from antibodies targeting (but not limited to) the following targets: CCR4, CD3, CD19, CD20, CD30, CD22, EGFR, EpCAM, HER2, PD-1, PD-L1, VEGF, VEGFA, VEGFR2, etc.; and autoimmune diseases; asthma, anti-infection, blood diseases, cardiovascular diseases, osteoporosis, multiple sclerosis, Alzheimer's disease and other diseases related proteins, such as C5 protein, CD2, IL -5, PCSK9, RSVF, platelet glycoprotein IIb/IIIa, RANK and other proteins and polypeptides.
  • targets such as C5 protein, CD2, IL -5, PCSK9, RSVF, platelet glycoprotein IIb/IIIa, RANK and other proteins and polypeptides.
  • the present invention provides a method for preparing an antibody drug conjugate, comprising the steps of:
  • the reaction temperature T1 is 37 ⁇ 10° C. and the pH is 3-5.
  • the reaction temperature T2 is 4 ⁇ 10°C and the pH is 5.1-6.5 .
  • mutant antibody may react with drug molecule A first, and then the resulting reaction product reacts with drug molecule B; or, the mutant antibody may first react with drug molecule B, and then the resulting reaction The product reacts with drug molecule A, as shown in Figure 3-4.
  • the -SH in the mutant antibody reacts with the maleimide group of drug molecule A.
  • the amino acid generating the free sulfhydryl group has the formula
  • R1 is selected from the group consisting of C1-C4 alkyl substituted with mercapto, C2-C4 alkenyl substituted with mercapto, and C2-C4 alkynyl substituted with mercapto.
  • the R1 is -CH 2 -SH or CH 2 CH 2 -SH.
  • the amino acid from which the free sulfhydryl group is derived is cysteine.
  • the amino acid from which the hydrocarbyl aryl group is derived has the formula
  • Said A is selected from the following group: substituted or unsubstituted C6-C10 aryl, substituted or unsubstituted including 1-3 5-10-membered heteroaryl groups selected from O, N, S;
  • R is selected from the group consisting of substituted or unsubstituted C1-C4 alkyl, substituted or unsubstituted C2-C4 alkenyl, substituted or unsubstituted C2-C4 alkynyl;
  • the amino acid producing the hydrocarbyl aryl group is selected from the group consisting of p-acetylphenylalanine, p-propionylphenylalanine.
  • drug molecule A and drug molecule B which can be selected according to needs, for example, small molecule drugs, nucleic acids, fluorescent molecules, etc., which are common in the field.
  • drug molecule A and drug molecule B which can be selected according to needs, for example, small molecule drugs, nucleic acids, fluorescent molecules, etc., which are common in the field.
  • the drug molecule A and drug molecule B can be independently selected from drugs targeting (but not limited to) the following targets: FoxO1, HDAC, DP-1, E2F, ABL, AMPK, BRK, BRSK I, BRSK2, BTK, CAMKK1, CAMKK ⁇ , CAMKK ⁇ , Rb, Suv39HI, SCF, p19INK4D, GSK-3, pi 8INK4, myc, cyclin E, CDK2, CDK9, CDG4/6, cyclin D, pl6INK4A, cdc25A, BMI1 , SCF, Akt, CHK1/2, C 1 ⁇ , CK1 ⁇ , C 2, CLK2, CSK, DDR2, DYRK1A/2/3, EF2K, EPH-A2/A4/B1/B2/B3/B4, EIF2A 3, Smad2, Smad3, Smad4, Smad7, p53, p21Cipl, PAX, Fyn, CAS, C3
  • the drug molecule A and the drug molecule B are drugs with two different mechanisms.
  • antibody drug conjugate As used herein, the terms “antibody drug conjugate”, “antibody drug conjugate”, “ADC” are used interchangeably.
  • the present invention also provides the antibody-drug conjugate prepared by the above method or a pharmaceutically acceptable salt thereof.
  • the antibody drug conjugate is a therapeutic or diagnostic drug. It has at least the biological activity of antibody, drug molecule A and drug molecule B. In addition, it can also achieve the synergistic effect when the mechanism drug is not used in combination, has the ability to target the drug delivery to cells, reduce the dosage of the drug, and reduce the drug resistance of the drug.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned antibody-drug conjugate or a pharmaceutically acceptable salt thereof as an active ingredient, and a pharmaceutically acceptable carrier.
  • the methods of the present invention can prepare various antibody-drug conjugates targeting specific cells. For example, but not limited to, tumor cells, immune cells, inflammatory cells, etc. can be targeted. Then, the antibody or the conjugated drug can play a role, and the targeted therapy can be achieved in a targeted manner.
  • “Pharmaceutically acceptable excipient” and “pharmaceutically acceptable carrier” refer to substances that aid in the formulation and/or administration and/or absorption of an active agent by an individual, and may be included in the compositions of the present disclosure without causing significant adverse toxicological effects in the individual.
  • Non-limiting examples of pharmaceutically acceptable carriers and excipients include water, NaCl, physiological saline solution, lactated Ringer's, conventional sucrose, conventional dextrose, binders, fillers, disintegrants, lubricants, Coatings, sweeteners, flavors, saline solutions (eg Ringer's solution), alcohols, oils, gelatin, carbohydrates such as lactose, amylose or starch, fatty acid esters, hydroxymethylcellulose, polyvinylpyrrole alkanes and pigments, etc.
  • Such formulations can be sterilized and, if desired, with adjuvants such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers that do not deleteriously react with or interfere with the activity of the compounds provided herein , a mixture of salts, buffers, colorants and/or fragrances that affect osmotic pressure.
  • adjuvants such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers that do not deleteriously react with or interfere with the activity of the compounds provided herein , a mixture of salts, buffers, colorants and/or fragrances that affect osmotic pressure.
  • compositions of the present invention may be in solid or liquid form.
  • the medicaments containing the active ingredients of the present invention may be in suitable oral dosage forms such as tablets, troches, troches, water-soluble or oily suspensions, dispersible powders or granules, emulsions, hard or soft capsules or syrups or elixirs.
  • Medicaments for oral use may be prepared according to the known procedures of the pharmaceutical ingredient manufacturers, and these compositions may include one or more of the following agents, such as sweetening, flavoring, coloring and protecting agents, in order to provide elegance and Delicious medicinal preparation.
  • Tablets contain the active ingredient in admixture with non-toxic pharmaceutically acceptable excipients which are suitable for the manufacture of tablets.
  • excipients examples include, inert diluents such as calcium carbonate, sodium carbonate, lactose, calcium phosphate or sodium phosphate; granulating, disintegrating agents such as corn starch or alginic acid; binding agents such as starch, Gelatin or acacia, and lubricants such as magnesium stearate, stearic acid, or talc.
  • the tablet may be uncoated or coated to delay degradation and absorption in the gastrointestinal tract, thereby maintaining activity over a longer period of time.
  • the active compounds may be administered to a subject by any suitable route, including orally, parenterally, by inhalation spray, topically, rectally, nasally, bucally, vaginally, or via an implanted kit.
  • parenteral as used herein includes subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and cranial intra-injection or infusion techniques.
  • the composition is administered orally, intraperitoneally or intravenously.
  • compositions of the present invention suitable for oral administration will typically be discrete units in solid form, such as in the form of tablets, capsules, cachets, powders, granules, lozenges, patches, suppositories, pills, or In liquid form, eg, liquid preparations, injectable or infusible solutions or suspensions.
  • the precise amount of compound to provide a therapeutically effective amount of the compound to an individual will depend on the mode of administration, the type and severity of the disease and/or disorder, and the characteristics of the individual, such as general health, age, sex, weight, and tolerance to the drug . Those of ordinary skill in the art will be able to determine appropriate dosages based on these and other factors. When administered in combination with other therapeutic agents, the "therapeutically effective amount" of any other therapeutic agent will depend on the type of drug used.
  • compositions should be formulated such that doses of the inhibitor from 0.01 to 100 mg/kg body weight/day can be administered to patients receiving these compositions.
  • the compositions of the present invention provide a dose of 0.01 mg to 50 mg. In other embodiments, a dose of 0.1 mg-25 mg or 5 mg-40 mg is provided.
  • subjects to which the pharmaceutical composition or therapeutic agent of the present invention is administered include mammals (eg, humans, mice, rats, hamsters, rabbits, cats, dogs, cows, sheep, monkeys, etc.).
  • the present invention also provides a method for preparing a pharmaceutical composition, comprising the steps of: mixing a pharmaceutically acceptable carrier with the antibody-drug conjugate of the present invention or a pharmaceutically acceptable salt thereof to form a pharmaceutical combination thing.
  • the present invention also provides a treatment method, which comprises the steps of: administering the antibody-drug conjugate of the present invention, or a pharmaceutically acceptable salt thereof, or administering the pharmaceutical composition of the present invention to a subject in need of treatment , for the selective inhibition of cancers (eg, HER2-mediated cancers).
  • a treatment method which comprises the steps of: administering the antibody-drug conjugate of the present invention, or a pharmaceutically acceptable salt thereof, or administering the pharmaceutical composition of the present invention to a subject in need of treatment , for the selective inhibition of cancers (eg, HER2-mediated cancers).
  • cancers eg, HER2-mediated cancers
  • the present invention provides a mutant antibody with double conjugation potential, the mutant antibody includes two reactive groups (thiol and acetyl aryl) that can be linked to a drug, and the mutant antibody can have a single introduction
  • the two kinds of reactions that cannot be done can be carried out in sequence, and the antibody conjugate with upgraded function can be obtained simply and efficiently.
  • two reactive groups are introduced, and two drugs with different mechanisms are coupled to produce dual-target drugs.
  • the method of the present invention can be prepared by a one-pot method at room temperature of 4-37°C, can maintain the structure and function of the antibody, is simple to operate, safe and efficient, requires low equipment, and is suitable for large-scale production.
  • the mutant antibody of the present invention provides two different reactive groups, and the reaction sites are in one-to-one correspondence according to different types of drug molecules during the reaction, and the prepared antibody-drug conjugates have the same structure and high purity.
  • the antibody-drug conjugate prepared by the method of the present invention has improved drug efficacy, is not easy to develop drug resistance, and has low toxicity.
  • the antibody-drug conjugate prepared by the method of the present invention has excellent pharmacodynamics and pharmacokinetic properties, and is suitable for finished medicine.
  • cysteine was inserted into position 124 of the light chain of the anti-HER2 antibody to obtain a mutated light chain plasmid.
  • pAcF p-acetylphenylalanine
  • amino acid sequence of the heavy chain is (SEQ ID No: 1):
  • amino acid sequence of the light chain is (SEQ ID No:2):
  • the 2YT medium was used for culture expansion. When the OD reached 0.6-0.8, 1 mM IPTG, 0.2% arabinose and 1 mM p-acetylphenylalanine were added and cultured at 16 degrees for 24 hours to obtain the target molecule DualFab. Sample protein profile identification.
  • Dualfab can completely distinguish cells with high HER2 expression from other cells, showing good specificity.
  • the double coupling reaction step is carried out as shown in Figure 8.
  • the purified DualFab was added to 100 mM sodium acetate buffer pH 4.5, followed by 20 equivalents of Alexa 488 with hydroxylamine groups, at 37°C for 16-18 hours. The pH was adjusted to 6.0 and 10 equivalents of Alexa 568 with maleimide were added overnight at 4°C. Molecular sieve chromatography was used to remove excess small molecules, and the eluate was collected and concentrated to obtain dual fluorescein DualFab conjugate.
  • the SDS-PAGE test results and grayscale analysis of the reaction solution of the double-coupling reaction are shown in Figure 9.
  • the double-coupling experimental group and the control group that only added Alexa 488 with hydroxylamine groups and Compared to the control group where only Alexa 568 with maleimide was added the double-coupling was indeed completed, and the ratio of the two-step coupling was found to be 1:1 by grayscale analysis based on the results of the fluorophotometer.
  • immunofluorescence staining was used to verify the specificity of Alexa 488 Alexa 568 DualFab biconjugate to recognize HER2. After the cells were fixed, permeabilized and blocked, Alexa 488 Alexa 568 DualFab double conjugate was added to incubate at 4°C overnight, washed and mounted with DAPI-containing mounting medium, and photographed to obtain experimental results.
  • Alexa 488 Alexa 568 DualFab double conjugate can specifically recognize the cell line SKBR3 with high expression of HER2.
  • the dual-fluorescein DualFab conjugate was used to detect the endocytosis of the dual-conjugate by live cell imaging.
  • Alexa 488 Alexa 568 DualFab biconjugate specifically bound to the cell membrane of SKBR3 shortly after being added to the live cell monitoring system, and the signals of Alexa 568 and Alexa 488 were almost co-localized.
  • the fluorescent signal of Alexa 488 Alexa 568 DualFab double conjugates in SKBR3 continuously accumulated from the cell membrane to the cytoplasm, which intuitively demonstrated the endocytosis process of ADCs.
  • Figure 11 records the endocytosis process of Alexa 488 Alexa 568 DualFab biconjugate in SKBR3 at different time points of 0h, 3h, 6h, 12h, 18h and 24h.
  • the Alexa 488 Alexa 568 DualFab biconjugate was only bound to the membrane of SKBR3.
  • the fluorescent signals of Alexa 488 and Alexa 568 accumulated continuously from the membrane to the cytoplasm as indicated by the yellow arrows.
  • the above experimental results show that the present invention introduces a naked sulfhydryl group in the antibody light chain by genetic engineering, introduces an unnatural amino acid p-acetylphenylalanine with a functional reactive group carbonyl in the CH1 heavy chain part, and then expresses it by constructing
  • the strain is massively expressed in Escherichia coli, and after purification, a Fab (DualFab) with dual-conjugated functional groups is obtained, which can be used to prepare dual-mechanism or multi-mechanism antibody-conjugated drugs for efficient treatment of tumors and other diseases.
  • the purified DualFab was added to 100 mM sodium acetate buffer at pH 4.5, followed by 20 equivalents of NH2O-PEG4-vc-PAB-PBD with hydroxylamine groups, at 37°C for 16-18 hours.
  • the pH was adjusted to 6.0, and 10 equivalents of A-PEG4-vc-PAB-MMAE linker drug conjugate with maleimide group was added, overnight at 4°C.
  • Molecular sieve chromatography removes excess small molecules, and the eluate is collected and concentrated to obtain a bifunctional antitumor conjugate.
  • the reaction formula is shown in Figure 12.
  • the double-conjugation method of the present invention can introduce two or more drug molecules on the same antibody through a one-pot method, and the prepared antibody-drug conjugate can simultaneously bring two drugs with different mechanisms into target cells, which is superior to Synergistic drug use at the same time and space reduces the risk of drug resistance and improves drug efficacy.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • Organic Chemistry (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • Biochemistry (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Mycology (AREA)
  • Biophysics (AREA)
  • Genetics & Genomics (AREA)
  • Molecular Biology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Peptides Or Proteins (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

本发明公开了一种抗体药物偶联物的制备方法。具体地,该制备方法包括如下步骤:(a)提供一种包含两个反应基团的突变抗体,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基;(b)按任意先后顺序,将所述突变抗体与带有羟胺(-O-NH 2)基团或修饰有羟胺基团的药物分子A和带有马来酰亚胺基团或修饰有马来酰亚胺基团的药物分子B反应,从而得到所述抗体药物偶联物。本发明的方法可以通过一锅法高效制备耐药性降低、药效提高的抗体药物偶联物。

Description

一种抗体药物偶联物的制备方法 技术领域
本发明属于生物医学领域,具体涉及一种双功能抗体药物偶联物的制备方法。
背景技术
恶性肿瘤是种高致死性疾病。传统的癌症治疗方式包括外科手术,化学治疗和物理治疗,治疗效果不太理想,肿瘤细胞容易产生耐药性且一旦复发转移其后果更为严重。靶向小分子药物和单克隆抗体是近年来兴起的肿瘤治疗方式,但是治疗病症相对局限以及长期使用易出现耐药现象而导致治疗效果下降。肿瘤免疫治疗是一种全身性治疗方法,其原理是通过外界的条件性辅助,增强患者体内免疫系统对功能,从而达到杀伤肿瘤细胞,缓解病症的目的。但是对于产生免疫耐受的肿瘤也常常收效甚微。由于当前任何一种疗法单一使用都有一定的局限性。
抗体偶联型药物(antibody drug conjugates,ADCs)就是集化疗药物的高效毒性和抗体免疫治疗的高度特异性而对病人进行高效低毒治疗的一类新型药物。ADCs是将具有抗肿瘤毒性的小分子抗癌药物和具有靶向特异性的单克隆抗体利用连接子共价偶联形成的复合药。这种免疫偶联物融合了小分子药物(300-1000Da,IC 50在nmol以下)的高效抗肿瘤活性和单克隆抗体的高度选择性,稳定性和良好的药代动力特性。在选择好合适靶点后,就需要制备对该靶点有良好亲和力的抗体。但抗体150KD的分子量使ADCs很难透过毛细血管内皮层以及细胞外的间隙,导致其进入实体肿瘤部位的量远小于给药的剂量,因此早期的ADCs主要是针对血液瘤。为了使ADCs可以用于治疗实体瘤,目前尝试在保证抗体与抗原特异性结合的能力的前提下去除一些非必需的片段,降低ADCs的分子量,之后再与高效的小分子化合物偶联,从而达到治疗实体瘤的目的。至今为止,随着临床研究的不断深入,ADCs在长期使用中也会渐渐出现了耐药的状况。
因此,肿瘤靶向治疗领域需要提供新的用于耐药性降低、药效提高的治疗药 物及其制备方法。
发明内容
本发明的目的是提供一种耐药性降低、药效提高的抗肿瘤药物及其制备方法。
本发明第一方面,提供了一种抗体药物偶联物的制备方法,包括步骤:
(a)提供一种包含两个反应基团的突变抗体,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基
Figure PCTCN2022086254-appb-000001
(b)按任意先后顺序,将所述突变抗体与带有羟胺(-O-NH 2)基团或修饰有羟胺基团的药物分子A和带有马来酰亚胺基团
Figure PCTCN2022086254-appb-000002
或修饰有马来酰亚胺基团的药物分子B反应,从而得到所述抗体药物偶联物;
其中,当所述突变抗体与带有羟胺基团或修饰有羟胺基团的药物分子A反应时,反应温度T1为37±10℃且pH为3-5;且
当所述突变抗体与带有马来酰亚胺基团或修饰有马来酰亚胺基团的药物分子B反应时,反应温度T2为4±10℃且pH为5.1-6.5。
在另一优选例中,所述抗体选自下组:全长抗体、抗原结合片段、双特异性抗体或纳米抗体。
在另一优选例中,所述抗体选自下组:所述抗体选自靶向以下靶点的抗体:CCR4、CD3、CD19、CD20、CD30、CD22、EGFR、EpCAM、HER2、PD-1、PD-L1,VEGF、VEGFA、VEGFR2、C5蛋白、CD2、IL-5、PCSK9、RSVF、血小板糖蛋白Ⅱb/Ⅲa、RANK、DLL3、EDAR、CLL1、BMPR1B、E16、STEAP1、0772P、MPF、NaPi2b、Sema 5b、PSCA hlg、ETBR、MSG783、STEAP2、TrpM4、CRIPTO、CD21、CD79b、FcRH2、B7-H4、HER2、NCA、MDP、IL20Rα、Brevican、EphB2R、ASLG659、PSCA、GEDA、BAFF-R、CD22、CD79a、CXCR5、HLA-DOB、P2X5、CD72、LY64、FcRH1、IRTA2、TENB2、PMEL17、TMEFF1、GDNF-Ra1、Ly6E、TMEM46、Ly6G6D、LGR5、RET、LY6K、GPR19、GPR54、ASPHD1、酪氨酸酶、TMEM118、GPR172A、MUC16和CD33。
在另一优选例中,所述抗体选自靶向以下靶点的抗体:
CCR4、CD3、CD19、CD20、CD30、CD22、EGFR、EpCAM、HER2、PD-1、PD-L1,VEGF、VEGFA、VEGFR2。
在另一优选例中,所述烃酰基芳基具有式
Figure PCTCN2022086254-appb-000003
其中,所述环A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基,
所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
在另一优选例中,所述烃酰基芳基具有式
Figure PCTCN2022086254-appb-000004
其中,R为C1-C4烷基,优选地,R为甲基、乙基或异丙基。
在另一优选例中,所述突变抗体为在野生型抗体中分别引入(替换或插入)能够产生游离巯基(-SH)和烃酰基芳基的天然氨基酸或非天然氨基酸而获得。
在另一优选例中,所述突变抗体包括具有式(I)的氨基酸,从而提供巯基:
Figure PCTCN2022086254-appb-000005
其中,R1选自下组:巯基取代的C1-C4烷基、巯基取代的C2-C4烯基、巯基取代的C2-C4炔基。
在另一优选例中,所述R1为-CH 2-SH或CH 2CH 2-SH。
在另一优选例中,产生所述游离巯基的氨基酸为半胱氨酸。
在另一优选例中,所述突变抗体包括具有式(II)的氨基酸,从而提供所述烃酰基芳基:
Figure PCTCN2022086254-appb-000006
所述A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基;且
所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
在另一优选例中,所述A为苯基,且R为C1-C4烷基,优选地,R为甲基、乙基或异丙基。
在另一优选例中,产生所述烃酰基芳基的氨基酸选自下组:对乙酰基苯丙氨酸、对丙酰基苯丙氨酸。在另一优选例中,所述突变抗体为在野生型抗体中将两个位点分别引入(替换或插入)半胱氨酸和对乙酰基苯丙氨酸而获得。
在另一优选例中,所述突变抗体为靶向HER2的抗原结合片段(Fab),所述抗原结合片段包括一重链和一轻链;
其中,所述重链的氨基酸序列为SEQ ID No:1;且
其中,所述轻链的氨基酸序列为SEQ ID No:2。
在另一优选例中,步骤(b)中的反应是通过“一锅法”进行的。即,当前一个反应完成后,将另一药物分子直接加入前一反应的反应液中,调节反应条件并进行反应。
在另一优选例中,所述方法还包括分离纯化的后处理步骤,优选地,所述分离纯化可使用选自下组的方法进行:萃取法、色谱法(如分子筛层析)、电泳,或其组合。
在另一优选例中,步骤(b)中,反应溶剂含有pH缓冲体系的水溶液,如醋酸钠缓冲液。
在另一优选例中,所述抗体与带有羟胺(-O-NH 2)基团或修饰有羟胺基团的药物分子A的反应,包括一个或多个如下特征:
(1)pH为3.5-4.8,如3.8、4.0、4.2或4.5;
(2)所述反应的温度为37±5℃、37±3℃、37±2℃或37±1℃;
(3)所述反应的时间为1-36h,较佳地,6-24h,如12h、16或18h;和/或
(4)所述抗体与药物分子A的用量比为1:1-50当量,较佳地,1:5-30当量,如1:10当量、1:15当量、1:20当量或1:25当量。
在另一优选例中,所述抗体与带有马来酰亚胺基团或修饰有马来酰亚胺基团的药物分子B的反应,包括一个或多个如下特征:
(1)pH为5.2-6.2,如5.5、5.8或6.0;
(2)所述反应的温度为4±5℃、4±3℃、4±2℃或4±1℃;
(3)所述反应的时间为1-36h,较佳地,6-24h,如12h、16或18h;和/或
(4)所述抗体与药物分子B的用量比为1:1-50当量,较佳地,1:2-30当量,如1:5当量、1:10当量、1:15当量、或1:20当量。
在另一优选例中,所述药物分子A与药物分子B独立地选自下组:
示踪剂,如荧光素,放射元素标记的化合物等;有确定功能的化合物如细胞毒素、抗肿瘤药物、抗生素等,以及有确定功能的分子如核酸等。
例如,所述药物分子A与药物分子B可独立地选自靶向以下靶点(但并不限于)的药物:FoxOl、HDAC、DP-1、E2F、ABL、AMPK、BRK、BRSK I、BRSK2、BTK、CAMKK1、CAMKKα、CAMKKβ、Rb、Suv39HI、SCF、p19INK4D、GSK-3、 pi 8INK4、myc、细胞周期蛋白E、CDK2、CDK9、CDG4/6、环化素D、pl6INK4A、cdc25A、BMI1、SCF、Akt、CHKl/2、C 1δ、CK1γ、C 2、CLK2、CSK、DDR2、DYRK1A/2/3、EF2K、EPH-A2/A4/B1/B2/B3/B4、EIF2A 3、Smad2、Smad3、Smad4、Smad7、p53、p21Cipl、PAX、Fyn、CAS、C3G、SOS、Tal、Raptor、RACK-1、CRK、Rapl、Rac、KRas、NRas、HRas、GRB2、FAK、PI3K、spred、Spry、mTOR、MPK、LKBl、PAK 1/2/4/5/6、PDGFRA、PYK2、Src、SRPK1、PLC、PKC、PKA、PKBα/β、PKCα/γ/δ、PKD、PLKl、PRAK、PRK2、RIPK2、WAVE-2、TSC2、DAPKl、BAD、IMP、C-TAK1、TAKl、TAOl、TBK1、TESK1、TGFBR1、TIE2、TLK1、TrkA、TSSK1、TTBK1/2、TTK、Tpl2/cotl、MEK1、MEK2、PLDL Erkl、Erk2、Erk5、Erk8、p90RSK、PEA-15、SRF、p27KIP1、TIF la、HMGN1、ER81、MKP-3、c-Fos、FGF-R1、GCK、GSK3β、HER4、HIPK1/2/3/、IGF-1R、cdc25、UBF、LAMTOR2、Statl、StaO、CREB、JAK、Src、PTEN、NF-κB、HECTH9、Bax、HSP70、HSP90、Apaf-1、Cyto c、BCL-2、Bcl-xL、Smac、XIAP、半胱天冬酶-9、半胱天冬酶-3、半胱天冬酶-6、半胱天冬酶-7、CDC37、TAB、IKK、TRADD、TRAF2、R1P1、FLIP、TAKl、JNKl/2/3、Lck、A-Raf、B-Raf、C-Raf、MOS、MLKl/3、MN l/2、MSKl、MST2/3/4、MPSK1、MEKKl、ME K4、MEL、ASK1、MINK1、MKK 1/2/3/4/6/7、NE 2a/6/7、NUAK1、OSR1、SAP、STK33、Syk、Lyn、PDK1、PHK、PIM 1/2/3、Ataxin-1、mTORCl、MDM2、p21Wafl、细胞周期蛋白Dl、Lamln A、Tpl2、Myc、连环蛋白、Wnt、IKK-β、IKK-γ、IKK-α、IKK-ε、ELK、p65RelA、IRAKI、IRA 2、IRAK4、IRR、FADD、TRAF6、TRAF3、MKK3、MKK6、ROCK2、RSK1/2、SGK1、SmMLCK、SIK2/3、ULK1/2、VEGFR1、WNK l、YES1、ZAP70、MAP4K3、MAP4K5、MAPKlb、MAPKAP-K2K3、p38α/β/δ/γMAPK、Aurora A、Aurora B、Aurora C、MCAK、Clip、MAPKAPK、FAK、MARK 1/2/3/4、Mucl、SHC、CXCR4、Gap-1、Myc、β-连环蛋白/TCF、Cbl、BRM、Mcl1、BRD2、BRD3、BRD4、AR、RAS、ErbB3、EGFR、IRE1、HPK1、RIPK2和ERα,包括其所有变体、突变体、剪接变体、插入缺失体和融合体。
在另一优选例中,所述药物分子独立地为小分子药物。
在另一优选例中,所述药物分子独立地为抗肿瘤药物。
在另一优选例中,所述药物分子A与药物分子B为通过两种不同机制作用的药物。
在另一优选例中,所述药物分子A与药物分子B独立地选自下组:
PARP1/2抑制剂、诱导癌细胞DNA损伤的化疗药物、DNA烷基化类疗药 物、DNA或RNA合成抑制剂、EGFR、ALK或FGFR酪氨酸受体激酶抑制剂、KRAS、MEK或ERK肿瘤信号通路抑制剂。
在另一优选例中,所述药物分子A与药物分子B独立地选自下组:奥拉帕尼、卢卡帕尼、尼拉帕尼、甲氨蝶呤、卡培他滨、吉西他滨、去氧氟尿苷、培美曲塞二钠、帕唑帕尼、伊马替尼、埃罗替尼、拉帕替尼、吉非替尼、凡德他尼、赫赛汀、紫杉醇、长春瑞滨、多西他赛、多柔比星、羟基喜树碱、丝裂霉素、表柔比星、吡柔比星、博来霉素、来曲唑、他莫西芬、氟维司群、曲谱瑞林、氟他胺、亮丙瑞林、阿那曲唑、异环磷酰胺、白消安、环磷酰胺、卡莫司汀、尼莫司汀、司莫司汀、氮芥、马法兰、瘤可宁、卡铂、顺铂、奥沙利铂、络铂、拓扑特肯、喜树碱、拓扑替康、依维莫司、西罗莫斯、特癌适、6-巯基嘌呤、6-硫鸟嘌呤、硫唑嘌呤、菌素D、柔红霉素、阿霉素、米托蒽醌、争光霉素、普卡霉素或氨鲁米特。
本发明第二方面,提供了一种突变抗体,所述突变抗体包含两个反应基团,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基
Figure PCTCN2022086254-appb-000007
在另一优选例中,所述抗体选自下组:全长抗体、抗原结合片段、双特异性抗体或纳米抗体。
在另一优选例中,所述突变抗体为在野生型抗体中将两个位点分别替换为能够产生游离巯基(-SH)和烃酰基芳基的天然氨基酸或非天然氨基酸而获得。
在另一优选例中,所述氨基酸选自下组:L-氨基酸、D-氨基酸,或其组合。
在另一优选例中,所述突变抗体具有式:
HS-ab-A-(CO)-R    (III)
其中,所述ab代表抗体;
A-(CO)-R具有式
Figure PCTCN2022086254-appb-000008
其中,所述环A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基,
所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
在另一优选例中,所述烃酰基芳基具有式
Figure PCTCN2022086254-appb-000009
其中,R为C1-C4 烷基,优选地,R为甲基、乙基或异丙基。
在另一优选例中,所述突变抗体为在野生型抗体中将两个位点分别替换为半胱氨酸和对乙酰基苯丙氨酸而获得。
在另一优选例中,所述突变抗体通过基因工程方法制备得到。
在另一优选例中,所述突变抗体为靶向HER2的抗原结合片段,所述突变抗原结合片段包括一重链和一轻链;其中,
所述重链的氨基酸序列为SEQ ID No:1;且
所述轻链的氨基酸序列为SEQ ID No:2。
本发明第三方面,提供了如本发明第二方面所述的突变抗体的用途,用于制备抗体药物偶联物和/或抗体荧光偶联试剂。
在另一优选例中,所述抗体药物偶联物和/或抗体荧光偶联试剂是靶向肿瘤细胞的。
在另一优选例中,所述抗体药物偶联物和/或抗体荧光偶联试剂是所述抗原结合片段偶联两种不同的药物分子得到的双功能偶联药物。
本发明第四方面,提供了如本发明第一方面所述的制备方法制备的抗体药物偶联物,或其药学上可接受的盐。
本发明第五方面,提供了一种药物组合物,所述药物组合物包括:
如本发明第四方面所述的抗体药物偶联物,或其药学上可接受的盐;以及药学上可接受的载体。
本发明第六方面,提供了如本发明第四方面所述的抗体药物偶联物、或其药学上可接受的盐、或包含其的组合物的用途,用于制备一药物,所述药物用于选自下组的一种或多种用途:
A1)预防和/或抑制肿瘤细胞增殖;
A2)预防和/或抑制肿瘤生长;
A3)预防和/或治疗癌症;
A4)制备抗肿瘤药物;
A5)制备肿瘤细胞增殖抑制剂;
A6)肿瘤细胞成像、示踪。
在另一优选例中,所述肿瘤或肿瘤细胞为HER2高表达的肿瘤或肿瘤细胞。
在另一优选例中,所述肿瘤或肿瘤细胞选自下组:乳腺癌、肺癌、胰腺癌、卵巢癌、胃癌、纤维肉瘤、膀胱癌、卵巢癌、腺癌、结肠癌、骨癌、脑癌、神经细胞瘤、头颈癌、直肠癌、结肠癌、家族性腺瘤性息肉性癌、遗传性非息肉性结直肠癌、食管癌、唇癌、喉癌、下咽癌、舌癌、唾液腺癌、腺癌、甲状腺髓样癌、乳头状甲状腺癌、肾癌、肾实质癌、宫颈癌、子宫体癌、子宫内膜癌、绒毛膜癌、前列腺癌、睾丸癌、泌尿癌、黑素瘤、急性淋巴性白血病、慢性淋巴性白血病、急性骨髓性白血病、慢性粒细胞白血病、肝细胞癌、胆囊癌、支气管癌、小细胞肺癌、非小细胞肺癌、多发性骨髓瘤等。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一累述。
附图说明
图1为一种本发明的双功能突变抗体的结构示意图;
图2为一种本发明的双偶联抗体药物偶联物的结构示意图;
图3为一锅法获得双机制肿瘤治疗抗体偶联药物反应方案1:
图4为一锅法获得双机制肿瘤治疗抗体偶联药物反应方案2;
图5显示了通过蛋白质质谱技术确认纯化获得的anti HER2 DualFab(抗HER2双功能Fab)分子量符合理论值。
图6显示了免疫荧光验证纯化获得的anti HER2 DualFab的特异性。
图7显示了细胞活力测试验证纯化获得的anti HER2 DualFab的特异性。
图8显示了双偶联反应步骤的一个实施例。
图9显示了双偶联反应的反应液SDS-PAGE测试结及灰度分析。如图所示,在双偶联反应进行时,双偶联实验组与仅加入带有羟胺基团的Alexa 488的对照组和仅加入带有马来酰亚胺的Alexa 568的对照组相比,确认双偶联完成,并且根据荧光成像仪的结果进行灰度分析,表明两步偶联的比率达到了1:1。
图10为Alexa 488 Alexa 568 DualFab免疫荧光染色结果。
图11为Alexa 488 Alexa 568 DualFab处理SKBR3 0h、3h、6h、12h、18h及 24h的荧光拍摄结果。
图12为实施例3的反应式。
具体实施方式
本发明人经过广泛而深入的研究,通过大量筛选和测试,提供了一种抗体偶联药物的制备方法。本发明在常规的野生型抗体上构建了同时具有巯基和酰基反应基团的突变抗体,修饰后的突变抗体能够通过马来酰亚胺连接子和羟胺连接子通过“一锅法”偶联两种药物分子。所述反应获得的抗体药物偶联物,对目标细胞(如肿瘤细胞)具有多种靶向抑制作用,从而可实现的多机制的靶向治疗。在此基础上完成本发明。
术语
除非另有定义,否则本文中所用的全部技术术语和科学术语均具有如本发明所属领域普通技术人员通常理解的相同含义。
如本文所用,在提到具体列举的数值中使用时,术语“约”意指该值可以从列举的值变动不多于1%。例如,如本文所用,表述“约100”包括99和101和之间的全部值(例如,99.1、99.2、99.3、99.4等)。
如本文所用,术语“含有”或“包括(包含)”可以是开放式、半封闭式和封闭式的。换言之,所述术语也包括“基本上由…构成”、或“由…构成”。
如本文所用,术语“室温”或“常温”是指温度为4-40℃,较佳地,25±5℃。
如文本所用,术语“烷基”作为整体或另一基团的一部分,是指包含若干个碳原子的直链或支链烷基,其中“C1-C4烷基”是指具有1-4个碳原子的直链或支链烷基,包括1、2、3或4个碳原子的烷基,烷基优选例如C1-C2、C1-C3。典型的“烷基”包括但不限于甲基、乙基、丙基、异丙基、正丁基、叔丁基、异丁基。烷基还可以为亚烷基,如1、2、3或4个碳原子的亚烷基。
如文本所用,术语“烯基”是指具有至少2个碳原子和至少一个双键的直链或支链烃。烯基可包括任何数量的碳原子数,其中,“C2-C4烯基”是指具有2-4个碳原子和至少一个双键的直链或支链烃,例如C 2、C 2-C 3、C 2-C 4。烯基可具有任何合适数量的双键,包括但不限于1或2个。烯基的实例包括但不限于乙烯基(乙烯基团))、丙烯基、异丙烯基、1-丁烯基、2-丁烯基、异丁烯基、丁二烯基。烯基还可以为亚烯基,如2、3或4个碳原子的亚烯基。
如文本所用,术语“炔基”是指具有至少2个碳原子和至少一个三键的直链或支链烃。炔基可包括任何数量的碳原子,“C2-C4炔基”是指具有2-4个碳原子和至少一个三键的直链或支链烃,例如C 2、C 2-C 3、C 2-C 4。炔基的实例包括但不限于乙炔基、丙炔基、1-丁炔基、2-丁炔基、异丁炔基、仲丁炔基、丁二炔基。炔基还可以为亚炔基,如2、3或4个碳原子的亚炔基。
术语“芳基”是指芳香环状烃类化合物基团,其中,“C6-C10芳基”是指包含6、7、8、9或10个环碳原子的芳香环状烃类化合物基团,具有1-2个环,尤其指单环和双环基团,如苯基、联苯基或萘基。凡含有两个或两个以上芳香环(双环等),芳基基团的芳香环可由单键联接(如联苯)。“取代芳基”是指芳基中的一个或多个位置被取代,尤其是1-3个取代基,可在任何位置上取代。
如文本所用,术语“杂芳基”是指含有1-3个选自N、O、S原子的杂芳族体系,其中,“5-10元杂芳基”是指含有1-3个选自N、O、S原子的5-10元杂芳族体系。杂芳基优选5至10元环,更优选为5元或6元,杂芳基包括但不限于吡咯基、吡唑基、咪唑基、噁唑基、异噁唑基、噻唑基、噻二唑基、异噻唑基、呋喃基、吡啶基、吡嗪基、嘧啶基、哒嗪基、三氮嗪基、三氮唑基及四氮唑基等。“杂芳基”可以是取代的或者未取代的,当被取代时,取代基可以位于C原子或杂原子上。
如本文所述,术语“多个”是指两个或以上,如2、3、4、5或6。
如本文所用,“卤素”或“卤原子”指F、Cl、Br、和I。
如未特别说明,所述取代指各基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
突变抗体
本发明中,所述“野生型抗体”指在引入本发明的反应基团之前的抗体。所述抗体可以为任何所需的抗体,如全长抗体、抗原结合片段、双特异性抗体、纳米抗体等功能蛋白。
在不破坏(或在可接受程度内)所述抗体与靶标结合能力的情况下,引入所述两个反应基团(游离巯基和烃酰基芳基)对本领域技术人员而言是已知的或容易实现的。如可以通过插入、替换、修饰等方法引入反应基团。所述游离巯基和烃酰基芳基可为位于抗体分子中相同或不同的肽链上,如巯基位于轻链上, 烃酰基芳基位于重链上。
典型地,所述突变抗体为在野生型抗体中分别引入能够产生游离巯基(-SH)和烃酰基芳基的天然氨基酸或非天然氨基酸而获得。所述“引入”可以通过替换、插入或修饰获得。
优选地,可通过在野生型抗体分子中将一个半胱氨酸替换一个已有的氨基酸,从而引入游离的巯基;而在另一位点将一个对乙酰基苯丙氨酸替换另一已有的氨基酸,从而引入乙酰基苯基。优选地,所述被替换的位点为远离与靶点结合的结构域的位置。
可参考本发明的实施例,上述突变抗体可以通过常用的基因工程的方法构建、表达获得,或通过化学方法合成。在突变抗体的肽链序列确定的情况下,通过基因工程的方法构建、表达从而获得所述突变抗体的方法是已知的。
例如,所述抗体可选自靶向以下靶点(但并不限于)的抗体:CCR4、CD3、CD19、CD20、CD30、CD22、EGFR、EpCAM、HER2、PD-1、PD-L1,VEGF、VEGFA、VEGFR2等;以及与自身免疫疾病;哮喘、抗感染、血液病、心血管病、骨质疏松、多发性硬化症、阿尔兹海默症等疾病的相关蛋白,如C5蛋白、CD2、IL-5、PCSK9、RSVF、血小板糖蛋白Ⅱb/Ⅲa、RANK等蛋白和多肽。
抗体药物偶联物的制备方法
本发明提供了一种抗体药物偶联物的制备方法,包括步骤:
(a)提供一种包含两个反应基团的突变抗体,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基
Figure PCTCN2022086254-appb-000010
(b)按任意先后顺序,将所述突变抗体与带有羟胺(-O-NH 2)基团或修饰有羟胺基团的药物分子A和带有马来酰亚胺基团
Figure PCTCN2022086254-appb-000011
或修饰有马来酰亚胺基团的药物分子B反应,从而得到所述抗体药物偶联物。
本发明中,当所述突变抗体与带有羟胺基团或修饰有羟胺基团的药物分子A反应时,反应温度T1为37±10℃且pH为3-5。
本发明中,当所述突变抗体与带有马来酰亚胺基团或修饰有马来酰亚胺基团的药物分子B反应时,反应温度T2为4±10℃且pH为5.1-6.5。
如本文所用,所述“任意先后顺序”指所述突变抗体可以先与药物分子A反应,然后所得反应产物与药物分子B反应;或者,所述突变抗体先与药物分 子B反应,然后所得反应产物与药物分子A反应,如图3-4所示。
突变抗体中的所述烃酰基芳基中的羰基与药物分子B的-O-NH 2反应,生成-C=N-O-连接基团。
突变抗体中的-SH与药物分子A的马来酰亚胺基团反应。
本发明人发现,上述两组基团具有合适的反应性能,使得两个反应可以在37℃及以下的温度反应,只需调节温度和pH即可经一锅法完成两种药物分子的偶联。
优选地,产生所述游离巯基的氨基酸具有式
Figure PCTCN2022086254-appb-000012
其中,R1选自下组:巯基取代的C1-C4烷基、巯基取代的C2-C4烯基、巯基取代的C2-C4炔基。
在另一优选例中,所述R1为-CH 2-SH或CH 2CH 2-SH。
更优选地,产生所述游离巯基的氨基酸为半胱氨酸。
优选地,产生所述烃酰基芳基的氨基酸具有式
Figure PCTCN2022086254-appb-000013
所述A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基;且
所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
更优选地,产生所述烃酰基芳基的氨基酸选自下组:对乙酰基苯丙氨酸、对丙酰基苯丙氨酸。
本发明中,对于药物分子A和药物分子B的种类没有要求,可根据需要选择,例如可以为本领域常见的小分子药物、核酸、荧光分子等。本领域技术人员理解,对于一个已知的药物分子,在不破坏其活性结构的基础上,引入马来酰亚胺基团或羟胺基团的方法是本领域已知的。
典型地,所述药物分子A与药物分子B可独立地选自靶向以下靶点(但并不限于)的药物:FoxOl、HDAC、DP-1、E2F、ABL、AMPK、BRK、BRSK I、BRSK2、BTK、CAMKK1、CAMKKα、CAMKKβ、Rb、Suv39HI、SCF、p19INK4D、 GSK-3、pi 8INK4、myc、细胞周期蛋白E、CDK2、CDK9、CDG4/6、环化素D、pl6INK4A、cdc25A、BMI1、SCF、Akt、CHKl/2、C 1δ、CK1γ、C 2、CLK2、CSK、DDR2、DYRK1A/2/3、EF2K、EPH-A2/A4/B1/B2/B3/B4、EIF2A 3、Smad2、Smad3、Smad4、Smad7、p53、p21Cipl、PAX、Fyn、CAS、C3G、SOS、Tal、Raptor、RACK-1、CRK、Rapl、Rac、KRas、NRas、HRas、GRB2、FAK、PI3K、spred、Spry、mTOR、MPK、LKBl、PAK 1/2/4/5/6、PDGFRA、PYK2、Src、SRPK1、PLC、PKC、PKA、PKBα/β、PKCα/γ/δ、PKD、PLKl、PRAK、PRK2、RIPK2、WAVE-2、TSC2、DAPKl、BAD、IMP、C-TAK1、TAKl、TAOl、TBK1、TESK1、TGFBR1、TIE2、TLK1、TrkA、TSSK1、TTBK1/2、TTK、Tpl2/cotl、MEK1、MEK2、PLDL Erkl、Erk2、Erk5、Erk8、p90RSK、PEA-15、SRF、p27KIP1、TIF la、HMGN1、ER81、MKP-3、c-Fos、FGF-R1、GCK、GSK3β、HER4、HIPK1/2/3/、IGF-1R、cdc25、UBF、LAMTOR2、Statl、StaO、CREB、JAK、Src、PTEN、NF-κB、HECTH9、Bax、HSP70、HSP90、Apaf-1、Cyto c、BCL-2、Bcl-xL、Smac、XIAP、半胱天冬酶-9、半胱天冬酶-3、半胱天冬酶-6、半胱天冬酶-7、CDC37、TAB、IKK、TRADD、TRAF2、R1P1、FLIP、TAKl、JNKl/2/3、Lck、A-Raf、B-Raf、C-Raf、MOS、MLKl/3、MN l/2、MSKl、MST2/3/4、MPSK1、MEKKl、ME K4、MEL、ASK1、MINK1、MKK 1/2/3/4/6/7、NE 2a/6/7、NUAK1、OSR1、SAP、STK33、Syk、Lyn、PDK1、PHK、PIM 1/2/3、Ataxin-1、mTORCl、MDM2、p21Wafl、细胞周期蛋白Dl、Lamln A、Tpl2、Myc、连环蛋白、Wnt、IKK-β、IKK-γ、IKK-α、IKK-ε、ELK、p65RelA、IRAKI、IRA 2、IRAK4、IRR、FADD、TRAF6、TRAF3、MKK3、MKK6、ROCK2、RSK1/2、SGK1、SmMLCK、SIK2/3、ULK1/2、VEGFR1、WNK l、YES1、ZAP70、MAP4K3、MAP4K5、MAPKlb、MAPKAP-K2K3、p38α/β/δ/γMAPK、Aurora A、Aurora B、Aurora C、MCAK、Clip、MAPKAPK、FAK、MARK 1/2/3/4、Mucl、SHC、CXCR4、Gap-1、Myc、β-连环蛋白/TCF、Cbl、BRM、Mcl1、BRD2、BRD3、BRD4、AR、RAS、ErbB3、EGFR、IRE1、HPK1、RIPK2和ERα,包括其所有变体、突变体、剪接变体、插入缺失体和融合体。
在另一优选例中,所述药物分子A与药物分子B为两种不同机制作用的药物。
抗体药物偶联物
如本文所用,术语“抗体药物偶联物”、“抗体偶联药物”、“ADC”可互换使用。
本发明还提供了上述方法制备的抗体药物偶联物或其药学上可接受的盐。
通常,所述抗体药物偶联物为用于治疗或诊断的药物。其至少具备抗体、药物分子A和药物分子B的生物活性。此外,其还可实现不用机理药物联合用药时产生的协同效果,对细胞具有靶向给药能力,减小用药量、并降低药物的耐药性。
药物组合物及应用
本发明提供了一种药物组合物,其包括上述抗体药物偶联物或其药学上可接受的盐作为活性成分,以及药学上可接受的载体。
特别地,本发明的方法可以制备各种靶向特定细胞的抗体药物偶联物。例如,可以靶向(但并不限于)肿瘤细胞、免疫细胞、炎症细胞等。进而使抗体或偶联的药物发挥作用,针对性的实现靶向治疗。
“药学上可接受的赋形剂”和“药学上可接受的载体”是指有助于活性剂的配制和/或施用和/或被个体吸收的物质,并且可以包含在本公开的组合物中而不引起对该个体的显著不利的毒理作用。药学上可接受的载体和赋形剂的非限制性实例包括水、NaCl、生理盐水溶液、乳酸林格氏液、常规蔗糖、常规葡萄糖、粘合剂、填充剂、崩解剂、润滑剂、包衣、甜味剂、风味剂、盐溶液(例如林格溶液)、醇、油、明胶、碳水化合物,诸如乳糖、直链淀粉或淀粉、脂肪酸酯、羟甲基纤维素、聚乙烯吡咯烷和颜料等。这样的制剂可以被灭菌,并且如果需要,与不会有害地与本文提供的化合物反应或干扰本文提供的化合物的活性的辅助剂例如润滑剂、防腐剂、稳定剂、润湿剂、乳化剂、影响渗透压的盐、缓冲剂、着色剂和/或芳香物质等混合。本领域普通技术人员将认识到其他药物载体和赋形剂适用于公开的化合物。
在某些实施例中,本发明的药物组合物可以以固体或液体形式。
含有本发明活性成分的药物可以是适宜的口服剂型,例如片剂,药片,含片,水溶性或油性悬液,分散乳胶粉或颗粒,乳剂,硬或软胶囊或糖浆或酏剂。口服使用的药物可根据药物成分制造商的已知工艺方法来制备,这些组合物可包括下述一种或多种药剂,例如甜味剂,调味剂,着色剂和保护剂,以便提供优雅和美味的药品制剂。药片含有与非毒性药学上可接受的赋形剂混合的活性成分,这些赋形剂适合于生产片剂。这些赋形剂的例子有,惰性稀释剂,例如碳酸钙、碳酸钠、乳糖、钙磷酸盐或磷酸钠;制粒,崩解剂,例如,玉米淀粉或褐藻酸;结合剂,例如如淀粉、明胶或阿拉伯胶,以及润滑剂,例如硬脂酸 镁、硬脂酸或滑石粉。该药片可无涂层,也可有涂层,以延迟在胃肠道的降解和吸收,从而在较长时期内维持活性。
通过任意合适的途径,包括口服地、肠胃外地、通过吸入喷雾、局部地、直肠地、鼻地、含服地、阴道地或经由植入型药盒,可以将活性化合物施用给受试者。本文使用的术语“肠胃外的”包括皮下的、静脉内的、肌肉内的、关节内的、滑膜内的、胸骨内的、鞘内的、肝内的、病灶内的(intralesional)和颅内的注射或输注技术。优选地,口服地、腹膜内地或静脉内地施用所述组合物。
适合于口服施用的本发明的药物组合物典型地将是以固体形式的离散单元,例如以片剂、胶囊、扁囊剂、粉末、颗粒、锭剂、贴片、栓剂、丸剂的形式,或以液体形式,例如液体制剂、可注射的或可输注的溶液或悬浮液。
向个体提供治疗有效量的化合物的精确量将取决于给药方式、疾病和/或病症的类型和严重程度以及个体的特征,例如一般健康状况、年龄、性别、体重和对药物的耐受性。本领域普通技术人员将能够根据这些和其他因素确定合适的剂量。当与其他治疗剂组合施用时,任何其他治疗剂的“治疗有效量”将取决于所用药物的类型。合适的剂量对于批准的治疗剂是已知的,并且可以由本领域普通技术人员根据个体的状况、治疗的病症类型和通过以下使用的本发明化合物的量进行调整,例如,在文献中报道和在Physician’s Desk Reference(第57版,2003)中推荐的剂量。优选地,应如此配制组合物,使得可以将0.01-100mg/kg体重/天的抑制剂剂量施用给接受这些组合物的患者。在某些实施方案中,本发明的组合物提供了0.01mg至50mg的剂量。在其它实施方案中,提供了0.lmg-25mg或5mg-40mg的剂量。
本发明的药物组合物或治疗剂的给药对象的实例包括哺乳动物(例如,人、小鼠、大鼠、仓鼠、兔、猫、狗、牛、绵羊、猴等)。
本发明还提供了一种药物组合物的制备方法,包括步骤:将药学上可接受的载体与本发明所述的抗体药物偶联物或其药学上可接受的盐进行混合,从而形成药物组合物。
本发明还提供了一种治疗方法,它包括步骤:给需要治疗的对象施用本发明中所述抗体药物偶联物,或其药学上可接受的盐,或施用本发明所述的药物组合物,用于选择性地抑制癌症(如HER2介导的癌症)。
本发明的主要优点包括:
1.本发明的提供了一种具有双偶联潜力的突变抗体,所述突变抗体包括能 与药物连接的两种反应基团(巯基和乙酰基芳基),所述突变抗体可以具有单引入不可做到的两种反应的依次连续进行,简单高效获得功能升级的抗体偶联物。同时引入两个反应基团,偶联2个不同机制的药物,产生双靶点药物。
2.本发明的方法可以在4-37℃的常温下,通过一锅法制备,可保持抗体的结构和功能,且操作简单、安全高效,设备要求低,适合大规模生产。
3.本发明的突变抗体提供了两种不同的反应基团,反应时根据药物分子类型的不同,反应位点是一一对应的,制备得到的抗体药物偶联物结构一致,纯度高。
4.本发明的方法制备的抗体药物偶联物具有提高的药效、不易产生耐药性,毒性低。
5.本发明的方法制备的抗体药物偶联物具有优异的药效和药代动力学性质,适于成药。
下面结合具体实施,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件,例如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
表1 实验仪器
Figure PCTCN2022086254-appb-000014
表2 试剂
Figure PCTCN2022086254-appb-000015
Figure PCTCN2022086254-appb-000016
实施例1
获得具有双偶联潜力的抗体片段
首先确定在抗HER2抗体轻链124位定点插入半胱氨酸,获得带有突变的轻链质粒。为使抗体片段具有良好羰基反应性,选择具有反应活性良好的对乙酰基苯丙氨酸(pAcF)作为掺入目标氨基酸,之后构建对乙酰基苯丙氨酸正交体系。然后在抗HER2抗体CH1重链121位定点插入琥珀密码子,获得带有突变的HC质粒,最后和已经构建完成的对乙酰基苯丙氨酸对应的氨酰tRNA合成酶质粒以及突变LC质粒共转化获得DualFab(双功能的Fab)的表达菌株(LC Q124C,HC A121X(X为4-乙酰基-L-苯丙氨酸)。
其中,所述重链的氨基酸序列为(SEQ ID No:1):
Figure PCTCN2022086254-appb-000017
其中,所述轻链的氨基酸序列为(SEQ ID No:2):
Figure PCTCN2022086254-appb-000018
利用2YT培养基进行培养扩增,OD达到0.6-0.8时加入1mM IPTG,0.2%阿拉伯糖和1mM对乙酰基苯丙氨酸,16度培养24小时,纯化获得目标分子 DualFab。送样蛋白质谱鉴定。
如图5所示。确认纯化获得的anti HER2 DualFab重轻链分子量符合理论值。
如图6所示。通过免疫荧光的实验方法确认Dualfab可以完全将HER2高表达的细胞和其他细胞区分开,体现出良好的特异性。
如图7所示。通过细胞活力测试实验表明,DualFab与trastuzumab(阳性对照)作用细胞120小时后,均对HER2高表达的SKBR3的细胞生长有特异性抑制而HER2低表达的MCF-7细胞的生长并没有受到影响。
实施例2
通过分步偶联法获得双荧光素偶联物
如图8进行双偶联反应步骤。
将纯化获得的DualFab加入到100mM pH 4.5的醋酸钠缓冲液中,然后加入20当量的带有羟胺基团的Alexa 488,37℃16-18小时。调节pH至6.0,加入10当量带有马来酰亚胺的Alexa 568,4℃过夜。分子筛层析去除过量的小分子,收集洗脱液,浓缩获得双荧光素DualFab偶联物。
双偶联反应的反应液SDS-PAGE测试结及灰度分析如图9所示,在双偶联反应进行时,双偶联实验组与仅加入带有羟胺基团的Alexa 488的对照组和仅加入带有马来酰亚胺的Alexa 568的对照组相比,双偶联确实已完成,并且根据荧光照胶仪的结果进行灰度分析发现,两步偶联的比率为1:1。之后利用免疫荧光染色验证Alexa 488 Alexa 568 DualFab双偶联物识别HER2的特异性。在细胞固定通透封闭之后,加入Alexa 488 Alexa 568 DualFab双偶联物4℃孵育过夜,洗涤后用含有DAPI的封片剂封片,拍照获得实验结果。
如图10所示:Alexa 488 Alexa 568 DualFab双偶联物可以特异性的识别HER2高表达的细胞系SKBR3。接下来利用双荧光素DualFab偶联物通过活细胞成像检测双偶联物的内吞情况。首先发现Alexa 488 Alexa 568 DualFab双偶联物在加入活细胞监测体系不久就特异的结合到SKBR3的细胞膜上,并且Alexa 568和Alexa 488的信号几乎是共定位。随着监测时间的增长,Alexa 488 Alexa 568 DualFab双偶联物在SKBR3中的荧光信号不断由细胞膜累积到细胞质中,直观的展示了ADCs的内吞过程。
图11记录了在0h,3h,6h,12h,18h和24h不同时间点Alexa 488 Alexa  568 DualFab双偶联物在SKBR3中的内吞过程。在0-3h时,Alexa 488 Alexa 568 DualFab双偶联物仅是结合在SKBR3的膜上。随着时间的推移,从6h,12h,18h到24h,随着黄色的箭头指示,Alexa 488和Alexa 568的荧光信号不断由胞膜到胞质内累积。
以上实验结果表明本发明通过基因工程方法在抗体轻链中引入裸露的巯基,在CH1重链部分引入带有功能性反应基团羰基的非自然氨基酸对乙酰基苯丙氨酸,然后通过构建表达菌株在大肠杆菌中大量表达,纯化后获得具有双偶联官能团的Fab(DualFab),可用来制备双机制或多机制的抗体偶联药物,高效治疗肿瘤以及其他疾病。
实施例3
通过分步偶联法获得双功能抗肿瘤偶联物
将纯化获得的DualFab加入到100mM pH 4.5的醋酸钠缓冲液中,然后加入20当量的带有羟胺基团的NH2O-PEG4-vc-PAB-PBD,37℃16-18小时。调节pH至6.0,加入10当量带有马来酰亚胺基团的A-PEG4-vc-PAB-MMAE连接子药物偶联物,4℃过夜。分子筛层析去除过量的小分子,收集洗脱液,浓缩获得双功能抗肿瘤偶联物。反应式如图12所示。
讨论
本发明的双偶联的方法可以通过一锅法在同一抗体上引入两种以上药物分子,所制备的抗体药物偶联物可同时将两种不同机制的药物带入目标细胞,起到优于协同用药的同时间同空间的作用,降低耐药出现的风险,提高药效。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (10)

  1. 一种抗体药物偶联物的制备方法,其特征在于,包括步骤:
    (a)提供一种包含两个反应基团的突变抗体,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基
    Figure PCTCN2022086254-appb-100001
    (b)按任意先后顺序,将所述突变抗体与带有羟胺(-O-NH 2)基团或修饰有羟胺基团的药物分子A和带有马来酰亚胺基团
    Figure PCTCN2022086254-appb-100002
    或修饰有马来酰亚胺基团的药物分子B反应,从而得到所述抗体药物偶联物;
    其中,当所述突变抗体与带有羟胺基团或修饰有羟胺基团的药物分子A反应时,反应温度T1为37±10℃且pH为3-5;且
    当所述突变抗体与带有马来酰亚胺基团或修饰有马来酰亚胺基团的药物分子B反应时,反应温度T2为4±10℃且pH为5.1-6.5。
  2. 如权利要求1所述的制备方法,其特征在于,所述抗体选自下组:全长抗体、抗原结合片段、双特异性抗体或纳米抗体。
  3. 如权利要求1所述的制备方法,其特征在于,所述烃酰基芳基具有式
    Figure PCTCN2022086254-appb-100003
    其中,所述环A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
    且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基,
    所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
  4. 如权利要求1所述的制备方法,其特征在于,所述突变抗体包括具有式(I)的氨基酸,从而提供巯基:
    Figure PCTCN2022086254-appb-100004
    其中,R1选自下组:巯基取代的C1-C4烷基、巯基取代的C2-C4烯基、巯基取代的C2-C4炔基。
  5. 如权利要求1所述的制备方法,其特征在于,所述突变抗体包括具有式(II)的氨基酸,从而提供所述烃酰基芳基:
    Figure PCTCN2022086254-appb-100005
    所述A选自下组:取代或未取代的C6-C10的芳基、取代或未取代的包括1-3个选自O、N、S的5-10元杂芳基;
    且R选自下组:取代或未取代的C1-C4烷基、取代或未取代的C2-C4烯基、取代或未取代的C2-C4炔基;且
    所述取代指基团上的一个或多个H独立地被选自下组的基团取代:卤素、C1-C4烷基、C1-C4烷氧基、C2-C4烯基、C2-C4炔基、-NO 2、-CN或-OH。
  6. 一种突变抗体,所述突变抗体包含两个反应基团,其中,所述反应基团一个为游离巯基(-SH),另一个为烃酰基芳基
    Figure PCTCN2022086254-appb-100006
  7. 如权利要求6所述的突变抗体的用途,其特征在于,用于制备抗体药物偶联物和/或抗体荧光偶联试剂。
  8. 如权利要求1所述的制备方法制备的抗体药物偶联物,或其药学上可接受的盐。
  9. 一种药物组合物,其特征在于,所述药物组合物包括:
    如权利要求8所述的抗体药物偶联物,或其药学上可接受的盐;以及药学上可接受的载体。
  10. 如权利要求4所述的抗体药物偶联物、或其药学上可接受的盐、或包含其的组合物的用途,其特征在于,用于制备一药物,所述药物用于选自下组的一种或多种用途:
    A1)预防和/或抑制肿瘤细胞增殖;
    A2)预防和/或抑制肿瘤生长;
    A3)预防和/或治疗癌症;
    A4)制备抗肿瘤药物;
    A5)制备肿瘤细胞增殖抑制剂;和/或
    A6)肿瘤细胞成像、示踪。
PCT/CN2022/086254 2021-04-12 2022-04-12 一种抗体药物偶联物的制备方法 WO2022218288A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110390274.6A CN115192731B (zh) 2021-04-12 2021-04-12 一种抗体药物偶联物的制备方法
CN202110390274.6 2021-04-12

Publications (1)

Publication Number Publication Date
WO2022218288A1 true WO2022218288A1 (zh) 2022-10-20

Family

ID=83570785

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/086254 WO2022218288A1 (zh) 2021-04-12 2022-04-12 一种抗体药物偶联物的制备方法

Country Status (2)

Country Link
CN (1) CN115192731B (zh)
WO (1) WO2022218288A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117143114A (zh) * 2023-10-30 2023-12-01 深圳大学 一种BRD4和Src双靶点抑制剂及其制备方法与应用

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201139666A (en) * 2010-03-26 2011-11-16 Kyowa Hakko Kirin Co Ltd Novel antibody having modification site introduced therein, and antibody fragment
WO2021031930A1 (zh) * 2019-08-19 2021-02-25 沈阳药科大学 抗体的突变体及其应用
CN112601522A (zh) * 2018-07-09 2021-04-02 辛瑟斯治疗股份有限公司 抗体-alk5抑制剂偶联物及其用途

Family Cites Families (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3019522B1 (en) * 2013-07-10 2017-12-13 Sutro Biopharma, Inc. Antibodies comprising multiple site-specific non-natural amino acid residues, methods of their preparation and methods of their use
RU2016134258A (ru) * 2013-10-15 2018-02-28 Сорренто Терапьютикс Инк. Конъюгат лекарственного средства с направляющей молекулой и двумя различными лекарственными средствами
CN113941007B (zh) * 2020-07-16 2024-06-07 成都科岭源医药技术有限公司 一种串联的双药物链接组装单元及其应用

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
TW201139666A (en) * 2010-03-26 2011-11-16 Kyowa Hakko Kirin Co Ltd Novel antibody having modification site introduced therein, and antibody fragment
CN112601522A (zh) * 2018-07-09 2021-04-02 辛瑟斯治疗股份有限公司 抗体-alk5抑制剂偶联物及其用途
WO2021031930A1 (zh) * 2019-08-19 2021-02-25 沈阳药科大学 抗体的突变体及其应用

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
J. Y. AXUP, K. M. BAJJURI, M. RITLAND, B. M. HUTCHINS, C. H. KIM, S. A. KAZANE, R. HALDER, J. S. FORSYTH, A. F. SANTIDRIAN, K. STA: "Synthesis of site-specific antibody-drug conjugates using unnatural amino acids", PROCEEDINGS OF THE NATIONAL ACADEMY OF SCIENCES, NATIONAL ACADEMY OF SCIENCES, vol. 109, no. 40, 2 October 2012 (2012-10-02), pages 16101 - 16106, XP055076259, ISSN: 00278424, DOI: 10.1073/pnas.1211023109 *
ZHANG LIN, WANG ZEWEI, WANG ZHIYUAN, LUO FANG, GUAN MINGFENG, XU MEIMEI, LI YUNDONG, ZHANG YAOYANG, WANG ZHAOYIN, WANG WENYUAN: "A Simple and Efficient Method to Generate Dual Site-Specific Conjugation ADCs with Cysteine Residue and an Unnatural Amino Acid", BIOCONJUGATE CHEMISTRY, AMERICAN CHEMICAL SOCIETY, US, vol. 32, no. 6, 16 June 2021 (2021-06-16), US , pages 1094 - 1104, XP055977120, ISSN: 1043-1802, DOI: 10.1021/acs.bioconjchem.1c00134 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN117143114A (zh) * 2023-10-30 2023-12-01 深圳大学 一种BRD4和Src双靶点抑制剂及其制备方法与应用
CN117143114B (zh) * 2023-10-30 2024-02-20 深圳大学 一种BRD4和Src双靶点抑制剂及其制备方法与应用

Also Published As

Publication number Publication date
CN115192731A (zh) 2022-10-18
CN115192731B (zh) 2023-09-26

Similar Documents

Publication Publication Date Title
JP7259104B2 (ja) 抗trop2抗体-薬物コンジュゲート
CN110464847B (zh) 抗her2抗体-药物偶联物
CA2925915C (en) Anti-epcam antibodies and methods of use
TW202122115A (zh) 包含抗人類ror1抗體之抗體—藥物結合物及其用途
EA038192B1 (ru) Фармацевтические композиции, содержащие диастереомеры макролида, способы их синтезирования и терапевтическое применение
BR112021011014A2 (pt) Anticorpos anti-claudina e usos destes
US20120219568A1 (en) Epidithiodioxopiprazines and uses thereof in treating cancer
WO2016108587A1 (ko) 리피바디 유도체-약물 복합체, 그 제조방법 및 용도
US10293025B2 (en) Oral administration compositions comprising an OB-fold protein variant
CN112010839B (zh) 靶向丝/苏氨酸激酶抑制剂的晶型
WO2022218288A1 (zh) 一种抗体药物偶联物的制备方法
CN108368143A (zh) 用于癌症疗法的单马来酰亚胺官能化的铂化合物
JP6705953B2 (ja) ドライバーオンコジーンの遺伝子変異を標的にアルキル化する新規アルキル化剤
WO2022262516A1 (zh) 连接子及其缀合物
WO2021016361A1 (en) Methods and compositions for treating cancer using peptide nucleic acid-based agents
JP7069135B2 (ja) アルブミンベースのナノ医薬品を使用するための組成物及び方法
CN108264510A (zh) 一种选择性抑制激酶化合物及其用途
CN110882251A (zh) 一种10-HCPT和Crizotinib偶联化合物合成方法及抗肿瘤应用
Yu et al. The first marine dual-drug cocrystal of cytarabine with 5-fluorouracil having synergistic antitumor effects shows superior biopharmaceutical peculiarities by oral administration
TW202104259A (zh) 癌治療用醫藥
CN108498518B (zh) 七元环小檗碱类似物及其药物组合物在制备治疗多发性骨髓瘤的药物中的应用
WO2019230669A1 (ja) Runx結合配列を標的とする医薬組成物およびrunx阻害剤
CN104974252B (zh) 一种抑制肿瘤生长的抗体-小分子药物偶联物及其制备方法和用途
WO2023103854A1 (zh) 一种亲和力改善的抗体-药物偶联物、其制备方法及应用
TWI852695B (zh) 抗體-藥物結合物之用途

Legal Events

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

Ref document number: 22787512

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 22787512

Country of ref document: EP

Kind code of ref document: A1