CN113813399A - Camptothecin compound, antibody drug conjugate thereof and application thereof - Google Patents

Camptothecin compound, antibody drug conjugate thereof and application thereof Download PDF

Info

Publication number
CN113813399A
CN113813399A CN202110420028.0A CN202110420028A CN113813399A CN 113813399 A CN113813399 A CN 113813399A CN 202110420028 A CN202110420028 A CN 202110420028A CN 113813399 A CN113813399 A CN 113813399A
Authority
CN
China
Prior art keywords
antibody
cancer
compound
val
formula
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202110420028.0A
Other languages
Chinese (zh)
Inventor
郭茂君
李辉
李海泓
庞彬彬
贾忠全
王威威
许喆
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Levena Biopharma Co ltd
Original Assignee
Levena Biopharma Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Levena Biopharma Co ltd filed Critical Levena Biopharma Co ltd
Priority to CN202110420028.0A priority Critical patent/CN113813399A/en
Publication of CN113813399A publication Critical patent/CN113813399A/en
Pending legal-status Critical Current

Links

Images

Classifications

    • 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
    • A61K47/54Medicinal 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 organic compound
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/4738Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/4745Quinolines; Isoquinolines ortho- or peri-condensed with heterocyclic ring systems condensed with ring systems having nitrogen as a ring hetero atom, e.g. phenantrolines
    • 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
    • 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
    • A61K47/54Medicinal 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 organic compound
    • A61K47/542Carboxylic acids, e.g. a fatty acid or an amino acid
    • 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
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/68Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an antibody, an immunoglobulin or a fragment thereof, e.g. an Fc-fragment
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Epidemiology (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Molecular Biology (AREA)
  • Immunology (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Medicines Containing Antibodies Or Antigens For Use As Internal Diagnostic Agents (AREA)

Abstract

The invention provides camptothecin compounds, antibody drug conjugates thereof and applications thereof. The camptothecin compounds of the invention have a structure shown in the following formula I, wherein R is1、R2And R3As described herein. The invention also provides an antibody drug conjugate containing the structure shown in the formula I. The compound and the antibody drug conjugate shown in the formula I have improved stability and antitumor activity.

Description

Camptothecin compound, antibody drug conjugate thereof and application thereof
Technical Field
The invention relates to a camptothecin compound, an antibody drug conjugate thereof and application thereof.
Background
Camptothecin (CPT), a cytotoxic quinoline alkaloid, selectively inhibits topoisomerase I (Topo I) and binds to the complex formed by Topo I-DNA, stabilizing the complex, thereby preventing rejoining of the broken DNA strands, preventing DNA replication and RNA synthesis. Camptothecin is a cell cycle S-phase specific drug, has no effect on G0-phase cells, and has slight lethality on G1, G2 and M-phase cells. Camptothecin can be used for the treatment of cancer, including gastric cancer, esophageal cancer, gastric cardia cancer, colon cancer, rectal cancer, primary liver cancer, acute and chronic myelogenous leukemia, chorioepithelial cancer, lung cancer, and bladder cancer.
7-ethyl-10-hydroxycamptothecin (SN38) is a metabolite of camptothecin in vivo, has better tumor inhibition effect, and is also an intermediate of irinotecan serving as an antitumor drug.
Disclosure of Invention
In a first aspect herein, there is provided a compound of formula I:
Figure BDA0003027506470000011
in the formula (I), the compound is shown in the specification,
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
R2selected from H and C1-C4 alkyl;
R3is selected from-NH2-OH and-NO2
A second aspect herein provides a compound represented by formula II:
Figure BDA0003027506470000021
in the formula (I), the compound is shown in the specification,
z is-NH-, -NMe or-O-;
d represents a drug molecule, said "-Z-D" meaning that drug molecule D is covalently linked to the remainder of the compound of formula II through the oxygen in its own hydroxyl group or the nitrogen in an amine group;
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
y is-NH-, -O-or-NH-SO2-;
L1is-La-Lb-, wherein LaSelected from C1-C6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene; l isbIs absent, or is-CO-, -NH-, -COO-or-NH-SO2-;
L2Is a peptide that can be recognized and hydrolyzed by enzymes in the pathological environment; and
R4a group which can react with a free amino group on a thiol group or a lysine residue in a cysteine residue in a polypeptide to thereby attach the compound represented by formula II to the polypeptide.
In a third aspect, herein is provided an antibody drug conjugate having the structure shown in formula III below:
Figure BDA0003027506470000022
in the formula (I), the compound is shown in the specification,
ab is an antibody or antigen-binding fragment thereof;
x represents the bond to R on the antibody or antigen-binding fragment thereof4A group formed after a coupling reaction;
m is an integer of 1 to 8;
R4、L1、L2、Y、Z、R1and D is as described in any embodiment herein.
In a fourth aspect, there is provided a pharmaceutical composition comprising an antibody drug conjugate according to any one of the embodiments herein and a pharmaceutically acceptable carrier.
A fifth aspect herein provides a use selected from:
(1) use of a compound according to any one of the embodiments herein for the preparation of an antibody drug conjugate;
(2) use of an antibody drug conjugate according to any of the embodiments herein in the manufacture of a medicament for the treatment or prevention of cancer.
The detailed description and preferred embodiments of each aspect herein are as detailed in other portions of this application.
Drawings
FIG. 1: MS spectrum of compound 7.
FIG. 2: HIC map of Trop2 ADC: LN 401-34-3.
FIG. 3: trop2 ADC: SEC profile of LN 401-34-3.
FIG. 4: trop2 expresses a dose response curve of low (+) to high (+++) BxPC-3, MDA-MB-468 and MDA-MB-231 and Trop 2-negative (-) SW620 human tumor cells after 5 days of treatment with anti-Trop 2 ADC.
Detailed Description
It is intended to provide a novel method for liberating camptothecin or derivatives from PAB and water in acetal of camptothecin or derivatives thereof, which are produced after hydrolysis of peptide chains, and corresponding compounds. The camptothecin derivative and the antibody drug conjugate thereof provided by the invention have more excellent anti-tumor effect and stability.
Term(s) for
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which the claimed subject matter belongs. All patents, patent applications, and publications cited herein are incorporated by reference in their entirety unless otherwise indicated.
It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the subject matter claimed. In this application, the use of the singular also includes the plural unless specifically stated otherwise. It must be noted that, as used in this specification and the appended claims, the singular forms "a," "an," and "the" include plural referents unless the context clearly dictates otherwise. It should also be noted that the use of "or", "or" means "and/or" unless stated otherwise. Furthermore, the term "comprising" as well as other forms, such as "includes," "including," and "containing," are not limiting and can be open, semi-closed, and closed. In other words, the term also includes the meaning of "consisting essentially of …," or "consisting of ….
Definitions for the terms of the standardization sector can be found in the literature references including Carey and Sundberg "ADVANCED ORGANIC CHEMISTRY 4TH ED." Vols.A (2000) and B (2001), Plenum Press, New York. Unless otherwise indicated, conventional methods within the skill of the art are employed, such as mass spectrometry, NMR, IR and UV/VIS spectroscopy, and pharmacological methods. Unless a specific definition is set forth, the terms used herein in the pertinent description of analytical chemistry, organic synthetic chemistry, and pharmaceutical chemistry are known in the art. Standard techniques can be used in chemical synthesis, chemical analysis, pharmaceutical preparation, formulation and delivery, and treatment of patients. For example, the reaction and purification can be carried out using the instructions of the kit from the manufacturer, or according to the methods known in the art or the instructions of the present invention. The techniques and methods described above can generally be practiced according to conventional methods well known in the art, as described in various general and more specific documents referred to and discussed in this specification. In the present specification, groups and substituents thereof may be selected by one skilled in the art to provide stable moieties and compounds.
When a substituent is described by a general formula written from left to right, the substituent also includes chemically equivalent substituents obtained when the formula is written from right to left. For example, -CH2O-is equivalent to-OCH2-。
Certain chemical groups defined herein are preceded by a shorthand notation to indicate the total number of carbon atoms present in the group. For example, C1-6 alkyl refers to an alkyl group as defined below having a total of 1 to 6 carbon atoms. The total number of carbon atoms in the shorthand notation excludes carbons that may be present in a substituent of the group.
In addition to the foregoing, the following terms, when used in the specification and claims of this application, have the meanings indicated below, unless otherwise specifically indicated.
In the present application, the term "halogen" refers to fluorine, chlorine, bromine or iodine.
"hydroxy" means an-OH group.
"hydroxyalkyl" refers to an alkyl group as defined below substituted with a hydroxyl group (-OH).
"carbonyl" refers to a-C (═ O) -group.
"nitro" means-NO2
"cyano" means-CN.
"amino" means-NH2
"carboxyl" means-COOH.
In the present application, "alkyl" as a group or as part of another group refers to a fully saturated straight or branched hydrocarbon chain radical consisting only of carbon and hydrogen atoms, having, for example, from 1 to 10 (preferably from 1 to 8, more preferably from 1 to 6) carbon atoms, and being attached to the rest of the molecule by a single bond. Alkyl groups include, but are not limited to, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, 2-methylbutyl, 2-dimethylpropyl, n-hexyl, heptyl, 2-methylhexyl, 3-methylhexyl, octyl, nonyl, decyl, and the like. In some embodiments, the alkyl group is a C1-C4 alkyl group.
In the present application, "alkenyl" as a group or as part of another group refers to a straight or branched hydrocarbon chain group consisting of only carbon and hydrogen atoms, containing at least one double bond, having, for example, 2 to 10 (preferably 2 to 8, more preferably 2 to 6, more preferably 2 to 4) carbon atoms, and connected to the rest of the molecule by a single bond. Alkenyl groups include, but are not limited to, ethenyl, propenyl, allyl, but-1-enyl, but-2-enyl, pent-1, 4-dienyl, and the like.
In the present application, "alkynyl" as a group or as part of another group refers to a straight or branched hydrocarbon chain group consisting solely of carbon and hydrogen atoms, containing at least one triple bond, having, for example, from 2 to 10 (preferably from 2 to 8, more preferably from 2 to 6, more preferably from 2 to 4) carbon atoms, and being attached to the rest of the molecule by a single bond. Alkynyl groups include, but are not limited to, ethynyl, propynyl, and the like.
In the present application, alkylene, alkenylene and alkynylene refer to divalent alkyl, divalent alkenyl and divalent alkynyl groups that lack 2H atoms. Exemplary alkylene groups include-CH2-、-CH2CH2-and the like; exemplary alkenylene groups include-CH ═ CH-, -CH ═ CH-CH2-and the like; exemplary alkynylene groups include-C.ident.C-, -C.ident.C-CH2-and the like.
It will also be appreciated by those skilled in the art that in the processes described below, the functional groups of the intermediate compounds may need to be protected by suitable protecting groups. Such functional groups include hydroxyl, amino, mercapto and carboxylic acid. Suitable hydroxy protecting groups include trialkylsilyl or diarylalkylsilyl groups (e.g.tert-butyldimethylsilyl, tert-butyldiphenylsilyl or trimethylsilyl), tetrahydropyranyl, benzyl, and the like. Suitable protecting groups for amino, amidino and guanidino include t-butyloxycarbonyl, benzyloxycarbonyl and the like. Suitable thiol protecting groups include-C (O) -R "(where R" is alkyl, aryl or aralkyl), p-methoxybenzyl, trityl and the like. Suitable carboxyl protecting groups include alkyl, aryl or aralkyl esters.
Protecting groups may be introduced and removed according to standard techniques known to those skilled in the art and as described herein. The use of protecting Groups is described in detail in Greene, T.W. and P.G.M.Wuts, Protective Groups in organic Synthesis, (1999),4th Ed., Wiley. In some embodiments, the protecting group herein is an amino protecting group, such as Boc (tert-butyloxycarbonyl). The protecting group may also be a polymeric resin.
Camptothecin derivatives
The camptothecin derivatives provided by the invention have improved stability. Provided herein are camptothecin derivatives represented by the following formula I:
Figure BDA0003027506470000061
in the formula (I), the compound is shown in the specification,
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
R2selected from H and C1-C4 alkyl;
R3is selected from-NH2-OH and-NO2
In formula I, preferably, R1Selected from H and C1-C4 alkyl, more preferably H. Preferably, R2Is H, methyl or ethyl, more preferably ethyl. Preferably, R3is-NH2or-OH, more preferably-NH2
In some embodiments, the compound of formula I is a compound represented by formula 3 or 4 below:
Figure BDA0003027506470000062
drugs covalently linked to linkers
Provided herein are drugs covalently linked by a linker compound represented by the following formula II:
Figure BDA0003027506470000071
in the formula (I), the compound is shown in the specification,
z is-NH-, -NMe or-O-;
d represents a drug molecule, said "-Z-D" meaning that drug molecule D is covalently linked to the remainder of the compound of formula II through the oxygen in its own hydroxyl group or the nitrogen in an amine group;
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
y is-NH-, -O-or-NH-SO2-;
L1Is selected from-La-Lb-, wherein LaSelected from C1-C6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene; l isbIs absent, or is-CO-, -NH-, -COO-, or-NH-SO2-;
L2Is a peptide that can be recognized and hydrolyzed by enzymes in the pathological environment;
R4a group which can react with a free amino group on a thiol group or a lysine residue in a cysteine residue in a polypeptide to thereby attach the compound represented by formula II to the polypeptide.
Herein, the linker refers to R4-L1-L2-a group.
In formula II, preferably, the drug is selected from: MMAE, Duo-5, DXD, irinotecan, camptothecin, 7-ethyl-10-hydroxycamptothecin, topotecan, fluorouracil, doxifluridine, cytarabine, etoposide, fludarabine, capecitabine, vincristine, epothilone B, paclitaxel, docetaxel, daunorubicin, epirubicin, methotrexate, gemcitabine, melphalan, nimustine, mitoxantrone, doxorubicin, and mitomycin. Preferably, the drug is camptothecin or 7-ethyl-10-hydroxycamptothecin. It will be appreciated that when the drug is attached to the remainder of formula II other than the "-Z-D" moiety through the nitrogen of its amine group or the oxygen of its hydroxyl group, the attachment site on the drug does not interfere with the biological function and activity of the drug itself. For example, the remainder of formula II may be covalently linked via an amino or hydroxyl group at a position remote from the active center, according to the known active center of these drugs.
In formula II, preferably, R1Selected from H and C1-C4 alkyl, more preferably H.
In formula II, preferably, LbAbsent or as a carbonyl group; preferably, L1Through LbAnd said L2Covalent attachment; preferably, LbAnd L2Form an amide group therebetween. Preferably, L1In, with R4Linked LaIs C1-C6 alkylene, with L2Linked Lbis-CO-. More preferably still, the first and second liquid crystal compositions are,L1is C1-C6 alkylene or C1-C6 alkylenecarbonyl.
In formula II, preferably, L is2The peptide is dipeptide, tripeptide, tetrapeptide or pentapeptide. In one or more embodiments, the amino acid residues in the peptide are selected from the group consisting of: one or more of valine, ornithine, lysine, serine, glycine, and citrulline. In one or more embodiments, the peptide is Val-Cit, Val-Orn, Val-Lys, Ser-Val-Lys, Gly-Val-Lys, Ser-Val-Cit, Gly-Gly-Gly-Val-Lys, or Gly-Gly-Val-Lys. In a preferred embodiment, the peptide is Val-Cit.
Preferably, in formula II, the enzyme is a proteolytic enzyme, a protease, or a peptidase. In one or more embodiments, the enzyme is selected from the group consisting of: one or more of a cysteine protease, an aspartic protease, a glutamic protease, a threonine protease, a gelatinase, a metalloprotease, and an asparagine peptide cleaving enzyme. Particularly preferably, the enzyme is asparagine endopeptidase (Legumain).
L1And L2And L2And R1The substituted phenyl groups can be each independently substituted by-CO-NH-, -COO-, -O-SO2-NH-and-NH-SO2-NH-is covalently linked. Preferably, L1And L2And L2And R1The substituted phenyl groups may each be covalently linked to one another via-CO-NH-or-COO-. Thus, in formula II, preferably Y is-O-or-NH-. Preferably, in formula II, Y is-O-or-NH-, Lbis-CO-.
In the formula II, R4And L1Can be selected from carbon-carbon single bond, amido (-CO-NH-), ester (-COO-), carbonyl (-CO-), -O-SO2-NH-and-NH-SO2-NH-covalently linked, these groups may be R4A portion of a group. L is1Through L thereofaAnd R4Covalent attachment, e.g., direct covalent attachment to such linking groups.
Preferably, R4Selected from the following groups:
Figure BDA0003027506470000091
wherein the wavy line represents R4And L1The location of the connection.
More preferred R4Selected from:
Figure BDA0003027506470000092
preferably, the polypeptide is a polypeptide having a desired biological activity, including but not limited to enzymes and antibodies, particularly the various antibodies described herein. The antibody is preferably a monoclonal antibody.
In a preferred embodiment, the compound of formula II is a compound represented by formula 7 below:
Figure BDA0003027506470000093
ADC
the compounds of formula II described herein may be conjugated to an antibody or antigen-binding fragment thereof to form an antibody conjugated drug (ADC). Accordingly, also provided herein is an ADC having the structure shown in formula III below:
Figure BDA0003027506470000094
Figure BDA0003027506470000101
in the formula (I), the compound is shown in the specification,
ab is an antibody or antigen-binding fragment thereof;
x represents the bond to R on the antibody or antigen-binding fragment thereof4A group formed after a coupling reaction;
m is an integer of 1 to 8;
R4、L1、L2、Y、Z、R1and D is as in any one of the embodiments hereinbefore.
In a preferred embodiment, in formula III, R4Selected from:
Figure BDA0003027506470000102
L1in, with R4Linked LaIs C1-C6 alkylene, with L2Linked Lbis-CO-; l is2Is Val-Cit, Val-Orn, Val-Lys, Ser-Val-Lys, Gly-Val-Lys, Ser-Val-Cit, Gly-Gly-Gly-Val-Lys or Gly-Gly-Val-Lys, preferably Val-Cit; y is-O-or-NH-; r1Is H or C1-C4 alkyl, preferably H; z is-NH-, -NMe or-O-; d is MMAE, Duo-5, DXD, irinotecan, camptothecin, 7-ethyl-10-hydroxycamptothecin, topotecan, fluorouracil, doxifluridine, cytarabine, etoposide, fludarabine, capecitabine, vincristine, epothilone B, paclitaxel, docetaxel, daunorubicin, epirubicin, methotrexate, gemcitabine, melphalan, nimustine, mitoxantrone, doxorubicin or mitomycin, preferably camptothecin or 7-ethyl-10-hydroxycamptothecin.
Herein, "antibody" has a meaning well known in the art and includes any form of antibody having the desired biological activity, such as monoclonal antibodies (including full length monoclonal antibodies), polyclonal antibodies, multispecific antibodies (e.g., bispecific antibodies), humanized antibodies, fully human antibodies, chimeric antibodies, camelized single domain antibodies, and the like.
By "monoclonal antibody" is meant an antibody obtained from a substantially homogeneous population of antibodies, i.e., the individual antibodies comprising the population are identical except for possible naturally occurring mutations that may be present in minor amounts. Monoclonal antibodies are highly specific, being directed against a single epitope. In contrast, conventional (polyclonal) antibody preparations typically include a large number of antibodies directed against (or specific for) different epitopes.
"full-length antibody" refers to an immunoglobulin molecule that, when naturally occurring, comprises at least four peptide chains: the two heavy (H) chains and the two light (L) chains are linked to each other by disulfide bonds. Each heavy chain consists of a heavy chain variable region (abbreviated herein as VH) and a heavy chain constant region (abbreviated herein as CH). The heavy chain constant region consists of 3 domains, CH1, CH2, and CH 3. Each light chain consists of a light chain variable region (abbreviated herein as VL) and a light chain constant region. The light chain constant region consists of one domain CL. The VH and VL regions can be further subdivided into Complementarity Determining Regions (CDRs) with high variability and regions that are spaced apart to be more conserved, called Framework Regions (FRs). Each VH or VL region is formed by, in the following order: FR1, CDR1, FR2, CDR2, FR3, CDR3, FR4 are composed of 3 CDRs and 4 FRs arranged from amino terminus to carboxy terminus. The variable regions of the heavy and light chains contain binding domains that interact with antigens. The constant region of an antibody may mediate the binding of the immunoglobulin to host tissues or factors, including various cells of the immune system (e.g., effector cells) and the first component of the classical complement system (Clq).
An "antigen-binding fragment" of an antibody includes a fragment or derivative of an antibody, typically including at least one fragment of an antigen-binding region or variable region (e.g., one or more CDRs) of a parent antibody, which retains at least some of the binding specificity of the parent antibody. Examples of antibody binding fragments include, but are not limited to, Fab ', F (ab')2, and Fv fragments; a diabody; a linear antibody; single chain antibody molecules, such as sc-Fv; nanobodies and multispecific antibodies formed from antibody fragments. When the binding activity of an antigen is expressed on a molar concentration basis, the binding fragment or derivative typically retains at least 10% of its antigen binding activity. Preferably, the binding fragment or derivative retains at least 20%, 50%, 70%, 80%, 90%, 95% or 100% or more of the antigen binding affinity of the parent antibody. It is also contemplated that antigen-binding fragments of an antibody may include conservative or non-conservative amino acid substitutions (referred to as "conservative variants" or "functionally conservative variants" of the antibody) that do not significantly alter its biological activity. The term "binding compound" refers to both antibodies and binding fragments thereof.
"Single chain Fv" or "scFv" antibodies refer to antibody fragments that comprise the VH and VL domains of an antibody, wherein these domains are present in a single polypeptide chain. Fv polypeptides also typically comprise a polypeptide linker between the VH and VL domains that enables the scFv to form the desired structure for antigen binding. A "domain antibody" is an immunologically functional immunoglobulin fragment that contains only the variable region of a heavy chain or the variable region of a light chain. In certain instances, two or more VH regions are covalently linked to a peptide linker to form a bivalent domain antibody. The 2 VH regions of the bivalent domain antibody may target the same or different antigens. A "bivalent antibody" comprises 2 antigen binding sites. In some cases, 2 binding sites have the same antigen specificity. However, bivalent antibodies may be bispecific. "diabodies" refer to small antibody fragments having two antigen-binding sites, which fragments comprise a heavy chain variable domain (VH) linked to a light chain variable domain (VL) in the same polypeptide chain (VH-VL or VL-VH). By using a linker that is short enough not to allow pairing between two domains of the same strand, this domain is forced to pair with the complementary domain of the other strand and two antigen binding sites are created.
Preferably, in formula III, the antibody binds to R through the free amino group of the thiol group of cysteine or lysine contained in the antibody4Are connected. Thus, X may be an-S-group or an-NH-group.
Preferably, in formula III, the antibody can be any of a variety of antibodies or antigen-binding fragments thereof known in the art to have the desired biological activity. For example, the antibody or functional fragment thereof may be selected from: anti-Her 2 antibody, anti-EGFR antibody, anti-VEGFR antibody, anti-CD 20 antibody, anti-CD 33 antibody, anti-PD-L1 antibody, anti-PD-1 antibody, anti-CTLA-4 antibody, anti-TNF α antibody, anti-CD 28 antibody, anti-4-1 BB antibody, anti-OX 40 antibody, anti-GITR antibody, anti-CD 27 antibody, anti-b-CD 40 antibody, or anti-ICOS antibody, anti-CD 25 antibody, anti-CD 30 antibody, anti-CD 3 antibody, anti-CD 22 antibody, anti-CCR 4 antibody, anti-CD 38 antibody, anti-CD 52 antibody, anti-complement C5 antibody, anti-RSV F protein, anti-GD 2 antibody, anti-CD GITR antibody, anti-receptor lib/ICla II glycoprotein, anti-ICOS antibody, anti-IL 2R antibody, anti-LAG 3 antibody, anti-Integrin α 4 antibody, anti-lgE antibody, anti-PDGFRa antibody, anti-RANKL antibody, anti-SLAMF 7 antibody, anti-LTIGIT antibody, anti-TIM-3 antibody, anti-VEGFR 2 antibody, anti-VISTA antibody, anti-C-Met antibody, anti-BCMA antibody, anti-Claudin 18 antibody, anti-Nectin-4 antibody, anti-CD 79b antibody, and anti-Trop 2 antibody. In some embodiments, the antibody or antigen-binding fragment thereof is an anti-Trop 2 antibody or anti-binding fragment thereof.
In formula III, the number of moieties conjugated to the antibody (i.e., the number of m) may be from 1 to 8, and this number is generally related to the number of disulfide bonds or lysine free amino groups in the antibody, as well as to the groups involved in the conjugation reaction. The number of m can be easily determined by those skilled in the art depending on the amino acid sequence of the antibody, the group participating in the coupling reaction, and the like. In some embodiments, m is an integer from 2 to 8. In some embodiments, m is 8.
In a preferred embodiment, the compound of formula III is an antibody of formula LN401-34-3 below, wherein m is 8:
Figure BDA0003027506470000131
preparation method
The compounds of formulae I, II and III herein can be prepared by methods described in the examples herein. The corresponding preparation process is described below by way of example.
A compound of formula I
In the present invention, the compound of formula I can be obtained by starting from an appropriately substituted or protected benzyl alcohol, reacting with DMSO in the presence of acetic acid and acetic anhydride to form thioacetal, reacting with sulfuryl chloride to form an intermediate of chloroacetal, and reacting with SN38 or its analogues.
A compound of formula II
Similar to the synthesis of the compound of formula I, starting from suitably substituted or protected benzyl alcohol, reacting with DMSO in the presence of acetic acid and acetic anhydride to form thioacetal, reacting with sulfuryl chloride to form a chloroacetal intermediate, and reacting with a suitable reactive molecule containing a hydroxyl group (HO-D) or an amino group (HN-D) or a azomethine HNMe-D to obtain the compound of formula IAn analog of I. The latter is then combined with the appropriate R4-L1-L2Condensation to give the compound of formula II.
Preparation of ADC
The ADC of the present invention can be obtained by reducing an antibody, mixing the reduced antibody with the compound of formula II of the present invention, standing at room temperature for a sufficient time, and then separating and purifying. Methods for reducing antibodies are well known in the art. The purpose of the reduction includes reduction of disulfide bonds in the antibody to sulfhydryl groups. Suitable reducing agents include TCEP and the like. The reducing agent and the buffer system can be selected according to different antibody types.
Pharmaceutical composition, use and method for treatment and prevention of diseases
The present application provides a pharmaceutical composition comprising an effective amount of an ADC according to any of the embodiments of the present application and a pharmaceutically acceptable carrier. The present application also provides a method of treating or preventing a disease comprising administering to a subject in need thereof a therapeutically effective amount or a prophylactically effective amount of an ADC of the present application or a pharmaceutical composition thereof. Also provided is the use of a compound of formula I or formula II as described herein for the preparation of an ADC, and the use of an ADC as described in any of the embodiments herein for the preparation of a medicament for the treatment or prevention of a disease.
Herein, "preventing" and "prevention" includes reducing the likelihood of occurrence or worsening of a disease or disorder in a subject; the term also includes: prevention of a disease or condition occurs in a mammal, particularly when such mammal is susceptible to the disease or condition, but has not yet been diagnosed as having the disease or condition. "treatment" and other similar synonyms include the following meanings: (i) inhibiting the disease or disorder, i.e., arresting its development; (ii) alleviating the disease or condition, i.e., causing regression of the state of the disease or condition; or (iii) alleviating the symptoms caused by the disease or disorder.
Herein, "administering" refers to a method capable of delivering a compound or composition to a desired site for biological action. Methods of administration well known in the art may be used in the present invention. These methods include, but are not limited to, oral routes, via the duodenal route, parenteral injection (including intrapulmonary, intranasal, intrathecal, intravenous, subcutaneous, intraperitoneal, intramuscular, intraarterial injection or infusion), topical administration, and rectal administration. Administration techniques useful for The compounds and methods described herein are well known to those skilled in The art, for example, in Goodman and Gilman, The pharmaceutical Basis of Therapeutics, current ed.; pergamon; and Remington's, Pharmaceutical Sciences (current edition), Mack Publishing Co., Easton, Pa.
Herein, an effective amount includes both a therapeutically effective amount and a prophylactically effective amount, and refers to an amount of an ADC of the present application that, when administered to a subject, either alone or in combination with other therapeutic agents, is effective to prevent or ameliorate one or more symptoms of a disease or condition, or the development of the disease or condition. A therapeutically effective amount also refers to an amount of ADC sufficient to result in an improvement in symptoms, such as an amount that treats, cures, prevents, or ameliorates a related medical condition or increases the rate of treatment, cure, prevention, or amelioration of such a condition. The specific effective amount will depend upon a variety of factors, such as the particular disease to be treated, the physical condition of the patient, such as weight, age and sex, the duration of the treatment, the treatment being co-administered (if any), and the specific formulation employed.
A pharmaceutically acceptable carrier refers to an ingredient of a pharmaceutical formulation or composition other than the active ingredient that is not toxic to the subject. Pharmaceutically acceptable carriers include, but are not limited to, adjuvants, carriers, excipients, glidants, sweeteners, diluents, preservatives, dyes/colorants, flavoring agents, surfactants, wetting agents, dispersing agents, suspending agents, stabilizing agents, isotonicity agents, solvents, or emulsifiers. The pharmaceutical compositions of the present application can be formulated using pharmaceutically acceptable carriers well known in the art.
The pharmaceutical compositions of the present application may be formulated into various suitable dosage forms, including but not limited to tablets, capsules, injections, and the like, and may be administered by any suitable route to achieve the intended purpose. For example, it may be administered parenterally, subcutaneously, intravenously, intramuscularly, intraperitoneally, transdermally, orally, intrathecally, intracranially, intranasally or externally. The dosage of the drug may depend on the age, health and weight of the patient, concurrent treatment, and frequency of treatment, etc. The pharmaceutical compositions of the present application can be administered to any subject in need thereof, e.g., a mammal, particularly a human.
Depending on the antibody or antigen-binding fragment thereof in the ADC and the drug, the pharmaceutical composition of the present application may be used to treat or prevent the corresponding disease. For example, when the drug is an anticancer drug, the pharmaceutical composition of the present application can be used to treat cancers treatable by the anticancer drug, including but not limited to bladder, brain, breast, cervix, colon-rectum, esophagus, kidney, liver, lung, nasopharynx, pancreas, prostate, skin, stomach, uterus, ovary, testis, and blood, among others. Specifically, the cancer includes bladder cancer, kidney cancer, urinary tract cancer, chorioepithelial cancer, glioblastoma multiforme, melanoma, brain cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, uterine cancer, ovarian cancer, testicular cancer, and leukemia. Preferably, the ADCs of the present application are for use in the treatment or prevention of solid and hematological tumors, such as lung, chorioepithelial, kidney, urinary, colon, rectal, prostate, glioblastoma multiforme, ovarian, pancreatic, breast, melanoma, liver, bladder, stomach, lung or esophageal cancer.
In some embodiments, the drug molecule in the ADCs described herein is camptothecin or SN38, and the diseases are indications for camptothecin and SN38, including gastric cancer, esophageal cancer, cardiac cancer, colon cancer, rectal cancer, primary liver cancer, acute and chronic myelogenous leukemias, chorioepithelial carcinoma, lung cancer, and bladder cancer.
The invention will now be further illustrated by reference to specific examples, which are intended to be illustrative only and not to be limiting of the scope of the invention. Test methods without specific conditions noted in the following examples are generally performed according to conventional conditions or according to conditions recommended by the manufacturer. All percentages, ratios, or parts are by weight unless otherwise specified. Unless otherwise defined, all technical and scientific terms used herein have the same meaning as is familiar to those skilled in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred embodiments and materials described herein are intended to be exemplary only.
Example 1: synthesis of Compound 1
Figure BDA0003027506470000161
P-nitrobenzyl alcohol (5g, 32.15mmol) was dissolved in DMSO (100mL) and Ac was added2O (45mL), AcOH (45 mL). The reaction was carried out at room temperature for 36 hours, and HPLC showed completion of the reaction of the starting materials, thereby terminating the reaction. The reaction was poured into 300mL saturated NaHCO3In aqueous solution, EA (300mL) was extracted twice, the organic phases were combined and then saturated NaHCO3The mixture was washed twice with an aqueous solution (200mL) and once with brine (200 mL). Dried over anhydrous sodium sulfate, filtered and concentrated to give 30g of crude product. Purifying by column chromatography, discharging the product from pure petroleum ether, carrying out spin drying twice by using DCM, and carrying out oil pump drying to obtain 6g of the product, namely the compound 1.1H NMR(CDCl3):8.23(2H,d,J=8Hz);7.54(2H,d,J=8Hz);4.76(2H,s);4.74(2H,s);2.22(3H,s)。
Example 2: synthesis of Compound 2
Figure BDA0003027506470000162
Compound 1(2g, 9.38mmol) was dissolved in anhydrous DCM (20mL), N2Protecting, cooling to-70 deg.C, adding SO2Cl2(1.39g,10.32mmol) was kept at a temperature of-60 to-70 ℃ for reaction for 2 hours. HPLC detects the reaction of the raw materials is complete, and the reaction is finished. Concentrating to obtain a primary product of the compound 2, and directly using the primary product in the next reaction.
Example 3: synthesis of Compound 3
Figure BDA0003027506470000163
Mixing SN38(1.8g, 4.59mmol) was added to DMF (20mL), N2Protecting, cooling to 0-5 ℃ in ice bath. The crude compound 2 obtained in example 2 was dissolved in 10mL of DCM. A solution of Compound 2 in DCM was added to a solution of SN38 in DMF. The reaction was carried out at room temperature for 3 hours. After SN38 remained by HPLC, the reaction of Compound 2 was completed and the reaction was terminated. The reaction mixture was dissolved with EA (500mL), and then saturated NaHCO with aqueous citric acid (300mL)3Water (300mL), brine (300mL), dried over anhydrous sodium sulfate, filtered, concentrated to give crude product, which is initially purified with DCM normal phase to give crude product. The crude product was further purified by reverse phase to give 440mg of Compound 3.
Gradient: 10-90%, B% eluent: a: water; b: acetonitrile; duration: 30 min; collecting: 20-29 min.
1H NMR(DMSO-d6):8.19(2H,d,J=8Hz);8.13(1H,d,J=8Hz);7.73(1H,s);7.62(3H,d,J=8Hz);7.29(1H,s);6.49(1H,s);5.65(2H,s);5.43(2H;s);5.31(2H,s);4.95(2H,s);3.14-3.19(2H,m);1.91-2.02(1H,m);1.82-1.90(2H,m);1.24-1.31(6H,m);0.84-0.91(3H,m)。
Example 4: synthesis of Compound 4
Figure BDA0003027506470000171
A mixture of compound 3(300mg, 0.538mmol), Fe (150mg,2.69mmol), NH4Cl (201mg, 3.77mmol) was added to ethanol (30mL), water (10mL), heated to 60 ℃ and reacted for 5 hours. HPLC detects the completion of the reaction of the starting materials. Filtration, cake washing with DCM (100mL) three times, filtrate combination, concentration and addition of saturated NaHCO3(200mL) of aqueous phase, aqueous phase extracted three times with DCM (200mL), combined organic phases, dried over anhydrous sodium sulfate, filtered, and concentrated to give 270mg of Compound 4.
1H NMR(DMSO-d6):8.13(1H,d,J=8Hz);7.72(1H,s);7.73(1H,s);7.62(1H,d,J=8Hz);7.29(1H,s);6.98(2H,d,J=8Hz);6.49-6.52(3H,m);5.49(2H,s);5.43(2H;s);5.31(2H,s);5.07(2H,s);4.56(2H,s)3.14-3.16(1H,m);1.86-1.90(1H,m);1.28-1.34(4H,m);0.87-0.91(3H,m)。
Example 5: synthesis of Compound 5
Figure BDA0003027506470000181
VC1000(5g,10mmol) was dissolved in 50mL of DMF, 10mL of DEA was added, and the mixture was stirred at room temperature for 4 hours, and LCMS showed completion of the reaction. The reaction mixture was slurried with 200mL of methyl tert-butyl ether and filtered to give Compound 5(2.2g, 80% yield).
Example 6: synthesis of Compound 6
Figure BDA0003027506470000182
Compound 5(2.2g,8mmol) and MCOSu (2.4g,8mmol) was dissolved in 30mL DMF and DIEA (1.29g,10mmol) was added and the reaction stirred at room temperature for 3 hours and LCMS showed completion. The reaction mixture was slurried with 100mL of methyl tert-butyl ether and filtered to give Compound 6(3.1g, 84% yield).
Example 7: synthesis of Compound 7
Figure BDA0003027506470000183
Compound 6(46mg,0.1mmol), compound 4(52mg,0.1mmol) and HATU (57mg,0.15mmol) were dissolved in 2mL DMF, DIEA (25mg,0.2mmol) was added and the reaction stirred at room temperature for 1 h and LCMS indicated completion. The reaction mixture was slurried with 10mL of methyl tert-butyl ether, filtered to give 90mg of crude product, which was dissolved in acetonitrile and purified by medium pressure reverse phase to give compound 7(53mg, 54% yield). MS (+)977.82, the MS map of which is shown in figure 1.
Example 8: synthesis of Compound CLA-SN38 (US Patent No.9,931,417B2)
The compound CLA-SN38 was synthesized as described in US Patent No.9,931,417B2.
Figure BDA0003027506470000191
Example 9: coupling preparation of ADC 1(Trop2-CLA SN38)
Antibody Trop 210 mg (10mg/mL, 1mL, shanghai shijian biotechnology limited) was added with 67 μ L TCEP (10mM) (10.0eq), stirred at room temperature for 2 hours, buffered by displacement using a 30KDa ultrafiltration tube, and excess reducing agent was removed (3-4 times, 10 xDV). This was removed to obtain 8.8mg (12.26mg/mL, 0.7mL) of the reduced antibody, and 140. mu.L of small molecule CLA-SN38(12.0EQ,5mM, DMSO) was added thereto, and the mixture was stirred at room temperature for 30 minutes, and then the buffer was replaced with a 30kDa ultrafiltration tube to remove excess small molecules (3-4 times, 10xDV), and then the mixture was taken out to obtain ADC 1(Trop2 ADC: LN 401-34-1; m.times.8).
Figure BDA0003027506470000192
Example 10: coupled preparation of ADC 2(Trop2 ADC: LN401-34-3)
10mg of Trop2 antibody (10mg/mL, 1mL, Shimejian Biotech Co., Ltd., Shanghai) was added 67. mu.L of TCEP (10mM) (10.0eq), stirred at room temperature for 2 hours, buffered by displacement using a 30kDa ultrafiltration tube, and excess reducing agent was removed (3-4 times, 10 xDV). This was removed to give 10mg (12.26mg/mL,0.8mL) of the reduced antibody, 160. mu.l of compound 7(12.0EQ, 5mM, DMSO) was added, stirred at room temperature for 30 minutes, the buffer was replaced with a 30KDa ultrafiltration tube, the excess small molecule (3-4 times, 10xDV) was removed, and ADC 2(Trop2 ADC: LN 401-34-3; m ═ 8) was obtained,
HIC and SEC tests were performed according to the following table.
HIC:
Figure BDA0003027506470000201
SEC:
Figure BDA0003027506470000202
Figure BDA0003027506470000211
HIC and SEC profiles of LN401-34-3 are shown in FIGS. 2 and 3, respectively. LN401-34-3 has the following structural formula:
Figure BDA0003027506470000212
example 11: ADC antitumor cell activity assay
Cell viability assay
Reagent
Solutions of 10mM toxin small molecules were prepared using 100% DMSO. Samples of anti-Trop 2 Antibody Drug Conjugates (ADCs) were all provided in PBS.
Cell culture
Human BxPC-3, MDA-MB-231, MDA-MB-468 and SW620 cancer cell lines used in the activity assay were purchased from Type Culture Collection (ATCC; Manassas, VA), Culture Medium (Gibco ThermoFisher; Waltham, Mass.), fetal bovine serum (FBS; Corning, N.Y.) supplemented with 10% heat inactivation and 1 XPcillin-streptomycin (Corning), and routinely tested in RPMI-1640 (BxPC-3; Gibco ThermoFisher; Waltham, Mass.) or DMEM: f-12(MDA-MB-231, MDA-MB-468 and SW 620; Gibco), 5% CO maintained at 37 ℃2Culturing in a humidified environment.
Cell viability assay
Tumor cells were harvested by non-enzymatic separation with a Cell stripe disease Reagent (Corning), seeded into 384-well flat-bottomed white wall plates (875 cells per well in 12.5 μ L of whole medium), and left to adhere for 2-4 hours at 37 ℃. Cells were then treated with 12.5 μ L of test sample at a final concentration of 2X in duplicate, then serially diluted in complete medium, which was incubated at 37 ℃ for 120 hours. According to the protocol of the instrument manufacturer, using CellTiter-
Figure BDA0003027506470000222
2.0 cell viability assay (Promega; Uygur)Madison, sconsin), the inhibitory effect on the growth of cancer cells was determined. Luminescence was measured using a Tecan Spark multimode microplate reader (Tecan Group Ltd.; Switzerland Mandoff).
Data analysis
Data were normalized using Microsoft Excel (redmond, washington) with control treated with no test sample and analyzed using GraphPad Prism software (version 8; rajohn, ca, usa). The half maximal effective concentration for inhibition was derived from the dose response curve using non-linear regression analysis fitted to a 4-parameter logistic equation (EC 50). The therapeutic window for ADC was defined as the ratio of EC50 between antigen negative and antigen positive cell lines.
Results
The in vitro cytotoxic activity against Trop2 ADC LN401-34-1 and LN401-34-3 was assessed in a panel of human cancer cell lines with no/low (SW620 and MDA-MB-231) to high (BxPC-3 and MDA-MB-468) Trop2 expression using standard cell viability assays. As shown in FIG. 4, LN401-34-1 and LN401-34-3 reduced cell viability dose-dependently for all cell lines tested. The determined mean (SD) EC50 is summarized in table 1.
Table 1: EC50(nM) of anti-Trop 2 ADC in human tumor cells
Figure BDA0003027506470000221

Claims (14)

1. A compound of the following formula I:
Figure FDA0003027506460000011
in the formula (I), the compound is shown in the specification,
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
R2selected from H and C1-C4 alkyl;
R3is selected from-NH2-OH and-NO2
2. The compound of claim 1,
R1selected from H and C1-C4 alkyl, more preferably H;
R2is H, methyl or ethyl, more preferably ethyl;
R3is-NH2
3. The compound of claim 1, wherein the compound of formula I is a compound of formula 3 or 4 below:
Figure FDA0003027506460000012
4. a compound represented by the following formula II:
Figure FDA0003027506460000021
in the formula (I), the compound is shown in the specification,
z is-NH-, -NMe-or-O-;
d represents a drug molecule, said "-Z-D" meaning that drug molecule D is covalently linked to the remainder of the compound of formula II through the oxygen in its own hydroxyl group or the nitrogen in an amine group;
R1selected from H, halogen, nitro, amino, hydroxyl, cyano, C1-C6 alkyl, halogenated C1-C6 alkyl, C1-C6 alkoxy or halogenated C1-C6 alkoxy;
y is-NH-, -O-, or-NH-SO2-;
L1is-La-Lb-, wherein LaSelected from C1-C6 alkylene, C2-C6 alkenylene or C2-C6 alkynylene; l isbIs absent, or is-CO-, -NH-, -COO-or-NH-SO2-;
L2Is a peptide that can be recognized and hydrolyzed by enzymes in the pathological environment;
R4a group which can react with a free amino group on a thiol group or a lysine residue in a cysteine residue in a polypeptide to thereby attach the compound represented by formula II to the polypeptide.
5. The compound of claim 4,
the drug is selected from: MMAE, Duo-5, DXD, irinotecan, camptothecin, 7-ethyl-10-hydroxycamptothecin, topotecan, fluorouracil, doxifluridine, cytarabine, etoposide, fludarabine, capecitabine, vincristine, epothilone B, paclitaxel, docetaxel, daunorubicin, epirubicin, methotrexate, gemcitabine, melphalan, nimustine, mitoxantrone, doxorubicin, and mitomycin; preferably, the drug is camptothecin or 7-ethyl-10-hydroxycamptothecin; and/or
R1Selected from H and C1-C4 alkyl, preferably H; and/or
Y is-O-or-NH-; and/or
LaIs C1-C6 alkylene; l isbAbsent or as a carbonyl group; r4Through LaAnd L1Covalent linkage, L1Through LbAnd L2Covalent attachment; preferably, LbAnd L2Through amide covalent linkage; and/or
L2The peptide is dipeptide, tripeptide, tetrapeptide or pentapeptide; preferably, the amino acid residues in the peptide are selected from: one or more of valine, ornithine, lysine, serine, glycine, and citrulline; more preferably, the peptide is Val-Cit, Val-Orn, Val-Lys, Ser-Val-Lys, Gly-Val-Lys, Ser-Val-Cit, Gly-Gly-Gly-Val-Lys, or Gly-Gly-Val-Lys; and/or
The enzyme is a proteolytic enzyme, a protease, or a peptidase; preferably, the enzyme is selected from: one or more of a cysteine protease, an aspartic protease, a glutamic protease, a threonine protease, a gelatinase, a metalloprotease, and an asparagine peptide cleaving enzyme; and/or
R4Selected from the following groups:
Figure FDA0003027506460000031
wherein the wavy line represents R4And L1The location of the connection; preferably, R4Selected from:
Figure FDA0003027506460000032
6. the compound of claim 4,
L1and L2And L2And R1Substituted phenyl groups are each linked via-CO-NH-, -COO-, -O-SO2-NH-and-NH-SO2-NH-is covalently linked; preferably, L1And L2And L2And R1The substituted phenyl groups are covalently linked through-CO-NH-or-COO-respectively; preferably, Y is-O-or-NH-, Lbis-CO-; and/or
R4And L1Through a carbon-carbon single bond, -CO-NH-, -COO-, -CO-, -O-SO2-NH-and-NH-SO2-NH-is covalently linked; preferably, these radicals are R4A portion of a group.
7. The compound of any one of claims 4-6, wherein the compound of formula II is a compound of formula 7:
Figure FDA0003027506460000041
8. an antibody drug conjugate having the structure shown in formula III below:
Figure FDA0003027506460000042
in the formula (I), the compound is shown in the specification,
ab is an antibody or antigen-binding fragment thereof;
x represents the bond to R on the antibody or antigen-binding fragment thereof4A group formed after a coupling reaction;
m is an integer of 1 to 8;
R4、L1、L2、Y、Z、R1and D is as defined in any one of claims 4 to 6.
9. The antibody drug conjugate of claim 8, wherein the antibody or functional fragment thereof is selected from the group consisting of: anti-Her 2 antibody, anti-EGFR antibody, anti-VEGFR antibody, anti-CD 20 antibody, anti-CD 33 antibody, anti-PD-L1 antibody, anti-PD-1 antibody, anti-CTLA-4 antibody, anti-TNF α antibody, anti-CD 28 antibody, anti-4-1 BB antibody, anti-OX 40 antibody, anti-GITR antibody, anti-CD 27 antibody, anti-b-CD 40 antibody, or anti-ICOS antibody, anti-CD 25 antibody, anti-CD 30 antibody, anti-CD 3 antibody, anti-CD 22 antibody, anti-CCR 4 antibody, anti-CD 38 antibody, anti-CD 52 antibody, anti-complement C5 antibody, anti-RSV F protein, anti-GD 2 antibody, anti-CD GITR antibody, anti-receptor lib/ICla II glycoprotein, anti-ICOS antibody, anti-IL 2R antibody, anti-LAG 3 antibody, anti-Integrin α 4 antibody, anti-lgE antibody, anti-PDGFRa antibody, anti-RANKL antibody, anti-SLAMF 7 antibody, anti-LTIGIT antibody, anti-TIM-3 antibody, anti-VEGFR 2 antibody, anti-VISTA antibody, anti-C-Met antibody, anti-BCMA antibody, anti-Claudin 18 antibody, anti-Nectin-4 antibody, anti-CD 79b antibody, and anti-Trop 2 antibody; preferably, the antibody or antigen-binding fragment thereof is an anti-Trop 2 antibody or anti-binding fragment thereof.
10. The antibody drug conjugate of claim 8 or 9,
R4selected from:
Figure FDA0003027506460000051
L1in, with R4Linked LaIs C1-C6 alkylene, with L2Linked Lbis-CO-;
L2is Val-Cit, Val-Orn, Val-Lys, Ser-Val-Lys, Gly-Val-Lys, Ser-Val-Cit, Gly-Gly-Gly-Val-Lys or Gly-Gly-Val-Lys, preferably Val-Cit;
y is-O-or-NH-;
R1is H or C1-C4 alkyl, preferably H;
z is-NH-, -NMe or-O-, preferably-O-;
d, the drug is selected from MMAE, Duo-5, DXD, irinotecan, camptothecin, 7-ethyl-10-hydroxycamptothecin, topotecan, fluorouracil, doxifluridine, cytarabine, etoposide, fludarabine, capecitabine, vincristine, epothilone B, paclitaxel, docetaxel, daunorubicin, epirubicin, methotrexate, gemcitabine, melphalan, nimustine, mitoxantrone, doxorubicin or mitomycin, and is preferably camptothecin or 7-ethyl-10-hydroxycamptothecin; drug D is covalently linked to the remainder of the compound of formula III through Z, wherein Z is represented by oxygen in the drug's own hydroxyl group or the-NH-or-NMe group in the amine group; and
x is-S-or-NH-.
11. The antibody drug conjugate of any one of claims 8 to 10, wherein the antibody drug conjugate of formula III is:
Figure FDA0003027506460000052
12. a pharmaceutical composition comprising the antibody drug conjugate of any one of claims 8-11 and a pharmaceutically acceptable carrier.
13. An application selected from the group consisting of:
(1) use of a compound according to any one of claims 1 to 7 for the preparation of an antibody drug conjugate;
(2) use of an antibody drug conjugate according to any one of claims 8 to 11 in the manufacture of a medicament for the treatment or prevention of cancer.
14. The use of claim 13, wherein the cancer is selected from the group consisting of: bladder cancer, chorioepithelial cancer, brain cancer, breast cancer, cervical cancer, colorectal cancer, esophageal cancer, kidney cancer, liver cancer, lung cancer, nasopharyngeal cancer, pancreatic cancer, prostate cancer, skin cancer, stomach cancer, uterine cancer, ovarian cancer, testicular cancer, and blood cancer;
preferably, the antibody drug conjugate of claims 8-11, wherein said drug is camptothecin or 7-ethyl-10-hydroxycamptothecin, and said cancer is selected from the group consisting of: gastric cancer, esophageal cancer, cardiac cancer, colon cancer, rectal cancer, primary liver cancer, acute and chronic myelogenous leukemias, chorioepithelial cancer, lung cancer, and bladder cancer.
CN202110420028.0A 2021-04-19 2021-04-19 Camptothecin compound, antibody drug conjugate thereof and application thereof Pending CN113813399A (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202110420028.0A CN113813399A (en) 2021-04-19 2021-04-19 Camptothecin compound, antibody drug conjugate thereof and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202110420028.0A CN113813399A (en) 2021-04-19 2021-04-19 Camptothecin compound, antibody drug conjugate thereof and application thereof

Publications (1)

Publication Number Publication Date
CN113813399A true CN113813399A (en) 2021-12-21

Family

ID=78923772

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202110420028.0A Pending CN113813399A (en) 2021-04-19 2021-04-19 Camptothecin compound, antibody drug conjugate thereof and application thereof

Country Status (1)

Country Link
CN (1) CN113813399A (en)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019042447A1 (en) * 2017-09-04 2019-03-07 江苏恒瑞医药股份有限公司 Novel toxin and method for preparing intermediate thereof
CN109928908A (en) * 2019-03-08 2019-06-25 联宁(苏州)生物制药有限公司 A kind of Preparation Method And Their Intermediate of the drug-linker MC-MMAF for antibody drug conjugates
CN111362926A (en) * 2018-12-26 2020-07-03 联宁(苏州)生物制药有限公司 Synthetic method of intermediate CLA-SN38 for antibody coupled drug and intermediate thereof
CN111499685A (en) * 2020-03-30 2020-08-07 联宁(苏州)生物制药有限公司 Antibody coupling drug intermediate with maleimide connector and synthetic method thereof
CN111689980A (en) * 2019-05-26 2020-09-22 四川百利药业有限责任公司 Camptothecin drug and antibody conjugate thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019042447A1 (en) * 2017-09-04 2019-03-07 江苏恒瑞医药股份有限公司 Novel toxin and method for preparing intermediate thereof
CN111362926A (en) * 2018-12-26 2020-07-03 联宁(苏州)生物制药有限公司 Synthetic method of intermediate CLA-SN38 for antibody coupled drug and intermediate thereof
CN109928908A (en) * 2019-03-08 2019-06-25 联宁(苏州)生物制药有限公司 A kind of Preparation Method And Their Intermediate of the drug-linker MC-MMAF for antibody drug conjugates
CN111689980A (en) * 2019-05-26 2020-09-22 四川百利药业有限责任公司 Camptothecin drug and antibody conjugate thereof
CN111499685A (en) * 2020-03-30 2020-08-07 联宁(苏州)生物制药有限公司 Antibody coupling drug intermediate with maleimide connector and synthetic method thereof

Similar Documents

Publication Publication Date Title
CN113816969B (en) Eptification control compound, antibody drug conjugate thereof and application thereof
JP7337907B2 (en) 1H-pyrazolo[4,3-d]pyrimidine compounds as toll-like receptor 7 (TLR7) agonists and methods and uses thereof
JP2024038168A (en) Bioactive molecule conjugates, their preparation and uses
CA3082160C (en) Anti-mesothelin antibody and antibody drug conjugate thereof
CN113816990B (en) Modified amino acids and their use in ADCs
JP2022058351A (en) Anti-EGFR antibody drug conjugate
KR20160045146A (en) Pharmaceutical compositions comprising macrolide diastereomers, methods of their synthesis and therapeutic uses
CN116920115A (en) Connector for antibody drug conjugate and application thereof
JP2021513844A (en) Glypican 3 antibody and its conjugate
TWI558399B (en) Combination therapy for cancer
WO2023131219A1 (en) Conjugates, compositions and methods of use
US20230372519A1 (en) Anti-c-met antibody drug conjugates
AU2024200147A1 (en) Exatecan derivatives, linker-payloads, and conjugates and thereof
CN112237634A (en) Antibody drug conjugate, intermediate thereof, preparation method and application
CN114106088A (en) Bromomethylpyrazine-based drug conjugates and ADCs
WO2022078279A1 (en) Antibody-drug conjugate and use thereof
CN114569739A (en) Antibody drug conjugates
WO2023174401A1 (en) Anti-gpc3 antibody drug conjugate and use thereof
CN113813399A (en) Camptothecin compound, antibody drug conjugate thereof and application thereof
WO2022262789A1 (en) Antitumor compound and use thereof
CA3226938A1 (en) Anti-dll3 antibody and preparation method therefor, drug conjugate and application thereof
WO2022014698A1 (en) Method for producing antibody-drug conjugate
JP2023550532A (en) Antibody-drug conjugate targeting TROP2, its production method and use
WO2024012566A2 (en) Antibody, linkers, payload, conjugates and applications thereof
RU2800137C1 (en) Antibody-drug conjugate, intermediate for its production, method of its production and its use

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination