US20230357259A1 - Bioactive conjugate, preparation method therefor and use thereof - Google Patents

Bioactive conjugate, preparation method therefor and use thereof Download PDF

Info

Publication number
US20230357259A1
US20230357259A1 US18/155,650 US202318155650A US2023357259A1 US 20230357259 A1 US20230357259 A1 US 20230357259A1 US 202318155650 A US202318155650 A US 202318155650A US 2023357259 A1 US2023357259 A1 US 2023357259A1
Authority
US
United States
Prior art keywords
compound
membered
alkyl
antibody
mmol
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
US18/155,650
Other languages
English (en)
Inventor
Jiaqiang Cai
Shuai SONG
Tongtong Xue
Liang Xiao
Hanwen DENG
Qiang Tian
Jing Wang
Dengnian LIU
Liping Liu
Haimin Yu
Zhouning YANG
Xu Cao
Guoqing Zhong
Lichun Wang
Jingyi Wang
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Original Assignee
Sichuan Kelun Biotech Biopharmaceutical Co Ltd
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 Sichuan Kelun Biotech Biopharmaceutical Co Ltd filed Critical Sichuan Kelun Biotech Biopharmaceutical Co Ltd
Priority to US18/155,650 priority Critical patent/US20230357259A1/en
Assigned to SICHUAN KELUN-BIOTECH BIOPHARMACEUTICAL CO., LTD. reassignment SICHUAN KELUN-BIOTECH BIOPHARMACEUTICAL CO., LTD. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: CAI, JIAQIANG, CAO, XU, DENG, Hanwen, LIU, Dengnian, LIU, LIPING, SONG, Shuai, TIAN, QIANG, WANG, JING, WANG, JINGYI, WANG, Lichun, XIAO, LIANG, XUE, Tongtong, YANG, Zhouning, YU, Haimin, ZHONG, Guoqing
Publication of US20230357259A1 publication Critical patent/US20230357259A1/en
Pending legal-status Critical Current

Links

Images

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/02Linear peptides containing at least one abnormal peptide link
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D491/00Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00
    • C07D491/22Heterocyclic compounds containing in the condensed ring system both one or more rings having oxygen atoms as the only ring hetero atoms and one or more rings having nitrogen atoms as the only ring hetero atoms, not provided for by groups C07D451/00 - C07D459/00, C07D463/00, C07D477/00 or C07D489/00 in which the condensed system contains four or more hetero rings
    • 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
    • A61K47/6889Conjugates wherein the antibody being the modifying agent and wherein the linker, binder or spacer confers particular properties to the conjugates, e.g. peptidic enzyme-labile linkers or acid-labile linkers, providing for an acid-labile immuno conjugate wherein the drug may be released from its antibody conjugated part in an acidic, e.g. tumoural or environment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/08Peptides having 5 to 11 amino acids
    • 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
    • A61K47/62Medicinal 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 a protein, peptide or polyamino acid
    • A61K47/65Peptidic linkers, binders or spacers, e.g. peptidic enzyme-labile linkers
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68031Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being an auristatin
    • 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
    • A61K47/6801Drug-antibody or immunoglobulin conjugates defined by the pharmacologically or therapeutically active agent
    • A61K47/6803Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates
    • A61K47/68037Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a camptothecin [CPT] or derivatives
    • 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
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • 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
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6851Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell
    • A61K47/6855Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site the antibody targeting a determinant of a tumour cell the tumour determinant being from breast cancer cell
    • 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
    • A61K47/6835Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment the modifying agent being an antibody or an immunoglobulin bearing at least one antigen-binding site
    • A61K47/6883Polymer-drug antibody conjugates, e.g. mitomycin-dextran-Ab; DNA-polylysine-antibody complex or conjugate used for therapy
    • A61K47/6885Polymer-drug antibody conjugates, e.g. mitomycin-dextran-Ab; DNA-polylysine-antibody complex or conjugate used for therapy the conjugate or the polymer being a starburst, a dendrimer, a cascade
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants
    • C07K16/30Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants from tumour cells
    • CCHEMISTRY; METALLURGY
    • 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
    • C07K19/00Hybrid peptides, i.e. peptides covalently bound to nucleic acids, or non-covalently bound protein-protein complexes
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K5/00Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof
    • C07K5/04Peptides containing up to four amino acids in a fully defined sequence; Derivatives thereof containing only normal peptide links
    • C07K5/06Dipeptides
    • C07K5/06008Dipeptides with the first amino acid being neutral
    • C07K5/06017Dipeptides with the first amino acid being neutral and aliphatic
    • C07K5/06034Dipeptides with the first amino acid being neutral and aliphatic the side chain containing 2 to 4 carbon atoms
    • C07K5/06052Val-amino acid
    • 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

Definitions

  • the disclosure belongs to the technical field of medical technology, and relates to a bioactive molecule conjugate, preparation method thereof, and use in the prevention and/or treatment of a disease associated with an abnormal cell activity, including but not limited to the use in the prevention and/or treatment of a neoplastic disease.
  • Chemotherapy was once a standard therapy for cancer, but bioactive molecules having high killing effect can mistakenly kill normal cells, resulting in serious side effects.
  • Targeted therapy has become a hot research topic in the field of oncology due to the targetability and anti-tumor activity.
  • bio-macromolecular drugs e.g., therapeutic antibodies or antibody fragments
  • targeted small molecule ligands e.g., targeted small molecule ligands.
  • bio-macromolecular drugs e.g., therapeutic antibodies or antibody fragments
  • bio-macromolecular drugs have limited curative effects on solid tumors; in addition, bioactive molecules often lack targetability and accidentally injure normal cells and cause serious toxic and side effects, despite of their high killing effect on cancer cells.
  • ADCs antibody-drug conjugates
  • the ADC combine the targeting effect of antibodies and the activity of bioactive molecules making it a “biological missile”.
  • the ADC is guided by antibodies to bind to target cells, and then is internalized by cells to release drugs thereby treating relevant diseases. Due to the specificity and targetability to tumor cell related targets, the application values of antibodies not only are reflected in the treatment, but also become an ideal carrier for drug targeted delivery, and reduce side effects of drugs.
  • Small molecule drug conjugates are designed on the basis of same principle as antibody-drug conjugates (ADCs); that is, coupling bioactive molecules with some small molecule ligands which can selectively bind to receptors on the surfaces of tumor cells through chemical processes, thereby improving the targetability of effector molecules (bioactive molecules) to tumor cells.
  • ADCs antibody-drug conjugates
  • the difference between the SMDCs and the ADCs is that the SMDCs use small molecule ligands instead of antibodies, and there is not yet SMDC available on the market.
  • An ADC generally consists of an antibody, a bioactive molecule and a linker.
  • the bioactive molecule is covalently coupled to the antibody via the linker; the antibody (e.g., monoclonal antibodies) can specifically recognize a specific target on the surface of a tumor cell, thus guiding the ADC to reach the surface of cancer cell and enabling the ADC to enter the cancer cell through endocytosis effect; then the bioactive molecule is released in the cancer cell to achieve the effect of specifically killing the cancer cell without damaging normal tissue cells.
  • the antibody e.g., monoclonal antibodies
  • Lysine is the most common linking site in antibodies, and ⁇ -amino groups thereof can react with activated carboxyl groups of linkers to form amide bonds.
  • Techniques for site-specific coupling are currently available, that is, carboxyl groups of linkers are activated, and then form amide bonds with specific lysine ⁇ -amino groups in antibodies to complete the coupling.
  • amide bonds are prone to hydrolysis under the action of enzymes enzymes in vivo, as a result, bioactive molecules and antibodies dissociate before reaching target cells resulted in increasing toxicity while losing tagetability of ADCs.
  • Thio groups of antibody cysteine usually exist in the the form of disulfide bonds.
  • the disulfide bonds in the antibody can be opened to provide multiple free sulfhydryl groups as coupling sites.
  • One method of coupling with the sulfhydryl groups of the antibody is Michael addition reaction between the free sulfhydryl groups of the antibody and maleimide, or two Michael addition reactions between a specific substrate and free sulfhydryl groups of the antibody to form a sulfur bridge bond in a unique structure.
  • ADCs obtained by thiol-Michael addition methods will undergo reverse Michael additions in systemic circulation, resulting in toxic reactions.
  • the patent WO2016142049 discloses amatoxins as bioactive molecules, and structure comprising bioactive molecules having the structure of methylsulfonyl-substituted benzobisoxadiazole and linkers, but details of coupling with antibodies are not specifically described.
  • the invention discloses a novel bioactive molecule conjugate, which is obtained by improving the coupling way of the drug and the targeting moiety in an ADC or SMDC.
  • the conjugate has high stability, extremely high coupling efficiency (90%) and high DAR (5-8).
  • the disclosure is based on the above findings.
  • the ADC of the invention e.g. BT001021 (example 32)
  • the exposure of the bioactive small molecular toxin in tumor is significantly higher than that in plasma
  • Immu-132 has significantly higher plasma exposure than tumor exposure under the same administration route. Therefore, the ADC of the invention has a better therapeutic window than Immu-132.
  • We were also surprised that the ADC of the invention has a better efficacy than Immu-132 in animal models of gastric cancer, breast cancer and non-small cell lung cancer.
  • a first aspect of the disclosure provides a compound as shown in formula (I) or a pharmaceutically acceptable salt thereof,
  • T is a fragment of a bioactive molecule, preferably a fragment of a molecule with antitumor bioactivity
  • L 1 is selected from an amino acid, a peptide composed of 2-10 amino acids, an oligosaccharide, -(CH 2 )t 1 -, -(CH 2 CH 2 O) t1 -(CH 2 ) t2 -,
  • L 1 is selected from Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, a peptide composed of 2-5 amino acids,
  • each of R, R′, R 1 and R 2 is independently H(hydrogen), D (deuterium), C 1-6 alkyl, C 2-10 alkenyl, C 2-10 alkynyl or C 3-6 cycloalkyl
  • Z 1 is Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, Val-Cit, Cit-Val, Cit-Ala, Val-Ala, Lys-Val, Val-Lys(Ac), Phe-Lys, Phe-Lys(Ac), D-Val-Leu-Lys, Gly-Gly-Arg or Ala-Ala-Asn
  • x 1 is 0, 1, 2 or 3
  • x 3 is 0, 1, 2, 3 or 4.
  • L 1 is selected from Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, Cit-Val, Val-Ala, Lys-Val, Val-Lys(Ac), Phe-Lys, Phe-Lys(Ac), D-Val-Leu-Lys, Gly-Gly-Arg, Ala-Ala-Asn,
  • each of R, R′ and R 1 is independently H (hydrogen), D (deuterium), C 1-6 alkyl, C 2-10 alkenyl, C 2-10 alkynyl or C 3-6 cycloalkyl
  • Z 1 is Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, Val-Cit, Cit-Val, Cit-Ala, Val-Ala, Lys-Val, Val-Lys(Ac), Phe-Lys, Phe-Lys(Ac), D-Val-Leu-Lys, Gly- Gly-Arg or Ala-Ala-Asn
  • each of x 1 and x 3 is independently 0, 1, 2 or 3.
  • L 1 is selected from Lys, Cit, Cit-Val, Val-Ala, Lys-Val,
  • each of R, R′ and R 1 is independently H(hydrogen), D (deuterium) or C 1-4 alkyl, Z 1 is Cit, Lys, Cit-Val, Cit-Ala, Val-Ala or Lys-Val, and each of x 1 and x 3 is independently 0, 1 or 2.
  • L 1 is selected from Lys, Cit, Cit-Val, Val-Ala, Lys-Val,
  • L 1 is selected from
  • L 2 is selected from Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, a peptide composed of 2-5 amino acids,
  • each of R 3 , R 4 , R 5 and R 6 is independently selected from H (hydrogen), D (deuterium), halogen, a carboxylic acid group, a sulfonic acid group, CF 3 , CN, CH 2 CN, C 1-4 alkyl, C 1-4 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl or C 3-6 cycloalkyl, each of y 1 and y 2 is independently 0, 1, 2, 3, 4, 5, 6, 7 or 8, and L 2 is bonded to L 1 at the position 1 of L 2 ;
  • n 1 0, 1, 2 or 3.
  • L 2 is selected from Val, Cit, Phe, Lys, D-Val, Leu, Gly, Ala, Asn, Val-Cit, Cit-Val, Val-Ala, Lys-Val, Val-Lys(Ac), Phe-Lys, Phe-Lys(Ac), D-Val-Leu-Lys, Gly-Gly-Arg, Ala-Ala-Asn,
  • each of R 3 , R 4 , R 5 and R 6 is independently selected from H (hydrogen), D (deuterium), halogen, a carboxylic acid group, a sulfonic acid group, CF 3 , CN, CH 2 CN, C 1-4 alkyl, C 1-4 alkoxy, C 2-6 alkenyl, C 2-6 alkynyl or C 3-6 cycloalkyl, each of y 1 and y 2 is independently 0, 1, 2, 3, 4, 5, 6, 7 or 8, and L 2 is bonded to L 1 at the position 1 of L 2 ;
  • n 1 0, 1 or 2.
  • L 2 is selected from
  • each of R 3 , R 4 , R 5 and R 6 is independently selected from H (hydrogen), D (deuterium) or C 1-4 alkyl, each of y 1 and y 2 is independently 0, 1, 2, 3, 4, 5, 6, 7 or 8, and L 2 is bonded to L 1 at the position 1 of L 2 ;
  • L 2 is selected from
  • L 2 is selected from
  • L 3 is selected from the following groups optionally substituted with one or more R 7 : amino, 3-8 membered cycloalkylene, 3-8 aliphatic heterocyclylene, 6-12 membered bridged heterocyclylene, 6-12 membered spiroheterocyclylene, 6-12 membered fused heterocyclylene, 6-10 membered arylene, 5-12 membered heteroarylene or 3-8 membered cycloalkylene-W-; wherein W is oxygen or NR 8 , R 7 is independently selected from H (hydrogen), D (deuterium), halogen, ⁇ O, CF 3 , CN, CH 2 CN, carboxyl, sulfonic acid group, C 1-4 alkyl, C 1-4 alkoxy, C 2-6 alkenyl or C 2-6 alkynyl; preferably, the 3-8 aliphatic heterocyclylene, 6-12 membered bridged heterocyclylene, 6-12 membered spirohetero
  • L 3 is selected from the following groups optionally substituted with one or more R 7 : amino, N-methylpiperidylene, pyrazolylene or triazolylene; wherein R 7 is independently selected from H (hydrogen), D (deuterium), halogen, ⁇ O, CF 3 , CN, CH 2 CN, carboxyl, sulfonic acid group, C 1-4 alkyl, C 1-4 alkoxy, C 2-6 alkenyl or C 2-6 alkynyl; m 2 is 0 or 1.
  • L 3 is selected from triazolylene; m 2 is 0 or 1.
  • L 3 is selected from
  • m 2 is 0 or 1. preferably, L 3 is bonded to L 2 at the position 1 of L 3 .
  • L 3 is selected from the following groups optionally substituted with one or more R 7 : amino,
  • L 3 is selected from
  • each R q is independently selected from C 1-6 alkyl, C 2-6 alkenyl, C 3-6 alkynyl or C 3-8 cycloalkyl; ( ⁇ 1 is 0, 1 or 2; and ⁇ 2 is 1, 2 or 3.
  • L 3 is selected from
  • L 4 is selected from
  • Z 4 is 6-10 membered aryl or 5-6 membered heteroaryl;
  • R 10 is selected from H (hydrogen) or C 1-6 alkyl;
  • Z 2 is selected from C 1-6 alkylene, C 2-10 alkenylene, C 2-10 alkynylene or C 3-8 cycloalkylene;
  • R 9 is selected from H (hydrogen) or C 1-6 alkyl;
  • Z 3 is absent or selected from C 1-6 alkylene; or R 9 and Z 3 together with the nitrogen atom attached thereto form a 4-8 membered heterocyclylene;
  • is independently 0, 1, 2, 3, 4, 5 or 6, and L 4 is bonded to E at the position 2 of L4;
  • n 3 0, 1, 2 or 3.
  • L 4 is selected from
  • Z 4 is a benzene ring
  • R 10 is selected from H (hydrogen) and C 1-6 alkyl
  • Z 2 is selected from C 1-6 alkylene, C 2-10 alkenylene, C 2-10 alkynylene or C 3-8 cycloalkylene
  • R 9 is selected from H (hydrogen) or C 1-6 alkyl
  • Z 3 is absent or selected from C 1-6 alkylene or R 9 and Z 3 together with the nitrogen atom attached thereto form a 4-8 membered heterocyclylene
  • is independently 0, 1, 2, 3, 4, 5 or 6, and L 4 is bonded to E at the position 2 of L 4 ;
  • n3 0, 1, 2 or 3.
  • L 4 is selected from
  • Z 4 is 5-6 membered heteroarylene;
  • R 10 is selected from H (hydrogen) or C 1-6 alkyl;
  • Z 2 is selected from C 1-6 alkylene, C 2-10 alkenylene, C 2-10 alkynylene or C 3-8 cycloalkylene;
  • R 9 is selected from H (hydrogen) or C 1- 6 alkyl;
  • Z 3 is absent or selected from C 1-6 alkylene; or R 9 and Z 3 together with the nitrogen atom attached thereto form a 4-8 membered heterocyclylene;
  • is independently 0, 1, 2, 3,4, 5 or 6; and
  • L 4 is bonded to E at the position 2 of L4;
  • n3 0, 1, 2 or 3.
  • L 4 is selected from
  • L 4 is selected from
  • L 4 is selected from
  • E is selected from 5-10 membered heteroarylene optionally substituted with one or more R 12 ; wherein R 12 is independently selected from H (hydrogen), D (deuterium), halogen, CN, nitro, C 1-4 alkyl or halogenated C 1-4 alkyl.
  • E is selected from the following groups optionally substituted with one or more R 12 : pyrimidylene, quinolylene or pyrrolo [2,3-d] pyrimidylene; wherein R 12 is independently selected from H (hydrogen), D (deuterium), halogen, CN, nitro, C 1- 2 alkyl or halogenated C 1-2 alkyl.
  • E is selected from pyrimidinyl optionally substituted with one or more R 12 ; wherein R 12 is independently selected from H(hydrogen) or D (deuterium).
  • G is selected from halogen, OMs, OTs, OTf, nitro, or anyone of the following groups which is optionally substituted with one or more R 13 : alkylthio, arylthio, heteroarylthio, alkyl sulfinyl, aryl sulfinyl, heteroaryl sulfinyl, alkyl sulfonyl, aryl sulfonyl or heteroaryl sulfonyl; wherein R 13 is independently selected from H (hydrogen), D (deuterium), halogen, CN, nitro, C 1-6 alkyl, halogenated C 1-6 alkyl, C 1-6 alkoxy, 6-10 membered aryl or 5-12 membered heteroaryl.
  • G is selected from F, Cl, Br, I, OMs, OTs, OTf, methylsulfonyl, ethylsulfonyl, p-toluenesulfonyl or naphthalenesulfonyl.
  • G is selected from F, Cl, Br, OMs, OTs, methylsulfonyl or p-toluenesulfonyl.
  • G is selected from Cl or methylsulfonyl.
  • G is preferably methylsulfonyl
  • E is preferably pyrimidylene
  • m 3 is 1.
  • m 4 is preferably an integer from 0 to 6
  • methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • m 5 is preferably an integer from 0 to 6
  • methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • m 6 is preferably an integer from 0 to 6
  • methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • m 7 is an integer from 1 to 5
  • methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • ms is an integer from 1 to 5
  • methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • m 9 is an integer from 1 to 5
  • R 13 is selected from hydrogen or C 1- 6 alkyl, methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • m 10 is an integer from 0 to 6, and Z 4 is selected from 5-6 membered heteroarylene; methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring.
  • Z 4 is selected from pyridylene, pyrimidylene, pyrazolylene, thiazolylene, oxazolylene or triazolylene; methylsulfonyl is a substituent on a carbon atom adjacent to a nitrogen atom in the pyrimidine ring; more preferably, m 10 is an integer from 0-6.
  • Z 4 is selected from pyridylene, pyrimidylene, pyrazolylene or triazolylene. More preferably, m 10 is an integer from 0-6.
  • Z 4 is selected from oxazolylene or thiazolylene, and methylsulfonyl is a substituent of a carbon atom adjacent to a nitrogen atom in the pyrimidine ring. More preferably, m 10 is an integer from 0-6.
  • m 10 is an integer from 0-6, and Z 4 is selected from 6-10 membered arylene; and methylsulfonyl is a substituent of a carbon atom adjacent to a nitrogen atom. More preferably, m 10 is an integer from 0-6.
  • m 10 is an integer from 0-6, and Z 4 is a benzene ring.
  • T is a fragment of a bioactive molecule.
  • the bioactive molecule is selected from a metal complex, such as a platinum metal complex (e.g., oxaliplatin) or a gold metal complex; a glycopeptide antibiotic such as bleomycin or pingyangmycin; a DNA topoisomerase inhibitor, such as a topoisomerase I inhibitor (e.g., camptothecin, hydroxycamptothecin, 9-aminocamptothecin, SN-38, irinotecan, topotecan, bellotencian or rubitecan) or a topoisomerase II inhibitor (e.g., actinomycin D, adriamycin, doxorubicin, duocarmycin, daunorubicin, mitoxantrone, podophyllotoxin or etoposide); a drug interfering with DNA synthesis, such as methotrexate, 5-fluorouracil, 5-fluorour
  • the bioactive molecule is selected from
  • R 14 is selected from acyl or sulfonyl, which is substituted with R 15 , and R 15 is selected from C 1-6 alkyl, halogenated C 1-6 alkyl, 6-10 membered aryl or 5-12 membered heteroaryl;
  • R 16 is selected from H (hydrogen), D (deuterium), C 1-6 alkyl or C 1-6 alkyl substituted with R 17 , and R 17 is selected from aryl or heteroaryl, including but not limited to phenyl and pyridyl, and m 11 is 0, 1 or 2.
  • the bioactive molecule is selected from
  • R 14 is selected from acyl or sulfonyl, which is substituted with R 15 , and R 15 is selected from C 1-6 alkyl, halogenated C 1-6 alkyl, 6-10 membered aryl or 5-12 membered heteroaryl;
  • R 16 is selected from H (hydrogen), D (deuterium), C 1-6 alkyl, C 1-6 alkyl substituted with R 17 , and R 17 is selected from aryl or heteroaryl, including but not limited to phenyl or pyridyl, and m 11 is 0, 1, or 2.
  • the bioactive molecule is selected from
  • the bioactive molecule is selected from
  • the bioactive molecule is selected from
  • the bioactive molecule is selected from
  • the bioactive molecule is selected from
  • T is selected from
  • T is selected from
  • T is selected from
  • T is selected from
  • T is selected from
  • the compound shown in formula (I) is selected from
  • the compound is selected from:
  • the disclosure provides a conjugate, comprising a bioactive molecule, a linker and a targeting moiety.
  • the targeting moiety is linked to the linker via an active group (e.g., a thiol group) to form a conjugate.
  • a target of A is selected from epidermal growth factor, Trop-2, CD37, HER2, CD70, EGFRvIII, Mesothelin, Folate receptor1, Mucin 1, CD138, CD20, CD19, CD30, SLTRK6, Nectin 4, Tissue factor, Mucin16, Endothelin receptor, STEAP1, SLC39A6, Guanylylcyclase C, PSMA, CCD79b, CD22, Sodium phosphate cotransporter 2B, GPNMB, Trophoblast glycoprotein, AGS-16, EGFR, CD33, CD66e, CD74, CD56, PD-L1, TACSTD2, DR5, E16, STEAP1, 0772P, MPF, Napi3b, Sema 5b, PSCA hlg, ETBR, MSG783, STEAP2, TrpM4, CRIPTO, CD21, CD79b, FcRH2, NCA, MDP, IL20R ⁇ , Brevican, Eph
  • A is a small molecule ligand, such as a folic acid derivative, a glutamate urea derivative, a somatostatin derivative, an arylsulfonamide derivative (e.g., a carbonic anhydrase IX inhibitor), a polyene connecting two aliphatic indoles, a cyanine dye or IR-783 or a derivative thereof.
  • a small molecule ligand such as a folic acid derivative, a glutamate urea derivative, a somatostatin derivative, an arylsulfonamide derivative (e.g., a carbonic anhydrase IX inhibitor), a polyene connecting two aliphatic indoles, a cyanine dye or IR-783 or a derivative thereof.
  • A is selected from
  • A is an antibody such as a monoclonal antibody or an antigen binding fragment thereof, wherein the monoclonal antibody or the antigen binding fragment thereof comprises Fab, Fab′, F(ab′) 2 , Fd, Fv, dAb, a complementary determinant fragment, a single chain antibody (e.g., scFv), a non-human antibody, a humanized antibody, a chimeric antibody, a completely humanized antibody, a probody, a bispecific antibody or a multispecific antibody.
  • a monoclonal antibody or an antigen binding fragment thereof comprises Fab, Fab′, F(ab′) 2 , Fd, Fv, dAb, a complementary determinant fragment, a single chain antibody (e.g., scFv), a non-human antibody, a humanized antibody, a chimeric antibody, a completely humanized antibody, a probody, a bispecific antibody or a multispecific antibody.
  • A is an anti-Her 2 monoclonal antibody, such as Trastuzumab, Pertuzumab; or an anti-Trop-2 monoclonal antibody, such as Sacituzumab.
  • A is an anti-Trop-2 monoclonal antibody, such as antibody M1, M2 or M3.
  • Antibody M1 M2 M3 Heavy chain CDR1 GYTFTNY (SEQ ID No.: 1) GYTFTNY (SEQ ID No.: 1) GYTFTNY (SEQ ID No.: 1) Heavy chain CDR2 NTDSGE (SEQ ID No.:2) NTDSGE (SEQ ID No.:2) NTDSGE (SEQ ID No.:2) Heavy chain CDR3 GGFGSSYWYFDV (SEQ ID No.:3) GGFGSSYWYFDV (SEQ ID No.:3) GGFGSSYWYFDV (SEQ ID No.:3) GGFGSSYWYFDV (SEQ ID No.:3) Light chain CDR1 KASQDVSSAVA (SEQ ID No.:4) KASQDVSSAVA (SEQ ID No.:4) KASQDVSIAVA (SEQ ID No.:8) Light chain CDR2 SASYRYT (SEQ ID No.:5) SASYRYT (SEQ ID No.:5) SASYRYT (SEQ ID No.:
  • Heavy chain and light chain sequences of the hydrophobically modified antibody M1 Amino acid sequence of heavy chain variable region of M1: (121 aa)
  • Heavy chain and light chain sequences of the hydrophobically modified antibody M2 Amino acid sequence of heavy chain variable region of M2: (121 aa)
  • Heavy chain and light chain sequences of the hydrophobically modified antibody M3 Amino acid sequence of heavy chain variable region of M3: (121 aa)
  • Terminal Lys of heavy chains is easily deleted, but such deletion does not affect bioactivity. See Dick, L.W. et al., Biotechnol. Bioeng., 100: 1132-1143.
  • the above monoclonal antibodies M1, M2, M3 and sequences or fragments thereof with deleted Lys at terminal of heavy chains all belong to the M1, M2, M3 monoclonal antibodies of this invention.
  • A is selected from a RGD peptide that recognizes cell surface integrin receptor; a growth factor that recognizes cell surface growth factor receptor, such as EGF, PDGF or VEGF; or a peptide capable of recognizing functional cell surface plasminogen activator, bombesin, bradykinin, somatostatin or prostate-specific membrane antigen receptor.
  • A is selected from CD40 ligand, CD30 ligand, OX40 ligand, PD-1 ligand, ErbB ligand, Her2 ligand, TACSTD2 ligand, or DR5 ligand.
  • the conjugate is selected from: or wherein, y is an integer or a decimal from 1 to 10, and mAb is an anti-Trop-2 monoclonal antibody or an anti-Her 2 monoclonal antibody; preferably, the anti-Trop-2 monoclonal antibody is selected from antibodies of Sacituzumab, M1, M2 or M3, and the anti-Her 2 monoclonal antibody is Trastuzumab or Pertuzumab; preferably, y is an integer or a decimal from 5 to 8 (e.g., 5, 6, 7 or 8).
  • the conjugate is selected from: wherein, y is an integer or a decimal from 1 to 10, and mAb is an anti-Trop-2 monoclonal antibody or an anti-Her 2 monoclonal antibody; preferably, the anti-Trop-2 monoclonal antibody is selected from Sacituzumab, and the anti-Her 2 monoclonal antibody is selected from Trastuzumab or Pertuzumab; preferably, y is an integer or a decimal from 5 to 8 (e.g., 5, 6, 7 or 8).
  • the conjugate is: or wherein, A1 is Sacituzumab, and y is an integer or a decimal from 1 to 10; preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A1 is Sacituzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A1 is Sacituzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: wherein, A1 is a fragment of Sacituzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A2 is Trastuzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A2 is Trastuzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: wherein, A2 is Trastuzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A3 is Pertuzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A3 is Pertuzumab, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A4 is antibody M1, and y is an integer or a decimal from 1 to 10; and preferably, y is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: wherein, A4 is antibody M1, and y is an integer or a decimal from 1 to 10; and preferably, ⁇ is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein, A5 is antibody M2, and ⁇ is an integer or a decimal from 1 to 10; and preferably, ⁇ is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: wherein, A5 is antibody M2, and ⁇ is an integer or a decimal from 1 to 10; and preferably, ⁇ is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: or wherein A6 is antibody M3, and ⁇ is an integer or a decimal from 1 to 10; and preferably, ⁇ is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the conjugate is: wherein A6 is antibody M3, and ⁇ is an integer or a decimal from 1 to 10; and preferably, ⁇ is an integer or a decimal from 5 to 8, such as an integer or a decimal from 6-7, 6-7.5, 6-8, 6.5-7, 6.5-7.5, 6.5-8, 7-8 or 7.5-8.
  • the disclosure provides a method for preparing the conjugate of the second aspect, comprising a step of coupling the linker of the compound of formula (I) with an active group of the targeting moiety.
  • the method comprises a step of opening a disulfide bond of the targeting moiety by a reductant (e.g., TCEP) to obtain a sulfydryl group.
  • a reductant e.g., TCEP
  • the method comprises a step of forming a C-S bond between the linker of the compound of formula (I) and the sulfydryl group of the targeting moiety.
  • the targeting moiety is an anti-Her 2 monoclonal antibody (e.g., Trastuzumab, Pertuzumab) or an anti-Trop-2 monoclonal antibody (e.g., Sacituzumab, M1, M2 or M3), or an active fragment or mutant thereof.
  • an anti-Her 2 monoclonal antibody e.g., Trastuzumab, Pertuzumab
  • an anti-Trop-2 monoclonal antibody e.g., Sacituzumab, M1, M2 or M3
  • the molar ratio of the targeting moiety to the compound of formula (I) is 1:(1-20); preferably, the coupling is carried out in water and/or an organic solvent; preferably, the organic solvent is selected from N,N-dimethylformamide, dimethylsulfoxide, N-methylpyrrolidone, nitriles (e.g., acetonitrile), alcohols (e.g., methanol, ethanol) or any combination thereof.
  • the organic solvent is selected from N,N-dimethylformamide, dimethylsulfoxide, N-methylpyrrolidone, nitriles (e.g., acetonitrile), alcohols (e.g., methanol, ethanol) or any combination thereof.
  • the method further comprises a step of purifying the coupling product; preferably, the coupling product is purified by chromatography (e.g., one or more of ion exchange chromatography, hydrophobic chromatography, reverse phase chromatography or affinity chromatography).
  • chromatography e.g., one or more of ion exchange chromatography, hydrophobic chromatography, reverse phase chromatography or affinity chromatography.
  • the disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compound of the first aspect of the disclosure or a pharmaceutically acceptable salt thereof, or the conjugate of the second aspect, and one or more pharmaceutical excipients.
  • the disclosure provides use of the compound of the first aspect or a pharmaceutically acceptable salt thereof or the conjugate of the second aspect in the manufacturer of a medicament for the treatment of a disease associated with an abnormal cell activity (e.g., cancer).
  • a disease associated with an abnormal cell activity e.g., cancer
  • the cancer is a solid tumor or a non-solid tumor, such as esophageal cancer (e.g., esophageal adenocarcinoma, esophageal squamous cell carcinoma), a brain tumor, lung cancer (e.g., small cell lung cancer, non-small cell lung cancer), squamous cell carcinoma, bladder cancer, stomach cancer, ovarian cancer, peritoneal cancer, pancreatic cancer, breast cancer, head and neck cancer, cervical cancer, endometrial cancer, colorectal cancer, liver cancer, kidney cancer, non Hodgkin’s lymphoma, central nervous system tumors (e.g., neuroglioma, glioblastoma multiforme, glioma or sarcoma), prostate cancer and thyroid cancer.
  • esophageal cancer e.g., esophageal adenocarcinoma, esophageal squamous cell carcinoma
  • a brain tumor e.g
  • the disclosure provides use of the compound of the first aspect or a pharmaceutically acceptable salt thereof or the conjugate or pharmaceutical composition of the second aspect in treating a disease associated with an abnormal cell activity (e.g., cancer).
  • a disease associated with an abnormal cell activity e.g., cancer
  • the disclosure provides a method of treating a disease associated with an abnormal cell activity (e.g., cancer), comprising a step of administering an effective amount of the compound of the first aspect or a pharmaceutically acceptable salt thereof or the conjugate or pharmaceutical composition of the second aspect to an individual in need thereof.
  • a disease associated with an abnormal cell activity e.g., cancer
  • the pharmaceutical excipients refer to excipients and additives used in drug manufacturing and formulating, and are substances that have been reasonably evaluated in terms of safety and are contained in pharmaceutical preparations in addition to active ingredients.
  • pharmaceutical excipients also have important functions such as solubilization, sustained release, and are important ingredients that may affect the quality, safety and efficacy of drugs.
  • compositions can be divided by sources into natural substances, semi-synthetic substances and full-synthetic substances; divided by effects and uses into solvents, propellants, solubilizers, cosolvents, emulsifiers, colorants, binders, disintegrants, filling agents, lubricants, wetting agents, osmotic pressure regulators, stabilizers, glidants, flavoring agents, preservatives, suspending agents, coating materials, aromatics, anti-adhesion agents, antioxidants, chelating agents, penetration enhancers, pH regulators, buffers, plasticizers, surfactants, foaming agents, defoamers, thickeners, inclusion agents, humectants, absorbents, diluents, flocculants and deflocculants, filter aids and release retardants; and divided by administration routes into oral administration, injection, mucosal, transdermal or local administration, nasal or oral inhalation and ophthalmic administration.
  • the same pharmaceutical excipient can
  • the pharmaceutical composition can be formulated into various suitable dosage forms depending on administration routes, such as tablets, capsules, granules, oral solutions, oral suspensions, oral emulsions, powders, tinctures, syrups, injections, suppositories, ointments, creams, pastes, ophthalmic preparations, pills, subdermals, aerosols, powders and sprays.
  • the pharmaceutical composition or suitable dosage forms may contain 0.01 mg to 1000 mg of the compound of the disclosure or a pharmaceutically acceptable salt or conjugate thereof, suitably 0.1 mg to 800 mg, preferably 0.5 to 500 mg, preferably 0.5 to 350 mg, and particularly preferably 1 to 1-250 mg.
  • the pharmaceutical composition can be administered in the form of injections, including liquids for injection, sterilized powders for injection, and concentrated solutions for injection.
  • Acceptable carriers and solvents include water, Ringer’s solution and isotonic sodium chloride solution.
  • sterilized non-volatile oil can also be used as a solvent or suspending medium, such as monoglyceride or diglyceride.
  • the term “individual” include a human individual or a non-human animal.
  • exemplary human individual includes a human individual with a disease (e.g., a disease described herein) (referred to as a patient) or a normal individual.
  • non-human animal in the disclosure includes all vertebrates, such as a non-mammal (e.g., a bird, an amphibian and a reptile) and a mammal, such as a non-human primate, a domestic animal, and/or a domesticated animal (e.g., a sheep, a dog, a cat, a cow and a pig).
  • the term “effective amount” refers to the amount of the compound that, after being administered, relieves one or more symptoms of the treated disease to some extent.
  • the term “conjugate” refers to a substance obtained by linking a bioactive molecule with a targeting moiety.
  • the bioactive molecule is linked to the targeting moiety via a linker.
  • the linker can be cleaved in a specific environment (e.g., an intracellular low pH environment) or under a specific action (e.g., the action of lysosomal protease), thereby dissociating the bioactive molecule from the target moiety.
  • the linker comprises cleavable or non-cleavable units, such as a peptide or disulfide bond.
  • the bioactive molecule is linked directly to the targeting moiety via a covalent bond that can be cleaved under a specific environment or action, thereby dissociating the bioactive molecule from the targeting moiety.
  • bioactive substance and “bioactive molecule” refer to a substance that inhibits or prevents cell functions and/or cause cell death or destruction.
  • bioactive substance or bioactive molecule in the conjugate is a molecule with anti-tumor bioactivity.
  • a radioisotope such as At 211 , I 131 , I 125 , Y 90 , Re 186 , Re 188 , Sm 153 , Bi 212 , P 32 , Pb 212 or a radioisotope of Lu; a metal complex such as a platinum metal complex, a gold metal complex or oxaliplatin; a glycopeptide antibiotic such as bleomycin or pingyangmycin; a DNA topoisomerase inhibitor such as a topoisomerase I inhibitor, e.g., camptothecin, hydroxycamptothecin, 9- aminocamptothecin, SN-38, irinotecan, topotecan, bellotencian or rubitecan, or a topoisomerase II inhibitor, e.g., actinomycin D, adriamycin, doxorubicin, duocarmycin, daunorubicin, mitoxantrone, podophyl
  • small molecule refers to a small molecule drug with bioactivity.
  • linker refers to a fragment linking a bioactive molecule with a targeting moiety.
  • targeting moiety refers to a moiety of the conjugate that can specifically bind to a target (or a portion of the target) on the cell surface.
  • the conjugate can be delivered to a specific cell population by interaction between the targeting moiety and the target.
  • the conjugate can be referred to as a “drug-antibody conjugate” when the targeting moiety of the conjugate is an antibody.
  • the “drug-antibody conjugate” and “immune conjugate” are interchangeable.
  • the term “antibody” is interpreted in its broadest sense, including a complete monoclonal antibody, polyclonal antibody, and a multispecific antibody (e.g., a bispecific antibody) formed from at least two complete antibodies, provided that the antibody has required bioactivity.
  • a multispecific antibody e.g., a bispecific antibody
  • the terms “antibody” and “immunoglobulin” are interchangeable.
  • the term “monoclonal antibody” refers to an antibody from a group of substantially uniform antibodies, i.e., antibodies that make up the group are identical except for a small number of possible natural mutations.
  • a monoclonal antibody has high specificity for one determinant (epitope) of an antigen, while a comparative polyclonal antibody contains different antibodies for different determinants (epitopes).
  • the monoclonal antibody has the advantage of being free from contamination by other antibodies during synthesis.
  • the modifier “monoclonal” here indicates that the antibody is characterized by coming from a substantially uniform antibody group and should not be construed as being prepared by a special method.
  • the monoclonal antibody also specifically includes a chimeric antibody, i.e., a portion of a heavy chain and/or a light chain is the same as or homologous to a type, a class, or a subclass of antibodies, while the rest is the same as or homologous to another type, another class, or another subclass of antibodies, provided the antibody has required bioactivity (see, e.g., US 4,816,567; and Morrison et al., 1984, PNAS, 81: 6851-6855).
  • the chimeric antibody available in the disclosure includes a primatized antibody containing a variable region antigen binding sequence from a non-human primate (e.g., an ancient monkey or an orangutan) and a human constant region sequence.
  • antibody fragment refers to a portion of the antibody, preferably an antigen binding region or a variable region.
  • antibody fragment includes Fab, Fab′, F(ab′) 2 , Fd, Fv, dAb and complementary determinant fragment, diabody, linear antibody and single chain antibody molecule.
  • bispecific antibody also known as “bifunctional antibody conjugate” refers to a conjugate formed by a first antibody (fragment) and a second antibody (fragment) through a coupling arm.
  • the conjugate retains the activity of each antibody and thus has bifunctional and bispecific properties.
  • multispecific antibody includes, for example, a trispecific antibody which is an antibody having three different antigen binding specificities, and a tetraspecific antibody which is an antibody having four different antigen binding specificities.
  • complete antibody refers to an antibody containing an antigen binding variable region, a light chain constant region (CL) and heavy chain constant regions (CH1, CH2 and CH3).
  • the constant regions can be natural sequences (e.g., human natural constant region sequences) or amino acid sequence variants thereof.
  • the complete antibody is preferably a complete antibody having one or more effector functions.
  • antibody is a modified antibody comprising an antibody or antibody fragment that can specifically bind to a target thereof and can be coupled with a masked group, and the masked group here refers that the cleavage constant for the binding capacity of the antibody or antibody fragment to the target is at least 100 times or 1000 times or 10000 times greater than that for the binding capacity of an antibody or antibody fragment not coupled with a masked group to a target thereof.
  • a “humanized” form of a non-human (e.g., mouse) antibody refers to a chimeric antibody that contains minimal non-human immunoglobulin sequences.
  • Most of the humanized antibodies are those in which residues in hypervariable regions of human recipient immunoglobulins are substituted with residues in non-human (e.g., mice, rats, rabbits or non-human primates) hypervariable regions (donor antibodies) with required specificity, affinity and functions.
  • residues in frame regions (FRs) of human immunoglobulins are also substituted with non-human residues.
  • the humanized antibody can also contain residues not present in recipient antibodies or donor antibodies. Such modifications are made to further optimize antibody performance.
  • a humanized antibody generally contains at least one variable region, typically two variable regions, in which all or almost all hypervanable loops correspond to non-human immunoglobulins, while all or almost all FRs are those of human immunoglobulin sequences.
  • a humanized antibody can also contain at least a portion of an immunoglobulin constant region (Fc, usually human immunoglobulin Fc).
  • Fc immunoglobulin constant region
  • Complete antibody can be classified into different “classes” according to amino acid sequences of heavy chain constant regions.
  • the main five classes are IgA, IgD, IgE, IgG and IgM, and several of which can also be further classified into different “subclasses” (isotypes), such as IgG1, IgG2, IgG3, IgG4, IgA1 and IgA2.
  • Different classes of heavy chain constant regions of antibodies are called ⁇ , ⁇ , ⁇ , ⁇ and ⁇ respectively.
  • Different classes of subunit structures and 3D configurations of immunoglobulins are well known in the art.
  • an antibody peptide chain can contain one or more D-amino acids.
  • Peptides containing D-amino acids may be more stable and less degradable in oral cavity, intestinal tract or plasma than peptides containing only L-amino acids.
  • Monoclonal antibody used in the disclosure can be produced by multiple methods.
  • the monoclonal antibody used in the disclosure can be obtained by hybridoma methods using multiple species (including cells of mice, hamsters, rats and humans) (see, for example, Kohler et al., 1975, Nature, 256: 495), or by recombinant DNA techniques (see, for example, US 4,816,567), or isolated from phage antibody libraries (see, for example, Clackson et al., 1991, Nature, 352: 624-628; and Marks et al., 1991, Journal of Molecular Biology, 222: 581-597).
  • Monoclonal antibody that can be used in the disclosure includes, but is not limited to anti-Her 2 monoclonal antibody such as Trastuzumab and Pertuzumab, or anti-Trop-2 monoclonal antibody such as Sacituzumab (i.e., Isactuzumab or hRS7 antibody), M1, M2 or M3.
  • anti-Her 2 monoclonal antibody such as Trastuzumab and Pertuzumab
  • anti-Trop-2 monoclonal antibody such as Sacituzumab (i.e., Isactuzumab or hRS7 antibody), M1, M2 or M3.
  • a target of A is selected from epidermal growth factor, Trop-2, CD37, HER2, CD70, EGFRvIII, Mesothelin, Folate receptor1, Mucin 1, CD138, CD20, CD19, CD30, SLTRK6, Nectin 4, Tissue factor, Mucin16, Endothelin receptor, STEAP1, SLC39A6, Guanylylcyclase C, PSMA, CCD79b, CD22, Sodium phosphate cotransporter 2B, GPNMB, Trophoblast glycoprotein, AGS-16, EGFR, CD33, CD66e, CD74, CD56, PD-L1, TACSTD2, DR5, E16, STEAP1, 0772P, MPF, Napi3b, Sema 5b, PSCA hlg, ETBR, MSG783, STEAP2, TrpM4, CRIPTO, CD21, CD79b, FcRH2, NCA, MDP, IL20Ra, Brevican, Eph
  • the target of the targeting moiety A is slected from a RGD peptide that recognizes cell surface integrin receptor; a growth factor that recognizes cell surface growth factor receptor, such as EGF, PDGF or VEGF; and a peptide capable of recognizing functional cell surface plasminogen activator, bombesin, bradykinin, somatostatin or prostate-specific membrane antigen receptor.
  • the target of the targeting moiety A is selected from a CD40 ligand, a CD30 ligand, an OX40 ligand, a PD-1 ligand, an ErbB ligand, a Her2 ligand, a TACSTD2 ligand and a DR5 ligand.
  • the targeting moiety A is an anti-Her 2 monoclonal antibody, such as Trastuzumab or Pertuzumab; or an anti-Trop-2 monoclonal antibody, such as Sacituzumab, M1, M2 or M3.
  • the targeting moiety is Trastuzumab or Pertuzumab.
  • Tratuzumab is an anti-Her 2 monoclonal antibody, an amino acid sequence thereof is known to a person skilled in the art, for a schematic sequence thereof, refer to, for example, CN103319599.
  • terminal Lys of heavy chains of the targeting moiety is easily deleted, but such deletion does not affect bioactivity. See Dick, L.W. et al., Biotechnol. Bioeng., 100: 1132-1143.
  • the targeting moiety is an anti-Trop-2 monoclonal antibody, such as Sacituzumab, M1, M2 or M3 deleted terminal Lys of heavy chains, for example, the targeting moiety is an anti-Her 2 monoclonal antibody, such as Trastuzumab or Pertuzumab deleted terminal Lys of heavy chains.
  • Exemplary heavy and light chain sequences of Trastuzumab refer to SEQ ID No.: 17 and SEQ ID No.:18.
  • heavy and light chain sequences of Trastuzumab referred to or involved are described using the sequences shown in SEQ ID No.: 17 and SEQ ID No.: 18, respectively.
  • Exemplary heavy and light chain sequences of Pertuzumab refer to SEQ ID No.: 16 and SEQ ID No.: 15 of US7560111.
  • the anti-Trop-2 antibody of the targeting moiety is RS7 (i.e., Sacituzumab of the disclosure) described in U.S. pat. 7,517,964; and hRS7 (i.e., Sacituzumab of the disclosure) described in US2012/0237518.
  • the anti-Trop-2 antibody available in the disclosure can also be obtained by screening through carrier design, construction and construction of an antibody library displaying antibodies as disclosed in CN103476941A, or can be obtained by screening a G-MAB® library of Sorrento Therapeutics, Inc.
  • heavy chain sequence and light chain amino acid sequence of the monoclonal antibody Sacituzumab refer to, for example, SEQ ID No.: 19 and SEQ ID No.:20, respectively.
  • Terminal K (or lys) of heavy chains is easily deleted, but such deletion does not affect bioactivity. See Dick, L.W. et al., Biotechnol. Bioeng., 100: 1132-1143.
  • ErbB2 and Her2/neu are interchangeable, both of which represent human Her2 proteins of natural sequences (Genebank CAS No.: X03363, see, for example, Semba et al., 1985, PNAS, 82: 6497-6501; and Yamamoto et al., 1986, Nature, 319: 230-234) and functional derivatives thereof, such as amino acid sequence variants.
  • ErbB2 represents a gene encoding human Her2 and neu represents a gene encoding rat p185neu.
  • the compound or conjugate of the disclosure can inhibit or kill cells that express ErbB2 receptors, such as breast cancer cells, ovarian cancer cells, gastric cancer cells, endometrial cancer cells, salivary gland cancer cells, lung cancer cells, kidney cancer cells, colon cancer cells, thyroid cancer cells, pancreatic cancer cells, bladder cancer cells or liver cancer cells.
  • cells that express ErbB2 receptors such as breast cancer cells, ovarian cancer cells, gastric cancer cells, endometrial cancer cells, salivary gland cancer cells, lung cancer cells, kidney cancer cells, colon cancer cells, thyroid cancer cells, pancreatic cancer cells, bladder cancer cells or liver cancer cells.
  • Trop-2 or TROP2 refers to human trophoblast cell-surface antigen 2, also known as TACSTD2, M1S1, GA733-1, EGP-1, which is a cell surface receptor expressed in many human tumors (e.g., breast cancer, colorectal cancer, lung cancer, pancreatic cancer, ovarian cancer, prostate cancer and cervical cancer).
  • the compound or conjugates of the disclosure can inhibit or kill cells that express TROP2 receptors, such as breast cancer cells, colorectal cancer cells, lung cancer cells, pancreatic cancer cells, ovarian cancer cells, prostate cancer cells or cervical cancer cells.
  • the conjugate of the invention indicates a specific linking mode of a sulfydryl group and a linker in the antibody when the targeting moiety is an antibody.
  • C 1-6 alkyl refers to linear or branched alkyl containing 1-6 carbon atoms, including, for example, “C 1-4 alkyl” and “C 1-3 alkyl”. Specific examples include, but are not limited to methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, iso-pentyl, 2-methyl butyl, neopentyl, 1-ethyl propyl, n-hexyl, isohexyl, 3-methyl pentyl, 2-methyl pentyl, 1-methyl pentyl, 3,3-dimethyl butyl, 2,2-dimethyl butyl, 1,1-dimethyl butyl, 1,2-dimethyl butyl, 1,3-dimethyl butyl, 2,3-dimethyl butyl, 2-ethyl buty
  • C 2-6 alkenyl refers to linear, branched or cyclic alkenyl containing at least a double bond and 2-6 carbon atoms, including, for example, “C 2-4 alkenyl”. Examples thereof include, but are not limited to vinyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 1,3-butadienyl, 1-pentenyl, 2-pentenyl, 3-pentenyl, 1,3-pentadienyl, 1,4-pentadienyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 1,4-hexadienyl, cyclopentenyl, 1,3-cyclopentadienyl, cyclohexenyl and 1,4-cyclohexadienyl.
  • C 2-6 alkynyl refers to linear or branched alkynyl containing at least a triple bond and 2-6 carbon atoms, including, for example, “C 2-4 alkynyl”. Examples thereof include, but are not limited to ethynyl, propynyl, 2-butynyl, 2-pentynyl, 3-pentynyl, 4-methyl-2-pentynyl, 2-hexynyl, 3-hexynyl and 5-methyl-2-hexynyl.
  • halogen includes fluorine, chlorine, bromine and iodine.
  • 3-8 membered cycloalkyl or “C 3-8 cycloalkyl” refers to saturated cyclic alkyl containing 3-8 carbon atoms, including, for example, “3-6 membered cycloalkyl”, “4-6 membered cycloalkyl”, “5-7 membered cycloalkyl” or “5-6 membered cycloalkyl”. Specific examples include, but are not limited to cyclopropanyl, cyclobutylalkyl, cyclopentanyl, cyclohexyl, cycloheptyl and cyclooctadecyl.
  • C 1-6 alkoxy refers to a group having a structure of C 1-6 alkyl-O-, wherein C 1-6 alkyl is as defined previously. Specific examples include, but are not limited to methoxy, ethoxy, propoxy, isopropoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, pentoxy and hexyloxy.
  • 3-8 membered aliphatic heterocyclyl refers to a cyclic group containing 3-8 ring-forming atoms (at least one of which is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom).
  • a heteroatom such as a nitrogen atom, an oxygen atom or a sulfur atom.
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the “3-8 membered aliphatic heterocyclyl” includes, for example, “3-8 membered nitrogen-containing aliphatic heterocyclyl,” “3-8 membered oxygen-containing aliphatic heterocyclyl,” “3-6 membered aliphatic heterocyclyl,” “3-6 membered oxygen-containing aliphatic heterocyclyl,” “4-7 membered aliphatic heterocyclyl,” “4-6 membered aliphatic heterocyclyl,” “5-7 membered aliphatic heterocyclyl,” “5-6 membered aliphatic heterocyclyl,” “5-6 membered nitrogen-containing aliphatic heterocyclyl,” including but not limited to oxiranyl, oxocyclobutyl, pyrrolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, tetrahydropyranyl and homopiperazinyl.
  • 6-12 membered spirocyclyl refers to a cyclic structure containing 6-12 ring-forming carbon atoms and formed by two or more cyclic structures sharing one carbon atom.
  • the carbon atoms in the cyclic structure can be substituted with oxygen.
  • the “6-12 membered spirocyclyl” includes, for example, “6-11 membered spirocyclyl”, “6-10 membered spirocyclyl”, “7-10 membered spirocyclyl”, “7-9 membered spirocyclyl”, “7-8 membered spirocyclyl”, “9-10 membered spirocyclyl” and “3-10 membered spirocyclyl”.
  • 6-12 membered bridged cyclyl refers to a cyclic structure containing 6-12 ring-forming carbon atoms and formed by two or more cyclic structures sharing two nonadjacent carbon atoms, Optionally, the carbon atoms in the cyclic structure can be substituted with oxygen.
  • the “6-12 membered bridged cyclyl” includes, for example, “6-11 membered bridged cyclyl”, “5-10 membered bridged cyclyl”, “7-10 membered bridged cyclyl”, “7-9 membered bridged cyclyl”, “7-8 membered bridged cyclyl”, “9-10 membered bridged cyclyl” and “3-10 membered bridged cyclyl”. Specific examples include, but are not limited to
  • 6-12 membered fused cyclyl refers to a cyclic structure containing 6-12 ring-forming carbon atoms and formed by two or more cyclic structures sharing two adjacent atoms, including “6-11 membered fused cyclyl”, “6-10 membered fused cyclyl”, “6-8 membered fused cyclyl”, “10-12 membered fused cyclyl”, “7-10 membered fused cyclyl”.
  • Examples thereof include, but are not limited to
  • 6-12 membered spiroheterocyclyl refers to a cyclic structure containing 6-12 ring-forming atoms (at least one of which is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom) and formed by two or more cyclic structures sharing one ring-forming atom.
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the ring-forming atoms in the cyclic structure can be substituted with oxygen.
  • the “6-12 membered spiroheterocyclyl” includes, for example, “6-11 membered spiroheterocyclyl”, “5-10 membered spiroheterocyclyl”, “7-11 membered spiroheterocyclyl”, “7-10 membered spiroheterocyclyl”, “7-9 membered spiroheterocyclyl”, “7-8 membered spiroheterocyclyl”, “9-10 membered spiroheterocyclyl” and “3-10 membered spiroheterocyclyl”. Specific examples include, but are not limited to
  • 6-12 membered bridged heterocyclyl refers to a cyclic structure containing 6-12 ring-forming atoms (at least one of which is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom) and formed by two or more cyclic structures sharing two nonadjacent ring-forming atoms.
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the “6-12 membered bridged heterocyclyl” includes, for example, “6-11 membered bridged heterocyclyl”, “6-9 membered bridged heterocyclyl”, “6-10 membered bridged heterocyclyl”, “7-10 membered bridged heterocyclyl”, “7-9 membered bridged heterocyclyl”, “7-8 membered bridged heterocyclyl”, “8 membered bridged heterocyclyl”, “9-10 membered bridged heterocyclyl” and “3-10 membered bridged heterocyclyl”. Specific examples include, but are not limited to
  • 6-12 membered fused heterocyclyl refers to a cyclic structure containing 6-12 ring-forming atoms (at least one of which is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom) and formed by two or more cyclic structures sharing two adjacent atoms.
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the “6-12 membered fused heterocyclyl” includes, for example, “6-11 membered fused heterocyclyl,” “5-10 membered fused heterocyclyl”, “7-10 membered fused heterocyclyl”, “3-10 membered fused heterocyclyl”, “3-10 membered nitrogen-containing fused heterocyclyl”, “9-10 membered fused heterocyclyl”, “9-10 membered nitrogen-containing fused heterocyclyl” and “6-12 membered oxygen-containing fused heterocyclyl”.
  • Specific examples include, but are not limited to tetrahydroimidazo [4,5-c]pyridyl, 3,4-dihydroquinazolinyl, 1,2-dihydroquinoxalinyl, benzo[d][1,3]dioxolyl, 1,3-dihydroisobenzofuranyl, 4H-1,3-benzoxazinyl, 4,6-dihydro-1H-furo[3,4- ⁇ d]imidazolyl, 3a,4,6,6a-tetrahydro-1H-furo[3,4-d]imidazolyl, 4,6-dihydro-1H-thieno[3,4-d]imidazolyl, 4,6-dihydro-1H-pyrrolo[3,4-d]imidazolyl, benzoimidazolidinyl, octahydro-benzo[d]imidazolyl, decahydroquinolyl, hexahydrothienoi
  • aryl refers to a monocyclic or polycyclic hydrocarbonyl having aromaticity, such as 6-20 membered aryl, 6-10 membered aryl and 5-8 membered aryl. Specific examples include but are not limited to phenyl, naphthyl, anthracenyl and phenanthryl.
  • the “6-20 membered aryl” refers to aryl containing 6-20 ring-forming atoms.
  • heteroaryl refers to a cyclic group having aromaticity, wherein at least one ring-forming atom is a heteroatom, such as a nitrogen atom, an oxygen atom or a sulfur atom.
  • ring-forming atoms e.g., carbon atoms, nitrogen atoms or sulfur atoms
  • the ring-forming atoms in the cyclic structure can be substituted with oxygen.
  • Specific examples include, but are not limited to 5-10 membered heteroaryl, 5-10 membered nitrogen-containing heteroaryl, 6-10 membered oxygen-containing heteroaryl, 6-8 membered nitrogen-containing heteroaryl and 5-8 membered oxygen-containing heteroaryl, such as furyl, thienyl, pyrrolyl, thiazolyl, isothiazolyl, thiadiazolyl, oxazolyl, isoxazolyl, oxadiazolyl, imidazolyl, pyrazolyl, 1,2,3-triazolyl, 1,2,4-triazolyl, 1,2,3-oxadiazolyl, 1,2,4-oxadiazolyl, 1,2,5-oxadiazolyl, 1,3,4-oxadiazolyl, pyridyl, 2-pyridone, 4-pyridone, pyrimidinyl, 1,4-dioxacyclohexadienyl, 2H-1,2-oxazinyl
  • the disclosure obtains a kind of novel bioactive molecule conjugate by improving the coupling way of drugs and targeting moieties in ADCs or SMDCs.
  • the bioactive molecule conjugate is obtained through nucleophilic substitutions of the heteroaryl ring on the ADC linker by the free sulfhydryl in the antibody.
  • the conjugate obtained by the coupling can achieve at least one of the following technical effects:
  • the coupling method described in the disclosure has broad application scope and can be widely used in coupling bioactive molecules with antibodies or targeted small molecule ligands.
  • FIG. 1 shows a TIC (total ion chromatogram) of BT001002.
  • FIG. 2 shows a deconvolution diagram of a coupled light chain of BT001002.
  • FIG. 3 shows a deconvolution diagram of a coupled heavy chain of BT001002.
  • FIG. 4 shows a TIC (total ion chromatogram) of BT001004.
  • FIG. 5 shows a deconvolution diagram of a coupled light chain of BT001004.
  • FIG. 6 shows a deconvolution diagram of a coupled heavy chain of BT001004.
  • FIG. 7 shows a SEC chromatogram of BT001002.
  • FIG. 8 shows a SEC chromatogram of molecular weight Marker of BT001002.
  • FIG. 9 shows a SEC chromatogram of BT001004.
  • FIG. 10 shows a deconvolution diagram of a coupled light chain of BT001012.
  • FIG. 11 shows a deconvolution diagram of a coupled heavy chain of BT001012.
  • FIG. 12 shows a deconvolution diagram of a coupled light chain of BT001013.
  • FIG. 13 shows a deconvolution diagram of a coupled heavy chain of BT001013.
  • FIG. 14 shows a deconvolution diagram of a coupled light chain of BT001018.
  • FIG. 15 shows a deconvolution diagram of a coupled heavy chain of BT001018.
  • FIG. 16 shows a deconvolution diagram of a coupled light chain of BT001021.
  • FIG. 17 shows a deconvolution diagram of a coupled heavy chain of BT001021.
  • FIG. 18 shows a deconvolution diagram of a coupled light chain of BT001023.
  • FIG. 19 shows a deconvolution diagram of a coupled heavy chain of BT001023.
  • FIG. 20 shows a deconvolution diagram of a coupled light chain of BT001040.
  • FIG. 21 shows a deconvolution diagram of a coupled heavy chain of BT001040.
  • FIG. 22 shows a deconvolution diagram of a coupled light chain of BT001041.
  • FIG. 23 shows a deconvolution diagram of a coupled heavy chain of BT001041.
  • FIG. 24 shows a deconvolution diagram of a coupled light chain of BT001042.
  • FIG. 25 shows a deconvolution diagram of a coupled heavy chain of BT001042.
  • FIG. 26 shows a deconvolution diagram of a coupled light chain of BT001043.
  • FIG. 27 shows a deconvolution diagram of a coupled heavy chain of BT001043.
  • FIG. 28 shows a deconvolution diagram of a coupled light chain of BT001044.
  • FIG. 29 shows a deconvolution diagram of a coupled heavy chain of BT001044.
  • FIG. 30 shows a deconvolution diagram of a coupled light chain of BT001046.
  • FIG. 31 shows a deconvolution diagram of a coupled heavy chain of BT001046.
  • FIG. 32 shows a deconvolution diagram of a coupled light chain of BT001047.
  • FIG. 33 shows a deconvolution diagram of a coupled heavy chain of BT001047.
  • FIG. 34 shows a SEC chromatogram of BT001012.
  • FIG. 35 shows a SEC chromatogram of BT001013.
  • FIG. 36 shows a SEC chromatogram of BT001018.
  • FIG. 37 shows a SEC chromatogram of BT001021.
  • FIG. 38 shows a SEC chromatogram of BT001023.
  • FIG. 39 shows a SEC chromatogram of BT001042.
  • FIG. 40 shows a SEC chromatogram of BT001043.
  • FIG. 41 shows a SEC chromatogram of BT001044.
  • FIG. 42 shows a SEC chromatogram of BT001046.
  • FIG. 43 shows a SEC chromatogram of BT001047.
  • FIG. 44 shows changes in growth of tumor volume of each group of mice in a NCI-N87 human gastric cancer model.
  • FIG. 45 shows changes in body weight of each group of mice in a NCI-N87 human gastric cancer model.
  • FIG. 46 shows changes in growth of tumor volume of each group of mice in an HCC1806 human breast cancer model.
  • FIG. 47 A shows changes in growth of tumor volume of each group of mice in a xenograft model of HCC827 human non-small cell lung cancer.
  • FIG. 47 B shows changes in body weight of each group of mice in a xenograft model of HCC827 human non-small cell lung cancer.
  • FIG. 48 A shows changes in growth of tumor volume of each group of mice in a xenograft model of NCI-N87 human gastric cancer.
  • FIG. 48 B shows changes in body weight of each group of mice in a xenograft model of NCI-N87 human gastric cancer.
  • FIG. 49 A shows changes in growth of tumor volume of each group of mice in a tumor-bearing mice model of MDA-MB-231 human breast cancer.
  • FIG. 49 B shows changes in body weight of each group of mice in a tumor-bearing mice model of MDA-MB-231 human breast cancer.
  • OMs methylsulfonyloxy FA
  • OTs Trifluoromethylsulfonyloxy ACN
  • CCK8 reagent 2-(2-methoxy-4-nitrophenyl)-3-(4-nitrophenyl)-5-(2,4-disulfophenyl)-2H-tetrazole monosodium salt
  • TBS Tert-butyldimethylsilyl
  • FBS Fetal bovine serum MMT p-methoxytriphenylmethyl DMSO
  • Dimethyl sulfoxide PB/PBS Phosphate buffered saline
  • NMR nuclear magnetic resonance
  • MS Mass spectra
  • Step 1 synthesis of tert-butyl (4-((2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5 methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl) carbamate
  • Step 2 synthesis of (2S)-N-((3R,4S,5S)-1-((2S)-2-((1R,2R)-3-((1-((4-aminobenzyl)amino)-1-oxo-3-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-propionyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-heptanoyl-4-yl)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutanamide
  • Example 2 Synthesis of (S)-N-((3R,4S,5S)-1-((S)-2-((1R,2R)-3-((S)-1-((4-aminobenzyl)amino) 1-oxo-3-phenylprop-2-yl)amino)-1-methoxy-2-methyl-3-oxopropyl) pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxoheptyl-4-yl)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyramide (T011)
  • Step 1 synthesis of tert-buty (S)-(4-((2-((((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-phenylpropionamido) methyl)phenyl) l carbamate
  • Step 2 synthesis of tert-butyl (S)-(4-((2-amino-3-phenylpropionamido) methyl)phenyl) carbamate
  • Lithium hydroxide monohydrate (21 mg, 0.51 mmol) was dissolved in water (1 mL) and added to a tetrahydrofuran (2 mL) solution of tert-butyl (S)-(4-((2-((((9H-fluoren-9-yl)methoxy)carbonyl)amino)-3-phenylpropionamido)methyl)phenyl) carbamate (102 mg, 0.17 mmol). The resulting mixture was reacted at room temperature for 2 hours. The reaction solution was added with water (20 mL) and extracted with ethyl acetate (30 mL ⁇ 4).
  • Step 3 synthesis of (4-((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-3-((methylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy -5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)carbamate
  • Step 4 synthesis of (S)-N-((3R,4S,5S)-1-((S)-2-((1R,2R)-3-((S)-1-((4-aminobenzyl)amino)-1-oxo-3-phenylprop-2-yl)amino)-1-methoxy-2-methyl-3-oxopropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxoheptyl-4-yl)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyramide
  • Trifluoroacetic acid (0.5 mL) was added to a solution of (4-((S)-2-((2R,3R)-3-((S)-1-(3R,4S,5S)-4-((S)-3-(methylamino)-3-methylbutyrylamino)-N, 3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanamido)-3-phenylpropionamido)methyl)phenyl) carbamate (14.0 mg, 0.015 mmol) in dichloromethane (1.5 mL). The resulting mixture was reacted at room temperature for 1 h.
  • Methylsulfonyl chloride (462 mg, 12.77 mmol, purity:about 70%) was added dropwise to a solution of belotecan hydrochloride (3 g, 6.38 mmol) and triethylamine (2.58 g, 25.54 mmol) in dichloromethane (40 mL). The resulting mixture was reacted at room temperature for 2 h. Suction filtration was performed, and the filter cake was washed three times with dichloromethane (3 mL) to obtain the title compound (2.2 g).
  • Step 1 synthesis of tert-butyl 4-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl) butyrate (compound 1-2)
  • Step 2 synthesis of 4-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)butyric acid (compound 1-3)
  • Step 3 sysnthesis of (9H-fluoren-9-yl)-methyl-((S)-1-(((S)-1-((4-(((tert-butoxycarbonyl)amino)methyl)phenyl)amino)-1-oxo-5-ureidopentyl-2-yl)amino)-3-methyl-1-oxobutyl-2-yl)-carbamate (compound 1-5)
  • Step 4 synthesis of (9H-fluoren-9-yl)-methyl-((S)-1-(((S)-1-((4-(aminomethyl)phenyl))amino)-1-oxo-5-5-ureidopentyl-2-yl)amino)-3-methyl-1-oxobutyl-2-yl)-carbamate (compound 1-6)
  • Step 5 synthesis of (9H-fluoren-9-yl)-methyl-((S)-1-(((S)-1-((4-(((R)-2-((t-butyloxycarboryl)amino)-3-phenylpropionamido)methyl)phenyl)amino-1-oxo-5-ureidopentyl-2-yl)amino)-3-methyl-1-oxobutyl-2-yl)-carbamate (compound 1-7)
  • Boc-D-phenylalanine 1.1 g, 4.2 mmol
  • the trifluoroacetate of compound 1-6 (3.0 g, 4.2 mmol) were dissolved in N,N-dimethylformamide (40 mL), cooled to 0° C., and then 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (1.2 g, 6.3 mmol), 1-hydroxybenzotriazole (0.9 g, 6.3 mmol) and N-methylmorpholine (1.7 g, 16.8 mmol) were successively added.
  • the reaction system was stirred for 1.0 h at the temperature.
  • Step 6 synthesis of (9H-fluoren-9-yl)-methyl-((S)-1-(((S)-1-((4-(((R)-2-amino-3-phenylpropionamido)methyl)phenyl)amino)-1-oxo-5-ureidopentyl-2-yl)amino)-3-methyl-1-oxobutyl-2-yl)-carbamate (compound 1-8)
  • Step 7 synthesis of (9H-fluoren-9-yl)-methyl-((S)-1-(((S)-1-((4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-butyrylamido)-N,3-dimethylamino)-3-methoxy-5-methylheptanoyl)pyrro-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)amino)-1-oxo-5-ureidopent-2-yl)amino)-3-methyl-1-oxobutan-2-yl)carbamate (compound 1-9)
  • Step 8 synthesis of (S)-2-((S)-2-amino-3-butyrylamino)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-butyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrro-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)-5-ureidovaleramide (compound 1-10)
  • Step 9 synthesis of (S)-2-((S)-2-(4-(4-chloro-7H-pyrrolo[2,3-d]pyrimidin-7-yl)butyrylamido)-3-methylbutyrylamido)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)-5-ureidovaleramide (compound TL001)
  • Step 1 synthesis of 4-(4-(methylthio)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)butyric acid (compound 2-2)
  • Step 2 synthesis of (S)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)-2-((S)-3-methyl-2-(4-(4-(methylthio)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-butyrylamido)-butyrylamido)-5-ureidovaleramide (compound 2-3)
  • Step 3 synthesis of (S)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)-2-((S)-3-methyl-2-(4-(4-(methylsulfonyl)-7H-pyrrolo[2,3-d]pyrimidin-7-yl)-butyrylamido)-butyrylamido)-5-ureidovaleramide (compound TL002)
  • Step 1 synthesis of methyl 6-(2-(methylthio)pyrimidin-5-yl)-5-hexynoate (compound 3-2)
  • methyl 5-hexynoate 500 mg, 3.97 mmol
  • 5-bromo-2-methylthiopyrimidine were dissolved in N,N-dimethylformamide (3 ml), then triethylamine (3 ml), cuprous iodide (75 mg, 0.4 mmol) and Bis (triphenylphosphine) palladium (II) dichloride (279 mg, 0.4 mmol) were successively added.
  • the resulting mixture was heated to 95° C. under nitrogen protection and reacted for 6 h under stirring, quenched with water, and extracted with ethyl acetate (20 mL ⁇ 3).
  • compound 3-2 (200 mg, 0.8 mmol) was dissolved in a mixed solution of tetrahydrofuran and water (4 mL/4 mL), and lithium hydroxide monohydrate (235 mg, 5.6 mmol) was added, and reacted at room temperature under stirring for 4 h, then diluted with water and extracted with ethyl acetate (20 ml x 2).
  • the desiccant was removed by filtration, and the solvent was evaporated under reduced pressure to obtain the title compound (120 mg).
  • Step 4 N-((S)-1-(((S)-1-((4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)amino)-1-oxo-5-ureido-2-yl)amino)-3-methyl-1-oxybutan-2-yl)-6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamide (compound TL003)
  • Example 8 (S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamide)-ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′ ⁇ ,4 ⁇ ′ ⁇ ,6,7]-indolizino[1,2-b]-quinolin-4-yl(4-((S)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-2-(3-ureidopropyl)-6,12,15,18,21,24,27,30,33-nonoxy-3,9,36-azatetracosan-41-amido)benzyl)carbonate
  • Step 1 synthesis of methyl (S)-(1-((4-(hydroxymethyl)phenyl)amino)-1-oxo-5-ureidopentan-2-yl)-(9H-fluorenyl) carbamate (compound 19-2)
  • Step 2 synthesis of (S)-2-amino-N-(4-(hydroxymethyl)phenyl)-5-ureidovaleramide (compound 19-3)
  • Step 3 synthesis of (S)-2-(32-azido-5-oxo-3,9,12,15,18,21,24,27,30-nonyloxa-6-azatriacetamido)-N-(4-(hydroxymethyl)phenyl)-5-ureidovaleramide (compound 19-4)
  • Step 4 synthesis of 4-((S)-35-azido-4,8-dioxo-2-(3-ureidopropyl)-6,12,15,18,21,24,27,30,33-nonoxy-3,9-azatetracosane)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonylamino)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound 19-5)
  • Step 5 synthesis of 4-((S)-35-amino-4,8-dioxo-2-(3-ureidopropyl)-6,12,15,18,21,24,27,30,33-nonoxy-3,9-azatetracosane)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonylamino)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound 19-6)
  • Step 6 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamide)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl(4-((S)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-2-(3-ureidopropyl)-6,12,15,18,21,24,27,30,33-nonoxy-3,9,36-azatetracosan-41-amido)benzyl)carbonate (compound TL019)
  • Step 1 methyl ((S)-1-(((S)-1-((4-(hydroxymethyl)phenyl)amino)-1-oxo-5-ureidovalerylamido-2-yl)amino))-3-methyl-butyramido-2-yl)-(9H-fluorenyl) carbamate
  • Step 2 synthesis of (S)-2-((S)-2-amino-3-methylbutyramido)-N-(4-(hydroxymethyl)phenyl)-5-ureidovaleramide (compound 28-2)
  • Step 3 synthesis of N-((S)-1-(((S)-1-((4-hydroxymethyl)phenyl)amino)-1-oxo-5-ureidopent-2-yl)amino)-3-methyl-1-oxobutan-2-yl)-6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamide(compound 28-4)
  • Step 4 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamide)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl-(4-((S)-2-((S)-3-methyl-2-(6-(2-(methylsulfonyl)pyrimidin-5-yl))-5-hexynamido)butyramido-5-ureidovalerylamido)benzyl)carbonate (compound TL028)
  • Step 1 synthesis of (S)-2-((S)-35-azido-2-isopropyl-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-azatetracosyl)-N-(4-(hydroxymethyl)phenyl)-5-ureidovaleramide (compound 29-1)
  • Step 2 synthesis of 4-((2S,SS)-38-azido-5-isopropyl-4,7,11-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonoxy-3,6,12-triazatritriacontylamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound 29-2)
  • Step 3 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamide)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′,6,7]indolizino[1,2-b]quinolin-4-yl-(4-((2S,5S)-5-isopropyl-38-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,7,11-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonoxy-3,6,12-triazotritriacontylamido)benzyl)carbonate (compound TL029)
  • Step 1 synthesis of (S)-2-((S)-35-amino-2-isopropyl-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)-N-(4-(hydroxymethyl)phenyl)-5-ureidovaleramide
  • compound 29-1 400 mg, 0.44 mmol was dissolved in methanol and tetrahydrofuran (2.0 mL: 4.0 mL). After complete dissolution, platinum dioxide (40 mg) was added in one batch under nitrogen protection, then the mixed solution was subject to hydrogen substitution for three times. Hydrogenation was conducted at 20° C. for 2 hours. The reaction solution was filtered. The filter cake was washed with methanol. The filtrate was concentrated under reduced pressure. The residue was purified by preparative high performance liquid chromatography (method D) to obtain the title compound (200 mg). ESI-MS (m/z): 890.4[M+H] + .
  • Step 2 synthesis of N-((6S,9S)-1-amino-6-((4-(hydroxymethyl)phenyl)carbamoyl)-9-isopropyl-1,8,11,15-tetraoxo-13,19,22,25,28,31,34,37,40-nonoxy-2,7,10,16-tetraazadotetracont-42-yl)-6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynylamide
  • compound 22-1 250 mg, 0.28 mmol was dissolved in N,N-dimethylformamide (1.0 mL), then HATU (160 mg, 0.42 mmol) and N,N-diisopropylethylamine (109 mg, 0.84 mmol) were successively added, followed by stirring overnight at room temperature. Purification was performed on preparative high performance liquid chromatography (method D) to obtain the title compound (250 mg).
  • Step 3 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((2S,5S)-5-isopropyl-45-(2-(methylsulfonyl)pyrimidin-5-yl)-4,7,11,40-tetraoxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonoxy-3,6, 12,39-tetraazapentatetracontane-44-carbamoyl)benzyl)carbonate (compound TL022)
  • Example 12 4-((S)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadedotetracontyl-41-alkynamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonyl)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((S)-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadotetracontyl-41-alkynamido)benzylcarbonate
  • 6-(-2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynoic acid (12 mg, 0.045 mmol) was dissolved in dichloromethane (2 mL), then 2-(7-azobenzotriazol)-N,N,N′,N′-tetramethylureahexafluorophosphate (21.2 mg, 0.056 mmol) and N,N-diisopropylethylamine (8.6 mg, 0.067 mmol) were added and stirred for 10 min, and compound 24-1 (35 mg, 0.022 mmol) was added and reacted for 1h under stirring. Purification was performed on preparative high performance liquid chromatography (method B) to obtain the title compound (20 mg). ESI-MS (m/z): 1821.8[M+H] + .
  • Step 2 synthesis of 4-((S)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadedotetracontyl-41-alkynamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonyl)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound TL024)
  • Step 1 preparation of (S)-(9H-fluoren-9-yl)-methyl(1-((4-(hydroxymethyl)phenyl)amino)-1 -oxopropyl-2-yl)carbamate
  • Step 3 preparation of (9H-fluoren-9-yl)-methyl((S)-1-(((S)-1-((4-(hydroxymethyl)phenyl)amino)-1-oxopropan-2-yl)amino)-3-methyl-1-oxobutan-2-yl)-carbamate
  • Step 4 preparation of (S)-2-amino-N-((S)-1-((4-(hydroxymethyl)phenyl)amino)-1-oxoprop-2-yl)-3-methylbutanamide
  • Step 5 preparation of (S)-2-(32-azido-5-oxo-3,9,12,15,18,21,24,27,30-nonoxy-6-diazapentatriacontamido)-N-((S)-1-((4-(hydroxymethyl)phenyl)amino)-1-oxopropan-2-yl)-3-methylbutyramide
  • Step 6 preparation of 4-((2S,5S)-38 azido-5-isopropyl-2-methyl-4,7,11-trioxo-9,15,18,21,24,27,30,33,36-nonoxy-3,6,12-triazatriacontamino)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonylamino)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 7 preparation of (S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamide)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7indolizino[1,2-b]quinolin-4-yl-4-((2S,5S)-5-isopropyl-2-methyl-38-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,7,11-trioxo-9,15,18,21,24,27,30,33,36-nonoxy-3,6,12-triazatriacontazanamido)benzylcarbonate (compound TL030)
  • cuprous bromide (5 mg, 0.04 mmol) and compound 30-7 (20 mg, 15 umol) were added dropwise to a solution of 6-(2-(methylsulfonyl)pyrimidin-5-yl)-N-(prop-2-yn-1-yl)-hex-5-ynylamide (9 mg, 0.007 mmol) in water and N,N-dimethylformamide (0.2 ml: 0.8 ml) and reacted for 4 hours under stirring. Purification was performed on preparative high performance liquid chromatography (method D) to obtain the title compound (4.15 mg). ESI-MS (m/z): 1672.7 [M+H] +
  • Example 14 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11 -(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of 6-(2-(methylsulfonyl)pyrimidin-5-yl)-N-(prop-2-yn-1-yl)hex-5-ynamide
  • prop-2-ynyl-1-amine (189 mg, 3.4 mmol) and compound 3-4 (800 mg, 2.83 mmol) were dissolved in dichloromethane (10 mL), then N,N-diisopropylethylamine (738 mg, 5.67 mmol) and O-(7-azabenzotriazol-1-yl)-N,N,N′,N′-tetramethyluronium hexafluorophosphate (1.63 g, 4.25 mmol) were successively added and reacted for 2 hours under stirring.
  • Step 2 synthesis of 4-((S)-35-azido-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonazo-3,9-diazapentatriacontamino)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamide)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-2H-pyrano[2,3-b]-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • T-030 250 mg, 0.49 mmol was dissolved in dichloromethane (10 mL) and cooled to 0° C., then a solution of 4-dimethylaminopyridine (478 mg, 3.91 mmol) in dichloromethane (3 mL) was added, followed by the slow and dropwise addition of a solution of triphosgene (72 mg, 0.24 mmol) in dichloromethane (10 mL) and reacted at 0° C. for 20 min under stirring.
  • Step 3 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((S)-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl)carbonate
  • Step 4 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound TL033)
  • Example 15 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl((S)-11-diethyl-9-hydroxy-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-carbonate
  • Step 1 synthesis of 4-((S)-35-azido-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-4,8-dioxo 6,12,15,18,21,24,27-nonoxy-((S)-9-((tert-butyldimethylsilyl)oxy)-4,11-diethyl-3,14-dioxo-3,4,12,14-tetrahydro-1,2,3,4-tetrahydroquinolin-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • compound 34-1 (100 mg, 0.2 mmol) was dissolved in anhydrous dichloromethane (2 ml) under nitrogen protection, then cooled to 0° C., followed by an addition of a solution of 4-dimethylaminopyridine (144 mg, 1.18 mmol) in anhydrous dichloromethane (0.5 ml), then a solution of triphosgene (41 mg, 0.14 mmol) in dry dichloromethane (0.5 ml) was slowly added dropwise. The resulting mixture was reacted at 0° C. for 1h under stirring.
  • Step 2 synthesis of (S)-9-(tert-butyldimethylsilyl)oxy)-4,11-diethyl-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl-4-((S)-2-(4-(((6-2-(methylsulfonyl)pyrimidin-5-yl)-35-(4-((6-2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-dioxo 6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)carbonate
  • Step 3 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl((S)-11-diethyl-9-hydroxy-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-carbonate(compound TL034)
  • Example 16 Synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of ((S)-35-azido-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1,2,3,6-triazacycloheptane-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 2 synthesis of (S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)-4-((S)-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-35-(4-((6-2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido]carbonate
  • Step 3 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound TL035)
  • Example 17 Synthesis of 4-((S,Z)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadotetracontyl-41-alkenamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of (Z)-6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-enoic acid
  • Step 2 synthesis of (S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((S,Z)-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadotetracontyl-41-alkenamido)benzylcarbonate
  • Step 3 synthesis of 4-((S,Z)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadedotetracontyl-41-alkenamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound TL045)
  • Example 18 4-((S)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadedotetracontyl-41-alkynamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of (S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((S)-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadotetracontyl-41-alkynamido)benzylcarbonate
  • Step 2 synthesis of 4-((S)-2-(4-aminobutyl)-42-(2-(methylsulfonyl)pyrimidin-5-yl)-4,8,37-trioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9,36-triazadedotetracontyl-41-alkynamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylacetamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate (compound TL048)
  • Example 19 4-((S)-2-(4-aminobutyl)-35-(4-((2-(2-((methylsulfonyl)pyrimidin-5-yl)thiazol-4-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of 2-(2-(methylthio)pyrimidin-5-yl)thiazol-4-carboxylic acid
  • Step 2 synthesis of 2-(2-(methylsulfonyl)pyrimidin-5-yl)thiazol-4-carboxylic acid
  • Step 3 synthesis of 2-(2-(methylsulfonyl)pyrimidin-5-yl)-N-(prop-2-yn-1-yl)thiazo-4-carboxamide
  • Step 4 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl-(4-((S)-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyl)-35-(4-((2-(2-(methylsulfonyl)pyrimidin-5-yl)thiazol-4-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonazeza-3,9-diazapentatriacontamino)benzyl)carbonate
  • Step 5 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((2-(2-((methylsulfonyl)pyrimidin-5-yl)thiazol-4-carboxamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrone[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Example 20 4-((S)-2-(4-aminobutyric acid)-35-(4-((2-(2-(methylsulfonyl)pyrimidin-5-yl)-oxazol-4-formylamino)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of ethyl 2-(2-(methylthio)pyrimidin-5-yl)oxazol-4-carboxylate
  • ethyl 2-bromooxazol-4-carboxylate 100 mg, 0.45 mmol
  • compound 49-1 126 mg, 0.50 mmol
  • potassium carbonate 125 mg, 0.9 mmol
  • [1,1′-bis(diphenylphosphino)ferrocen]palladium dichloride 33 mg, 0.05 mmol
  • Step 2 synthesis of 2-(2-(methylthio)pyrimidin-5-yl)oxazol-4-carboxylic acid
  • compound 50-1 (50 mg, 0.19 mmol) was dissolved in a mixed solvent of tetrahydrofuran and water (4 mL/2 mL), after complete dissolution, lithium hydroxide monohydrate (40 mg, 0.94 mmol) was added thereto and reacted at 25° C. for 1 h.
  • the reaction solution was diluted with water (15 mL) and extracted with ethyl acetate (20 mL ⁇ 2).
  • Step 3 synthesis of 2-(2-(methylsulfonyl)pyrimidin-5-yl)oxazol-4-carboxylic acid
  • Step 4 synthesis of 2-(2-(methylsulfonyl)pyrimidin-5-yl)-N-(prop-2-yn-1-yl)-oxazol-4-formamide
  • Step 5 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl4-((S)-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-35-(4-((2-(2-(methylsulfonyl)pyrimidin-5-yl)oxazol-4-formylamino)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl)carbonate
  • Step 6 synthesis of 4-((S)-2-(4-aminobutyric acid)-35-(4-((2-(2-(methylsulfonyl)pyrimidin-5-yl)oxazol-4-formylamino)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl-((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of 9-fluorenylmethyl ((S)-1-(((S)-1-((4-(((S)2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-butyrylamido)-N,3-dimethylbutyrylamido)-3-methoxy-5-methylheptanoyl)pyrro-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)amino)-1-oxo-5-ureidopent-2-yl)amino)-3-methyl-1-oxobutan-2-yl) carbamate
  • Step 2 synthesis of (S)-2-((S)-2-amino-3-butyrylamino)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-butyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptanoyl)pyrro-2-yl)-3-methoxy-2-methylpropionamido)-3-phenylpropionamido)methyl)phenyl)-5-ureidovaleramide
  • Step 3 synthesis of (S)-2-((S)-35-azido-2-isopropyl-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)-N-(4-(((S)-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-((S)-2-(dimethylamino)-3-methylbutanamido)-N,3-dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanamido)-3-phenylpropanamido)methyl)phenyl)-5-ureidovaleramide
  • Step 4 synthesis of N-((1-((6S,9S)-1-amino-6-((4-(((S)-2-((2R,3R)-3-((S)1-((3R,4S,SS)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutanamido)-N,3-dimethylbutanamido)-3-methoxy-5-methylheptanoyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropanamido)-3-phenylpropanamido)methyl)phenyl)carbamoyl)-9-isopropyl-1,8,11,15-tetraoxy-13,19,22,25,28,31,34,37,40-nonaoxa-2,7,10,16-tetraazaanthracen-42-yl)-1H-1,2,3-triazol-4-yl)methyl)-6-(
  • Example 22 4-((2S,5S)-5-isopropyl-38-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,7,1 1-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonaoxa-3,6,12-triazadotetracontyl)benzyl-((S)-1-(((S)-1-(((3R,4S,SS)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-yl)
  • Step 1 4-((2S,SS)-38-azido-5-isopropyl-4,7,11-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonaoxa-3,6, 12-triazadotetracontyl)benzyl-((S)-1-(((S)1-(((3R,4S,SS)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-yl)(methyl)amino)-3-methyl-1-oxobutan-2-yl)(methyl)carbamate
  • Step 2 synthesis of 4-((2S,5S)-5-isopropyl-38-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,7, 11-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonaoxa-3,6, 12-triazadotetracontyl)benzyl-((S)-1-(((S)-1-(((3R,4S,SS)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-y
  • Step 1 synthesis of 4-((2S,5S)-38-azido-5-isopropyl-4,7,11-trioxo-2-(3-ureidopropyl)-9,15,18,21,24,27,30,33,36-nonoxy- ⁇ 3,6,12-triazaoctatriacontamino)benzyl-(4-nitrophenyl)-carbonate
  • Step 2 synthesis of (S)-2-((2R ⁇ ,3R)-3-((2S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-((((4-((S)-2-((S)-2-(32-azido-5-oxo-3,9,12,15,18,21,24,27,30-nonoxy-6-azatriacontamido)-3-methylbutyrylamino)-5-ureidopentanoylamino)benzyl)oxy)carbonyl)(methyl)amino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionyl)-L-phenylalanine
  • compound 53-1 60 mg, 0.06 mmol
  • compound 53-1 60 mg, 0.06 mmol
  • ((2R)-3-((2S)-1-((3R, 5S)-4-((S)-N,3-dimethyl-2-((S)-3-methyl-2-(methylamino)butyrylamino)butyrylamino)-3-methoxy-5-methylheptyl)pyrrolidine-2-yl)-3-methoxy-2-methypropionyl)-L-phenylalanine 41 mg, 0.06 mmol
  • N,N-dimethylformamide 2 mL
  • 1-hydroxybenzotriazole 8 mg, 0.06 mmol
  • Step 3 synthesis of (S)-2-((2R,3R)-3-((2S)-1-((3R,4S,5S)-4-((S)-N,3-dimethyl-2-((S)-3-methyl2-(methyl(((4-((S)-2-((S)-3-methyl-2-(32-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-carbamoyl)methyl)-1H-1,2,3-triazol-1-yl)-5-oxo-3,9,12,15,18,21,24,27,30-nonoxy-6-azatriacontamino)butyrylamino)-5-ureidovalerylamino)benzyl)oxy)carbonyl)amino)butyrylamino)-3-methoxy-5-methylheptyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionyl)-L-pheny
  • Example 24 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl-((S)-1-(((S)-1-(((3R,4S,5S)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-yl)(methyl)amino)-3-methyl-1-o
  • Step 1 synthesis of (S)-4-(35-azido-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyryl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl-(4-nitrophenyl)-carbonate
  • Step 2 synthesis of 4-((S)-35-azide-2-(4-(((4-methoxyphenyl) benzhydryl)amino)butyryl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl((S)-1-(((S)-1-(((3R,4S,SS)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-yl)(methyl)amino)-3-methyl-1-oxobutan-2-yl)amino)-3-methyl-1-oxobutan-2-yl)(methyl
  • Step 3 synthesis of 4-((S)-2-(4-(((4-methoxyphenyl)benzhydryl)amino)butyryl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl((S)-1-(((S)-1-(((3R,4S,SS)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyhept
  • Step 4 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)-5-hexynamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonaoxa-3,9-diazapentatriacontamino)benzyl-((S)-1-(((S)-1-(((3R,4S,5S)-1-((S)-2-((1R,2R)-3-(((1S,2R)-1-hydroxy-1-phenylpropan-2-yl)amino)-1-methoxy-2-methyl-3-oxypropyl)pyrrolidin-1-yl)-3-methoxy-5-methyl-1-oxyheptan-4-yl)(methyl)amino)-3-methyl
  • Example 25 Synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((4-(2-(methylsulfonyl)pyrimidin-5-yl)benzamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3 ⁇ ′ ⁇ ,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • Step 1 synthesis of methyl 4-(2-(methylthio)pyrimidin-5-yl)benzoate
  • Step 3 synthesis of 4-(2-(methylsulfonyl)pyrimidin-5-yl)benzoic acid
  • Step 4 synthesis of 4-(2-(methylsulfonyl)pyrimidin-5-yl)-N-(prop-2-yn-1-yl)benzamide
  • benzotriazole-N,N,N′,N′-tetramethyluronium hexafluorophosphate 100 mg, 0.26 mmol
  • dichloromethane 10 mL
  • propynylamine 10 mg, 0.2 mmol
  • N,N-diisopropylethylamine 70 mg, 0.5 mmol
  • the reaction solution was purified by silica gel column chromatography to obtain the title compound (20 mg).
  • Step 5 synthesis of (S)-4-ethyl-11-(2-(N-isopropylmethanesulfonamido)ethyl)-3,14-dioxo-3,4,12,14-tetrahydro-1H-pyrano[3′,4′:6,7]indolizino[1,2-b]quinolin-4-yl(4-((S)-2-(4-(((4-methoxyphenyl)diphenylmethyl)amino)butyl)-35-(4-((4-(2-(methylsulfonyl)pyrimidin-5-yl)benzamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl)carbonate
  • cuprous iodide (10 mg, 0.05 mmol) and water (2 mL) were successively added to dimethyl sulfoxide solution (2 mL) of compound 55-4 (16 mg, 0.05 mmol) and compound 33-1 (80 mg, 0.05 mmol) and reacted for 1 h under stirring.
  • Purificaiton (method B) was performed to obtain the title compound (79 mg).
  • Step 6 synthesis of 4-((S)-2-(4-aminobutyl)-35-(4-((4-(2-(methylsulfonyl)pyrimidin-5-yl)benzamido)methyl)-1H-1,2,3-triazol-1-yl)-4,8-dioxo-6,12,15,18,21,24,27,30,33-nonoxy-3,9-diazapentatriacontamido)benzyl((S)-4-ethyl-11-(2-(N-isopropylmethylsulfonamido)ethyl)- ⁇ 3,14-dioxo- ⁇ 3,4,12,14-tetrahydro-1H-pyrano[3 ⁇ ′ ⁇ ,4′:6,7]indolizino[1,2-b]quinolin-4-yl)carbonate
  • compound 55-5 (55 mg, 0.029 mmol) was added to trifluoroacetic acid (0.5 mL) in a mixed solvent of water/acetonitrile (0.1 mL/0.5 mL), and reacted for 15 min under stirring.
  • the reaction solution was purified by preparative high performance liquid chromatography (method C) to obtain the trifluoroacetate of the title compound (42 mg).
  • ESI-MS (m/z):821.0[M/2+H] + .
  • Step 1 synthesis of 32-(4-((6-(2-(methylsulfonyl)pyrimidin-5-yl)hex-5-ynamido)methyl)-1H-1,2,3-triazol-1-yl)-5-oxo-3,9,12,15,18,21,24,27,30-nonoxy-6-azadotriacontanoic acid
  • Step 2 synthesis of (9H-fluoren-9-yl)methyl((S)-1-(((S)-1-((4-((S)-3-azido-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)propyl)phenyl)amino)-1-oxo-5-ureidopentanoylamino-2-yl)amino)-3-methyl-1 -oxobutan-2-yl)carbamate
  • Step 3 synthesis of (S)-2-((S)-2-amino-3-methylbutyrylamino)-N-(4-((S)-3-azido-2-((2R,3R)-3-((S)-l-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)propyl)phenyl)-5-ureidovaleramide
  • Step 4 synthesis of N-((1-((6S,9S)-1-amino-6-((4-((S)-3-azido-2-((2R,3R)-3-((S)-1-((3R,4S,5S)-4-((S)-2-((S)-2-(dimethylamino)-3-methylbutyrylamino)-N,3-dimethylbutyrylamino)-3-methoxy-5-methylheptyl)pyrrolidin-2-yl)-3-methoxy-2-methylpropionamido)propyl)phenyl)carbamoyl)-9-isopropyl-1,8,11,15-tetraoxo-13,19,22,25,28,31,34,37,40-nonoxy-2,7,10,16-tetraazadotetracont-42-yl)-1H-1,2,3-triazol-4-yl)methyl)-6-(2-(
  • antibody Sacituzumab (anti-Trop-2, 33.5 mg/mL) was diluted with 0.25 ml of a solution (pH 7.6) containing 20 mM PB, 150 mM NaCl and 20 mM sodium edetate, to which 0.45 ml of a solution (pH 7.6) containing 20 mM PB and 150 mM NaCl was added and evenly mixed.
  • TCEP tris(2-carboxyethyl)phosphine
  • TL003 dissolved in dimethyl sulfoxide was added in an amount of 15 equiv. and evenly mixed, which was allowed to stand at room temperature for 2 hours. After the addition, 6.1 ⁇ l of 100 mM cysteine was added to terminate the reaction. At last, the buffer was replaced with a 20 mM PB buffer solution of pH 6.44 by G-25 gel column to obtain the coupling product of TL003 with Sacituzumab, which was named as BT001002.
  • Sacituzumab (anti-Trop-2, 17.6 mg/mL) was diluted with 0.095 mL of a diluent (a solution containing 20 mM PB, 150 mM NaCl and 20 mM sodium edetate, pH 7.6). Then the diluted solution was adjusted with 1 M Na 2 HPO 4 solution to pH 7.4, and 10 mM TCEP solution was added and evenly mixed, which was allowed to stand at room temperature for 30 min. To the solution system, TL019 dissolved in dimethyl sulfoxide was added in an amount of 9 equiv. and evenly mixed, which was allowed to stand at room temperature for 2 h. At last, the buffer was replaced with a PBS buffer solution of pH 6.5 by G-25 gel column to obtain the coupling product of TL019 with Sacituzumab, which was named as BT001004.
  • a diluent a solution containing 20 mM PB, 150 mM NaCl and 20 mM sodium
  • Sacituzumab (anti-Trop-2, 33.5 mg/mL) was diluted with 0.25 ml of a solution (pH 7.6) containing 20 mM PB, 150 mM NaCl and 20 mM sodium edetate, then 0.45 mL of a solution (pH 7.6) containing 20 mM PB and 150 mM NaCl was added and evenly mixed.
  • TCEP tris(2-carboxyethyl)phosphine
  • 0.3 mL of antibody M3 (anti-Trop-2, 33.5 mg/mL) was diluted with 0.25 ml of a solution (pH 7.6) containing 20 mM PB, 150 mM NaCl and 20 mM sodium edetate, then 0.45 mL of a solution (pH 7.6) containing 20 mM PB and 150 mM NaCl was added and evenly mixed.
  • TCEP tris(2-carboxyethyl)phosphine
  • Example 48 Determination of Molecular Weight of BT001002 by LC-MS
  • mAb stands for a monoclonal antibody
  • LC stands for the light chain of an antibody
  • HC stands for the heavy chain of an antibody
  • DAR1 stands for a conjugate containing a light chain/heavy chain of an antibody and a bioactive molecule
  • DAR2 stands for a conjugate containing a light chain/heavy chain of an antibody and two bioactive molecules
  • DAR3 stands for a conjugate containing a light chain/heavy chain of an antibody and three bioactive molecules
  • DAR4 stands for a conjugate containing a light chain/heavy chain of an antibody and four bioactive molecules
  • glycoform stands for the structure of glycan of the two heavy chains:
  • G0F stands for fucosylation and free of galactosylation.
  • both the molecular weights of the light chain and the heavy chain of the antibody are changed after being coupled with TL003, wherein the light chain was coupled with 1 bioactive molecule and the heavy chain was coupled with 3 bioactive molecules. Therefore, it could be inferred that the DAR of the antibody to the bioactive molecule was 8.
  • the molecular weight of BT001004 obtained from coupling was analyzed by LC-MS.
  • LC stands for the light chain of an antibody
  • HC stands for the heavy chain of an antibody
  • the light chain of the antibody was coupled with 0-1 bioactive molecule (LC and DAR1 accounted for 14% and 86%, respectively), and the heavy chain was coupled with 1-3 bioactive molecules (DAR1, DAR2 and DAR3 accounted for 13%, 19% and 68%, respectively). Therefore, it could be calculated that the DAR of the antibody to the bioactive molecule was 7.0.
  • Example 50 Determination of Molecular Weight of BT001012 by LC-MS

Landscapes

  • Chemical & Material Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • General Health & Medical Sciences (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Epidemiology (AREA)
  • Biochemistry (AREA)
  • Molecular Biology (AREA)
  • Genetics & Genomics (AREA)
  • Biophysics (AREA)
  • Cell Biology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Oncology (AREA)
  • Biotechnology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Peptides Or Proteins (AREA)
  • Medicinal Preparation (AREA)
  • Medicines That Contain Protein Lipid Enzymes And Other Medicines (AREA)
  • Pyrrole Compounds (AREA)
  • Nitrogen Condensed Heterocyclic Rings (AREA)
  • Plural Heterocyclic Compounds (AREA)
US18/155,650 2017-12-15 2023-01-17 Bioactive conjugate, preparation method therefor and use thereof Pending US20230357259A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
US18/155,650 US20230357259A1 (en) 2017-12-15 2023-01-17 Bioactive conjugate, preparation method therefor and use thereof

Applications Claiming Priority (9)

Application Number Priority Date Filing Date Title
CN201711347535 2017-12-15
CN201711347535.6 2017-12-15
CN201810230346.9 2018-03-20
CN201810230346 2018-03-20
CN201811071947 2018-09-14
CN201811071947.6 2018-09-14
PCT/CN2018/120125 WO2019114666A1 (zh) 2017-12-15 2018-12-10 生物活性物偶联物及其制备方法和用途
US202016758980A 2020-04-24 2020-04-24
US18/155,650 US20230357259A1 (en) 2017-12-15 2023-01-17 Bioactive conjugate, preparation method therefor and use thereof

Related Parent Applications (2)

Application Number Title Priority Date Filing Date
PCT/CN2018/120125 Division WO2019114666A1 (zh) 2017-12-15 2018-12-10 生物活性物偶联物及其制备方法和用途
US16/758,980 Division US11970506B2 (en) 2017-12-15 2018-12-10 Bioactive molecule conjugate, preparation method and use thereof

Publications (1)

Publication Number Publication Date
US20230357259A1 true US20230357259A1 (en) 2023-11-09

Family

ID=66819957

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/758,980 Active 2040-07-31 US11970506B2 (en) 2017-12-15 2018-12-10 Bioactive molecule conjugate, preparation method and use thereof
US18/155,650 Pending US20230357259A1 (en) 2017-12-15 2023-01-17 Bioactive conjugate, preparation method therefor and use thereof

Family Applications Before (1)

Application Number Title Priority Date Filing Date
US16/758,980 Active 2040-07-31 US11970506B2 (en) 2017-12-15 2018-12-10 Bioactive molecule conjugate, preparation method and use thereof

Country Status (7)

Country Link
US (2) US11970506B2 (zh)
EP (1) EP3725798A4 (zh)
JP (2) JP7446993B2 (zh)
KR (1) KR20200099123A (zh)
CN (5) CN111295389B (zh)
CA (1) CA3080236A1 (zh)
WO (1) WO2019114666A1 (zh)

Families Citing this family (37)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11207420B2 (en) * 2017-04-19 2021-12-28 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Cytotoxin and conjugate, uses of same and preparation method therefor
CN111295389B (zh) 2017-12-15 2021-10-22 四川科伦博泰生物医药股份有限公司 生物活性物偶联物及其制备方法和用途
CN113286796A (zh) * 2019-01-30 2021-08-20 四川科伦博泰生物医药股份有限公司 喜树碱衍生物及其水溶性前药、包含其的药物组合物及其制备方法和用途
CN112390885B (zh) * 2019-08-12 2024-01-19 凯惠科技发展(上海)有限公司 一种trop2抗体及其制备方法、其偶联物和应用
KR20220130160A (ko) * 2020-01-22 2022-09-26 상하이 센후이 메디슨 컴퍼니 리미티드 에리불린 유도체의 약물 접합체, 이를 제조하기 위한 방법 및 의료분야에서 이의 용도
BR112022014189A2 (pt) * 2020-01-22 2022-11-29 Jiangsu Hengrui Medicine Co Conjugado de anticorpo anti-trop-2 análogo de exatecano e uso médico do mesmo
CR20220580A (es) * 2020-05-15 2023-04-11 Sichuan Kelun Biotech Biopharmaceutical Co Ltd Conjugado anticuerpo-fármaco, método de preparación del mismo y uso del mismo
CN116234564A (zh) 2020-09-16 2023-06-06 四川科伦博泰生物医药股份有限公司 抗Nectin-4抗体、包含所述抗体的缀合物及其应用
MX2023003448A (es) * 2020-09-30 2023-04-19 Jiangsu Hengrui Pharmaceuticals Co Ltd Composicion farmaceutica que comprende conjugado de anticuerpo-farmaco y uso de la misma.
TW202241521A (zh) * 2021-02-09 2022-11-01 大陸商蘇州宜聯生物醫藥有限公司 生物活性物偶聯物及其製備方法和用途
WO2022184082A1 (en) * 2021-03-03 2022-09-09 Sorrento Therapeutics, Inc. Antibody-drug conjugates comprising an anti-bcma antibody
CN117279664A (zh) 2021-04-10 2023-12-22 普方生物制药美国公司 Folr1结合剂、其偶联物及其使用方法
JP2024516631A (ja) 2021-04-23 2024-04-16 プロファウンドバイオ ユーエス カンパニー 抗cd70抗体、そのコンジュゲートおよびこれを使用する方法
CN114106088A (zh) * 2021-04-28 2022-03-01 联宁(苏州)生物制药有限公司 基于溴甲基吡嗪的药物偶联物及adc
JP2024516217A (ja) * 2021-04-30 2024-04-12 シチュアン ケルン-バイオテック バイオファーマシューティカル カンパニー リミテッド 腫瘍疾患の治療における医薬の使用
CN113181373B (zh) * 2021-05-10 2024-03-01 深圳安特生物医药科技有限公司 一种抗体药物偶联制剂及其制备方法和应用
AU2022287001A1 (en) * 2021-06-02 2023-10-26 Klus Pharma Inc. Antibody drug conjugate, and preparation method therefor and use thereof
TW202320857A (zh) 2021-07-06 2023-06-01 美商普方生物製藥美國公司 連接子、藥物連接子及其結合物及其使用方法
CN118043080A (zh) * 2021-08-13 2024-05-14 昆山新蕴达生物科技有限公司 一种基于微管抑制剂的抗体偶联药物
WO2023030064A1 (zh) * 2021-09-01 2023-03-09 四川科伦博泰生物医药股份有限公司 一种提高抗体药物偶联物产品质量的方法
CN115991784A (zh) 2021-10-19 2023-04-21 宝船生物医药科技(上海)有限公司 抗cd47-cldn18.2双特异性抗体及其用途
WO2023098515A1 (zh) * 2021-12-02 2023-06-08 四川科伦博泰生物医药股份有限公司 一种药物-接头偶联物的制备方法
WO2023122228A1 (en) * 2021-12-23 2023-06-29 Adcentrx Therapeutics Inc. Novel auristatin analogs and immunoconjugates thereof
WO2023134706A1 (zh) * 2022-01-13 2023-07-20 四川科伦博泰生物医药股份有限公司 抗trop-2抗体偶联药物和其他治疗剂的组合用途
WO2023143208A1 (zh) * 2022-01-26 2023-08-03 苏州宜联生物医药有限公司 一种药物连接体偶联物的制备方法
WO2023143154A1 (zh) * 2022-01-28 2023-08-03 四川科伦博泰生物医药股份有限公司 一种药物组合物及其制备方法和应用
WO2023155808A1 (zh) * 2022-02-16 2023-08-24 苏州宜联生物医药有限公司 抗体-艾日布林或其衍生物的偶联物、其中间体、制备方法、药物组合物和用途
WO2023201267A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating trop-2 expressing cancers
WO2023201268A1 (en) 2022-04-13 2023-10-19 Gilead Sciences, Inc. Combination therapy for treating tumor antigen expressing cancers
WO2023221975A1 (zh) * 2022-05-18 2023-11-23 苏州宜联生物医药有限公司 包含蛋白降解剂类生物活性化合物的抗体药物偶联物及其制备方法和用途
CN117143177A (zh) * 2022-05-30 2023-12-01 苏州宜联生物医药有限公司 一种药物连接体偶联物的制备方法及其中间体
WO2024002042A1 (zh) * 2022-06-27 2024-01-04 百奥泰生物制药股份有限公司 治疗实体瘤的方法
WO2024012524A1 (zh) * 2022-07-14 2024-01-18 苏州宜联生物医药有限公司 抗体药物偶联物及其制备方法和用途
WO2024012566A2 (en) * 2022-07-15 2024-01-18 Genequantum Healthcare (Suzhou) Co., Ltd. Antibody, linkers, payload, conjugates and applications thereof
WO2024027708A1 (zh) * 2022-08-02 2024-02-08 诺纳生物(苏州)有限公司 Msln抗体药物偶联物
WO2024078586A1 (en) 2022-10-14 2024-04-18 Sichuan Kelun-Biotech Biopharmaceutical Co., Ltd. Antibody-drug conjugate binding to human ptk7 and method for preparation and use thereof
WO2024097812A1 (en) 2022-11-04 2024-05-10 Gilead Sciences, Inc. Therapy for treating bladder cancer

Family Cites Families (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4816567A (en) 1983-04-08 1989-03-28 Genentech, Inc. Recombinant immunoglobin preparations
AP9300587A0 (en) * 1992-11-12 1995-05-05 Glaxo Inc Water soluble camptothecin derivatives.
US5410024A (en) 1993-01-21 1995-04-25 Arizona Board Of Regents Acting On Behalf Of Arizona State University Human cancer inhibitory pentapeptide amides
JP3469580B2 (ja) 1993-10-01 2003-11-25 帝国臓器製薬株式会社 新規なペプチド誘導体
DK3483183T3 (da) 2002-03-01 2021-06-21 Immunomedics Inc Immunokonjugat omfattende humaniserede rs7-antistoffer
EP3120861B1 (en) 2003-11-06 2018-08-15 Seattle Genetics, Inc. Intermediate for conjugate preparation comprising auristatin derivatives and a linker
US7560111B2 (en) 2004-07-22 2009-07-14 Genentech, Inc. HER2 antibody composition
ES2647927T3 (es) 2008-03-18 2017-12-27 Seattle Genetics, Inc. Conjugados enlazadores del fármaco auriestatina
US8609092B2 (en) 2010-02-08 2013-12-17 Agensys, Inc. Antibody drug conjugates (ADC) that bind to 161P2F10B proteins
WO2011130305A2 (en) 2010-04-12 2011-10-20 Sorrento Therapeutics Inc. Method for displaying antibodies
JPWO2011155579A1 (ja) 2010-06-10 2013-08-15 北海道公立大学法人 札幌医科大学 抗Trop−2抗体
PL3327027T3 (pl) 2011-11-17 2021-06-28 Pfizer Inc. Cytotoksyczne peptydy i ich koniugaty przeciwciało lek
CN107982545B (zh) 2012-05-15 2021-04-09 联宁(苏州)生物制药有限公司 药物偶联物,偶联方法,及其用途
US20140017265A1 (en) 2012-07-05 2014-01-16 Mersana Therapeutics, Inc. Terminally Modified Polymers and Conjugates Thereof
KR20230078823A (ko) 2012-12-13 2023-06-02 이뮤노메딕스, 인코오포레이티드 개선된 효능 및 감소된 독성을 위한 항체 및 sn-38의 면역컨쥬게이트의 투약
WO2017004144A1 (en) 2015-07-01 2017-01-05 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
WO2015012904A2 (en) 2012-12-13 2015-01-29 Immunomedics, Inc. Antibody-sn-38 immunoconjugates with a cl2a linker
WO2014144878A2 (en) * 2013-03-15 2014-09-18 The Scripps Research Institute Novel thiol & amino modifying reagents for protein chemistry and methods of use thereof
CN103319599B (zh) 2013-05-27 2015-02-18 上海交联药物研发有限公司 一种抗人ErbB2抗体—美登木素偶联物及其应用
CN110075295A (zh) * 2013-07-23 2019-08-02 免疫医疗公司 具有cl2a接头的抗体-sn-38免疫缀合物
JP6752204B2 (ja) * 2014-12-03 2020-09-09 ジェネンテック, インコーポレイテッド 四級アミン化合物及びその抗体−薬物コンジュゲート
EP4115910A1 (en) 2015-03-09 2023-01-11 Heidelberg Pharma Research GmbH Amatoxin-antibody conjugates
CN115089722A (zh) 2015-06-19 2022-09-23 森托瑞恩生物制药公司 用于控制药物释放的递送系统
AU2016308539B2 (en) * 2015-08-14 2019-01-24 Remegen Co., Ltd. Covalent linkers in antibody-drug conjugates and methods of making and using the same
CN106729743B (zh) 2015-11-23 2021-09-21 四川科伦博泰生物医药股份有限公司 抗ErbB2抗体-药物偶联物及其组合物、制备方法和应用
CN107029244B (zh) 2016-02-04 2021-04-27 浙江昭华生物医药有限公司 抗her2抗体-药物偶联物及其应用
CA3032559C (en) * 2016-08-03 2022-04-19 Pfizer Inc. Heteroaryl sulfone-based conjugation handles, methods for their preparation, and their use in synthesizing antibody drug conjugates
RU2758234C2 (ru) 2017-03-27 2021-10-26 Иммьюномедикс, Инк. ЛЕЧЕНИЕ ТРИЖДЫ НЕГАТИВНОГО РАКА МОЛОЧНОЙ ЖЕЛЕЗЫ, ХАРАКТЕРИЗУЮЩЕГОСЯ ЭКСПРЕССИЕЙ Trop-2, С ПОМОЩЬЮ САЦИТУЗУМАБА ГОВИТЕКАНА И ИНГИБИТОРА Rad51
WO2018227023A1 (en) * 2017-06-07 2018-12-13 Silverback Therapeutics, Inc. Antibody construct conjugates
CN111295389B (zh) * 2017-12-15 2021-10-22 四川科伦博泰生物医药股份有限公司 生物活性物偶联物及其制备方法和用途
CN110903395A (zh) * 2018-09-14 2020-03-24 四川科伦博泰生物医药股份有限公司 抗体、偶联物及其制备方法和用途

Also Published As

Publication number Publication date
WO2019114666A1 (zh) 2019-06-20
CN113651831A (zh) 2021-11-16
US11970506B2 (en) 2024-04-30
CA3080236A1 (en) 2019-06-20
CN113683622A (zh) 2021-11-23
EP3725798A1 (en) 2020-10-21
CN113698414A (zh) 2021-11-26
US20200347075A1 (en) 2020-11-05
JP2021506743A (ja) 2021-02-22
CN111295389A (zh) 2020-06-16
JP7446993B2 (ja) 2024-03-11
EP3725798A4 (en) 2021-11-10
JP2024038168A (ja) 2024-03-19
KR20200099123A (ko) 2020-08-21
US20210101906A2 (en) 2021-04-08
CN113603703A (zh) 2021-11-05
CN111295389B (zh) 2021-10-22

Similar Documents

Publication Publication Date Title
US20230357259A1 (en) Bioactive conjugate, preparation method therefor and use thereof
US20210113710A1 (en) Hydrophilic self-immolative linkers and conjugates thereof
US20160051695A1 (en) Her2 antibody-drug conjugates
US9950077B2 (en) Anti-folate receptor alpha (FRA) antibody-drug conjugates and methods of using thereof
WO2019149116A1 (zh) 制备偶联物的方法
US20160015831A1 (en) Anti-cd22 antibody-drug conjugates and methods of using thereof
CN116209679A (zh) 含有抗间皮素抗体的抗体-药物共轭物及其用途
US11207420B2 (en) Cytotoxin and conjugate, uses of same and preparation method therefor
CN111542324B (zh) 细胞毒性剂及其偶联物、其制备方法及用途
US20230330258A1 (en) Conjugates comprising a phosphorus (v) and a drug moiety

Legal Events

Date Code Title Description
AS Assignment

Owner name: SICHUAN KELUN-BIOTECH BIOPHARMACEUTICAL CO., LTD., CHINA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:CAI, JIAQIANG;SONG, SHUAI;XUE, TONGTONG;AND OTHERS;REEL/FRAME:062565/0919

Effective date: 20200220

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION