WO2021000067A1 - 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法 - Google Patents

一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法 Download PDF

Info

Publication number
WO2021000067A1
WO2021000067A1 PCT/CN2019/093946 CN2019093946W WO2021000067A1 WO 2021000067 A1 WO2021000067 A1 WO 2021000067A1 CN 2019093946 W CN2019093946 W CN 2019093946W WO 2021000067 A1 WO2021000067 A1 WO 2021000067A1
Authority
WO
WIPO (PCT)
Prior art keywords
compound
acid
group
reaction
structural formula
Prior art date
Application number
PCT/CN2019/093946
Other languages
English (en)
French (fr)
Inventor
赵珞博
杨庆良
赵林尧
黄圆圆
叶杭波
盖顺
赖娟
李雯君
白露
曹敏君
Original Assignee
杭州多禧生物科技有限公司
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 杭州多禧生物科技有限公司 filed Critical 杭州多禧生物科技有限公司
Priority to JP2022500117A priority Critical patent/JP2022540395A/ja
Priority to EP19936127.0A priority patent/EP3991752A4/en
Priority to IL289458A priority patent/IL289458A/en
Priority to AU2019452577A priority patent/AU2019452577B2/en
Priority to US17/623,360 priority patent/US20230025327A1/en
Priority to KR1020227003562A priority patent/KR20220029725A/ko
Priority to CA3145050A priority patent/CA3145050A1/en
Priority to CN201980095886.3A priority patent/CN114040778A/zh
Priority to BR112021025984A priority patent/BR112021025984A2/pt
Priority to PCT/CN2019/093946 priority patent/WO2021000067A1/zh
Priority to US17/596,956 priority patent/US20220249594A1/en
Priority to SG11202113022UA priority patent/SG11202113022UA/en
Priority to CA3142960A priority patent/CA3142960C/en
Priority to BR112021026009A priority patent/BR112021026009A2/pt
Priority to PCT/CN2020/075709 priority patent/WO2020258893A1/en
Priority to MX2021015878A priority patent/MX2021015878A/es
Priority to CN202080032694.0A priority patent/CN114040781A/zh
Priority to JP2021576898A priority patent/JP2022544442A/ja
Priority to KR1020227002437A priority patent/KR20220024915A/ko
Priority to EP20831886.5A priority patent/EP3986476A4/en
Priority to AU2020301115A priority patent/AU2020301115A1/en
Priority to TW109116404A priority patent/TWI785334B/zh
Publication of WO2021000067A1 publication Critical patent/WO2021000067A1/zh
Priority to TW110104872A priority patent/TW202144009A/zh
Priority to IL288954A priority patent/IL288954A/en
Priority to CL2021003468A priority patent/CL2021003468A1/es
Priority to ZA2022/01370A priority patent/ZA202201370B/en
Priority to JP2023203751A priority patent/JP2024028815A/ja

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N25/00Biocides, pest repellants or attractants, or plant growth regulators, characterised by their forms, or by their non-active ingredients or by their methods of application, e.g. seed treatment or sequential application; Substances for reducing the noxious effect of the active ingredients to organisms other than pests
    • A01N25/32Ingredients for reducing the noxious effect of the active substances to organisms other than pests, e.g. toxicity reducing compositions, self-destructing compositions
    • 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
    • A61K47/6811Drugs conjugated to an antibody or immunoglobulin, e.g. cisplatin-antibody conjugates the drug being a protein or peptide, e.g. transferrin or bleomycin
    • 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
    • A61K38/05Dipeptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/62Medicinal 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
    • A61K47/64Drug-peptide, drug-protein or drug-polyamino acid conjugates, i.e. the modifying agent being a peptide, protein or polyamino acid which is covalently bonded or complexed to a therapeutically active agent
    • A61K47/641Branched, dendritic or hypercomb peptides
    • 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
    • A61K47/6835Medicinal 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
    • A61K47/6863Medicinal 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 stomach or intestines 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
    • A61K47/6889Conjugates 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
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K39/00Medicinal preparations containing antigens or antibodies
    • A61K2039/505Medicinal preparations containing antigens or antibodies comprising antibodies
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides

Definitions

  • the present invention relates to coupling a Tubulysin derivative (homolog) with a cell-binding molecule with a branch-containing (side chain) linker, and the resulting conjugate has better pharmacokinetic properties, thereby enabling more precise targeted killing. Dead abnormal cells.
  • the present invention also relates to a method for synthesizing Tubulysin homologues and cell binding agents and the molecules contained therein, as well as methods for using the conjugate to target treatment of cancer, infection and autoimmune diseases.
  • Adcetris in the treatment of relapsed or refractory Hodgkin’s lymphoma (Okeley, N. et al. Hematol Oncol. Clin. North. Am, 2014, 28, 13-25; Gopal, A., et al., Blood 2015, 125, 1236-43) and the success of Kadcyla for the treatment of relapsed HER2-positive breast cancer (Peddi, P., Hurvitz, S., Ther. Adv. Med. Oncol. 2014, 6(5), 202-9; Lambert, J . And Chari, R., J. Med. Chem. 2014, 57, 6949-64) proved that antibody-drug conjugate (ADC) is a very promising cancer targeted therapy.
  • ADC antibody-drug conjugate
  • Important factors for ADC success include three important components, namely monoclonal antibodies, cytotoxic molecules and linkers, and linker-cytotoxic molecules linking sites (L. Ducry and B Stump, Bioconjugate Chem., 2010 ,21,5-13; GS Hamilton, Biologicals 2015,43,318-32).
  • the research on each component of ADC has a history of thirty years.
  • the linker needs to be able to react with the specific reactive functional groups on the drug, have stability in the human blood circulatory system, and be easy to release the drug after binding to the antigen and endocytosis.
  • the most important thing is that the linker-cytotoxic molecule once After being off-target in the blood circulation, normal tissues cannot be damaged, and the existing connection technology is still limited (Ponte, J.
  • T-DM1 A stable (uncleavable) MCC linker is used, which is of great benefit to patients who are HER2 positive with metastatic breast cancer (mBC) or who have received corresponding treatment or HER2 tumor recurrence within six months of adjuvant therapy (Peddi, P. and Hurvitz, S., Ther. Adv. Med. Oncol. 2014, 6(5), 202-209; Piwko C. et al., Clin Drug Investig. 2015, 35(8), 487-93; Lambert, J. and Chari, R., J. Med. Chem. 2014, 57, 6949-64).
  • mBC metastatic breast cancer
  • T-DM1 has failed as a first-line treatment for patients with HER2-positive unresectable locally advanced or metastatic breast cancer.
  • As a second-line treatment for HER2-positive advanced gastric cancer compared with its side effects, it has The benefit for patients is not great (Ellis, PA, et al., J.Clin.Oncol. 2015, 33 (2015 ASCO meeting abstract 507); Shen, K. et al., Sci Rep. 2016, 6, 23262; de Goeij, BE and Lambert, JMCurr Opin Immunol 2016, 40, 14-23; Barrios, CH et al., J Clin Oncol 2016, 34, (2016 ASCO Conference Abstract 593).
  • one direction of ADC chemistry research and development is to expand the linker-cytotoxic agent component and coupling chemistry, not only using a single cytotoxic agent, but also solving the ADC linker-cell
  • the problem of the activity of toxic agents against target diseases (Lambert, JM Ther Deliv 2016, 7, 279-82; Zhao, RY et al., 2011, J. Med. Chem. 54, 3606-23).
  • Many drug developers and academic institutions have concentrated their efforts to develop new and reliable specific coupling linkers, site-specific ADC coupling methods. It seems that these ADCs have a longer circulating half-life, higher efficacy, and can reduce off-target toxicity.
  • ADCs have narrow The in vivo pharmacokinetic (PK) characteristics of the pharmacokinetics and the consistency of the production process are better (Hamblett, KJ, etc. Clin. Cancer Res. 2004, 10, 7063-70; Adem, YT, etc., Bioconjugate Chem. 2014, 25, 656- 664; Boylan, NJ Bioconjugate Chem. 2013, 24, 1008-1016; Strop, P. et al., Chem. Biol. 2013, 20, 161-67; Wakankar, A. mAbs, 2011, 3, 161-172).
  • site-directed coupling methods that have been reported include: the introduction of modified cysteine into antibodies (Junutula, JR et al., Nat. Biotechnol.
  • Patent 8,871,908 or use microorganisms Glutamine tag introduced by transglutaminase (MTGase) (Dennler, P., et al., 2014, Bioconjug. Chem. 25, 569-78; Siegmund, V. et al. Angew. Chemie-Int. Ed. 2015, 54, 13420-4; U.S. Patent Application No. 20130189287; U.S. Patent 7,893,019), isopeptide bond-peptide bond is formed outside the protein backbone by enzyme or bacteria (Kang, HJ, et al. Science 2007,318,1625-8; Zakeri, B .Etc. Proc. Natl. Acad. Sci. USA 2012, 109, E690-7; Zakeri, B. & Howarth, MJ Am. Chem. Soc. 2010, 132, 4526-7).
  • MMGase transglutaminase
  • ADCs prepared with these linkers and coupling methods have a larger therapeutic window.
  • a Tubulysin conjugate containing a long chain linker can prevent the antibody-drug conjugate from being hydrolyzed by hydrolytic enzymes such as protease or esterase, making the conjugate more stable in the circulatory system.
  • Tubulysin is a class of highly effective cytotoxic agents, which is well known in the art and can be isolated from natural products based on known methods or prepared by organic synthesis methods (such as Balasubramanian, R., etc. J. Med. Chem. ,2009,52,238-40; Wipf,P.,et al. Org.Lett.,2004,6,4057-60; Pando,O.,et al. J.Am.Chem.Soc.,2011,133,7692-5 ; Reddy, JA, etc. Mol. Pharmaceutics, 2009, 6, 1518-25; Raghavan, B., etc. J. Med. Chem., 2008, 51, 1530-33; Patterson, AW, etc. J.
  • WO2010033733 WO 2009002993; Ellman, J., etc., PCT WO200 9134279; WO 2009012958; US Patent Application 20110263650, 20110021568; Matschiner, G., etc., WO2009095447; Vlahov, I., etc., WO2009055562, WO 2008112873; Low, P., etc., WO2009026177; Richter, W., WO2008138561; Kjems , J., et al., WO 2008125116; Davis, M.; et al., WO2008076333; Diener, J.; et al., U.S.
  • Tubulysin conjugate (PCT/IB2012/053554) for targeted treatment of cancer, infection and autoimmune diseases.
  • the Tubulysin conjugate containing a long-chain branched linker in the present invention has an increased half-life during targeted delivery, and minimizes exposure to non-target cells, tissues or organs in the blood circulation, resulting in reduced off-target toxicity.
  • the invention relates to coupling a Tubulysin homologue and a cell binding molecule with a branched chain (side chain) linker, and the produced conjugate has better pharmacokinetic properties, thereby being able to target and kill abnormal cells more accurately.
  • the present invention also relates to the coupling of Tubulysin homologues and cell binding agents and the methods for synthesizing the molecules contained therein, as well as methods for using the conjugates to target treatment of cancer, infection and autoimmune diseases.
  • the present invention relates to an antibody-Tubulysin B derivative conjugate, characterized in that the conjugate has the structure shown in the following formula (I):
  • P 1 is H, COCH 3 , COH, PO(OH) 2 , CH 2 OPO(OH) 2 , SO 2 CH 3 , C 6 H 11 O 5 (glycoside), CONHCH 3 , CON(CH 3 ) 2 , CON(CH 2 CH 2 ) 2 NCH 3 , CON(CH 2 CH 3 ) 2 or CON(CH 2 CH 2 ) 2 CHN(CH 2 CH 2 ) 2 CH 2 ;
  • R 1 , R 2 , R 3 and R 4 are each independently H, C 1 -C 6 alkane group, C 1 -C 6 alkenyl group, C 1 -C 6 alkane ether group (R 1 OR 2 ), C 1 -C 6 alkyl carbonyl group (R 1 COR 2 ), C 1 -C 6 alkyl ester group (R 1 COOR 2 ), C 1 -C 6 alkyl carboxyl group ((R 1 COOH) or C 1 -C 6 alkyl amide group ( (R 1 CONHR 2 );
  • R 5 is H, OC 1 -C 6 alkane group, C(O)-H, C(O)-C 1 -C 6 (straight or branched chain) alkane group, C(O)-NH-C 1- C 6 (straight or branched chain) alkyl or C(O)-N(C 1 -C 6 (straight or branched) alkyl) 2 ;
  • R 6 , R 7 and R 8 are each independently H, C 1 -C 6 alkane group, C 1 -C 6 alkane ether group (R 1 OR 2 ), C 1 -C 6 alkane carbonyl group (R 1 COR 2 ) , C 1 -C 6 alkyl ester group (R 1 COOR 2 ), C 1 -C 6 alkyl carboxyl group ((R 1 COOH) or C 1 -C 6 alkyl amide group ((R 1 CONHR 2 )); preferably R 6 , R 7 and R 8 are each independently H or CH 3 ;
  • mAb is an antibody, antibody fragment, monoclonal antibody, polyclonal antibody, nanobody, prodrug antibody (probody), or antibody and antibody fragment modified with a synthetic molecule or protein;
  • L is a linker containing a hydrophilic branch, and its main framework is C 2 -C 100 peptide units (1-12 natural or unnatural amino acids), hydrazone bond groups, disulfide groups, ester groups, An oxime group, an amide group or a thioether bond group.
  • the structure of L is:
  • Aa is L- or D-natural or unnatural amino acid
  • r is an integer between 0-12; when r is not 0, (Aa) r is a peptide unit composed of the same or different amino acids;
  • R 1 , m 2 and (Aa) r are as described in claim 1 and the above definition.
  • the cell surface receptor binding molecule mAb can be any form of cell conjugate, including peptides or peptide-like structures: antibodies, single-chain antibodies, antibody fragments that can bind to target cells, monoclonal antibodies, single-chain single-chain Cloned antibodies, monoclonal antibody fragments that can bind to target cells, chimeric antibodies, chimeric antibody fragments that can bind to target cells, functional domain antibodies, functional domain antibody fragments that can bind to target cells, genetically engineered proteins that mimic antibodies , Fibronectin conjugate adnectin, pre-designed ankyrin repeat protein (DARPin), lymphokines, hormones, vitamins, growth factors, colony stimulating factors, nutrient transport molecules, transferrin, cell surface small molecule ligands, or connected with Albumin, macromolecules, or dendrimers of cell conjugates, macromolecular materials, proteins, liposomes, nanoparticles, vesicles containing cell-binding molecules (binding peptides, proteins, antibodies, antibodies
  • L’ is (II-0) and (II-00):
  • the process of preparing the above-mentioned conjugate is characterized in that the preparation method of the coupled mAb-SH includes any one of the following a) to c):
  • reducing agent preferably, tris (2-carboxyethyl) phosphine (TCEP), dithiothreitol (DTT), dithiopentaerythritol (DTE), L-glutathione (GSH), 2- Mercaptoethylamine ( ⁇ -MEA) or/and ⁇ -mercaptoethanol ( ⁇ -ME, 2-ME)
  • TCEP tris (2-carboxyethyl) phosphine
  • DTT dithiothreitol
  • DTE dithiopentaerythritol
  • GSH L-glutathione
  • 2- Mercaptoethylamine ⁇ -MEA
  • ⁇ -ME, 2-ME 2-mercaptoethanol
  • the process of preparing the above-mentioned conjugate is characterized in that the buffer system used in the synthesis of the conjugate is: pH 5.0-9.5, and the concentration of phosphoric acid, acetic acid, lemon Buffer solution of acid, boric acid, carbonic acid, barbituric acid, Tris (tris(hydroxymethylaminomethane)), benzoic acid or triethanolamine, or their mixture, and contains a water-soluble organic solvent with a volume ratio of 0% to 35%: Methanol, ethanol, n-propanol, isopropanol, n-butanol, isobutanol, acetonitrile, acetone, DMF, DMA or DMSO, the coupling reaction temperature is controlled at 0°C ⁇ 45°C, the coupling reaction is 5 minutes ⁇ 96 hour.
  • the buffer system used in the synthesis of the conjugate is: pH 5.0-9.5, and the concentration of phosphoric acid, acetic acid, lemon Buffer solution of acid, boric acid
  • the process of preparing the above-mentioned conjugate is characterized in that after the coupling reaction is completed, ultrafiltration or column chromatography is used for purification to obtain the conjugate of structural formula (I).
  • column chromatography includes molecular sieve column, cation column, anion column, hydrophobic (HIC) column, reverse phase column or protein A or G affinity column.
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of the structural formula (II) is condensed by the Tubulysin B derivative of the structural formula (III) and the compound of the structural formula (L') Response obtained:
  • X is OH, halogen (F, Cl, Br, or I), phenol, pentachlorophenol, trifluoromethanesulfonic acid, imidazole, dichlorophenol, tetrachlorophenol, 1-hydroxybenzotriazole, p- Toluenesulfonic acid, methanesulfonic acid, 2-ethyl-5-phenylisoxazole-3'-sulfonic acid, Self-acid anhydrides or acid anhydrides formed with other acid anhydrides such as acetyl anhydride and formic anhydride; or polypeptide condensation reaction intermediates or Mitsunobu reaction intermediates;
  • the condensation reaction is carried out in the organic solvent methylene chloride, dichloroethane, DMF, DMA, tetrahydrofuran (THF), DMSO containing 1%-100% by volume of pyridine, triethylamine or diisopropylethylamine. , Acetone, isopropanol, n-butanol or acetonitrile, or a mixed solvent of two or more of the above solvents, or with or without inert gas (nitrogen, argon, helium) protection, the temperature is controlled at -20 Under the condition of °C-150°C, the reaction time is 5 minutes-120 hours to complete;
  • the condensation reaction is carried out under the following buffer system and the following conditions.
  • the buffer system is: phosphoric acid, acetic acid, citric acid, boric acid, carbonic acid, barbituric acid, Tris (three) with a pH of 5.0 to 9.5 and a concentration of 1 mM to 1000 mM.
  • Hydroxymethylaminomethane benzoic acid or triethanolamine, or their mixture buffer solution, and containing 0% to 35% by volume of water-soluble organic solvents: methanol, ethanol, n-propanol, isopropanol, n-butyl Alcohol, isobutanol, acetonitrile, acetone, DMF, DMA or DMSO, the coupling reaction temperature is controlled at 0°C to 45°C, and the coupling reaction is 5 minutes to 96 hours.
  • the NH 2 group in the structural formula (III) is ideally based on trifluoroacetate, hydrochloride, formate, acetate, sulfate, phosphate, nitrate, citrate, succinic acid Condensation occurs in the form of salt, benzoic acid, and sulfonate.
  • the condensation reagent can be alternatively: 1-(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride (EDC), two Cyclohexylcarbodiimide (DCC), N,N'-diisopropylcarbodiimide (DIC), 1-cyclohexyl-2-morpholinoethylcarbodiimide p-toluenesulfonate ( CMC or CME-CDI), carbonyl diimidazole (CDI), O-benzotriazole-N,N,N',N'-tetramethylurea tetrafluoroborate (TBTU), O-benzotriazole -Tetramethylurea hexafluorophosphate (HBTU), benzotriazol-1-yloxytris(dimethylamino)phosphonium hexafluorophosphate (EDC), two Cyclohexylcarbodiimide (DCC), N,N
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of Tubulysin B derivative of structural formula (III) includes one or more of the following steps:
  • R 5 ′ is H, C 1 -C 6 alkane group, C 1 -C 6 alkane group, or C 1 -C 6 (linear or branched) aminoalkyl group; the definitions of other groups are as described above.
  • Tubulysin B derivative of structural formula (III) includes one or more of the following steps:
  • Step 1 Stir the aqueous solution of diethoxyacetonitrile and ammonium sulfide at room temperature to obtain compound 1, namely 2,2-diethoxythioacetamide;
  • Step 2 Heat compound 1 and bromopyruvate in anhydrous solvent (such as anhydrous tetrahydrofuran, dichloromethane, acetonitrile, N,N-dimethylformamide, methanol, isopropanol) to condense compound 2 ;
  • anhydrous solvent such as anhydrous tetrahydrofuran, dichloromethane, acetonitrile, N,N-dimethylformamide, methanol, isopropanol
  • Step 3 Dissolve compound 2 in a solvent (such as tetrahydrofuran, dichloromethane, ethyl acetate, n-heptane, dioxane, acetonitrile), and use Lewis acid or protic acid (including hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonate) Acid, formic acid, oxalic acid, acetic acid, p-toluenesulfonic acid, pyridine p-toluenesulfonate, AlCl 3 , FeCl 3 , ZnCl 2 , BF 3 , BCl 3 , BBr 3 , TiCl 4 , ZnBr 2 , LiBF 4 ) hydrolyze to obtain the compound 3;
  • a solvent such as tetrahydrofuran, dichloromethane, ethyl acetate, n-heptane, dioxane, acetonit
  • Step 4 Unsaturated sulfinamide is dehydrogenated by n-butyl lithium and other bases under low temperature conditions (such as -45°C to -78°C), and then undergoes addition reaction with compound 3 in the presence of Lewis acid to obtain compound 4;
  • Lewis acids are selected to include hydrochloric acid, sulfuric acid, phosphoric acid, methanesulfonic acid, formic acid, oxalic acid, acetic acid, p-toluenesulfonic acid, pyridine p-toluenesulfonate, AlCl 3 , FeCl 3 , ZnCl 2 , BF 3 , BCl 3 , BBr 3 , TiCl 4 , ZnBr 2 , LiBF 4 ;
  • Step 5 Compound 4 is selectively reduced by reducing agents (such as NaBH 4 , LiBH 4 , Na(OAc) 3 BH, Na(CN)BH 3, etc.) under low temperature conditions (such as -45°C to -78°C) , By adding Lewis acid (such as Ti(Oet)4) to control its stereochemistry to obtain compound 5;
  • reducing agents such as NaBH 4 , LiBH 4 , Na(OAc) 3 BH, Na(CN)BH 3, etc.
  • Lewis acid such as Ti(Oet)4
  • Step 6 Dissolve compound 5 in a solvent (such as methanol, ethanol, isopropanol, tetrahydrofuran, acetonitrile), and remove the tert-butylsulfinyl group with acids such as hydrochloric acid, sulfuric acid, and phosphoric acid to obtain compound 6;
  • a solvent such as methanol, ethanol, isopropanol, tetrahydrofuran, acetonitrile
  • Step 7 Dissolve compound 6 and azido acid in a solvent (such as n-heptane, tetrahydrofuran, dichloromethane, N,N-dimethylformamide), and then use a condensation reagent (such as DIC/HOBt, DCC/HOBt, In the presence of EDC/HOBt, HATU, BOP, T3P, BrOP), or through a condensation reaction route, a condensation reaction occurs to obtain compound 7;
  • a solvent such as n-heptane, tetrahydrofuran, dichloromethane, N,N-dimethylformamide
  • a condensation reagent such as DIC/HOBt, DCC/HOBt, In the presence of EDC/HOBt, HATU, BOP, T3P, BrOP
  • hydrazoic acid and isobutyl chloroformate are reacted in THF in the presence of an organic base (such as triethylamine, diisopropylethylamine, N-methylmorpholine, etc.) to obtain a mixed anhydride, and then compound 6
  • an organic base such as triethylamine, diisopropylethylamine, N-methylmorpholine, etc.
  • azido acid is reacted with oxalyl chloride, triethylamine and a catalyst amount of DMF in a solvent (such as n-heptane, n-hexane, dichloromethane, tetrahydrofuran) to be converted into acid chloride, and then compound 6 hydrochloride Condensation occurs to obtain compound 7;
  • a solvent such as n-heptane, n-hexane, dichloromethane, tetrahydrofuran
  • Step 8 In a solvent (such as dichloromethane, tetrahydrofuran, acetonitrile), the hydroxyl group on compound 7 and a hydroxyl protecting reagent (such as TESCl), under the action of an organic base (such as imidazole, triethylamine, pyridine) to obtain compound 8 ;
  • a solvent such as dichloromethane, tetrahydrofuran, acetonitrile
  • an organic base such as imidazole, triethylamine, pyridine
  • Step 9 Compound 8 is dissolved in a solvent (such as tetrahydrofuran, dichloromethane, acetonitrile), under the action of a base (such as KHMDS, LiHMDS, NaHMDS, KOtBu, NaH, KH), the amide is deprotonated, and then combined with iodine Methane, methyl bromide, dimethyl sulfate, methyl trifluoromethanesulfonate, or ethyl iodide undergoes alkylation reactions to obtain compound 9;
  • a solvent such as tetrahydrofuran, dichloromethane, acetonitrile
  • a base such as KHMDS, LiHMDS, NaHMDS, KOtBu, NaH, KH
  • Step 10 Compound 9 is dissolved in a solvent (such as tetrahydrofuran, dichloromethane, ethyl acetate), where the azide group is under certain conditions, such as hydrogen and palladium-carbon catalyst, triphenylphosphine and water (Staudinger In the presence of reaction), it is reduced to an amino group, and then condensed with an acid or a reactive acid derivative to obtain compound 10;
  • a solvent such as tetrahydrofuran, dichloromethane, ethyl acetate
  • the azide group is under certain conditions, such as hydrogen and palladium-carbon catalyst, triphenylphosphine and water (Staudinger In the presence of reaction)
  • Step 11 The hydroxyl protecting group PG 1 in compound 10 is deprotected under appropriate conditions (for example, the TES protecting group can be deprotected in hydrochloric acid, THF/MeOH/AcOH, nBu 4 NF or pyridine hydrofluoride in THF) Is deprotected to obtain compound 11;
  • Step 12 The ester group in compound 11 is converted into carboxylic acid under the action of alkali (such as LiOH, NaOH, KOH) or other appropriate conditions (such as methyl ester can be converted into carboxylic acid under the action of reagents such as LiCl, LiI, Me3SiOK) Is converted into acid compound 12;
  • alkali such as LiOH, NaOH, KOH
  • reagents such as LiCl, LiI, Me3SiOK
  • Step 13 In the presence of a base (such as triethylamine, N,N-diisopropylethylamine, pyridine) and a catalyst (such as DMAP), under certain temperature conditions (such as 0°C to 23°C), compound 12 and Acetic anhydride, propionic anhydride, isopropionic anhydride and other acid anhydrides, acetyl halide, propionic acid halide, propionic acid halide, formamide halide, isoamide halide, diformamide halide and other acid halides can be reacted to obtain compound 13.
  • a base such as triethylamine, N,N-diisopropylethylamine, pyridine
  • a catalyst such as DMAP
  • Step 14 Compound 13 will undergo condensation reaction with appropriate hydroxyl-containing compounds such as pentafluorophenol or N-hydroxysuccinimide in the presence of condensation reagents (such as EDC, DIC, DCC, HATU, HBTU) to obtain reactive The ester compound 14;
  • condensation reagents such as EDC, DIC, DCC, HATU, HBTU
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 16 The nitro group in compound 16 is reduced to an amino group under reducing conditions such as hydrogen and palladium-carbon catalyst, hydrazine hydrate and FeCl 3 , iron powder and acetic acid, etc., to obtain compound III;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of the compound of the structural formula (L') includes one or more of the following steps:
  • Step 1 Compound 1-1 and compound 1-2 are condensed under the action of a condensing agent (such as EDC, HATU, DIC, DCC), or a condensation reaction occurs through a condensation reaction route (such as compound 1-2 in a condensation reagent such as DIC and Under the action of EDC, it undergoes condensation reaction with pentafluorophenol, nitrophenol or N-hydroxysuccinimide to produce the corresponding active ester, and then reacts with compound 1-1) to obtain compound 1a;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • a condensation reaction route such as compound 1-2 in a condensation reagent such as DIC and Under the action of EDC, it undergoes condensation reaction with pentafluorophenol, nitrophenol or N-hydroxysuccinimide to produce the corresponding active ester, and then reacts with compound 1-1) to obtain compound 1a;
  • compound 1-3 and compound 1-4 undergo condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 1b;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 2 The carboxyl protecting group PG 2 in compound 1 is removed under the action of a deprotection reagent (for example, the tert-butyl ester group under the action of acid), to obtain compound 2;
  • a deprotection reagent for example, the tert-butyl ester group under the action of acid
  • Step 3 The carboxyl-containing compound 2 and the amino-containing compound 3 undergo a condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 4;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 4 The amino protecting group PG 1 on compound 4 is removed under deprotection conditions.
  • the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst, and the Boc protecting group on the amino can be acidic Was removed under conditions to obtain compound 5;
  • Step 5 Carboxyl-containing compound 6 and amino-containing compound 5 undergo a condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 7;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 6 The carboxyl protecting group PG 3 on compound 7 is removed under deprotection conditions (for example, the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc. ) To obtain compound 8;
  • Step 7 Compound 8 and hydroxyl-containing compounds (such as pentafluorophenol or N-hydroxysuccinimide) in the presence of condensation reagents (such as EDC, HATU, DIC, DCC), condensation reaction occurs to obtain reactive esters Compound, or react with other acid activating groups to obtain compound L'with condensation reaction activity;
  • condensation reagents such as EDC, HATU, DIC, DCC
  • Step 1 The amino protecting group PG 1 on compound 1 is removed under deprotection conditions.
  • the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst, and the Boc protecting group on the amino can be acidic It is excised under conditions to obtain compound 2;
  • Step 2 Amino-containing compound 2 and carboxyl-containing compound 3 undergo condensation reaction under the action of a condensation reagent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 4;
  • a condensation reagent such as EDC, HATU, DIC, DCC
  • Step 3 The carboxyl protecting group PG 2 on compound 4 is removed under deprotection conditions.
  • the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc. , To obtain compound 5;
  • Step 4 The carboxyl-containing compound 5 and the amino-containing compound 6 undergo a condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 7;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 5 The carboxyl protecting group PG 3 on compound 7 is removed under deprotection conditions.
  • the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc. , To obtain compound 8;
  • Step 6 Compound 8 and hydroxyl-containing compounds (such as pentafluorophenol or N-hydroxysuccinimide) undergo condensation reaction in the presence of a condensation reagent to obtain a reactive ester compound, or react with other acid-activated groups Obtain compound 9 with condensation reaction activity;
  • the NH 2 group in structural formula (V) is preferably trifluoroacetate, hydrochloride, formate, acetate, sulfate, phosphate, nitrate, citrate, succinate, Condensation occurs in the form of benzoic acid or sulfonate.
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (IV) includes one or more of the following steps:
  • Carboxylic acid compound 1 and hydroxyl-containing compounds such as pentafluorophenol or N-hydroxysuccinimide
  • condensation reagents such as EDC, DIC, DCC, HATU, HBTU
  • the carboxylic acid compound 1 is reacted with ethyl chloroformate, isobutyl chloroformate, etc. in the presence of an organic base (such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases) to obtain Reactive mixed acid anhydrides;
  • an organic base such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases
  • the carboxylic acid compound 1 is reacted with oxalyl chloride in the presence of an organic base such as triethylamine and a catalyst amount (for example, 0.01 equivalent to 0.5 equivalent) of DMF to obtain acid chloride.
  • an organic base such as triethylamine
  • a catalyst amount for example, 0.01 equivalent to 0.5 equivalent
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (V) includes one or more of the following steps:
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 2 The amino protecting group PG 4 on compound 3 is removed under deprotection conditions (for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst), and the Boc protecting group on the amino can be removed under deprotection conditions. It is excised under acidic conditions to obtain compound V;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of compound 2 includes one or more of the following steps:
  • compound 8 (compound XIVa) obtained in this synthesis step is the target compound 2; PG 4 is an amino protecting group.
  • the synthesis of compound 2 includes one or more of the following steps:
  • Step 1 Dissolve L-tyrosine ester derivatives 1 in a suitable solvent, such as acetone, tetrahydrofuran, acetonitrile, dichloromethane, etc., or a mixed solvent of these solvents and water, and benzyl chloride or benzyl bromide Or other benzyl compounds to react at 0 to 60°C, appropriate organic or inorganic bases can be added to the reaction system, such as sodium carbonate, potassium carbonate, sodium bicarbonate, potassium bicarbonate, sodium hydroxide, potassium hydroxide, three Ethylamine, DBU, sodium hydride, etc., and appropriate additives such as sodium iodide or phase transfer catalysts, such as benzyltriethylammonium chloride (TEBA), tetrabutylammonium bromide (TBAB), Tetrabutylammonium chloride, tetrabutylammonium hydrogen sulfate, trioctylmethylammonium chloride
  • Step 2 Dissolve compound 2 in an organic solvent such as dichloromethane, tetrahydrofuran, methanol, ethanol, ether, etc., using a reducing agent, such as lithium aluminum hydride, DIBAL, sodium borohydride, lithium borohydride, dihydrobis(2-methyl) Oxyethoxy) sodium aluminate (Red-Al), diborane, etc. reduction, additives can be added to the reaction system, such as I 2 , ferric chloride, zinc chloride, magnesium chloride, lithium chloride, calcium chloride Control the activity of the reducing agent and obtain compound 3;
  • a reducing agent such as lithium aluminum hydride, DIBAL, sodium borohydride, lithium borohydride, dihydrobis(2-methyl) Oxyethoxy) sodium aluminate (Red-Al), diborane, etc. reduction
  • additives can be added to the reaction system, such as I 2 , ferric chloride, zinc chloride, magnesium chloride,
  • Alcohol compound 3 is oxidized into aldehyde under appropriate oxidation conditions, such as swern oxidation (oxalyl chloride, DMSO, triethylamine), Parikh-Doering oxidation (sulfur trioxide pyridine oxidation), Dess-Martin oxidation, etc. 4;
  • Step 4 Reaction of aldehyde 4 with phosphate (Horner-Wadsworth-Emmons reaction) or phosphorous ylide reaction (Wittig reaction) for carbon chain extension to obtain compound 5;
  • Step 5 The double bond in compound 5 is hydrogenated and reduced under the action of a homogeneous or two-phase catalyst, and the benzyl group is also removed at the same time to obtain a chiral compound with a single stereo structure, or two diastereomers
  • the catalyst includes Pd/C, Pd(OH) 2 /C, Pd/BaSO 4 , PtO 2 , Pt/Al 2 O 3 , Ru/C, Raney nickel and other two-phase catalysts, homogeneous asymmetric hydrogenation catalyst , Such as Crabtree catalyst, [Ru(II)-(BINAP)] catalyst, [(Ph3P)CuH] 6 catalyst, etc.;
  • Step 6 Dissolve compound 6 in an organic solvent, such as tetrahydrofuran, acetonitrile, dichloromethane, and be nitrated under nitration reaction conditions.
  • organic solvent such as tetrahydrofuran, acetonitrile, dichloromethane
  • Nitrating reagents include nitric acid, nitric acid/acetic acid, potassium nitrate/sulfuric acid, tert-butyl nitroso, Nitric acid/trifluoroacetic anhydride, NO 2 BF 4 , nitropyridinium salt, etc.;
  • Step 7. 7-nitro compound is reduced to an amino group under the following conditions, these conditions include H 2 / Pd / C, Fe or Zn / HOAc, SnCl 2 / HCl .
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of compound 2 includes one or more of the following steps:
  • the compound 8 (compound XIVb) obtained in this synthesis step is the target compound 2.
  • the synthesis of compound 2 includes one or more of the following steps:
  • Step 2 The hydroxyl group on compound 3 is removed under the following conditions. These conditions include the Barton-McCombie deoxygenation reaction, that is, the alcohol is first converted into thioacyl derivatives, such as alkyl xanthates, benzene thiochloroformate Ester, thiocarbonyl imidazole ester, and then treated with Bu 3 SnH, free radical cleavage occurs to obtain the dehydroxylated product; the conditions for free radical cleavage include: n-Bu 3 SnH/AIBN, n-Bu 3 SnH/AIBN/n- BuOH/PMHS, (Bu 4 N) 2 S 2 O 8 /HCO 2 Na;
  • Step 3 Dissolve compound 4 in tetrahydrofuran, the Evans chiral prosthetic group is cut under the condition of LiOH/H 2 O 2 to obtain the corresponding acid 5;
  • Step 4 Compound 5 is dissolved in an organic solvent, such as ethyl acetate, methanol, dichloromethane, ethanol or acetic acid, etc., and the benzyl group is catalytically hydrogenolyzed in the presence of a palladium-carbon catalyst to obtain compound 6;
  • an organic solvent such as ethyl acetate, methanol, dichloromethane, ethanol or acetic acid, etc.
  • Step 5 Dissolve compound 6 in an organic solvent, such as tetrahydrofuran, acetonitrile, dichloromethane, and nitrate under nitration reaction conditions.
  • organic solvent such as tetrahydrofuran, acetonitrile, dichloromethane, and nitrate under nitration reaction conditions.
  • Nitrating reagents include nitric acid, nitric acid/acetic acid, potassium nitrate/sulfuric acid, tert-butyl nitroso, Nitric acid/trifluoroacetic anhydride, NO 2 BF 4 , nitropyridinium salt, etc.;
  • Step 6 The nitro group in compound 7 is reduced to an amino group under the following conditions. These conditions include H 2 /Pd/C, Fe or Zn/HOAc, SnCl 2 /HCl, etc., to obtain a chiral compound 8 with a single stereoconfiguration .
  • the synthesis of structural formula (VI) includes one or more of the following steps:
  • Step 1 Compound 1 and an appropriate hydroxyl-containing compound (such as pentafluorophenol or N-hydroxysuccinimide) undergo a condensation reaction in the presence of a condensation reagent to obtain a reactive acid derivative compound 2;
  • an appropriate hydroxyl-containing compound such as pentafluorophenol or N-hydroxysuccinimide
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 3 The amino protecting group PG 4 on compound 4 is selectively removed under deprotection conditions (for example, the Cbz protecting group on the amino group can be cleaved off under the action of hydrogen and palladium-carbon catalyst), and the Boc on the amino group is protected The group can be cleaved under acidic conditions to obtain compound 5;
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as When Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3 ), a condensation reaction occurs to obtain compound 6.
  • no base is used in the reaction, but appropriate reaction temperature and reaction time need to be controlled;
  • Step 5 The amino protecting group PG 1 on compound 6 is removed under deprotection conditions (for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst), and the Boc protecting group on the amino can be It is excised under acidic conditions to obtain compound VI;
  • deprotection conditions for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst
  • Boc protecting group on the amino can be It is excised under acidic conditions to obtain compound VI;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (VII) includes one or more of the following steps:
  • Step 1 Carboxylic acid compound 1 and hydroxyl-containing compounds (such as pentafluorophenol or N-hydroxysuccinimide) undergo condensation reaction in the presence of condensation reagents (such as EDC, HATU, DIC, DCC) to obtain reactive ⁇ Esters;
  • condensation reagents such as EDC, HATU, DIC, DCC
  • the carboxylic acid compound 1 is reacted with ethyl chloroformate, isobutyl chloroformate, etc. in the presence of an organic base (such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases) to obtain Reactive mixed acid anhydrides;
  • an organic base such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases
  • carboxylic acid compound 1 and oxalyl chloride are reacted to obtain acid chloride in the presence of an organic base such as triethylamine and a catalyst amount (such as 0.01 equivalent to 0.5 equivalent) of DMF;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (II) is obtained by the condensation reaction of structural formula (VIII) and structural formula (IX):
  • the definition of X and the condensation reaction conditions are as described above.
  • the NH 2 group in structural formula (VIII) is ideally based on trifluoroacetate, hydrochloride, formate, acetate, sulfate, phosphate, nitrate, citrate, succinate, Condensation occurs in the form of benzoic acid or sulfonate.
  • Step 1 The carboxyl protecting group PG 3 on compound 1 is removed under deprotection conditions (for example, the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc.) , To obtain compound 2;
  • deprotection conditions for example, the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc.
  • Step 2 Compound 2 and hydroxyl-containing compounds (such as pentafluorophenol or N-hydroxysuccinimide) in the presence of condensation reagents (such as EDC, HATU, DIC, DCC), condensation reaction occurs to obtain reactive esters Compound 3;
  • condensation reagents such as EDC, HATU, DIC, DCC
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 4 The amino protecting group PG 3 on compound 5 is removed under deprotection conditions (for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst), and the Boc protecting group on the amino can be It is excised under acidic conditions to obtain compound 6;
  • deprotection conditions for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst
  • Boc protecting group on the amino can be It is excised under acidic conditions to obtain compound 6;
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as When Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3 ), a condensation reaction occurs to obtain compound 7; optionally, no base is used in the reaction, but the appropriate reaction temperature and reaction time need to be controlled;
  • Step 6 The amino protecting group PG 1 on compound 7 is removed under deprotection conditions.
  • the Cbz protecting group on the amino can be removed under the action of hydrogen and palladium-carbon catalyst, and the Boc protecting group on the amino can be acidic Was removed under conditions to obtain compound VIII;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (IX) includes one or more of the following steps:
  • Carboxylic acid compound 1 and an appropriate hydroxyl-containing compound undergo a condensation reaction in the presence of a condensation reagent to obtain a reactive ester IX;
  • the carboxylic acid compound 1 is reacted with ethyl chloroformate, isobutyl chloroformate, etc. in the presence of an organic base (such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases) to obtain Reactive mixed acid anhydride IX;
  • an organic base such as N-methylmorpholine, triethylamine, diisopropylethylamine and other organic bases
  • carboxylic acid compound 1 and oxalyl chloride are reacted to obtain acid chloride IX in the presence of an organic base such as triethylamine and a catalyst amount of DMF;
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (II) is obtained by the condensation reaction of structural formula (X) and structural formula (XI):
  • Y 1 and Y 2 groups condense to form Y group; Y 1 and Y 2 are NH 2 ,- + NH 3 , COOH, COX, SO 2 Cl, P(O)Cl 2 , NHCOX, NHSO 2 Cl, NHP(O)Cl 2 , NHP(O)(OH)Cl,
  • Step 1 The carboxyl-containing compound 1 and compound VI undergo a condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 2; wherein Z 1 is the precursor of Y 1 , Such as amino group, carboxyl group, amide group, phosphoramido group and sulfonamide group, carboxylate, phosphate, phosphonate, etc. protected by appropriate groups;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 2 The amino protecting group PG 1 on compound 2 is removed under deprotection conditions.
  • the Cbz protecting group on the amino group can be removed under the action of hydrogen and palladium-carbon catalyst, and the Boc protecting group on the amino group can be acidic Was removed under conditions to obtain compound 3;
  • Step 3 The carboxyl-containing compound 4 and the amino-containing compound 3 undergo a condensation reaction under the action of a condensing agent or through a condensation reaction route to obtain compound 5;
  • Step 4 The functional group Z 1 in compound 5 undergoes appropriate chemical transformation, such as deprotection of carboxyl and amino groups, to generate functional group Y 1 to obtain compound X;
  • the process of preparing the conjugate described above is characterized in that the synthesis of structural formula (XI) includes one or more of the following steps:
  • Step 1 Dissolve compound 1 in an organic solvent, such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide, and extract hydrogen with a base such as sodium hydride, sodium, and sodium hydroxide. Then it is stirred with compound 2 (where X is a halogen such as chlorine, bromine, iodine, or other leaving groups) at a certain temperature to react to obtain compound 3;
  • organic solvent such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide
  • a base such as sodium hydride, sodium, and sodium hydroxide.
  • Step 2 The carboxyl protecting group PG1 on compound 3 is removed under deprotection conditions.
  • the tert-butyl ester protecting group can be cleaved under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc., to obtain compound XIa -1;
  • Step 3 Dissolve compound 1 in an organic solvent, such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide, and extract hydrogen with a base such as sodium hydride, sodium, sodium hydroxide, etc. Then with compound 4 (stir at a certain temperature, react to obtain compound 5;
  • organic solvent such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide
  • Step 4 The carboxyl protecting group PG 1 on compound 5 is removed under deprotection conditions.
  • the tert-butyl ester protecting group can be cleaved under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc., to obtain a compound XIa-2;
  • Step 5 Dissolve compound 6 in an organic solvent, such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethylsulfoxide, etc., and add a suitable organic base, such as triethylamine, N,N- Diisopropylethylamine, pyridine, etc. are reacted with methylsulfonyl chloride, 4-toluenesulfonyl chloride, etc. at 0-5°C to obtain compound 7;
  • organic solvent such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethylsulfoxide, etc.
  • a suitable organic base such as triethylamine, N,N- Diisopropylethylamine, pyridine, etc. are reacted with methylsulfonyl chloride, 4-toluenesulfonyl chloride, etc. at 0-5°C to obtain compound 7;
  • Step 6 Compound 7 reacts with ammonia in an aqueous phase or an organic solvent, such as methanol, ethanol, acetonitrile, tetrahydrofuran, epoxy hexacyclic ring, etc., and the reaction can be appropriately heated to obtain compound XIb.
  • an organic solvent such as methanol, ethanol, acetonitrile, tetrahydrofuran, epoxy hexacyclic ring, etc.
  • Step 7 Compound 7 is reacted with sodium azide in an organic solvent, such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide, etc., to obtain compound 8;
  • organic solvent such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethyl sulfoxide, etc.
  • Step 8 The azide compound 8 is reduced by hydrogenation in the presence of a palladium-carbon catalyst, or reduced under the action of triphenylphosphine and water to obtain compound XIb;
  • Step 9 Compound 7 and dibenzylamine in an organic solvent, such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethylsulfoxide, etc., preferably N,N-dimethylformamide, at 100 Reaction occurs at °C to obtain compound 9;
  • organic solvent such as tetrahydrofuran, dichloromethane, N,N-dimethylformamide, dimethylsulfoxide, etc., preferably N,N-dimethylformamide, at 100 Reaction occurs at °C to obtain compound 9;
  • Step 10 Dissolve compound 9 in a solvent, such as ethyl acetate, methanol, ethanol, acetic acid, tetrahydrofuran, etc., and reduce it on a palladium-carbon catalyst under a certain hydrogen pressure.
  • a solvent such as ethyl acetate, methanol, ethanol, acetic acid, tetrahydrofuran, etc.
  • the reaction can be appropriately heated to 45°C to obtain the compound XIb.
  • the NH 2 group in the structural formula (XII) is trifluoroacetate, hydrochloride, formate, acetate, sulfate, phosphate, nitrate, citrate, succinic acid
  • the condensation reaction takes place in the form of salt, benzoic acid or sulfonate.
  • the process of preparing the above-mentioned conjugate is characterized in that the synthesis of structural formula (XII) includes one or more of the following steps:
  • Step 1 Compound 1 and an appropriate hydroxyl-containing compound (such as pentafluorophenol or N-hydroxysuccinimide) undergo a condensation reaction in the presence of a condensation reagent to obtain a reactive acid derivative compound 2;
  • an appropriate hydroxyl-containing compound such as pentafluorophenol or N-hydroxysuccinimide
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 3 The amino protecting group PG 4 on compound 4 is selectively removed under deprotection conditions (for example, the Cbz protecting group on the amino group can be cleaved off under the action of hydrogen and palladium-carbon catalyst), and the Boc on the amino group is protected The group can be cleaved under acidic conditions to obtain compound 5;
  • organic bases such as TEA, DBU, DIPEA
  • inorganic bases such as Na 2 CO 3 , Cs 2 CO 3 , K 2 CO 3 , NaHCO 3
  • Step 5 The amino protecting group PG1 on compound 6 is removed under deprotection conditions (for example, the Cbz protecting group on the amino can be cleaved under the action of hydrogen and palladium-carbon catalyst), and the Boc protecting group on the amino can be acidic It is excised under conditions to obtain compound XII;
  • the process of preparing the conjugate described above is characterized in that the synthesis of structural formula (XIII) includes one or more of the following steps:
  • Step 1 The carboxyl-containing compound 1 and the amino-containing compound 2 undergo a condensation reaction under the action of a condensing agent (such as EDC, HATU, DIC, DCC) or through a condensation reaction route to obtain compound 3;
  • a condensing agent such as EDC, HATU, DIC, DCC
  • Step 2 The carboxyl protecting group PG 1 on compound 3 is removed under deprotection conditions (for example, the tert-butyl ester protecting group on the carboxyl group can be removed under the action of formic acid, acetic acid, trifluoroacetic acid, hydrochloric acid, phosphoric acid, etc. ) To obtain compound 4;
  • Step 3 Carboxylic acid compound 4 and a hydroxyl-containing compound (such as pentafluorophenol or N-hydroxysuccinimide) undergo a condensation reaction in the presence of a condensation reagent to obtain a reactive ester structure (XIII);
  • a hydroxyl-containing compound such as pentafluorophenol or N-hydroxysuccinimide
  • the carboxylic acid compound 4 reacts with ethyl chloroformate, isobutyl chloroformate, etc., in the presence of an organic base such as N-methylmorpholine, triethylamine, diisopropylethylamine, etc., to obtain a reaction Active mixed acid anhydride structure (XIII);
  • the carboxylic acid compound 4 reacts with oxalyl chloride in the presence of an organic base such as triethylamine and a catalyst amount of DMF to obtain the acid chloride structure (XIII),
  • a pharmaceutical composition comprises the conjugate described in any one of the above or the conjugate formed by reacting the compound containing the linker described above with a cell binding molecule, and a pharmaceutically Acceptable excipients.
  • the conjugate described in any one of the above is used in the preparation of drugs for the treatment of cancer, infection or autoimmune diseases.
  • FIG. 1 Shows the synthesis of Tubulysin derivative fragments 13 and 18.
  • FIG. 1 shows the synthesis of Tubulysin derivative fragment 34.
  • FIG. 3 Shows the synthesis of Tubulysin derivative fragments 37, 38 and 45.
  • Figure 4. shows the synthesis of Tubulysin derivative fragment 57.
  • Figure 5 shows the synthesis of Tubulysin derivative fragment 71.
  • Figure 6 shows the synthesis of a Tubulysin derivative 72 that can be coupled.
  • Figure 7 shows the in vivo anti-tumor activity of the conjugate on BALB/c nude mice bearing NCI-N87 xenograft tumors.
  • Alkyl refers to linear or cyclic linear or branched aliphatic hydrocarbons containing 1 to 8 carbon atoms. Branched chain refers to a linear alkyl group with one or more lower alkyl groups, such as methyl, ethyl or propyl connected.
  • alkyl groups include methyl, ethyl, n-propyl, isopropyl, n-butyl, tert-butyl, n-pentyl, 3-pentyl, octyl, nonyl, decyl, Cyclopentyl, cyclohexane, 2,2-dimethylbutyl, 2,3-dimethylbutyl, 2,2-dimethylpentyl, 2,3-dimethylpentyl, 3,3-Dimethylpentyl, 2,3,4-Trimethylpentyl, 3-Methylhexyl, 2,2-Dimethylhexyl, 2,4-Dimethylhexyl, 2,5- Dimethylhexyl, 3,5-dimethylhexyl, 2,4-dimethylpentyl, 2-methylheptyl, 3-methylheptyl, n-heptyl, isoheptyl, n-octyl And isooc
  • the C 1 -C 8 alkyl group may be unsubstituted or substituted by one or more of the following but not limited to: C 1 -C 8 alkyl group, C 1 -C 8 alkoxy group, aryl group, acyl group , Acyloxy group, ester group, -C(O)NH 2 , -C(O)NHR', -C(O)N(R') 2 , -NHC(O)R',-S(O) 2 R', -S(O)R', -OH, halogen (-F, -Cl, -Br, -I), -N 3 , -NH 2 , -NHR', -N(R') 2 and- CN; wherein R'refers to C 1 -C 8 alkyl or aryl.
  • C 3 -C 8 carbocyclic ring refers to a saturated or unsaturated non-aromatic hydrocarbon cyclic compound containing 3, 4, 5, 6, 7, or 8 carbon atoms.
  • Typical C 3 -C 8 carbocyclic rings include but are not limited to cyclopropyl, cyclobutyl, cyclopentyl, cyclopentadienyl, cyclohexyl, cyclohexenyl, 1,3-cyclohexadienyl, 1 ,4-Cyclohexadienyl, cycloheptyl, 1,3-cycloheptadienyl, 1,3,5-cycloheptatrienyl, cyclooctyl and cyclooctadienyl.
  • the C 3 -C 8 carbocyclic ring may be unsubstituted or substituted by one or more of the following but not limited to: C 1 -C 8 alkyl, C 1 -C 8 alkoxy, aryl, Acyl, acyloxy, ester, -C(O)NH 2 ,-C(O)NHR',-C(O)N(R') 2 ,-NHC(O)R',-S(O) 2 R',-S(O)R',-OH, halogen (-F,-Cl,-Br,-I), -N 3 ,-NH 2 ,-NHR',-N(R') 2 and -CN; wherein R'is C 1 -C 8 alkyl or aryl.
  • the C 3 -C 8 carbocyclic group refers to a group in which one hydrogen atom on the aforementioned C 3 -C 8 carbocyclic ring is replaced by a chemical bond.
  • Alkenyl refers to straight or branched aliphatic hydrocarbons containing one carbon-carbon double bond, with 2 to 8 carbon atoms in the carbon chain.
  • alkenyl groups include vinyl, propenyl, n-butenyl, isobutenyl, 3-methyl-2-butenyl, n-pentenyl, hexenyl, heptenyl, and octenyl.
  • Alkynyl refers to a straight or branched aliphatic hydrocarbon containing one carbon-carbon triple bond, with 2 to 8 carbon atoms in the carbon chain.
  • alkynyl groups include ethynyl, propynyl, n-butynyl, 2-butynyl, 3-methylbutynyl, n-pentynyl, hexynyl, heptynyl, and octynyl.
  • Heteroalkyl refers to an alkyl group containing 2 to 8 carbon atoms and having 1 to 4 carbon atoms substituted with O, S or N.
  • Aryl or aromatic group refers to an aromatic hydrocarbon or heteroaromatic hydrocarbon group composed of 3 to 14 carbon atoms (6 to 10 carbon atoms in most cases) containing one or more rings.
  • Heteroaromatic hydrocarbon group refers to one or more carbon atoms (in most cases 1, 2, 3 or 4 carbon atoms) by O, N, Si, Se, P or S (preferably O, S, N) The aromatic hydrocarbon group produced by the substitution.
  • Aryl or aromatic group also refers to an aromatic hydrocarbon group in which one or more hydrogen atoms are substituted.
  • R 13 and R 14 are respectively H, alkyl, alkene, alkynyl, heteroalkyl, aryl, aralkyl, carbonyl, or a pharmaceutically acceptable salt.
  • the halogen atom refers to fluorine, chlorine, bromine, and iodine atoms, preferably fluorine and chlorine.
  • the heterocyclic ring refers to an aromatic containing 2 to 8 carbon atoms, and 1 to 4 carbon atoms of a non-aromatic or heterocyclic ring are replaced by hetero elements. These miscellaneous elements are O, N, S, Se and P, preferably O, N and S.
  • the available heterocycles can also be found in "The Handbook of Chemistry and Physics", 78th edition, CRC Press, 1997-1998, pages 225 to 226.
  • Suitable non-heteroaryl groups include, but are not limited to, epoxy, azaethyl, sulfiethane, pyrrolidinyl, pyrazolidinyl, imidazolidinyl, oxiranyl, tetrahydrofuranyl, and dioxy Pentyl, tetrahydropyranyl, dioxanyl, piperidinyl, piperazinyl, morpholinyl, pyranyl, imidazolinyl, pyrrolinyl, pyrazolinyl, thiazolinyl , Tetrahydrothiopyranyl, dithianyl, thiomorpholinyl, dihydropyranyl, tetrahydropyranyl, tetrahydropyridyl, dihydropyridyl, tetrahydropyrimidinyl, dihydrothiopyran Groups, azepanyl groups and their fused rings with phenyl groups
  • Heteroaryl refers to an aromatic heteromonocyclic, bicyclic or polycyclic structure containing 3 to 14 (preferably 5 to 10) atoms.
  • alkyl, cycloalkyl, alkene, alkynyl, aryl, heteroaryl and heterocyclyl also refer to alkylene, cycloalkylene, and alkenylene formed by the loss of two hydrogen atoms from the corresponding hydrocarbon , Alkynylene, arylene, heteroarylene and heterocyclic ring etc.
  • Alkyl refers to a carbon atom attached to a hydrogen atom of an acyclic alkyl radical, usually a terminal or sp3 hybridized carbon atom replaced by an aryl group.
  • Typical arylalkyl groups include benzyl, 2-phenylethyl, 2-styryl, naphthylmethyl, 2-naphthylethyl, 2-naphthalenevinyl, naphthylbenzyl, 2-naphthylbenzeneethane Base etc.
  • Heteroaralkyl refers to an acyclic alkyl radical with a hydrogen atom linked to a carbon atom, usually a terminal or sp3 hybridized carbon atom replaced by a heteroaryl group. Heteroaralkyl represents, for example, 2-benzimidazolemethyl, 2-furanethyl.
  • “Hydroxy Protective Group” refers to methyloxymethyl ether (MOM), 2-methyloxyethyloxymethyl ether (2-MOEOM), tetrahydropyran ether, benzyl ether, p-methyloxybenzyl ether , Trimethylsilyl ether, triethylsilyl ether, triisopropylsilyl ether, tert-butyl dimethylsilyl ether, triphenylmethylsilyl ether, ethyl acetate, ethyl acetate with substituents, benzene Formate, benzyl formate, chloroacetate, methoxyacetate, phenoxyacetate, pivaloate, adamantanoate, trimethyl benzoate (mesitoate ), methylsulfonate and tosylate.
  • MOM methyloxymethyl ether
  • 2-MOEOM 2-methyloxyethyloxymethyl ether
  • Amino acids can be natural and/or unnatural amino acids, usually L-type or D-type, preferably alpha amino acids.
  • Natural amino acids are arranged by genes, they are alanine, arginine, asparagine, aspartic acid, cysteine, glutamic acid, glutamine, glycine, histidine, isoleucine Acid, leucine, lysine, methionine, phenylalanine, proline, serine, threonine, tyrosine, tryptophan and valine.
  • Unnatural amino acids are derived forms of protein amino acids.
  • 2-aminoisobutyric acid dehydroalanine
  • ⁇ -aminobutyric acid neurotrans
  • amino acid also includes amino acid analogs and analogs.
  • the analogues have the same structure of H 2 N(R)CHCO 2 H natural amino acids, but the R group is not found in natural amino acids.
  • analogs include serine, hexine, methionine sulfoxide, and methionine methylsulfonium salt.
  • the analogous amino acids have a structure different from the conventional chemical structure of alpha amino acids, but their functions are similar.
  • the term "unnatural amino acid” is in the form of "D", and natural amino acid is in the form of "L".
  • the amino acid sequence should preferably be recognized and cleaved by proteases.
  • amino acid sequence is selected as Val-Cit, Ala-Val, Ala-Lys, Gly-Lys, Ala-Ala, Val -Val, Val-Ala-Val, Lys-Lys, Ala-Asn-Val, Val-Leu-Lys, Cit-Cit, Val-Lys, Asp-Lys, Glu-Lys, Ala-Ala-Asn, Lys, Cit , Ser, and Glu molecules.
  • a "peptide” is formed by combining two or more amino acids with the amino group of one amino acid and the carboxyl group of another amino acid by a peptide bond (ie, an amide bond).
  • a compound with two amino acids connected by a peptide bond is called a dipeptide; a compound with three amino acids connected by a peptide bond is called a tripeptide, and so on, a compound with thirty amino acids connected by a peptide bond is called a tria peptide.
  • a peptide composed entirely of natural alpha amino acids is a natural peptide (natural protein).
  • Peptides containing one or more unnatural amino acids or amino acid analogs are unnatural peptides (peptoid compounds).
  • a peptide of two or more amino acids is a peptide unit.
  • a "glycoside” is a molecule that connects a sugar group to another group through its anomeric carbon bond through its sugar group bond. Glycosides can be linked by O-(O-glycoside), N-(sugar amine), S-(thioglycoside) or C-(C-glycoside) glycosidic bonds.
  • the core is that the empirical formula is C m (H 2 O) n (where m can be different from n, m and n are ⁇ 36).
  • glycosides in this article include glucose (glucose), fructose (levulose) allose, and Altro Sugar, mannose, gulose, idose, galactose, troose, galactosamine, glucosamine, sialic acid, N-acetylglucosamine, sulfoquinolone (6-deoxy-6-sulfo -D-glucopyranose), ribose, arabinose, xylose, soluble glucose, sorbitol, mannitol, sucrose, lactose, maltose, trehalose, maltodextrin, raffinose, glucuronic acid (glucuronide) And stachyose.
  • Glycosides can be in D or L form, 5-atom cyclic furanose form, 6-atom cyclic pyranose form, or non-cyclic form, ⁇ -isomer (the end group different carbon below the carbon atom in Haworth projection) -OH) or ⁇ -isomer (-OH of an anomeric carbon above the Haworth projection plane).
  • the glycosides commonly used herein are monosaccharides, disaccharides, polyols or oligosaccharides (containing 3-6 sugar units).
  • Antibody as used here, is used in the broadest sense, specifically covering complete monoclonal antibodies, polyclonal antibodies, specific antibodies, and multispecific antibodies (for example, bispecific antibodies and antibody fragments). For the biological activity of the antibody fragment, the necessary number of drug binding sites is linked.
  • the native form of an antibody is a tetramer consisting of two identical immunoglobulin chain pairs, each pair having a light chain and a heavy chain. In each pair, the light chain and heavy chain variable regions (VL and VH) are jointly responsible for binding to the antigen.
  • the light chain and heavy chain variable regions are interrupted by a framework region and three hypervariable regions, which are also called "complementarity determining regions" or "services". The constant area may be recognized for interaction with the immune system.
  • An antibody can be of any type (eg, IgG, IgE, IgM, IgD, and IgA), major class (eg, IgG1, IgG2, IgG3, IgG4, IgA1, and IgA2) or subclass.
  • Antibodies can be obtained from any suitable species. In some aspects, antibodies are of human or murine origin. The antibody can be human, humanized or chimeric.
  • the terms “specifically binds” and “specifically binds” mean that an antibody or antibody derivative will bind to its corresponding target antigen in a highly selective manner, instead of binding to many other antigens.
  • the antibody or antibody has an affinity derivative binding of at least about 1 ⁇ 10 ⁇ 7 M. It is best to combine 1x10 -8 M to 10 -9 M, 10 -10 M, 10 -11 M, or 10 -12 M.
  • the affinity of the predetermined antigen is at least twice greater than the affinity of the bound non-specific antigen (such as bovine serum albumin, casein).
  • Medical use or “medicinal use” means that the corresponding compound or compound composition does not produce harmful, allergic or other adverse reactions in animals or humans.
  • compositions include all carriers, diluents, adjuvants or forming agents, such as preservatives, antioxidants, fillers, disintegrating agents, wetting agents, emulsifiers, suspending agents, solvents, dispersion media, coatings, Antibacterial agents, antifungal agents, isotonic and absorption delaying agents, etc.
  • active auxiliary components can also be added to the pharmaceutical ingredients.
  • pharmaceutically acceptable salts refer to salt derivatives of the compounds of the present invention. After appropriate modification, the compounds of the present invention can form corresponding acid or base salts. Pharmaceutically acceptable salts include common non-toxic salts or quaternary ammonium salts. These salts can be prepared from the compounds of the present invention and corresponding non-toxic inorganic or organic acids.
  • inorganic acids include hydrochloric acid, hydrobromic acid, sulfuric acid, sulfamic acid, phosphoric acid and nitric acid
  • organic acids include acetic acid, propionic acid, succinic acid, tartaric acid, citric acid, methanesulfonic acid, benzenesulfonic acid, glucaldehyde Acid, glutamic acid, benzoic acid, salicylic acid, toluenesulfonic acid, oxalic acid, fumaric acid and lactic acid can all be made into medicinal salts.
  • Other salts include ammonium salts such as tromethamine, aminotriethanol, meglumine, pyrrole ethanol, and metal salts such as sodium, potassium, calcium, zinc, and magnesium.
  • the pharmaceutically acceptable salt of the present invention can be prepared by conventional chemical methods. Generally speaking, these salts can be formed by adding other suitable equivalent amounts of bases or acids to the free acid or alkali aqueous solution or organic solution or a mixed solution of the compounds of the present invention.
  • the reaction medium of the non-aqueous phase is generally ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • the list of applicable salts can be found in "Remington’s Pharmaceutical Sciences", 17th edition. Mack Publishing Company, Easton, PA, 1985, page 1418.
  • pharmaceutically acceptable salt refers to a pharmaceutically acceptable organic or inorganic salt of a ligand drug conjugate or a linker drug conjugate.
  • the conjugate may contain at least one amino group, and therefore may form an acid addition salt with the amino group.
  • the pharmaceutically acceptable salt may include additional molecules such as acetate ion, succinate ion or other counter ion.
  • the counterion can be any organic or inorganic moiety that stabilizes the charge on the parent compound.
  • pharmaceutically acceptable salts may have more than one charged atom in their structure. Embodiments where multiple charged atoms are part of a pharmaceutically acceptable salt may have multiple counter ions. Therefore, a pharmaceutically acceptable salt may have one or more charged atoms and/or one or more counterions.
  • phrases "pharmaceutically acceptable solvate” or “solvate” refers to the association of one or more solvent molecules and a ligand drug conjugate or a linker drug conjugate.
  • solvents that form pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, butanol, tert-butanol, acetone, glycerol, DMSO, ethyl acetate, formic acid, acetic acid, triethanolamine and Ethanolamine.
  • Hydrate refers to a compound containing water.
  • the water can be connected to other parts by coordination bonds, such as a metal ion hydrated ligand to form a complex, or it can be combined by covalent bonds, such as hydrated chloroacetaldehyde. It can also refer to crystals or liquid molecules formed by certain compounds and moisture under certain temperature and pressure conditions.
  • the water in the hydrate is present in a certain amount.
  • the composition of anhydrous sodium sulfate Na 2 SO 4 hydrate is Na 2 SO 4 ⁇ 10H 2 O.
  • Water may not be directly combined with cations or anions, but may exist in the crystal in a certain proportion, occupying a certain position in the crystal lattice. This combined form of water is called lattice water and generally contains 12 water molecules. Some crystalline compounds also contain water, but there is no certain proportion.
  • the hydrate salt refers to a pharmaceutically acceptable salt formed on the basis of the hydrate.
  • Optical isomers are also called enantiomers, antipodal isomers, optical isomers, mirror image isomers, enantiomers or chiral isomers, which cannot be completely overlapped with each other's stereoisomers.
  • a substance contains a chiral carbon atom
  • Enantiomers have the same optical rotation power, but the direction of rotation is opposite, and their physical and chemical properties are very likely to be similar.
  • a molecule containing two carbon atoms with the same properties has three optical isomers.
  • the number of optical isomers is 2 n
  • n is the number of atoms with different chirals.
  • patients or “subjects” include, but are not limited to, humans, rats, mice, guinea pigs, monkeys, pigs, goats, cows, horses, dogs, cats, birds, and poultry.
  • the patient or subject is a human.
  • administering refers to any means of transferring, delivering, introducing or delivering drugs or other agents to a subject, including oral, topical, intravenous, intraperitoneal, intramuscular, intralesional, intranasal, subcutaneous or sheath Internal administration
  • the present invention also contemplates the use of devices or devices to administer medicaments. Such devices can utilize active or passive transport, and can be slow-release or rapid-release delivery devices.
  • Boc tert-butoxycarbonyl
  • BroP bromotripyrrolidine phosphonium hexafluorophosphate
  • CDI 1,1'-carbonyldiimidazole
  • DCC dicyclohexyl carbon Diimine
  • DCE dichloroethane
  • DCM methylene chloride
  • DIAD diisopropyl azodicarboxylate
  • DIBAL-H diisobutylaluminum hydride
  • DIPEA diisopropylethylamine
  • DEPC Dicyano diethyl phosphate
  • DMA N, N-dimethylacetamide
  • DMAP 4-(N, N-dimethylamino) pyridine
  • DMF N, N-dimethylformamide
  • DMSO Dimethyl sulfoxide
  • DTT dithiothreitol
  • EDC 1-(3-dimethylamino
  • the present invention includes compounds of formula (I) having at least one desired atomic isotope substitution in an amount higher than the natural abundance (ie enrichment) of the isotope and uses of the compounds.
  • Isotopes are atoms that have the same atomic number but different mass numbers, that is, the same number of protons but different numbers of neutrons.
  • Isotopic substitutions such as deuterium substitutions, can be partial or complete. Partial deuterium substitution means that at least one hydrogen is replaced by deuterium.
  • the isotope is enriched by 90%, 95%, or 99% or more at any location of interest. In one embodiment, deuterium is enriched by 90%, 95%, or 99% at the desired location.
  • C 1 -C 6 means a group containing 1 to 6 carbons.
  • linker containing hydrophilic branches means that the main framework is C 2 -C 100 peptide unit (1-12 natural or unnatural amino acids), hydrazone bond group, disulfide group, ester group, An oxime group, an amide group or a thioether bond group.
  • salt means a salt of a compound suitable for use in pharmaceutical preparations.
  • the salt may be an acid addition salt, such as sulfate, hydrobromide, tartrate, methanesulfonate, maleate, citrate, phosphoric acid Salt, acetate, pamoate, hydroiodide, nitrate, hydrochloride, lactate, methyl sulfate, fumarate, benzoate, succinate, methanesulfonate, Lactobionate, suberate, tosylate, etc.
  • acid addition salt such as sulfate, hydrobromide, tartrate, methanesulfonate, maleate, citrate, phosphoric acid Salt, acetate, pamoate, hydroiodide, nitrate, hydrochloride, lactate, methyl sulfate, fumarate, benzoate, succinate, methanesulfonate, Lactobionate, suberate, tosylate, etc.
  • the salt may be the following salts, such as calcium salt, potassium salt, magnesium salt, meglumine salt, ammonium salt, zinc salt, piperazine salt, tromethamine salt , Lithium salt, choline salt, diethylamine salt, 4-phenylcyclohexylamine salt, benzathine salt, sodium salt, tetramethylammonium salt, etc. Polymorphic crystalline forms and solvates are also included in the scope of the present invention.
  • the pharmaceutically acceptable salt of the present invention can be prepared by conventional chemical methods. Generally speaking, these salts can be formed by adding other suitable equivalent amounts of bases or acids to the free acid or alkali aqueous solution or organic solution or a mixed solution of the compounds of the present invention.
  • the reaction medium of the non-aqueous phase is generally ether, ethyl acetate, ethanol, isopropanol or acetonitrile.
  • compositions include all carriers, diluents, adjuvants or forming agents, such as preservatives, antioxidants, fillers, disintegrating agents, wetting agents, emulsifiers, suspending agents, solvents, dispersion media, coatings, Antibacterial agents, antifungal agents, isotonic and absorption delaying agents, etc.
  • active auxiliary components can also be added to the pharmaceutical ingredients.
  • formula (I) Indicates the chiral carbon atom site, which can be selected as pure R, pure S or a mixed structure of different ratios of R/S.
  • Another aspect of the present invention is the production and preparation of antibodies. Including its production process or combination process in vivo, in vitro.
  • the production methods of polyclonal antibodies against receptor peptides are well known, such as US Patent No. 4,493,795 (Nestor et al.).
  • the classic method of preparing monoclonal antibodies is to immunize mice with specific antigens and fuse the isolated mouse spleen cells with myeloma cells (Kohler, G; Milstein, C. 1975. Nature 256:495-497).
  • monoclonal antibodies can be obtained by immunizing mice, rats, hamsters or other mammals with the antigen of interest.
  • the target antigens include: intact cells, antigens isolated from cells, intact viruses, weakened intact viruses and viral proteins.
  • the spleen cells and myeloma cells were fused with PEG6000.
  • the hybridomas obtained after the fusion are screened using their sensitivity to HAT.
  • Monoclonal antibodies produced by hybridoma cells react with specific target cell receptors or inhibit receptor activity, which plays a role in the implementation of this invention.
  • the monoclonal hybridoma cells obtained after fusion can secrete monoclonal antibodies against specific antigens.
  • the monoclonal antibodies used in the invention are enriched by culturing monoclonal hybridoma cells in a nutrient-rich medium. The culture conditions need to ensure that the hybridoma cells have enough time to secrete the antibodies produced into the culture medium. After collecting the antibody-containing culture supernatant, the antibody is purified by well-known techniques.
  • Separation methods include: protein A affinity chromatography; anion exchange chromatography, cation exchange chromatography, hydrophobic chromatography and molecular sieve chromatography (especially affinity chromatography and molecular sieves using antigen cross-linked protein A) Chromatography is widely used); centrifugation; precipitation or other standard purification methods
  • the effective medium and artificial synthetic medium required for hybridoma culture can be obtained through technical synthesis or commercial channels.
  • a typical synthetic medium DMEM (Dulbecco et al. Virol 8: 396 (1959)) added 4.5mg/L glucose, 20mM glutamine, 20% fetal bovine serum and antifoaming agents, such as: polyoxyethylene polyoxygen Propylene copolymer.
  • antibody-producing cell lines can also be constructed by other methods, such as: directly transfecting tumorigenic DNA into B lymphocytes, or introducing oncogenic virus genes (such as EBV, also known as HHV-4 or KSHV)
  • oncogenic virus genes such as EBV, also known as HHV-4 or KSHV
  • EBV also known as HHV-4 or KSHV
  • B lymphocytes see US Patent Nos. 4,341,761; 4,399,121; 4,427,783; 4,444,887; 4,515,70; 4,466,917; 4,472,500; 4,491,632; 4,493,890.
  • Monoclonal antibodies can also be prepared by anti-receptor polypeptides or carboxy-terminal polypeptides. For details, see Niman et al. Proc. Natl. Acad. Sci.
  • anti-receptor polypeptides or polypeptide analogs can be used alone or cross-linked immunogenic carriers as immunogens to prepare anti-receptor polypeptide monoclonal antibodies.
  • Phage display technology obtains fully human antibodies that specifically bind to known antigens from a fully human antibody library through affinity screening.
  • the phage display technology itself, vector construction and library screening are all well documented in the literature. For details, see Dente et al. Gene.148(1): 7-13 (1994); Little et al. Biotechnol Adv. 12(3): 539-55 (1994); Clackson et al. Nature 352:264-628 (1991); Huse et al. Science 246: 1275-1281 (1989).
  • the monoclonal antibodies obtained from other species (such as mice) using hybridoma technology need to be humanized.
  • the modified antibody can greatly reduce the immune side reaction caused by heterologous antibodies to the human body.
  • the more common method of antibody humanization is the transplantation and remodeling of complementary determining domains.
  • U.S. Patent Nos. 5859205 and 6797492 Liu et al., Immunol Rev. 222: 9-27 (2008); Almagro et al., Front Biosci. 1; 13: 1619-33 (2008); Lazar et al. Mol Immunol. 44(8) ): 1986-98 (2007); Li et al. Proc. Natl. Acad. Sci. USA.
  • Fully human antibodies can also be prepared by immunizing transgenic mice, rabbits, monkeys and other mammals carrying a large number of human immunoglobulin light and heavy chains with antigen. Take mice as an example: Xenomouse (Abgenix, Inc.), HuMab-Mouse (Medarex/BMS), VelociMouse (Regeneron), see: U.S. Patent No.: 6596541,6207418,6150584,6111166,6075181,5922545,5661016,5545806 , 5436149 and 5569825.
  • Antibodies that are immunospecific to malignant cell antigens can be obtained through commercial channels or some mature technical methods, such as chemical synthesis or recombinant expression technology.
  • the coding genes of such antibodies can also be obtained through commercial channels, such as GenBank database or other similar databases, published literature, or conventional cloning and sequencing methods.
  • polypeptides or proteins can also be used as binding molecules to bind, block, attack or interact with corresponding receptors or epitopes on the surface of target cells by other means. As long as these polypeptides or proteins can specifically bind to specific epitopes or their corresponding receptors, they do not necessarily belong to the immunoglobulin family. These polypeptides can also be isolated by techniques similar to phage display antibodies (Szardenings, J Recept Signal Transduct Res. 2003; 23(4): 307-49). The peptide fragments obtained from random peptide libraries are similar to the application of antibodies and antibody fragments. A polypeptide or protein molecule can maintain its antigen binding specificity by connecting its binding molecules with some macromolecules or mediators. These macromolecules and media include albumin, polymers, liposomes, nanoparticles or dendrimers.
  • antibodies used for coupling are as follows (but not limited to this): 3F8 (anti-GD2 antibody), Abbavozumab (anti-CA-125 antibody), Abciximab (anti-CD41 antibody (integrin ⁇ -IIB), adalimumab (anti-TNF- ⁇ antibody), adelimumab (anti-EpCAM antibody, CD326), afelimumab (anti- -TNF- ⁇ ); Aftu beads (anti-CD20 antibody), Alacizumab pegol (anti-VEGFR2 antibody), ALD518 (anti-IL-6 antibody), alemtuzumab (alias: Campath, MabCampath, Campas, Anti-CD52 antibody), Atutumomab (anti-CEA antibody), Anatumomab (anti-TAG-72 antibody), Anrukinzumab (alias: IMA-638, anti-IL-13 antibody), Apolizumab (alias: IMA-638, anti-IL
  • J591 Anti-PSMA antibody, treatment of prostate cancer, Weill Cornell Medical College
  • 225.28S anti-HMW-MAA (high molecular weight melanoma-associated antigen) antibody, Sorin Radiofarmaci SRL (Milan, Italy) for treatment of melanoma
  • COL-1 Anti-CEACAM3 antibody, CGM1, nat.cancer inst..
  • Treatment of colorectal cancer and gastric cancer Treatment of colorectal cancer and gastric cancer
  • CYT-356 Treatment of prostate cancer
  • HNK20 OraVax, for the treatment of respiratory syncytial virus
  • ImmuRAIT from IMMUNOMEDICS for the treatment of non-Hodgkin's lymphoma
  • Lym-1 anti-HLA-DR10 antibody, Peregrine Pharmaceuticals, for cancer
  • MAK-195F anti-TNF antibody (also known as: tumor necrosis factor; TNFA, tumor necrosis factor- ⁇ ; TNFSF2), Abbott/Nor, for the treatment of septic toxic shock]
  • MEDI-500 [alias: T10B9 , Anti-CD3 antibody, TR ⁇ (T cell receptor ⁇ / ⁇ ), complex, MedImmune company for the treatment of graft-versus-host disease], RING SCAN [anti-TAG72 (tumor-associated glycoprotein 72 antibody), Neoprobe Group, for Treatment of breast, colon and rectal cancer.
  • Avicidin anti-EpCAM antibody (epithelial cell adhesion molecule), anti-TACSTD1 antibody (tumor-associated calcium signal transduction 1), anti-GA733-2 (gastrointestinal tumor-associated protein 2), anti-EGP-2 antibody (epiglycoprotein 2) ); Anti-KSA antibody; KS1/4 antigen; M4S; tumor antigen 17-1A; CD326, from NeoRx company for the treatment of colon cancer, ovarian cancer, prostate cancer and non-Hodgkin’s lymphoma; LymphoCide (IMMUNOMEDICS, NJ) , Smart ID10 (Protein Design Labs), Oncolym (Techniclone, California), Allomune (BioTransplant, CA), anti-VEGF antibody (Genentech, CA); CEAcide (IMMUNOMEDICS, NJ), IMC-1C11 (ImClone, NJ ) And Cetuximab (ImClone, New Jersey).
  • IMMUNOMEDICS Smart ID10 (Protein Design Labs), On
  • antibodies used to bind antigen include (but are not limited to): aminopeptidase N (CD13), annexin A1, B7-H3 (CD276, various cancers), CA125, CA15-3 (cancer), CA19- 9 (cancer), L6 (cancer), Lewis Y (cancer), Lewis X (cancer), alpha-fetoprotein (cancer), CA242, placental alkaline phosphatase (cancer), prostate specific antigen (prostate cancer), prostate Acid phosphatase (prostate), epidermal growth factor (carcinoma), CD2 (Hodgkin’s disease, non-Hodgkin’s lymphoma, multiple myeloma), CD3 epsilon (T cell lymphoma, lung cancer, breast cancer) , Gastric cancer, ovarian cancer, autoimmune disease, malignant ascites), CD19 (B-cell malignant tumor), CD20 (non-Hodgkin's lymphoma), CD22 (leukemia, lympho
  • antigens are: other different clusters (CD1, CD1a, CD1b, CD1c, CD1d, CD1e, CD2, CD3, CD3d, CD3e, CD3g, CD4, CD5, CD6, CD7, CD8, CD8a, CD8b, CD9 , CD10, CD11a, CD11b, CD11c, CD11d, CD12w, CD14, CD15, CD16, CD16a, CD16b, CDw17, CD18, CD19, CD20, CD21, CD22, CD23, CD24, CD25, CD26, CD27, CD28, CD29, CD30 , CD31, CD32, CD32a, CD32b, CD33, CD34, CD35, CD36, CD37, CD38, CD39, CD40, CD41, CD42, CD42a, CD42b, CD42c, CD42d, CD43, CD44, CD45, CD46, CD47, CD48, CD49b , CD49c, CD49c, CD49d, CD
  • the conjugate of the present invention is suitable for cancer treatment.
  • cancers include, but are not limited to, adrenal cortex cancer, rectal cancer, bladder cancer, brain tumors (adults: brainstem glioma, children, cerebellar astrocytoma, astrocytoma, ependymoma, medulloblastoma) Tumor, supratentorial primitive neuroectodermal tumor, pineal gland, visual pathway and hypothalamic glioma), breast cancer, carcinoid tumor, gastrointestinal tract, unknown primary cancer, cervical cancer, colon cancer, endometrial cancer , Esophageal cancer, extrahepatic cholangiocarcinoma, necessarily family tumors (PNET), extracranial malignant germ cell tumors, eye cancer, intraocular melanoma, gallbladder cancer, gastric cancer (stomach), germ cell tumors, extragonadal, gestational trophoblastic tumors , Head and neck cancer, hypopharyngeal cancer, islet cell carcinoma
  • the conjugate of the present invention is suitable for the prevention and treatment of autoimmune diseases.
  • Autoimmune diseases include, but are not limited to, achlorhydria, autoimmune chronic active hepatitis, acute disseminated encephalomyelitis, acute hemorrhagic leukoencephalitis, Addison's disease, gammaglobulinemia, alopecia areata, muscle atrophy Lateral sclerosis, ankylosing spondylitis, anti-glomerular basement membrane/tubular basement membrane nephritis, antiphospholipid syndrome, antisynthetic syndrome, arthritis, atopic hypersensitivity, allergic dermatitis, autoimmunity Aplastic anemia, autoimmune cardiomyopathy, autoimmune hemolytic anemia, autoimmune hepatitis, autoimmune inner ear disease, autoimmune lymphoproliferative syndrome, autoimmune peripheral nervous system disease, autoimmune Pancreatitis autoimmune polyendocrine disease type I, II, III, autoimmune progesterone dermatitis,
  • the antigen-binding molecules for coupling for the treatment or prevention of autoimmune diseases include, but are not limited to: anti-elastin antibody; Abyss anti-epithelial cell antibody; anti-basement membrane type IV collagen antibody ; Anti-nuclear antibody; Anti-double-stranded DNA antibody; Anti-single-stranded DNA antibody, anti-cardiolipin antibody IgM, IgG; Anti-celiac (anti-celiac antibody) antibody; Anti-phospholipid antibody IgK, IgG; Anti-riboprotein antibody; Anti-mitochondrial antibody ; Thyroid antibody; Microsomal antibody, T-cell antibody; Thyroglobulin antibody, anti-scleroderma-70 antibody (anti-SCL-70); Human anti-Jo antibody (anti-jo); Anti-systemic lupus erythematosus itself Antibodies; Anti-Sjogren’s Syndrome Antibodies (Anti-La/SSB); Anti-Systemic Lupus Erythematosus
  • the binding molecules used for conjugation in the present invention can bind to receptors or receptor complexes expressed by activated lymphocytes associated with autoimmune diseases.
  • immunoglobulin gene superfamily members such as CD2, CD3, CD4, CD8, CD19, CD20, CD22, CD28, CD30, CD33, CD37, CD38, CD70, CD79, CD79b, CD90, CD123, CD125, CD138, CD152/ CTLA-4, PD-1, or ICOS
  • members of the TNF receptor superfamily such as CD27, CD40, CD95/Fas, CD134/OX40, CD137/4-1BB, INF-R1, TNFR-2, RANK, TACI, BCMA, osteoprotegerin, Apo2/TRAIL-R1, TRAIL-R2, TRAIL-R3, TRAIL-R4, Trop2 and 30 APO-3
  • integrins such as CD27, CD40, CD95/Fas, CD134/OX40, CD137/4-1BB, INF-R1,
  • useful binding partners immunospecific for viral or bacterial antigens are human or human monoclonal antibodies.
  • viral antigen includes, but is not limited to: any viral peptide that can induce an immune response, polypeptide protein (for example, HIV gp120, HIV nef, RSV F glycoprotein, influenza virus neuraminidase, influenza virus blood Lectin, human T lymphocyte virus infection regulator tax, herpes simplex virus glycoproteins (eg, gB, gC, gD and gE) and hepatitis B surface antigen).
  • polypeptide protein for example, HIV gp120, HIV nef, RSV F glycoprotein, influenza virus neuraminidase, influenza virus blood Lectin, human T lymphocyte virus infection regulator tax, herpes simplex virus glycoproteins (eg, gB, gC, gD and gE) and hepatitis B surface antigen).
  • bacteria antigen includes, but is not limited to: any microbial peptides, polypeptide proteins, carbohydrates, polysaccharides, lipid molecules (for example, bacteria, fungi, pathogenic protozoa, Yeast polypeptides include, for example, lipopolysaccharides and capsular polysaccharides (5/8).
  • Useful type I antibodies that can be used to treat viral or bacterial infections include, but are not limited to: Palivizumab, a human anti-respiratory syncytial virus monoclonal antibody used to treat RSV infection; PRO542, a CD4 fusion antibody for the treatment of HIV infection; Ostavir, a humanized antibody for the treatment of hepatitis B virus; PROTVIR, a humanized antibody for the treatment of cytomegalovirus IgG.sub.1, and anti-lipopolysaccharide (anti- LPS) antibodies.
  • the conjugate of the present invention can be used to treat infectious diseases.
  • infectious diseases include, but are not limited to: Acinetobacter infection, actinomycosis, African sleeping sickness (African trypanosomiasis), AIDS (acquired immunodeficiency syndrome), amebiasis, microsporidiosis, anthrax , Cryptobacillus hemolyticus infection, Argentine hemorrhagic fever, ascariasis, aspergillosis, astrovirus infection, Babesia, Bacillus cereus infection, bacterial pneumonia, bacterial vaginosis, Bacteroides infection, small bagworm disease, Bailey roundworm infection, BK virus infection, black sarcoidosis, Blastocystis hominis infection, Blastomyces, Perun hemorrhagic fever, Borrelia infection, botulism (and infant botulism), Brazilian hemorrhagic fever, Brucella Bacillosis, Burkholderia infection, Buruli ulcer, Calicivirus
  • the antibody binding molecules previously described in this patent can be used against pathogenic strains, including, but not limited to: Acinetobacter baumannii, Actinomyces israelia, Actinomyces gordonii and Propionibacterium propionate, Trypanosoma brucei Insects, HIV (Human Immunodeficiency Virus), Entamoeba histolytica, Anaplasma, Bacillus anthracis, Cryptobacterium hemolyticus, Junin virus, Ascaris, Aspergillus, Astrovirus family, Babesia, Bacillus cereus Bacillus, Polybacteria, Bacteroides, Colonic ciliates, Bailey Ascaris, BK virus, Nodularia hoodia, Blastocystis hominis, Blastomyces dermatitis, Arenavirus, Borrelia, Clostridium botulinum Bacteria, Chrysalis, Brucella, usually Burkholderia cepacia and other Burkholderi
  • Antibodies acting on pathogenic virus antigens include the following examples but not limited to: variola virus, herpes virus, adenovirus, papovavirus, intestine Daoviridae, Parvoviridae, Parvoviridae, Reovirus, Retroviridae, Influenza virus, Parainfluenza virus, Mumps, Measles, Respiratory syncytial virus, Rubella, Arbovirus, Rhabdovirus, Arenaviridae, Non-A/Non-B hepatitis virus, rhinovirus, coronavirus, rotaviridae, tumor virus [e.g., hepatitis B virus (hepatocellular carcinoma), human papilloma virus (cervical cancer, Anal cancer), Kaposi's sarcoma-associated herpes virus (Kaposi's sarcoma), Epstein-Barr virus (n
  • the present invention also includes the above-mentioned conjugate components combined with other feasible drug carriers as therapeutic drugs for cancer and autoimmune diseases.
  • the methods of the present invention for treating cancer and autoimmune diseases include in vitro, in vivo or indirect in vivo therapy.
  • in vitro therapy applications include treating cells cultured in vitro with drugs to kill all cells except cells that do not express the target antigen; or killing cells that express unwanted antigens.
  • hematopoietic stem cells are processed in vitro, the diseased or malignant cells are killed and then returned to the original patient.
  • the tumor cells or lymphocytes in the bone marrow are removed by indirect in vivo treatment and then returned to the original patient to treat cancer and autoimmune diseases, or the T cells and other lymphocytes in the bone marrow are removed before transplantation to prevent transplantation.
  • Immune antagonistic response The method of implementation is as follows: bone marrow cells are obtained from patients or other individuals, and then cultured in a serum-containing medium containing the conjugate of the present invention at 37°C.
  • the drug concentration ranges from 1 pM to 0.1 mM, and the culture time is about 30 minutes to about 48 hours. .
  • the specific drug concentration and incubation time are determined by experienced clinicians.
  • the bone marrow cells are washed with serum-containing medium and then injected back into the human body by intravenous injection. If the patient needs other treatments before obtaining bone marrow cells and reinfusion treatment, such as ablative chemotherapy or systemic radiotherapy, the processed bone marrow cells can be stored in qualified liquid nitrogen medical equipment.
  • the conjugate of the present invention When used in in vivo clinical applications, the conjugate of the present invention will be provided in the form of a solution or a freeze-dried solid that can be dissolved in sterile water and injected.
  • suitable conjugate administration methods are as follows: the conjugate is injected intravenously once a week for 4-12 weeks. A single dose is dissolved in 50 to 500 ml of normal saline, and normal saline can be added to human serum albumin (for example, 0.5 to 1 ml of 100 mg/ml concentrated human serum albumin).
  • the drug dose is about 50 ⁇ g to 20mg per kilogram of body weight per week, intravenous injection (each injection of 10ug to 200mg/kg body weight). After 4 to 12 weeks of treatment, the patient can receive a new round of treatment.
  • the detailed treatment method including the route of administration, excipients, diluents, drug dosage, treatment time, etc. can be determined by experienced surgeons.
  • diseases that can selectively kill cell populations in vivo or indirectly in vivo include any kind of malignant tumors, autoimmune diseases, transplant rejection and infections (including viruses, bacteria or parasites).
  • the amount of conjugate required to achieve the desired biological effect will vary depending on many factors, including the nature of the compound, the efficacy and the bioavailability of the conjugate, the type of disease, and the race of the patient, The patient's disease state, route of administration, all these factors together determine the schedule and method of administration.
  • the conjugate of the present invention can be used for parenteral administration by dissolving in a physiological buffer at a mass-volume ratio of 0.1 to 10%.
  • the typical dose of the drug ranges from 1 ug to 0.1 g per kilogram of body weight per day; the recommended dose of drug ranges from 0.01 mg to 20 mg per kilogram of body weight per day or equivalent doses for children.
  • the recommended dosage depends on many variables, including the type of disease or dysfunction, the overall health status of the individual patient, the relative biological activity of the coupled drug, the dosage form of the compound, and the method of administration (intravenous injection, intramuscular injection) , Or other), the pharmacokinetic properties of the selected administration mode, as well as the speed of administration (single injection or continuous infusion) and the schedule of administration (the number of administrations within a certain period of time).
  • the conjugate of the present invention can also be administered in the form of a unit dose.
  • the "unit dose” here refers to the dose administered by a patient at a time.
  • the unit dose of the drug can be simply and conveniently packaged and used, and the unit dose of the drug is to maintain The physically and chemically stable active conjugate itself, or a pharmaceutically acceptable mixture as described later.
  • a typical daily dose ranges from 0.01 to 100 mg per kilogram of body weight. In general, the daily unit dose for humans ranges from 1 to 3000 mg.
  • the recommended unit dose is 1 mg to 500 mg, administered four times a day, or 10 mg to 500 mg, once a day.
  • the conjugate of the present invention can be prepared into pharmaceutical preparations by adding one or more pharmaceutically acceptable excipients.
  • the unit dose of the drug can be used for oral administration, such as tablets, simple capsules or soft capsules; or intranasal administration, such as powder, nose drops, or spray; or through the skin, such as ointment Preparations, creams, lotions, gels or sprays or skin patches.
  • the medicament can be conveniently administered in the form of a unit dose, and prepared by any known pharmaceutical method, such as Remington: The Science and Practice of Pharmacy, 21th ed.; Lippincott Williams & Wilkins: Philadelphia, PA, 2005. Methods.
  • the pharmaceutical dosage form containing the compound of the present invention includes oral or parenteral administration of the pharmaceutical composition.
  • dosage forms such as tablets, powders, capsules, tablets (lozenges), etc. may contain one or more of the following raw materials or other compounds with similar properties: binders, such as microcrystalline cellulose or Tragacanth; diluents, such as starch or lactose; dispersants, such as starch and cellulose derivatives; lubricants, such as magnesium stearate; glidants, such as colloidal silica; sweeteners, such as sucrose or saccharin ; Flavour enhancers, such as peppermint or methyl salicylate.
  • Capsules can be in the form of hard capsules or soft capsules.
  • a mixture of gelatin is selectively mixed with a plasticizer, and the same applies to starch capsules.
  • the physical form of the unit dose can be changed by adding a variety of different materials, such as sugar coating, shellac or enteric agent.
  • Other oral dosage forms such as syrups or elixirs may contain sweetening agents, preservatives, pigments, coloring agents and flavoring agents.
  • the active compound can be made into a fast-dissolving dosage form, a slow-release dosage form or a sustained-release agent through different treatments and formulations, of which the sustained-release agent is a better dosage form.
  • the tablet preferably contains lactose, corn starch, magnesium silicate, croscarmellose sodium, polyvinylpyrrolidone, magnesium stearate, talc and other combinations.
  • Liquid medicines for parenteral administration include sterile aqueous or non-aqueous solutions, suspensions and emulsions. Liquid medicines may also contain binders, buffers, preservatives, chelating agents, sweeteners, flavoring and coloring agents, etc.
  • Non-aqueous solvents include ethanol, propylene glycol, polyethylene glycol, vegetable oils such as olive oil and organic lipids such as ethyl oleate.
  • Aqueous solvents include a mixture of water, ethanol, buffer reagents and salts, especially biocompatible, degradable lactide polymers, lactide/glycolide polymers or polyethylene glycol/polyglycerol Alcohol copolymers can be used as excipients to control the release of active drugs.
  • Excipients for intravenous injection can include fluid and nutritional supplements, electrolyte supplements, and Ringer's dextrose-based excipients, and similar excipients.
  • Other feasible parenteral administration systems for the active drugs of the present invention include ethylene-vinyl acetate copolymer particles, implantable osmotic pumps and liposomes.
  • inhalants including dry powders, aerosols and water drops.
  • the inhalant can be a solution containing, for example, polyoxyethylene-9-lauryl ether, glycocholate, deoxycholate or oil, and can be administered in the form of nasal drops or intranasal colloid.
  • Buccal agents include, for example, lozenges, candy lozenges, etc., which may contain flavoring agents such as sucrose, gum arabic, and other auxiliary materials such as glycocholate.
  • Suppositories are suitable in unit dose form, with solids such as cocoa butter as a carrier, and salicylic acid can also be added.
  • plasters, emulsions, lotions, patches, gels, sprays, aerosols or oils are the first choice.
  • Vaseline, lanolin, polyethylene glycol, alcohols and their mixtures can be used as drug carriers.
  • the dosage form for skin administration can be patch, emulsion, buffer solution, dissolved or dispersed in polymer or adhesive.
  • the conjugate of the present invention can work with other known or unknown therapeutic drugs, such as chemotherapy drugs, radiotherapy, immunotherapy drugs, autoimmune disease drugs, anti-infective drugs or other antibody drug conjugates, Achieve synergy.
  • Synergistic drugs or radiotherapy can be administered or performed before or after administration of the conjugate of the invention. It can be 1 hour, 12 hours, one day, one week, one month, or several months before or after taking the conjugate of the present invention.
  • the synergistic drugs include but are not limited to:
  • Chemotherapeutic drugs a). Alkylation reagents: such as [Chlorambucil: (Chlorambucil, Cyclophosphamide, Ifosfamide, Chlorambucil, Melphalan, Cyclophosphamide); Nitroureas: (Carmustine, Lomustine); Alkyl Sulfonates: (Busulfan, Susulfan); Triazenes: (Dacarbazine); Platinum Compounds: (Carboplatin , Cisplatin, oxaliplatin)]; b).
  • Alkylation reagents such as [Chlorambucil: (Chlorambucil, Cyclophosphamide, Ifosfamide, Chlorambucil, Melphalan, Cyclophosphamide); Nitroureas: (Carmustine, Lomustine); Alkyl Sulfonates: (Busulfan, Susulfan); Triazenes: (Dacarbazine);
  • Plant alkaloids such as [vinca alkaloids: (vinblastine, vincristine, vindesine, vinorelbine); taxane compounds: (paclitaxel , Taxotere)]; c).
  • DNA topoisomerase inhibitors such as [epitopodophyll resin: (9-aminocamptothecin, camptothecin, clinato, etoposide, etoposide phosphate) , Irinotecan, teniposide, topotecan,); mitomycin: (mitomycin C)]; d).
  • Antimetabolites such as ⁇ [antifolate: dihydrofolate reductase inhibitor : (Methotrexate, Trimetrexate); IMP dehydrogenase inhibitors (mycophenolic acid, formamidothiazole, ribavirin, EICAR); ribonucleotide reductase inhibitors (hydroxyurea, iron Amine)]; [Pyrimidine analogues: Uracil analogues: (5-Fluorouracil, deoxyfluridine, raltitrexed (Tuoyoude)); Cytosine analogues: (cytarabine, arabinocytosine) Glycoside, fludarabine); purine analogues: (azathioprine, mercaptopurine, guanine)] ⁇ ; e).
  • Hormones such as ⁇ receptor antagonist: [antiestrogens: (megestrol, Raloxifene, tamoxifen); LHRH agonists: (goserelin, leuprolide acetate); antiandrogens: (bicalutamide, flutamide)]; tretinoin/deltoid muscle: [Vitamin D3 analogs (CB 1093, EB 1089 KH 1060, cholecalciferol, vitamin D2); photodynamic therapy: (for this, phthalocyanine photosensitizer, PC4, demethoxymethyl); cytokines: (interferon - ⁇ , interferon- ⁇ , tumor necrosis factor (tumor necrosis factor), human protein contains TNF domain)] ⁇ f).
  • ⁇ receptor antagonist [antiestrogens: (megestrol, Raloxifene, tamoxifen); LHRH agonists: (goserelin, leuprolide acetate
  • Kinase inhibitors such as bibw 2992 (anti-EGFR/Erb2), imatinib, gemfibrine Tinib, pagatanib, sorafenib, dasatinib, sunitinib, erlotinib, nilotinib, lapatinib, axitinib, pazopanib, pona Tinib, Vandetanib, Flumatinib, e7080 (anti-VEGFR2), Moritinib, Meditinib, Pranatinib (ap24534), HQP1351, Bafitinib (INNO-406), Bosu Tinib (SKI-606), Sunitinib, Cabotinib, Volitinib, Vermodil, Iniparib, Ruxolitinib, CYT387, Axitinib, Tivozanib, Bevacizumab, Sorafenib, tras
  • gemcitabine such as lenalidomide
  • bortezomib Thalidomide such as lenalidomide
  • lenalidomide such as lenalidomide
  • pomalidomide such as lenalidomide
  • toxedote zybrestat
  • PLX4 032 sta-9090
  • Stimuvax such as lovastatin
  • dopaminergic neurotoxins such as 1-methyl-4-phenylpyridine ion
  • Cell cycle inhibitors such as staurosporine
  • actinomycin such as actinomycin D, dactinomycin
  • pingyangmycin such as bleomycin, bleomycin A2, B2, peiro Anthracycline
  • anthracycline antibiotics such as erythromycin, doxorubicin (adriamycin), idarubicin, epirubicin, pirarubici
  • Anti-autoimmune disease agents include, but are not limited to: cyclosporine, cyclosporine A, azathioprine, aminocaproic acid, bromocriptine, chlorambucil, chloroquine, cyclophosphamide, glucocorticoids (Such as hormonal drugs, betamethasone, budesonide, flunisolide, fluticasone propionate, hydrocortisone, dexamethasone, fluocondrodanazole, triamcinolone acetonide, beclomethasone propionate), Dehydroepiandrosterone, etanercept, hydroxychloroquine, infliximab, meloxicam, methotrexate, mycophenolate mofetil, sirolimus, tacrolimus, prednisone
  • Anti-infectious agents include, but are not limited to: a). Aminoglycosides: Amikacin, Wuyimycin, Gentamicin (netilmicin, sisomicin, isepamicin ), hygromycin, kanamycin (amikacin, arbekacin, aminodeoxykanamycin, dibekacin, tobramycin) neomycin (neomycin B, Paromomycin, Ribomycin), Netilmicin, Spectinomycin, Streptomycin, Tobramycin, Methyl stilomycin; b) Amido alcohols: chloramphenicol azide, chlorine Thiamphenicol, thiamphenicol, thiamphenicol; c).
  • Ansamycins Geldanamycin, herbimycin; d).
  • Carbapenems Biapenem, Donipel South, ertapenem, imipenem/cilastatin, meropenem, panipenem; e).
  • Cephalosporins carbocephalosporin (chlorocarbef), cefacetonitrile, cefaclor, cefradine, cephalosporin Amoxicillin, Ceflanine, Cefotaxime, Cefalotin or Cephalosporin, Cephalexin, Cephalosporin, Cefmandol, Cefpirin, Ceftriazine, Cefazolin, Cefazidone, Cefazolin, Cephalosporin Lazon, Cefcapine, Cefdioxime, Cefepime, Cefminox, Cefoxitin, Cefprozil, Cefoxadine, Cefotiazole, Cefuroxime, Cefdioxime, Cefdinir, Cefditoren, Cefdioxime Pyroxime, ceftazime, cefmenoxime, cefodizime, cefnixi, cefoperazone, cefradit, cefotaxime
  • Glycopeptides Bleomycin, Vancomycin (Olivancin, Special Lavancin), teicoplanin (dalbavancin), ramoranine, daptomycin; g). Glycyl: such as tigecycline; h). ⁇ -lactamase inhibitor: green Mycin (sulbactam, tazobactam), clavulatan (clavulanic acid); i). Lincosamides: clindamycin, lincomycin; j). lipopeptide: daptomycin , A54145, calcium-dependent antibiotic (CDA); k).
  • CDA calcium-dependent antibiotic
  • Macrolides azithromycin, quinerythromycin, quinerythromycin, clarithromycin, dirithromycin, erythromycin, fluorored Josamycin, josamycin, ketolactones (telithromycin, quinerythromycin quinerythromycin quinerythromycin quinerythromycin quinerythromycin), mediocin, meocamycin, oleomycin, Famycin (rifampicin, rifampicin, rifabutin, rifapentin), rotamycin, roxithromycin, spectinomycin, spiramycin, tacrolimus (FK506), vinegar Eleamycin, telithromycin; l).
  • Monocyclic ⁇ -lactam antibiotics aztreonam, tigemonam; m). oxazolidinones: linezolid; n).
  • Penicillins amoxicillin , Ampicillin (Pimacillin, Hetacillin, Bazamicillin, Metancillin, Phthalocillin) Azociillin, Alocillin, Penicillin, Benzathine penicillin, Phenoxybenzaprine, Clomethacillin, General Lucaine penicillin, Carbenicillin (Carbenicillin), o-Clocicillin, Dicloxacillin, Pioneermycin, Flucloxacillin, Mecillin (Azemidine penicillin diester), Mezlocillin, Methicillin, Nafcillin, Benzazole Cillin, acemethicillin, penicillin, necillin, penicillin, piperacillin, phenprocillin, sulbenicillin, temoxicillin, ticarcillin; o).
  • Peptides bacitracin, polymyxin E, polymyxa Su B; p).
  • Quinolones alafloxacin, balofloxacin, ciprofloxacin, clinfloxacin, danofloxacin, difloxacin, enoxacin, enrofloxacin, Floxin, plus Rafloxacin, Gatifloxacin, Gemifloxacin, Gapafloxacin, Travafloxacin Kano, Levofloxacin, Lomefloxacin, Marbofloxacin, Moxifloxacin, Nafloxacin, Norfloxacin, Orbifloxacin, Ofloxacin, Pefloxacin, Travafloxacin, Gapafloxacin, Sitafloxacin, Sparfloxacin, Temafloxacin, Tosufloxacin, Travafloxacin; q ).
  • Sulfa drugs Sulfamethuron, Pyramidol, Sulfaacetamide, Sulfamethan, Sulfa, Sulfasalazine , Sulfisoxazole, trimethoprim, trimethoprim-sulfamethoxazole (compound trimethoprim); s).
  • Steroid antibacterial drugs such as fusidic acid; t).
  • Tetracyclines powerful Chlortetracycline, Chlortetracycline, Chlortetracycline, Dimecycline, Lymecycline, Chlormethene oxytetracycline, Metacycline, Minocycline, Oxytetracycline, Penicillin, Rolicycline, Tetracycline, Glycine Aminoacyl (such as tigecycline); u).
  • antibiotics Annonaceae, Arsavanamine, bacterial terpene alcohol inhibitor (Bacitracin), Dadal/AR inhibitor (cycloserine), dictyostatin, sponge Lactone, Eurisalox, Epothilone, Ethambutol, Etoposide, Faropenem, Fusidic Acid, Fusidic Acid, Isoniazid, Laulimalide, Metronidazole, Mupirocin, mycolactones, NAM synthesis inhibitors (such as fosfomycin), nitrofurantoin, paclitaxel, slabmycin, pyrazinamide, quinupristin/dalfopristin, rifampin (rifampicin), tazobactam tinid Azole, annonaceous lactone;
  • Antiviral drugs a). Entry/fusion inhibitors: aplaviroc, Maraviro, Veriveiro, gp41 (Enfuvir), PRO140, CD4 (Ibalizumab); b). Integrase inhibition Agents: Lategravir, elvitegravir, globoidnan A; c). Maturation inhibitor: Bevirima, becon; d). Neuraminidase inhibitor: Oseltamivir, Zanamivir, Pala Mivir; e).
  • Nucleosides and nucleotides abacavir, acyclovir, adefovir, amdosovo, apricitabine, brivudine, cidofovirclavudine, dextrovir Tabine, didanosine (DDI), ifetabine, emtricitabine (FTC), entecavir, famciclovir, fluorouracil (5-FU), 3'-fluoro substituted 2', 3'-di Deoxynucleoside analogues (e.g., 3'-fluoro-2', 3'-dideoxythymidine (FLT) and 3'-fluoro-2', 3'-dideoxy (FLG) fomivirsen, ganciclones Lovir, iodoside, lamivudine (3TC), L-nucleoside (such as ⁇ -L-thymidine, ⁇ -L-2'-deoxycy
  • Non-nucleosides amantadine, ateviridine, capvirin, diarylpyrimidine (etravirin, ripavirin), deira Viridine, behenyl alcohol, emivirine, efavirenz, foscarnet (phosphoryl formic acid), imiquimod, interferon alpha, loviramide, lordadenosine, tapazole, nevirapine, NOV-205, peginterferon alpha, podophyllotoxin, rifampicin, rimantadine, resiquimod (R-848), acetamine adamantane; g).
  • Protease inhibitor amprenavir , Atazanavir, Beaucepvir, Darunavir, Fosamprenavir, Indinavir, Lopinavir, Nefinavir, pleconaril, Ritonavir, Saquinavir, Terra Pivir (VX-950) Telanavir; h).
  • antiviral drugs abzyme, arbidol, calanolides A, cerulenin, cyanobacteria-N, diarylpyrimidine, epigallium Catechin Gallate (EGCG), Foscarnet Sodium, Grifson, Talivirin (Viramididine), Hydroxyurea, KP-1461, Miltefosine, Praconaride, Synthesis Inhibitor , Ribavirin, seliciclib;
  • immunotherapy drugs such as imiquimod, interferons (such as ⁇ , ⁇ ), granulocyte colony stimulating factors, cytokines, interleukins (IL-1 ⁇ IL-35), antibodies (such as trast Mab, Pertuzumab, Bevacizumab, Cetuximab, Panitumumab, Infliximab, Adalimumab, Basiliximab, Daclizumab, Omalizumab Monoclonal antibody), protein-binding drug (for example, Abraxane), an antibody-binding drug selected from calicheamicin derivatives, maytansine derivatives (DM1 and DM4), CC-1065 and dokamycin minor groove agents, Effective taxol derivatives, doxorubicin, aritatin anti-mitotic drugs (such as trastuzumab-DM1, Inotuzumab monoclonal antibody, brentuximab vedotin, Glembatumumab vedotin,
  • the present invention also relates to the preparation process of the antibody drug conjugate.
  • the conjugate of the present invention can be prepared by a variety of well-known methods in this field.
  • the antimitotic agent in the conjugate of the present invention can be synthesized according to the following method or an improved method described below.
  • these improvement methods are well-known and obvious methods that are easily obtained from scientific and technological literature. In particular, these methods are introduced in the book "Comprehensive Organic Transformations" (written by R.C. Larock, 1999, Wiley-VCH Publishing, 2nd Edition).
  • reaction temperature that is easier to handle is usually between -80°C to 150°C (better between room temperature and 100°C).
  • time required for the reaction can also vary widely. Of course, this depends on many factors, especially the reaction temperature and the nature of the solvent used. Generally speaking, for an ideal reaction, a reaction time of 3 to 20 hours is appropriate.
  • the operation treatment after the completion of the reaction can be carried out in a conventional manner.
  • the reaction product can be recovered by distilling off the solvent from the reaction system.
  • the solvent is evaporated, the remainder can be poured into water, and then extracted with an organic solvent that is immiscible with water.
  • the extraction solvent is evaporated, the reaction product can be obtained.
  • it can be further purified by various common methods, such as recrystallization, sedimentation or various chromatographic methods. Generally speaking, column chromatography and preparative thin-plate chromatography are more commonly used.
  • the present invention is further illustrated by the following examples, and the content of these examples is not intended to limit the scope of the present invention.
  • the cell lines except for special instructions, are stored under the conditions specified by the American Standard Culture Collection (ATCC), the German Culture Collection (DSMZ) or the Shanghai Cell Culture Center of the Chinese Academy of Sciences. Except for special instructions, cell culture reagents are from Invitrogen. All anhydrous reagents are commercially obtained and stored in Sure-Seal sealed bottles. Other reagents and solvents were purchased according to the highest specifications and were used without further treatment. Use Varain PreStar HPLC for preparative HPLC separation. The NMR spectrum was obtained on a Bruker 500MHz instrument.
  • the crude product is mixed with silica gel (1.5kg) and mixed well, and purified by silica gel column (10kg) column chromatography (10-20% ethyl acetate/petroleum ether gradient elution) to obtain a brown oil, which is the target compound (509g, 92% yield).
  • the acetal (300g, 1.16mol) solution in acetone (3.0L) was heated to reflux, and 4N HCl solution (250mL) was added dropwise within 1.0 hour. TLC showed that the reaction of the starting material was complete.
  • the reaction solution was concentrated under reduced pressure and the two phases were separated.
  • the organic phase was diluted with ethyl acetate (1.5L), washed with saturated NaHCO 3 aqueous solution (1.0L), water (1.0L) and brine (1.0L), and then dried with anhydrous sodium sulfate. Combine all the aqueous phases and back-extract with ethyl acetate, and dry the organic phase with anhydrous sodium sulfate.
  • the organic phase was concentrated, and the crude product obtained was slurried with petroleum ether/ethyl acetate (5:1) solution, the precipitated solid was collected by vacuum filtration, and washed with petroleum ether/ethyl acetate (10:1) solution.
  • the filtrate was concentrated and purified by column chromatography (0-15% ethyl acetate/petroleum ether), and all the solids were combined to obtain 40 g (43% yield) of the target product, which was a white or bright yellow solid.
  • Azido-Ile-OH (8,153g, 0.97mol) was dissolved in tetrahydrofuran (1.5L) and cooled to 0°C, and NMM (214mL, 1.94mol) and isobutyl chloroformate (95mL, 0.73mol) were added in sequence. After stirring at 0°C for 1 hour, compound 7 (150 g, 0.49 mmol) was added in portions. After stirring for 30 minutes at 0°C, it was gradually raised to room temperature and stirring was continued for 2 hours. The reaction was quenched with ice water at 0°C and extracted three times with ethyl acetate.
  • reaction solution was diluted with water (6L), stirred for 30 minutes, the aqueous phase was extracted with dichloromethane (2L ⁇ 2), the organic phases were combined, washed with water (2L), brine (2L), dried and filtered, and concentrated to obtain a white solid 542g (95% yield).
  • the reaction was allowed to rise slowly, and stirring was continued for about 1 hour.
  • the reaction solution was heated to about -30°C, and TLC monitoring showed that the reaction was complete.
  • HATU (39.9 g, 105 mmol) was added to a solution of 4-(((benzyloxy)carbonyl)amino)butyric acid (26.1 g, 110 mmol) in DMF (300 mL). After stirring at room temperature for 30 minutes, the reaction mixture was added to a solution of compound 30 (39.4 g, 100 mmol) and triethylamine (20.2 g, 200 mmol) in DMF (300 mL). The reaction was stirred at room temperature for 2 hours, diluted with water, and extracted with ethyl acetate. The organic layer was washed with saturated brine and dried over sodium sulfate.
  • a DMF (60 mL) solution of compound 36 (36.0 g, based on 25.4 mmol) was added to the reaction flask and cooled to 5° C. in an ice water bath.
  • the concentrated solution was diluted with dichloromethane, cooled to 5°C in an ice water bath, and formic acid was slowly added dropwise to adjust the pH to 3.0-4.0. After that, it was concentrated until no solvent was evaporated, and the residue was transferred to a silica gel column, eluted with n-hexane/ethyl acetate/formic acid and dichloromethane/methanol/formic acid, and concentrated to obtain a purified crude product.
  • propionyl chloride (315g, 3.4mol) dropwise, after the dropwise addition, react at -70°C for 1 hour, slowly warm up to room temperature, pour the reaction solution into ice saturated aqueous salt solution (7L), and extract with ethyl acetate (3 ⁇ 2L), combine the organic phases, wash once with water (2L), wash once with saturated salt solution (2L), dry with anhydrous sodium sulfate, filter, spin dry, and purify with silica gel column (pure petroleum ether to 5:1 petroleum Ether/ethyl acetate) to obtain 500 g of a colorless oil with a yield of 87%.
  • silica gel column pure petroleum ether to 5:1 petroleum Ether/ethyl acetate
  • compound 38 (92.6g, 0.50mol) was dissolved in anhydrous dichloromethane (1.5L), the temperature was reduced to -10°C, and diisopropylethylamine (70.5g, 0.54mol) and n-Bu 2 BOTf (1.0M dichloromethane solution, 500 mL, 0.50 mol) was added dropwise to the reaction flask, reacted at 0°C for 1 hour, and then cooled to -78°C, and compound 25 (161 g, 0.45 mol) The methyl chloride (1L) solution was added dropwise into the reaction flask, and the temperature of the solution did not exceed -70°C.
  • compound 39 (200g, 0.37mol) was dissolved in anhydrous tetrahydrofuran (3.5L), and dithiocarbonylimidazole (198g, 1.11mol) was added. The reaction was refluxed for 8 hours, and dithiocarbonylimidazole (65g) was added. , 0.37mol), react overnight. The reaction was cooled to room temperature the next day, concentrated under reduced pressure to remove the solvent, and purified on a silica gel column (pure petroleum ether to 5:1 petroleum ether/ethyl acetate) to obtain an oily liquid (170 g, yield 83%).
  • compound 40 (210g, 323mmol) was dissolved in dry toluene (3L), and tri-n-butylstannane (182g, 646mmol) and azobisisobutyronitrile (0.5g, 3.23mmol) were added under nitrogen protection. ), react at reflux for 2 hours, cool to room temperature, spin dry, and purify by silica gel column (pure petroleum ether to 5:1 petroleum ether/ethyl acetate) to obtain an oily substance (141 g, 83% yield).
  • Dissolve compound 52 (67g, 94.2mmol) in 0.75L DMF, cool with ice water, add DIPEA (48.6g, 376.8mmol), keep the reaction solution at 10 to 20 degrees, slowly add compound 53 (105g, crude , Based on 94.2mmol). React at room temperature for 1 hour, concentrate, dilute with dichloromethane, wash with water, and extract the aqueous phase with dichloromethane. The organic phases were combined, washed with 0.2N hydrochloric acid, saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated. Purification by column chromatography (50%-100% ethyl acetate/petroleum ether, then 10% methanol/dichloromethane) yielded 80.5 g of the product with a yield of 98%.
  • the concentrated solution was diluted with dichloromethane, cooled to 5°C in an ice water bath, and formic acid was slowly added dropwise to adjust the pH to 3.0-4.0. After that, it was concentrated on a rotary evaporator until no solvent was evaporated, and the residue was transferred to a silica gel column, eluted with n-hexane/ethyl acetate/formic acid and dichloromethane/methanol/formic acid, and concentrated to obtain a purified crude product.
  • Dissolve compound 71 (8.2g, 7.6mmol) in DMF (80mL) solution, cool the reaction flask to 0-5°C with an ice-water bath, add 18 (5.2g, 7.6mmol) DMF solution (20mL), add slowly DIPEA (4mL, 22.8mmol) into the reaction flask. Control the dropping rate to keep the temperature of the reaction solution between 5-10°C during the entire dropping process. After the dropping is completed, remove the ice-water bath, allow the reaction to warm to room temperature, and stir for 1.5 hours. Concentrate, add dichloromethane (100mL), add formic acid under ice bath to adjust the pH to 3-4, concentrate, and purify on silica gel column.
  • the eluent is 20-100% ethyl acetate/n-hexane and 0-20% methanol/ Dichloromethane (each containing 0.1% formic acid), the crude product (11.44g) obtained by purification on a silica gel column was purified by preparative HPLC. The eluent was 20-30% acetonitrile/water (each containing 0.1% formic acid). Compound 72 (6.8 g, yield 60%) was obtained by drying. ESI m/z C 71 H 115 N 10 O 22 S[M+H] + : calculated value 1491.78, actual measured value 1492.01.
  • Dissolve compound 76 (16.7 g, 24.8 mmol) in 200 mL of DMF, cool in ice water, add DIPEA (12.9 g, 0.10 mol), keep the reaction solution at 10 degrees to 20 degrees, slowly add compound 53 (30 g, crude , Based on 24.8mmol). React at room temperature for 1 hour, concentrate, dilute with dichloromethane, wash with water, and extract the aqueous phase with dichloromethane. The organic phases were combined, washed with 0.2N hydrochloric acid, saturated brine, dried over anhydrous sodium sulfate, filtered, and concentrated. Purification by column chromatography (50%-100% ethyl acetate/petroleum ether, then 10% methanol/dichloromethane) gave 20 g of the product with a yield of 98%.
  • H 2 N-PEG 4 -CH 2 CH 2 CO 2 H (3.0g, 11.3mmol, 1.0eq.) and K 2 CO 3 (4.7g, 33.93mmol, 3.0eq.) were dissolved in 50mL In water and cool on an ice water bath.
  • BocHN-PEG 4 -CH 2 CH 2 CO 2 H (0.81 g, 2.22 mmol, 1.0 eq.)
  • K 2 CO 3 (0.92 g, 6.66 mmol, 3.0 eq.
  • NaI 0.033g, 0.222mmol, 0.1eq.
  • BnBr (0.57g, 3.33mmol, 1.5eq.) was added dropwise, the mixture was warmed to room temperature and stirred overnight.
  • H 2 N-PEG 4 -CH 2 CH 2 CO 2 H (2.8g, 10.4mmol, 1.0eq.) and K 2 CO 3 (4.3g, 31.2mmol, 3.0eq.) were dissolved in 40mL
  • a solution of compound 79 in tetrahydrofuran (40 mL) (3.8 g, 10.4 mmol, 1.0 eq.) was added dropwise and the mixture was warmed to room temperature and stirred overnight.
  • reaction mixture was adjusted to pH 4-5 with 1N KHSO 4 , extracted with dichloromethane (150 mL ⁇ 1, 100 mL ⁇ 2), washed with water (200 mL ⁇ 1) and brine (200 mL ⁇ 1), dried with anhydrous sodium sulfate, and concentrated. The residue was dissolved in a small amount of dichloromethane and loaded onto a silica gel column and eluted with 4-6% methanol/dichloromethane to obtain a colorless oil (5.18 g, 81% yield).
  • ESI m/z C 27 H 53 N 2 O 13 [M+H] + calculated value 613.3, actual measured value 613.3.
  • H 2 N-PEG 4 -CH 2 CH 2 CO 2 Bn (crude product from the previous step) was dissolved in 3 mL DMF, cooled on an ice/water bath, and DIPEA (0.78 g, 6.0 mmol, 4.0 eq. ), and then add compound 80 (0.93g, 1.5mmol, 1.0eq.) in DMF (7mL) solution and HATU (1.72g, 4.5mmol, 3.0eq.).
  • compound 82 (0.64 g, 1.05 mmol, 1.0 eq.) was stirred in a mixed solution of 5 mL dichloromethane and 2 mL TFA for 2 hours, and then concentrated. The residue was concentrated three times with dichloromethane and placed under a high vacuum pump. The above crude product was redissolved in 3 mL DMF and cooled on an ice water bath. To this was added compound 80 (0.64g, 1.05mmol, 1.0eq.) in DMF (7mL) solution, and then DIPEA (0.54g, 4.20mmol, 4.0eq.) and HATU (1.2g, 3.15mmol, 3.0eq.) ).
  • Raney-Ni (7.5 g, suspended in water) was washed with water (three times) and isopropanol (three times), and mixed with compound 103 (5.0 g, 16.5 mmol) in isopropanol.
  • the mixture was stirred under a hydrogen balloon at room temperature for 16 hours, then filtered through a pad of Celite, the pad was washed with isopropanol and the filtrate was concentrated, and purified by column chromatography (5-25% methanol/dichloromethane) to give a pale yellow oil (2.60 g, 57% yield).
  • HATU (0.50 g, 1.32 mmol) and triethylamine (0.06 mL, 1.32 mmol) were added to a dichloromethane (10 mL) solution of compound 111 (1.00 g, 1.32 mmol).
  • the reaction was stirred at 0°C for 30 minutes, then Z-Lys-OH (0.40 g, 1.43 mmol) was added, stirred at room temperature for 1 hour, then diluted with water (20 mL) and extracted with ethyl acetate (3 ⁇ 20 mL). The combined organic phase was washed with brine (30 mL), dried over anhydrous sodium sulfate, filtered and concentrated.
  • HMDS hexamethyldisilazane, 9.0 mL, 43.15 mmol
  • ZnCl 2 (16 mL, 1.0 M ether solution) were added.
  • the mixture was heated to 115-125°C and the toluene was collected through a Dean-Stark trap.
  • the reaction mixture was heated at 120°C for 6 hours. During this period, 2 ⁇ 40 mL of anhydrous toluene was added to maintain the volume of the mixture at about 50 mL. Then the mixture was cooled and 1 mL 1:10 HCl (concentrated)/methanol was added.
  • Example 150 General preparation method of conjugates prepared by reducing antibody disulfide bonds to Tubulysin derivatives.
  • the mixture was incubated at room temperature to 37.5°C for 2 to 18 hours, and then DHAA (135 ⁇ l, 50 mm) was added. After continuous incubation at RT overnight, the mixture is purified by G-25 column or cation chromatography column or anion chromatography column with 10-100 mM phosphoric acid, or citric acid, 50-200 mM NaCl pH 6-7.5 buffer solution to obtain the conjugate Compound (75% to 99% yield). This purification step can also be purified by dialysis and filtration, using 10-100 mM phosphoric acid, or citric acid, 50-200 mM NaCl, pH 6-7.5 buffer solution, to (3-30 times the volume of the dialyzed solution) to obtain the conjugate.
  • Example 151 Additional preparation method of conjugate.
  • Cell binding molecules can be coupled to the compounds of this patent through amide, thioether or disulfide bonds.
  • Dilute the antibody >5mg/mL) with PBS buffer (pH8.0) containing 50mM sodium borate, add dithiothreitol (final concentration of 10mM), and treat it at 35°C for 30 minutes.
  • the antibody can release free sulfhydryl groups.
  • Antibodies and Traut's reagent (2-iminothiophene) (Jue, R., et al. Biochem. 1978, 17(25): 5399-5405) can also release sulfhydryl groups, or under pH 7-8 conditions, and SATP (N -Succinimide-S-acetylthiopropionate) or N-succinimide-S-acetyl (thiotetraoxalic acid) (SAT(PEG)4) and other different linkers react, after Through the action of hydroxylamine, sulfhydryl groups are formed (Duncan, R, et al, Anal. Biochem. 1983, 132, 68-73, Fuji, N. et al, Chem. Pharm. Bull. 1985, 33, 362-367). Basically, 5-9 sulfhydryl molecules are attached to each antibody molecule.
  • each antibody molecule will connect 3.2-4.8 tubulysin derivatives on average.
  • linkers can be divided into dimethyl(phenyl)silyl (DMPS), SMDP, 4-succinimidyloxycarbonyl-methyl- ⁇ (2-pyridyldisulfide)toluene (SMPT), N-succinimide-4-(2-pyridinethio) valerate (SPP), N-succinimide-4-(2-pyridinethio) propionate (SPDP), N-succinimide -4-(2-pyridinethio)butyrate (SPDB), succinimidyl 4-(N-maleimidomethyl)cyclohexane-1-carboxylate (SMCC), N-hydroxy Succinimide-(polyethylene glycol) n-maleimide (SM(PEG)n) and so on.
  • DMPS dimethyl(phenyl)silyl
  • SMDP 4-succinimidyloxycarbonyl-methyl- ⁇ (2-pyridyldisulfide)toluene
  • the antibody (>5mg/mL) is diluted in a buffer (pH 6.5 ⁇ 7.5, 5mM PBS, 50mM NaCl, 1mM EDTA) and reacts with the linker for 2 hours, and the molar ratio of the linker to the antibody is more than 6-10 times.
  • the reaction mixture can be separated by Sephadex G25 gel chromatography, and its lower molecular weight molecules will be removed.
  • This purification step can also use a cation chromatography column or an anion chromatography column, using 10-100mM phosphoric acid, or citric acid, 50-200mM NaCl pH 6-7.5 buffer solution to obtain the conjugate compound (75% to 99% yield rate).
  • This purification step can also use dialysis and filtration, using 10-100mM phosphoric acid, or citric acid, 50-200mM NaCl pH 6-7.5 buffer solution, to (3-30 times the volume of the dialyzed solution), to obtain the conjugate Compound (75% to 99% yield).
  • concentration of the antibody is determined by spectrophotometry, and the linker contains a pyridinethio group.
  • the extinction coefficient of the antibody at 280nm is 2067550M -1 cm -1 . After the modified antibody was treated with an excess of dithiothreitol (20 times equivalent), the released 2-thiopyridine group was measured, and the extinction coefficients at 343 and 280 nm were 8080 and 5100 M -1 cm -1 ).
  • 1.2-1.5 equivalents of Tubulysin derivative molecules with sulfhydryl groups are added to the modified antibody.
  • the reaction is carried out at room temperature for 5 to 18 hours.
  • the reaction mixture is passed through Sephadex G25 gel chromatography to remove unlinked drugs or other low molecular weight substances. Then the concentration of the ligation product was determined by measuring the absorbance at 280nm and 252nm.
  • the ligation product is in the form of a monomer, with an average of 3.2-4.8 drug molecules attached to each antibody molecule.
  • Example 152 In vitro cytotoxicity evaluation of Her2 antibody conjugates C-37, C-59, C-72, C-123 and C-134 (compared with T-DM1):
  • the cell line used for cytotoxicity assay was human gastric cancer cell line NCI-N87; cells were grown in RPMI-1640 containing 10% FBS. During the measurement, cells (180 ⁇ L, 6000 cells) were added to each well of a 96-well plate and incubated at 37° C., 5% CO 2 for 24 hours. Next, in a suitable cell culture medium (total volume, 0.2 mL), the cells are treated with different concentrations of test compound (20 ⁇ L), and the control wells contain cells and culture medium but no test compound. The plate was incubated at 37°C and 5% CO 2 for 120 hours, then MTT (5 mg/ml, 20 ⁇ L) was added to the well, and the plate was incubated at 37°C for 1.5 hours.
  • Example 153 In vivo anti-tumor activity study on BALB/c nude mice bearing NCI-N87 xenograft tumors.
  • mice Five-week-old female BALB/c nude mice (66 animals) were subcutaneously inoculated with N-87 cancer cells (5 ⁇ 10 6 cells/mouse) in 0.1 mL of serum-free medium in the right shoulder area. The tumor grew to an average size of 140mm 3 after 8 days of growth. The animals were then randomly divided into 10 groups (6 animals in each group). The first group of mice served as a control group and was injected with phosphate buffered saline.
  • Example 154 Toxicity study of Tubulysin B derivative Her2 antibody conjugate (compared with T-DM1).
  • the weight change (usually decrease) of an animal is a macroscopic reflection of its toxicity to drugs.
  • Fifty-six female ICR mice aged 6-7 weeks were divided into 7 groups, each group contained 8 animals, and the drugs C-37, C-49, C-72, and C-123 were given intravenously at a dose of 150 mg/kg. , C-134 and T-DM1.
  • the control group (8 animals) was given PBS buffered saline.
  • the body weight of the mice in the control group and all conjugate groups except T-DM1 did not decrease by more than 5%.

Abstract

涉及含支链(侧链)连接体偶联Tubulysin衍生物(同系物)与细胞结合分子,产生的偶联物具有更好的药代动力学性质、从而能更精确地靶向杀死异常细胞。还涉及Tubulysin 同系物与细胞结合剂的偶联及其所含分子的合成方法,以及使用该偶联物靶向治疗癌症、感染和自身免疫疾病的方法。含长支链连接体的Tubulysin偶联物在靶向传送期间半表期增加,在血液循环中对非靶细胞、组织或器官的暴露最小,导致脱靶毒性降低。

Description

一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法 技术领域
本发明涉及以含支链(侧链)连接体偶联Tubulysin衍生物(同系物)与细胞结合分子,产生的偶联物具有更好的药代动力学性质、从而能更精确地靶向杀死异常细胞。本发明还涉及Tubulysin同系物与细胞结合剂的及其所含分子的合成方法,以及使用该偶联物靶向治疗癌症、感染和自身免疫疾病的方法。
背景技术
Adcetris治疗复发或难治性霍奇金淋巴瘤的临床成功(Okeley,N.等Hematol Oncol.Clin.North.Am,2014,28,13-25;Gopal,A.,等,Blood 2015,125,1236-43)和Kadcyla用于治疗复发的HER2阳性乳腺癌的成功(Peddi,P.,Hurvitz,S.,Ther.Adv.Med.Oncol.2014,6(5),202–9;Lambert,J.和Chari,R.,J.Med.Chem.2014,57,6949-64)证明了抗体药物偶联物(ADC)是一种非常有前景的癌症靶向治疗方法。ADC成功的重要因素包括其中的三个重要组成部分,即单克隆抗体、细胞毒性分子和连接体,以及连接体-细胞毒性分子的连接位点(L.Ducry和B Stump,Bioconjugate Chem.,2010,21,5–13;GS Hamilton,Biologicals 2015,43,318-32)。对于ADC各组分的研究已持续有三十年的历史。连接体需要满足可以和药物上特定的反应性官能团反应,在人体血液循环系统具有稳定性,并且在与抗原结合和细胞内吞后要容易释放药物,尤其重要的是连接体-细胞毒性分子一旦在血液循环中脱靶后不能损害正常组织,现有的连接技术仍然有限(Ponte,J.等,Bioconj.Chem.,2016,27(7),1588-98;Dovgan,I.等Sci.Rep.2016,6,30835;Ross,P.L.和Wolfe,J.L.,J Pharm.Sci.105(2),391-7;Chen,T.等J.Pharm.Biomed.Anal.,2016,117,304-10)。
早期ADC主要用于靶向治疗液体肿瘤,使用的连接体很不稳定,游离药物在血液循环中被释放和进而出现脱靶毒性(Bander N.H.等,Clin.Adv.Hematol.Oncol.,2012,10,1-16)。当前一代ADC上的连接体更稳定,细胞毒 性剂地活性也更高(Behrens,C.R.和Liu,B.,mAbs,2014.6,46-53)。然而,脱靶毒性目前仍然是ADC药物开发中的主要挑战之一(Roberts,S.A.等,Regul.Toxicol.Pharmacol.2013,67,382-91)。例如在临床实践中T-DM1
Figure PCTCN2019093946-appb-000001
使用了稳定的(不可切割的)MCC连接体,它对HER2阳性患有转移性乳腺癌(mBC)或已经接受了相应治疗或在辅助治疗的六个月内HER2肿瘤复发的患者有很大益处(Peddi,P.和Hurvitz,S.,Ther.Adv.Med.Oncol.2014,6(5),202-209;Piwko C.等,Clin Drug Investig.2015,35(8),487-93;Lambert,J.和Chari,R.,J.Med.Chem.2014,57,6949-64)。但是,在临床试验中T-DM1作为HER2阳性无法切除的局部晚期或转移性乳腺癌患者的一线治疗药物已经失败,作为HER2阳性晚期胃癌的二线治疗药物,与其产生的毒副作用相比,它对患者的益处不大(Ellis,PA,等,J.Clin.Oncol.2015,33(2015ASCO会议摘要507);Shen,K.等,Sci Rep.2016,6,23262;de Goeij,B.E.和Lambert,J.M.Curr Opin Immunol 2016,40,14-23;Barrios,C.H.等,J Clin Oncol 2016,34,(2016ASCO会议摘要593)。
为了解决脱靶毒性的问题,ADC化学研发的一个方向就是,将连接体-细胞毒性剂组分和偶联化学扩展,而不仅仅应用单个的细胞毒性剂,同时还解决了ADC的连接体-细胞毒性剂对目标疾病的活性问题(Lambert,JM Ther Deliv 2016,7,279-82;Zhao,RY等,2011,J.Med.Chem.54,3606-23)。许多药物的研发者和学术机构都集中力量来开发新型的可靠的特异偶联连接体,定点ADC偶联方法,似乎这些ADC的循环半衰期更长,疗效更高,能降低脱靶毒性,ADC具有窄的体内药代动力学(PK)特性,生产工艺的批间一致性更好(Hamblett,K.J.等Clin.Cancer Res.2004,10,7063-70;Adem,Y.T.等,Bioconjugate Chem.2014,25,656-664;Boylan,N.J.Bioconjugate Chem.2013,24,1008–1016;Strop,P.等,Chem.Biol.2013,20,161-67;Wakankar,A.mAbs,2011,3,161–172)。已见报导的这些定点偶联方法包括:抗体中引入经改造的半胱氨酸(Junutula,J.R.等,Nat.Biotechnol.2008,26,925–32;Junutula,JR,等2010Clin.Cancer Res.16,4769;US专利8,309,300;7,855,275;7,521,541;7,723,485,WO2008/141044),硒代半胱氨酸(Hofer,T.等,Biochemistry 2009,48,12047–57;Li,X.等,Methods 2014,65,133–8;US专利8,916,159),含全氟芳香族试剂标签的半胱氨酸(Zhang,C.等,Nat.Chem.2015,8,1–9),硫代海藻糖(Okeley,N.M.等,Bioconjugate Chem.2013, 24,1650),非天然氨基酸(Axup,J.Y.等,Proc.Nat.Acad.Sci.USA.2012,109,16101-6;Zimmerman,E.S.等,2014,Bioconjug.Chem.25,351–361;Wu,P.,等,2009 Proc.Natl.Acad.Sci.106,3000-5;Rabuka,D.,等,Nat.Protoc.2012,7,1052-67;美国专利8,778,631和美国专利申请号.20100184135,WO2010/081110;WO2006/069246,2007/059312;美国专利7,332,571,7,696,312和7,638,299;WO2007/130453,美国专利7,632,492和7,829,659);分子间双硫键被还原后通过下列基团再成桥:二溴马来酰胺(Jones,MW等.J.Am.Chem.Soc.2012,134,1847–52),双砜试剂(Badescu,G.等.Bioconjug.Chem.2014,25,1124–36;WO2013/190272,WO2014/064424)和双溴哒嗪二酮(Maruani,A.等.Nat.Commun.2015,6,6645);半乳糖和唾液酸转移酶(Zhou,Q.等.Bioconjug.Chem.2014,25,510–520;美国专利申请号20140294867 for Sanofi-Genzyme),甲酰甘氨酸生成酶(FGE)(Drake,PM等.Bioconj.Chem.2014,25,1331–41;Carrico,IS等美国专利7,985,783;8,097,701;8,349,910,和美国专利申请号20140141025,20100210543),磷酸泛酰巯基乙胺基转移酶(PPTases)(Grünewald,J.等.Bioconjug.Chem.2015,26,2554–62),分选酶A(Beerli,RR,等.PLoS One 2015,10,e0131177),用茂原链轮丝菌转谷氨酰胺酶引入的谷氨酰胺标签(mTG)(Strop,P.,Bioconj.Chem.,2014,25,855–62;Strop,P.,等.,Chem.Biol.2013,20,161–7;美国专利8,871,908),或是用微生物转谷氨酰胺酶(MTGase)引入的谷氨酰胺标签(Dennler,P.,等,2014,Bioconjug.Chem.25,569–78;Siegmund,V.等.Angew.Chemie-Int.Ed.2015,54,13420–4;美国专利申请号20130189287;美国专利7,893,019),通过酶或细菌在蛋白质主链外形成异肽键-肽键(Kang,HJ,等.Science 2007,318,1625–8;Zakeri,B.等.Proc.Natl.Acad.Sci.USA 2012,109,E690–7;Zakeri,B.&Howarth,MJ Am.Chem.Soc.2010,132,4526–7)。
我们已经公开了几种偶联方法,重新桥接天然抗体链间二硫化物被还原产生的一对硫醇,例如使用溴马来酰亚胺和二溴马来酰亚胺连接体(WO2014/009774),2,3-二取代琥珀酸/2-单取代/2,3-二取代富马酸或马来酸连接体(WO2015/155753,WO20160596228),乙炔二羧基连接体(WO2015/151080,WO20160596228)或肼连接体(WO2015/151081)。相较于与抗体上的半胱氨酸或赖氨酸残基偶联的传统非选择性方法相比,用这些 连接体和偶联方法制备的ADC的治疗窗口更大。在此我们公开一种含有长支链连接体的Tubulysin偶联物。长支链连接体可以防止抗体药物偶联物被水解酶如蛋白酶或酯酶等水解,使偶联物在循环系统中更加稳定。
Tubulysin是一类高效的细胞毒性剂,它在本领域广为熟知,可以基于已知方法从天然产物中分离或通过有机合成的方法制备(如Balasubramanian,R.,等.J.Med.Chem.,2009,52,238–40;Wipf,P.,等.Org.Lett.,2004,6,4057–60;Pando,O.,等.J.Am.Chem.Soc.,2011,133,7692–5;Reddy,J.A.,等.Mol.Pharmaceutics,2009,6,1518–25;Raghavan,B.,等.J.Med.Chem.,2008,51,1530–33;Patterson,A.W.,等.J.Org.Chem.,2008,73,4362–9;Pando,O.,等.Org.Lett.,2009,11(24),5567–9;Wipf,P.,等.Org.Lett.,2007,9(8),1605–7;Friestad,G.K.,Org.Lett.,2004,6,3249–52;Peltier,H.M.,等.J.Am.Chem.Soc.,2006,128,16018–9;Chanrasekhar,S.,等J.Org.Chem.,2009,74,9531–4;Liu,Y.,等.Mol.Pharmaceutics,2012,9,168–75;Friestad,G.K.,等.Org.Lett.,2009,11,1095–8;Kubicek,K.,等.,Angew Chem Int Ed Engl,2010.49:4809-12;Chai,Y.,等.,Chem Biol,2010,17:296-309;Ullrich,A.,等.,Angew Chem Int Ed Engl,2009,48,4422-5;Sani,M.,等.Angew Chem Int Ed Engl,2007,46,3526-9;Domling,A.,等.,Angew Chem Int Ed Engl,2006,45,7235-9;Zanda,M.,等,Can.Pat.Appl.CA 2710693(2011);Chai,Y.,等.Eur.Pat.Appl.2174947(2010),WO 2010034724;Leamon,C.等,WO2010033733,WO 2009002993;Ellman,J.,等,PCT WO2009134279;WO 2009012958;美国专利申请20110263650,20110021568;Matschiner,G.,等,WO2009095447;Vlahov,I.,等,WO2009055562,WO 2008112873;Low,P.,等,WO2009026177;Richter,W.,WO2008138561;Kjems,J.,等,WO 2008125116;Davis,M.;等,WO2008076333;Diener,J.;等,美国专利申请20070041901,WO2006096754;Matschiner,G.,等,WO2006056464;Vaghefi,F.,等,WO2006033913;Doemling,A.,Ger.Offen.DE102004030227,WO2004005327,WO2004005326,WO2004005269;Stanton,M.,等,美国专利申请20040249130;Hoefle,G.,等,Ger.Offen.DE10254439,DE10241152,DE10008089;Leung,D.,等,WO2002077036;Reichenbach,H.,等,Ger.Offen.DE19638870;Wolfgang,R.,US20120129779;Chen,H.,美国专利申请 20110027274。我们先前公开了用于靶向治疗癌症,感染和自身免疫疾病的Tubulysin偶联物(PCT/IB2012/053554)的构建。本发明中的含有长支链连接体的Tubulysin偶联物在靶向传送期间半衰期增加,在血液循环中对非靶细胞、组织或器官的暴露最小,导致脱靶毒性降低。
发明内容
本发明涉及以含支链(侧链)连接体偶联Tubulysin同系物与细胞结合分子,产生的偶联物具有更好的药代动力学性质、从而能更精确地靶向杀死异常细胞。本发明还涉及Tubulysin同系物与细胞结合剂的偶联及其所含分子的合成方法,以及使用该偶联物靶向治疗癌症、感染和自身免疫疾病的方法。
在一个方面,本发明涉及一种抗体-Tubulysin B衍生物偶联物,其特征在于,所述偶联物具有如下式(I)所示的结构:
Figure PCTCN2019093946-appb-000002
或者以具有式I所示结构为母体的药学上可接受的盐、水合物或水合盐,具有式I所示结构的多晶型物,式I所示结构的光学异构体,式I所示结构的一个或多个氘( 2H)原子代替氢( 1H)原子,或式I所示结构的一个或多个 13C原子代替 12C原子;
其中,P 1是H、COCH 3、COH、PO(OH) 2、CH 2OPO(OH) 2、SO 2CH 3、C 6H 11O 5(糖苷)、CONHCH 3、CON(CH 3) 2、CON(CH 2CH 2) 2NCH 3、CON(CH 2CH 3) 2或CON(CH 2CH 2) 2CHN(CH 2CH 2) 2CH 2
R 1、R 2、R 3和R 4分别独立地是H、C 1-C 6烷烃基、C 1-C 6烯烃基、C 1-C 6烷醚基(R 1OR 2)、C 1-C 6烷羰基(R 1COR 2)、C 1-C 6烷酯基(R 1COOR 2)、C 1-C 6烷羧基((R 1COOH)或C 1-C 6烷酰胺基((R 1CONHR 2);
或者,R 1和R 2一起,R 1和R 3一起,R 2和R 3一起,或R 3和R 4一起构成C 2-C 7杂环基或C 2-C 7环烷基结构;
R 5是H、O-C 1-C 6烷烃基、C(O)-H、C(O)-C 1-C 6(直链或支链)烷烃基、C(O)-NH-C 1-C 6(直链或支链)烷基或C(O)-N(C 1-C 6(直链或支链)烷基) 2
R 6、R 7和R 8分别独立地是H、C 1-C 6烷烃基、C 1-C 6烷醚基(R 1OR 2)、C 1-C 6烷羰基(R 1COR 2)、C 1-C 6烷酯基(R 1COOR 2)、C 1-C 6烷羧基((R 1COOH)或C 1-C 6烷酰胺基((R 1CONHR 2);优选R 6、R 7和R 8分别独立地是H或CH 3
mAb是抗体、抗体片段、单克隆抗体、多克隆抗体、纳米抗体、前药抗体(probody),或者用合成分子或蛋白进行修饰过的抗体及抗体片段;
L是含有亲水支链的连接体,其主要构架是C 2-C 100的肽单元(1~12个天然或非天然氨基酸)、腙键基团、二硫化物基团、酯基团、肟基团、酰胺基团或硫醚键基团构成。
在一个方面,在本发明的偶联物中,L的结构为:
Figure PCTCN2019093946-appb-000003
其中Aa是L-或D-天然或非天然氨基酸;
r是0-12之间的整数;当r不是0时,(Aa) r是由相同或不同的氨基酸构成的肽单元;
m 1=1-18之间的整数;m 2=1-100之间的整数;m 3=1-8之间的整数;m 4=0-8之间的整数;m 5=1-8之间的整数;
Y是NHC(=O)、NHS(O 2)、NH(SO)、NHS(O 2)NH、NHP(O)(OH)NH或C(O)NH;
R 9是H、(O=)CR 1、(O=)CNHR 1、R 1COOH、R 1(COCH 2NH) m2H、R 1(Aa) r或R 1(COCH 2NCH 3) m2H,以及
R 1、m 2和(Aa) r定义如权利要求1和上述定义所述。
此外,所述细胞表面受体结合分子mAb可以为任何形式的细胞结合体,包括肽或类似肽的结构:抗体,单链抗体,可以与目标细胞结合的抗体片段,单克隆抗体,单链单克隆抗体,可以与目标细胞结合的单克隆抗体片段,嵌合抗体,可以与目标细胞结合的嵌合抗体片段,功能区抗体,可以与目标细胞结合的功能区抗体片段,仿抗体的基因工程蛋白质,纤连蛋白结合体adnectin,预设计锚蛋白重复蛋白(DARPin),淋巴因子,激素,维他命,生长因子,集落刺激因子,营养传送分子,转铁蛋白,细胞表面小分子配体,或连接有细胞结合体的白蛋白、高分子、 或树枝高分子,表面含有细胞结合分子(结合肽、蛋白、抗体或细胞表面小分子配体)的高分子材料、蛋白、脂质体、纳米颗粒、泡馕或(病毒)微囊。
细胞结合分子-Tubulysin B衍生物偶联物的制备
在一具体实施例中,所述细胞结合分子-Tubulysin B衍生物偶联物的合成
包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000004
其中结构式(II)中的P 1
Figure PCTCN2019093946-appb-000005
R 1、R 2、R 3、R 4、R 5、R 6、R 7和R 8以及mAb如结构式(I)中所述;
L’的结构为(II-0)和(II-00):
Figure PCTCN2019093946-appb-000006
其中m 1、m 2、m 3、m 4、m 5、Aa、r、Y和R 9如结构式(I)所述。
具体理想的L’的结构是:
Figure PCTCN2019093946-appb-000007
Figure PCTCN2019093946-appb-000008
其中m 1、m 2、m 3、m 4、m 5、Aa、r和R 9如权利要求1-2中任一项所述。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,进行偶连的mAb-SH的制备方法包括以下a)~c)中的任一个:
a).由还原剂(优选,三(2-羧基乙基)膦(TCEP)、二硫苏糖醇(DTT)、二硫季戊四醇(DTE)、L-谷胱甘肽(GSH)、2-巯基乙胺(β-MEA)或/和β-巯基乙醇(β-ME,2-ME))还原抗体、抗体片段、单克隆抗体、多克隆抗体、纳米抗 体、前药抗体(probody)或用合成分子或蛋白进行修饰过的抗体及抗体片段重轻链间、重重链间或自身链间的二硫键来获取;
b).通过Traut试剂或硫内酯与抗体分子的胺反应形成硫醇来制备:
Figure PCTCN2019093946-appb-000009
c).在缓冲体系条件下,通过生化反应,在抗体上引入较易还原的二硫键基团,然后用TCEP、DTT、GSH、β-MEA、β-ME还原:
Figure PCTCN2019093946-appb-000010
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,偶联物的合成中使用的缓冲体系是:pH 5.0~9.5,浓度为1mM~1000mM的磷酸、醋酸、柠檬酸、硼酸、碳酸、巴比妥酸、Tris(三羟甲基氨基甲烷)、苯甲酸或三乙醇胺、或它们的混合物缓冲溶液,并含有体积比为0%~35%的水溶性有机溶剂:甲醇、乙醇、正丙醇、异丙醇、正丁醇、异丁醇、乙腈、丙酮、DMF、DMA或DMSO,偶联反应温度控制在0℃~45℃,偶联反应是5分钟~96小时。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,在偶联反应完成后,采用超滤或柱层析法进行纯化获得结构式为(I)的偶联物。常用的柱层析包括分子筛柱、阳离子柱、阴离子柱、疏水(HIC)柱、反相柱 或者蛋白A或G亲和柱。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)的合成由结构式为(III)的Tubulysin B衍生物和结构式为(L’)的化合物缩合反应获得:
Figure PCTCN2019093946-appb-000011
其中X是OH、卤素(F、Cl、Br、或I)、苯酚、五氯酚、三氟甲基磺酸、咪唑、二氯苯酚、四氯苯酚、1-羟基苯并三氮唑、对甲苯磺酸、甲磺酸、2-乙基-5-苯基异噁唑-3'-磺酸、
Figure PCTCN2019093946-appb-000012
Figure PCTCN2019093946-appb-000013
Figure PCTCN2019093946-appb-000014
自我酸酐或与其他酸酐如乙酰酐、甲酸酐形成的酸酐;或多肽缩合反应中间体或Mitsunobu反应中 间体;
其中,缩合反应是在含有体积比为1%-100%的吡啶、三乙胺或二异丙基乙胺的有机溶剂二氯甲烷、二氯乙烷、DMF、DMA、四氢呋喃(THF)、DMSO、丙酮、异丙醇、正丁醇或乙腈中,或上面二种或三种以上溶剂的混合溶剂,或有或无惰性气体(氮气、氩气、氦气)保护下,温度控制在-20℃–150℃条件下,反应时长5分钟–120小时完成;
或者,缩合反应在如下缓冲体系及如下条件下进行,所述缓冲体系是:pH 5.0~9.5,浓度为1mM~1000mM的磷酸、醋酸、柠檬酸、硼酸、碳酸、巴比妥酸、Tris(三羟甲基氨基甲烷)、苯甲酸或三乙醇胺、或它们的混合物缓冲溶液,并含有体积比为0%~35%的水溶性有机溶剂:甲醇、乙醇、正丙醇、异丙醇、正丁醇、异丁醇、乙腈、丙酮、DMF、DMA或DMSO,偶联反应温度控制在0℃~45℃,偶联反应是5分钟~96小时。
另外,结构式(III)中的NH 2基团理想地是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸、磺酸盐形式进行缩合反应。
当X是OH时,上述缩合反应需借助缩合试剂,通常缩合试剂可选择地是:1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(EDC),二环己基碳二亚胺(DCC),N,N'-二异丙基碳二酰亚胺(DIC),1-环已基-2-吗啉乙基碳二亚胺对甲苯磺酸盐(CMC或CME-CDI),羰基二咪唑(CDI),O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸(TBTU),O-苯并三氮唑-四甲基脲六氟磷酸酯(HBTU),苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐(BOP),六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷(PyBOP),焦碳酸二乙酯(DEPC),N,N,N',N'-四甲基氯甲脒六氟磷酸盐,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(HATU),1-[(二甲胺)(吗啉基)亚甲基]-1[1,2,3]三唑并[4,5-b]1-吡啶-3-氧六氟磷酸盐(HDMA),2-氯-1,3-二甲基咪唑鎓六氟磷酸盐(CIP),氯代三吡咯烷基鏻六氟磷酸盐(PyCloP),双(四亚甲基)氟代甲酰胺(BTFFH),N,N,N',N'-四甲基-硫-(1-氧代-2-吡啶基)硫脲鎓六氟磷酸盐,2-(2-吡啶酮-1-基)-1,1,3,3-四甲基脲四氟硼酸盐(TPTU),硫-(1-氧代-2-吡啶基)-N,N,N',N'-四甲基硫脲六氟磷酸盐,O-[(乙氧基羰基)氰基甲胺]-N,N,N',N'-四甲基硫脲六氟磷酸盐(HOTU),(1-氰基-2-乙氧基-2-氧代亚乙基氨基氧基)二甲基氨基-吗啉-碳鎓六氟磷酸盐(COMU),(苯并三氮唑-1-基氧基)二吡咯烷碳六氟磷酸盐 (HBPyU),N-苄基-N′-环己基碳二亚胺(或荷载在聚合物上),二吡咯烷基(N-琥珀酰亚氨氧基)碳鎓六氟磷酸盐(HSPyU),1-(氯-1-吡咯烷基亚甲基)吡咯烷六氟磷酸盐(PyClU),2-氯-1,3-二甲基咪唑四氟硼酸盐(CIB),(苯并三氮唑-1-基氧基)二哌啶碳六氟磷酸盐(HBPipU),6-氯苯并三氮唑-1,1,3,3-四甲基脲四氟硼酸酯(TCTU),溴化三(二甲基氨基)膦六氟磷酸(BrOP),1-正丙基磷酸酐(PPACA,
Figure PCTCN2019093946-appb-000015
),2-异氰基乙基吗啉(MEI),N,N,N',N'-四甲基脲-氧-(N-琥珀酸亚胺基)六氟磷酸盐(HSTU),2-溴-1-乙基吡啶四氟硼酸盐(BEP),氧-[(乙氧基羰基)氰基甲胺]-N,N,N',N'-四甲基硫尿四氟硼酸盐(TOTU),4-(4,6-二甲氧基三嗪-2-基)-4-甲基吗啉盐酸盐(MMTM,DMTMM),2-琥珀酰亚胺基-1,1,3,3-四甲基脲四氟硼酸酯(TSTU),N,N,N',N'-四甲基-O-(3,4-二氢-4-氧代-1,2,3-苯并三嗪-3-基)脲四氟硼酸盐(TDBTU),偶氮二甲酰二哌啶(ADD),双(4-氯苄基)偶氮二甲酸酯(DCAD),偶氮二甲酸二叔丁酯(DBAD),偶氮二甲酸二异丙酯(DIAD)或偶氮二甲酸二乙酯(DEAD)。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(III)的Tubulysin B衍生物的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000016
Figure PCTCN2019093946-appb-000017
其中R 5’是H、C 1-C 6烷烃基、C 1-C 6烷烃基、或C 1-C 6(直链或支链)胺烷基;其他基团的定义如前述。
理想条件下,结构式(III)的Tubulysin B衍生物的合成包括下列步骤中的一种或多种:
步骤1.将二乙氧基乙腈与硫化铵的水溶液在室温搅拌,得到化合物1即2,2-二乙氧基硫代乙酰胺;
Figure PCTCN2019093946-appb-000018
步骤2.将化合物1与溴丙酮酸酯在无水溶剂(如无水四氢呋喃、二氯甲烷、乙腈、N,N-二甲基甲酰胺、甲醇、异丙醇)中加热,缩合得化合物2;
Figure PCTCN2019093946-appb-000019
步骤3.将化合物2溶解在溶剂(如四氢呋喃、二氯甲烷、乙酸乙酯、正庚烷、二氧六环、乙腈)中,用路易斯酸或质子酸(包括盐酸、硫酸、磷酸、甲磺酸、甲酸、草酸、乙酸、对甲苯磺酸、对甲苯磺酸吡啶、AlCl 3、FeCl 3、 ZnCl 2、BF 3、BCl 3、BBr 3、TiCl 4、ZnBr 2、LiBF 4)水解,得到化合物3;
Figure PCTCN2019093946-appb-000020
步骤4.不饱和亚磺酰胺在低温条件下(如-45℃至-78℃)被正丁基锂等碱脱氢,然后在路易斯酸存在的条件下与化合物3发生加成反应,得到化合物4;
Figure PCTCN2019093946-appb-000021
路易斯酸选择为包括盐酸、硫酸、磷酸、甲磺酸、甲酸、草酸、乙酸、对甲苯磺酸、对甲苯磺酸吡啶、AlCl 3、FeCl 3、ZnCl 2、BF 3、BCl 3、BBr 3、TiCl 4、ZnBr 2、LiBF 4
步骤5.化合物4在低温条件(如-45℃至-78℃)被下被还原剂(如NaBH 4、LiBH 4、Na(OAc) 3BH、Na(CN)BH 3等)选择性地还原,通过加入路易斯酸(如Ti(Oet)4),控制其立体化学,得到化合物5;
Figure PCTCN2019093946-appb-000022
步骤6.将化合物5溶解在溶剂(如甲醇、乙醇、异丙醇、四氢呋喃、乙腈)中,被盐酸、硫酸和磷酸等酸脱去叔丁基亚磺酰基,得到化合物6;
Figure PCTCN2019093946-appb-000023
步骤7.将化合物6和叠氮酸溶解在溶剂(如正庚烷、四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺)中,在缩合试剂(如DIC/HOBt、DCC/HOBt、EDC/HOBt、HATU、BOP、T3P、BrOP)存在下,或通过缩合反应路线,发生缩合反应,得到化合物7;
或者,叠氮酸与氯甲酸异丁酯在有机碱(如三乙胺、二异丙基乙胺、N-甲基吗啡啉等)存在时在THF中反应,得到混合酸酐,再和化合物6的盐酸盐发生缩合,得到化合物7;
或者,叠氮酸在溶剂(如正庚烷、正己烷、二氯甲烷、四氢呋喃)中与草酰氯、三乙胺和催化剂量的DMF反应,被转换成酰氯,再和化合物6的盐酸盐发生缩合,得到化合物7;
Figure PCTCN2019093946-appb-000024
步骤8.在溶剂(如二氯甲烷、四氢呋喃、乙腈)中,化合物7上的羟基与羟基保护试剂(如TESCl),在有机碱(如咪唑、三乙胺、吡啶)的作用下得到化合物8;
Figure PCTCN2019093946-appb-000025
步骤9.化合物8被溶解在溶剂(如四氢呋喃、二氯甲烷、乙腈)中,在碱(如KHMDS,LiHMDS,NaHMDS,KOtBu,NaH,KH)的作用下,酰胺发生去质子化,然后与碘甲烷、溴甲烷、硫酸二甲酯、三氟甲磺酸甲酯或碘乙烷等发生烷基化反应,得到化合物9;
Figure PCTCN2019093946-appb-000026
步骤10.化合物9被溶解在溶剂(如四氢呋喃、二氯甲烷、乙酸乙酯)中,其中的叠氮基在一定的条件下,如在氢气和钯碳催化剂、三苯基膦和水(Staudinger反应)的存在下,被还原成氨基,然后与酸或者具有反应活性的酸衍生物发生缩合,得到化合物10;
Figure PCTCN2019093946-appb-000027
步骤11.化合物10中的羟基保护基PG 1在适当的条件(如TES保护基团可以在盐酸、THF/MeOH/AcOH、nBu 4NF或吡啶氢氟酸盐的THF溶液中被脱保护)下被脱保护,得到化合物11;
Figure PCTCN2019093946-appb-000028
步骤12.化合物11中的酯基在碱(如LiOH、NaOH、KOH)的作用下,或其他适当条件(如甲酯可以在LiCl、LiI、Me3SiOK等试剂的作用下被转化成羧酸)下被转化成酸化合物12;
Figure PCTCN2019093946-appb-000029
步骤13.在碱(如三乙胺、N,N-二异丙基乙胺、吡啶)和催化剂(如DMAP)存在时,一定的温度条件(如0℃至23℃)下,化合物12与乙酸酐,丙酸酐、异丙酸酐等酸酐、乙酰卤、丙酸卤、丙酸卤、甲酰胺卤、异酰胺卤、二甲酰胺卤等酸卤作用,得到化合物13,该反应也可不使用碱或者催化剂;
Figure PCTCN2019093946-appb-000030
步骤14.化合物13与适当的含羟基的化合物如五氟苯酚或N-羟基琥珀酰亚胺在缩合试剂(如EDC、DIC、DCC、HATU、HBTU)存在时,发生缩合反应,得到具有反应活性的酯化合物14;
Figure PCTCN2019093946-appb-000031
步骤15.化合物15与化合物14,在水相、一定的pH值条件(如pH=5.0-8.0)下,或者有机相、存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物16;反应也任选不使用任何碱,条件是需要控制一定的反应温度(如0℃至23℃)和反应时间(如30分钟至18小时);
Figure PCTCN2019093946-appb-000032
步骤16.化合物16中的硝基在还原条件下,如在氢气和钯碳催化剂、水合肼和FeCl 3、铁粉和醋酸等的存在的条件下,被还原成氨基,得到化合物III;
Figure PCTCN2019093946-appb-000033
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000034
Figure PCTCN2019093946-appb-000035
理想条件下,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
步骤1.化合物1-1和化合物1-2在缩合剂(如EDC、HATU、DIC、DCC)的作用下缩合,或者经过缩合反应路线发生缩合反应(如化合物1-2在缩合试剂如DIC和EDC的作用下与五氟苯酚、硝基苯酚或N-羟基琥珀酰亚胺发生缩合反应,生产相应的活性酯,然后再与化合物1-1反应),得到化合物物1a;
或者,化合物1-3和化合物1-4在缩合剂(如EDC、HATU、DIC、DCC)的作用下,或者经过缩合反应路线发生缩合反应,得到化合物物1b;
Figure PCTCN2019093946-appb-000036
步骤2.化合物1中的羧基保护基PG 2在脱保护试剂作用下(如叔丁酯基在酸的作用下),被去除,得到化合物2;
Figure PCTCN2019093946-appb-000037
步骤3.含羧基的化合物2和含氨基的化合物3在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化 合物4;
Figure PCTCN2019093946-appb-000038
步骤4.化合物4上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
Figure PCTCN2019093946-appb-000039
步骤5.含羧基化合物6和含氨基的化合物5在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物7;
Figure PCTCN2019093946-appb-000040
步骤6.化合物7上的羧基保护基团PG 3在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除),得到化合物8;
Figure PCTCN2019093946-appb-000041
步骤7.化合物8与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯化合物,或与其他酸活化基团反应得到具有缩合反应活性的化合物L’;
Figure PCTCN2019093946-appb-000042
16.如权利要求9-13中任一项所述的偶联物,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000043
理想条件下,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
步骤1.化合物1上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物2;
Figure PCTCN2019093946-appb-000044
步骤2.含氨基的化合物2和含羧基的化合物3在缩合试剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物4;
Figure PCTCN2019093946-appb-000045
步骤3.化合物4上的羧基保护基团PG 2在去保护条件下被去除,如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物5;
Figure PCTCN2019093946-appb-000046
步骤4.含羧基化合物5和含氨基的化合物6在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物7;
Figure PCTCN2019093946-appb-000047
步骤5.化合物7上的羧基保护基团PG 3在去保护条件下被去除,如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物8;
Figure PCTCN2019093946-appb-000048
步骤6.化合物8与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯化合物,或与其他酸活化基团反应得到具有缩合反应活性的化合物9;
Figure PCTCN2019093946-appb-000049
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)化合物的合成由结构式(IV)和结构式(V)缩合反应获得:
Figure PCTCN2019093946-appb-000050
其中X定义及缩合反应条件如前面所述;
结构式(V)中的NH 2基团优选地是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(IV)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000051
理想条件下,结构式(IV)的合成包括下列步骤中的任一种:
羧酸化合物1与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、DIC、DCC、HATU、HBTU)存在时,发生缩合反应,得到具有反应活性的酯;
或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐;
或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量(如0.01当量至0.5当量)的DMF存在条件下,反应得到酰氯。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(V)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000052
理想条件下,结构式(V)的合成包括下列步骤中的一种或多种:
步骤1.化合物1与化合物2,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物3;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000053
步骤2.化合物3上的氨基保护基团PG 4在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物V;
Figure PCTCN2019093946-appb-000054
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000055
其中,该合成步骤中得到的化合物8(化合物XIVa)即为所述目标化合物2;PG 4是氨基保护基团。
理想条件下,化合物2的合成包括下列步骤中的一种或多种:
步骤1.将L-酪氨酸的酯类衍生物1溶解于适当的溶剂,如丙酮、四氢呋喃、乙腈、二氯甲烷等,或者这些溶剂与水的混合溶剂,与苄基氯、苄基溴或其它苄基化合物,在0至60℃反应,可在反应体系中加入适当的有机或无机碱,如碳酸钠、碳酸钾、碳酸氢钠、碳酸氢钾、氢氧化钠、氢氧化钾、三乙胺、DBU、氢化钠等,也可在体系内加入适当的添加剂如碘化钠或相转移催化剂,如苄基三乙基氯化铵(TEBA)、四丁基溴化铵(TBAB)、四丁基氯化铵、四丁基硫酸氢铵、三辛基甲基氯化铵、十二烷基三甲基氯化铵、十四烷基三甲基氯化铵等,得到化合物2;
步骤2.将化合物2溶解于有机溶剂如二氯甲烷、四氢呋喃、甲醇、乙醇、乙醚等,用还原剂,如氢化铝锂、DIBAL、硼氢化钠、硼氢化锂、二氢双(2-甲氧乙氧基)铝酸钠(Red-Al)、乙硼烷等还原,可在反应体系中加入添加剂,如I 2、三氯化铁、氯化锌、氯化镁、氯化锂、氯化钙等控制还原剂的活性,得到化合物3;
步骤3.醇类化合物3在适当的氧化条件下,如swern氧化(草酰氯、DMSO、三乙胺)、Parikh-Doering氧化(三氧化硫吡啶氧化)、Dess-Martin氧化等,被氧化成醛4;
步骤4.醛4与磷酸酯反应(Horner-Wadsworth-Emmons反应)或者磷叶立德反应(Wittig反应),进行碳链延长,得到化合物5;
步骤5.化合物5中的双键在均相或两相催化剂的作用下被氢化还原,其中的苄基也同时被脱去,得到立体结构单一的手性化合物,或者两个非对映异构体的混合物;催化剂包括Pd/C、Pd(OH) 2/C、Pd/BaSO 4、PtO 2、Pt/Al 2O 3、Ru/C、Raney镍等两相催化剂,均相不对称氢化催化剂,如Crabtree催化剂、[Ru(II)-(BINAP)]类催化剂、[(Ph3P)CuH] 6催化剂等;
步骤6.将化合物6溶解在有机溶剂中,如四氢呋喃、乙腈、二氯甲烷,在硝化反应条件下被硝化,硝化试剂包括硝酸、硝酸/醋酸、硝酸钾/硫酸、亚硝基叔丁酯、硝酸/三氟乙酸酐、NO 2BF 4、硝基吡啶鎓盐等;
步骤7.在以下条件下化合物7中的硝基被还原成氨基,这些条件包括H 2/Pd/C,Fe或Zn/HOAc,SnCl 2/HCl。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000056
其中,该合成步骤中得到的化合物8(化合物XIVb)即为所述目标化合物2。
理想条件下,化合物2的合成包括下列步骤中的一种或多种:
步骤1.化合物1在-78℃至-45℃条件下,与Evans手性N-酰基噁唑烷酮或硫酮2,其中X=O或S,R 16=H,甲基,苯基,R 17=H,甲基,异丙基、苯基、苄基等,进行Aldol反应,得到立体构型单一的化合物3;
步骤2.化合物3上的羟基在以下条件下被脱去,这些条件包括Barton–McCombie去氧反应,即醇首先转化为硫代酰基衍生物,如烷基黄原酸盐、硫代氯甲酸苯酯、硫代羰基咪唑酯,然后用Bu 3SnH处理,发生自由基断裂得到脱羟基产物;自由基断键的条件包括:n-Bu 3SnH/AIBN,n-Bu 3SnH/AIBN/n-BuOH/PMHS,(Bu 4N) 2S 2O 8/HCO 2Na;
步骤3.将化合物4溶解在四氢呋喃中,Evans手性辅基在LiOH/H 2O 2的条件下被切断得到相应的酸5;
步骤4.化合物5被溶解在有机溶剂中,如乙酸乙酯、甲醇、二氯甲烷、乙醇或乙酸等,其中的苄基在钯炭催化剂存在下被催化氢解,得到化合物6;
步骤5.将化合物6溶解在有机溶剂中,如四氢呋喃、乙腈、二氯甲烷,在硝化反应条件下被硝化,硝化试剂包括硝酸、硝酸/醋酸、硝酸钾/硫酸、亚硝基叔丁酯、硝酸/三氟乙酸酐、NO 2BF 4、硝基吡啶鎓盐等;
步骤6.在以下条件下化合物7中的硝基被还原成氨基,这些条件包括H 2/Pd/C,Fe或Zn/HOAc,SnCl 2/HCl等,得到立体构型单一的手性化合物8。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)的合成由结构式(VI)和结构式(VII)缩合反应获得:
Figure PCTCN2019093946-appb-000057
其中X定义及缩合反应条件如权利要求9-11中任一项所述。
具体实施例中,结构式(VI)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000058
Figure PCTCN2019093946-appb-000059
理想条件下,结构式(VI)的合成包括下列步骤中的一种或多种:
步骤1.化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酸的衍生物化合物2;
Figure PCTCN2019093946-appb-000060
步骤2.化合物2与化合物3,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物4;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000061
步骤3.化合物4上的氨基保护基团PG 4在去保护条件下被选择性地去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
Figure PCTCN2019093946-appb-000062
步骤4.化合物5与结构式(IV),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物6;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000063
步骤5.化合物6上的氨基保护基团PG 1在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物VI;
Figure PCTCN2019093946-appb-000064
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(VII)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000065
理想条件下,结构式(VII)的合成包括下列步骤中的一种或多种:
步骤1.羧酸化合物1与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯;
或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐;
或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量(如0.01当量至0.5当量)的DMF存在条件下,反应得到酰氯;
Figure PCTCN2019093946-appb-000066
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)的合成由结构式(VIII)和结构式(IX)缩合反应获得:
Figure PCTCN2019093946-appb-000067
其中X定义及缩合反应条件如上面所述。结构式(VIII)中的NH 2基团理想地是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
具体实施条件下,结构式(VIII)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000068
Figure PCTCN2019093946-appb-000069
理想实施条件下,结构式(VIII)的合成包括下列步骤中的一种或多种:
步骤1.化合物1上的羧基保护基PG 3在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除),得到化合物2;
Figure PCTCN2019093946-appb-000070
步骤2.化合物2与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯化合物3;
Figure PCTCN2019093946-appb-000071
步骤3.化合物3与化合物4,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、 Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物5;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000072
步骤4.化合物5上的氨基保护基团PG 3在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物6;
Figure PCTCN2019093946-appb-000073
步骤5.化合物6与结构式(IV),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物7;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000074
步骤6.化合物7上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物VIII;
Figure PCTCN2019093946-appb-000075
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构 式(IX)的合成包括下列步骤中的一种或多种:
羧酸化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯IX;
或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐IX;
或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量的DMF存在条件下,反应得到酰氯IX;
Figure PCTCN2019093946-appb-000076
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)的合成由结构式(X)和结构式(XI)缩合反应获得:
Figure PCTCN2019093946-appb-000077
其中Y 1和Y 2基团缩合构成Y基团;Y 1和Y 2分别是NH 2、- +NH 3、COOH、COX、SO 2Cl、P(O)Cl 2、NHCOX、NHSO 2Cl、NHP(O)Cl 2、NHP(O)(OH)Cl、
Figure PCTCN2019093946-appb-000078
Figure PCTCN2019093946-appb-000079
在另一具体实施例制备上面所述的偶联物的过程中,结构式(X)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000080
Figure PCTCN2019093946-appb-000081
理想条件下,结构式(X)的合成包括下列步骤中的一种或多种:
步骤1.含羧基的化合物1和化合物VI在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线,发生缩合反应,得到化合物2;其中Z 1为Y 1的前体,如经过适当基团保护的氨基、羧基、酰胺基、磷酰胺基和磺酰胺基、羧酸酯、磷酸酯、膦酸酯等;
Figure PCTCN2019093946-appb-000082
步骤2.化合物2上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物3;
Figure PCTCN2019093946-appb-000083
步骤3.含羧基化合物4和含氨基的化合物3在缩合剂作用下,或者经过缩合反应路线发生缩合反应,得到化合物5;
Figure PCTCN2019093946-appb-000084
步骤4.化合物5中的官能团Z 1经过适当的化学转换,如羧基和氨基的脱保护等操作,生成官能团Y 1,得到化合物X;
Figure PCTCN2019093946-appb-000085
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(XI)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000086
理想条件下,结构式(XI)的合成包括下列步骤中的一种或多种:
步骤1.将化合物1溶解在有机溶剂,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜中,用碱,如氢化钠、钠、氢氧化钠等拔氢,然后与化合物2(其中X为氯、溴、碘等卤素或者其他离去基团)在一定的温度下搅拌,反应得到化合物3;
步骤2.化合物3上的羧基保护基团PG1在去保护条件下被去除,如叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物XIa-1;
步骤3.将化合物1溶解在有机溶剂,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜中,用碱,如氢化钠、钠、氢氧化钠等拔氢,然后 与化合物4(在一定的温度下搅拌,反应得到化合物5;
步骤4.化合物5上的羧基保护基团PG 1在去保护条件下被去除,如叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物XIa-2;
步骤5.将化合物6溶解在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,加入适当的有机碱,如三乙胺、N,N-二异丙基乙胺、吡啶等,与甲基磺酰氯、4-甲苯磺酰氯等在0-5℃反应,得到化合物7;
步骤6.化合物7与氨水在水相或有机溶剂中,如甲醇、乙醇、乙腈、四氢呋喃、环氧六环等,发生反应,反应可适当加热,得到化合物XIb。
步骤7.化合物7与叠氮化钠在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,发生反应,得到化合物8;
步骤8.叠氮化合物8在钯碳催化剂存在的条件下被氢化还原,或者三苯基膦和水的作用下被还原,得到化合物XIb;
步骤9.化合物7与二苄胺在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,优选N,N-二甲基甲酰胺,在100℃发生反应,得到化合物9;
步骤10.将化合物9溶解在溶剂中,如乙酸乙酯、甲醇、乙醇、乙酸、四氢呋喃等,在一定的氢气压力下,在钯碳催化剂上被还原,反应可适当加热至45℃,得到化合物XIb。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式为(II)的合成由结构式(XII)和结构式(XIII)缩合反应获得:
Figure PCTCN2019093946-appb-000087
Figure PCTCN2019093946-appb-000088
其中X定义及缩合反应条件如前面所述。
理想条件下,结构式(XII)中的NH 2基团是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(XII)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000089
理想条件下,结构式(XII)的合成包括下列步骤中的一种或多种:
步骤1.化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酸的衍生 物化合物2;
Figure PCTCN2019093946-appb-000090
步骤2.化合物2与化合物3,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物4;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000091
步骤3.化合物4上的氨基保护基团PG 4在去保护条件下被选择性地去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
Figure PCTCN2019093946-appb-000092
步骤4.化合物5与结构式(IV)(即权利要求18-21中任一项所述的结构式(IV)),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物6;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
Figure PCTCN2019093946-appb-000093
步骤5.化合物6上的氨基保护基团PG1在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物XII;
Figure PCTCN2019093946-appb-000094
在另一具体实施例制备上面所述的偶联物的过程中,其特征在于,结构式(XIII)的合成包括下列步骤中的一种或多种:
Figure PCTCN2019093946-appb-000095
优选条件下,结构式(XIII)的合成包括下列步骤中的一种或多种:
步骤1.含羧基化合物1和含氨基的化合物2在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物3;
Figure PCTCN2019093946-appb-000096
步骤2.化合物3上的羧基保护基团PG 1在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除),得到化合物4;
Figure PCTCN2019093946-appb-000097
步骤3.羧酸化合物4与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯结构式(XIII);
或者,羧酸化合物4与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱存在下反应,得到具有反应活性的混合酸酐结构式(XIII);
或者,羧酸化合物4与草酰氯,在三乙胺等有机碱和催化剂量的DMF存在条件下,反应得到酰氯结构式(XIII),
Figure PCTCN2019093946-appb-000098
所述式(II)化合物的优选结构如下:
Figure PCTCN2019093946-appb-000099
Figure PCTCN2019093946-appb-000100
在另一具体实施例中,一种药物组合物,其包含上述任一项所述的偶联物或以上面所述含有链接体的化合物同细胞结合分子反应构成的偶联物,以及药学上可接受的辅料。上面任一项所述的偶联物在用于制备治疗癌症、感染或自身免疫疾病的药物中用途。
附图说明
图1.显示合成Tubulysin衍生物片段13和18。
图2.显示合成Tubulysin衍生物片段34。
图3.显示合成Tubulysin衍生物片段37,38和45。
图4.显示合成Tubulysin衍生物片段57。
图5.显示合成Tubulysin衍生物片段71。
图6.显示合成可偶联Tubulysin衍生物72。
图7.显示偶连物在荷载NCI-N87异种移植肿瘤的BALB/c裸鼠上的体内抗肿瘤活性.
图8.显示Tubulysin衍生物的Her2抗体偶联物的毒性研究(与T-DM1对照)
相关术语解释:
烷基指含有1至8个碳原子的线性或者环状直链或者支链脂肪烃。支链指在线性的烷基上有一个或者多个低级烷基,比如甲基,乙基或者丙基相连接。烷基实施例包括甲基,乙基,正丙基,异丙基,正丁基,叔丁基,正戊烷基,3-戊烷基,辛烷基,壬烷基,癸烷基,环戊烷基,环己烷基,2,2-二甲基丁基,2,3-二甲基丁基,2,2-二甲基戊基,2,3-二甲基戊基,3,3-二甲基戊基,2,3,4-三甲基戊基,3-甲基己基,2,2-二甲基己基,2,4-二甲基己基,2,5-二甲基己基,3,5-二甲基己基,2,4-二甲基戊基,2-甲基庚基,3-甲基庚基,正庚基,异庚基,正辛烷基和异辛烷基。C 1-C 8烷基既可以未被取代也可以被以下但不限于以下的一个或者多个基团取代:C 1-C 8烷基,C 1-C 8烷氧基,芳基,酰基,酰氧基,酯基,-C(O)NH 2,-C(O)NHR’,-C(O)N(R’) 2, -NHC(O)R’,-S(O) 2R’,-S(O)R’,-OH,卤素(-F,-Cl,-Br,-I),-N 3,-NH 2,-NHR’,-N(R’) 2及-CN;其中R’指C 1-C 8烷基或者芳基。
C 3-C 8碳环指含3,4,5,6,7,或8个碳原子的饱和或不饱和的非芳烃环状化合物。典型的C 3-C 8碳环包括但不限于环丙基,环丁基,环戊基,环戊二烯基,环己基,环己烯基,1,3-环己二烯基,1,4-环己二烯基,环庚基,1,3-环庚二烯基,1,3,5-环庚三烯基,环辛基及环辛二烯基。C 3-C 8碳环既可以未被取代,也可以被以下但不限于以下的一个或者多个基团取代:C 1-C 8烷基,C 1-C 8烷氧基,芳基,酰基,酰氧基,酯基,-C(O)NH 2,-C(O)NHR’,-C(O)N(R’) 2,-NHC(O)R’,-S(O) 2R’,-S(O)R’,-OH,卤素(-F,-Cl,-Br,-I),-N 3,-NH 2,-NHR’,-N(R’) 2及-CN;其中R’是C 1-C 8烷基或者芳基。
C 3-C 8碳环基指上述C 3-C 8碳环上的一个氢原子被化学键所取代而产生的基团。
烯烃基指含有一个碳碳双键的直链或者支链脂肪烃,碳链上有2至8个碳原子。烯烃基的实施例包括乙烯基,丙烯基,正丁烯基,异丁烯基,3-甲基-2-丁烯基,正戊烯基,己烯基,庚烯基和辛烯基。
炔烃基指含有一个碳碳三键的直链或者支链脂肪烃,碳链上有2至8个碳原子。炔烃基的实施例包括乙炔基,丙炔基,正丁炔基,2-丁炔基,3-甲基丁炔基,正戊炔基,己炔基,庚炔基和辛炔基。
杂烷基指含2至8个碳原子而且有1至4个碳原子被O,S或者N所取代的烷基。
芳基或者芳香基指含有一个或者多个环的由3至14个碳原子(多数的情形是6至10个碳原子)组成的芳香烃或者杂芳香烃基团。杂芳香烃基团指有一个或多个碳原子(多数的情形是1,2,3或4个碳原子)被O,N,Si,Se,P或者S(最好是O,S,N)所取代所产生的芳香烃基团。芳基或者芳香基也指一个或多个氢原子被取代的芳香烃基团,这些取代基团有:R 13,F,Cl,Br,I,OR 13,SR 13,NR 13R 14,N=NR 13,N=R 13,NR 13R 14,NO 2,SOR 13R 14,SO 2R 13,SO 3R 13,OSO 3R 13,PR 13R 14,POR 13R 14,PO 2R 13R 14,OPO 3R 13R 14或者PO 3R 13R 14。其中R 13和R 14分别为H,烷基,烯烃基,炔烃基,杂烷基,芳基,芳烷基,羰基或者可以药用的盐。
卤素原子指氟,氯,溴,碘原子,最好是氟和氯。
杂环指含2至8个碳原子的芳香,非芳香性环或杂环的环上有1至4个碳原子被杂元素所取代。这些杂元素为O,N,S,Se和P,最好为O,N和S。可用的杂环亦可参见《The Handbook of Chemistry and Physics》,第78版,CRC Press,1997-1998,第225至226页。适合的非杂芳基包括但不限于环氧基,环氮乙烷基,环硫乙烷基,吡咯烷基,吡唑烷基,咪唑烷基,环氧乙烷基,四氢呋喃基,二氧戊环基,四氢吡喃基,二氧杂环己烷基,哌啶基,哌嗪基,吗啉基,吡喃基,咪唑啉基,吡咯啉基,吡唑啉基,噻唑烷基,四氢硫吡喃基,二噻烷基,硫吗啉基,二氢吡喃基,四氢吡喃基,四氢吡啶基,二氢吡啶基,四氢嘧啶基,二氢硫吡喃基,氮杂环庚烷基以及它们与苯基生成的稠环。
杂芳基指含3至14个(最好为5至10个)原子的芳香性杂单环,二环或多环结构。例如吡咯基,吡啶基,吡唑基,噻吩基,嘧啶基,吡嗪基,四氮唑基,吲哚基,喹啉基,嘌呤基,咪唑基,噻吩基,噻唑基,苯并噻唑基,呋喃基,苯并呋喃基,1,2,4-噻二唑基,异噻唑基,三氮唑基,四氮唑基,异喹啉基,苯并噻吩基,异苯并呋喃基,吡唑基,咔唑基,苯并咪唑基,异恶唑基,N-氧吡啶基以及他们与苯基生成的稠环。
烷基,环烷基,烯烃基,炔烃基,芳基,杂芳基和杂环基等术语也指由相应的烃失去两个氢原子生成的亚烷基,环亚烷基,亚烯基,亚炔基,亚芳基,杂亚芳基和杂亚环等。
“芳烷基”指的是一个非环状的烷基自由基的一个氢原子连接的碳原子,通常是一个终端或sp3杂化的碳原子由一个芳基取代。典型的芳基烷基包括苄基,2-苯乙烷基,2-苯乙烯基,萘甲基,2-萘乙烷基,2-萘乙烯基,萘苄基,2-萘苯乙烷基等。
“杂芳烷基”指非环型烷基自由基的一个氢原子链接到一个碳原子上,通常是一个终端或sp3杂化的碳原子由一个杂芳基取代。杂芳烷基代表如2-苯并咪唑甲基,2-呋喃乙基。
“羟基保护集团”是指,甲基氧甲基醚(MOM),2-甲基氧乙基氧甲基醚(2-MOEOM),四氢吡喃醚,苄醚,对甲基氧苄醚,三甲基矽醚,三乙基矽醚,三异丙基矽醚,叔丁基二甲基硅醚,三苯基甲基硅醚,乙酸乙酯,含取代基的乙酸乙酯,苯甲酸酯,甲酸苄酯,氯代乙酸酯,甲氧基乙酸酯, 苯氧基乙酸酯,特戊酸酯(pivaloate),金刚烷酸酯,三甲基苯甲酸酯(mesitoate),甲基磺酸酯和甲苯磺酸盐。
氨基酸可以是天然的和/或非自然的氨基酸,通常是L型或D型,优选是α氨基酸。天然氨基酸是由基因编码的编排,它们是丙氨酸、精氨酸、天门冬酰胺、天门冬氨酸、半胱氨酸、谷氨酸、谷氨酰胺、甘氨酸、组氨酸、异亮氨酸、亮氨酸、赖氨酸、蛋氨酸、苯丙氨酸、脯氨酸、丝氨酸、苏氨酸、酪氨酸、色氨酸和缬氨酸。非天然氨基酸是蛋白氨基酸衍生的形式。例如羟脯氨酸、羊毛硫氨酸;、2-氨基异丁酸,脱氢丙氨酸、γ-氨基丁酸(神经传递分子)、鸟氨酸、瓜氨酸、β-丙氨酸(3-胺基丙酸)、γ-羧基谷氨酸盐,硒代半胱氨酸(目前在许多非真核生物以及大多数真核生物中,而不是直接由DNA编码构成)、吡咯赖氨酸(仅在一些古细菌和细菌中发现的),N-甲酰甲硫氨酸(它是细菌,线粒体和叶绿体蛋白最初的氨基酸),5-羟基色氨酸,L-二羟基苯丙氨酸、三碘甲腺原氨酸、L-3,4-二羟基苯丙氨酸(DOPA),和O-磷酸丝氨酸。氨基酸的术语还包括氨基酸类似物和近似物。类似物具有相同的H 2N(R)CHCO 2H天然氨基酸的结构,但R基团在天然氨基酸未发现。例如类似物包括类丝氨酸、己氨酸、蛋氨酸亚砜,和蛋氨酸甲基锍盐。优选地,类氨基酸具有与α氨基酸的常规化学结构不同的结构,但其功能类似。术语“非天然氨基酸”以“D”的立体形态、天然氨基酸的以“L”立体形态。在本专利申请中使用1~12个氨基酸时,氨基酸序列最好能被蛋白酶的识别和裂解。许多识别和切割氨基酸序列已被熟知(如Matayoshi et al.Science 247:954(1990);Dunn et al.Meth.Enzymol.241:254(1994);Seidah et al.Meth.Enzymol.244:175(1994);Thornberry,Meth.Enzymol.244:615(1994);Weber et al.Meth.Enzymol.244:595(1994);Smith et al.Meth.Enzymol.244:412(1994);and Bouvier et al.Meth.Enzymol.248:614(1995),其中文献中还含带引述文献),特别是,氨基酸序列选择为Val-Cit,Ala-Val,Ala-Lys,Gly-Lys,Ala-Ala,Val-Val,Val-Ala-Val,Lys-Lys,Ala-Asn-Val,Val-Leu-Lys,Cit-Cit,Val-Lys,Asp-Lys,Glu-Lys,Ala-Ala-Asn,Lys,Cit,Ser,和Glu的分子。
“肽”是由两个或多个氨基酸通过一个氨基酸的氨基与另一个氨基酸的羧基以肽键(即酰胺键)结合而成。两个氨基酸以肽键相连的化合物称为二肽;三个氨基酸以肽键相连的化合物称为三肽,以此类推,三十个氨基 酸以肽键相连的化合物称为三十肽。完全以天然α氨基酸构成的肽为天然肽(天然蛋白)。含有一个或多个非天然氨基酸或氨基酸类似物的肽为非天然肽(类肽化合物)。两个或以上氨基酸的肽为一个肽单元。
“糖苷”是通过其糖基键将糖基团通过其端基异构体碳键连接到另一基团的分子。糖苷可以通过O-(O-糖苷),N-(糖胺),S-(硫代糖苷)或C-(C-糖苷)糖苷键连接。其核心是经验公式为C m(H 2O) n(其中m可以不同于n,m和n是<36),本文中的糖苷包括葡萄糖(葡萄糖),果糖(levulose)阿洛糖,阿卓糖,甘露糖,古洛糖,艾杜糖,半乳糖,托洛糖,半乳糖胺,葡糖胺,唾液酸,N-乙酰葡糖胺,磺基喹诺酮(6-脱氧-6-磺基-D-吡喃葡萄糖),核糖,阿拉伯糖,木糖,溶血糖,山梨醇,甘露醇,蔗糖,乳糖,麦芽糖,海藻糖,麦芽糖糊精,棉子糖,葡萄糖醛酸(葡糖苷酸)和水苏糖。糖苷可以是D型式或L型式,5个原子环状呋喃糖形式,6个原子环状吡喃糖形式,或非环形式,α-异构体(Haworth投影的碳原子下方的端基异碳的-OH)或β-异构体(在Haworth投影平面上方的端基异构碳的-OH)。其在本文中常用糖苷是单糖,二糖,多元醇或寡糖(含有3-6个糖单元)。
“抗体”,这里所用的是用在最广泛的意义上,具体涵盖完整的单克隆抗体,多克隆抗体,特异性抗体,多特异性抗体(例如,双特异性抗体和抗体片段),具有理想的生物活性,抗体片段的药物结合位点的必要数量的链接。一种抗体的原生形态是一个四聚体,由两个相同的免疫球蛋白链对,每对有一个轻链和重链。在每一对中,轻链和重链可变区(VL和VH)共同负责与抗原结合。轻链和重链可变区由一个框架区三高变区中断,也被称为“互补决定区”或“服务”。不断的区域可能是公认的,与免疫系统的相互作用。一种抗体可以是任何类型(如IgG、IgE、IgM、IgD、和IgA)、大类(例如,IgG1、IgG2、IgG3、IgG4、IgA1和IgA2)或子类。抗体可以从任何合适的物种中获得。在某些方面,抗体是人或鼠的起源。抗体可以是人的,人源化的或嵌合的。
术语“特异结合”和“特异结合”意味着抗体或抗体衍生物会以高度选择性的方式与其相应的靶抗原结合,而不是与许多其他抗原结合。通常情况下,抗体或抗体具有至少约1x10 -7M的亲和性衍生物结合。最好是1x10 -8M 到10 -9M、10 -10M、10 -11M或10 -12M结合。预定抗原的亲和力至少是两倍大于结合的非特异性抗原的亲和性(如牛血清白蛋白,酪蛋白)。
“药用”或“可以药用”指相应的化合物或化合物组成在动物或人体上不产生有害的,过敏性的或其他不良反应。
药学上可接受的辅料包括所有载体,稀释剂,助剂或者成形剂,比如防腐剂,抗氧化剂,填料,崩解剂,润湿剂,乳化剂,悬浮剂,溶剂,分散介质,涂层,抗菌剂,抗真菌剂,等渗和吸收延缓剂等等。在医药领域,在具有活性的药物成分中加入这些辅料是十分常见的做法。可以说,除非辅料与药物活性组分并不相容,在药物成分中加入辅料并不是没有道理。为取得好的结果,具有活性的辅助组分也可以被加入药物成分中。
在本发明中,可以药用的盐指本发明化合物的盐类衍生物。经过适当的修饰,本发明化合物可以形成相应的酸盐或碱盐。可以药用的盐包括常见的没有毒性的盐或季铵盐,这些盐可以用本发明化合物和相应的没有毒性的无机酸或有机酸制成。例如,用无机酸包括盐酸,氢溴酸,硫酸,氨基磺酸,磷酸以及硝酸等和有机酸包括乙酸,丙酸,丁二酸,酒石酸,柠檬酸,甲磺酸,苯磺酸,葡萄糖醛酸,谷氨酸,苯甲酸,水杨酸,甲苯磺酸,草酸,富马酸以及乳酸等都可以做成药用盐。其它的盐包括氨基丁三醇,氨基三乙醇,甲葡胺,吡咯乙醇等铵盐以及钠,钾,钙,锌,镁等金属盐。
本发明可以药用的盐可以用常规的化学方法制成。一般地说,这些盐可以通过在本发明化合物的游离酸或碱的水溶液或有机溶液或两者的混合溶液中加入其它适当的等当量的碱或酸而形成。非水相的反应介质一般为乙醚,乙酸乙酯,乙醇,异丙醇或者乙腈。可以适用盐的列表可参见《Remington’s Pharmaceutical Sciences》,第17版.Mack Publishing Company,Easton,PA,1985,第1418页.
术语“药学上可接受的盐”是指配体药物偶联物或接头药物缀合物的药学上可接受的有机或无机盐。缀合物可以含有至少一个氨基,因此可以与氨基形成酸加成盐。硝酸盐,硫酸氢盐,磷酸盐,酸式磷酸盐,异烟酸盐,乳酸盐,水杨酸盐,酸式柠檬酸盐,酒石酸盐,油酸盐,鞣酸盐,泛酸盐,酒石酸盐,抗坏血酸盐,琥珀酸盐,马来酸盐,龙胆酸盐,富马酸盐,葡萄糖酸盐,葡糖醛酸盐,糖酸盐,甲酸盐,苯甲酸盐,谷氨酸盐,甲磺酸 盐,乙磺酸盐,苯磺酸盐,对甲苯磺酸盐和双羟萘酸盐(即1,1'-亚甲基双-(2-羟基萘甲酸盐))。药学上可接受的盐可以包括另外的分子,例如乙酸根离子,琥珀酸根离子或其他抗衡离子。抗衡离子可以是使母体化合物上的电荷稳定的任何有机或无机部分。此外,药学上可接受的盐在其结构中可以具有多于一个的带电原子。多个带电原子是药学上可接受的盐的一部分的实施例可以具有多个抗衡离子。因此,药学上可接受的盐可以具有一个或多个带电原子和/或一个或多个抗衡离子。
短语“药学上可接受的溶剂合物”或“溶剂合物”是指一种或多种溶剂分子和配体药物偶联物或接头药物缀合物的缔合。形成药学上可接受的溶剂合物的溶剂的实施例包括但不限于水,异丙醇,乙醇,丁醇,叔丁醇,丙酮、甘油,DMSO,乙酸乙酯,甲酸,乙酸,三乙醇胺和乙醇胺。
水合物(Hydrate)指的是含有水的化合物。其中水可以以配位键与其他部分相连,如金属离子水合配体形成络合物,也可以是以共价键相结合,如水合三氯乙醛。也可以指是某些化合物与水分在一定温度、压力条件下形成的晶体或液体分子。水合物中的水是以确定的量存在的,例如无水硫酸钠Na 2SO 4的水合物的组成为Na 2SO 4·10H 2O。水合物中的水有几种不同的结合方式:一种是作为配体,配位在金属离子上,称为配位结晶水;另一种则结合在阴离子上,称为阴离子结晶水。水也可以不直接与阳离子或阴离子结合而依一定比例存在于晶体内,在晶格中占据一定的部位。这种结合形式的水称为晶格水,一般含有12个水分子。有些晶形化合物也含水,但无一定比例。水合物的盐是指在此水合物基础上形成的药学上可接受的盐。
旋光异构体又称对映异构体,对掌异构物、光学异构物、镜像异构物、对映异构体或手性异构体,不能与彼此立体异构体镜像完全重叠的分子。含有一个物质含一个手性碳原子时,有两个旋光异构体,它们具有互为实物和镜像的关系,故也称对映体。对映异构体具有相等的旋光能力,但旋转方向相反,其物理和化学性质极可能相似。含有两个相同属性碳原子的分子,有3个旋光异构体。分子中当含有几个不同的手性原子时,其旋光异构体的数目为2 n,n为不同手性原子的个数。等量互为旋光异构体的两种物质均匀混合,旋光性互相抵消,就组成了外消旋体。
“患者”或“受试者”的实施例包括但不限于人,大鼠,小鼠,豚鼠,猴,猪,山羊,牛,马,狗,猫,鸟和家禽。在示例性实施例中,患者或受试者是人。
“给药”是指将药物或其他药剂转移,输送,引入或运送给受试者的任何方式,包括口服,局部接触,静脉内,腹膜内,肌肉内,病灶内,鼻内,皮下或鞘内给药本发明也考虑了使用装置或器械来施用药剂,这种装置可以利用主动或被动的运输,并且可以是缓释或快速释放递送装置。
本文可以使用以下缩写,并具有指定的定义:Boc,叔丁氧基羰基;BroP,溴代三吡咯烷鏻六氟磷酸盐;CDI,1,1'-羰基二咪唑;DCC,二环己基碳二亚胺;DCE,二氯乙烷;DCM,二氯甲烷;DIAD,偶氮二羧酸二异丙酯;DIBAL-H,二异丁基氢化铝;DIPEA,二异丙基乙胺;DEPC,二氰基磷酸二乙酯;DMA,N,N-二甲基乙酰胺;DMAP,4-(N,N-二甲基氨基)吡啶;DMF,N,N-二甲基甲酰胺;DMSO,二甲亚砜;DTT,二硫苏糖醇;EDC,1-(3-二甲基氨基丙基)-3-乙基碳二亚胺盐酸盐;ESI-MS,电喷雾质谱;HATU,O-(7-氮杂苯并三唑-1-基)-N,N,N',N'-四甲基脲六氟磷酸盐;HOBt,1-羟基苯并三唑;HPLC,高压液相色谱;NHS,N-羟基琥珀酰亚胺;MMP,4-甲基吗啉;PAB,对氨基苄基;PBS,磷酸盐缓冲盐水(pH 7.0~7.5);PEG,聚乙二醇;SEC,大小排阻色谱法;TCEP,三(2-羧乙基)膦;TFA,三氟乙酸;THF,四氢呋喃;Val,缬氨酸。
具体实施方式
下面将参照附图更详细地描述本发明的具体实施例。虽然附图和后面实施例中显示了本发明的具体实施例子,然而应当理解,可以以各种形式实现本发明而不应被这里阐述的实施例所限制。相反,提供这些实施例是为了能够更透彻地理解本发明,并且能够将本发明的范围完整的传达给本领域的技术人员。
本公开主题可以以许多不同的形式来体现,并且不应该被解释为限于本文阐述的实施方案。实际上,对于本公开主题所属领域的技术人员来说,将会想到具有本文所包括的描述中给出的教导的益处的本公开主题的许多修改和其他实施方案。因此,应该理解的是,本公开主题不限于所公开的具体实施方案,并且修改和其他实施方案意在被包括在所公开的主题的范围内。
尽管本文使用了特定的术语,但它们仅用于一般性和描述性的意义,而不是为了限制的目的。除非另有定义,否则本文使用的所有技术和科学术语具有与本公开描述的主题所属领域的普通技术人员通常理解的相同的含义。
使用标准命名法来描述化合物。除非另有定义,否则本文使用的所有技术和科学术语具有与本发明所属领域的技术人员通常理解的相同的含义。
术语“一”不表示数量的限制,而是表示存在至少一个所引用的项目。数值范围的叙述仅仅意在作为单独提及落入该范围内的每个单独值的速记方法,除非本文另有指出,并且每个单独值被合并到说明书中,就好像其在本文中单独列举一样。所有范围的终点都包括在范围内并且可以独立组合。本文描述的所有方法可以以合适的顺序执行,除非本文另有指示或者与上下文明显矛盾。除非另有声明,否则实例或示例性语言(如“例如”)的使用仅意在更好地说明本发明,并且不构成对本发明范围的限制。
本发明包括具有至少一个期望的原子同位素取代,其量高于同位素的天然丰度(即富集)的式(I)化合物以及化合物的用途。同位素是具有相同原子序数但不同质量数,即相同数量的质子但不同数量的中子的原子。同位素取代,例如氘取代,可以是部分的或完全的。部分氘取代是指至少一个氢被氘取代。在某些实施方案中,同位素在感兴趣的任何位置富集90%、95%或99%或更多。在一个实施方案中,氘在所需位置富集90%、95%或99%。
需要说明的是,在说明书及权利要求当中使用了某些词汇来指称特定组件。本领域技术人员应可以理解,技术人员可能会用不同名词来称呼同一个组件。本说明书及权利要求并不以名词的差异来作为区分组件的方式,而是以组件在功能上的差异来作为区分的准则。如在通篇说明书及权利要求当中所提及的“包含”或“包括”为一开放式用语,故应解释成“包含但不限定于”。说明书后续描述为实施本发明的较佳实施方式,然所述描述乃以说明书的一般原则为目的,并非用以限定本发明的范围。本发明的保护范围当视所附权利要求所界定者为准。
术语“C 1-C 6”表示含1~6个碳的基团。
术语“含有亲水支链的连接体”表示主要构架是C 2-C 100的肽单元(1~12个天然或非天然氨基酸)、腙键基团、二硫化物基团、酯基团、肟基团、酰 胺基团或硫醚键基团构成。
术语“药学上可接受的盐”意味着适用于药物制剂的化合物的盐。在化合物具有一个或多个碱性基团的情况下,盐可以是酸加成盐,例如硫酸盐、氢溴酸盐、酒石酸盐、甲磺酸盐、马来酸盐、柠檬酸盐、磷酸盐、乙酸盐,扑酸盐、氢碘酸盐、硝酸盐、盐酸盐、乳酸盐、甲基硫酸盐、富马酸盐、苯甲酸盐、琥珀酸盐、甲磺酸盐、乳糖酸盐、辛二酸盐、甲苯磺酸盐等等。在化合物具有一个或多个酸性基团的情况下,盐可以是下列盐,诸如钙盐、钾盐、镁盐、葡甲胺盐、铵盐、锌盐、哌嗪盐、氨基丁三醇盐、锂盐、胆碱盐、二乙胺盐、4-苯基环己胺盐、苄星(benzathine)盐、钠盐、四甲基铵盐等等。多晶型结晶形式和溶剂化物也包含在本发明的范围内。
本发明药学上可接受的盐可以用常规的化学方法制成。一般地说,这些盐可以通过在本发明化合物的游离酸或碱的水溶液或有机溶液或两者的混合溶液中加入其它适当的等当量的碱或酸而形成。非水相的反应介质一般为乙醚,乙酸乙酯,乙醇,异丙醇或者乙腈。可以适用盐的列表可参见《Remington’s Pharmaceutical Sciences》,第17版.Mack Publishing Company,Easton,PA,1985,第1418页。
药学上可接受的辅料包括所有载体,稀释剂,助剂或者成形剂,比如防腐剂,抗氧化剂,填料,崩解剂,润湿剂,乳化剂,悬浮剂,溶剂,分散介质,涂层,抗菌剂,抗真菌剂,等渗和吸收延缓剂等等。在医药领域,在具有活性的药物成分中加入这些辅料是十分常见的做法。可以说,除非辅料与药物活性组分并不相容,在药物成分中加入辅料并不是没有道理。为取得好的结果,具有活性的辅助组分也可以被加入药物成分中。
在本文中,式(I)中的
Figure PCTCN2019093946-appb-000101
表示手性碳原子位点,可选择为纯R,纯S或R/S不同比例的混合结构。
除非特别指出特定的立体异构体(例如,通过结构式中相关立构中心处的粗或虚线键、通过描述结构式中的双键为具有E或Z构型、或通过使用立体化学指定命名法),所有立体异构体都作为纯化合物及其混合物包括在本发明的范围内。除非另有说明,个别的对映异构体、非对映异构体、几何异构体及其组合和混合物都被本发明包含。
本领域技术人员将理解,化合物可具有互变异构形式(例如,酮和烯醇形式)、共振形式和两性离子形式,其等同于本文中使用的结构式中所描绘 的那些,并且该结构式包括这种互变异构、共振或两性离子形式。
本发明的另一方面内容是抗体的生产制备。其中包括其在生物体内、体外的产生过程或者组合过程。抗受体肽段的的多克隆抗体的生产方法已是众所周知,如美国专利编号4,493,795(Nestor等)。制备单克隆抗体的经典方法是采用特定抗原免疫小鼠,分离得到的小鼠脾细胞与骨髓瘤细胞融合(Kohler,G;Milstein,C.1975.Nature 256:495-497)。详细的操作步骤见antibodies-A Laboratory Manual,Harlow and Lane,eds.,cold spring harbor laboratory press,new York(1988),此方法移收入本文所引用的文献。特别地,可通过运用目的抗原免疫小鼠,大鼠,仓鼠或者其他哺乳动物的方法来获得特殊的单克隆抗体。其中目的抗原包括:完整的细胞,从细胞中分离得到的抗原,完整的病毒,弱化的完整病毒以及病毒蛋白。采用PEG6000将脾细胞与骨髓瘤细胞融合。融合后得到的杂交瘤,利用其对HAT的敏感性,对其进行筛选。杂交瘤细胞产生的单克隆抗体通过其与特定的靶细胞受体发生免疫反应或者抑制受体活性,这在实施这项发明中起作用。
融合后得到的单克隆杂交瘤细胞可分泌针对特定抗原的单克隆抗体。目前,发明中使用的单克隆抗体,正是通过在营养丰富的培养基中培养单克隆杂交瘤细胞从而富集抗体。培养条件需确保杂交瘤细胞有足够的时间将产生的抗体分泌进入培养基中。将含抗体的培养上清收集后,通过人们所熟知的技术对抗体进行纯化。分离方法包括:蛋白A亲和层析法;阴离子交换层析法,阳离子交换层析法,疏水层析法以及分子筛层析法(特别是运用抗原交联蛋白A的亲和层析法以及分子筛层析法运用较为广泛);离心法;沉淀法或其它标准的纯化方法
杂交瘤培养所需有效培养基以及人工合成培养基可以通过技术合成或者商业途径获取。其中,典型的人工合成培养基:DMEM(Dulbecco等Virol 8:396(1959))加入4.5mg/L葡萄糖,20mM谷氨酰胺,20%胎牛血清以及消泡剂,如:聚氧乙烯聚氧丙烯共聚物。
除细胞融合技术外,构建生产抗体的细胞系还可以通过其它方法,如:直接将肿瘤原性DNA转染B淋巴细胞,或者将致癌病毒基因(如:EBV又称HHV-4或KSHV)导入B淋巴细胞,详见美国专利编号.4341761;4399121;4427783;4444887;4451570;4466917;4472500;4491632;4493890.单克隆抗体还可以通过抗受体多肽或者含羧基末端的多肽来制备。详见Niman等 Proc.Natl.Acad.Sci.USA,80:4949-4953(1983);Geysen等Proc.Natl.Acad.Sci.USA,82:178-182(1985);Lei等Biochemistry 34(20):6675-6688(1995)。通常,抗受体多肽或者多肽类似物可以单独使用或者交联免疫原性载体来作为免疫原制备抗受体多肽的单克隆抗体。
发明中作为结合分子的抗体还有其它成熟的生产方法。其中特别受关注的是生产全人源抗体的工艺方法。噬菌体展示技术通过亲和筛选从全人源抗体文库中获得与已知抗原特异性结合的全人源抗体。噬菌体展示技术本身,载体构建以及文库筛选都在文献中有详尽的记录。详见Dente等Gene.148(1):7-13(1994);Little等Biotechnol Adv.12(3):539-55(1994);Clackson等Nature 352:264-628(1991);Huse等Science 246:1275-1281(1989)。
运用杂交瘤技术通过其它种属(如:小鼠)获得的单克隆抗体需对其进行人源化改造。经过改造的抗体可以大大减少异源抗体对人类机体造成的免疫副反应。其中,抗体的人源化较为常见的方法是互补决定域的移植与重塑。详见:美国专利编号5859205及6797492;Liu等,Immunol Rev.222:9-27(2008);Almagro等,Front Biosci.1;13:1619-33(2008);Lazar等Mol Immunol.44(8):1986-98(2007);Li等Proc.Natl.Acad.Sci.USA.103(10):3557-62(2006)上述文章作为参考文献并入本文。全人源抗体也可以通过抗原免疫携带大量人免疫球蛋白轻链及重链的转基因小鼠,兔子,猴子等其它哺乳动物来制备。以小鼠为例:Xenomouse(Abgenix,Inc.),HuMab-Mouse(Medarex/BMS),VelociMouse(Regeneron),详见:美国专利编号:6596541,6207418,6150584,6111166,6075181,5922545,5661016,5545806,5436149以及5569825。人类治疗过程中,将鼠源抗体可变区基因与人源抗体恒定区基因整合构建的嵌合抗体在人体中所产生的免疫源性反应要大大低于鼠抗(Kipriyanov等,Mol Biotechnol.26:39-60(2004);Houdebine,Curr Opin Biotechnol.13:625-9(2002)上述文章作为参考文献并入本文.此外,对抗体可变区的部分位点进行定点突变可以有效提高抗体的亲和力和特异性(Brannigan等,Nat Rev Mol Cell Biol.3:964-70(2002);Adams等,J Immunol Methods.231:249-60(1999))。对抗体恒定区进行部分替换也可以有效增进其与免疫效应因子的亲和力从而增强细胞毒性作用。
对恶性细胞抗原有免疫特异性的抗体,可以通过商业途径或者一些成熟的技术方法来获得,如:化学合成或者重组表达技术。这类抗体的编码 基因同样可以通过一些商业途径,如GenBank数据库或者其它类似的数据库,出版文献,或者常规的克隆测序方法来获得。
除抗体以外,多肽或者蛋白也同样可以作为结合分子来结合,阻断、进攻或通过其它方式与靶细胞表面相对应的受体或者表位相互作用。只要这些多肽或者蛋白能特异结合特定表位或其对应受体,那么它们不一定要属于免疫球蛋白家族。这些多肽也可以通过类似噬菌体展示抗体的技术来进行分离(Szardenings,J Recept Signal Transduct Res.2003;23(4):307-49)。从随机的多肽文库获取的肽段与抗体及抗体片段的应用相似。多肽或蛋白分子可通过其结合分子与一些大分子或介质连接来保持其抗原结合的特异性。这些大分子及介质包括:白蛋白、聚合物、脂质体、纳米颗粒或者树状聚合物。
在癌症、自身免疫性疾病和感染性疾病的治疗中,用于偶联的抗体举例如下(但不限于此):3F8(抗GD2抗体),阿巴伏单抗(抗CA-125抗体),阿昔单抗(抗CD41抗体(整合素α-IIB),阿达木单抗(抗-TNF-α抗体),阿德木单抗(抗-EpCAM抗体,CD326),阿非莫单抗(抗-TNF-α);阿夫土珠(抗-CD20抗体),Alacizumab pegol(抗VEGFR2抗体),ALD518(抗-IL-6抗体),阿仑单抗(别名:Campath,MabCampath,坎帕斯,抗CD52抗体),阿妥莫单抗(抗CEA抗体),Anatumomab(抗-TAG-72抗体),Anrukinzumab(别名:IMA-638,抗-IL-13抗体),阿泊珠单抗(抗-HLA-DR抗体),阿西莫单抗(抗CEA抗体),阿塞珠单抗(抗-L-选择素(CD62L)抗体,Atlizumab(别名:托珠单抗,Actemra,RoActemra,抗-IL-6受体抗体),Atorolimumab(抗恒河猴因子抗体),bapineuzumab(抗β-淀粉样蛋白抗体),巴利昔单抗(舒莱,antiCD25(IL-2受体的α链)抗体,巴维昔单抗(抗磷脂酰丝氨酸抗体),贝妥莫单抗(别名:LymphoScan,抗-CD22抗体),贝利木单抗(别名:Benlysta,LymphoStat-B,抗BAFF抗体),Benralizumab(抗CD125抗体),柏替木单抗(抗CCL11(嗜酸性粒细胞趋化因子-1)抗体),贝索单抗(别名:Scintimun,抗CEA-相关抗原抗体),贝伐单抗(别名:阿瓦斯丁,抗VEGF-A抗体)比西单抗(别名:FibriScint,抗血纤维蛋白Ⅱβ链抗体),Bivatuzumab(抗CD44v6抗体),blinatumomab(别名:BiTE,抗CD19抗体),Brentuximab(CAC10,抗-CD30 TNFRSF8抗体),Briakinumab(抗-IL-12,IL-23抗体),康纳单抗(别名:Ilaris, 抗-IL-1抗体),Cantuzumab(别名:C242,抗CanAg抗体),卡罗单抗,卡妥索单抗(别名:removab,抗EpCAM,抗-CD3抗体),CC49(抗-TAG-72抗体),Cedelizumab(抗CD4抗体),赛妥珠单抗(别名Cimzia抗TNF-α抗体),西妥昔单抗(别名:爱必妥,IMC-C225,抗-EGFR抗体),西他土珠(抗-EpCAM抗体),Cixutumumab(抗-IGF-1抗体),克立昔单抗(抗CD4抗体),Clivatuzumab(抗MUC1抗体),Conatumumab(抗-TRAIL-R2抗体),CR6261(抗A型流感血凝素抗体),Dacetuzumab(抗-CD40抗体),达利珠单抗(别名:赛尼哌,抗CD25(IL-2受体的α链)抗体),Daratumumab(抗-CD38(环化ADP核糖水解酶)抗体),,狄诺塞麦(别名:Prolia,抗RANKL抗体),地莫单抗(抗B-淋巴瘤细胞抗体),阿托度单抗,Dorlixizumab,Ecromeximab(抗-GD3神经节苷脂抗体),依库丽单抗(别名:Soliris,抗-C5抗体),埃巴单抗(抗内毒素抗体),依决洛单抗(别名:Panorex,MAb17-1A,抗-EpCAM抗体),依法利珠单抗(别名:Raptiva,抗LFA-1(CD11a)抗体),依芬古单抗(别名:Mycograb,抗Hsp90抗体),Elotuzumab(抗SLAMF7抗体),伊斯利莫(抗IL-6抗体),恩莫单抗(抗ICAM-1(CD54)抗体),Epitumomab(抗episialin抗体),依帕珠单抗(抗-CD22抗体),Erlizumab(抗ITGB2(CD18)抗体),Ertumaxomab(别名:Rexomun,抗HER2/neu,CD3抗体),伊瑞西珠(别名:Abegrin,抗整合素αvβ3抗体),艾韦单抗(抗乙型肝炎表面抗原抗体),Fanolesomab(别名:NeutroSpec,抗CD15抗体),法拉莫单抗(抗干扰素受体抗体),Farletuzumab(抗叶酸受体1抗体),Felvizumab(抗呼吸道合胞病毒抗体),Fezakinumab(抗IL-22抗体),Figitumumab(抗IGF-1受体抗体),Fontolizumab(抗IFN-γ抗体),夫瑞韦如(抗狂犬病毒糖蛋白抗体),Fresolimumab(抗TGF-β抗体),加利昔单抗(抗CD80抗体),Gantenerumab(抗-β淀粉样蛋白抗体),Gavilimomab(抗-CD147(basigin)抗体),吉妥单抗(抗CD33抗体),Girentuximab(抗碳酸酐酶9抗体),Glembatumumab(别名:CR011,抗GPNMB抗体),戈利木单抗(别名:SIMPONI,抗TNF-α抗体),Gomiliximab(抗CD23(IgE受体)抗体),Ibalizumab(抗CD4抗体),替伊莫单抗(抗CD20抗体),伊戈伏单抗(别名:Indimacis-125,抗CA-125抗体),英西单抗(别名:Myoscint,抗心肌肌球蛋白抗体),英夫利昔单抗(别名:英利昔单抗,抗TNF-α抗体),Intetumumab (抗CD51抗体),伊诺莫单抗(抗CD25(IL-2受体α链)抗体),Inotuzumab(抗CD22抗体),易普利姆玛(抗CD152抗体),Iratumumab(抗CD30(TNFRSF8)抗体),Keliximab(抗CD4抗体),Labetuzumab(别名:CEA-Cide,抗CEA抗体),Lebrikizumab(抗IL-13抗体),Lemalesomab(抗NCA-90(粒细胞抗原)抗体),乐德木单抗(抗TGFβ-2抗体),来沙木单抗(抗TRAIL-R2抗体),利韦单抗(抗乙型肝炎表面抗原抗体),林妥珠单抗(抗CD33抗体),Lucatumumab(抗CD40抗体),鲁昔单抗(抗CD23(IgE受体)抗体),Mapatumumab(抗TRAIL-R1抗体),马司莫单抗(抗T-细胞受体抗体),马妥珠单抗(抗-EGFR抗体),美泊利单抗(别名:Bosatria,抗IL-5抗体),Metelimumab(抗TGFβ-1抗体),Milatuzumab(抗CD74抗体),Minretumomab(抗TAG-72抗体),米妥莫单抗(别名BEC-2,抗-GD3神经节苷脂抗体),Morolimumab(抗恒河猴因子抗体),莫维珠单抗(别名:NUMAX,抗呼吸道合胞病毒抗体),莫罗单抗-CD3(别名:OKT3 ORTHOCLONE,抗CD3抗体),他那可单抗(抗C242抗体),他那莫单抗(抗5T4抗体),那他珠单抗(别名:Tysabri,抗整合素α4抗体),奈巴库单抗(抗内毒素抗体),Necitumumab(抗EGFR抗体),Nerelimomab(抗TNF-α抗体),尼妥珠单抗(别名:Theracim,Theraloc,抗EGFR抗体),Nofetumomab,ocrelizumab(抗CD20抗体),奥度莫单抗(别名:Afolimomab,抗LFA-1(CD11A)抗体),木单抗(别名:Arzerra,抗CD20抗体),Olaratumab(抗PDGF-Rα抗体),奥马珠单抗(别名:索雷尔,抗IgE Fc区抗体),Oportuzumab(抗EpCAM抗体),奥戈伏单抗(别名:OvaRex,抗CA-125抗体),Otelixizumab(抗CD3抗体),帕吉昔单抗(抗脂磷壁酸抗体),帕利珠单抗(别名:Synagis,Abbosynagis,抗呼吸道合胞病毒抗体),帕尼单抗(别名:维克替比,ABX-EGF,抗-EGFR抗体),Panobacumab(抗绿脓杆菌抗体),Pascolizumab(抗IL-4抗体),Pemtumomab(别名:Theragyn,抗MUC1抗体),帕妥珠单抗(别名:OMNITARG,2C4,抗HER2/neu抗体),培克珠单抗(抗C5抗体),Pintumomab(抗腺癌抗原抗体),普立昔单抗(抗CD4抗体),普托木单抗(抗波形蛋白抗体),PRO140(抗CCR5抗体),Racotumomab(别名:1E10,抗-(N-羟乙酰神经氨酸(NeuGc,NGNA)-神经节苷脂GM3)抗体),瑞非韦鲁(抗狂 犬病毒糖蛋白抗体),Ramucirumab(抗VEGFR2抗体),雷珠单抗(别名:Lucentis,抗VEGF-A抗体),瑞西巴库(抗炭疽毒素,保护性抗原抗体),瑞加韦单抗(抗巨细胞病毒的糖蛋白B抗体),Reslizumab(抗IL-5抗体),Rilotumumab(抗HGF抗体),利妥昔单抗(别名:美罗华,Rituxanmab,抗CD20抗体),Robatumumab(抗IGF-1受体抗体),Rontalizumab(抗IFN-α抗体),Rovelizumab(别名:LeukArrest,抗CD11,CD18抗体),Ruplizumab(别名:Antova,抗CD154(CD40L)抗体),沙妥莫单抗(抗TAG-72抗体),司韦单抗(抗巨细胞病毒抗体),Sibrotuzumab(抗FAP抗体),Sifalimumab(抗IFN-α抗体),Siltuximab(抗IL-6抗体),Siplizumab(抗CD2抗体),(Smart)MI95(抗CD33抗体),Solanezumab(抗β-淀粉样蛋白抗体),Sonepcizumab(抗神经鞘氨醇-1-磷酸抗体),索土珠单抗(抗episialin抗体),司他莫鲁(抗肌肉生长抑制素抗体),硫索单抗(别名:LeukoScan,(抗-NCA-90(粒细胞抗原)抗体))),Tacatuzumab(抗α-甲胎蛋白抗体),他度珠单抗(抗整合素αIIbβ3抗体),他利珠单抗(抗IgE抗体),他尼珠(抗NGF抗体),Taplitumomab(抗CD19抗体),Tefibazumab(别名:Aurexis,抗聚集因子A抗体),阿替莫单抗,Tenatumomab(抗固生蛋白C抗体),替奈昔单抗(抗CD40抗体),Teplizumab(抗CD3抗体),TGN1412(抗CD28抗体),Ticilimumab(别名:Tremelimumab(抗CTLA-4抗体),Tigatuzumab(抗TRAIL-R2抗体),TNX-650(抗IL-13抗体),托珠单抗(别名Atlizumab,Actemra,RoActemra,(抗IL-6受体抗体),Toralizumab(抗CD154(CD40L)抗体),托西莫单抗(抗CD20抗体),曲妥珠单抗(赫赛汀,(抗HER2/neu蛋白抗体),Tremelimumab(抗CTLA-4抗体),Tucotuzumab celmoleukin(抗EpCAM抗体),妥韦单抗(抗B型肝炎病毒抗体),Urtoxazumab(抗大肠杆菌抗体),优斯它单抗(别名:Stelara,抗IL-12,IL-23抗体),伐利昔单抗(抗AOC3(VAP-1)抗体),Vedolizumab,(抗整合素α4β7抗体),Veltuzumab(抗CD20抗体),维帕莫单抗(抗AOC3(VAP-1)抗体,Visilizumab(别名:Nuvion,抗CD3抗体),Vitaxin(抗血管整合素avb3抗体),Volociximab(抗整合素α5β1),伏妥莫单抗(别名:HumaSPECT,抗肿瘤抗原CTAA16.88抗体),扎妥木单抗(别名:HUMAX-EGFR,(抗EGFR抗体),Zanolimumab(别名:HUMAX-CD4,抗CD4抗体),Ziralimumab(抗CD147(基础免疫球蛋白) 抗体),阿佐莫单抗(抗CD5抗体),依那西普
Figure PCTCN2019093946-appb-000102
阿法赛特
Figure PCTCN2019093946-appb-000103
阿巴西普
Figure PCTCN2019093946-appb-000104
利纳西普(ARCALYST),14F7[抗IRP-2(铁离子调节蛋白2)抗体],14G2a(抗GD2神经节苷脂抗体,治疗黑色素瘤和实体瘤,from nat.cancer inst.),J591(抗PSMA抗体,治疗前列腺癌,威尔康乃尔医学院),225.28S[抗HMW-MAA(高分子量黑色素瘤相关抗原)抗体,索林Radiofarmaci SRL(意大利米兰)治疗黑色素瘤],COL-1(抗CEACAM3抗体,CGM1,nat.cancer inst.。治疗大肠癌和胃癌),CYT-356(
Figure PCTCN2019093946-appb-000105
治疗前列腺癌),HNK20(OraVax公司,用于治疗呼吸道合胞病毒),ImmuRAIT(来自IMMUNOMEDICS治疗非霍奇金淋巴瘤),Lym-1(抗HLA-DR10抗体,百富勤医药,用于癌症),MAK-195F[抗TNF抗体(又称:肿瘤坏死因子;TNFA,肿瘤坏死因子-α;TNFSF2),阿伯特/诺尔,用于治疗败血症中毒性休克],MEDI-500[别名:T10B9,抗CD3抗体,TRαβ(T细胞受体α/β),复合物,MedImmune公司用于治疗移植物抗宿主病],RING SCAN[抗TAG72(肿瘤相关糖蛋白72抗体),Neoprobe集团,用于治疗乳腺癌、结肠癌和直肠癌。Avicidin(抗-EpCAM抗体(上皮细胞粘附分子),抗TACSTD1抗体(肿瘤相关钙信号转导1),抗GA733-2(胃肠肿瘤相关蛋白2),抗EGP-2抗体(上皮糖蛋白2);抗KSA抗体;KS1/4抗原;M4S;肿瘤抗原17-1A;CD326,来自NeoRx公司用于治疗结肠癌,卵巢癌,前列腺癌和非霍奇金淋巴瘤;LymphoCide(IMMUNOMEDICS公司,NJ),smart ID10(Protein Design Labs),Oncolym(Techniclone公司,加利福尼亚州),Allomune(BioTransplant,CA),抗VEGF抗体(Genentech公司,CA);CEAcide(IMMUNOMEDICS公司,NJ),IMC-1C11(ImClone,NJ)和西妥昔单抗(ImClone公司,新泽西州)。
用于结合抗原的其它抗体包括(但不仅限于此):氨肽酶N(CD13),膜联蛋白A1,B7-H3(CD276,各种癌症),CA125,CA15-3(癌),CA19-9(癌),L6(癌),Lewis Y(癌),Lewis X(癌),甲胎蛋白(癌),CA242,胎盘碱性磷酸酶(癌),前列腺特异性抗原(前列腺癌),前列腺酸性磷酸酶(前列腺),表皮生长因子(癌),CD2(霍奇金病,非霍奇金淋巴瘤的淋巴瘤,多发性骨髓瘤),CD3的ε(T细胞淋巴瘤,肺癌,乳腺癌,胃癌,卵巢癌,自身免疫性疾病,恶性腹水),CD19(B细胞恶性肿瘤),CD20(非霍奇金 淋巴瘤),CD22(白血病,淋巴瘤,多发性骨髓瘤,系统性红斑狼疮),CD30,CD33,CD37,CD38(多发性骨髓瘤),CD40(淋巴瘤,多发性骨髓瘤,白血病),CD51(转移性黑素瘤,肉瘤),CD52,CD56(小细胞肺癌癌,卵巢癌,Merkel细胞癌,以及液体肿瘤,多发性骨髓瘤),CD66e(癌症),CD70(转移性肾细胞癌和非霍奇金淋巴瘤),CD74(多发性骨髓瘤),CD79,CD80(淋巴瘤),CD98(癌症),粘蛋白(癌),CD221(实体瘤),CD227(乳腺癌,卵巢癌),CD262(非小细胞肺癌和其他癌),CD309(卵巢癌),CD326(实体瘤),CEACAM3(大肠癌,胃癌),CEACAM5(癌胚抗原;CEA,CD66e)(乳腺癌,结肠直肠癌和肺癌),DLL4(Δ状-4),EGFR(表皮生长因子受体,各种癌症),CTLA4(黑色素瘤),CXCR4(CD184,血红素肿瘤,实体肿瘤),内皮糖蛋白(CD105,实体瘤),EPCAM(上皮细胞粘附分子,膀胱癌,头,颈,结肠,前列腺非霍奇金淋巴瘤,和卵巢癌),ERBB2(表皮生长因子受体2;肺癌,乳腺癌,前列腺癌),FCGR1(自身免疫性疾病),FOLR(叶酸受体,卵巢癌),GD2神经节苷脂(癌症),G-28(一种细胞表面抗原glyvolipid,黑素瘤),独特型GD3(癌症),热休克蛋白(癌症),HER1(肺,胃癌),HER2(乳腺癌,肺癌和卵巢癌),HLA-DR10(NHL),HLA-DRB(非霍奇金淋巴瘤,B细胞白血病),人绒毛膜促性腺激素(癌),IGF1R(胰岛素样生长因子1受体,实体瘤,血液癌症),IL-2受体(白细胞介素2受体,T-细胞白血病和淋巴瘤),IL-6R(白细胞介素6受体,多发性骨髓瘤,类风湿性关节炎,Castleman病,IL6依赖性肿瘤),整合素(素αvβ3,α5β1,α6β4,αllβ3,α5β5,αvβ5细胞附着因子,对各种癌症),MAGE-1(癌),MAGE-2(癌),MAGE-3(癌),MAGE4(癌),抗转铁蛋白受体(癌),P97(黑色素瘤),MS4A1(跨膜域4亚科A成员1,非霍奇金B细胞淋巴瘤,白血病),MUC1或MUC1-KLH(乳腺癌,卵巢癌,子宫颈癌,支气管和胃肠道癌症),MUC16(CA125)(卵巢癌),CEA(大肠),GP100(黑色素瘤),MART1(黑色素瘤)MPG(黑色素瘤),MS4A1(跨膜域4蛋白A,小细胞肺癌,非霍奇金淋巴瘤),核仁,神经癌基因产物(癌),P21(癌),抗(N-羟乙酰神经氨酸,乳腺癌,黑色素瘤的癌症),PLAP样睾丸碱性磷酸酶(卵巢癌,睾丸癌),PSMA(前列腺肿瘤),PSA(前列腺),ROBO4,TAG 72(肿瘤相关糖蛋白72,白血病,胃癌,结肠直肠癌,卵巢癌)中,T细胞的跨膜蛋白(癌症),Tie(CD202b), TNFRSF10B(肿瘤坏死因子受体超家族成员10B,癌症),TNFRSF13B(肿瘤坏死因子受体超家族成员13B,多发性骨髓瘤,非霍奇金淋巴瘤,以及其他癌症,类风湿性关节炎和系统性红斑狼疮),TPBG(滋养层糖蛋白,肾细胞癌),TRAIL-R1(肿瘤坏死凋亡诱导配体受体1,淋巴瘤,非霍奇金淋巴瘤,大肠癌,肺癌),VCAM-1(CD106,黑色素瘤),血管内皮生长因子,血管内皮生长因子-A,VEGF-2(CD309)(各种癌症)。通过抗体识别的一些其他的肿瘤相关抗原已审阅(Gerber等,mAbs 1:3,247-253(2009);Novellino等,cancer immunol immunother.54(3),187-207(2005)Franke等,cancer biother radiopharm.2000,15,459-76)。还有许多其他的抗原是:其他不同的集群(CD1,CD1a,CD1b,CD1c,CD1d,CD1e,CD2,CD3,CD3d,CD3e,CD3g,CD4,CD5,CD6,CD7,CD8,CD8a,CD8b,CD9,CD10,CD11a,CD11b,CD11c,CD11d,CD12w,CD14,CD15,CD16,CD16a,CD16b,CDw17,CD18,CD19,CD20,CD21,CD22,CD23,CD24,CD25,CD26,CD27,CD28,CD29,CD30,CD31,CD32,CD32a,CD32b,CD33,CD34,CD35,CD36,CD37,CD38,CD39,CD40,CD41,CD42,CD42a,CD42b,CD42c,CD42d,CD43,CD44,CD45,CD46,CD47,CD48,CD49b,CD49c,CD49c,CD49d,CD49f,CD50,CD51,CD52,CD53,CD54,CD55,CD56,CD57,CD58,CD59,CD60,CD60a,CD60b,CD60c,CD61,CD62E,CD62L,CD62P,CD63,CD64,CD65,CD65s,CD66,CD66a,CD66b,CD66c,CD66d,CD66e,CD66f,CD67,CD68,CD69,CD70,CD71,CD72,CD73,CD74,CD75,CD75s,CD76,CD77,CD78,CD79,CD79a,CD79b,CD80,CD81,CD82,CD83,CD84,CD85,CD85a,CD85b,CD85c,CD85d,CD85e,CD85f,CD85g,CD85g,CD85i,CD85j,CD85k,CD85m,CD86,CD87,CD88,CD89,CD90,CD91,CD92,CD93,CD94,CD95,CD96,CD97,CD98,CD99,CD100,CD101,CD102,CD103,CD104,CD105,CD106,CD107,CD107a,CD107b,CD108,CD109,CD110,CD111,CD112,CD113,CD114,CD115,CD116,CD117,CD118,CD119,CD120,CD120a,CD120b,CD121,CD121a,CD121b,CD122,CD123,CD123a,CD124,CD125,CD126,CD127,CD128,CD129,CD130,CD131,CD132,CD133,CD134,CD135,CD136,CD137,CD138,CD139,CD140,CD140a,CD140b,CD141,CD142,CD143,CD144,CD145,CDw145,CD146,CD147,CD148,CD149,CD150,CD151,CD152,CD153,CD154, CD155,CD156,CD156a,CD156b,CD156c,CD156d,CD157,CD158,CD158a,CD158b1,CD158b2,CD158c,CD158d,CD158e1,CD158e2,CD158f2,CD158g,CD158h,CD158i,CD158j,CD158k,CD159,CD159a,CD159b,CD159c,CD160,CD161,CD162,CD163,CD164,CD165,CD166,CD167,CD167a,CD167b,CD168,CD169,CD170,CD171,CD172,CD172a,CD172b,CD172g,CD173,CD174,CD175,CD175s,CD176,CD177,CD178,CD179,CD179a,CD179b,CD180,CD181,CD182,CD183,CD184,CD185,CD186,CDw186,CD187,CD188,CD189,CD190,CD191,CD192,CD193,CD194,CD195,CD196,CD197,CD198,CD199,CDw198,CDw199,CD200,CD201,CD202,CD202(a,b),CD203,CD203c,CD204,CD205,CD206,CD207,CD208,CD209,CD210,CDw210a,CDw210b,CD211,CD212,CD213,CD213a1,CD213a2,CD214,CD215,CD216,CD217,CD218,CD218a,CD218,CD21b9,CD220,CD221,CD222,CD223,CD224,CD225,CD226,CD227,CD228,CD229,CD230,CD231,CD232,CD233,CD234,CD235,CD235a,CD235b,CD236,CD237,CD238,CD239,CD240,CD240ce,CD240d,CD241,CD242,CD243,CD244,CD245,CD246,CD247,CD248,CD249,CD250,CD251,CD252,CD253,CD254,CD255,CD256,CD257,CD258,CD259,CD260,CD261,CD262,CD263,CD264,CD265,CD266,CD267,CD268,CD269,CD270,CD271,CD272,CD273,CD274,CD275,CD276,CD277,CD278,CD279,CD281,CD282,CD283,CD284,CD285,CD286,CD287,CD288,CD289,CD290,CD291,CD292,CD293,CD294,CD295,CD296,CD297,CD298,CD299,CD300,CD300a,CD300b,CD300c,CD301,CD302,CD303,CD304,CD305,CD306,CD307,CD307a,CD307b,CD307c,CD307d,CD307e,CD307f,CD308,CD309,CD310,CD311,CD312,CD313,CD314,CD315,CD316,CD317,CD318,CD319,CD320,CD321,CD322,CD323,CD324,CD325,CD326,CD327,CD328,CD329,CD330,CD331,CD332,CD333,CD334,CD335,CD336,CD337,CD338,CD339,CD340,CD341,CD342,CD343,CD344,CD345,CD346,CD347,CD348,CD349,CD350,CD351,CD352,CD353,CD354,CD355,CD356,CD357,CD358,CD359,CD360,CD361,CD362,CD363,CD364,CD365,CD366,CD367,CD368,CD369,CD370,CD371,CD372,CD373,CD374,CD375,CD376,CD377,CD378, CD379,CD381,CD382,CD383,CD384,CD385,CD386,CD387,CD388,CD389,CRIPTO,CR,CR1,CRGF,CRIPTO,CXCR5,LY64,TDGF1,4-1BB,APO2,ASLG659,BMPR1B,4-1BB,5AC,5T4),APO2,ASLG659,BMPR1B(骨形态发生蛋白受体),CRIPTO,膜联蛋白A1,核仁,内皮糖蛋白(CD105),ROBO4,氨基肽酶N,Δ-样3(DLL3),Δ-样4(DLL4),VEGFR-2(CD309),CXCR4 9CD184),Tie2,B7-H3,WT1,MUC1,LMP2,HPV E6 E7,EGFRvIII,HER-2/neu,个体基因型,MAGE A3,P53 nonmutant,NY-ESO-1,GD2,CEA,MelanA/MART1,Napi3b(NAPI-3B,NPTIIb,SLC34A2,溶质载体家族34,成员2,II型钠依赖的磷转运3b),Ras突变,gp100,p53突变体,Proteinase3(PR1),BCR-abl,Tetratocarcinoma衍生的生长因子),EphA受体,EphB受体,EGFR,EGFRvIII,ETBR(内皮素),HER2/neu,HER3,HLA-DOB(MHC II类分子IA抗原),整合素,IRTA2,MPF(MPF,MSLN,SMR,巨核细胞强化因子,间皮素),CRIPTO,Sema 5b(FLJ10372,KIAA1445,Mm42015,SEMA5B,5EMAG,semaphoring 5 bHlog,sdema域,七个血小板重复序列,胞浆区),PSCA,STEAP1(6个跨膜上皮前列腺抗原),和STEAP2(HGNC 8639,IPCA-1,PCANP1,STAMP1,STEAP2,STMP,前列腺),酪氨酸酶,生存素,hTERT,肉瘤易位断点,EphA2,PAP,ML-IAP,AFP,EpCAM,ERG(TMPRSS2 ETS融合基因),NA17,PAX3,ALK,雄激素受体,细胞周期素B1,聚唾液酸,MYCN,RhoC,TRP-2,GD3,岩藻糖神经节苷脂,间皮素,PSMA,MAGE A1,sLe(a),CYP1B1,PLAC1,GM3,BORIS,Tn,GloboH,ETV6-AML,NY-BR-1,RGS5,SART3,STn,碳酸酐酶IX,PAX5,OY-TES1,精子蛋白17,LCK,HMWMAA,AKAP-4,SSX2,XAGE 1,B7H3,豆荚蛋白,Tie 2,Trop2,Page4,VEGFR2,MAD-CT-1,FAP,PDGFR-β,MAD-CT-2,Fos蛋白相关抗原1。
本发明的偶联物适用于癌症治疗。这些癌症包含,并不限于,肾上腺皮质癌,直肠癌,膀胱癌,脑肿瘤(成人:脑干胶质瘤,儿童,小脑星形细胞瘤,星形细胞瘤,室管膜瘤,髓母细胞瘤,幕上原始神经外胚层肿瘤,松果体,视觉通路和下丘脑胶质瘤),乳腺癌,类癌肿瘤,胃肠道,未知原 发癌,宫颈癌,结肠癌,子宫内膜癌,食管癌,肝外胆管癌,尤文家族肿瘤(PNET),颅外恶性生殖细胞肿瘤,眼癌,眼内黑色素瘤,胆囊癌,胃癌(胃),生殖细胞肿瘤,性腺外,妊娠滋养细胞肿瘤,头颈部肿瘤,下咽癌,胰岛细胞癌,肾癌(肾细胞癌),喉癌,白血病(急性淋巴细胞,急性髓细胞,慢性淋巴细胞,慢性髓细胞,多毛细胞),唇和口腔癌,肝癌,肺癌(非小细胞,小细胞,淋巴瘤(爱滋病相关,中枢神经系统,皮肤T细胞,霍奇金病,非霍奇金病,恶性间皮瘤,黑色素瘤,默克尔细胞癌,原发性隐匿性鳞状颈部转移癌,多发性骨髓瘤和其他浆细胞肿瘤,蕈样肉芽肿,骨髓增生异常综合征,骨髓增殖性疾病,鼻咽癌,神经母细胞瘤,口腔癌,口咽癌,骨肉瘤,卵巢癌(上皮细胞,生殖细胞瘤,低度恶性潜能肿瘤),胰腺癌(外分泌,胰岛细胞癌),鼻窦和鼻腔癌,甲状旁腺癌,阴茎癌,嗜铬细胞瘤癌,垂体肿瘤,浆细胞肿瘤,前列腺癌横纹肌肉瘤,直肠癌,肾细胞癌(肾癌),肾盂和输尿管(移行细胞),涎腺癌,塞扎综合症,皮肤癌,皮肤癌(皮样T细胞淋巴瘤,卡波西氏肉瘤,黑色素瘤),小肠癌,软组织肉瘤,胃癌,睾丸癌,胸腺瘤(恶性),甲状腺癌,尿道癌,子宫癌(肉瘤),儿童异常肿瘤,阴道癌,外阴癌,肾母细胞瘤。
本发明的偶联物适用于自身免疫疾病的预防和治疗。自身免疫疾病包括,但不限于,胃酸缺乏自身免疫性慢性活动性肝炎,急性播散性脑脊髓炎,急性出血性脑白质炎,阿狄森氏病,丙种球蛋白血症,斑秃,肌萎缩性侧索硬化症,强直性脊柱炎,抗肾小球基底膜/肾小管基底膜肾炎,抗磷脂综合征,抗合成酶综合征,关节炎,异位性过敏症,过敏性皮炎,自身免疫性再生障碍性贫血,自身免疫性心肌病,自身免疫性溶血性贫血,自身免疫性肝炎,自身免疫性内耳病,自身免疫性淋巴组织增生综合征,自身免疫性周围神经系统疾病,自身免疫性胰腺炎自体免疫多内分泌症I,II,III型,自身免疫性孕酮皮炎,自身免疫性血小板减少性紫癜,自身免疫性色素层炎,巴洛病/巴洛同心性硬化,黑奇特综合征,Berger病,Bickerstaff脑干脑炎,Blau综合征,大苞性类天苞疮,巨淋巴结增生,美洲锥虫病,慢性疲劳免疫功能紊乱综合征,慢性炎症性脱髓鞘性多发神经病,慢性复发性多病灶性骨髓炎,慢性莱姆病,慢性阻塞性肺疾病,变应性肉芽肿性血管炎,瘢痕性类天疱疮,腹部疾病,耳蜗前庭综合征,冷凝集素病,补体C2缺乏症,颅动脉炎,肢端硬皮综合征,克隆氏病(一种特发性炎症性 肠病),柯兴氏症,皮肤白细胞碎裂性血管炎,恶性萎缩性丘疹病,痛性肥胖病,疱疹样皮炎,皮肌炎,1型糖尿病,弥漫性皮肤硬皮病,心肌梗塞后综合征,盘状红斑狼疮,湿疹,子宫内膜异位症,幼年特发性关节炎,嗜酸细胞性筋膜炎,嗜酸细胞性筋膜炎,结节性红斑,特发性混合性冷球蛋白血症,埃文斯综合征,渐进性骨化性纤维组织结构不良,纤维肌痛,纤维肌炎,纤维性肺泡炎,胃炎,胃肠类天疱疮,巨细胞动脉炎,肾小球肾炎,肺出血肾炎综合征,格雷夫斯氏病,格林-巴利神经根炎,桥本脑炎,淋巴瘤性甲状腺肿,溶血性贫血,过敏性紫癜多,妊娠期疱疹,化脓性汗腺炎,休斯综合症(抗磷脂抗体综合征),低丙球蛋白血症,特发性炎性脱髓鞘疾病,特发性肺纤维化,特发性血小板减少性紫癜(自身免疫性血小板减少性紫癜),IgA肾病(Berger病),包涵体肌炎,发炎性脱髓鞘病变,间质性膀胱炎,肠易激综合征,幼年特发性关节炎,幼儿型类风湿性关节炎,皮肤粘膜淋巴结综合征,兰伯特肌无力综合征,白细胞分裂性血管炎,扁平苔藓,硬化性苔藓,线状IgA病(LAD),肌萎缩性侧索硬化症,狼疮样肝炎,红斑狼疮,马吉德综合征,美尼尔氏综合症,显微镜下多血管炎,米勒·费希尔综合征,混合性结缔组织病,硬斑病,穆哈二氏病,韦尔斯综合征,多发性骨髓瘤,多发性硬化症,重症肌无力,肌炎,发作性睡病,视神经脊髓炎(德维克病),神经性肌强直,眼部瘢痕性类天疱疮,眼阵挛-肌阵挛综合征,奥德氏甲状腺炎,复发性风湿病,熊猫症候群(合并链球菌感染的儿童自体免疫神经精神异常),类肿瘤性小脑变性,阵发性睡眠性血红蛋白尿症,进行性一侧面萎缩,巴-特二氏综合征,睫状体扁平部炎,天疱疮,寻常型天疱疮,恶性贫血,静脉周围炎,POEMS综合征,结节性多动脉炎,风湿性多肌痛,多发性肌炎,原发性胆汁性肝硬化,原发性硬化性胆管炎,进行性炎性神经病变,银屑病,银屑病关节炎,坏疽性脓皮,纯红细胞再生障碍性贫血,罗斯默森氏脑炎,雷诺病,复发性多软骨炎,莱特尔综合征,腿多动综合征,腹膜后纤维化,类风湿关节炎,类风湿发热,结节病,精神分裂症,施密特综合征,施尼茨勒综合征,巩膜炎,硬皮病,干燥综合征,脊椎关节病,粘血综合征,斯蒂尔病,僵人综合征,亚急性细菌性心内膜炎,Susac氏综合征,急性热性嗜中性皮肤病,西德纳姆舞蹈病,交感性眼炎,高安氏动脉炎,颞动脉炎(巨细胞动脉炎),痛性眼肌麻痹综合征,横贯性脊髓炎,溃疡性结肠炎(一种特发性炎症性肠疾病),未分化 的结缔组织病,未分化脊柱关节病,血管炎,白癜风,韦格纳氏肉芽肿病,威尔逊氏综合征,威斯科特-奥尔德里奇综合征。
在另一具体实施方案中,用于治疗或预防自身免疫疾病的用于偶联的抗原结合分子包括,但不限于:抗弹性蛋白抗体;Abys抗上皮细胞抗体;抗基底膜IV型胶原蛋白抗体;抗核抗体;抗双链DNA抗体;抗单链DNA抗体,抗心肌磷脂抗体IgM,IgG;抗celiac(anti-celiac antibody)抗体;抗磷脂抗体IgK,IgG;抗核糖蛋白抗体;抗线粒体抗体;甲状腺抗体;微粒体抗体,T-细胞抗体;甲状腺球蛋白抗体,抗硬皮病-70抗体(anti-SCL-70);人抗Jo抗体(anti-jo);抗系统性红斑狼疮患者自身抗体;抗干燥综合症抗体(Anti-La/SSB);抗系统性红斑狼疮抗体;抗壁细胞抗体;抗组蛋白抗体;抗核糖核蛋白抗体(anti-RNP);中性粒细胞胞质抗体(C-ANCA);细胞核周围抗中性粒细胞抗体(P-ANCA);抗着丝粒抗体;抗核纤维蛋白抗体,以及抗肾小球基底膜抗体(GBM)抗体,抗神经节苷脂(Anti-ganglioside)抗体;抗桥粒芯糖蛋白3抗体(anti-Desmogein 3);抗人P62抗体;抗人sp100抗体;抗线粒体M2抗体;类风湿因子抗体;抗突变型瓜氨酸波形蛋白抗体(anti-MCV);抗拓扑异构酶抗体;抗中性粒细胞胞浆(CANCA)抗体。
在某些优选的实施方案中,本发明中用于共轭偶联的结合分子,可以与自身免疫性疾病相关的活化淋巴细胞表达的受体或受体复合物结合。包括免疫球蛋白基因超家族成员(如CD2,CD3,CD4,CD8,CD19,CD20,CD22,CD28,CD30,CD33,CD37,CD38,CD70,CD79,CD79b,CD90,CD123,CD125,CD138,CD152/CTLA-4,PD-1,或ICOS),TNF受体超家族成员(如CD27,CD40,CD95/Fas,CD134/OX40,CD137/4-1BB,INF-R1,TNFR-2,RANK,TACI,BCMA,骨保护素,Apo2/TRAIL-R1,TRAIL-R2,TRAIL-R3,TRAIL-R4,Trop2和30 APO-3),整联蛋白,细胞因子受体,趋化因子受体,主要组织相容性蛋白,凝集素(C型,S型或I型)或补体调控蛋白。
在另一具体实施方案中,对病毒性或细菌性抗原具有免疫特异性的有用的结合体是人源的或人单克隆抗体。如本文所用术语“病毒性抗原”包括,但不限于:任何能诱发免疫反应的病毒肽段,多肽蛋白(例如,HIVgp120,HIV nef,RSV F糖蛋白,流感病毒神经氨酸酶,流感病毒血凝素,人类T淋巴细胞病毒感染调节因子tax,单疱疹病毒糖蛋白(例如,gB,gC, gD和gE)和乙型肝炎表面抗原)。如本文所用的术语“细菌性抗原”包括,但不限于:任何能诱发免疫反应的微生物肽段,多肽蛋白,糖类,多糖,脂质分子(例如,细菌,真菌,致病性原生动物,酵母多肽包括,例如,脂多糖和荚膜多糖5/8)。可用于治疗病毒性或细菌性感染的有用I型抗体包括,但不限于:帕利珠单抗,一种用于治疗RSV感染的人源的抗呼吸道合胞体病毒单克隆抗体;PRO542,一种用于治疗HIV感染的CD4融合抗体;Ostavir,一种治疗乙肝病毒人源性抗体;PROTVIR,一种用于治疗巨细胞病毒人源化抗体IgG.sub.1,还有抗脂多糖(anti-LPS)抗体。
本发明的偶联物可以用于治疗感染性疾病。这些传染病包括,但不限于:不动杆菌感染,放线菌病,非洲昏睡病(非洲锥虫病),艾滋病(获得性免疫缺陷综合症),阿米巴病,微粒孢子虫病,炭疽,溶血隐秘杆菌感染,阿根廷出血热,蛔虫病,曲霉菌病,星状病毒感染,巴贝虫病,蜡样芽胞杆菌感染,细菌性肺炎,细菌性阴道病,类杆菌感染,小袋虫病,贝利蛔线虫感染,BK病毒感染,黑色发结节病,人芽囊原虫感染,芽生菌,玻利维亚出血热,疏螺旋体感染,肉毒中毒(和婴儿肉毒中毒),巴西出血热,布氏杆菌病,伯霍尔德杆菌感染,布鲁里溃疡,感染杯状病毒(诺罗病毒和札幌病毒),弯曲菌病,念珠菌感染(念珠菌病,鹅口疮),猫抓病,蜂窝组织炎,查格斯病(美洲锥虫病),软下疳,水痘,衣原体,肺炎衣原体感染,霍乱,着色真菌病,肝吸虫病,难辨梭状芽孢杆菌感染,球孢子菌病,科罗拉多蜱热,普通感冒(急性病毒鼻咽炎;急性鼻炎),克雅氏病,克里米亚-刚果出血热,隐球菌病,隐孢子虫病,皮肤幼虫移行症,环孢子虫感染,囊虫病,巨细胞病毒感染,登革热,双核阿米巴病,白喉,裂头绦虫,龙线虫病,埃博拉出血热,包虫病,埃里希体病,蛲虫病(蛲虫感染),肠球菌感染,肠病毒感染,流行性斑疹伤寒,传染性红斑(第五病),幼儿急疹,姜片虫病,家族致命性失眠症,丝虫病,产气荚膜梭菌引起的食物中毒,非寄生阿米巴感染,梭杆菌感染,气性坏疽(梭菌性肌坏死),地丝菌病,吉斯特曼-施特劳斯综合征,贾第虫病,马鼻疽,腭口线虫病,淋病,腹股沟肉芽肿,A组链球菌感染,B组链球菌感染,流感嗜血杆菌感染,手足口病(HFMD),汉坦病毒肺综合征,幽门螺杆菌感染,溶血性尿毒综合征,出血热肾病综合征,甲型肝炎,乙型肝炎,丙型肝炎,丁型肝炎,戊型肝炎,单纯疱疹,组织胞浆菌病,钩虫感染,人鲍坎病毒感染,人埃 文氏埃立克体病,人粒细胞无形体病,人类偏肺病毒感染,人体单核细胞埃立克体病,人乳头状瘤病毒感染,人副流感病毒感染,膜壳绦虫病,爱泼斯坦-巴尔病毒传染性单核细胞增多症(单一),流感,等孢子球虫病,川崎病,角膜炎,金氏金氏杆菌感染,库鲁病,拉沙热,军团杆菌病(退伍军人协会会员病),军团杆菌病(庞蒂亚克热),利什曼病,麻风病,钩端螺旋体病,李氏杆菌病,莱姆病(莱姆疏螺旋体病),淋巴丝虫病(象皮肿),淋巴细胞性脉络丛脑膜炎,疟疾,马尔堡出血热,麻疹,类鼻疽(惠特莫尔氏病),脑膜炎,脑膜炎球菌病,后殖吸虫病,微孢子虫病,传染性软疣,流行性腮腺炎,斑疹伤寒(地方性斑疹伤寒),支原体肺炎,足菌肿,蝇蛆病,新生儿结膜炎(新生儿眼炎),克—亚综合征(vCJD,nvCJD),诺卡氏菌病,盘尾丝虫病(河盲症),副球孢子菌病(南美芽生菌病),肺吸虫病,巴氏杆菌病,头虱病(头虱),体虱病(体虱),阴虱病(阴虱),盆腔炎,百日咳,鼠疫,肺炎球菌感染,肺孢子虫性肺炎,肺炎,脊髓灰质炎,普氏菌感染,原发性阿米巴脑膜脑炎,进行性多病灶脑白质病,鹦鹉热,Q热,狂犬病,鼠咬热,呼吸道合胞病毒感染,鼻孢子菌病,鼻病毒感染,立克次体感染,立克次氏体,裂谷热,洛矶山斑疹热,轮状病毒感染,风疹,沙门氏菌病,SARS(严重急性呼吸系统综合症),疥疮,血吸虫病,败血症,痢疾(菌痢),带状疱疹(带状疱疹),天花(天花),孢子丝菌病,葡萄球菌食物中毒,葡萄球菌感染,线虫,梅毒,绦虫病,破伤风(牙关紧闭症),须癣,头癣,体癣,股癣,手癣,黑糠疹,足癣,甲癣,花斑癣,弓蛔虫病(眼幼虫移行症),弓蛔虫病(内脏幼虫移行症),弓形体病,旋毛虫病,滴虫病,鞭虫病(鞭虫感染),肺结核,兔热病,尿素分解尿素支原体感染,委内瑞拉马脑炎,委内瑞拉出血热,病毒性肺炎,西尼罗热,白色毛结节菌病(白色毛结节菌病),假结核菌感染,耶尔森菌病,黄热病,接合菌病。
本篇专利先前描述的抗体的结合分子可用于抗致病菌株,包括,但不限于:鲍曼不动杆菌,以色列放线菌,戈氏放线菌和丙酸丙酸盐杆菌,布氏锥虫,HIV(人类免疫缺陷病毒),溶组织内阿米巴,无形体属,炭疽杆菌,溶血隐秘杆菌,胡宁病毒,蛔虫,曲霉属,星状病毒家庭,巴贝斯虫属,蜡样芽胞杆菌,多杆菌,类杆菌属,结肠小袋纤毛虫,贝利蛔线虫属,BK病毒,何德毛结节菌,人芽囊原虫,皮炎芽生菌,沙粒病毒,疏螺旋体属,肉毒梭菌,清风藤,布鲁菌属,通常洋葱伯克霍尔德菌和其他伯克霍 尔德杆菌种,溃疡分枝杆菌,杯状病毒科家族,弯曲杆菌属,通常是白色念珠菌和其他念珠菌属,巴尔通体,A组链球菌和葡萄球菌,克氏锥虫,杜克雷嗜血杆菌,水痘带状疱疹病毒(VZV),沙眼衣原体,肺炎衣原体,霍乱弧菌,裴氏着色霉,华支睾吸虫,难辨梭状芽孢杆菌,粗球孢子菌和Coccidioides posadasii,科罗拉多蜱热病毒,鼻病毒,冠状病毒,朊病毒克雅氏病,克里米亚-刚果出血热病毒,新型隐球菌,隐孢子虫属,猫钩虫;multiple parasites,,环孢子,猪带绦虫,巨细胞病毒,登革热病毒(DEN-1,DEN-2,DEN-3和DEN-4)虫媒病毒,脆弱双核阿米巴,白喉棒状杆菌,裂头属,麦地那龙线虫,埃博拉病毒,棘球属,埃立克体属,蛲虫,肠球菌属,肠道病毒属,普氏立克次体,细小病毒B19,人类疱疹病毒6型和人类疱疹病毒7型,布氏姜片吸虫,肝片吸虫和大片吸虫,FFI朊病毒,丝虫目超家族,产气荚膜杆菌,梭杆菌属,产气荚膜梭菌,其他梭菌属,白地霉,GSS朊病毒,肠贾第鞭毛虫,鼻疽伯克霍尔德氏菌,棘颚口线虫和刚棘颚口虫,淋病奈瑟菌,肉芽肿杆菌,化脓性链球菌,无乳链球菌,流感嗜血杆菌,肠道病毒,大多数柯萨奇A病毒和肠病毒71型,辛诺瓦病毒,幽门螺旋杆菌,大肠杆菌O157:H7,布尼亚病毒科家族,甲型肝炎病毒,乙型肝炎病毒,丙型肝炎病毒,丁型肝炎病毒,戊型肝炎病毒,单纯疱疹病毒1型,单纯疱疹病毒2型,组织胞浆菌,十二指肠钩虫和美洲板口线虫,流感嗜血杆菌,人类博卡病毒,埃文氏埃立克体,无形体科,人类偏肺病毒,沙费埃里希体,人乳头瘤病毒,人副流感病毒,微小膜壳绦虫和缩小包膜绦虫,EB病毒,正粘病毒科,贝利等孢球虫,金氏金氏杆菌,肺炎克雷伯菌,Klebsiella ozaenas,库鲁病朊病毒,拉沙病毒,嗜肺军团菌,嗜肺军团菌,利什曼原虫属,麻风分枝杆菌和弥漫型痳疯分枝杆菌,钩端螺旋体属,李斯特菌,博氏包柔螺旋体和其他疏螺旋体属,班氏丝虫和马来丝虫,淋巴细胞性脉络丛脑膜炎病毒(LCMV),疟原虫属,马尔堡病毒,麻疹病毒,类鼻疽伯克氏菌,脑膜炎奈瑟菌,横川后殖吸虫,微孢子虫门,传染性软疣病毒(MCV),腮腺炎病毒,立克次体杆菌,肺炎支原体,多种细菌(马杜拉分枝菌病)和真菌(马杜拉分枝菌病),寄生双翅目蝇蛆,沙眼衣原体和淋球菌,vCJD朊病毒,星状诺卡氏菌和其他诺卡氏菌属物种,旋盘尾丝虫,巴西芽生菌,卫氏并殖吸虫和其他并殖吸虫属,巴斯德氏菌属,虱头癣,体虱,阴虱,百日咳博德特氏菌,鼠疫 耶尔森氏菌,肺炎链球菌,卡氏肺囊虫,脊髓灰质炎病毒,普雷沃氏菌属,福氏耐格里阿米巴,JC病毒,鹦鹉热衣原体,立克次体,狂犬病毒,念珠状链杆菌和鼠咬热螺旋体,呼吸道合胞病毒,西伯鼻孢子虫,鼻病毒,立克次体属,螨立克次体,立夫特山谷热病毒,立氏立克次体,轮状病毒,风疹病毒,沙门氏菌属,非典型肺炎冠状病毒,疥螨,血吸虫属,志贺氏菌属,水痘带状疱疹病毒,重型天花或类天花,申克孢子丝菌,葡萄球菌属,金黄色葡萄球菌,化脓性链球菌,粪类圆线虫,梅毒螺旋体,绦虫属,破伤风梭菌,毛癣菌属,断发毛癣菌,发癣菌属,絮状表皮癣菌,红色毛癣菌和须癣毛癣菌,威尼克外瓶霉,毛癣菌属,马拉色菌属,犬弓首蛔虫或猫弓蛔虫,弓形虫,旋毛虫,阴道毛滴虫,鞭虫,结核杆菌,土拉热弗朗西丝菌,解脲支原体,委内瑞拉马脑炎病毒,霍乱弧菌,瓜纳瑞托病毒,西尼罗病毒,白色毛孢子菌,假结核耶尔森菌,小肠结肠炎耶尔森菌,黄热病毒,毛霉目(毛霉菌病)和虫霉目(虫霉菌病),铜绿假单胞菌,胎儿弯曲杆菌(弧菌),嗜水气单胞菌,迟缓爱德华氏菌,耶尔森菌,志贺痢疾杆菌,福氏痢疾杆菌,宋内志贺氏菌,鼠伤寒沙门氏菌,雅司螺旋体,Treponema carateneum,奋森疏螺旋体,博氏包柔螺旋体,出血性黄疸钩端螺旋体,卡氏肺囊虫,布鲁氏菌,猪布鲁氏菌,布氏杆菌,支原体属,斑疹伤寒病原体,Rickettsia tsutsugumushi,披衣菌属;病原真菌(曲霉,白色念珠菌,荚膜组织胞浆菌);原生动物(溶组织内阿米巴,阴道毛滴虫,人毛滴虫,Tryoanosoma gambiense,罗得西亚锥虫,杜氏利什曼原虫,热带利什曼原虫,巴西利什曼原虫,肺囊虫肺炎,间日疟原虫,恶性疟原虫,恶性疟疾);或蠕虫(日本血吸虫,曼氏血吸虫,埃及血吸虫和钩虫)
其他抗体作为本发明结合配体用于治疗病毒性疾病包括,但不限于:抗体作用于病原病毒的抗原包括以下例子但不限于:天花病毒,疱疹病毒,腺病毒,乳多空病毒科,肠道病毒科,细小RNA病毒科,细小病毒科,呼肠孤病毒,逆转录病毒科,流感病毒,副流感病毒,腮腺炎,麻疹,呼吸道合胞病毒,风疹,虫媒病毒,弹状病毒,沙粒病毒科,Non-A/Non-B肝炎病毒,鼻病毒,冠状病毒,轮状病毒科,肿瘤病毒[例如,乙型肝炎病毒(肝细胞癌),人乳头状瘤病毒(宫颈癌,肛门癌),卡波西肉瘤相关疱疹病毒(卡波济氏肉瘤),EB病毒(鼻咽癌,伯基特淋巴瘤,原发性中枢神经系统淋巴瘤),MCPyV(梅克尔细胞癌),SV40(猿猴病毒40),丙型肝炎病毒(肝 癌),人类嗜T淋巴细胞病毒1型(成人T细胞白血病/淋巴瘤),免疫失调引起病毒:[如人类免疫缺陷病毒(艾滋病)];中枢神经系统病毒:[如,JCV(进行性多病灶脑白质病),MeV(亚急性硬化性全脑炎),LCV(淋巴细胞性脉络丛脑膜炎),虫媒病毒性脑炎,正粘病毒科(可能的)(昏睡性脑炎),RV(狂犬病),水疱性口炎-印度病毒属,疹病毒性脑膜炎,拉姆齐·亨特综合征II型;脊髓灰质炎(小儿麻痹症,后小儿麻痹症候群),人类嗜T淋巴细胞病毒1型(热带痉挛性截瘫)];巨细胞病毒(巨细胞病毒性视网膜炎,HSV(疱疹性角膜炎));心血管病病毒[如柯萨奇病毒(心包炎,心肌炎)];呼吸系统/急性鼻咽炎病毒/病毒性肺炎:[非洲淋巴细胞瘤病毒(疱疹病毒4型感染/传染性单核细胞增多),巨细胞病毒;SARS冠状病毒(严重急性呼吸器官综合征)正粘病毒:流感病毒A/B/C(流感/禽流感),副粘病毒:人类副流感病毒(副流感),呼吸道合胞病毒(人类呼吸道合胞病毒),肺病毒];消化系统病毒[MUV(腮腺炎),巨细胞病毒(巨细胞病毒性食管炎);腺病毒(腺病毒感染);轮状病毒,诺如病毒,星状病毒,冠状病毒;HBV(乙肝病毒),柯萨奇病毒,甲型肝炎(甲肝病毒),HCV(丙型肝炎病毒),HDV(丁型肝炎病毒),HEV(戊型肝炎病毒),HGV(庚型肝炎病毒)];泌尿生殖系统的病毒[如,BK病毒,MUV(腮腺炎)]。
按照更进一步的目标,本发明还包括上述偶联物成分结合其它可行的药物载体作为癌症和自身免疫疾病的治疗药物。本发明治疗癌症和自身免疫疾病的方法包括体外,体内或间接体内疗法。体外疗法应用实例,包括药物处理体外培养细胞,杀死除了没有表达目标抗原的细胞以外的所有细胞;或者杀死表达非想要的抗原的细胞。作为间接体内疗法的治疗方法的一个示例:在体外处理造血干细胞,杀死患病或恶性细胞后输回原患者体内。例如,临床上先通过间接体内治疗除去骨髓中的肿瘤细胞或淋巴细胞然后输回原患者来治疗癌症和自体免疫疾病,或者在移植前去除骨髓中的T细胞和其它淋巴细胞以防止移植物的免疫拮抗反应。实施方法如下:从病人或其他个体获取骨髓细胞,然后在加入本发明偶联物的含血清培养基中37℃培养,药物浓度范围为1pM到0.1mM,培养时间为30分钟左右到48小时左右。药物具体浓度和培养时间由经验丰富的临床医生决定。培养结束后,骨髓细胞用含血清培养基洗涤后通过静脉注射输回人体内。如果病人在获取骨髓细胞和回输处理前需要接受其它治疗,比如烧蚀化疗或全身 放射疗法的情况下,处理后的骨髓细胞可以保存在合格的液氮医疗设备中。
用于体内临床应用时,本发明的偶联物将以溶液或能被无菌水溶解后注射的冻干固体的形式提供。合适的偶联物给药方法事例如下:偶联物每星期通过静脉注射一次,持续4~12星期。单次剂量通过溶解于50到500毫升的生理盐水,生理盐水可以加入人血清白蛋白(例如,0.5到1毫升的100mg/ml的浓缩人血清白蛋白)。药物剂量大概在50μg到20mg每千克体重每星期,静脉注射(每次注射10ug到200mg/kg体重)。4~12周的治疗结束后,病人可以再接受新一轮的治疗。详细的治疗方法包括给药途径,赋形剂,稀释剂,药物剂量,治疗时间等等可以由有经验的外科医生决定。
可以通过体内或间接体内的方法选择性杀死细胞群来治疗疾病的例子包括任何种类的恶性肿瘤,自体免疫疾病,移植排斥和感染(包括病毒,细菌或寄生虫)。
达到理想的生物学效果而需要的偶联物的量,将因多个因素而各异,这些因素包括化合物的性质特点,疗效和偶联物的生物利用度,疾病的类型,病人的种族,病人患病的状态,给药的途径,所有这些因素共同决定给药时间表和给药方式。
概括的说,本发明中的偶联物可以通过以0.1到10%质量体积比溶解在生理缓冲液中用于非肠道给药。典型的药物剂量范围从1ug到0.1g每千克体重每天;推荐的药物剂量范围从0.01mg到20mg每千克体重每天或者等效剂量的儿童用量。推荐的给药量取决于多个变量,包括疾病或功能紊乱的类型,病人个体的整体的健康状态,偶联药物的相对生物学活性,化合物的剂型,给药的方式(静脉注射,肌肉注射,或其它),在选中给药方式下的药物动力学特性,以及给药的速度(单次注射或者连续滴注)和给药的时间表(在一定时间内的给药次数)。
本发明的偶联物同样可以以单位剂量的形式给药,这里的“单位剂量”是指一个病人一次给药的剂量,单位剂量的药物可以简单方便的包装和使用,单位剂量的药物是保持物理和化学稳定的活性偶联物本身,或者是像后面介绍的药学上可接受的混合物。典型的一天的剂量范围从0.01到100mg每千克体重。一般而言,人每天的单位剂量范围从1到3000mg。推荐的单位剂量是1mg到500mg,每天给药四次,或者10mg到500mg,每天一次。本发明的偶联物可以通过加入一种或多种药学上可接受的辅料,制成 药物制剂。此单位剂量的药物可以用于口服给药,比如是片剂,简单的胶囊或软胶囊;或鼻内给药,比如粉末状,滴鼻剂,或喷雾剂;或者通过皮肤给药,比如软膏剂,乳膏剂,洗液,凝胶剂或喷雾剂或皮肤贴片。药剂可以方便的以单位剂量的形式给药,且以任何已知的制药方法制得,比如Remington:The Science and Practice of Pharmacy,21th ed.;Lippincott Williams&Wilkins:Philadelphia,PA,2005年.中所述的方法。
包含本发明的化合物的药物剂型包括药物组合物首选口服或非肠道给药方式。对于口服给药剂型,如片剂,粉剂,胶囊剂,片剂(锭剂)等等可以包含一种或多种以下原料或有类似性质的其它化合物:粘合剂,比如微晶纤维素或黄耆胶;稀释剂,比如淀粉或乳糖;分散剂,比如淀粉和纤维素衍生物;润滑剂,比如硬脂酸镁;助流剂,比如二氧化硅胶体;甜味剂,比如蔗糖或糖精;增味剂,比如薄荷或水杨酸甲酯。胶囊可以是硬胶囊或软胶囊的形式,一般由明胶混合物选择性的与塑化剂混合,淀粉胶囊也是如此。另外,单位剂量的物理形式可以通过加入多种不同的原料而改变,例如,糖衣,虫胶或肠溶剂。其它的口服剂型如糖浆或酏剂可以含有甜味剂,防腐剂,颜料,着色剂和调味剂。另外,活性化合物可以通过不同的处理和配方,使其成为可快速溶解的剂型,缓慢释放剂型或缓释剂,其中的缓释剂是较好的剂型。片剂首选包含乳糖,玉米淀粉,硅酸镁,交联羧甲基纤维素钠,聚乙烯吡咯酮,硬脂酸镁,滑石等组合的剂型中。
非肠道给药的液体药剂包括无菌的水溶液或非水溶液,悬浮液和乳剂。液体药剂也可以含有粘合剂,缓冲液,防腐剂,螯合剂,甜味剂,调味剂和着色剂等等。非水溶剂包括乙醇,丙二醇,聚乙二醇,植物油比如橄榄油和有机脂类,比如油酸乙酯。水性溶剂包括了水,乙醇,缓冲试剂和盐的混合物,特别是,生物相容性的,可降解的丙交酯聚合物,丙交酯/乙交酯聚合物或者聚乙二醇/聚丙三醇共聚物可作为控制活性药物释放的辅料。静脉注射中的赋形剂可以包括液体和营养补充物,电解质补充物,以及基于林格氏葡萄糖的辅料,以及类似的辅料。对于本发明的活性药物其它可行的非肠道给药系统包括乙烯-醋酸乙烯酯共聚物微粒,植入式渗透泵和脂质体。
其它可行的给药方式包括吸入剂,吸入剂包括干粉剂,气雾剂和水滴剂。吸入剂可以是含有如聚氧乙烯-9-月桂醚,甘胆酸盐,脱氧胆酸盐或油 质的溶液,可以通过滴鼻剂,鼻内胶体的形式给药。口含药剂包括如锭剂,糖果锭剂等,可含有调味剂如蔗糖,阿拉伯胶以及其它辅料如甘胆酸盐等。栓剂适合于单位剂量的形式,以固体如可可油为载体,也可以加入水杨酸。皮肤局部用药剂型,以膏药,乳剂,洗液,贴片,凝胶,喷雾剂,气雾剂或油剂为首选。凡士林,羊毛脂,聚乙二醇,醇类以及它们的混合物可以作为药物载剂。皮肤给药的剂型可以是贴片,乳剂,缓冲溶液,溶解或分散在聚合物或粘合剂中。
特别地,本发明的偶联物可以与其它的已知或未知的治疗药物,如化疗药物,放射疗法,免疫疗法药物,自身免疫疾病药物,抗感染药物或其它抗体药物偶联物共同作用,达到协同效果。协同药物或放射治疗可以在本发明的偶联物给药之前或之后给药或施行。可以是在服用本发明的偶联物之前或之后1小时,12小时,一天,一星期,一个月,也可以是几个月。
在其它的实施方式中,起协同作用的药物包括但不限于:
1).化疗药物:a).烷基化试剂:比如[氮芥:(苯丁酸氮芥,环磷酰胺,异环磷酰胺,氮芥,美法仑,氯乙环磷酰胺);亚硝基脲:(卡莫司汀,洛莫司汀);烷基磺酸盐:(白消安,苏消安);三氮烯类:(达卡巴嗪);含铂化合物:(卡铂,顺铂,奥沙利铂)];b).植物生物碱:比如[长春花生物碱:(长春碱,长春新碱,长春地辛,长春瑞滨);紫杉烷类化合物:(紫杉醇,泰素帝)];c).DNA拓扑异构酶抑制剂:比如[表鬼臼树脂:(9-氨基喜树碱,喜树碱,克立那托,足叶乙甙,磷酸依托泊苷,伊立替康,替尼泊甙,拓扑替康,);丝裂霉素:(丝裂霉素C)];d).抗代谢药物:如{[抗叶酸:二氢叶酸还原酶抑制剂:(甲氨蝶呤,三甲曲沙);IMP脱氢酶抑制剂(霉酚酸,甲酰胺基噻唑,利巴韦林,EICAR);核糖核苷酸还原酶抑制剂(羟基脲,去铁胺)];[嘧啶类似物:尿嘧啶类似物:(5-氟尿嘧啶,去氧氟尿苷,雷替曲塞(拓优得));胞嘧啶类似物:(阿糖胞苷,阿糖胞苷,氟达拉滨);嘌呤类似物:(硫唑嘌呤,巯基嘌呤,鸟嘌呤)]};e).激素类:如{受体拮抗剂:[抗雌激素:(甲地孕酮,雷洛昔芬,他莫昔芬);LHRH激动剂:(戈舍瑞林,醋酸亮丙瑞林);抗雄激素:(比卡鲁胺,氟他胺)];维甲酸/三角肌:[维生素D3类似物(CB 1093,EB 1089 KH 1060,胆钙化醇,维生素D2);光动力疗法:(对此,酞菁光敏剂,PC4,去甲氧甲素);细胞因子:(干扰素-α,干扰素-γ,肿瘤坏死因子(肿瘤坏死因子),人类的蛋白质含TNF域)]}f).激酶抑制剂,如bibw 2992(抗 EGFR/Erb2),伊马替尼,吉非替尼,哌加他尼,索拉非尼,达沙替尼,舒尼替尼,厄洛替尼,尼罗替尼,拉帕替尼,阿西替尼,帕唑帕尼,颇纳替尼,凡德他尼,氟马替尼,e7080(抗VEGFR2),莫立替尼,美迪替尼,普纳替尼(ap24534),HQP1351,巴非替尼(INNO-406),波舒替尼(SKI-606),苏尼替尼,卡博替尼,沃利替尼,维莫德吉,Iniparib,鲁索利替尼,CYT387,阿西替尼,tivozanib,贝伐单抗,索拉非尼,曲妥珠单抗,西妥昔单抗,兰尼单抗,帕尼单抗,ispinesib;g).其它类:如吉西他滨,epoxomicins(例如来那度胺),硼替佐米沙利度胺,来那度胺,,泊马度胺,托舍多特,zybrestat,PLX4032,sta-9090,Stimuvax,allovectin-7,xegeva,Provenge,Yervoy,异戊二烯化抑制剂(如洛伐他汀),多巴胺能神经毒素(如1-甲基-4-苯基吡啶离子),细胞周期抑制剂(如星形孢菌素),放线菌素(如放线菌素D,更生霉素),平阳霉素(如博莱霉素博莱霉素A2,B2,培洛霉素),蒽环类抗生素(如红霉素,阿霉素(阿霉素),伊达比星,表阿霉素,吡柔比星,佐柔比星,米托蒽醌,MDR抑制剂(如维拉帕米),Ca 2+ATP酶抑制剂(如毒胡萝卜素),组蛋白去乙酰化酶抑制剂(伏立诺他,罗米地辛,帕比司他,丙戊酸,莫提诺他(MGCD0103),belinostat,PCI-24781,恩替诺特,SB939,resminostat,givinostat,AR-42,一种,萝卜硫素,曲古抑菌素A);毒胡萝卜素,塞来昔布,格列酮类,表没食子儿茶素没食子酸酯,5双硫仑,salinosporamide A.
2).抗自身免疫病剂包括,但不限于:环孢素,环孢素A,硫唑嘌呤,氨基己酸,溴隐亭,苯丁酸氮芥,氯喹,环磷酰胺,糖皮质激素(如激素类药,倍他米松,布地奈德,氟尼缩松,氟替卡松丙酸酯,氢化可的松,地塞米松,氟可龙丹那唑,曲安奈德,丙酸倍氯米松),脱氢表雄酮,依那西普,羟氯喹,英夫利昔单抗,美洛昔康,甲氨蝶呤,霉酚酸酯,西罗莫司,他克莫司,强的松
3).抗传染病剂包括,但不限于:a).氨基糖苷类:丁胺卡那霉素,武夷霉素,庆大霉素(奈替米星,西索米星,异帕米星),潮霉素,卡那霉素(丁胺卡那霉素,阿贝卡星,氨基去氧卡那霉素,地贝卡星,妥布霉素)新霉素(新霉素B,巴龙霉素,核糖霉素),奈替米星,,大观霉素,链霉素,妥布霉素,甲基姿苏霉素;b).酰胺醇类:叠氮氯霉素,氯霉素,氟甲砜霉素,甲砜霉素;c).安沙霉素类:格尔德霉素,除莠霉素;d).碳青霉烯类:比阿培南,多尼培南,厄他培南,亚胺培南/西司他丁,美罗培南,帕尼培南;e).头孢 类:碳头孢烯(氯碳头孢),头孢乙腈,头孢克洛,头孢拉定,头孢羟氨苄,头孢罗宁,头孢噻啶,头孢噻吩或头孢菌素,头孢氨苄,头孢来星,头孢孟多,头孢匹林,头孢三嗪,头孢氮氟,头孢西酮,头孢唑啉,头孢拉宗,头孢卡品,头孢达肟,头孢吡肟,头孢米诺,头孢西丁,头孢丙烯,头孢沙定,头孢替唑,头孢呋辛,头孢克肟,头孢地尼头孢妥仑,头孢吡肟,头孢他美,头孢甲肟,头孢地嗪,头孢尼西,头孢哌酮,头孢雷特,头孢噻肟,头孢替安,头孢唑兰,头孢氨苄,头孢咪唑,头孢匹胺,头孢匹罗,头孢泊肟,头孢丙烯,头孢喹肟,头孢磺啶,头孢他啶,头孢特仑,头孢布烯,头孢噻林,头孢唑肟,头孢,头孢曲松,头孢呋辛,头孢唑喃,头霉素(头孢西丁,头孢替坦,头孢美唑)氧头孢烯(氟氧头孢,拉氧头孢);f).糖肽:博来霉素,万古霉素(奥利万星,特拉万星),替考拉宁(达巴万星)雷莫拉宁,达托霉素;g).甘氨酰:如替加环素;h).β-内酰胺酶抑制剂:青霉烷(舒巴坦,他唑巴坦),克拉维烷(克拉维酸);i).林可酰胺类:克林霉素,林可霉素;j).脂肽:达托霉素,A54145,钙依赖性抗生素(CDA);k).大环内酯类:阿奇霉素,喹红霉素喹红霉素喹红霉素,克拉霉素,地红霉素,红霉素,氟红霉素,交沙霉素,酮内酯类(泰利霉素,喹红霉素喹红霉素喹红霉素喹红霉素)麦迪霉素,美奥卡霉素,竹桃霉素,利福霉素(利福平,利福平,利福布汀,利福喷丁),罗他霉素,罗红霉素,壮观霉素,螺旋霉素,他克莫司(FK506),醋竹桃霉素,泰利霉素;l).单环β-内酰胺抗生素:氨曲南,替吉莫南;m).唑烷酮类:利奈唑胺;n).青霉素类:阿莫西林,氨苄西林(匹氨西林,海他西林,巴氨西林,美坦西林,酞氨西林)叠氮西林,阿洛西林,青霉素,苄星青霉素,苯氧基苄星青霉素,氯甲西林,普鲁卡因青霉素,羧苄青霉素(卡茚西林),邻氯青霉素,双氯青霉素,先锋霉素,氟氯西林,美西林(氮卓脒青霉素双酯),美洛西林,甲氧西林,萘夫西林,苯唑西林,醋甲西林,青霉素,奈西林,青霉素,哌拉西林,苯丙西林,磺苄西林,替莫西林,替卡西林;o).多肽:杆菌肽,多粘菌素E,多粘菌素B;p).喹诺酮类药物:阿拉沙星,巴洛沙星,环丙沙星,克林沙星,达氟沙星,二氟沙星,依诺沙星,恩诺沙星,Floxin,加雷沙星,加替沙星,吉米沙星,格帕沙星,曲伐沙星卡诺,左氧氟沙星,洛美沙星,麻保沙星,莫西沙星,那氟沙星,诺氟沙星,奥比沙星,氧氟沙星,培氟沙星,曲伐沙星,格帕沙星,西他沙星,司帕沙星,替马沙星,妥舒沙星,曲伐沙星;q).菌素: 普那霉素,奎奴普丁/达福普汀);r).磺胺类药物:磺胺米隆,百浪多息,磺胺醋酰,磺胺甲,磺胺,柳氮磺胺吡啶,磺胺异噁唑,甲氧苄氨嘧啶,甲氧苄啶-磺胺甲基异噁唑(复方新诺明);s).类固醇抗菌药物:如夫西地酸;t).四环素类:强力霉素,金霉素,氯羟四环素,地美环素,赖甲环素,氯甲烯土霉素,美他环素,米诺环素,土霉素,青哌四环素,罗利环素,四环素,甘氨酰(如替加环素);u).其他类型的抗生素:番荔枝科,胂凡纳明,细菌萜醇抑制剂(杆菌肽),Dadal/AR抑制剂(环丝氨酸),dictyostatin,海绵内酯,艾榴塞洛素,埃博霉素,乙胺丁醇,依托泊苷,法罗培南,夫西地酸,痢特灵,异烟肼,laulimalide,甲硝唑,莫匹罗星,mycolactones,NAM合成抑制剂(如磷霉素),呋喃妥因,紫杉醇,平板霉素,吡嗪酰胺,奎奴普丁/达福普汀,利福平(利福平),他唑巴坦替硝唑,番荔枝内酯;
4).抗病毒的药物:a).进入/融合抑制剂:aplaviroc,马拉韦罗,维立韦罗,gp41的(恩夫韦),PRO140,CD4(Ibalizumab);b).整合酶抑制剂:拉替拉韦,埃替拉韦,globoidnan A;c).成熟抑制剂:贝韦立马,vivecon;d).神经氨酸酶抑制剂:奥司他韦,扎那米韦,帕拉米韦;e).核苷和核苷酸:阿巴卡韦,阿昔洛韦,阿德福韦,氨多索韦,apricitabine,溴夫定,西多福韦克拉夫定,右艾夫他滨,去羟肌苷(DDI),艾夫他滨,恩曲他滨(FTC),恩替卡韦,泛昔洛韦,氟脲嘧啶(5-FU),3'-氟取代的2',3'-二脱氧核苷类似物(例如,3'-氟-2',3'-二脱氧胸苷(FLT)和3'-氟-2',3'-双脱氧(FLG)福米韦生,更昔洛韦,碘苷,拉米夫定(3TC),L-核苷(如β-L-胸苷,β-L-2'-脱氧胞苷),喷昔洛韦,Racivir,利巴韦林,stampidine,司他夫定(d4T的),他利韦林(伟拉咪定),替比夫定,替诺福韦,伐昔洛韦三氟胸苷,缬更昔洛韦,扎西他滨(DDC),齐多夫定(AZT);f).非核苷类:金刚烷胺,ateviridine,卡普韦林,二芳基嘧啶(依曲韦林,利匹韦林),地拉韦啶,二十二烷醇,乙米韦林,依法韦仑,膦甲酸(磷酰基甲酸),咪喹莫特,干扰素α,洛韦胺,洛德腺苷,他巴唑,奈韦拉平,NOV-205,聚乙二醇干扰素α,鬼臼毒素,利福平,金刚乙胺,瑞喹莫德(R-848),醋胺金刚烷;g).蛋白酶抑制剂:安普那韦,阿扎那韦,博赛泼维,达芦那韦,福沙那韦,茚地那韦,洛匹那韦,奈非那韦,pleconaril,利托那韦,沙奎那韦,特拉匹韦(VX-950)替拉那韦;h).其他类型的抗病毒的药物:抗体酶,阿比朵尔,calanolides A,浅蓝菌素,蓝藻-N,二芳基嘧啶,表没食子儿茶素没食子酸酯(EGCG),膦甲酸钠,格瑞 弗森,他利韦林(伟拉咪定),羟基脲,KP-1461,米替福新,普来可那立,合成抑制剂,利巴韦林,seliciclib;
5).其它免疫治疗药物:如咪喹莫特,干扰素(如α,β),粒细胞集落刺激因子,细胞因子,白细胞介素(IL-1~IL-35),抗体(例如曲妥单抗,帕妥珠单抗,贝伐单抗,西妥昔单抗,帕尼单抗,英夫利昔单抗,阿达木单抗,巴利昔单抗,达克珠单抗,奥马珠单抗),蛋白结合药物(例如,Abraxane),一种抗体结合药物选自卡奇霉素衍生物,美登素衍生物(DM1 and DM4),CC-1065和多卡霉素小沟剂,有效的紫杉醇衍生物,阿霉素,阿里他汀抗有丝分裂药物(如曲妥珠单抗-DM1,Inotuzumab单抗,brentuximab vedotin,Glembatumumab vedotin,lorvotuzumab mertansine,AN-152 LMB2,TP-38,VB4-845,Cantuzumabmertansine,AVE9633,SAR3419,CAT-8015(抗-CD22),IMGN388,IMGN529,IMGN853,milatuzumab-阿霉素,SGN-75(抗CD70),抗-CD22-MCC-DM1)。
作为本发明更进一步的目标,本发明还涉及其中的抗体药物共轭体的制备过程。本发明的共轭体可以用多种该领域的熟知方法制备,例如,本发明共轭体中的抗有丝分裂剂可以按照下述方法或改进的下述方法合成。对于该领域的专业人士来说,这些改进方法都是很容易从科技文献中获得的、人所共知的、很明显的方法。特别是,这些方法在《Comprehensive Organic Transformations》(R.C.Larock著,1999,Wiley-VCH出版,第2版)一书中有很多的介绍。
在本文描述的反应过程中,有时会有必要保护可能参与反应的活性官能团,例如羟基、氨基、亚氨基、巯基和羧基,以避免副反应的发生。常规保护官能团的常见使用方法可参考P.G.Wuts和T.W.Greene所著的《Greene's Protective Groups in Organic Synthesis》(2006年,Wiley-Interscience出版,第4版)。有的反应可以在适当的含有酸或碱的溶液中进行。这类反应的酸、碱和溶剂并没有特别的限制,只要没有不利的影响,任何常规的酸、碱以及溶剂都可以在这里使用。而且,这些反应可以在广泛的温度范围内进行。不过,总的来说,比较容易操作的反应温度通常在-80℃ to 150℃之间(在室温与100℃之间更好)。反应所需的时间同样可以有很大的变化范围,当然,这取决于多种因素,尤其是反应温度和所用溶剂的性质。一般地说,对于比较理想的反应来说,3至20小时的反应时间比较合适。
反应结束后的操作处理可以按常规方法进行。例如,可以通过把溶剂从反应体系中蒸掉的方式回收反应产物。或者,如果必要的话,在把溶剂蒸出后,可以先把剩余物倒入水中,再用与水不互溶的有机溶剂萃取。最后,在把萃取溶剂蒸走以后,就可以获得反应产物。此外,如果有更高纯度的需要,还可通过各种常见的方法进一步纯化,例如重结晶、沉降或各种色谱层析的方法。一般地说,柱层析和制备薄板层析的方法更为常用。
实施例
本发明用下面的实施例进一步地说明,这些实施例的内容并不旨在限制本发明的范围。在实施例中细胞系,除了特殊说明之外,均是依据美国标准培养物保藏中心(ATCC)、德国菌种保藏中心(DSMZ)或中国科学院上海细胞培养中心规范的条件保存。除了特殊说明外,细胞培养试剂来自于Invitrogen公司。所有的无水试剂均由商业途径获得,并储存于Sure-Seal密封瓶中。其他的试剂和溶剂均按照最高规格购买,使用时未经进一步的处理。用Varain PreStar HPLC进行制备HPLC分离。NMR谱在Bruker 500MHz仪器上获得,化学位移以ppm为单位,四甲基硅烷为参考(0ppm),耦合常数(J)的单位是Hz。质谱数据在Waters Xevo Q Tof质谱仪(连接Waters Acquity UPLC高效液相色谱仪和TUV检测器)上获得。
实施例1 化合物1的合成
Figure PCTCN2019093946-appb-000106
在10-L反应釜中将二乙氧基乙腈(1.00kg,7.74mol)溶解于甲醇(6.0L),在室温下加入(NH 4) 2S(48%水溶液,1.41kg,9.29mol)。釜内温度升至33℃而后又降回室温。搅拌过夜后,浓缩反应液。向残留物中加入乙酸乙酯(5L),用饱和NaHCO 3溶液(4×1.0L)洗,水相用乙酸乙酯(5×1.0L)反萃。合并有机相,用饱和食盐水(3L)洗,之后用无水硫酸钠干燥,过滤后减压浓缩。粗品用石油醚打浆,真空抽滤,收集固体并用石油醚洗涤。滤液浓缩后再用石油醚打浆,合并收集所得固体,总共得到1.1kg(87%产率)目标产物,为白色或亮黄色固体。 1H NMR(500MHz,CDCl 3)δ7.81(d,J=71.1Hz,2H),5.03(s,1H),3.73(dq,J=9.4,7.1Hz,2H),3.64(dq,J=9.4,7.0Hz,2H),1.25(t,J=7.1Hz,6H)。
实施例2 化合物2的合成
Figure PCTCN2019093946-appb-000107
在5-L三颈圆底瓶上装备一个回流冷凝管和一个恒压滴液漏斗。在其中加入分子筛(
Figure PCTCN2019093946-appb-000108
500g)和硫酰胺2(350g,2.14mol)的乙醇溶液(3L),30分钟内滴入3-溴丙酮酸乙酯(纯度80%,404mL,2.57mol)。滴加过程中内温稍有上升,随后将反应液加热至回流并搅拌30分钟。将反应液冷却至室温后用硅藻土过滤除去不溶物,并用乙酸乙酯洗滤饼。滤液浓缩后的粗产物和硅胶(1.5kg)混合拌匀,通过硅胶柱(10kg)柱层析(10-20%乙酸乙酯/石油醚梯度洗脱)纯化,得到棕色油状物,为目标化合物(509g,92%产率)。
实施例3 化合物3的合成
Figure PCTCN2019093946-appb-000109
将缩醛(300g,1.16mol)的丙酮(3.0L)溶液加热至回流,在1.0小时内滴加4N HCl溶液(250mL)。TLC显示起始原料反应完全。反应液减压浓缩后分离两相。有机相加入乙酸乙酯稀释(1.5L),并依次用饱和NaHCO 3水溶液(1.0L)、水(1.0L)和食盐水(1.0L)洗,然后用无水硫酸钠干燥。合并所有的水相并用乙酸乙酯反萃,并用无水硫酸钠干燥有机相。滤出干燥剂后浓缩有机相,所得粗产品用石油醚/乙酸乙酯(5:1)溶液打浆,析出的固体经真空过滤收集,并用石油醚/乙酸乙酯(10:1)溶液洗。滤液浓缩用柱层析纯化(0-15%乙酸乙酯/石油醚),所有固体合并,得目标产物40g(43%产率),为白色或亮黄色固体。 1H NMR(500MHz,CDCl 3)δ10.08–10.06(m,1H),8.53–8.50(m,1H),4.49(q,J=7.1Hz,2H),1.44(t,J=7.1Hz,3H)。MS ESI m/z C 7H 8NO 3S[M+H] +:计算值186.01,实测值186.01。
实施例4 化合物4的合成
Figure PCTCN2019093946-appb-000110
在N 2保护下室温下向(S)-叔丁基亚磺酸胺(100g,0.825mol)的四氢呋喃(1L)溶液中加入Ti(OEt) 4(345mL,1.82mol)和3-甲基-2-丁酮(81mL, 0.825mol)。加热反应液,回流16小时后冷却至室温,然后倒入冰水(1L)。过滤并且用乙酸乙酯洗滤饼。分离滤液中的有机相,无水硫酸钠干燥后减压浓缩,其残留物减压蒸馏(15-20torr,95℃)得目标产物4(141g,90%产率),为黄色油状物。 1H NMR(500MHz,CDCl3)δ2.54–2.44(m,1H),2.25(s,3H),1.17(s,9H),1.06(dd,J=6.9,5.1Hz,6H)。MS ESI m/z C 9H 19NaNOS[M+Na] +:计算值212.12,实测值212.11。
实施例5 化合物5的合成
Figure PCTCN2019093946-appb-000111
在N 2保护下,于-78℃下向二异丙胺(264mL,1.87mol)的四氢呋喃溶液中加入正丁基锂溶液(2.5M,681mL,1.70mol)。反应在30分钟内升温至0℃然后重新冷却至-78℃,向其中加入化合物10(258g,1.36mol)并用四氢呋喃(50mL)冲洗。搅拌1小时后滴加ClTi(O iPr) 3(834g,3.17mol)的四氢呋喃(1.0L)溶液。滴加完成1小时后再缓慢滴入化合物4(210g,1.13mol)的四氢呋喃(500mL)溶液,耗时1小时。所得溶液在-78℃继续搅拌3小时。TLC监测反应进行完全后,用乙酸和四氢呋喃(体积比1:1,300mL)的混合液淬灭反应,而后将反应液倒入盐水(2L),用乙酸乙酯萃取(8×1L)。有机相用水和食盐水洗,无水硫酸钠干燥,过滤并浓缩。残留物经柱层析纯化(二氯甲烷/乙酸乙酯/石油醚2:1:2),得化合物5(298g,74%产率),为无色油状物。 1H-NMR(500MHz,CDCl3)δ8.13(s,1H),6.63(d,J=8.2Hz,1H),5.20–5.11(m,1H),4.43(q,J=7.0Hz,2H),3.42–3.28(m,2H),2.89(dt,J=13.1,6.5Hz,1H),1.42(t,J=7.1Hz,3H),1.33(s,9H),1.25–1.22(m,6H)。MS ESI m/z C 16H 26NaN 2O 4S 2[M+Na] +:计算值397.13,实测值397.11。
实施例6 化合物6的合成
Figure PCTCN2019093946-appb-000112
将化合物5(509g,1.35mol)溶于四氢呋喃(200mL),冷却至-78℃,向其中缓慢加入Ti(OEt) 4(570mL,2.72mol),加完后搅拌1小时。然后在90分钟内分批加入NaBH 4(51.3g,1.36mol),反应在-78℃搅拌3 小时。TLC监测发现起始原料仍有剩余。缓慢加入乙醇(50mL),继续搅拌1.5小时后将反应液倒入饱和食盐水(2L,含250mL乙酸),升至室温。经过硅藻土过滤,分离有机相。水洗、饱和食盐水洗、无水硫酸钠干燥有机相、过滤并浓缩。残留物经柱层析(乙酸乙酯/石油醚1:1)纯化得化合物6(364g,71%产率),为白色固体。 1H NMR(500MHz,CDCl 3)δ8.10(s,1H),5.51(d,J=5.8Hz,1H),5.23–5.15(m,1H),4.41(q,J=7.0Hz,2H),3.48–3.40(m,1H),3.37(d,J=8.3Hz,1H),2.29(t,J=13.0Hz,1H),1.95–1.87(m,1H),1.73–1.67(m,1H),1.40(t,J=7.1Hz,3H),1.29(s,9H),0.93(d,J=7.3Hz,3H),0.90(d,J=7.2Hz,3H)。MS ESI m/z:计算值C 16H 28NaN 2O 4S 2[M+Na] +:399.15,实测值399.14。
实施例7 化合物7的合成
Figure PCTCN2019093946-appb-000113
在0℃下于化合物6(600g,1.60mol)的乙醇(590mL)溶液中加入4N HCl的1,4-二氧六环(590mL)溶液。反应逐渐升至室温后搅拌2.5小时。过滤收集析出的白色固体并用乙酸乙酯洗。滤液浓缩后用乙酸乙酯打浆。合并两次获得的白色固体,共446g(90%产率)。
实施例8 化合物8的合成
Figure PCTCN2019093946-appb-000114
将叠氮化钠(740g,11.4mol)溶于水(2.0L)后加入二氯甲烷(2.0L)并冷却至0℃,向该溶液中加入Tf 2O(700mL,4.10mol),历时1.5小时。加完后在0℃下继续搅拌3小时。分离出有机相,水相用二氯甲烷萃取(2×500mL)。合并有机相,用饱和NaHCO 3(3×1.0L)洗。室温下将此二氯甲烷溶液加入到(L)-异亮氨酸(300g,2.28mol),碳酸钾(472g,3.42mol),五水硫酸铜(5.7g,22.8mmol)的混合甲醇/水(1:1v/v,6.0L)溶液中。加料过程中反应体系内温度会稍微升高。混合液在室温下搅拌16小时,减压蒸去溶剂,水相用浓盐酸(大约280mL)酸化至pH 6-6.5(约280mL),然后用磷酸盐缓冲液稀释(0.25M,pH 6.2,6.0L),用乙酸乙酯(6×2.0L)洗去磺酸胺副产物。水相用浓盐酸(大约400mL)酸化至pH 3后用乙酸乙酯 (4×2.0L)萃取。合并有机相,饱和食盐水洗(2.0L),无水硫酸钠干燥,过滤浓缩得产物8(320g,89%产率),为亮黄色油状物。 1H NMR(500MHz,CDCl 3)δ12.01(s,1H),3.82(d,J=5.9Hz,1H),2.00(ddd,J=10.6,8.6,5.5Hz,1H),1.54(dqd,J=14.8,7.5,4.4Hz,1H),1.36–1.24(m,1H),1.08–0.99(m,3H),0.97–0.87(m,3H)。
实施例9 化合物9的合成
Figure PCTCN2019093946-appb-000115
将Azido-Ile-OH(8,153g,0.97mol)溶于四氢呋喃(1.5L)并冷却至0℃,依次加入NMM(214mL,1.94mol)和氯甲酸异丁酯(95mL,0.73mol)。在0℃搅拌1小时后分批加入化合物7(150g,0.49mmol)。在0℃搅拌30分钟后,逐渐升至室温并继续搅拌2小时。反应在0℃下加冰水淬灭并用乙酸乙酯萃取三遍。合并有机相用1N HCl洗,饱和NaHCO 3洗和食盐水洗,无水硫酸钠干燥。过滤浓缩,残留物经柱层析(0-30%乙酸乙酯/石油醚)纯化得到白色固体(140g,70%产率)。 1H NMR(500MHz,CDCl3)δ8.14(s,1H),6.57(d,J=8.9Hz,1H),4.91(d,J=11.1Hz,1H),4.44(dd,J=13.2,6.3Hz,2H),4.08–3.95(m,2H),2.21(dd,J=24.4,11.5Hz,2H),1.90–1.79(m,3H),1.42(t,J=6.6Hz,3H),1.37–1.27(m,2H),1.11(d,J=6.4Hz,3H),1.01–0.94(m,9H)。MS ESI m/z C 18H 30N 5O 4S[M+H] +:计算值412.19,实测值412.19。
实施例10 化合物10的合成
Figure PCTCN2019093946-appb-000116
在0℃下依次向化合物9(436g,1.05mol)的二氯甲烷(50mL)溶液中加入咪唑(94g,1.37mmol)和三乙基氯硅烷(222mL,1.32mol)。反应液历时1小时升至室温后,继续搅拌1小时。加入饱和食盐水淬灭,分出有机相,水相用乙酸乙酯萃取。合并后的有机相经干燥、过滤、浓缩后用柱层析(15-35%乙酸乙酯/石油醚)纯化得产物10(557.4g,95%产率),为无色油状物。 1H NMR(500MHz,CDCl 3)δ8.12(s,1H),6.75(d, J=8.0Hz,1H),5.20–5.12(m,1H),4.44(q,J=7.0Hz,2H),4.06–3.97(m,1H),3.87(d,J=3.8Hz,1H),2.14(d,J=3.8Hz,1H),2.01–1.91(m,3H),1.42(t,J=7.1Hz,3H),1.34–1.25(m,2H),1.06(d,J=6.8Hz,3H),1.00–0.93(m,18H),0.88(dd,J=19.1,6.8Hz,6H)。MS ESI m/z C 24H 44N 5O 4SSi[M+H] +:计算值526.28,实测值526.28。
实施例11 化合物11的合成
Figure PCTCN2019093946-appb-000117
在0℃下向化合物10(408g,0.77mol)和碘甲烷(145mL,2.32mol)的四氢呋喃(4L)溶液中加入氢化钠(60%,62.2g,1.55mol)。将所得的反应液在0℃搅拌过夜,而后倒入激烈搅拌的冰水饱和氯化铵(5L)溶液中。用乙酸乙酯(3×500mL)萃取。合并后的有机相经干燥、过滤、浓缩后用柱层析(15-35%乙酸乙酯/石油醚)纯化得产物11(388g,93%产率),为亮黄色油状物。 1H NMR(500MHz,CDCl 3)δ8.09(s,1H),4.95(d,J=6.6Hz,1H),4.41(q,J=7.1Hz,2H),3.56(d,J=9.5Hz,1H),2.98(s,3H),2.27–2.06(m,4H),1.83–1.70(m,2H),1.41(t,J=7.2Hz,3H),1.29(ddd,J=8.9,6.8,1.6Hz,3H),1.01(d,J=6.6Hz,3H),0.96(dt,J=8.0,2.9Hz,15H),0.92(d,J=6.6Hz,3H),0.90(d,J=6.7Hz,3H)。MS ESI m/z C 25H 46N 5O 4SSi[M+H] +:计算值540.30,实测值540.30。
实施例12 化合物12的合成
Figure PCTCN2019093946-appb-000118
将2-甲基丙氨酸(500g,4.85mol),甲醛(37%水溶液,1.0L,12.1mol)和甲酸(1.0L)的混合液加热至回流(80℃)。搅拌3.0小时后冷却至室温,加入6N HCl(850mL),而后浓缩反应液。过滤收集所得的固体并用乙酸乙酯洗三遍(1.0L)。将固体溶于水(1.5L)并用4N NaOH(约1.0L)中和至pH 7。浓缩并用乙醇(2.0L)共沸以除水。残留物用甲醇(2.0L)溶解,过滤除去NaCl固体,乙酸乙酯洗。滤液浓缩后得白色固体639.2g,包含有 少量NaCl,可直接使用不用进一步纯化。
实施例13 化合物13的合成
Figure PCTCN2019093946-appb-000119
向化合物12(97g,0.74mol)的乙酸乙酯(1L)溶液中加入五氟苯酚(163g,0.88mol)和DIC(126mL,0.81mol)。反应室温搅拌24小时后用硅藻土过滤,用10mL乙酸乙酯洗。滤液不经进一步纯化直接使用。
实施例14 化合物14的合成
Figure PCTCN2019093946-appb-000120
向上述五氟苯基酯13的乙酸乙酯溶液中加入化合物11(200g,0.37mol)和干的Pd/C(10wt%,10g)。反应液在氢气(1atm)环境中搅拌27小时。硅藻土过滤,乙酸乙酯洗。合并的有机相浓缩后经柱层析(0-5%甲醇/乙酸乙酯)纯化得化合物14(184g,79%产率)。MS ESI m/z C 31H 58N 4O 5SSi[M+H] +:计算值627.39,实测值627.39。
实施例15 化合物15的合成
Figure PCTCN2019093946-appb-000121
将化合物14(200g,0.32mmol)溶于乙酸/水/四氢呋喃(v/v/v 3:1:1,638mL)混合溶液中,室温搅拌4天。反应液浓缩后用甲苯共沸带干,此步骤重复两遍后得化合物15,直接用于下一步反应。MS ESI m/z C 25H 45N 4O 5S[M+H] +:计算值513.30,实测值513.30。
实施例16 化合物16的合成
Figure PCTCN2019093946-appb-000122
在0℃下将氢氧化锂(0.4N,600mL,2.55mol)的水溶液加入化合物15(160g,0.319mol,1.0eq.)的甲醇(1.2L)溶液中。反应在室温搅拌2小时后浓缩。经柱层析(100%二氯甲烷至80:20:1二氯甲烷/甲醇/氨水)得化合物16(140g,两步产率91%)为无定形白色固体。MS ESI m/z  C 23H 40N 4O 5S[M+H] +:计算值485.27,实测值485.27。
实施例17 化合物17的合成
Figure PCTCN2019093946-appb-000123
将化合物16(143g,0.30mol)和DMAP(0.36g,2.95mmol)溶于无水四氢呋喃(1.4L)与无水DMF(75mL)的混合溶液中。冷却至0℃,加入三乙胺(82.2mL,0.59mmol)和乙酸酐(56mL,0.59mmol)。反应逐渐升至室温并搅拌24小时。浓缩后经柱层析(5-50%甲醇/二氯甲烷)纯化得化合物17(147g,95%产率),为无定形白色固体。 1H NMR(500MHz,DMSO)δ8.37(s,1H),7.63(d,J=9.5Hz,1H),5.54(dd,J=11.2,2.5Hz,1H),4.64(dd,J=9.4,7.2Hz,1H),4.34(s,1H),2.95(s,3H),2.27-2.19(m,1H),2.19-2.12(m,1H),2.11(s,6H),2.08(s,3H),1.82-1.66(m,2H),1.54-1.42(m,1H),1.10(s,3H),1.06–0.95(m,1H),0.99(s,3H),0.91(d,J=6.5Hz,3H),0.88(d,J=6.7Hz,3H),0.83(t,J=7.4Hz,3H),0.65(d,J=6.6Hz,3H)。 13C NMR(126MHz,DMSO)δ175.35,172.78,169.70,169.58,162.23,148.03,128.29,69.51,63.00,55.10,52.37,38.86,36.46,33.83,29.25,28.82,23.64,21.09,20.60,19.96,19.40,18.38,15.65,10.77。MS ESI m/z C 25H 44N 4O 6S[M+H] +:计算值527.3,实测值527.4。
实施例18 化合物18的合成
Figure PCTCN2019093946-appb-000124
在室温下,向化合物17(41.0g,77.9mmol,1.0eq)的无水二氯甲烷(600mL)溶液中加入EDC·HCl(44.8g,233mmol,3.0eq)和五氟苯酚(35.9g,194mmol,2.5eq)。将混合物在室温下搅拌2小时,用盐水(300mL)洗涤,经无水硫酸钠干燥,过滤、浓缩,并通过硅胶柱层析法(25-100%乙酸乙酯/正己烷洗脱)纯化,得到白色固体(43.0g,产率80%)。 1H NMR(500MHz,DMSO)δ9.06(s,1H),7.65(d,J=9.4Hz,1H),5.60(dd,J=11.0,2.8Hz,1H),4.64(dd,J=9.4,7.2Hz,1H),4.35(s,1H),2.97(s, 3H),2.34–2.16(m,2H),2.12(s,6H),2.11(s,3H),1.88–1.65(m,2H),1.57–1.37(m,1H),1.11(s,3H),1.06–0.96(m,1H),1.00(s,3H),0.92(d,J=6.5Hz,3H),0.88(d,J=6.7Hz,3H),0.83(t,J=7.4Hz,3H),0.66(d,J=6.6Hz,3H)。 13C NMR(126MHz,DMSO)δ175.24,172.78,171.75,169.81,156.32,141.69,141.56,139.71,138.59,136.60,134.68,69.49,63.11,55.16,52.41,38.83,36.40,33.64,29.42,28.82,23.62,21.01,20.55,19.93,19.39,18.35,15.62,10.73。MS ESI m/z C 31H 42F 5N 4O 6S[M+H] +:计算值693.3,实测值693.3。
实施例19 化合物19的合成
Figure PCTCN2019093946-appb-000125
在室温下将钠氢(60%,8g,200mmol)加入HO-PEG 9-OMe(42.8g,100mmol)的四氢呋喃(1L)的溶液中。搅拌30分钟后,将溴乙酸叔丁酯(48.8g,250mmol)加入,在室温下搅拌1小时,然后倒入冰水中,用二氯甲烷萃取,有机层用饱和食盐水洗,无水硫酸钠干燥。硅胶柱层析纯化(0-5%甲醇/二氯甲烷)得到化合物19,为黄色油状物状物(32g,59%产率)。
实施例20 化合物20的合成
Figure PCTCN2019093946-appb-000126
将化合物432(40g,73.8mmol)溶解在二氯甲烷(400mL)中,然后加入甲酸(600mL),25℃搅拌过夜。减压蒸馏除去所有的挥发物,得到黄色油状物(36g,约100%产率)。ESI m/z C 21H 43O 12[M+H] +:计算值487.27,实测值487.24。
实施例21 化合物21的合成
Figure PCTCN2019093946-appb-000127
将化合物20(36g,73.8mmol)溶解在二氯甲烷(640mL)中,依次加入草酰氯(100mL)和DMF(52g,0.74mmol)。将得到的溶液在室温下搅拌4小时,减压蒸馏除去所有的挥发物,得到黄色油状物。
实施例22 化合物22的合成
Figure PCTCN2019093946-appb-000128
将Z-L-Lys-OH(41.4g,147.6mmol)、碳酸钠(23.4g,221.4mmol)和NaOH(5.9g,147.6mmol)溶解于水(720mL)中,冷却至0℃,然后加入化合物21(37.2g,73.8mmol)的四氢呋喃溶液(20mL)。得到的混合物在室温下搅拌1小时,减压蒸馏除去THF,在冰浴下用浓盐酸调节至pH 3。溶液用二氯甲烷萃取,饱和食盐水洗,无水硫酸钠干燥,得到黄色油状物(55g,99%产率)。ESI m/z C 35H 60N 2O 15[M+H] +:计算值749.40,实测值749.39。
实施例23 化合物23的合成
Figure PCTCN2019093946-appb-000129
向Boc-L-酪氨酸甲酯(2.2kg,7.45mol),碳酸钾(1.54kg,11.2mol)和碘化钾(48g,0.29mol)的乙腈(8.8L)溶液中缓慢加入苄基溴(1.33kg,7.78mol)。室温搅拌过夜,加入水(8L)溶解固体,用乙酸乙酯(2×4L)萃取。合并后的有机相用水(4L)洗,饱和食盐水(4L)洗,无水硫酸钠干燥,过滤,浓缩后用石油醚(20L)打浆,得白色固体98(2.73kg,95%产率)。 1H NMR(500MHz,CDCl 3)δ7.43(d,J=7.0Hz,2H),7.38(t,J=7.4Hz,2H),7.32(t,J=7.2Hz,1H),7.04(d,J=8.5Hz,2H),6.91(d,J=8.6Hz,2H),5.04(s,2H),4.55(d,J=6.9Hz,1H),3.71(s,3H),3.03(qd,J=14.0,5.8Hz,2H),1.43(s,9H)。MS ESI m/z C 22H 28NO 5[M+H] +:计算值386.19,实测值386.19。
实施例24 化合物24的合成
Figure PCTCN2019093946-appb-000130
向2.4L的乙醇和2.4L的二氯甲烷的混合溶剂中加入NaBH 4(122g,3.2mol)和LiCl(136g,3.2mol),冷却至0℃。加入化合物23(616g,1.6mol)的二氯甲烷(2.4L)溶液,加毕,向反应液再加入2.4L的二氯甲烷,自然升至室温,有大量气泡产生,搅拌过夜。将反应液用水(6L)稀释,搅拌30分钟后,水相用二氯甲烷(2L×2)萃取,合并有机相,用水(2L)洗、盐水(2L)洗,干燥过滤,浓缩得白色固体542g(产率95%)。
实施例25 化合物25的合成
Figure PCTCN2019093946-appb-000131
将草酰氯(1.02kg,8.0mol)溶解在二氯甲烷(4L)中,冷却至-75℃,滴加DMSO(1.25kg,16mol)的二氯甲烷(400mL)溶液,维持温度在-65℃以下,滴加完成后搅拌30分钟,再滴加化合物24(1.90kg,5.33mol)的二氯甲烷(8L)溶液,滴加完成后溶液温度升高至约-65℃,搅拌30分钟后,继续滴加三乙胺(1.62kg,16mol),维持温度在-50℃以下,滴加完成后搅拌15分钟。让反应缓慢升温,持续搅拌约1小时,反应液升温至约-30℃,TLC监测显示反应完成。向反应液中加入水(6L),搅拌后分层,将水层用二氯甲烷(2L)洗,合并有机层,用10%的HCl(4L)和盐水(2L)各洗一次,干燥,过滤浓缩。将浓缩液用5:1石油醚/乙酸乙酯打浆,真空抽滤,得淡黄色固体1.36kg(72%产率)。
实施例26 化合物26的合成
Figure PCTCN2019093946-appb-000132
将2-溴丙酸叔丁酯(255g,1.22mol)和三苯基膦(320g,1.22mol)的干燥乙腈(1L)溶液在室温搅拌18小时。减压旋去乙腈后加入甲苯使白色固体析出。倾倒出甲苯后将白色固体溶于二氯甲烷(1L)并转移至分液漏斗。加入10%NaOH水溶液(1L),摇晃后有机相很快变黄。分出有机相,水相用二氯甲烷(1L)反萃一次。合并二氯甲烷相,饱和食盐水(400mL)洗,无水硫酸钠干燥,过滤后浓缩得叶立德26(280g,58%),为一黄色固体。
实施例27 化合物27的合成
Figure PCTCN2019093946-appb-000133
向化合物25(450g,1.27mol)的干燥二氯甲烷(3L)溶液中加入叶立德26(546g,1.40mmol)。室温搅拌过夜。TLC监测反应完全后,经柱层析(10-50%乙酸乙酯/石油醚)纯化得化合物27(444g,75%产率),为一白色固体。ESI m/z C 28H 38NO 5[M+H] +:计算值468.27,实测值468.22。
实施例28 化合物28的合成
Figure PCTCN2019093946-appb-000134
将化合物27(63g,0.13mol)溶于甲醇(315mL),加入Pd/C(10wt%,6.3g),在氢气(1atm)环境下室温搅拌过夜。过滤除去催化剂后,浓缩滤液得化合物28(45.8g,93%产率)。
实施例29 化合物29的合成
Figure PCTCN2019093946-appb-000135
在室温下向化合物28(390g,1.03mol)的四氢呋喃(4L)溶液中加入亚硝酸叔丁酯(1.06k g,10.3mol)。搅拌过夜后减压浓缩除去四氢呋喃,残留物经柱层析(10-50%乙酸乙酯/石油醚)纯化得化合物29(314g,72%产率),为一亮黄色固体。
实施例30 化合物30的合成
Figure PCTCN2019093946-appb-000136
在氮气保护下向化合物30(166g,0.392mol)的乙酸乙酯(500mL)溶液中加入Pd/C(10wt%,16g)。通入氢气,真空置换三次。反应液在氢气(1atm)环境中于室温搅拌16小时。硅藻土过滤,浓缩得化合物30(146g,97%产率),为一黄色泡沫状固体。 1H NMR(400MHz,CDCl 3)δ6.62(d,J=7.9Hz,1H),6.55(s,1H),6.43(d,J=7.3Hz,1H),4.39(dd,J=53.0,44.2Hz,1H),3.77(s,4H),2.72–2.29(m,3H),1.83–1.58(m,1H),1.40(d,J=7.6Hz,18H),1.24(s,1H),1.06(t,J=5.7Hz,3H)。MS ESI m/z C 21H 35N 2O 5[M+H] +:计算值394.25,实测值395.25。
实施例31 化合物31的合成
Figure PCTCN2019093946-appb-000137
将HATU(39.9g,105mmol)加入到4-(((芐氧基)羰基)氨基)丁酸(26.1g, 110mmol)的DMF(300mL)溶液。在室温下搅拌30分钟后,将该反应混合物加入到化合物30(39.4g,100mmol)和三乙胺(20.2g,200mmol)的DMF(300mL)溶液。反应在室温搅拌2小时,用水稀释,经乙酸乙酯萃取,有机层用饱和食盐水洗,硫酸钠干燥。浓缩后通过硅胶柱层析纯化(20-70%乙酸乙酯/石油醚),得到白色固体(45g,产率73%)。ESI m/z C 33H 48N 3O 8[M+H] +:计算值614.34,实测值614.15。
实施例32 化合物32的合成
Figure PCTCN2019093946-appb-000138
将化合物31(100g,163mmol)溶解在甲醇(500mL)中,加入Pd/C催化剂(10wt%,10g),在室温氢化反应(1atm H 2)过夜。将催化剂过滤掉后,滤液减压浓缩,得到棕色泡沫状固体32(75.8g,产率97%)。 1H NMR(400MHz,CDCl3)δ7.11(s,1H),6.83(d,J=10.3Hz,2H),5.04–4.52(m,6H),3.90–3.56(m,1H),2.81(d,J=5.3Hz,2H),2.63(dd,J=12.5,6.1Hz,2H),2.54-2.26(dd,J=14.0,7.6Hz,4H),1.94-1.64(m,3H),1.44–1.36(m,18H),1.08(d,J=6.9Hz,3H)。ESI m/z C 25H 42N 3O 6[M+H] +:计算值480.30,实测值480.59。
实施例33 化合物33的合成
Figure PCTCN2019093946-appb-000139
在0℃,向化合物22(130g,174mmol)的DMF(500mL)溶液中加入三乙胺(66mL,474mmol)和HATU(72g,190mmol),然后将反应混合物升温至室温,搅拌2小时。将化合物32(75.8g,158mmol)的DMF(500mL)溶液在0℃下加入到上述溶液中,反应在室温下搅拌1小时。将反应液倒入水(4L)中,用乙酸乙酯萃取(3×500mL),合并有机层,用饱和食盐水洗(2L),硫酸钠干燥,过滤浓缩后粗品33(190g)直接用于下一步反应。ESI m/z C 60H 100N 5O 20[M+H] +:计算值1210.69,实测值1210.69。
实施例34 化合物34的合成
Figure PCTCN2019093946-appb-000140
将上一步反应得到的粗品33(190g)溶解在甲醇(900mL)中,加入Pd/C催化剂(10wt%,19g),和在室温氢化(1atm H 2)反应过夜。滤除催化剂,将滤液减压浓缩,用硅胶柱纯化(0-10%甲醇/二氯甲烷)得到一棕色油状物(105g,两步产率62%)。ESI m/z C 52H 95N 5O 18[M+H] +:计算值1077.65,实测值1077.65。
实施例35 化合物35的合成
Figure PCTCN2019093946-appb-000141
室温下在化合物34(105g,97.1mmol)的EtOH(5.3L)溶液中,加入化合物4-马来酰亚胺基丁酸-N-琥珀酰亚胺酯(54.4g,194.2mmol)和0.1N磷酸二氢钠溶液(1.1L),反应在室温下搅拌过夜。减压蒸馏除去EtOH,残留水溶液倒入水(3L)中,然后用乙酸乙酯萃取(4×500mL),合并有机相,用饱和食盐水洗(2L),硫酸钠干燥、浓缩,粗产品用硅胶柱纯化(0-10%甲醇/二氯甲烷)得到一黄色油状物(100g,83%产率)。 1H NMR(500MHz,DMSO)δ9.53(s,0.7H),9.52(s,0.3H),9.22(s,0.7H),9.21(s,0.3H),7.95-7.87(m,2H),7.65(t,J=5.9Hz,1H),7.51-7.44(m,1H),6.99(s,2H),6.77–6.66(m,2H),6.65-6.57(m,1H),4.13(dt,J=5.4,8.1Hz,1H),3.84(s,2H),3.55(s,2H),3.52(s,2H),3.51–3.45(m,30H),3.42(dd,J=5.8,3.7Hz,2H),3.38(t,J=6.9Hz,2H),3.23(s,3H),3.14–3.01(m,4H),2.64–2.44(m,1H),2.41–2.22(m,4H),2.16–2.04(m,2H),1.76–1.64(m,4H),1.64–1.52(m,2H),1.52–1.35(m,2H),1.37(s,3H),1.35(s,6H),1.31(s,9H),0.97(t,J=8.5Hz,3H)。 13C NMR(126MHz,DMSO)δ175.35(次要),174.88,171.69,171.42,171.29,171.10,169.04,155.19(次要),155.05,145.97,134.47,129.36,126.00,125.20,122.92,115.69,79.32(次 要),79.17,77.35(次要),77.27,71.29,70.25,69.95,69.79,69.60,69.53,58.06,52.57,50.13,49.55(次要),41.25,38.12,37.94,37.45,36.84,36.80(次要),33.38,32.37,31.86,28.96,28.28,28.23,27.69,27.57,25.42,24.19,22.86,18.04,16.49(次要)。ESI m/z C 60H 101N 6O 21[M+H] +:计算值1241.7,实测值1241.8。
实施例36 化合物36的合成
Figure PCTCN2019093946-appb-000142
将化合物35(31.5g,25.4mmol)溶解在二氯甲烷(125mL)中,加入三氟乙酸(125mL),加毕,室温反应3小时。反应结束,将反应液在旋转蒸发仪上浓缩至无溶剂蒸出。再在真空油泵上真空浓缩至重量基本不变,得粗品。于粗品中加入乙醚(200mL)萃洗,分出产物层,减压浓缩至无溶剂蒸出。再在真空油泵上真空浓缩,直至其重量基本不变,得到化合物36(36.0g,含溶剂)。 1H NMR(500MHz,DMSO)δ9.18(s,1H),7.97-7.87(m,2H),7.79(s,2H),7.71–7.61(m,2H),7.00(s,2H),6.85-6.73(d,J=5.4Hz,2H),4.17–4.07(m,1H),3.84(s,2H),3.55(s,2H),3.52(s,2H),3.50(s,30H),3.42(dd,J=5.8,3.7Hz,2H),3.39(t,J=7.0Hz,2H),3.32-3.23(m,1H),3.23(s,3H),3.14–3.01(m,4H),2.82-2.71(m,1H),2.71–2.61(m,1H),2.58–2.50(m,1H),2.38(t,J=7.3Hz,2H),2.11(dt,J=7.8,3.0Hz,2H),1.88–1.77(m,1H),1.76–1.64(m,4H),1.59(dt,J=15.1,5.8Hz,1H),1.53–1.32(m,4H),1.31–1.11(m,2H),1.05(d,J=7.0Hz,2.1H),1.00(d,J=6.9Hz,0.9H)。 13C NMR(126MHz,DMSO)δ176.78(次要),176.55,171.74,171.42,171.34,171.12,169.07,146.63(次要),146.57,134.49,126.51,126.31,125.19,122.85,115.80,71.31,70.27,69.96,69.81,69.61,69.55,58.07,54.93(次要),52.64,50.72,50.13(次要),38.30(次要),38.14,37.95,36.85,35.75,35.38(次要),34.87,34.81(次要),33.46,32.38,31.83,28.98,25.40,24.21,22.89,17.49,16.74(次要)。ESI m/z C 51H 85N 6O 19[M+H] +:计算值1085.6,实测值 1085.4。
实施例37 化合物37的合成
Figure PCTCN2019093946-appb-000143
向反应瓶中投加化合物36(36.0g,以25.4mmol计)的DMF(60mL)溶液,冰水浴冷却至5℃。加入化合物18(19.3g,27.9mmol)的DMF(150mL)溶液,然后滴加DIPEA(25mL,139mmol)。滴加完毕,撤冰水浴,升至室温,搅拌18小时。反应结束后,将反应液真空油泵减压浓缩至无溶剂蒸出,浓缩完毕,将浓缩液用二氯甲烷稀释,冰水浴冷却至5℃,缓慢滴加甲酸,调pH至3.0-4.0。之后,浓缩至无溶剂蒸出,将残留物转移至硅胶柱上,用正己烷/乙酸乙酯/甲酸和二氯甲烷/甲醇/甲酸洗脱,浓缩得到纯化粗品。用水/甲醇/甲酸溶解纯化粗品,用制备HPLC进一步纯化,水/乙腈/甲酸洗脱,收集合适的洗脱液,浓缩后,用水稀释浓缩液,平均分配至冻干瓶中冻干,得淡黄色泡沫状固体(24g,60%产率)。 1H NMR(500MHz,DMSO)δ9.60(bs,1H),9.20(s,1H),8.19(s,0.33H),8.17(s,0.67H),8.02(d,J=9.0Hz,0.33H),7.98(d,J=9.0Hz,0.67H),7.94–7.83(m,2H),7.65(t,J=5.8Hz,1H),7.63(s,1H),7.56(s,0.33H),7.55(s,0.67H),6.99(s,2H),6.82–6.74(m,1H),6.74–6.67(m,1H),5.62–5.54(m,1H),4.69–4.59(m,1H),4.39(s,1H),4.25–4.05(m,2H),3.85(s,2H),3.55(s,2H),3.52(s,2H),3.50(s,30H),3.44-3.36(m,4H),3.23(s,3H),3.14–3.02(m,4H),2.96(s,3H),2.83–2.71(m,1H),2.71–2.57(m,1H),2.44–2.32(m,3H),2.32–2.14(m,2H),2.14–2.05(m,2H),2.12(s,6H),2.09(s,3H),1.99–1.65(m,7H),1.64–1.53(m,2H),1.53–1.32(m,5H),1.31–1.14(m,2H),1.11(s,3H),1.05(d,J=7.2Hz,2H),1.03(d,J=6.9Hz,1H),0.99(s,3H),0.93(d,J=6.4Hz,3H),0.86(d,J=6.7Hz,3H),0.82(t,J=7.3Hz,3H),0.67(d,J=6.4Hz,3H)。 13C NMR(126MHz,DMSO)δ177.52(次要),177.01,175.44,172.75,171.69,171.43,171.29,171.09,169.74,169.55(次要),169.46,169.04, 159.92(次要),159.88,149.86,149.74(次要),146.07,134.46,129.13(次要),129.08,126.12(次要),126.07,125.18,124.28(次要),124.11,122.95,122.86(次要),115.67,71.30,70.26,69.96,69.80,69.60,69.54,69.48,63.01,58.06,55.09,52.58,52.39,49.03,47.96(次要),40.35,38.89,38.11,37.94,37.39,36.84,36.53,36.02,35.78(次要),33.87,33.38,32.38,31.86,29.02,28.97,25.37,24.19,23.60,22.86,21.15,20.62,19.99,19.41,18.34,18.11,16.17(次要),15.69,10.81。ESI m/z C 76H 125N 10O 24S[M+H] +:计算值1593.9,实测值1593.8。
实施例38 化合物38的合成
Figure PCTCN2019093946-appb-000144
在在氮气保护下,将(S)-4-异丙基噁唑烷-2-酮(400g,3.09mol)溶于无水四氢呋喃(8L)中,降温至-70℃左右,将正丁基锂(2.5M正己烷溶液,1.36L,3.4moL)滴加进反应釜中,滴加完成后在-70℃下反应1小时。然后滴加丙酰氯(315g,3.4mol),滴加完毕后,在-70℃下反应1小时,缓慢升温至室温,将反应液倒入冰的饱和食盐水溶液(7L)中,乙酸乙酯萃取(3×2L),合并有机相,用水(2L)洗涤一次,饱和食盐水溶液(2L)洗涤一次,无水硫酸钠干燥,过滤,旋干,用硅胶柱纯化(纯石油醚至5:1石油醚/乙酸乙酯),得到无色油状物500g,产率87%。MS ESI m/z C 9H 16NO 3[M+H] +:计算值186.10,实测值186.10。 1H NMR(400MHz,CDCl 3)δ4.48–4.39(m,1H),4.27(t,J=8.7Hz,1H),4.21(dd,J=9.1,3.1Hz,1H),3.06–2.82(m,2H),2.38(dtd,J=14.0,7.0,4.0Hz,1H),1.17(t,J=7.4Hz,3H),0.90(dd,J=17.0,7.0Hz,6H)。
实施例39 化合物39的合成
Figure PCTCN2019093946-appb-000145
在氮气保护下,将化合物38(92.6g,0.50mol)溶于无水二氯甲烷(1.5L)中,降温至-10℃,将二异丙基乙基胺(70.5g,0.54mol)和n-Bu 2BOTf(1.0M二氯甲烷溶液,500mL,0.50mol)滴加进反应瓶中,0℃下反应1小时, 再降温至-78℃,将化合物25(161g,0.45mol)的二氯甲烷(1L)溶液滴加进反应瓶中,溶液温度不超过-70℃,滴加完毕反应2小时,然后缓慢升温至室温,并反应过夜。次日,将磷酸盐缓冲液(0.1N,pH 7.0,2L)加进反应瓶中,分液,水相用二氯甲烷(2×500mL)萃取,合并有机相,用饱和食盐水溶液(200mL)洗涤一次,无水硫酸钠干燥,过滤浓缩。将粗品溶于甲醇(2L)中,降温至0℃,将H 2O 2(30%水溶液,500mL)滴入,5℃下反应1小时,加水(3L),用二氯甲烷萃取(3×800mL),合并有机相,水(500mL)洗,饱和NaHCO 3溶液(500mL)洗,饱和食盐水(500mL)洗一次,无水硫酸钠干燥,过滤,浓缩,硅胶柱纯化(纯石油醚至5:1石油醚/乙酸乙酯),得到白色固体150g,产率60%。 1H NMR(400MHz,CDCl 3)δ7.36(ddd,J=24.2,14.2,7.1Hz,5H),7.12(d,J=8.4Hz,2H),6.90(d,J=8.5Hz,2H),5.02(s,2H),4.69(d,J=9.0Hz,1H),4.45(d,J=4.1Hz,1H),4.33(t,J=8.4Hz,1H),4.15(d,J=8.6Hz,1H),3.90(dd,J=16.6,8.0Hz,1H),3.85–3.77(m,2H),2.81(d,J=7.6Hz,2H),2.27(dd,J=11.4,6.7Hz,1H),1.35(s,9H),0.89(dd,J=14.3,6.9Hz,6H)。MS ESI m/z C 30H 41N 2O 7[M+H] +:计算值541.28,实测值541.30。
实施例40 化合物40的合成
Figure PCTCN2019093946-appb-000146
在氮气保护下,将化合物39(200g,0.37mol)溶于无水四氢呋喃(3.5L)中,加入二硫羰基咪唑(198g,1.11mol),回流反应8小时,补加二硫羰基咪唑(65g,0.37mol),反应过夜。次日冷却反应至室温,减压浓缩除去溶剂,硅胶柱纯化(纯石油醚至5:1石油醚/乙酸乙酯),得到油状液体(170g,产率83%)。 1H NMR(400MHz,CDCl 3)δ8.41(s,1H),7.67(s,1H),7.36(dt,J=16.0,6.9Hz,6H),7.09(s,1H),7.05(d,J=8.4Hz,2H),6.86(d,J=8.4Hz,2H),6.32(d,J=9.5Hz,1H),5.01(s,2H),4.56–4.43(m,2H),4.32(ddd,J=16.2,15.6,7.8Hz,3H),4.19(d,J=8.7Hz,1H),2.96(dd,J=14.6,4.4Hz,1H),2.49(dd, J=14.5,10.5Hz,1H),2.29(td,J=13.4,6.7Hz,1H),1.73(s,1H),1.29(s,9H),0.91(dd,J=13.9,6.9Hz,6H)。MS ESI m/z C 34H 43N 4O 7S[M+H] +:计算值651.27,实测值651.39。
实施例41 化合物41的合成
Figure PCTCN2019093946-appb-000147
在氮气保护下,将化合物40(210g,323mmol)溶于无水甲苯(3L)中,在氮气保护下加入三正丁基锡烷(182g,646mmol)和偶氮二异丁腈(0.5g,3.23mmol),回流反应2小时,冷却至室温,旋干,硅胶柱纯化(纯石油醚至5:1石油醚/乙酸乙酯),得到油状物(141g,83%产率)。 1H NMR(400MHz,CDCl 3)δ7.36(ddd,J=24.5,14.5,7.1Hz,5H),7.08(d,J=8.5Hz,2H),6.90(d,J=8.5Hz,2H),5.04(d,J=5.1Hz,2H),4.48(d,J=4.2Hz,1H),4.33(t,J=8.4Hz,1H),4.22(d,J=9.7Hz,1H),4.15(d,J=8.8Hz,1H),3.81(s,2H),2.73(dd,J=14.1,5.9Hz,1H),2.61(dd,J=14.0,7.2Hz,1H),2.29(dq,J=13.5,6.8Hz,1H),2.11–2.00(m,1H),1.60(dd,J=15.2,6.2Hz,2H),1.35(s,9H),1.20(d,J=6.9Hz,3H),0.89(dd,J=14.0,6.9Hz,6H)。MS ESI m/z C 30H 41N 2O 6[M+H] +:计算值525.28,实测值525.37。
实施例42 化合物42的合成
Figure PCTCN2019093946-appb-000148
将化合物41(208g,390mmol)溶于四氢呋喃(2.1L)和水(0.7L)的混合液中,冰浴下滴加LiOH(23.7g,0.99mol)的H 2O 2(30%水溶液,336mL,2.97mol)溶液,控制温度不超过5℃,反应3小时后,滴加入亚硫酸钠(1.5M,2L)溶液,用2N HCl调节pH至4,乙酸乙酯萃取(3×800mL),合并有机相,水洗(500mL)和饱和食盐水(500mL)洗一次,无水硫酸钠干燥,过滤,浓缩,硅胶柱纯化(纯石油醚至3:1石油醚/乙酸乙酯),得到油状物(158g,产率96%)。 1H NMR(400MHz,CDCl 3)δ7.46–7.28(m,5H),7.07(d,J=7.7Hz,2H),6.91(d,J=7.8Hz,2H),5.04(s,2H),4.52 (d,J=8.5Hz,1H),3.87(d,J=41.8Hz,1H),2.82–2.43(m,3H),1.85(t,J=12.2Hz,1H),1.41(s,9H),1.17(d,J=6.9Hz,3H)。MS ESI m/z C 24H 32NO 5[M+H] +:计算值414.22,实测值414.21。
实施例43 化合物43的合成
Figure PCTCN2019093946-appb-000149
将化合物42(158g,0.38mmol)溶于甲醇(1.5L)中,加入Pd/C(10wt%,15g),催化加氢(1atm H 2)反应16小时,过滤,浓缩滤液得到油状物(123g,产率100%)。 1H NMR(400MHz,CDCl 3)δ7.00(d,J=7.5Hz,2H),6.80(s,2H),4.51(d,J=9.0Hz,1H),3.88(s,1H),2.66(dd,J=65.6,22.6Hz,4H),1.88(t,J=12.2Hz,1H),1.42(s,9H),1.14(d,J=6.6Hz,3H)。MS ESI m/z C 17H 26NO 5[M+H] +:计算值324.17,实测值324.16。
实施例44 化合物44的合成
Figure PCTCN2019093946-appb-000150
将化合物43(113g,0.35mol)溶于无水四氢呋喃(1.5L)中,滴加亚硝酸叔丁酯(360g,3.5mol),室温反应3小时,反应完成后,浓缩反应液,硅胶柱纯化(纯石油醚至2:1石油醚/乙酸乙酯)得到黄色固体(85g,产率61%)。 1H NMR(400MHz,DMSO)δ12.00(s,1H),10.68(s,1H),7.67(s,1H),7.34(d,J=8.4Hz,1H),7.03(d,J=8.4Hz,1H),6.69(d,J=8.9Hz,1H),3.56(d,J=3.8Hz,1H),2.67(dd,J=13.5,5.1Hz,1H),2.41(dd,J=13.8,6.6Hz,1H),1.78–1.65(m,1H),1.27(s,9H),1.18(s,1H),1.05(d,J=7.1Hz,3H)。MS ESI m/z C 17H 25N 2O 7[M+H] +:计算值369.15,实测值369.14。
实施例45 化合物45的合成
Figure PCTCN2019093946-appb-000151
将化合物44(80g,217mmol)溶于甲醇(500mL)中,加入Pd/C(10wt%, 2.0g),催化加氢(1atm H 2)反应1小时,过滤,旋干得到白色固体(73g,产率93%)。MS ESI m/z C 17H 27N 2O 5[M+H] +:计算值339.18,实测值339.17。 1H NMR(400MHz,MeOD)δ6.60(d,J=7.9Hz,2H),6.44(d,J=7.3Hz,1H),3.71(d,J=6.3Hz,1H),2.62–2.37(m,3H),1.83(ddd,J=13.7,9.9,3.7Hz,1H),1.39(s,9H),1.13(d,J=7.1Hz,3H)。
实施例46 化合物46的合成
Figure PCTCN2019093946-appb-000152
将八甘醇单甲醚(115.2g,0.3mol)溶解于干燥的四氢呋喃(3L)中。室温下投入氢化钠(60wt%,24g,0.6mol),搅拌反应1小时后加入溴乙酸叔丁酯(146.3g,0.75mol)。室温反应1小时,将反应液加至4L二氯甲烷中,搅拌下加入碎冰2kg,分出水相,用1L二氯甲烷萃取,合并有机相,水洗,浓缩,柱层析纯化(20%乙酸乙酯/石油醚,然后0至5%甲醇/二氯甲烷)得产物108g(产率72%)。
实施例47 化合物47的合成
Figure PCTCN2019093946-appb-000153
向无水甲酸(1L)和二氯甲烷(500mL)的混合溶剂中投加化合物46(210g,0.422mol)。室温搅拌过夜,浓缩得到产物200g(产率100%)。
实施例48 化合物48的合成
Figure PCTCN2019093946-appb-000154
将化合物48(198g,0.422mol)溶解于2.6L二氯甲烷中。室温下滴入0.5mL的DMF和草酰氯(275mL)。搅拌反应3小时,浓缩得到产物210g。
实施例49 化合物49的合成
Figure PCTCN2019093946-appb-000155
将化合物Cbz-L-赖氨酸(236.3g,0.844mol),碳酸钠(89.5g,0.844mol)和氢氧化钠(33.8g,0.844mol)在1.6L水中混合。冰盐浴降温至零度,滴加化合物48(210g,粗品,以0.422mol计)的四氢呋喃(160mL)溶液。加完,室温反应1小时,加乙酸乙酯1L,搅拌后分出水相,浓盐酸调pH至3至4,二氯甲烷萃取,有机相用饱和食盐水洗,无水硫酸钠干燥。过滤,浓缩得 产物290g(产率97%)。
实施例50 化合物50的合成
Figure PCTCN2019093946-appb-000156
将化合物49(182.5g,0.26mol)溶于2L二氯甲烷中。室温下加入五氟苯酚(95.4g,0.52mol)和DIC(131g,1.04mol)。搅拌反应1小时,浓缩得到产物粗品430g。
实施例51 化合物51的合成
Figure PCTCN2019093946-appb-000157
将4-氨基丁酸叔丁酯(62g,0.39mol)溶于1.5L的DMF中。冰水降温,加入DIPEA(134.2g,1.04mol),保持10度至20度下慢慢加入化合物50(430g,粗品,以0.26mol计)。室温反应1小时,浓缩,加二氯甲烷稀释,水洗,水相用二氯甲烷萃取。合并有机相并用0.2N盐酸洗,饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(25%-100%乙酸乙酯/石油醚,再0至5%甲醇/二氯甲烷)得到产物180g,产率82%。
实施例52 化合物52的合成
Figure PCTCN2019093946-appb-000158
将化合物51(78g,92.3mmol),钯碳(10wt%,13g)混合于500mL甲醇中。室温下在氢气球下反应过夜。过滤,浓缩滤液,柱层析纯化(0至20%甲醇/二氯甲烷),得产物70.2g(产率92%)。
实施例53 化合物53的合成
Figure PCTCN2019093946-appb-000159
将化合物52(17.3g,94.2mmol)溶解于500mL二氯甲烷中。室温下依次加入五氟苯酚(34.7g,188.5mmol)和DIC(47.5g,377mmol)。搅拌反应1小时,浓缩得到粗产物105g。
实施例54 化合物54的合成
Figure PCTCN2019093946-appb-000160
将化合物52(67g,94.2mmol)溶于0.75L的DMF中,冰水降温,加入DIPEA(48.6g,376.8mmol),保持反应液在10度至20度,慢慢加入化合物53(105g,粗品,以94.2mmol计)。室温反应1小时,浓缩,加二氯甲烷稀释,水洗,水相用二氯甲烷萃取。合并有机相,用0.2N盐酸洗,饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(50%-100%乙酸乙酯/石油醚,再10%甲醇/二氯甲烷)得到产物80.5g,产率98%。
实施例55 化合物55的合成
Figure PCTCN2019093946-appb-000161
向无水甲酸(400mL)和二氯甲烷(200mL)的混合溶剂中投加化合物54(80.5g,91.9mmol)。室温反应过夜,浓缩,加二氯甲烷稀释,饱和食盐水洗,无水硫酸钠干燥,过滤,柱层析纯化(0%至20%甲醇/二氯甲烷),得到产物70g(产率93%)。
实施例56 化合物56的合成
Figure PCTCN2019093946-appb-000162
将化合物55(104g,0.127mol)溶于二氯甲烷(1000mL)中,在室温下,依次加入N-羟基琥珀酰亚胺(16g,0.14mol)和EDC·HCl(37g,0.2mol)。室温下反应1小时,将反应液用饱和食盐水洗,无水硫酸钠干燥,过滤, 浓缩得到产物120g(产率100%)。
实施例57 化合物57的合成
Figure PCTCN2019093946-appb-000163
将化合物56(120g,127mmol)和化合物45(45.0g,133mmol)在1L四氢呋喃中混合。升温至回流,过夜后浓缩,加二氯甲烷2L稀释,用饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩。在粗品中加水1.5L,乙酸乙酯1.5L,搅拌半小时,分出水相,乙酸乙酯层再用水萃取两次(600mL×2),合并水相,再用乙酸乙酯(700mL)洗去杂质。水相中加氯化钠至饱和,用二氯甲烷萃取(1L×2),合并二氯甲烷,用无水硫酸钠干燥,过滤,浓缩。柱层析纯化(0%至20%甲醇/二氯甲烷),得产物83.6g(产率58%)。 1H NMR(600MHz,DMSO)δ9.51(bs,1H),9.23(s,1H),7.97–7.83(m,1H),7.64(t,J=5.8Hz,1H),7.46(s,1H),6.99(s,1H),6.73(s,1H),6.61(d,J=8.7Hz,0H),4.14(dt,J=5.4,8.3Hz,1H),3.84(s,2H),3.55(s,2H),3.52(s,2H),3.51–3.46(m,26H),3.42(dd,J=5.7,3.8Hz,2H),3.39(t,J=7.0Hz,2H),3.23(s,3H),3.14–3.01(m,4H),2.54(dd,J=13.5,7.0Hz,1H),2.45(dd,J=13.5,7.0Hz,1H),2.41–2.32(m,3H),2.11(td,J=7.1,1.9Hz,2H),1.75–1.64(m,4H),1.64–1.54(m,1H),1.52–1.43(m,1H),1.43–1.35(m,2H),1.32(s,9H),1.32–1.15(m,4H),1.02(d,J=7.1Hz,3H)。 13C NMR(151MHz,DMSO)δ177.18,171.68,171.43,171.28,171.08,169.02,155.15,145.99,134.45,129.41,125.91,125.30,123.04,115.71,77.29,71.29,70.25,69.95,69.79,69.59,69.53,58.05,52.57,50.33,40.71,38.12,37.93,37.64,36.83,35.92,33.35,32.37,31.85,28.95,28.26,27.86(次要),25.39,24.18,22.85,18.10。ESI m/z C 54H 89N 6O 20[M+H] +:计算值1141.6,实测值1141.9。
实施例58 化合物58的合成
Figure PCTCN2019093946-appb-000164
将化合物57(59.0g,51.7mmol)溶解在二氯甲烷(345mL)中,加入三氟乙酸(172mL),加毕,室温反应2小时。反应结束,将反应液在旋转蒸发仪上浓缩至无溶剂蒸出。再在真空油泵上真空浓缩至重量基本不变,得粗品。于粗品中加入乙醚(600mL)萃洗,分出产物层,减压浓缩至无溶剂蒸出。再在真空油泵上真空浓缩,直至其重量基本不变,得到化合物58(75.5g,含溶剂)。 1H NMR(600MHz,DMSO)δ9.82(s,1H),9.18(s,1H),7.95-7.88(m,2H),7.83(s,2H),7.67(s,1H),7.66(t,J=6.1Hz,1H),6.99(s,2H),6.84-6.78(m,2H),4.16–4.09(m,1H),3.84(s,2H),3.55(s,2H),3.52(s,2H),3.52–3.44(m,26H),3.42(dd,J=5.6,3.9Hz,2H),3.39(t,J=7.0Hz,2H),3.33–3.25(m,1H),3.23(s,3H),3.14–3.02(m,4H),2.76(dd,J=13.9,6.1Hz,1H),2.65(dd,J=13.9,6.1Hz,1H),2.55(dq,J=14.0,7.0Hz,1H),2.38(t,J=7.3Hz,2H),2.14–2.08(m,2H),1.82(ddd,J=14.1,8.7,5.5Hz,1H),1.74–1.65(m,4H),1.64–1.55(m,1H),1.52–1.43(m,2H),1.43–1.34(m,2H),1.30–1.13(m,2H),1.05(d,J=7.0Hz,3H)。 13C NMR(151MHz,DMSO)δ176.54,171.73,171.43,171.34,171.11,169.07,146.59,134.48,126.50,126.33,125.19,122.85,115.84,71.30,70.27,69.96,69.81,69.61,69.55,58.06,52.65,50.72,38.31,38.13,37.95,36.85,35.75,34.88,33.45,32.38,31.82,28.97,25.40,24.20,22.88,17.48。ESI m/z C 49H 81N 6O 18[M+H] +:计算值1041.6,实测值1041.7。
实施例59 化合物59的合成
Figure PCTCN2019093946-appb-000165
向反应瓶中投加化合物58(75.5g,以51.7mmol计)的DMF(160mL)溶液,冰水浴冷却至5℃。加入化合物18(35.8g,51.7mmol)的DMF(240mL)溶液,然后滴加DIPEA(36.8g,285mmol)。滴加完毕,撤冰水浴,升至室温,搅拌10小时。反应结束后,将反应液真空油泵减压浓缩至无溶剂蒸出,浓缩完毕,将浓缩液用二氯甲烷稀释,冰水浴冷却至5℃,缓慢滴加甲酸,调pH至3.0-4.0。之后,在旋转蒸发仪上浓缩至无溶剂蒸出,将残留物转移至硅胶柱上,用正己烷/乙酸乙酯/甲酸和二氯甲烷/甲醇/甲酸洗脱,浓缩得到纯化粗品。用水/甲醇/甲酸溶解纯化粗品,用制备HPLC进一步纯化,水/乙腈/甲酸洗脱,收集合适的洗脱液,浓缩后,用水稀释浓缩液,平均分配至冻干瓶中冻干,得淡黄色泡沫状固体(48g,60%产率)。 1H NMR(600MHz,DMSO)δ9.20(s,1H),8.17(s,1H),8.03-7.95(m,1H),7.95–7.86(m,2H),7.77-7.61(m,2H),7.55(s,1H),6.98(s,2H),6.77(d,J=8.1Hz,1H),6.71(d,J=8.1Hz,1H),5.58(d,J=10.7Hz,1H),4.65(dd,J=9.3,7.1Hz,1H),4.40(s,1H),4.18–4.08(m,2H),3.85(s,2H),3.55(s,2H),3.52(s,2H),3.51-3.47(m,26H),3.44–3.36(m,4H),3.23(s,3H),3.14–3.02(m,4H),2.96(s,3H),2.75(dd,J=13.5,6.7Hz,1H),2.63(dd,J=13.5,6.7Hz,1H),2.43–2.32(m,3H),2.31–2.23(m,1H),2.23–2.09(m,3H),2.13(s,6H),2.09(s,3H),1.88–1.73(m,3H),1.75–1.65(m,4H),1.65–1.53(m,2H),1.53–1.44(m,3H),1.44–1.34(m,2H),1.31–1.15(m,2H),1.12(s,3H),1.05(d,J=6.9Hz,3H),1.01(s,3H),0.93(d,J=6.4Hz,3H),0.86(d,J=6.6Hz,3H),0.82(t,J=7.3Hz,3H),0.67(d,J=6.1Hz,3H)。 13C NMR(151MHz,DMSO)δ177.05,175.28,172.76,171.74,171.47,171.34,171.11,169.78,169.49,169.09,159.91,149.88,146.10,134.47,129.10,126.11,125.23, 124.12,122.97,115.72,71.33,70.30,69.99,69.83,69.63,69.57,69.52,63.21,58.08,55.05,52.63,52.49,49.07,40.37,38.89,38.14,37.97,37.43,36.87,36.50,36.06,33.91,33.41,32.41,31.87,29.06,28.99,25.40,24.22,23.66,22.88,21.08,20.63,20.00,19.43,18.41,18.13,15.68,10.81。ESI m/z C 74H 121N 10O 23S[M+H] +:计算值1549.8,实测值1550.2。
实施例60 化合物60的合成
Figure PCTCN2019093946-appb-000166
将八甘醇单甲醚(10g,26mmol,1.0eq)溶于100mL无水二氯甲烷,加入DMAP(32mg,0.26mmol,0.01eq),冰浴下滴加三乙胺(10.5g,104mmol,4.0eq)和TsCl(14.9g,78mmol,3.0eq)固体,完毕,反应10min,升温至室温反应过夜。次日,将反应液用1N HCl洗涤(100mL),水洗(100mL),盐水洗(100mL),无水硫酸钠干燥,旋干,用少量二氯甲烷溶解,上样过柱,洗脱液为5%-100%石油醚/乙酸乙酯和1%-3%甲醇/二氯甲烷,得到11.6g黄色油状物,产率83%。ESI m/z C 24H 43O 11S[M+H] +:计算值539.2,实测值539.2。
实施例61 化合物61的合成
Figure PCTCN2019093946-appb-000167
将化合物60(11.6g,21.5mmol,1.0eq)溶于20mL无水DMF中,加入二苄胺(5.5g,27.8mmol,1.5eq),100℃反应过夜。次日,用300mL二氯甲烷稀释,水洗(300mL×3),盐水洗(300mL),无水硫酸钠干燥,旋干,用少量二氯甲烷溶解,上样过柱,洗脱液为5%-100%石油醚/乙酸乙酯,得到8.2g浅黄色油状物,产率66%。ESI m/z C 31H 50NO 8[M+H] +:计算值564.3,实测值564.3。
实施例62 化合物62的合成
Figure PCTCN2019093946-appb-000168
将化合物61(8.6g,15.2mmol,1.0eq)溶于100mL无水甲醇中,加入干钯碳催化剂(0.9g,10wt%),加热至回流回流,反应过夜。次日,过滤反应液,用甲醇洗涤,旋干,得到5.3g无色油状物,产率90%。ESI m/z  C 17H 38NO 8[M+H] +:计算值384.3,实测值384.3。
实施例63 化合物63的合成
Figure PCTCN2019093946-appb-000169
将Z-L-谷氨酸叔丁酯(0.96g,2.86mmol)和化合物62(1.1g,2.86mmol)溶于DMF(20mL)中,0℃下加入HATU(1.2g,3.15mmol)和DIPEA(1mL,6.3mmol),加完慢慢升至室温反应1小时。将反应液倒入冰水中,用二氯甲烷萃取(3×50mL),合并有机相,水洗(30mL)、饱和碳酸氢钠洗(30mL)、饱和食盐水洗(30mL),硫酸钠干燥,过滤浓缩,用硅胶柱纯化(流动相为甲醇/二氯甲烷)得到产品63(1.5g,产率75%)。ESI m/z C 34H 59N 2O 13[M+H] +:计算值703.4,实测值703.4。
实施例64 化合物64的合成
Figure PCTCN2019093946-appb-000170
将化合物63(0.66g,0.93mmol)溶于甲醇(10mL)中,加入Pd/C(10wt%,60mg),氢气球下反应2小时。过滤浓缩得到化合物64(450mg,85%产率)。ESI m/z C 26H 53N 2O 11[M+H] +:计算值569.4,实测值569.4。
实施例65 化合物65的合成
Figure PCTCN2019093946-appb-000171
将化合物64(0.45g,0.79mmol)和4-马来酰亚胺基丁酸-N-琥珀酰亚胺酯(0.33g,1.18mmol)溶于乙醇(5mL)中,加入NaH 2PO 4(0.1N,1mL),室温反应过夜。减压浓缩除去大部分乙醇后加水(20mL),用二氯甲烷萃取(3×30mL),合并有机相,水洗(20mL)、盐水洗(20mL)、硫酸钠干燥、过滤浓缩,粗品用硅胶柱纯化(流动相为甲醇/二氯甲烷)得到化合物65(380mg,65%产率)。ESI m/z C 34H 60N 3O 14[M+H] +:计算值734.4,实测值734.4。
实施例66 化合物66的合成
Figure PCTCN2019093946-appb-000172
将化合物65(230mg,0.31mmol)溶于二氯甲烷(2mL)中,加入三氟乙酸(2mL),室温反应1小时。浓缩后再用二氯甲烷带3次,并用油泵抽干,得到化合物66(208mg,100%产率)。ESI m/z C 30H 52N 3O 14[M+H] +:计算值678.3,实测值678.3。
实施例67 化合物67的合成
Figure PCTCN2019093946-appb-000173
将化合物66(208mg,0.3mmol)溶于二氯甲烷(5mL)中,加入五氟苯酚(113mg,0.6mmol)和EDC·HCl(117mg,0.6mmol),加完后在室温反应过夜。用二氯甲烷(20mL)稀释,水洗(5mL)、硫酸钠干燥、过滤浓缩得到化合物67(252mg,100%产率)。ESI m/z C 36H 51F 5N 3O 14[M+H] +:计算值844.3,实测值844.3。
实施例68 化合物68的合成
Figure PCTCN2019093946-appb-000174
将化合物45(7.3g,21.7mmol)和N-Boc-丙氨酸羟基琥珀酰亚胺酯(7.2g,21.7mmol)溶于乙醇(300mL)中,加入0.1N NaH 2PO 4(150mL),室温反应过夜。将反应液浓缩,加水(100mL),乙酸乙酯(3×50mL)萃取,合并有机相,盐水洗(50mL)、硫酸钠干燥、过滤浓缩,用硅胶柱纯化(流动相为乙酸乙酯/石油醚),得到化合物68(6.7g,55%产率)。ESI m/z C 29H 40N 3O 8[M+H] +:计算值558.3,实测值558.3。
实施例69 化合物69的合成
Figure PCTCN2019093946-appb-000175
将化合物68(6.7g,12mmol)溶于甲醇(100mL)中,加入Pd/C(10wt%,0.67g),在氢气球下反应2小时。过滤,浓缩滤液得到化合物69(5g,100%产率)。ESI m/z C 21H 34N 3O 6[M+H] +:计算值424.2,实测值424.2。
实施例70 化合物70的合成
Figure PCTCN2019093946-appb-000176
将化合物67(252mg,0.3mmol)和化合物69(190mg,0.45mmol)溶于DMF(5mL)中,冷却至0℃,加入DIPEA(0.13mL,0.75mmol),加完后慢慢升到室温,反应1小时。加水(20mL)稀释,用二氯甲烷萃取(3×10mL),合并有机相,水洗(10mL)、1N HCl洗(10mL)、盐水洗(10mL)、硫酸钠干燥、过滤浓缩,粗品用硅胶柱(流动相为甲醇/二氯甲烷)纯化,得到化合物70(180mg,55%产率)。ESI m/z C 51H 83N 6O 19[M+H] +:计算值1083.6,实测值1083.6。
实施例71 化合物71的合成
Figure PCTCN2019093946-appb-000177
将化合物70(8.2g,7.6mmol)溶于二氯甲烷(56.8mL),加入三氟乙酸(18.9mL),室温反应2小时,浓缩,并用二氯甲烷共浓缩两遍,油泵抽干。向残留液体中加入乙醚(100mL),剧烈搅拌1小时,静置,弃去上清液,再重复操作两次,将下层液体在旋转蒸发仪上浓缩,并用油泵抽干,得化合物71(9.2g,>100%产率)。ESI m/z C 46H 75N 6O 17[M+H] +:计算值983.51, 实测值983.37。
实施例72 化合物72的合成
Figure PCTCN2019093946-appb-000178
将化合物71(8.2g,7.6mmol)溶于DMF(80mL)溶液,用冰水浴冷却反应瓶至0-5℃,加入18(5.2g,7.6mmol)的DMF溶液(20mL),慢慢滴加DIPEA(4mL,22.8mmol)至反应瓶中。控制滴加速度,使整个滴加过程中反应液温度保持在5-10℃之间,滴加完成后,撤除冰水浴,让反应升温至室温,搅拌1.5小时。浓缩,并加二氯甲烷(100mL),冰浴下加入甲酸调节pH为3-4,浓缩,硅胶柱纯化,洗脱液为20-100%乙酸乙酯/正己烷和0-20%甲醇/二氯甲烷(各含0.1%甲酸),将硅胶柱纯化所得的粗产品(11.44g)再用制备HPLC纯化,洗脱液为20-30%乙腈/水(各含0.1%甲酸),浓缩冻干得到化合物72(6.8g,收率60%)。ESI m/z C 71H 115N 10O 22S[M+H] +:计算值1491.78,实测值1492.01。
实施例73 化合物73的合成
Figure PCTCN2019093946-appb-000179
向无水甲酸(500mL)和二氯甲烷(250mL)的混合溶剂中投加化合物63(22.4g,0.03mol)。室温搅拌过夜,浓缩得到产物19g(产率100%)。
实施例74 化合物74的合成
Figure PCTCN2019093946-appb-000180
将化合物73(19.0g,0.03mol)溶于二氯甲烷(200mL)中。室温下加入五氟苯酚(11.0g,0.06mol)和DIC(15.1g,0.12mol)。搅拌反应1小时,浓缩得到产物粗品45g。
实施例75 化合物75的合成
Figure PCTCN2019093946-appb-000181
将4-氨基丁酸叔丁酯(6.40g,0.04mol)溶于500mL的DMF中。冰水降温,加入DIPEA(15.5g,0.12mol),保持10度至20度下慢慢加入化合物74(45g,粗品,以0.03mol计)。室温反应1小时,浓缩,加二氯甲烷稀释,水洗,水相用二氯甲烷萃取。合并有机相并用0.2N盐酸洗,饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(25%-100%乙酸乙酯/石油醚,再0-5%甲醇/二氯甲烷)得到产物19.5g,产率83%。
实施例76 化合物76的合成
Figure PCTCN2019093946-appb-000182
将化合物75(19.5g,24.8mmol),钯碳(10wt%,5g)混合于200mL甲醇中。室温下在氢气球下反应过夜。过滤,浓缩滤液,得产物16.7g(产率100%)。
实施例77 化合物77的合成
Figure PCTCN2019093946-appb-000183
将化合物76(16.7g,24.8mmol)溶于200mL的DMF中,冰水降温,加入DIPEA(12.9g,0.10mol),保持反应液在10度至20度,慢慢加入化合物53(30g,粗品,以24.8mmol计)。室温反应1小时,浓缩,加二氯甲烷稀释,水洗,水相用二氯甲烷萃取。合并有机相,用0.2N盐酸洗,饱和食盐水洗,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(50%-100%乙酸乙酯/石油醚,再10%甲醇/二氯甲烷)得到产物20g,产率98%。
实施例78 化合物78的合成
Figure PCTCN2019093946-appb-000184
将化合物77(16.8g,20.5mmol)溶于二氯甲烷(60mL)中,加入无水甲酸(120mL)。室温反应过夜,浓缩,加乙酸乙酯(150mL)稀释,用水(300mL)萃取,弃去有机相,水相加固体氯化钠至饱和,然后用二氯甲烷(200ml×2)萃取,收集有机相,无水硫酸钠干燥,过滤,浓缩,柱层析纯化(0-20%甲醇/二氯甲烷),得到化合物78(16.4g,收率>100%,含有部分甲酸)。ESI m/z C 34H 59O 15N 4[M+H] +:计算值763.39,实测值763.29。
实施例79 化合物79的合成
Figure PCTCN2019093946-appb-000185
将化合物78(15.6g,20.5mol)溶于二氯甲烷(200mL)中,室温下,依次加入N-羟基琥珀酰亚胺(NHS,3.7g,32.3mol)和EDC·HCl(8.3g,43mol)。室温反应30分钟,盐水洗,无水硫酸钠干燥,过滤,浓缩得到化合物79(17.6g,收率100%)。ESI m/z C 38H 62O 17N 5[M+H] +:计算值860.41,实测值860.29。
实施例80 化合物80的合成
Figure PCTCN2019093946-appb-000186
方法一:
将化合物79(8.8g,10.2mmol)和化合物45(3.5g,10.2mmol)溶于200mL四氢呋喃中。加热至回流,搅拌过夜,浓缩,向粗品中加水(300mL)和乙酸乙酯(100mL),搅拌,分出水相。向水相中加氯化钠至饱和,用二氯甲烷(2×150mL)萃取,合并二氯甲烷,无水硫酸钠干燥,过滤,浓缩。柱层析纯(0-20%甲醇/二氯甲烷),得化合物80(4.0g,收率36%)。ESI m/z C 51H 83O 19N 6[M+H] +:计算值1083.56,实测值1083.47。
方法二:
将化合物79(4.4g,5.12mmol)和化合物45(1.73g,5.12mmol)溶于100 mL EtOH中。然后加入0.1N NaH 2PO 4溶液(20mL),搅拌过夜。浓缩,粗品中加水(200mL)和乙酸乙酯(100mL),搅拌,分出水相。向水相中加氯化钠至饱和,用二氯甲烷(2×100mL)萃取,合并二氯甲烷,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(0-20%甲醇/二氯甲烷),得化合物80(2.0g,收率36%)。
方法三:
将化合物79(4.4g,5.12mmol)和化合物45(1.73g,5.12mmol)溶于100mL乙腈中。然后加入0.1N NaH 2PO 4溶液(20mL),搅拌过夜。浓缩,粗品中加水(200mL)和乙酸乙酯(100mL),搅拌,分出水相。向水相中加氯化钠至饱和,用二氯甲烷(2×100mL)萃取,合并二氯甲烷,无水硫酸钠干燥,过滤,浓缩。柱层析纯化(0-20%甲醇/二氯甲烷),得化合物80(2.2g,收率40%)。
实施例81 化合物73的合成
Figure PCTCN2019093946-appb-000187
将化合物49(20g,28.4mmol,1.0eq.)溶解在350mL无水二氯甲烷中,并在冰水浴上冷却。依次加入NHS(3.9g,34.1mmol,1.2eq.)和EDC(27g,142.0mmol,5.0eq.)。在室温下搅拌反应物过夜,然后用水(200mL×2),盐水(200mL×1)洗涤,用无水硫酸钠干燥,浓缩。将残余物溶于少量二氯甲烷中,并加载到硅胶柱上,用2:49:49至4:48:48甲醇/乙酸乙酯/二氯甲烷洗脱。得到产物为黄色油状物(14.2g,62%收率)。ESI m/z C 37H 60N 3O 16[M+H] +:计算值802.4,实测值:802.4.
实施例82 化合物74的合成.
Figure PCTCN2019093946-appb-000188
向化合物69(6.4g,15.1mmol,1.0eq)在40mL乙醇和10mL 0.1M NaH 2PO 4的混合物中加入化合物73(12.7g,15.9mmol,1.05eq)。将反应混合物搅拌过夜,浓缩并溶解在二氯甲烷中,经无水硫酸钠干燥,过滤,浓 缩,并通过硅胶柱(3-5%甲醇/二氯甲烷)纯化,得到白色泡沫(11.7g,70%收率))。ESI m/z C 54H 88N 5O 19[M+H] +:计算值,实测值:1110.6,实测值:1110.6。
实施例83 化合物75的合成.
Figure PCTCN2019093946-appb-000189
将化合物74(4.2g,3.79mmol,1.0eq.)和钯碳(0.4g,10wt%)混合于5mL甲醇中。室温下在氢气球下搅拌过夜,滤除催化剂并用甲醇洗涤,将滤液浓缩,得到0.32g粗产物,将其直接用于下一步骤(产率87%)。ESI m/z C 46H 82N 5O 17[M+H] +:计算值1997.1,实测值:1997.1。
实施例84 化合物76的合成.
Figure PCTCN2019093946-appb-000190
在500mL烧瓶中,将H 2N-PEG 4-CH 2CH 2CO 2H(3.0g,11.3mmol,1.0eq.)和K 2CO 3(4.7g,33.93mmol,3.0eq.)溶解在50mL水中,并在冰水浴上冷却。逐滴加入Boc 2O(3.2g,14.7mmol,1.3)的50mL四氢呋喃溶液。将反应温热至室温,然后搅拌过夜。用1N KHSO 4将反应混合物调节至pH4-5,用二氯甲烷(200mL×1,100mL×3)萃取,用水(500mL×1)和盐水(500mL×1)洗涤,用无水硫酸钠干燥并且集中。将残余物溶于少量二氯甲烷中,然后加载到硅胶柱上,用2-4%甲醇/二氯甲烷洗脱,合并各组分并浓缩,得到3.8g无色油状物(收率93%)。ESI m/z C 16H 32NO 8[M+H] +计算值:366.2,实测值:366.2。
实施例85 化合物77的合成
Figure PCTCN2019093946-appb-000191
在50mL单颈烧瓶中,混合BocHN-PEG 4-CH 2CH 2CO 2H(0.81g,2.22mmol,1.0eq.),K 2CO 3(0.92g,6.66mmol,3.0eq.)和NaI(0.033g,0.222mmol,0.1eq.)在10mL DMF中,用冰水浴冷却,滴加BnBr(0.57g,3.33mmol,1.5eq.),将混合物温热至室温并搅拌过夜。将反应混合物用100mL 水稀释,用二氯甲烷(100mL×2)萃取,用水(200mL×1)和盐水(200mL×1)洗涤,用无水硫酸钠干燥,并浓缩。将残余物溶于少量二氯甲烷中,加载到硅胶柱上,用70-90%乙酸乙酯/石油醚洗脱,得到0.69g无色油状物(69%收率)。ESI m/z C 23H 38NO 8[M+H] +计算值:446.3,实测值:446.3。
实施例86 化合物78的合成
Figure PCTCN2019093946-appb-000192
将BocHN-PEG 4-CH 2CH 2CO 2Bn(0.69g,1.5mmol,1.0eq.)的6mL二氯甲烷和3mL TFA溶液在室温下搅拌30分钟。除去溶剂,将残余物与二氯甲烷共浓缩三次,置于高真空泵上粗产物直接用于下一反应。ESI m/z C 18H 30NO 6[M+H] +:计算值356.2,实测值356.2。
实施例87 化合物79的合成
Figure PCTCN2019093946-appb-000193
向BocHN-PEG 4-CH 2CH 2CO 2H(3.8g,10.4mmol,1.0eq.)在50mL无水二氯甲烷中的溶液中加入NHS(1.4g,12.5mmol,1.2eq.)和EDC(10.0g,52.0mmol,5.0eq.),在室温下搅拌过夜,然后用水(50mL×2),盐水(100mL×1)洗涤,用无水硫酸钠干燥,浓缩。粗产物直接用于下一步骤。ESI m/zC 20H 35N 2O 10[M+H] +:计算值463.2,,实测值463.2.。
实施例88 化合物80的合成
Figure PCTCN2019093946-appb-000194
在300mL烧瓶中,将H 2N-PEG 4-CH 2CH 2CO 2H(2.8g,10.4mmol,1.0eq.)和K 2CO 3(4.3g,31.2mmol,3.0eq.)溶解在40mL水中,在冰水浴上冷却,并且逐滴加入化合物79的四氢呋喃(40mL)溶液(3.8g,10.4mmol,1.0eq.)并将混合物温热至室温并搅拌过夜。使用1N KHSO 4将反应混合物调节至pH4-5,用二氯甲烷(150mL×1,100mL×2)萃取,用水(200mL×1)和盐水(200mL×1)洗涤,干燥无水硫酸钠,浓缩。将残余物溶于少量二氯甲烷中,并加载到硅胶柱上,用4-6%甲醇/二氯甲烷洗脱,得到无色油状物 (5.18g,81%收率)。ESI m/z C 27H 53N 2O 13[M+H] +:计算值613.3,,实测值613.3。
实施例89 化合物81的合成
Figure PCTCN2019093946-appb-000195
将H 2N-PEG 4-CH 2CH 2CO 2Bn(来自前一步骤的粗产物)溶解在3mL DMF中,在冰/水浴上冷却,滴加DIPEA(0.78g,6.0mmol,4.0eq.),然后加入化合物80(0.93g,1.5mmol,1.0eq.)的DMF(7mL)溶液和HATU(1.72g,4.5mmol,3.0eq.)。将反应物在冰浴上搅拌2小时,并用100mL水稀释,用二氯甲烷(100mL×3)萃取,用1N KHSO 4(200mL×1),饱和碳酸氢钠(200mL×1)和盐水(200mL×1)洗涤,用无水硫酸钠干燥,浓缩。将残余物溶于少量二氯甲烷中,加载到硅胶柱上,并用0-5%甲醇/二氯甲烷洗脱。合并浓缩,得到1.0g淡黄色油状物(71%收率)。ESI m/z C 45H 80N 3O 18[M+H] +:计算值950.5,实测值950.5.
实施例90 化合物82的合成
Figure PCTCN2019093946-appb-000196
在11-氨基十一酸苄酯(2.91g,10.0mmol)和Boc-Glu(OBzl)-OH(3.37g,10.0mmol)的DMF(50mL)溶液中,加入EDC(1.91g,12.0mmol)和三乙胺(3.5mL,25.0mmol)。在室温下搅拌8小时,用水(100mL)稀释,用乙酸乙酯(3×100mL)萃取。将合并的有机相用100mL盐水洗涤一次,然后经无水硫酸钠干燥,过滤并浓缩。通过硅胶柱色谱(乙酸乙酯/二氯甲烷,1:15)纯化残余物,得到标题化合物,为无色油状物(5.37g,88%收率)。
实施例91 化合物83的合成
Figure PCTCN2019093946-appb-000197
在室温下,将化合物82(0.64g,1.05mmol,1.0eq.)在5mL二氯甲烷和2mL TFA的混合溶液中搅拌2小时,然后浓缩。将残余物与二氯甲烷共 浓缩三次并置于高真空泵下。将上述粗产物重新溶解在3mL DMF中,并在冰水浴上冷却。向其中加入化合物80(0.64g,1.05mmol,1.0eq.)的DMF(7mL)溶液中,然后加入DIPEA(0.54g,4.20mmol,4.0eq.)和HATU(1.2g,3.15mmol,3.0eq.)。将反应物在冰浴上搅拌1小时,然后加入100mL水,并用二氯甲烷(150mL×1,100mL×1)萃取。将有机相用1N KHSO 4(200mL×1),饱和碳酸氢钠(200mL×1)和盐水(200mL×1)洗涤,经无水硫酸钠干燥,过滤并浓缩。将粗产物溶于少量二氯甲烷中,并加载到硅胶柱上,然后用0-10%甲醇/二氯甲烷洗脱。合并组分并浓缩,得到0.94g淡黄色油状物(81%收率)。ESI m/z C 57H 92N 4O 17[M+H] +:计算值1104.6,实测值:1104.6。
实施例92 化合物84的合成
Figure PCTCN2019093946-appb-000198
在0℃下,向4-氨基丁酸叔丁酯(1.03g,6.12mmol)和化合物49(3.91g,5.56mmol)的DMF(18mL)溶液中,加入HATU(2.32g,6.12mmol)和TEA(1.2mL,8.34mmol)。将反应搅拌1小时,然后用水(300mL)稀释,并用乙酸乙酯(3×250mL)萃取。将有机溶液用盐水洗涤,用无水硫酸钠干燥,过滤,浓缩并通过硅胶柱色谱法(32:1二氯甲烷/甲醇)纯化,得到标题化合物(5.10g,99%产率).ESI MS m/z 846.50([M+H] +)
实施例93 化合物85的合成
Figure PCTCN2019093946-appb-000199
将化合物84(1.0g,1.18mmol)和Pd/C(10wt%,0.10g)加入到含有甲醇(50mL)的氢化瓶中。将混合物振荡2小时,通过硅藻土(助滤剂)过滤,浓缩滤液,得到化合物85(0.93g,产率>100%).ESI MS m/z 712.50([M+H] +).
实施例94 化合物86的合成
Figure PCTCN2019093946-appb-000200
将化合物85溶解在95%EtOH(50mL)和NaH 2PO 4溶液(0.1M,pH 5.0,10mL)中,加入N-琥珀酰亚胺基4-马来酰亚胺基丁酸酯(0.50g,1.77mmol,1.5eq),将混合物搅拌过夜,然后浓缩,用水(50mL)稀释,用二氯甲烷(80mL×3)萃取,经无水硫酸钠干燥,过滤,浓缩,并通过硅胶柱色谱法(25:1二氯甲烷/甲醇)纯化。得到标题化合物,为浅黄色油状物(0.82g,80%)。
实施例95 化合物87的合成。
Figure PCTCN2019093946-appb-000201
将化合物86(0.82g,0.94mmol)溶解在HCOOH(50mL)中并在室温下搅拌1小时。浓缩反应混合物并与甲苯共浓缩两次,将残余物置于真空泵上,得到化合物87(0.80g,粗产物)。ESI MS m/z([M+H] +)值:820.45。
实施例96 化合物88的合成
Figure PCTCN2019093946-appb-000202
向化合物87(0.80g,粗品,0.94mmol)的DMA(5.0mL)溶液中加入NHS(0.12g,1.03mmol)和EDC·HCl(0.27g,1.41mmol),并将反应混合物搅拌2小时,然后用水(15mL)稀释,并用乙酸乙酯(3×10mL)萃取。将合并的有机相用盐水(10mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过硅胶柱(10-50%乙酸乙酯/石油醚)纯化残余物,得到无色油状化合物(0.67g,78%产率)。ESI MS m/z([M+H] +)值:918.55
实施例97 化合物89的合成
Figure PCTCN2019093946-appb-000203
将N-Boc-乙二胺(5.6mL,35.4mmol,1.1eq.)和饱和NaHCO 3(60mL)的混合物冷却至0℃,向其中分批添加加入N-甲氧基羰基马来酰亚胺(5.00g,32.2mmol,1.0eq.)。在0℃下搅拌30分钟后,将反应温热至室温,搅拌1小时。过滤收集沉淀物,用冷水洗涤,然后溶于乙酸乙酯中,用盐水洗涤,用无水硫酸钠干燥,浓缩,得到白色固体(6.69g,87%收率)。ESI MS m/z([M+H] +)值:241.12。
实施例98 化合物90的合成
Figure PCTCN2019093946-appb-000204
在高压管中,将化合物89(6.00g,25.0mmol),呋喃(18.0mL)的甲苯(120mL)溶液加热至回流并搅拌16小时。无色溶液在反应过程中变黄,然后将混合物冷却至室温并且浓缩。将得到的白色固体用乙醚打浆,得到化合物90(6.5g,84%收率)。ESI MS m/z([M+H] +)值:309.13。
实施例99 化合物91的合成
Figure PCTCN2019093946-appb-000205
在室温下,将化合物90(9.93g,32.2mmol)的二恶烷(15mL)溶液与浓HCl(15mL)反应3小时,过滤收集所得固体,用乙酸乙酯洗涤滤饼。将固体在烘箱(50℃)中干燥过夜,得到化合物91(6.94g,88%收率)。ESI MS m/z([M+H]+)值:206.05。
实施例100 化合物92的合成
Figure PCTCN2019093946-appb-000206
在-10℃下,向化合物91(1.22g,5mmol)的四氢呋喃(10mL)溶液中加入POCl 3(0.47mL,5mmol)。搅拌10分钟后,加入 2,5,8,11,14,17,20,23,26-九氧二十八烷-28-胺(2.14g,5mmol),然后加入DIPEA(0.87mL,5mmol)。将反应温热至0℃并搅拌3小时,然后浓缩。将残余物用二氯甲烷(10mL)稀释,用硅藻土过滤,将滤液直接用于下一步骤。ESI MS m/z([M+H] +)值:716.29。
实施例101 化合物93的合成
Figure PCTCN2019093946-appb-000207
将琥珀酸二甲酯(20.0g,136.9mmol)和二羟乙胺(7.20g,68.7mmol)的无水甲苯(500mL)和吡啶(50mL)的混合物在150℃加热28小时。浓缩混合物,在硅胶柱上纯化,用5-25%乙酸乙酯/二氯甲烷洗脱,得到标题化合物(12.5g,83%收率)。ESI MS m/z([M+Na] +)值:242.42。
实施例102 化合物94的合成
Figure PCTCN2019093946-appb-000208
向化合物93(12.0g,49.56mmol)的无水吡啶(350mL)溶液中加入甲磺酰氯(20.0g,175.4mmol)。搅拌过夜后,浓缩混合物,用乙酸乙酯(350mL)稀释,用冷的1M NaH 2PO 4(2×300mL)洗涤,经MgSO 4干燥,过滤并浓缩,得到粗产物(18.8g,>100%产率))。粗产物无需进一步纯化即可用于下一步骤。ESI MS m/z([M+H] +)值:376.06
实施例103 化合物95的合成
Figure PCTCN2019093946-appb-000209
向马来酰亚胺(10.0g,103.0mmol)的甲苯(200mL)溶液中加入呋喃(10.0mL,137.4mmol)。将混合物在100℃下加热8小时,冷却至室温,浓缩并在乙酸乙酯/己烷中结晶,用甲醇洗涤固体,得到16.7g(99%)标题化合物。 1H NMR(CDC l3):11.12(s,1H),6.68~6.64(m,2H),5.18~5.13(m,2H),2.97~2.92(m,2H)。ESI MS m/z([M+Na] +)值:188.04。
实施例104 化合物96的合成
Figure PCTCN2019093946-appb-000210
向化合物94(新鲜制备,90%纯,8.5g,约20mmol)的DMA(350mL)溶液中,加入化合物95(10.2g,61.8mmol),碳酸钠(8.0g,75.5mmol)和碘化钠(0.3g,2.0mmol)。将混合物在室温下搅拌过夜,浓缩,用乙酸乙酯(350mL)稀释,用饱和NaHCO 3溶液(300mL),饱和NaCl溶液(300mL)和1M NaH 2PO 4(300mL)洗涤。用硫酸钠干燥有机层,过滤,浓缩,上样于硅胶柱上,用10-30%乙酸乙酯/正己烷洗脱,得到标题化合物(7.9g,77%收率。ESI MS m/z([M+Na] +)值:536.4。
实施例105 化合物97的合成
Figure PCTCN2019093946-appb-000211
将化合物96(3.0g,5.8mmol)和三甲基甲锡烷醇(4.8g,26.4mmol)的1,2-二氯乙烷(150mL)溶液在80℃下回流8小时,然后冷却至室温,并使残余物通过短硅胶柱,用二氯甲烷/甲醇洗脱,除去过量的三甲基氢氧化锡。合并组分,浓缩后用DMA和甲苯稀释,加热至120℃并搅拌过夜。将反应混合物上样到硅胶柱,用5-10%甲醇/二氯甲烷洗脱,得到标题化合物(1.62g,76%收率)。ESI MS m/z([M+Na] +)值:386.2。
实施例106 化合物98的合成
Figure PCTCN2019093946-appb-000212
向化合物97(1.62g,4.20mmol)和化合物85(2.71g,3.82mmol)的DMA(20mL)溶液中加入EDC·HCl(0.81g,4.20mmol)。在室温下搅拌反应。过夜,然后倒入水(50mL)中并用乙酸乙酯(3×40mL)萃取。将 合并的有机相用盐水(40mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过柱色谱(10-50%乙酸乙酯/石油醚)纯化残余物,得到无色油状物(3.20g,80%收率)。ESI MS m/z([M+H] +)值:1057.85。
实施例107 化合物99的合成
Figure PCTCN2019093946-appb-000213
将化合物98(3.20g,3.03mmol)的甲酸(10mL)溶液在室温下搅拌过夜。然后将溶液浓缩并与甲苯共浓缩三次,得到无色油状物(3.00g,粗品),其无需进一步纯化即可使用。ESI MS m/z([M+H] +)值:1001.50。
实施例108 化合物100的合成
Figure PCTCN2019093946-appb-000214
向化合物99(3.00g,粗产物,3.03mmol)的DMA(15.0mL)溶液中加入NHS(0.38g,3.33mmol)和EDC·HCl(0.87g,4.55mmol),并将反应混合物室温搅拌2小时,然后用水(50mL)稀释,并用乙酸乙酯(3×30mL)萃取。将合并的有机相用盐水(30mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过硅胶柱(10-50%乙酸乙酯/石油醚)纯化残余物,得到无色油状物(2.90g,90%收率)。ESI MS m/z([M+H] +)值:1098.50。
实施例109 化合物101的合成
Figure PCTCN2019093946-appb-000215
向2,2'-(乙烷-1,2-二基双(氧基))二乙醇(55.0mL,410.75mmol,3.0eq.)的无水四氢呋喃(200mL)溶液中加入钠(0.1g)搅拌混合物直至Na消失,然后滴加丙烯酸叔丁酯(20.0mL,137.79mmol,1.0eq.)。将混合物搅拌过夜,然后在0℃下用HCl溶液(20.0mL,1N)淬灭。通过旋转浓缩除去四氢呋喃,加入盐水(300mL),用乙酸乙酯(3×100mL)萃取。 将有机层用盐水(3×300mL)洗涤有机相,经无水硫酸钠干燥,过滤并浓缩,得到无色油状物(30.20g,79.0%产率),其不经进一步纯化而使用。MS ESI m/z([M+H] +)值:278.17。
实施例110 化合物102的合成
Figure PCTCN2019093946-appb-000216
在0℃下,向3-(2-(2-(2-羟基乙氧基)乙氧基)乙氧基)丙酸叔丁酯(30.20g,108.5mmol,1.0eq.)无水二氯甲烷(220mL)溶液中,加入TsCl(41.37g,217.0mmol,2.0eq.)和TEA(30.0mL,217.0mmol,2.0eq.)。将混合物在室温下搅拌过夜,然后用水(3×300mL)和盐水(300mL)洗涤,经无水硫酸钠干燥,过滤,浓缩并通过硅胶柱色谱法(3:1己烷/乙基)纯化。得到无色油状物(39.4g,收率84.0%)。MS ESI m/z([M+H] +)值:433.28。.
实施例111 化合物103的合成
Figure PCTCN2019093946-appb-000217
向3-(2-(2-(2-(甲苯磺酰氧基)乙氧基)乙氧基)乙氧基)丙酸叔丁酯(39.4g,91.1mmol,1.0eq.)无水DMF(100mL)溶液中加入NaN 3(20.67g,316.6mmol,3.5eq.)。将混合物在室温下搅拌过夜。加入水(500mL)并用乙酸乙酯(3×300mL)萃取。将合并的有机层用水(3×900mL)和盐水(900mL)洗涤,用无水硫酸钠干燥,过滤,浓缩并通过硅胶柱色谱法(5:1己烷/乙酸乙酯)纯化,得到无色黄色油状物(23.8g,收率85.53%)。MS ESI m/z([M+Na] +)值:326.2。
实施例112 化合物104的合成
Figure PCTCN2019093946-appb-000218
将Raney-Ni(7.5g,悬浮在水中)用水(三次)和异丙醇(三次)洗涤,并与化合物103(5.0g,16.5mmol)在异丙醇中混合。将混合物在氢气球下室温下搅拌16小时,然后用硅藻土垫过滤,用异丙醇洗涤垫并浓缩滤液,通过柱色谱(5-25%甲醇/二氯甲烷)纯化,得到浅黄色油状物(2.60g,57%收率)。MS ESI m/z([M+H] +)值:279.19。
实施例113 化合物105的合成
Figure PCTCN2019093946-appb-000219
向十四烷二酸(2.06g,8mmol)的DMF(30mL)溶液中加入K 2CO 3(1.1g,8mmol)和苄基溴(1.36g,8mmol)。将混合物在室温下搅拌过夜,然后浓缩并通过柱色谱(乙酸乙酯/石油醚)纯化,得到标题化合物105(1.2g,45%收率)。ESI MS m/z([M+H] +)值:349.23。
实施例114 化合物106的合成
Figure PCTCN2019093946-appb-000220
向化合物104(2.60g,9.35mmol)和化合物105(3.91g,11.2mmol)的二氯甲烷(50mL)溶液中加入EDC·HCl(2.15g,11.2mmol)和DIPEA(3.6mL,20.6mmol)。将反应混合物在室温下搅拌1小时,然后用50mL二氯甲烷稀释,倒入含有50mL水的分液漏斗中。分离有机相,用盐水(50mL)洗涤,经无水硫酸钠干燥,过滤并浓缩。通过柱色谱(0-10%甲醇/二氯甲烷)纯化残余物,得到标题化合物(4.94g,87%产率)。ESI m/z([M+H] +)值:608.40。
实施例115 化合物107的合成
Figure PCTCN2019093946-appb-000221
向化合物106(4.94g,8.14mmol)的二氯甲烷(20mL)溶液中加入TFA(20mL)。将反应物在室温下搅拌1小时,然后浓缩至干燥并与二氯甲烷共浓缩两次,并将残余物置于泵上,得到化合物107(4.50g,粗产物)。ESI MS m/z([M+H] +)值:552.35。
化合物116 化合物108的合成
Figure PCTCN2019093946-appb-000222
向化合物107(4.50g,粗产物,8.14mmol)和化合物104(1.95g,7.00mmol)的二氯甲烷(50mL)溶液中加EDC·HCl(1.56g,8.14mmol)和DIPEA(2.7mL,15.4mmol)。将反应混合物在室温下搅拌1小时,然后用 50mL二氯甲烷稀释,倒入含有50mL水的分液漏斗中。分离有机相,用盐水(50mL)洗涤,经无水硫酸钠干燥,过滤并浓缩。通过柱色谱(0-10%甲醇/二氯甲烷)纯化残余物,得到标题化合物108(5.22g,92%收率)。ESI m/z([M+H] +)值:811.52。
化合物117 化合物109的合成
Figure PCTCN2019093946-appb-000223
向化合物108(5.22g,6.44mmol)的二氯甲烷(20mL)溶液中加入TFA(5mL)。将反应物在室温下搅拌1小时,然后浓缩至干燥并与二氯甲烷共浓缩两次,并将残余物置于泵上,得到化合物109(4.90g,粗产物。ESI MS m/z([M+H] +)值755.46。
实施例118 化合物110的合成
Figure PCTCN2019093946-appb-000224
向化合物109(4.90g,粗产物,6.44mmol)的二氯甲烷(30mL)溶液中添加NHS(0.81g,7.08mmol),EDC·HCl(1.85g,9.66mmol)和DIPEA(2.8mL,16.1mmol)。将反应混合物在室温下搅拌2小时,然后用水(50mL)稀释,并用乙酸乙酯(3×30mL)萃取。将合并的有机相用盐水(30mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过硅胶柱(10-50%乙酸乙酯/石油醚)纯化残余物,得到无色油236(4.90g,90%收率)。ESI MS m/z([M+H] +)值:852.48。
实施例119 化合物111的合成
Figure PCTCN2019093946-appb-000225
在氢化瓶中,向化合物110(4.90g,5.75mmol)的四氢呋喃(20mL)溶液中加入Pd/C(10wt%,0.20g)。将混合物在1atm氢气下搅拌过夜,通过硅藻土(助滤剂)过滤,并将过滤的溶液浓缩,得到化合物111(4.50g,>100%产率)。ESI MS m/z([M+H] +)值:762.44。
实施例120 化合物112的合成
Figure PCTCN2019093946-appb-000226
在0℃下,向化合物111(1.00g,1.32mmol)的二氯甲烷(10mL)溶液中加入HATU(0.50g,1.32mmol)和三乙胺(0.06mL,1.32mmol)。将反应在0℃下搅拌30分钟,然后加入Z-Lys-OH(0.40g,1.43mmol),在室温下搅拌1小时,然后用水(20mL)稀释,并用乙酸乙酯(3×20mL)萃取。将合并的有机相用盐水(30mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过硅胶柱(0-10%甲醇/二氯甲烷)纯化残余物,得到无色油状物112(1.28g,95%收率)。ESI MS m/z([M+H] +)值:1017.60。
实施例121 化合物113的合成
Figure PCTCN2019093946-appb-000227
向化合物112(1.28g,1.26mmol)的二氯甲烷(10mL)溶液中加入NHS(0.17g,1.51mmol)和EDC·HCl(0.29g,1.51mmol),然后加入三乙胺(0.38mL,2.77mmol)。在室温下搅拌反应2小时,然后用水(20mL)稀释,并用乙酸乙酯(3×15mL)萃取。将合并的有机相用盐水(30mL)洗涤,用无水硫酸钠干燥,过滤并浓缩。通过硅胶柱(0-10%甲醇/二氯甲烷)纯化残余物,得到无色油状物113(1.28g,91%收率)。ESI MS m/z([M+H] +)值:1114.62。
实施例122 化合物114的合成
Figure PCTCN2019093946-appb-000228
将丙烯酸叔丁酯(12.81g,0.10mmol)和乙基-1,2-二胺(24.3g,0.40mol)的四氢呋喃(150mL)溶液在45℃下搅拌24小时。将混合物浓缩并在Al 2O 3凝胶柱上纯化,用甲醇/二氯甲烷(三乙胺)(5%:15%:80%)洗脱,得到标题化合物(17.50g,92%收率)。ESI MS m/z([M+H] +):值189.20。
实施例123 化合物115的合成
Figure PCTCN2019093946-appb-000229
将3-((2-氨基乙基)氨基)丙酸叔丁酯(17.00g,90.33mmol)的1,4-二恶烷(50mL)溶液与浓盐酸(15毫升)在室温下搅拌30分钟,浓缩并用纯水(150mL)和乙酸乙酯/己烷(40mL,1:5)稀释。分离混合物,用水(2×10mL)萃取有机层。浓缩水层并经真空泵干燥,得到标题化合物(18.70g,100%收率,通过LC-MS纯度为96%)。ESI MS m/z([M+H] +)值:133.20。
实施例124 化合物116的合成
Figure PCTCN2019093946-appb-000230
在0℃下,向3-((2-氨基乙基)氨基)丙酸(18.70g,90.33mmol)的四氢呋喃(150mL)溶液中加入马来酸酐(8.85g,90.33mmol)。将混合物在0-4℃下搅拌4小时,浓缩,得到(Z)-4-(2-((2-羧乙基)氨基)乙基)氨基)-4-氧代丁-2-烯酸,然后向混合物中加入甲苯(150mL)和DMA(50mL),在90℃下搅拌,用Dean-Stark分水器回流。在捕集器中收集30mL溶剂后,加入HMDS(六甲基二硅氮烷,9.0mL,43.15mmol)和ZnCl 2(16mL,1.0M的乙醚溶液)。将混合物加热至115-125℃,并通过Dean-Stark分水器收集甲苯。将反应混合物在120℃下加热6小时。在此期间,加入2×40mL无水甲苯以保持混合物体积约50mL。然后冷却混合物并加入1mL 1:10HCl(浓)/甲醇。浓缩混合物,在硅胶柱上纯化,用水/乙腈(1:15)洗脱,浓缩,真空泵干燥,得到标题化合物14.75g(收率77.0%)。ESI MS m/z([M+H] +)值:213.10。
实施例125 化合物117的合成
Figure PCTCN2019093946-appb-000231
在四氢呋喃(300mL),DIPEA(50mL)和HSAc(10.0g,0.131mol)的混合物中加入化合物60(57.30g,0.106mol)。将混合物搅拌过夜,浓缩并在硅胶柱上纯化,用乙酸乙酯/二氯甲烷(1:2至4:1)洗脱,浓缩,并经真空泵干燥,得到标题化合物40.51g(86%收率)。ESI MS m/z([M+H] +)值:443.35。
实施例126 化合物118的合成
Figure PCTCN2019093946-appb-000232
在乙酸(200mL)和30%H 2O 2(100mL)的混合物中加入化合物117(40.40g,0.091mol)。在35℃下搅拌过夜。浓缩混合物,用纯水(200mL)和甲苯(150mL)稀释,分离,有机层用水(2×25mL)萃取。合并水溶液,浓缩并经真空泵干燥,得到标题化合物40.50g(收率99%,LC-MS纯度95%)ESI MS m/z([M+H] +)值:449.30
实施例127 化合物119的合成
Figure PCTCN2019093946-appb-000233
向化合物118(20.0g,44.62mmol)的四氢呋喃(100mL)和二氯甲烷(100mL)的混合物中依次加入草酰氯(25.21g,200.19mmol)和DMF(0.015mL)。将混合物在室温下搅拌2小时,浓缩,与二氯甲烷/甲苯(1:1,2×50mL)共浓缩,然后再溶于四氢呋喃(50mL)中。加入化合物116(7.50g,35.36mmol)的四氢呋喃(100mL)溶液。将混合物搅拌过夜,真空浓缩并在硅胶柱上纯化,用甲醇/二氯甲烷(1:6至1:5)洗脱,并经真空泵干燥,得到标题化合物14.76g(65%收率)。ESI MS m/z([M+H] +)值:643.35。
实施例128 化合物120的合成
Figure PCTCN2019093946-appb-000234
将化合物119(7.50g,11.67mmol),N-羟基琥珀酰亚胺(1.50g,13.04mmol)和EDC(10.10g,52.60mmol)的四氢呋喃(100mL)溶液搅拌过夜,真空浓缩并在硅胶柱上纯化,用乙酸乙酯/二氯甲烷(1:4至2:1)洗脱,并经真空泵干燥,得到标题化合物6.30g(73%收率)。ESI MS m/z([M+H] +)值:740.40
实施例129 化合物121的合成
Figure PCTCN2019093946-appb-000235
在0℃下,向2-(2-(2-(2-氨基乙酰氨基)乙酰氨基)乙酰氨基)乙酸(gly-gly-gly)(0.50g,2.03mmol)和化合物120(1.65g,2.22mmol)的DMF(15mL)溶液中加入DIPEA(3mL)。将反应混合物在0℃下搅拌0.5小时,然后在室温下搅拌4小时。然后浓缩反应混合物,并通过硅胶色谱法(流动相:乙腈/水=95:5,含有0.1%甲酸)纯化,得到标题化合物121(1.04g,63%收率)。MS-ESI m/z C 32H 56N 5O 17S[M+H] +计算值14.33,实测值814.46。
实施例130 化合物122的合成
Figure PCTCN2019093946-appb-000236
将化合物121(0.70g,0.86mmol),N-羟基琥珀酰亚胺(0.20g,1.73mmol)和EDC(1.21g,6.36mmol)的四氢呋喃(20mL)溶液,室温搅拌过夜,真空浓缩并在硅胶柱上纯化。用乙酸乙酯/二氯甲烷(1:4至2:1)洗脱,并经真空泵干燥,得到标题化合物0.540g(69%收率)。MS-ESI m/z C 36H 59N 6O19S[M+H] +:计算值911.34,实测值911.42。
实施例131 化合物123的合成
Figure PCTCN2019093946-appb-000237
在0℃下,(2S,4R)-5-(3-氨基-4-羟基苯基)-4-(2-((6S,9R,11R)-6-((S)-仲丁基)-9-异丙基的溶液-2,3,3,8-四甲基-4,7,13-三氧-12-氧杂-2,5,8-三氮杂十一烷-11-基)噻唑-4-甲酰胺基)-2-甲基戊酸(Tub-039,R.Zhao,et al,PCT/CN2017/120454;R.Zhao,et al,14th PEGS Boston,Boston,MA,USA,3rd May 2018)(83mg,0.106mmol)和化合物122(122mg,0.134mmol)的DMF(8mL)溶液中加入DIPEA(2mL)。将反应混合物 在0℃下搅拌0.5小时,然后在室温下搅拌4小时。然后浓缩反应混合物,并通过制备型HPLC(流动相:乙腈/水=10%至80%,含有0.1%甲酸)纯化,得到化合物123(95.5mg,58%收率)。MS-ESI m/z C 69H 112N 11O 24S[M+H] +:计算值1542.72,实测值1542.76。
实施例132 化合物124的合成
Figure PCTCN2019093946-appb-000238
将(S)-1-苄基-5-叔丁基2-氨基戊二酸盐酸盐(8.70g,26.39mmol),14-(苄氧基)-14-氧代十四烷酸(9.19mmol),DIPEA(8.0mL,46.0mmol)和EDC(15.3g,80.50mmol)的二氯甲烷(200mL)溶液在室温下搅拌6小时。将混合物用水(100mL)稀释并分离。用二氯甲烷(100mL)萃取水相。合并有机相,用盐水洗涤,用硫酸钠干燥,过滤,浓缩并在硅胶柱上纯化(二氯甲烷/乙酸乙酯=20:1至5:1),得到标题化合物314。MS-ESI m/z C 37H 54NO 7[M+H] +:计算值624.38,实测值624.38。
实施例133 化合物125的合成
Figure PCTCN2019093946-appb-000239
在4℃下将化合物124(12.50g,20.05mmol)溶解在二恶烷(30mL)中,并与盐酸(10mL,36%)搅拌0.5小时。将反应混合物用甲苯(20mL)和DMF(20mL)稀释,在15℃浓缩,得到标题化合物(11.26g,99%收率)。MS-ESI m/z C 33H 46NO 7[M+H] +:计算值568.32,实测值568.34。
实施例134 化合物126的合成
Figure PCTCN2019093946-appb-000240
将化合物125(10.70g,18.86mmol),1-氨基-15-氧代-3,6,9,12,19,22,25,28-辛氧基-16-氮杂三十烷-31-叔丁基酯盐酸盐(11.45g, 18.93mmol),EDC(9.51g,50.01mmol)和DIPEA(4.00mL,23.00mol)的二氯甲烷(200mL)溶液室温搅拌过夜,用盐水(100mL)稀释并分离。用二氯甲烷(100mL)萃取水相。合并有机相,用盐水洗涤,用硫酸钠干燥,过滤,浓缩并在硅胶柱上纯化(二氯甲烷/乙酸乙酯=10:1至4:1),得到标题化合物(18.15g,86%收率)。MS-ESI m/z C 59H 96N 3O 17[M+H] +:计算值1118.67,实测值1118.80。
实施例135 化合物127的合成
Figure PCTCN2019093946-appb-000241
在4℃下将化合物126(10.50g,9.39mmol)溶解在二恶烷(45mL)中,并与盐酸(15mL,36%)搅拌0.5小时。将反应混合物用甲苯(20mL)和DMF(20mL)稀释,在15℃浓缩,并在硅胶柱上纯化(二氯甲烷/甲醇=10:1至6:1),得到标题化合物(8.67g,87%收率)。MS-ESI m/z C 55H 88N 3O 17[M+H] +:计算值1062.60,实测值1062.68。
实施例136 化合物128的合成
Figure PCTCN2019093946-appb-000242
将化合物127(8.50g,8.01mmol),N-羟基琥珀酰亚胺(3.20g,27.82mmol),EDC(10.28g,54.10mmol)和DIPEA(6.00mL,34.51mmol)的四氢呋喃(150mL)溶液室温搅拌6小时后,真空浓缩,得到NHS酯,无需纯化即可用于下一步骤。向(S)-6-氨基-2-((叔丁氧基羰基)氨基)己酸盐酸盐(2.75g,9.73mmol)的DMF(100mL)和1.0M Na 2PO 4(pH 7.5,55mL)的混合溶液中,在1小时内分四份加入上面制备的N-琥珀酰亚胺酯。将反应混合物在室温下搅拌3小时。浓缩后,将残余物在硅胶柱上纯化(二氯甲烷/甲醇=10:1至4:1),得到标题化合物(8.16g,79%收率)。MS-ESI m/z C 66H 108N 5O 20[M+H] +:计算值1289.75,实测值1289.90。
实施例137 化合物129的合成
Figure PCTCN2019093946-appb-000243
在4℃下将化合物128(8.10g,6.28mmol)溶解在二恶烷(40mL)中,并与盐酸(15mL,36%)搅拌0.5小时。将反应混合物用甲苯(20mL)和DMF(20mL)稀释,在15℃浓缩,得到标题化合物(7.71g,100%收率),其不经进一步纯化用于下一步。MS-ESI m/z C 61H 88N 3O 17,[M+H]+:计算值1190.70,实测值1190.78。
实施例138 化合物130的合成
Figure PCTCN2019093946-appb-000244
在0℃下,向4-马来酰亚胺-N-琥珀酰胺酯(7.10g,25.35mmol)和丙氨酸(3.01g,33.80mmol)的DMF(50mL)溶液中加入DIPEA(10mL)。将反应混合物在0℃下搅拌0.5小时,然后在室温下搅拌1小时。然后将反应混合物浓缩并在硅胶柱(流动相:二氯甲烷/甲醇=10:1,含有0.1%甲酸)上纯化,得到化合物130(5.21g,81%收率)。MS-ESI m/z C 11H 14N 2O 5[M+H] +:计算值255.09,实测值255.15。
实施例139 化合物131的合成
Figure PCTCN2019093946-appb-000245
将化合物130(5.15g,20.26mmol),N-羟基琥珀酰亚胺(2.80g,24.34mmol),EDC(10.28g,54.10mmol)和DIPEA(5.50mL,31.63mmol)的二氯甲烷(70mL)溶液室温搅拌6小时,真空浓缩,在硅胶柱上纯化(流动相:二氯甲烷/乙酸乙酯=10:1),得到化合物131(5.83g,82%收率)。MS-ESI m/z C 15H 17N 3O 7[M+H] +:计算值351.11,实测值351.20。
实施例140 化合物132的合成
Figure PCTCN2019093946-appb-000246
在0℃下,向化合物129(7.61g,6.39mmol)和化合物131(2.90g,8.280mmol)的DMF(40mL)溶液中加入DIPEA(7mL)。将反应混合物在0℃下搅拌0.5小时,然后在室温下搅拌1小时。然后将反应混合物浓缩并在硅胶柱上纯化(流动相:二氯甲烷/甲醇=10:1,含有0.1%甲酸),得到化合物132(7.10g,78%收率)。MS-ESI m/z C 11H 14N 2O 5[M+H] +:计算值1426.7782,实测值1426.7820。
实施例141 化合物133的合成
Figure PCTCN2019093946-appb-000247
将化合物132(7.05g,4.94mmol),N-羟基琥珀酰亚胺(0.92g,8.00mmol),EDC(3.01g,15.84mmol)和DIPEA(1.00mL,5.75mmol)的四氢呋喃(50mL)溶液室温搅拌6小时后,真空浓缩,得到NHS酯,无需纯化即可用于下一步骤。
向2-(2-(2-氨基乙酰氨基)乙酰氨基)乙酸(gly-gly-gly)盐酸盐(1.67g,7.40mmol)的DMF(40mL)和1.0M Na 2PO 4(pH 7.5,15mL)的混合溶液中,在1小时内分四份加入上述化合物。将反应混合物在室温下搅拌3小时。浓缩后,将残余物在硅胶柱上纯化(二氯甲烷/甲醇=10:1至7:1),得到标题化合物(8.16g,79%收率)。MS-ESI m/z C 11H 14N 2O 5[M+H] +:计算值597.84,实测值1597.84。
实施例142 化合物134的合成
Figure PCTCN2019093946-appb-000248
向化合物133(150.3mg,0.0935mmol),Tub-039(60.2mg,0.0769mmol)和DIPEA(0.030mL,0.172mmol)在DMA(5mL)中的溶液,加入EDC(100mg,0.526mmol)。将反应混合物在室温下搅拌6小时,真空浓缩,再溶于甲醇/二氯甲烷(0.5mL:3mL)中,并通过短硅胶柱,甲醇/二氯甲烷(1:3)洗脱,真空浓缩,得到粗化合物,用于下一步骤。MS-ESI m/z值:2326.25。
将上述化合物的二氯甲烷(1mL)溶液与TFA(3mL)搅拌1小时。将反应混合物用甲苯(3mL)和DMF(3mL)稀释,浓缩,并通过制备型HPLC(流动相:含有0.1%甲酸的2%至50%乙腈水溶液)纯化,得到化合物134(69.0mg,72%收率)。MS-ESI m/z C 11H 14N 2O 5[M+H] +:计算值2146.1497,实测值2146.1588。
实施例143 化合物135的合成
Figure PCTCN2019093946-appb-000249
向(S)-30-(4-(2,5-二氧代-2,5-二氢-1H-吡咯-1-基)丁酰胺基)-27-氧代-2,5,8,11,14,17,20,23-八氧杂-26-氮杂三十烷-31-酸(20mg,0.029mmol)的二氯甲烷(5mL)溶液中加入EDC(11mg,0.059mmol)和五氟苯酚(10.8mg,0.059mmol)。将反应混合物在室温下搅拌2小时,浓缩并在硅胶柱上纯化,用乙酸乙酯/二氯甲烷(1:4)洗脱,得到标题化合物(24mg,100%收率)。MS-ESI m/z C 36H 50F 5N 3O 14[M+H] +:计算值844.32,实测值844.32。
实施例144 化合物136的合成
Figure PCTCN2019093946-appb-000250
将(S)-2-(((S)-2-(((苄氧基)羰基)氨基)丙酰胺基)丙酸叔丁酯(10g,0.028mol)的甲醇溶液(100mL)和10%钯碳(1.0g)在氢气(5psi)下搅拌3小时。滤出固体,浓缩过滤的溶液,得到无色油状产物(6.1g,100%收率)。ESI m/z C 10H 20N 2O 3[M+H] +:计算值217.15,实测值217.15.
实施例145 化合物137的合成
Figure PCTCN2019093946-appb-000251
向(S)-30-(((苄氧基)羰基)氨基)-27-氧代-2,5,8,11,14,17,20,23-八氧杂-26-氮杂三十烷-31-酸(100mg,0.154mmol)的二氯甲烷(5mL)溶液中加入EDC(59mg,0.309mmol)和五氟苯酚(PFP)(57mg,0.309mmol),将混合物在室温下搅拌2小时,用二氯甲烷(20mL)稀释,用水(5mL)洗涤,经硫酸钠干燥,过滤并浓缩。将残余物重新溶解在DMF(5mL)中,然后加入化合物136(49mg,0.23mmol)和DIPEA(90mg,0.69mmol)。将混合物在室温下搅拌1小时,浓缩,并在短硅胶柱上纯化,用甲醇/二氯甲烷(1:10)洗脱,得到标题化合物137(80mg,61%收率)。ESI m/z C 40H 68N 4O 15[M+H] +:计算值845.47,实测值845.47。
实施例146 化合物138的合成
Figure PCTCN2019093946-appb-000252
将化合物137(80mg,0.094mmol)的甲醇(5mL)溶液和10%钯碳(10mg)在氢气(5psi)下搅拌2小时。滤出固体,浓缩滤液,得到无色油状产物(66mg,100%收率),其不经进一步纯化用于下一步。MS-ESI m/z C 32H 62N 4O 13[M+H] +:计算值711.43,实测值711.43。
实施例147 化合物139的合成
Figure PCTCN2019093946-appb-000253
向化合物138(66mg,0.094mmol)中加入乙醇(5mL),2,5-二氧代吡咯烷-1-基4-(2,5-二氧代-2,5-二氢-1H-吡咯-1-基)丁酸酯(39mg,0.141mmol)和PBS(0.1M,pH 7.5,1.0mL)。将反应混合物搅拌过夜,浓缩并在硅胶柱上纯化(二氯甲烷/甲醇=100:0至10:1),得到标题化合物139(37mg,45%收率)。ESI m/z C 40H 69N 5O=[M+H] +:计算值876.47,实测值876.47。
实施例148 化合物140的合成
Figure PCTCN2019093946-appb-000254
将化合物139(50mg,0.057mmol)的二氯甲烷(3mL)溶液在室温下与TFA(1mL)搅拌2小时。将反应混合物浓缩至干,然后重新溶解在二氯甲烷(5mL)中,向其中加入EDC(16mg,0.084mmol)和五氟苯酚(15mg,0.084mmol)。将混合物在室温下搅拌4小时,浓缩,并在硅胶柱上纯化(二氯甲烷/乙酸乙酯=100:10至3:1),得到标题化合物140(41mg,73%收率)。ESI m/z C 42H 60F 5N 5O 16[M+H] +计算值:986.40,实测值986.42。
实施例149 化合物141的合成
Figure PCTCN2019093946-appb-000255
向4-(双(2-(2,5-二氧代-2,5-二氢-1H-吡咯-1-基)乙基)氨基)-4-氧代丁酸(100mg,0.27mmol)的二氯甲烷溶液中加入(5mL)EDC(210mg,1.10mmol)和五氟苯酚(101mg,0.55mmol)。将混合物在室温下搅拌3小时,浓缩并在硅胶柱上纯化(二氯甲烷/乙酸乙酯=20:1至5:1),得到 标题化合物141(114mg,80%收率)。MS-ESI m/z C 22H 16F 5N 3O 7[M+H] +计算值:530.0,实测值530.09。
实施例150.通过还原抗体二硫键同Tubulysin衍生物偶连而制备的偶连物的一般制备方法。
在2.0毫升10毫克/毫升pH值为6.0~8.0的HER2抗体的溶液中,加入0.70ml~2.0ml,100mM磷酸(PBS),pH值为6.5~8.5缓冲液,TCEP(16-20μl,20mM的水溶液),室温下~37.5℃孵育0.5~4小时,加入同当量的叠氮化合物(叠氮苯甲酸,或2-(2-(2-羟基乙氧基)乙氧基)乙氧基叠氮化合物)室温~37.5℃下孵育1~4小时,随后加入含能同巯基基团反应的Tubulysin衍生物分子(28-32μl,20mM的DMA溶液)(如化合物39,57,72,123,或134)。混合物在室温~37.5℃下孵育2~18小时,然后DHAA(135μl,50毫米)加入。在RT隔夜连续培养后,混合物经G-25柱或阳离子层析柱或阴离子层析柱,用10~100mM磷酸,或柠檬酸,50~200mM NaCl pH 6~7.5的缓冲溶液纯化得到共轭体化合物(75%~99%收率)。该纯化步骤也可以用渗析过滤方式,用10~100mM磷酸,或柠檬酸,50~200mM NaCl pH 6~7.5的缓冲溶液,以(3-30倍的被渗析液体积),纯化得到共轭体化合物(75%~99%收率)。通过液相色谱-质谱法质谱测定药物/抗体比率(DAR)是3.1~4.9,HPLC分析为95~99%单体(Tosoh Bioscience,Tskgel G3000SW柱子,内径7.8毫米x 30厘米,0.5毫升/分钟,100分钟)。用化合物39,57,72,123,或134制备的抗体偶连物的结构分别如下:
Figure PCTCN2019093946-appb-000256
C-37、
Figure PCTCN2019093946-appb-000257
C-59或
Figure PCTCN2019093946-appb-000258
C-72
Figure PCTCN2019093946-appb-000259
实施例151.偶联物另外制备方法。
细胞结合分子,(抗体)可通过酰胺、硫醚或二硫键与本专利的化合物进行偶联。用含50mM硼酸钠的PBS缓冲液(pH8.0)稀释抗体(>5mg/mL),加入二硫苏糖醇(终浓度为10mM),35℃处理30分钟,抗体可释放自由巯基。用G-25柱凝胶过滤层析(PBS缓冲液中添加1mM EDTA)后,通过Ellman试剂[5,5’-二硫代双(2-硝基苯甲酸)]测定,每个抗体上大约偶联8个巯基。抗体与Traut's试剂(2-亚氨基噻吩)(Jue,R.,et al.Biochem.1978,17(25):5399-5405)也可释放巯基,或在pH7~8条件下,与SATP(N-琥珀酰亚胺-S-乙酰硫代丙酸盐)或N-琥珀酰亚胺-S-乙酰基(硫代四乙二酸)(SAT(PEG)4)等不同的连接物反应,后经羟胺作用,形成巯基(Duncan,R,et al,Anal.Biochem.1983,132,68-73,Fuji,N.et al,Chem.Pharm.Bull.1985,33,362-367)。基本上,每个抗体分子上连接5~9个巯基分子。
4℃条件下,向冰冷的含有自由巯基的抗体的二甲基乙酰胺(DMA)(2~20%v/v)中加入含有马来酰亚胺或溴乙酰胺基团(需0.5M的硼酸钠溶液 (pH 9)促成抗体与溴乙酰胺的烷基化)的药物(连接的药物与巯基的摩尔比应为1.2~1.5:1)。1~2小时后,添加过量半胱氨酸使反应终止;通过超滤、凝胶层析(G-25,缓冲液为PBS)、无菌过滤后得到浓缩的偶联产物。通过测定280nm和252nm处的吸光度测定蛋白浓度和每个抗体所连接的药物个数。分子排阻HPLC可用来测定连接物的单体形式所占的比重,而低于0.5%的未结合的药物可用RP-HPLC用来测定。对于硫醚键连接形成的单体药物而言,每个抗体分子平均会连接3.2-4.8个tubulysin衍生物。
连接接头的种类可分为二甲基(苯基)甲硅烷基(DMPS),SMDP,4-琥珀酰亚胺基氧羰基-甲基-α(2-吡啶基二硫)甲苯(SMPT),N-琥珀酰亚胺-4-(2-吡啶硫)戊酸酯(SPP),N-琥珀酰亚胺-4-(2-吡啶硫)丙酸酯(SPDP),N-琥珀酰亚胺-4-(2-吡啶硫)丁酸酯(SPDB),琥珀酰亚胺基4-(N-马来酰亚胺甲基)环己烷-1-羧化酯(SMCC),N-羟基琥珀酰亚胺-(聚乙二醇)n-马来酰亚胺(SM(PEG)n)等。抗体(>5mg/mL)稀释于缓冲液(pH 6.5~7.5,5mM PBS,50mM NaCl,1mM EDTA)中,与链接反应2小时,而链接体与抗体的摩尔比为6~10倍以上。反应混合物可通过Sephadex G25凝胶层析分离,其分子量较低的分子会被除去。该纯化步骤也可以用阳离子层析柱或阴离子层析柱,用10~100mM磷酸,或柠檬酸,50~200mM NaCl pH 6~7.5的缓冲溶液纯化得到共轭体化合物(75%~99%收率)。该纯化步骤也可以用渗析过滤方式,用10~100mM磷酸,或柠檬酸,50~200mM NaCl pH 6~7.5的缓冲溶液,以(3-30倍的被渗析液体积),纯化得到共轭体化合物(75%~99%收率)。抗体的浓度通过分光光度法测定,连接物包含吡啶硫基。抗体在280nm处的消光系数为2067550M -1cm -1。修饰的抗体用过量的二硫苏糖醇(20倍以上当量)处理后,测定释放的2-硫吡啶基团,在343、280nm处的消光系数分别为8080和5100M -1cm -1)。向修饰抗体中加入1.2~1.5当量带有巯基基团的Tubulysin衍生物分子。该反应在是室温下进行5~18小时。反应混合物通过Sephadex G25凝胶层析,除去未连接的药物或其他低分子量的物质。再通过测定280nm和252nm处的吸光度来测定连接产物的浓度。连接产物是单体形式,平均每个抗体分子连接3.2~4.8个药物分子。
实施例152.Her2抗体偶联物C-37、C-59、C-72、C-123和C-134的体外细胞毒性评价(与T-DM1对照):
用于细胞毒性测定的细胞系是人胃癌细胞系NCI-N87;细胞在含有10%FBS的RPMI-1640中生长。测定时,将细胞(180μL,6000个细胞)加入到96孔板的每个孔中,并在37℃,5%CO 2下温育24小时。接下来,在合适的细胞培养基(总体积,0.2mL)中,用不同浓度的测试化合物(20μL)处理细胞,对照孔含有细胞和培养基但不含测试化合物。将平板在37℃和5%CO 2下温育120小时,然后向孔中加入MTT(5mg/ml,20μL),并将孔板在37℃下孵育1.5小时。小心去除培养基,然后加入DMSO(180μL)。震荡15分钟,在490nm和570nm测量吸光度,620nm为参考。依照以下公式计算抑制率:抑制率%=[1-(分析值-空白对照值)/(控制值-空白对照值)]×100。化合物细胞毒性结果总结如表1。
表1 本发明申请中的Her-amatoxin类似物偶联物的细胞毒性
化合物 DAR(药物/抗体比) NCI-N87细胞IC 50(nM)
C-37 3.9 0.22
C-59 4.0 0.11
C-72 3.8 0.10
C-123 7.8 0.15
C-134 3.9 0.25
T-DM1 3.5 0.32
实施例153 在荷载NCI-N87异种移植肿瘤的BALB/c裸鼠上的体内抗肿瘤活性研究。
在人胃癌N-87细胞系肿瘤异种移植模型中评估偶联物C-37,C-49,C-72,C-123,C-134和T-DM1的体内疗效。五周龄的雌性BALB/c裸鼠(66只动物)在右肩下区域用0.1mL无血清培养基里的N-87癌细胞(5×10 6个细胞/小鼠)进行皮下接种。肿瘤生长8天后长至平均大小140mm 3。然后将动物随机分成10组(每组6只动物)。第一组小鼠用作对照组,并用以磷酸盐缓冲的盐水注射。剩余的6组分别用偶联物C-37,C-49,C-72,C-123,C-134和T-DM1以6mg/kg的剂量静 脉内注射。每3或者4天测量肿瘤的三维尺寸,并使用公式:肿瘤体积=1/2(长×宽×高)计算肿瘤体积,同时还测量动物的体重。当满足下列任何一项标准时,处死小鼠:(1)体重减少超过注射前体重的20%,(2)肿瘤体积大于1500mm 3,(3)病态严重至无法进食和进水或(4)皮肤坏死。如果肿瘤是未可感知的,则认为小鼠是无肿瘤的。
结果绘制在图7中。在6.0mg/kg剂量,所有6种偶联物都未导致动物体重减轻。对照PBS溶液,所有偶联物均显示抗肿瘤活性。所有测试Tubulysin偶联物均显示比T-DM1优异的抗肿瘤活性。
实施例154.Tubulysin B衍生物的Her2抗体偶联物的毒性研究(与T-DM1对照)。
动物的体重变化(通常是减小)是其对药物的毒性的宏观反映。将56只6-7周龄的雌性ICR小鼠分为7组,每组包含8只动物,分别静脉注射给予150mg/kg剂量的药物C-37,C-49,C-72,C-123,C-134和T-DM1。控制组(8只动物)给予PBS缓冲的生理盐水。如图8显示,在12天的实验中,对照组和T-DM1之外的所有偶联物组中的小鼠的体重均未降低超过5%。相反,T-DM1组中动物体重持续下降,最大减少给药前的24%,并且在研究结束时没有观察到恢复趋势。体重变化结果表明,动物对这些含有支链连接体的Tubulysin B衍生物的Her2抗体偶联物,耐受性高于具有常规单连接体的T-DM1。
前面仅仅示出了本发明的原理,应理解,本发明的范围不预期限制在本文所述的示例性方面,而应包括所有当前已知的和未来开发的等同物。另外,应当指出,在不脱离本发明技术原理的前提下,还可以作出若干改进和修改,这些改进和修改也应被视为本发明的范围。

Claims (54)

  1. 一种抗体-Tubulysin B衍生物(同系物)偶联物,其特征在于,所述偶联物具有如下式(I)所示的结构:
    Figure PCTCN2019093946-appb-100001
    或者以具有式I所示结构为母体的药学上可接受的盐、水合物或水合盐,具有式I所示结构的多晶型物,式I所示结构的光学异构体,式I所示结构的一个或多个氘( 2H)原子代替氢( 1H)原子,或式I所示结构的一个或多个 13C原子代替 12C原子;
    其中,P 1是H、COCH 3、COH、PO(OH) 2、CH 2OPO(OH) 2、CONHCH 3、CON(CH 3) 2、CON(CH 2CH 2) 2NCH 3、CON(CH 2CH 3) 2或CON(CH 2CH 2) 2CHN(CH 2CH 2) 2CH 2;
    R 1、R 2、R 3和R 4分别独立地是H、C 1-C 6烷烃基、C 1-C 6烯烃基、C 1-C 6烷醚基、C 1-C 6烷羰基、C 1-C 6烷酯基、C 1-C 6烷羧基或C 1-C 6烷酰胺基;
    或者,R 1和R 2一起,R 1和R 3一起,R 2和R 3一起,或R 3和R 4一起构成C 2-C 7杂环基或C 2-C 7环烷基结构;
    R 5是H、O-C 1-C 6烷烃基、C(O)-H、C(O)-C 1-C 6(直链或支链)烷烃基、C(O)-NH-C 1-C 6(直链或支链)烷基或C(O)-N(C 1-C 6(直链或支链)烷基) 2
    R 6、R 7和R 8分别独立地是H、C 1-C 6烷烃基、C 1-C 6烷醚基、C 1-C 6烷羰基、C 1-C 6烷酯基、C 1-C 6烷羧基或C 1-C 6烷酰胺基;优选R 6、R 7和R 8分别独立地是H或CH 3
    mAb是抗体、抗体片段、单克隆抗体、多克隆抗体、纳米抗体、前药抗体,或者用合成分子或蛋白进行修饰过的抗体及抗体片段;
    L是含有亲水支链的连接体,其主要构架是C 2-C 100的肽单元(1~12个天然或非天然氨基酸)、腙键基团、二硫化物基团、酯基团、肟基团、酰胺基团或硫醚键基团构成;
    n=1-30。
  2. 如权利要求1所述的偶联物,其特征在于,L的结构为:
    Figure PCTCN2019093946-appb-100002
    其中Aa是L-或D-天然或非天然氨基酸;
    r是0-12之间的整数;当r不是0时,(Aa) r是由相同或不同的氨基酸构成的肽单元;
    m 1=1-18之间的整数;m 2=1-100之间的整数;m 3=1-8之间的整数;m 4=0-8之间的整数;m 5=1-8之间的整数;
    Y是NHC(=O)、NHS(O 2)、NH(SO)、NHS(O 2)NH、NHP(O)(OH)NH或C(O)NH;
    R 9是H、(O=)CR 1、(O=)CNHR 1、R 1COOH、R 1(COCH 2NH) m2H、R 1(Aa) r或R 1(COCH 2NCH 3) m2H,以及
    R 1、m 2和(Aa) r定义如权利要求1和上述定义所述。
  3. 如权利要求1-2中任一项所述的偶联物,其特征在于,所述偶联物的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100003
    其中结构式(II)中的P 1
    Figure PCTCN2019093946-appb-100004
    R 1、R 2、R 3、R 4、R 5、R 6、R 7和R 8以及mAb如权利要求1-2中任一项所述;
    L’的结构为:
    Figure PCTCN2019093946-appb-100005
    其中m 1、m 2、m 3、m 4、m 5、Aa、r和R 9如权利要求1-2中任一项所述。
  4. 如权利要求3所述的偶联物,其特征在于,L’的结构为:
    Figure PCTCN2019093946-appb-100006
    其中m 1、m 2、m 3、m 4、m 5、Aa、r和R 9如权利要求1-2中任一项所述。
  5. 如权利要求3-4中任一项所述的偶联物,其特征在于,mAb-SH的制 备方法包括以下a)~c)中的任一个:
    a).由还原剂(优选,三(2-羧基乙基)膦(TCEP)、二硫苏糖醇(DTT)、二硫季戊四醇(DTE)、L-谷胱甘肽(GSH)、2-巯基乙胺(β-MEA)或/和β-巯基乙醇(β-ME,2-ME))还原抗体、抗体片段、单克隆抗体、多克隆抗体、纳米抗体、前药抗体(probody)或用合成分子或蛋白进行修饰过的抗体及抗体片段重轻链间、重重链间或自身链间的二硫键来获取;
    b).通过Traut试剂或硫内酯与抗体分子的胺反应形成硫醇来制备:
    Figure PCTCN2019093946-appb-100007
    c).在缓冲体系条件下,通过生化反应,在抗体上引入较易还原的二硫键基团,然后用TCEP、DTT、GSH、β-MEA、β-ME还原:
    Figure PCTCN2019093946-appb-100008
  6. 如权利要求3-5中任一项所述的偶联物,其特征在于,偶联物的合成中使用的缓冲体系是:pH 5.0~9.5,浓度为1mM~1000mM的磷酸、醋酸、柠檬酸、硼酸、碳酸、巴比妥酸、Tris(三羟甲基氨基甲烷)、苯甲酸或三乙醇胺、或它们的混合物缓冲溶液,并含有体积比为0%~35%的水溶性有机溶剂:甲醇、乙醇、正丙醇、异丙醇、正丁醇、异丁醇、乙腈、丙酮、DMF、DMA或DMSO,偶联反应温度控制在0℃~45℃,偶联反应是5分 钟~96小时。
  7. 如权利要求3-6中任一项所述的偶联物,其特征在于,在偶联反应完成后,采用超滤或柱层析法进行纯化获得结构式为(I)的偶联物。
  8. 如权利要求7所述的偶联物,其特征在于,所述柱层析包括分子筛柱、阳离子柱、阴离子柱、疏水(HIC)柱、反相柱或者蛋白A或G亲和柱。
  9. 如权利要求3-8中任一项所述的偶联物,其特征在于,结构式为(II)的合成由结构式为(III)的Tubulysin B衍生物和结构式为(L’)的化合物缩合反应获得:
    Figure PCTCN2019093946-appb-100009
    其中X是OH、卤素(F、Cl、Br、或I)、苯酚、五氯酚、三氟甲基磺酸、咪唑、二氯苯酚、四氯苯酚、1-羟基苯并三氮唑、对甲苯磺酸、甲磺酸、2-乙基-5-苯基异噁唑-3'-磺酸、
    Figure PCTCN2019093946-appb-100010
    Figure PCTCN2019093946-appb-100011
    Figure PCTCN2019093946-appb-100012
    自我酸酐或与其他酸酐如乙酰酐、甲酸酐形成的酸酐;或多肽缩合反应中间体或Mitsunobu反应中间体;
    其中,
    缩合反应是在含有体积比为1%-100%的吡啶、三乙胺或二异丙基乙胺的有机溶剂二氯甲烷、二氯乙烷、DMF、DMA、四氢呋喃(THF)、DMSO、丙酮、异丙醇、正丁醇或乙腈中,或上面二种或三种以上溶剂的混合溶剂,或有或无惰性气体(氮气、氩气、氦气)保护下,温度控制在-20℃–150℃条件下,反应时长5分钟–120小时完成;
    或者
    缩合反应在如下缓冲体系及如下条件下进行,所述缓冲体系是:pH 5.0~9.5,浓度为1mM~1000mM的磷酸、醋酸、柠檬酸、硼酸、碳酸、巴比妥酸、Tris(三羟甲基氨基甲烷)、苯甲酸或三乙醇胺、或它们的混合物缓冲溶液,并含有体积比为0%~35%的水溶性有机溶剂:甲醇、乙醇、正丙醇、异丙醇、正丁醇、异丁醇、乙腈、丙酮、DMF、DMA或DMSO,偶联反应温度控制在0℃~45℃,偶联反应是5分钟~96小时。
  10. 如权利要求9所述的偶联物,其特征在于,结构式(III)中的NH 2基团是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸、磺酸盐形式进行缩合反应。
  11. 如权利要求9-10中任一项所述的偶联物,其特征在于,当X是OH时,上述缩合反应需借助缩合试剂,所述缩合试剂是:1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸盐(EDC),二环己基碳二亚胺(DCC),N,N'-二异丙基碳二酰亚胺(DIC),1-环已基-2-吗啉乙基碳二亚胺对甲苯磺酸盐(CMC或CME-CDI),羰基二咪唑(CDI),O-苯并三氮唑-N,N,N',N'-四甲基脲四氟硼酸(TBTU),O-苯并三氮唑-四甲基脲六氟磷酸酯(HBTU),苯并三氮唑-1-基氧基三(二甲基氨基)磷鎓六氟磷酸盐(BOP),六氟磷酸苯并三唑-1-基-氧基三吡咯烷基磷(PyBOP),焦碳酸二乙酯(DEPC),N,N,N',N'-四甲基氯甲脒六氟磷酸盐,2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯(HATU),1-[(二甲胺)(吗啉基)亚甲基]-1[1,2,3]三唑并[4,5-b]1-吡啶-3-氧六氟磷酸盐(HDMA),2-氯-1,3-二甲基咪唑鎓六氟磷酸盐(CIP),氯代三吡咯烷基鏻六氟 磷酸盐(PyCloP),双(四亚甲基)氟代甲酰胺(BTFFH),N,N,N',N'-四甲基-硫-(1-氧代-2-吡啶基)硫脲鎓六氟磷酸盐,2-(2-吡啶酮-1-基)-1,1,3,3-四甲基脲四氟硼酸盐(TPTU),硫-(1-氧代-2-吡啶基)-N,N,N',N'-四甲基硫脲六氟磷酸盐,O-[(乙氧基羰基)氰基甲胺]-N,N,N',N'-四甲基硫脲六氟磷酸盐(HOTU),(1-氰基-2-乙氧基-2-氧代亚乙基氨基氧基)二甲基氨基-吗啉-碳鎓六氟磷酸盐(COMU),(苯并三氮唑-1-基氧基)二吡咯烷碳六氟磷酸盐(HBPyU),N-苄基-N′-环己基碳二亚胺(或荷载在聚合物上),二吡咯烷基(N-琥珀酰亚氨氧基)碳鎓六氟磷酸盐(HSPyU),1-(氯-1-吡咯烷基亚甲基)吡咯烷六氟磷酸盐(PyClU),2-氯-1,3-二甲基咪唑四氟硼酸盐(CIB),(苯并三氮唑-1-基氧基)二哌啶碳六氟磷酸盐(HBPipU),6-氯苯并三氮唑-1,1,3,3-四甲基脲四氟硼酸酯(TCTU),溴化三(二甲基氨基)膦六氟磷酸(BrOP),1-正丙基磷酸酐(PPACA,
    Figure PCTCN2019093946-appb-100013
    ),2-异氰基乙基吗啉(MEI),N,N,N',N'-四甲基脲-氧-(N-琥珀酸亚胺基)六氟磷酸盐(HSTU),2-溴-1-乙基吡啶四氟硼酸盐(BEP),氧-[(乙氧基羰基)氰基甲胺]-N,N,N',N'-四甲基硫尿四氟硼酸盐(TOTU),4-(4,6-二甲氧基三嗪-2-基)-4-甲基吗啉盐酸盐(MMTM,DMTMM),2-琥珀酰亚胺基-1,1,3,3-四甲基脲四氟硼酸酯(TSTU),N,N,N',N'-四甲基-O-(3,4-二氢-4-氧代-1,2,3-苯并三嗪-3-基)脲四氟硼酸盐(TDBTU),偶氮二甲酰二哌啶(ADD),双(4-氯苄基)偶氮二甲酸酯(DCAD),偶氮二甲酸二叔丁酯(DBAD),偶氮二甲酸二异丙酯(DIAD)或偶氮二甲酸二乙酯(DEAD)。
  12. 如权利要求9-11中任一项所述的偶联物,其特征在于,结构式(III)的Tubulysin B衍生物的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100014
    Figure PCTCN2019093946-appb-100015
    R 5’是H、C 1-C 6烷烃基、C 1-C 6烷烃基、或C 1-C 6(直链或支链)胺烷基;
  13. 如权利要求12所述的偶联物,其特征在于,结构式(III)的Tubulysin B衍生物的合成包括下列步骤中的一种或多种:
    步骤1.将二乙氧基乙腈与硫化铵的水溶液在室温搅拌,得到化合物1即2,2-二乙氧基硫代乙酰胺;
    Figure PCTCN2019093946-appb-100016
    步骤2.将化合物1与溴丙酮酸酯在无水溶剂(如无水四氢呋喃、二氯甲烷、乙腈、N,N-二甲基甲酰胺、甲醇、异丙醇)中加热,缩合得化合物2;
    Figure PCTCN2019093946-appb-100017
    步骤3.将化合物2溶解在溶剂(如四氢呋喃、二氯甲烷、乙酸乙酯、正庚烷、二氧六环、乙腈)中,用路易斯酸或质子酸(包括盐酸、硫酸、磷酸、甲磺酸、甲酸、草酸、乙酸、对甲苯磺酸、对甲苯磺酸吡啶、AlCl 3、FeCl 3、ZnCl 2、BF 3、BCl 3、BBr 3、TiCl 4、ZnBr 2、LiBF 4)水解,得到化合物3;
    Figure PCTCN2019093946-appb-100018
    步骤4.不饱和亚磺酰胺在低温条件下(如-45℃至-78℃)被正丁基锂等碱脱氢,然后在路易斯酸存在的条件下与化合物3发生加成反应,得到化合物4;
    Figure PCTCN2019093946-appb-100019
    路易斯酸选择为包括盐酸、硫酸、磷酸、甲磺酸、甲酸、草酸、乙酸、对甲苯磺酸、对甲苯磺酸吡啶、AlCl 3、FeCl 3、ZnCl 2、BF 3、BCl 3、BBr 3、TiCl 4、ZnBr 2、LiBF 4
    步骤5.化合物4在低温条件(如-45℃至-78℃)被下被还原剂(如NaBH 4、LiBH 4、Na(OAc) 3BH、Na(CN)BH 3等)选择性地还原,通过加入路易斯酸(如Ti(Oet)4),控制其立体化学,得到化合物5;
    Figure PCTCN2019093946-appb-100020
    步骤6.将化合物5溶解在溶剂(如甲醇、乙醇、异丙醇、四氢呋喃、乙腈)中,被盐酸、硫酸和磷酸等酸脱去叔丁基亚磺酰基,得到化合物6;
    Figure PCTCN2019093946-appb-100021
    步骤7.将化合物6和叠氮酸溶解在溶剂(如正庚烷、四氢呋喃、二氯 甲烷、N,N-二甲基甲酰胺)中,在缩合试剂(如DIC/HOBt、DCC/HOBt、EDC/HOBt、HATU、BOP、T3P、BrOP)存在下,或通过缩合反应路线,发生缩合反应,得到化合物7;
    或者,叠氮酸与氯甲酸异丁酯在有机碱(如三乙胺、二异丙基乙胺、N-甲基吗啡啉等)存在时在THF中反应,得到混合酸酐,再和化合物6的盐酸盐发生缩合,得到化合物7;
    或者,叠氮酸在溶剂(如正庚烷、正己烷、二氯甲烷、四氢呋喃)中与草酰氯、三乙胺和催化剂量的DMF反应,被转换成酰氯,再和化合物6的盐酸盐发生缩合,得到化合物7;
    Figure PCTCN2019093946-appb-100022
    步骤8.在溶剂(如二氯甲烷、四氢呋喃、乙腈)中,化合物7上的羟基与羟基保护试剂(如TESCl),在有机碱(如咪唑、三乙胺、吡啶)的作用下得到化合物8;
    Figure PCTCN2019093946-appb-100023
    步骤9.化合物8被溶解在溶剂(如四氢呋喃、二氯甲烷、乙腈)中,在碱(如KHMDS,LiHMDS,NaHMDS,KOtBu,NaH,KH)的作用下,酰胺发生去质子化,然后与碘甲烷、溴甲烷、硫酸二甲酯、三氟甲磺酸甲酯或碘乙烷等发生烷基化反应,得到化合物9;
    Figure PCTCN2019093946-appb-100024
    步骤10.化合物9被溶解在溶剂(如四氢呋喃、二氯甲烷、乙酸乙酯)中,其中的叠氮基在一定的条件下,如在氢气和钯碳催化剂、三苯基膦和水(Staudinger反应)的存在下,被还原成氨基,然后与酸或者具有反应活性的酸衍生物发生缩合,得到化合物10;
    Figure PCTCN2019093946-appb-100025
    步骤11.化合物10中的羟基保护基PG 1在适当的条件(如TES保护基团可以在盐酸、THF/MeOH/AcOH、nBu 4NF或吡啶氢氟酸盐的THF溶液中被脱保护)下被脱保护,得到化合物11;
    Figure PCTCN2019093946-appb-100026
    步骤12.化合物11中的酯基在碱(如LiOH、NaOH、KOH)的作用下,或其他适当条件(如甲酯可以在LiCl、LiI、Me3SiOK等试剂的作用下被转化成羧酸)下被转化成酸化合物12;
    Figure PCTCN2019093946-appb-100027
    步骤13.在碱(如三乙胺、N,N-二异丙基乙胺、吡啶)和催化剂(如DMAP)存在时,一定的温度条件(如0℃至23℃)下,化合物12与乙酸酐,丙酸酐、异丙酸酐等酸酐、乙酰卤、丙酸卤、丙酸卤、甲酰胺卤、异酰胺卤、二甲酰胺卤等酸卤作用,得到化合物13,该反应也可不使用碱或者催化剂;
    Figure PCTCN2019093946-appb-100028
    步骤14.化合物13与适当的含羟基的化合物如五氟苯酚或N-羟基琥珀酰亚胺在缩合试剂(如EDC、DIC、DCC、HATU、HBTU)存在时,发生缩合反应,得到具有反应活性的酯化合物14;
    Figure PCTCN2019093946-appb-100029
    步骤15.化合物15与化合物14,在水相、一定的pH值条件(如pH=5.0-8.0)下,或者有机相、存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物16;反应也任选不使用任何碱,条件是需要控制一定的反应温度(如0℃至23℃)和反应时间(如30分钟至18小时);
    Figure PCTCN2019093946-appb-100030
    步骤16.化合物16中的硝基在还原条件下,如在氢气和钯碳催化剂、水合肼和FeCl 3、铁粉和醋酸等的存在的条件下,被还原成氨基,得到化合物III;
    Figure PCTCN2019093946-appb-100031
  14. 如权利要求9-13中任一项所述的偶联物,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100032
    Figure PCTCN2019093946-appb-100033
  15. 如权利要求14所述的偶联物,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
    步骤1.化合物1-1和化合物1-2在缩合剂(如EDC、HATU、DIC、DCC)的作用下缩合,或者经过缩合反应路线发生缩合反应(如化合物1-2在缩合试剂如DIC和EDC的作用下与五氟苯酚、硝基苯酚或N-羟基琥珀酰亚胺发生缩合反应,生产相应的活性酯,然后再与化合物1-1反应),得到化合物物1a;
    或者,化合物1-3和化合物1-4在缩合剂(如EDC、HATU、DIC、DCC)的作用下,或者经过缩合反应路线发生缩合反应,得到化合物物1b;
    Figure PCTCN2019093946-appb-100034
    步骤2.化合物1中的羧基保护基PG 2在脱保护试剂作用下(如叔丁酯基 在酸的作用下),被去除,得到化合物2;
    Figure PCTCN2019093946-appb-100035
    步骤3.含羧基的化合物2和含氨基的化合物3在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物4;
    Figure PCTCN2019093946-appb-100036
    步骤4.化合物4上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
    Figure PCTCN2019093946-appb-100037
    步骤5.含羧基化合物6和含氨基的化合物5在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物7;
    Figure PCTCN2019093946-appb-100038
    步骤6.化合物7上的羧基保护基团PG 3在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除),得到化合物8;
    Figure PCTCN2019093946-appb-100039
    步骤7.化合物8与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯化合物,或与其他酸活化基团反应得到具有缩合反应活性的化合物L’;
    Figure PCTCN2019093946-appb-100040
  16. 如权利要求9-13中任一项所述的偶联物,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100041
  17. 如权利要求16所述的偶联物,其特征在于,结构式为(L’)的化合物的合成包括下列步骤中的一种或多种:
    步骤1.化合物1上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物2;
    Figure PCTCN2019093946-appb-100042
    步骤2.含氨基的化合物2和含羧基的化合物3在缩合试剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物4;
    Figure PCTCN2019093946-appb-100043
    步骤3.化合物4上的羧基保护基团PG 2在去保护条件下被去除,如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物5;
    Figure PCTCN2019093946-appb-100044
    步骤4.含羧基化合物5和含氨基的化合物6在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物7;
    Figure PCTCN2019093946-appb-100045
    步骤5.化合物7上的羧基保护基团PG 3在去保护条件下被去除,如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物8;
    Figure PCTCN2019093946-appb-100046
    步骤6.化合物8与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯化合物,或与其他酸活化基团反应得到具有缩合反应活性的化合物9;
    Figure PCTCN2019093946-appb-100047
  18. 如权利要求3-11中任一项所述的偶联物,其特征在于,结构式为(II)化合物的合成由结构式(IV)和结构式(V)缩合反应获得:
    Figure PCTCN2019093946-appb-100048
    其中X定义及缩合反应条件如权利要求9-11中任一项所述。
  19. 如权利要求18所述的偶联物,其特征在于,结构式(V)中的NH 2基团是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、 柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
  20. 如权利要求18-19中任一项所述的偶联物,其特征在于,结构式(IV)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100049
  21. 如权利要求20所述的偶联物,其特征在于,结构式(IV)的合成包括下列步骤中的任一种:
    羧酸化合物1与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、DIC、DCC、HATU、HBTU)存在时,发生缩合反应,得到具有反应活性的酯;
    或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐;
    或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量(如0.01当量至0.5当量)的DMF存在条件下,反应得到酰氯。
  22. 如权利要求18-21中任一项所述的偶联物,其特征在于,结构式(V)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100050
    Figure PCTCN2019093946-appb-100051
  23. 如权利要求22所述的偶联物,其特征在于,结构式(V)的合成包括下列步骤中的一种或多种:
    步骤1.化合物1与化合物2,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物3;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100052
    步骤2.化合物3上的氨基保护基团PG 4在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物V;
    Figure PCTCN2019093946-appb-100053
  24. 如权利要求22-23中任一项所述的偶联物,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100054
    其中,该合成步骤中得到的化合物8(化合物XIVa)即为所述目标化合物2。
  25. 如权利要求24所述的偶联物,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
    步骤1.将L-酪氨酸的酯类衍生物1溶解于适当的溶剂,如丙酮、四氢呋喃、乙腈、二氯甲烷等,或者这些溶剂与水的混合溶剂,与苄基氯、苄基溴或其它苄基化合物,在0至60℃反应,可在反应体系中加入适当的有机或无机碱,如碳酸钠、碳酸钾、碳酸氢钠、碳酸氢钾、氢氧化钠、氢氧化钾、三乙胺、DBU、氢化钠等,也可在体系内加入适当的添加剂如碘化钠或相转移催化剂,如苄基三乙基氯化铵(TEBA)、四丁基溴化铵(TBAB)、四丁基氯化铵、四丁基硫酸氢铵、三辛基甲基氯化铵、十二烷基三甲基氯化铵、十四烷基三甲基氯化铵等,得到化合物2;
    步骤2.将化合物2溶解于有机溶剂如二氯甲烷、四氢呋喃、甲醇、乙醇、乙醚等,用还原剂,如氢化铝锂、DIBAL、硼氢化钠、硼氢化锂、二氢双(2-甲氧乙氧基)铝酸钠(Red-Al)、乙硼烷等还原,可在反应体系中加入添加剂,如I 2、三氯化铁、氯化锌、氯化镁、氯化锂、氯化钙等控制还原剂的活性,得到化合物3;
    步骤3.醇类化合物3在适当的氧化条件下,如swern氧化(草酰氯、DMSO、三乙胺)、Parikh-Doering氧化(三氧化硫吡啶氧化)、Dess-Martin氧化等,被氧化成醛4;
    步骤4.醛4与磷酸酯反应(Horner-Wadsworth-Emmons反应)或者磷叶立 德反应(Wittig反应),进行碳链延长,得到化合物5;
    步骤5.化合物5中的双键在均相或两相催化剂的作用下被氢化还原,其中的苄基也同时被脱去,得到立体结构单一的手性化合物,或者两个非对映异构体的混合物;催化剂包括Pd/C、Pd(OH) 2/C、Pd/BaSO 4、PtO 2、Pt/Al 2O 3、Ru/C、Raney镍等两相催化剂,均相不对称氢化催化剂,如Crabtree催化剂、[Ru(II)-(BINAP)]类催化剂、[(Ph3P)CuH] 6催化剂等;
    步骤6.将化合物6溶解在有机溶剂中,如四氢呋喃、乙腈、二氯甲烷,在硝化反应条件下被硝化,硝化试剂包括硝酸、硝酸/醋酸、硝酸钾/硫酸、亚硝基叔丁酯、硝酸/三氟乙酸酐、NO 2BF 4、硝基吡啶鎓盐等;
    步骤7.在以下条件下化合物7中的硝基被还原成氨基,这些条件包括H 2/Pd/C,Fe或Zn/HOAc,SnCl 2/HCl。
  26. 如权利要求22-23中任一项所述的偶联物,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100055
    其中,该合成步骤中得到的化合物8(化合物XIVb)即为所述目标化合物2。
  27. 如权利要求26所述的偶联物,其特征在于,化合物2的合成包括下列步骤中的一种或多种:
    步骤1.化合物1在-78℃至-45℃条件下,与Evans手性N-酰基噁唑烷酮或硫酮2,其中X=O或S,R 16=H,甲基,苯基,R 17=H,甲基,异丙基、苯基、苄基等,进行Aldol反应,得到立体构型单一的化合物3;
    步骤2.化合物3上的羟基在以下条件下被脱去,这些条件包括 Barton–McCombie去氧反应,即醇首先转化为硫代酰基衍生物,如烷基黄原酸盐、硫代氯甲酸苯酯、硫代羰基咪唑酯,然后用Bu 3SnH处理,发生自由基断裂得到脱羟基产物;自由基断键的条件包括:n-Bu 3SnH/AIBN,n-Bu 3SnH/AIBN/n-BuOH/PMHS,(Bu 4N) 2S 2O 8/HCO 2Na;
    步骤3.将化合物4溶解在四氢呋喃中,Evans手性辅基在LiOH/H 2O 2的条件下被切断得到相应的酸5;
    步骤4.化合物5被溶解在有机溶剂中,如乙酸乙酯、甲醇、二氯甲烷、乙醇或乙酸等,其中的苄基在钯炭催化剂存在下被催化氢解,得到化合物6;
    步骤5.将化合物6溶解在有机溶剂中,如四氢呋喃、乙腈、二氯甲烷,在硝化反应条件下被硝化,硝化试剂包括硝酸、硝酸/醋酸、硝酸钾/硫酸、亚硝基叔丁酯、硝酸/三氟乙酸酐、NO 2BF 4、硝基吡啶鎓盐等;
    步骤6.在以下条件下化合物7中的硝基被还原成氨基,这些条件包括H 2/Pd/C,Fe或Zn/HOAc,SnCl 2/HCl等,得到立体构型单一的手性化合物8。
  28. 权利要求3-11中任一项所述的偶联物,其特征在于,结构式为(II)的合成由结构式(VI)和结构式(VII)缩合反应获得:
    Figure PCTCN2019093946-appb-100056
    其中X定义及缩合反应条件如权利要求9-11中任一项所述。
  29. 如权利要求28所述的偶联物,其特征在于,结构式(VI)的合成包括下 列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100057
  30. 如权利要求29所述的偶联物,其特征在于,结构式(VI)的合成包括下列步骤中的一种或多种:
    步骤1.化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酸的衍生物化合物2;
    Figure PCTCN2019093946-appb-100058
    步骤2.化合物2与化合物3,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物4;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100059
    步骤3.化合物4上的氨基保护基团PG 4在去保护条件下被选择性地去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
    Figure PCTCN2019093946-appb-100060
    步骤4.化合物5与结构式(IV),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物6;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100061
    步骤5.化合物6上的氨基保护基团PG 1在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物VI;
    Figure PCTCN2019093946-appb-100062
  31. 如权利要求28-30中任一项所述的偶联物,其特征在于,结构式(VII) 的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100063
  32. 如权利要求31所述的偶联物,其特征在于,结构式(VII)的合成包括下列步骤中的一种或多种:
    步骤1.羧酸化合物1与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯;
    或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐;
    或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量(如0.01当量至0.5当量)的DMF存在条件下,反应得到酰氯;
    Figure PCTCN2019093946-appb-100064
  33. 权利要求3-11中任一项所述的偶联物,其特征在于,结构式为(II)的合成由结构式(VIII)和结构式(IX)缩合反应获得:
    Figure PCTCN2019093946-appb-100065
    Figure PCTCN2019093946-appb-100066
    其中X定义及缩合反应条件如权利要求9-11中任一项所述。
  34. 如权利要求33所述的偶联物,其特征在于,结构式(VIII)中的NH 2基团是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
  35. 如权利要求33-34中任一项所述的偶联物,其特征在于,结构式(VIII)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100067
    Figure PCTCN2019093946-appb-100068
  36. 如权利要求35所述的偶联物,其特征在于,结构式(VIII)的合成包括下列步骤中的一种或多种:
    步骤1.化合物1上的羧基保护基PG 3在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除),得到化合物2;
    Figure PCTCN2019093946-appb-100069
    步骤2.化合物2与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂(如EDC、HATU、DIC、DCC)存在时,发生缩合反应,得到具有反应活性的酯化合物3;
    Figure PCTCN2019093946-appb-100070
    步骤3.化合物3与化合物4,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物5;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100071
    步骤4.化合物5上的氨基保护基团PG 3在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc 保护基可以在酸性条件下被切除,得到化合物6;
    Figure PCTCN2019093946-appb-100072
    步骤5.化合物6与结构式(IV)(即权利要求18-21中任一项所述的结构式(IV)),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物7;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100073
    步骤6.化合物7上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物VIII;
    Figure PCTCN2019093946-appb-100074
  37. 如权利要求33-36中任一项所述的偶联物,其特征在于,结构式(IX)的合成包括下列步骤中的一种或多种:
    羧酸化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯IX;
    或者,羧酸化合物1与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱(如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱)存在下反应,得到具有反应活性的混合酸酐IX;
    或者,羧酸化合物1与草酰氯,在三乙胺等有机碱和催化剂量的DMF存在条件下,反应得到酰氯IX;
    Figure PCTCN2019093946-appb-100075
  38. 如权利要求3-11中任一项所述的偶联物,其特征在于,结构式为(II)的合成由结构式(X)和结构式(XI)缩合反应获得:
    Figure PCTCN2019093946-appb-100076
    其中Y 1和Y 2基团缩合构成Y基团;Y 1和Y 2分别是NH 2、- +NH 3、COOH、COX、SO 2Cl、P(O)Cl 2、NHCOX、NHSO 2Cl、NHP(O)Cl 2、NHP(O)(OH)Cl、
    Figure PCTCN2019093946-appb-100077
  39. 如权利要求38所述的偶联物,其特征在于,结构式(X)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100078
  40. 如权利要求39所述的偶联物,其特征在于,结构式(X)的合成包括下列步骤中的一种或多种:
    步骤1.含羧基的化合物1和化合物VI在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线,发生缩合反应,得到化合物2;其中Z 1为Y 1的前体,如经过适当基团保护的氨基、羧基、酰胺基、磷酰胺基和磺酰胺基、羧酸酯、磷酸酯、膦酸酯等;
    Figure PCTCN2019093946-appb-100079
    步骤2.化合物2上的氨基保护基团PG 1在去保护条件下被去除,如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除,氨基上的Boc保护基可以在酸性条件下被切除,得到化合物3;
    Figure PCTCN2019093946-appb-100080
    步骤3.含羧基化合物4和含氨基的化合物3在缩合剂作用下,或者经过缩合反应路线发生缩合反应,得到化合物5;
    Figure PCTCN2019093946-appb-100081
    步骤4.化合物5中的官能团Z 1经过适当的化学转换,如羧基和氨基的脱保护等操作,生成官能团Y 1,得到化合物X;
    Figure PCTCN2019093946-appb-100082
  41. 如权利要求38-40中任一项所述的偶联物,其特征在于,结构式(XI)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100083
  42. 如权利要求41所述的偶联物,其特征在于,结构式(XI)的合成包括下列步骤中的一种或多种:
    步骤1.将化合物1溶解在有机溶剂,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜中,用碱,如氢化钠、钠、氢氧化钠等拔氢,然后与化合物2(其中X为氯、溴、碘等卤素或者其他离去基团)在一定的温度下搅拌,反应得到化合物3;
    步骤2.化合物3上的羧基保护基团PG1在去保护条件下被去除,如叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物XIa-1;
    步骤3.将化合物1溶解在有机溶剂,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜中,用碱,如氢化钠、钠、氢氧化钠等拔氢,然后与化合物4(在一定的温度下搅拌,反应得到化合物5;
    步骤4.化合物5上的羧基保护基团PG 1在去保护条件下被去除,如叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用下被切除,得到化合物XIa-2;
    步骤5.将化合物6溶解在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,加入适当的有机碱,如三乙胺、N,N-二异丙 基乙胺、吡啶等,与甲基磺酰氯、4-甲苯磺酰氯等在0-5℃反应,得到化合物7;
    步骤6.化合物7与氨水在水相或有机溶剂中,如甲醇、乙醇、乙腈、四氢呋喃、环氧六环等,发生反应,反应可适当加热,得到化合物XIb。
    步骤7.化合物7与叠氮化钠在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,发生反应,得到化合物8;
    步骤8.叠氮化合物8在钯碳催化剂存在的条件下被氢化还原,或者三苯基膦和水的作用下被还原,得到化合物XIb;
    步骤9.化合物7与二苄胺在有机溶剂中,如四氢呋喃、二氯甲烷、N,N-二甲基甲酰胺、二甲亚砜等,优选N,N-二甲基甲酰胺,在100℃发生反应,得到化合物9;
    步骤10.将化合物9溶解在溶剂中,如乙酸乙酯、甲醇、乙醇、乙酸、四氢呋喃等,在一定的氢气压力下,在钯碳催化剂上被还原,反应可适当加热至45℃,得到化合物XIb。
  43. 权利要求3-11中任一项所述的偶联物,其特征在于,结构式为(II)的合成由结构式(XII)和结构式(XIII)缩合反应获得:
    Figure PCTCN2019093946-appb-100084
    其中X定义及缩合反应条件如权利要求9-11中任一项所述。
  44. 权利要求43所述的偶联物,其特征在于,结构式(XII)中的NH 2基团 是以三氟乙酸盐、盐酸盐、甲酸盐、醋酸盐、硫酸盐、磷酸盐、硝酸盐、柠檬酸盐、丁二酸盐、苯甲酸或磺酸盐形式进行缩合反应。
  45. 如权利要求43-44中任一项所述的偶联物,其特征在于,结构式(XII)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100085
  46. 如权利要求45所述的偶联物,其特征在于,结构式(XII)的合成包括下列步骤中的一种或多种:
    步骤1.化合物1与适当的含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酸的衍生物化合物2;
    Figure PCTCN2019093946-appb-100086
    步骤2.化合物2与化合物3,在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、 Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物4;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100087
    步骤3.化合物4上的氨基保护基团PG 4在去保护条件下被选择性地去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物5;
    Figure PCTCN2019093946-appb-100088
    步骤4.化合物5与结构式(IV)(即权利要求18-21中任一项所述的结构式(IV)),在水相,一定的pH值条件(如pH=5.0-8.0)下,或者有机相,存在有机碱(如TEA、DBU、DIPEA)或无机碱(如Na 2CO 3、Cs 2CO 3、K 2CO 3、NaHCO 3)时,发生缩合反应,得到化合物6;反应也任选不使用任何碱,但是需要控制合适的反应温度和反应时间;
    Figure PCTCN2019093946-appb-100089
    步骤5.化合物6上的氨基保护基团PG1在去保护条件下被去除(如氨基上的Cbz保护基可以在氢气和钯炭催化剂的作用下被切除),氨基上的Boc保护基可以在酸性条件下被切除,得到化合物XII;
    Figure PCTCN2019093946-appb-100090
  47. 如权利要求43-46中任一项所述的偶联物,其特征在于,结构式(XIII)的合成包括下列步骤中的一种或多种:
    Figure PCTCN2019093946-appb-100091
  48. 如权利要求47所述的偶联物,其特征在于,结构式(XIII)的合成包括下列步骤中的一种或多种:
    步骤1.含羧基化合物1和含氨基的化合物2在缩合剂(如EDC、HATU、DIC、DCC)作用下,或者经过缩合反应路线发生缩合反应,得到化合物3;
    Figure PCTCN2019093946-appb-100092
    步骤2.化合物3上的羧基保护基团PG 1在去保护条件下被去除(如羧基上的叔丁基酯保护基可以在甲酸、乙酸、三氟乙酸、盐酸、磷酸等的作用 下被切除),得到化合物4;
    Figure PCTCN2019093946-appb-100093
    步骤3.羧酸化合物4与含羟基的化合物(如五氟苯酚或N-羟基琥珀酰亚胺)在缩合试剂存在时,发生缩合反应,得到具有反应活性的酯结构式(XIII);
    或者,羧酸化合物4与氯甲酸乙酯、氯甲酸异丁酯等,在有机碱如N-甲基吗啡啉、三乙胺、二异丙基乙胺等有机碱存在下反应,得到具有反应活性的混合酸酐结构式(XIII);
    或者,羧酸化合物4与草酰氯,在三乙胺等有机碱和催化剂量的DMF存在条件下,反应得到酰氯结构式(XIII),
    Figure PCTCN2019093946-appb-100094
  49. 如权利要求1-48中任一项所述的偶联物,其特征在于,所述式(I)化合物的结构如下:
    Figure PCTCN2019093946-appb-100095
    Figure PCTCN2019093946-appb-100096
  50. 一种化合物,其特征在于,所述式(II)化合物结构如下:
    Figure PCTCN2019093946-appb-100097
    Figure PCTCN2019093946-appb-100098
  51. 一种药物组合物,其包含权利要求1-49中任一项所述的偶联物或以权利要求50所述的化合物构成的偶联物,以及药学上可接受的辅料。
  52. 权利要求1-49中任一项所述的偶联物在用于制备治疗癌症、感染或自身免疫疾病的药物中用途。
  53. 权利要求50所述的化合物在用于制备治疗癌症、感染或自身免疫疾病的药物中用途。
  54. 权利要求51所述的药物组合物在用于制备治疗癌症、感染或自身免疫疾病的药物中用途。
PCT/CN2019/093946 2019-06-24 2019-06-29 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法 WO2021000067A1 (zh)

Priority Applications (27)

Application Number Priority Date Filing Date Title
JP2022500117A JP2022540395A (ja) 2019-06-29 2019-06-29 細胞結合性分子-チューブリシン誘導体共役体及びその調製方法
EP19936127.0A EP3991752A4 (en) 2019-06-29 2019-06-29 CELL BINDING MOLECULE-TUBULYSIN DERIVATIVE CONJUGATE AND METHOD OF PREPARATION THEREOF
IL289458A IL289458A (en) 2019-06-29 2019-06-29 A cell-binding conjugated derivative of the tubolisin molecule and a method for its production
AU2019452577A AU2019452577B2 (en) 2019-06-29 2019-06-29 Cell-binding molecule-Tubulysin derivative conjugate and preparation method therefor
US17/623,360 US20230025327A1 (en) 2019-06-29 2019-06-29 Conjugates of tubulysin derivatives and cell binding molecules and methods of making
KR1020227003562A KR20220029725A (ko) 2019-06-29 2019-06-29 세포-결합 분자-튜불리신 유도체 접합체 및 이의 제조 방법
CA3145050A CA3145050A1 (en) 2019-06-29 2019-06-29 Conjugates of tubulysin derivatives and cell binding molecules and methods of making
CN201980095886.3A CN114040778A (zh) 2019-06-29 2019-06-29 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法
BR112021025984A BR112021025984A2 (pt) 2019-06-29 2019-06-29 Conjugado de anticorpo-derivado de tubulisina b (análogo), composto, composição farmacêutica, e, usos do conjugado, dos compostos e das composições farmacêuticas
PCT/CN2019/093946 WO2021000067A1 (zh) 2019-06-29 2019-06-29 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法
BR112021026009A BR112021026009A2 (pt) 2019-06-24 2020-02-18 Composições líquida e farmacêutica, conjugados dos análogos de tubulisina, análogo de tubulisina, agentes de ligação celular e sinérgico, e, célula tumoral
EP20831886.5A EP3986476A4 (en) 2019-06-24 2020-02-18 FORMULATION OF A CONJUGATE OF A TUBULYSIN ANALOG TO A CELL BINDING MOLECULE
CA3142960A CA3142960C (en) 2019-06-24 2020-02-18 A formulation of a conjugate of a tubulysin analog to a cell-binding molecule
US17/596,956 US20220249594A1 (en) 2019-06-24 2020-02-18 A formulation of a conjugate of a tubulysin analog to a cell-binding molecule
PCT/CN2020/075709 WO2020258893A1 (en) 2019-06-24 2020-02-18 A formulation of a conjugate of a tubulysin analog to a cell-binding molecule
MX2021015878A MX2021015878A (es) 2019-06-24 2020-02-18 Una formulación de un conjugado de un análogo de tubulisina con una molécula de unión celular.
CN202080032694.0A CN114040781A (zh) 2019-06-24 2020-02-18 Tubulysin衍生物与细胞结合分子偶联物的制剂配方
JP2021576898A JP2022544442A (ja) 2019-06-24 2020-02-18 チューブリシン類縁体と細胞結合分子との共役体の製剤
KR1020227002437A KR20220024915A (ko) 2019-06-24 2020-02-18 튜불리신 유사체 대 세포-결합 분자의 접합체의 제형
SG11202113022UA SG11202113022UA (en) 2019-06-24 2020-02-18 A formulation of a conjugate of a tubulysin analog to a cell-binding molecule
AU2020301115A AU2020301115A1 (en) 2019-06-24 2020-02-18 A formulation of a conjugate of a tubulysin analog to a cell-binding molecule
TW109116404A TWI785334B (zh) 2019-06-29 2020-05-18 一種細胞結合分子-tubulysin衍生物偶聯物及其製備方法
TW110104872A TW202144009A (zh) 2019-06-24 2021-02-09 Tubulysin衍生物與細胞結合分子偶聯物的製劑配方
IL288954A IL288954A (en) 2019-06-24 2021-12-13 Conjugation formulation of a tubulosin analog to a cell-binding molecule
CL2021003468A CL2021003468A1 (es) 2019-06-24 2021-12-24 Una formulación de un conjugado de un análogo de tubulisina con una molécula de unión celular
ZA2022/01370A ZA202201370B (en) 2019-06-29 2022-01-28 Cell-binding molecule-tubulysin derivative conjugate and preparation method therefor
JP2023203751A JP2024028815A (ja) 2019-06-24 2023-12-01 チューブリシン類縁体と細胞結合分子との共役体の製剤

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
PCT/CN2019/093946 WO2021000067A1 (zh) 2019-06-29 2019-06-29 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法

Publications (1)

Publication Number Publication Date
WO2021000067A1 true WO2021000067A1 (zh) 2021-01-07

Family

ID=74100095

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2019/093946 WO2021000067A1 (zh) 2019-06-24 2019-06-29 一种细胞结合分子-Tubulysin衍生物偶联物及其制备方法

Country Status (12)

Country Link
US (1) US20230025327A1 (zh)
EP (1) EP3991752A4 (zh)
JP (1) JP2022540395A (zh)
KR (1) KR20220029725A (zh)
CN (1) CN114040778A (zh)
AU (1) AU2019452577B2 (zh)
BR (1) BR112021025984A2 (zh)
CA (1) CA3145050A1 (zh)
IL (1) IL289458A (zh)
TW (1) TWI785334B (zh)
WO (1) WO2021000067A1 (zh)
ZA (1) ZA202201370B (zh)

Cited By (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3862023A1 (en) * 2020-02-05 2021-08-11 Hangzhou DAC Biotech Co, Ltd Conjugates of cell-binding molecules with cytotoxic agents
US11767294B2 (en) 2012-07-12 2023-09-26 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents
US11873281B2 (en) 2012-07-12 2024-01-16 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115536530A (zh) * 2022-10-10 2022-12-30 浙江大学 一种Vulgarisin型四环二萜化合物的化学全合成方法

Citations (77)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4341761A (en) 1980-07-25 1982-07-27 E. I. Du Pont De Nemours And Company Antibodies to immunogenic peptides and their use to purify human fibroblast interferon
US4399121A (en) 1981-11-04 1983-08-16 Miles Laboratories, Inc. Iodothyronine immunogens and antibodies
US4427783A (en) 1981-12-14 1984-01-24 Hoffmann-La Roche Inc. Immunoassay of thymosin α1
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4451570A (en) 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
US4466917A (en) 1981-02-12 1984-08-21 New York University Malaria vaccine
US4472500A (en) 1980-07-07 1984-09-18 National Research Development Corporation Rat myeloma cell lines
US4491632A (en) 1979-10-22 1985-01-01 The Massachusetts General Hospital Process for producing antibodies to hepatitis virus and cell lines therefor
US4493890A (en) 1981-03-23 1985-01-15 Miles Laboratories, Inc. Activated apoglucose oxidase and its use in specific binding assays
US4493795A (en) 1983-10-17 1985-01-15 Syntex (U.S.A.) Inc. Synthetic peptide sequences useful in biological and pharmaceutical applications and methods of manufacture
US5436149A (en) 1993-02-19 1995-07-25 Barnes; Wayne M. Thermostable DNA polymerase with enhanced thermostability and enhanced length and efficiency of primer extension
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
DE19638870A1 (de) 1996-09-23 1998-03-26 Biotechnolog Forschung Gmbh Verbindungen mit antimykotischer und cytostatischer Wirkung, Herstellungsverfahren, Mittel und DSM 11 092
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6111166A (en) 1994-09-19 2000-08-29 Medarex, Incorporated Transgenic mice expressing human Fcα and β receptors
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6207418B1 (en) 1996-10-11 2001-03-27 Abgenix, Inc. Production of a multimeric protein by cell fusion method
DE10008089A1 (de) 2000-02-22 2001-10-31 Biotechnolog Forschung Gmbh Syntheseverfahren zur Herstellung von Tubulysinen
WO2002077036A2 (en) 2001-03-21 2002-10-03 Cell Therapeutics, Inc. Recombinant production of polyanionic polymers, and uses thereof
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
WO2004005327A1 (de) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Neue tubulysinanaloga
WO2004005269A1 (de) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Verfahren zur herstellung von substituierten thiazol-2-ylmethylestern
WO2004005326A2 (de) 2002-07-09 2004-01-15 Morphochem Aktiengellschaft Fu Tubulysinkonjugate
DE10241152A1 (de) 2002-09-05 2004-03-18 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysin-Biosynthese-Gene
DE10254439A1 (de) 2002-11-21 2004-06-03 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysine, Herstellungsverfahren und Tubulysin-Mittel
US6797492B2 (en) 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
DE102004030227A1 (de) 2004-06-23 2006-01-26 Dömling, Alexander, Dr. Wirkstoffe mit antiangiogenetischen Eigenschaften
WO2006033913A2 (en) 2004-09-16 2006-03-30 Synta Pharmaceuticals Corp. Bis (thio-hydrazide amides) for treament of hyperplasia
WO2006056464A2 (en) 2004-11-26 2006-06-01 Pieris Ag Compound with affinity for the cytotoxic t lymphocyte-associated antigen (ctla-4)
WO2006069246A2 (en) 2004-12-22 2006-06-29 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
WO2006096754A2 (en) 2005-03-07 2006-09-14 Archemix Corp. Stabilized aptamers to psma and their use as prostate cancer therapeutics
US20070041901A1 (en) 2002-06-18 2007-02-22 Diener John L Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
WO2007059312A2 (en) 2005-11-16 2007-05-24 Ambrx, Inc. Methods and compositions comprising non-natural amino acids
WO2007130453A2 (en) 2006-05-02 2007-11-15 Allozyne, Inc. Non-natural amino acid substituted polypeptides
WO2008076333A2 (en) 2006-12-13 2008-06-26 Insert Therapeutics, Inc. Polymer-drug conjugates with tether groups for controlled drug delivery
WO2008112873A2 (en) 2007-03-14 2008-09-18 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2008125116A2 (en) 2007-03-31 2008-10-23 Aarhus Universitet Upar inhibition
WO2008138561A1 (en) 2007-05-10 2008-11-20 R & D Biopharmaceuticals Gmbh Tubulysine derivatives
WO2008141044A2 (en) 2007-05-08 2008-11-20 Genentech, Inc. Cysteine engineered anti-muc16 antibodies and antibody drug conjugates
WO2009002993A1 (en) 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
WO2009012958A2 (en) 2007-07-20 2009-01-29 Helmholtz-Zentrum für Infektionsforschung GmbH Tubulysin d analogues
WO2009026177A1 (en) 2007-08-17 2009-02-26 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
WO2009055562A1 (en) 2007-10-25 2009-04-30 Endocyte, Inc. Tubulysins and processes for preparing
WO2009095447A1 (en) 2008-01-30 2009-08-06 Pieris Ag Muteins of tear lipocalin having affinity to human c-met receptor tyrosine kinase and methods for obtaining the same
WO2009134279A1 (en) 2007-07-20 2009-11-05 The Regents Of The University Of California Tubulysin d analogues
US7638299B2 (en) 2004-07-21 2009-12-29 Ambrx, Inc. Biosynthetic polypeptides utilizing non-naturally encoded amino acids
WO2010033733A1 (en) 2008-09-17 2010-03-25 Endocyte, Inc. Folate receptor binding conjugates of antifolates
WO2010034724A1 (en) 2008-09-25 2010-04-01 Universität des Saarlandes Bioactive pre-tubulysins and use thereof
WO2010081110A1 (en) 2009-01-12 2010-07-15 Sutro Biopharma, Inc. Dual charging system for selectively introducing non-native amino acids into proteins using an in vitro synthesis method
US20100210543A1 (en) 2009-02-17 2010-08-19 David Rabuka Aldehyde-Tagged Protein-Based Drug Carriers and Methods of Use
US7829659B2 (en) 2006-05-02 2010-11-09 Allozyne, Inc. Methods of modifying polypeptides comprising non-natural amino acids
CA2710693A1 (en) 2009-07-22 2011-01-22 Kemotech S.R.L. Pharmaceutical compositions
US20110027274A1 (en) 2009-08-03 2011-02-03 Medarex, Inc. Antiproliferative compounds, conjugates thereof, methods therefor, and uses thereof
US7893019B2 (en) 2006-08-11 2011-02-22 Bio-Ker S.R.L. G-CSF site-specific mono-conjugates
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
US20130189287A1 (en) 2011-12-23 2013-07-25 Paul Scherrer Institut Enzymatic conjugation of polypeptides
WO2013190272A1 (en) 2012-06-18 2013-12-27 Polytherics Limited Conjugation reagents
WO2014009774A1 (en) 2012-07-12 2014-01-16 Hangzhou Dac Biotech Co., Ltd Conjugates of cell binding molecules with cytotoxic agents
WO2014064424A1 (en) 2012-10-24 2014-05-01 Polytherics Limited Drug-protein conjugates
US20140141025A1 (en) 2012-11-16 2014-05-22 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US20140294867A1 (en) 2013-03-11 2014-10-02 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
US8871908B2 (en) 2011-11-11 2014-10-28 Rinat Neuroscience Corp. Antibodies specific for Trop-2 and their uses
US8916159B2 (en) 2007-04-02 2014-12-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Selenocysteine mediated hybrid antibody molecules
WO2015151081A2 (en) 2015-07-12 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Bridge linkers for conjugation of a cell-binding molecule
WO2015151080A2 (en) 2015-07-04 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Specific conjugation of a cell-binding molecule
WO2015155753A2 (en) 2015-08-10 2015-10-15 Suzhou M-Conj Biotech Co., Ltd Novel linkers and their uses in specific conjugation of drugs to a biological molecule
WO2016059628A1 (en) 2014-10-12 2016-04-21 Korol Oleg System and method for determining watering needs for field or landscape irrigation
CN105641707A (zh) * 2014-11-11 2016-06-08 杭州多禧生物科技有限公司 细胞毒素分子同细胞结合受体分子的共轭体
CN109912683A (zh) * 2017-12-13 2019-06-21 杭州多禧生物科技有限公司 一种细胞毒素分子、偶联物及其制备方法和应用
WO2019127607A1 (en) * 2017-12-31 2019-07-04 Hangzhou Dac Biotech Co. Ltd A conjugate of a tubulysin analog with branched linkers
CN110099682A (zh) * 2016-11-14 2019-08-06 杭州多禧生物科技有限公司 偶联连接体,含有此连接体的细胞结合分子-药物偶联物及其制备和应用

Patent Citations (91)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4491632A (en) 1979-10-22 1985-01-01 The Massachusetts General Hospital Process for producing antibodies to hepatitis virus and cell lines therefor
US4444887A (en) 1979-12-10 1984-04-24 Sloan-Kettering Institute Process for making human antibody producing B-lymphocytes
US4472500A (en) 1980-07-07 1984-09-18 National Research Development Corporation Rat myeloma cell lines
US4341761A (en) 1980-07-25 1982-07-27 E. I. Du Pont De Nemours And Company Antibodies to immunogenic peptides and their use to purify human fibroblast interferon
US4466917A (en) 1981-02-12 1984-08-21 New York University Malaria vaccine
US4493890A (en) 1981-03-23 1985-01-15 Miles Laboratories, Inc. Activated apoglucose oxidase and its use in specific binding assays
US4451570A (en) 1981-03-26 1984-05-29 The Regents Of The University Of California Immunoglobulin-secreting human hybridomas from a cultured human lymphoblastoid cell line
US4399121A (en) 1981-11-04 1983-08-16 Miles Laboratories, Inc. Iodothyronine immunogens and antibodies
US4427783A (en) 1981-12-14 1984-01-24 Hoffmann-La Roche Inc. Immunoassay of thymosin α1
US4493795A (en) 1983-10-17 1985-01-15 Syntex (U.S.A.) Inc. Synthetic peptide sequences useful in biological and pharmaceutical applications and methods of manufacture
US5859205A (en) 1989-12-21 1999-01-12 Celltech Limited Humanised antibodies
US6075181A (en) 1990-01-12 2000-06-13 Abgenix, Inc. Human antibodies derived from immunized xenomice
US6150584A (en) 1990-01-12 2000-11-21 Abgenix, Inc. Human antibodies derived from immunized xenomice
US5545806A (en) 1990-08-29 1996-08-13 Genpharm International, Inc. Ransgenic non-human animals for producing heterologous antibodies
US5569825A (en) 1990-08-29 1996-10-29 Genpharm International Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US5661016A (en) 1990-08-29 1997-08-26 Genpharm International Inc. Transgenic non-human animals capable of producing heterologous antibodies of various isotypes
US6797492B2 (en) 1991-05-17 2004-09-28 Merck & Co., Inc. Method for reducing the immunogenicity of antibody variable domains
US5436149A (en) 1993-02-19 1995-07-25 Barnes; Wayne M. Thermostable DNA polymerase with enhanced thermostability and enhanced length and efficiency of primer extension
US5922545A (en) 1993-10-29 1999-07-13 Affymax Technologies N.V. In vitro peptide and antibody display libraries
US6111166A (en) 1994-09-19 2000-08-29 Medarex, Incorporated Transgenic mice expressing human Fcα and β receptors
DE19638870A1 (de) 1996-09-23 1998-03-26 Biotechnolog Forschung Gmbh Verbindungen mit antimykotischer und cytostatischer Wirkung, Herstellungsverfahren, Mittel und DSM 11 092
US6207418B1 (en) 1996-10-11 2001-03-27 Abgenix, Inc. Production of a multimeric protein by cell fusion method
DE10008089A1 (de) 2000-02-22 2001-10-31 Biotechnolog Forschung Gmbh Syntheseverfahren zur Herstellung von Tubulysinen
US6596541B2 (en) 2000-10-31 2003-07-22 Regeneron Pharmaceuticals, Inc. Methods of modifying eukaryotic cells
WO2002077036A2 (en) 2001-03-21 2002-10-03 Cell Therapeutics, Inc. Recombinant production of polyanionic polymers, and uses thereof
US20070041901A1 (en) 2002-06-18 2007-02-22 Diener John L Stabilized aptamers to PSMA and their use as prostate cancer therapeutics
US20040249130A1 (en) 2002-06-18 2004-12-09 Martin Stanton Aptamer-toxin molecules and methods for using same
WO2004005327A1 (de) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Neue tubulysinanaloga
WO2004005269A1 (de) 2002-07-09 2004-01-15 Morphochem Ag Komb Chemie Verfahren zur herstellung von substituierten thiazol-2-ylmethylestern
WO2004005326A2 (de) 2002-07-09 2004-01-15 Morphochem Aktiengellschaft Fu Tubulysinkonjugate
DE10241152A1 (de) 2002-09-05 2004-03-18 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysin-Biosynthese-Gene
DE10254439A1 (de) 2002-11-21 2004-06-03 GESELLSCHAFT FüR BIOTECHNOLOGISCHE FORSCHUNG MBH (GBF) Tubulysine, Herstellungsverfahren und Tubulysin-Mittel
DE102004030227A1 (de) 2004-06-23 2006-01-26 Dömling, Alexander, Dr. Wirkstoffe mit antiangiogenetischen Eigenschaften
US7638299B2 (en) 2004-07-21 2009-12-29 Ambrx, Inc. Biosynthetic polypeptides utilizing non-naturally encoded amino acids
WO2006033913A2 (en) 2004-09-16 2006-03-30 Synta Pharmaceuticals Corp. Bis (thio-hydrazide amides) for treament of hyperplasia
US7521541B2 (en) 2004-09-23 2009-04-21 Genetech Inc. Cysteine engineered antibodies and conjugates
US8309300B2 (en) 2004-09-23 2012-11-13 Genentech, Inc. Cysteine engineered antibodies and conjugates
US7855275B2 (en) 2004-09-23 2010-12-21 Genentech, Inc. Cysteine engineered antibodies and conjugates
WO2006056464A2 (en) 2004-11-26 2006-06-01 Pieris Ag Compound with affinity for the cytotoxic t lymphocyte-associated antigen (ctla-4)
US7332571B2 (en) 2004-12-22 2008-02-19 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
US7696312B2 (en) 2004-12-22 2010-04-13 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
WO2006069246A2 (en) 2004-12-22 2006-06-29 Ambrx, Inc. Compositions containing, methods involving, and uses of non-natural amino acids and polypeptides
WO2006096754A2 (en) 2005-03-07 2006-09-14 Archemix Corp. Stabilized aptamers to psma and their use as prostate cancer therapeutics
WO2007059312A2 (en) 2005-11-16 2007-05-24 Ambrx, Inc. Methods and compositions comprising non-natural amino acids
US7829659B2 (en) 2006-05-02 2010-11-09 Allozyne, Inc. Methods of modifying polypeptides comprising non-natural amino acids
WO2007130453A2 (en) 2006-05-02 2007-11-15 Allozyne, Inc. Non-natural amino acid substituted polypeptides
US7632492B2 (en) 2006-05-02 2009-12-15 Allozyne, Inc. Modified human interferon-β polypeptides
US7893019B2 (en) 2006-08-11 2011-02-22 Bio-Ker S.R.L. G-CSF site-specific mono-conjugates
US8097701B2 (en) 2006-09-21 2012-01-17 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
US8349910B2 (en) 2006-09-21 2013-01-08 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
US7985783B2 (en) 2006-09-21 2011-07-26 The Regents Of The University Of California Aldehyde tags, uses thereof in site-specific protein modification
WO2008076333A2 (en) 2006-12-13 2008-06-26 Insert Therapeutics, Inc. Polymer-drug conjugates with tether groups for controlled drug delivery
WO2008112873A2 (en) 2007-03-14 2008-09-18 Endocyte, Inc. Binding ligand linked drug delivery conjugates of tubulysins
WO2008125116A2 (en) 2007-03-31 2008-10-23 Aarhus Universitet Upar inhibition
US8916159B2 (en) 2007-04-02 2014-12-23 The United States Of America, As Represented By The Secretary, Department Of Health And Human Services Selenocysteine mediated hybrid antibody molecules
WO2008141044A2 (en) 2007-05-08 2008-11-20 Genentech, Inc. Cysteine engineered anti-muc16 antibodies and antibody drug conjugates
US7723485B2 (en) 2007-05-08 2010-05-25 Genentech, Inc. Cysteine engineered anti-MUC16 antibodies and antibody drug conjugates
WO2008138561A1 (en) 2007-05-10 2008-11-20 R & D Biopharmaceuticals Gmbh Tubulysine derivatives
US20120129779A1 (en) 2007-05-10 2012-05-24 R&D-Biopharmaceuticals Gmbh Tubulysine derivatives
WO2009002993A1 (en) 2007-06-25 2008-12-31 Endocyte, Inc. Conjugates containing hydrophilic spacer linkers
WO2009012958A2 (en) 2007-07-20 2009-01-29 Helmholtz-Zentrum für Infektionsforschung GmbH Tubulysin d analogues
US20110021568A1 (en) 2007-07-20 2011-01-27 The Regents Of The University Of California Tubulysin d analogues
US20110263650A1 (en) 2007-07-20 2011-10-27 Helmholtz-Zentrum Für Infektions-Forschung Gmbh Tubulysin D Analogues
WO2009134279A1 (en) 2007-07-20 2009-11-05 The Regents Of The University Of California Tubulysin d analogues
WO2009026177A1 (en) 2007-08-17 2009-02-26 Purdue Research Foundation Psma binding ligand-linker conjugates and methods for using
WO2009055562A1 (en) 2007-10-25 2009-04-30 Endocyte, Inc. Tubulysins and processes for preparing
WO2009095447A1 (en) 2008-01-30 2009-08-06 Pieris Ag Muteins of tear lipocalin having affinity to human c-met receptor tyrosine kinase and methods for obtaining the same
WO2010033733A1 (en) 2008-09-17 2010-03-25 Endocyte, Inc. Folate receptor binding conjugates of antifolates
WO2010034724A1 (en) 2008-09-25 2010-04-01 Universität des Saarlandes Bioactive pre-tubulysins and use thereof
EP2174947A1 (en) 2008-09-25 2010-04-14 Universität des Saarlandes Bioactive pre-tubulysins and use thereof
US8778631B2 (en) 2009-01-12 2014-07-15 Sutro Biopharma, Inc. Mono charging system for selectively introducing non-native amino acids into proteins using an in vitro protein synthesis system
US20100184135A1 (en) 2009-01-12 2010-07-22 Sutro Biopharma, Inc. Mono charging system for selectively introducing non-native amino acids into proteins using an in vitro protein synthesis system
WO2010081110A1 (en) 2009-01-12 2010-07-15 Sutro Biopharma, Inc. Dual charging system for selectively introducing non-native amino acids into proteins using an in vitro synthesis method
US20100210543A1 (en) 2009-02-17 2010-08-19 David Rabuka Aldehyde-Tagged Protein-Based Drug Carriers and Methods of Use
CA2710693A1 (en) 2009-07-22 2011-01-22 Kemotech S.R.L. Pharmaceutical compositions
US20110027274A1 (en) 2009-08-03 2011-02-03 Medarex, Inc. Antiproliferative compounds, conjugates thereof, methods therefor, and uses thereof
US8871908B2 (en) 2011-11-11 2014-10-28 Rinat Neuroscience Corp. Antibodies specific for Trop-2 and their uses
US20130189287A1 (en) 2011-12-23 2013-07-25 Paul Scherrer Institut Enzymatic conjugation of polypeptides
WO2013190272A1 (en) 2012-06-18 2013-12-27 Polytherics Limited Conjugation reagents
WO2014009774A1 (en) 2012-07-12 2014-01-16 Hangzhou Dac Biotech Co., Ltd Conjugates of cell binding molecules with cytotoxic agents
WO2014064424A1 (en) 2012-10-24 2014-05-01 Polytherics Limited Drug-protein conjugates
US20140141025A1 (en) 2012-11-16 2014-05-22 Redwood Bioscience, Inc. Hydrazinyl-indole compounds and methods for producing a conjugate
US20140294867A1 (en) 2013-03-11 2014-10-02 Genzyme Corporation Site-specific antibody-drug conjugation through glycoengineering
WO2016059628A1 (en) 2014-10-12 2016-04-21 Korol Oleg System and method for determining watering needs for field or landscape irrigation
CN105641707A (zh) * 2014-11-11 2016-06-08 杭州多禧生物科技有限公司 细胞毒素分子同细胞结合受体分子的共轭体
WO2015151080A2 (en) 2015-07-04 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Specific conjugation of a cell-binding molecule
WO2015151081A2 (en) 2015-07-12 2015-10-08 Suzhou M-Conj Biotech Co., Ltd Bridge linkers for conjugation of a cell-binding molecule
WO2015155753A2 (en) 2015-08-10 2015-10-15 Suzhou M-Conj Biotech Co., Ltd Novel linkers and their uses in specific conjugation of drugs to a biological molecule
CN110099682A (zh) * 2016-11-14 2019-08-06 杭州多禧生物科技有限公司 偶联连接体,含有此连接体的细胞结合分子-药物偶联物及其制备和应用
CN109912683A (zh) * 2017-12-13 2019-06-21 杭州多禧生物科技有限公司 一种细胞毒素分子、偶联物及其制备方法和应用
WO2019127607A1 (en) * 2017-12-31 2019-07-04 Hangzhou Dac Biotech Co. Ltd A conjugate of a tubulysin analog with branched linkers

Non-Patent Citations (92)

* Cited by examiner, † Cited by third party
Title
"Antibodies--A Laboratory Manual", 1988, COLD SPRING HARBOR LABORATORY PRESS
"The Handbook of Chemistry and Physics", 1997, CRC PRESS, INC., pages: 225 - 226
ADAMS ET AL., J IMMUNOL METHODS., vol. 231, 1999, pages 249 - 60
ALMAGRO ET AL., FRONT BIOSCI, vol. 13, 2008, pages 1619 - 33
ASCO ANNUAL MEETING, 2015
ASCO ANNUAL MEETING, 2016
AXUP, J. Y ET AL., PROC. NAT. ACAD. SCI. USA., vol. 109, 2012, pages 16101 - 6
BALASUBRAMANIAN, R. ET AL., J. MED. CHEM., vol. 52, 2009, pages 238 - 40
BANDER, N. H. ET AL., CLIN. ADV. HEMATOL. ONCOL., vol. 10, 2012, pages 1 - 16
BARRIOS, C. H. ET AL., J. CLIN. ONCOL., vol. 34, 2016
BEERLI, R. R. ET AL., PLOS ONE, vol. 10, 2015, pages e0131177
BEHRENS, C. R.LIU, B., MABS, vol. 6, 2014, pages 46 - 53
BOUVIER ET AL., METH. ENZYMOL., vol. 248, 1995, pages 614
BOYLAN, N. J., BIOCONJ. CHEM.,, vol. 24, 2013, pages 1008 - 1016
BRANNIGAN ET AL., NAT REV MOL CELL BIOL., vol. 3, 2002, pages 964 - 70
CHAI, Y. ET AL., CHEM BIOL, vol. 17, 2010, pages 296 - 309
CHANDRASEKHAR, S. ET AL., J. ORG. CHEM., vol. 74, 2009, pages 9531 - 4
CHEN, T., BIOMED. ANAL., vol. 117, 2016, pages 304 - 10
CLACKSON ET AL., NATURE, vol. 352, 1991, pages 264 - 8
DENTE ET AL., GENE, vol. 148, no. 1, 1994, pages 595 - 13
DOMLING, A. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 45, 2006, pages 7235 - 9
DOVGAN, I. ET AL., SCI. REP., vol. 6, 2016, pages 30835
DULBECCO ET AL.: "Virol", vol. 8, 1959, pages: 396
DUNCAN, R ET AL., ANAL. BIOCHEM., vol. 132, 1983, pages 68 - 73
ELLIS, P. A. ET AL., J. CLIN. ONCOL., vol. 33, 2015
FRANKE ET AL., CANCER BIOTHER RADIOPHARM, vol. 15, 2000, pages 459 - 76
FRIESTAD, G. K. ET AL., ORG. LETT., vol. 11, no. 24, 2009, pages 1095 - 8
FRIESTAD, G. K., ORG. LETT., vol. 6, 2004, pages 3249 - 52
FUJI, N ET AL., CHEM. PHARM. BULL., vol. 33, 1985, pages 362 - 367
G.S. HAMILTON, BIOLOGICALS, vol. 43, 2015, pages 318 - 32
GERBE ET AL., MABS, vol. 1, no. 3, 2009, pages 247 - 53
GEYSEN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 82, 1985, pages 1418 - 82
GOEIJ, B. E.LAMBERT, J. M., CURR. OPIN. IMMUNOL., vol. 40, 2016, pages 14 - 23
GOPAL, A. ET AL., BLOOD, vol. 125, 2015, pages 1236 - 43
GRIINEWALD, J. ET AL., BIOCONJ. CHEM., vol. 26, 2015, pages 2554 - 62
HAMBLETT, K. J. ET AL., CLIN. CANCER RES., vol. 10, 2004, pages 7063 - 70
HOFER, T. ET AL., BIOCHEMISTRY, vol. 48, 2009, pages 12047 - 57
HOUDEBINE, CURR OPIN BIOTECHNOL, vol. 13, 2002, pages 625 - 9
HUSE ET AL., SCIENCE, vol. 246, 1989, pages 1275 - 81
JONES, M. W., SOC., vol. 134, 2012, pages 1847 - 52
JUE, R. ET AL., BIOCHEM, vol. 17, no. 25, 1978, pages 5399 - 5405
JUNUTULA, J. R. ET AL., NAT. BIOTECHNOL., vol. 26, 2008, pages 925 - 32
JUNUTULAJ. R. ET AL., CLIN. CANCER RES., vol. 16, 2010, pages 4769
KANG, H. J. ET AL., SCIENCE, vol. 318, 2007, pages 1625 - 8
KIPRIYANOV ET AL., MOL BIOTECHNOL, vol. 26, 2004, pages 39 - 60
KOHLER, G.MILSTEIN, C., NATURE, vol. 256, 1975, pages 495 - 7
KUBICEK, K. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 49, 2010, pages 4809 - 12
L. DUCRYB STUMP, BIOCONJ. CHEM., vol. 21, 2010, pages 5 - 13
LAMBERT, J. M., THER. DELIV., vol. 7, 2016, pages 279 - 82
LAMBERT, J.CHARI, R., J. MED. CHEM., vol. 57, 2014, pages 6949 - 64
LAZAR ET AL., MOL IMMUNOL, vol. 44, no. 8, 2007, pages 1986 - 98
LEI ET AL., BIOCHEMISTRY, vol. 34, no. 20, 1995, pages 6675 - 88
LI ET AL., PROC. NATL. ACAD. SCI. USA., vol. 103, no. 10, 2006, pages 3557 - 62
LI, X. ET AL., METHODS, vol. 65, 2014, pages 133 - 8
LITTLE ET AL., BIOTECHNOLADV, vol. 12, no. 3, 1994, pages 539 - 55
LIU ET AL., IMMUNOL REV., vol. 222, 2008, pages 9 - 27
LIU, Y. ET AL., MOL. PHARMACEUTICS, vol. 9, 2012, pages 168 - 75
MARUANI, A ET AL., NAT. COMMUN., vol. 6, 2015, pages 6645
MATAYOSHI ET AL., SCIENCE, vol. 247, 1990, pages 954
NIMAN ET AL., PROC. NATL. ACAD. SCI. USA, vol. 80, 1983, pages 4949 - 53
NOVELLINO ET AL., CANCER IMMUNOL IMMUNOTHER, vol. 54, no. 3, 2005, pages 187 - 207
OKELEY, N. ET AL., HEMATOL ONCOL. CLIN. NORTH. AM., vol. 28, 2014, pages 13 - 25
OKELEY, N. M. ET AL., BIOCONJ. CHEM., vol. 24, 2013, pages 1650
P. G. WUTST.W GREENE: "Greene's Protective Groups in Organic Synthesis", 2006, WILEY-INTERSCIENCE
PANDO, O. ET AL., J. AM. CHEM. SOC., vol. 133, 2011, pages 7692 - 5
PATTERSON, A. W. ET AL., J. ORG. CHEM., vol. 73, 2008, pages 4362 - 9
PEDDI, P.HURVITZ, S., THER. ADV. MED. ONCOL., vol. 6, no. 5, 2014, pages 202 - 209
PELTIER, H. M. ET AL., J. AM. CHEM. SOC., vol. 128, 2006
PIWKO C ET AL., CLIN. DRUG INVESTIG., vol. 35, no. 8, 2015, pages 487 - 93
PONTE, J. ET AL., BIOCONJ. CHEM.,, vol. 27, no. 7, 2016, pages 1588 - 98
R ZHAO ET AL., 14TH PEGS BOSTON, 3 May 2018 (2018-05-03)
RABUKA, D. ET AL., NAT. PROTOC., vol. 7, 2012, pages 1052 - 67
RAGHAVAN, B. ET AL., J. MED. CHEM., vol. 51, 2008, pages 1530 - 33
REDDY, J. A. ET AL., MOL. PHARMACEUTICS, vol. 6, 2009, pages 1518 - 25
RICHARD C. LAROCK: "Remington: The Science and Practice of Pharmacy", 1999, LIPPINCOTT WILLIAMS & WILKINS
ROBERTS, S. A. ET AL., REGUL. TOXICOL. PHARMACOL., vol. 67, 2013, pages 382 - 91
ROSS, P. L.WOLFE, J. L., J. PHARM. SCI., vol. 105, no. 2, pages 391 - 7
SANI, M. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 46, 2007, pages 3526 - 9
SEIDAH ET AL., METH. ENZYMOL., vol. 244, 1994, pages 412
SHEN, K. ET AL., SCI REP., vol. 6, 2016, pages 23262
SIEGMUND, V. ET AL., ANGEW. CHEMIE - INT. ED., vol. 54, 2015, pages 13420 - 4
STROP, P. ET AL., CHEM. BIOL., vol. 20, 2013, pages 161 - 67
SZARDENINGS, J RECEIVE SIGNAL TRANSFER RES, vol. 23, no. 4, 2003, pages 307 - 49
ULLRICH, A. ET AL., ANGEW. CHEM. INT. ED. ENGL., vol. 48, 2009, pages 4422 - 5
WAKANKAR, A, MABS, vol. 3, 2011, pages 161 - 172
WIPF, P. ET AL., ORG. LETT., vol. 9, no. 8, 2007, pages 1605 - 7
WU, P. ET AL., PROC. NATL. ACAD. SCI., vol. 106, 2009, pages 3000 - 5
ZAKERI, B. ET AL., PROC. NATL. ACAD. SCI. USA, vol. 109, 2012, pages E690 - 7
ZAKERI, B.HOWARTH, M., J. AM. CHEM. SOC., vol. 132, 2010, pages 4526 - 7
ZHANG, C ET AL., NAT. CHEM., vol. 8, 2015, pages 1 - 9
ZHAO, R. Y. ET AL., J. MED. CHEM., vol. 54, 2011, pages 3606 - 23
ZIMMERMAN, E.S. ET AL., BIOCONJ. CHEM., vol. 25, 2014, pages 1331 - 4 1

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11767294B2 (en) 2012-07-12 2023-09-26 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents
US11834406B2 (en) 2012-07-12 2023-12-05 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents
US11873281B2 (en) 2012-07-12 2024-01-16 Hangzhou Dac Biotech Co., Ltd. Conjugates of cell binding molecules with cytotoxic agents
EP3862023A1 (en) * 2020-02-05 2021-08-11 Hangzhou DAC Biotech Co, Ltd Conjugates of cell-binding molecules with cytotoxic agents

Also Published As

Publication number Publication date
US20230025327A1 (en) 2023-01-26
TW202114740A (zh) 2021-04-16
TWI785334B (zh) 2022-12-01
BR112021025984A2 (pt) 2022-04-12
EP3991752A4 (en) 2023-03-29
IL289458A (en) 2022-07-01
CN114040778A (zh) 2022-02-11
AU2019452577B2 (en) 2023-08-24
EP3991752A1 (en) 2022-05-04
CA3145050A1 (en) 2021-01-07
JP2022540395A (ja) 2022-09-15
AU2019452577A1 (en) 2022-02-24
ZA202201370B (en) 2022-10-26
KR20220029725A (ko) 2022-03-08

Similar Documents

Publication Publication Date Title
CN109912683B (zh) 一种细胞毒素分子、偶联物及其制备方法和应用
JP7295640B2 (ja) アマニタ毒素の誘導体及びそれらと細胞結合分子との共役体
CN110099682B (zh) 偶联连接体,含有此连接体的细胞结合分子-药物偶联物及其制备和应用
KR102641565B1 (ko) 분지형 링커를 갖는 튜불리신 유사체의 접합체
JP6700321B2 (ja) 細胞結合分子の特異的共役体
JP7429987B2 (ja) 架橋ピロロベンゾジアゼピン二量体(pbd)誘導体及びその共役体
AU2012385228B2 (en) Conjugates of cell binding molecules with cytotoxic agents
CN110621673A (zh) 双链连接的细胞毒性药物偶联物
TW202108179A (zh) 細胞毒性劑經由支鏈連接子與細胞結合分子的偶聯物
TWI785334B (zh) 一種細胞結合分子-tubulysin衍生物偶聯物及其製備方法
TW202216206A (zh) 細胞結合分子與喜樹鹼類似物的偶聯物
JP2022544442A (ja) チューブリシン類縁体と細胞結合分子との共役体の製剤
CN113423430A (zh) 含支链连接子的鹅膏毒素偶联物
JP2020063254A (ja) 細胞結合分子の特異的共役体
TW202041237A (zh) 含支鏈連接體的Tubulysin同系物偶聯物

Legal Events

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

Ref document number: 19936127

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3145050

Country of ref document: CA

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2022500117

Country of ref document: JP

Kind code of ref document: A

REG Reference to national code

Ref country code: BR

Ref legal event code: B01A

Ref document number: 112021025984

Country of ref document: BR

ENP Entry into the national phase

Ref document number: 20227003562

Country of ref document: KR

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 2019936127

Country of ref document: EP

Effective date: 20220131

ENP Entry into the national phase

Ref document number: 2019452577

Country of ref document: AU

Date of ref document: 20190629

Kind code of ref document: A

ENP Entry into the national phase

Ref document number: 112021025984

Country of ref document: BR

Kind code of ref document: A2

Effective date: 20211221