WO2023103854A1 - 一种亲和力改善的抗体-药物偶联物、其制备方法及应用 - Google Patents

一种亲和力改善的抗体-药物偶联物、其制备方法及应用 Download PDF

Info

Publication number
WO2023103854A1
WO2023103854A1 PCT/CN2022/135429 CN2022135429W WO2023103854A1 WO 2023103854 A1 WO2023103854 A1 WO 2023103854A1 CN 2022135429 W CN2022135429 W CN 2022135429W WO 2023103854 A1 WO2023103854 A1 WO 2023103854A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
antibody
pharmaceutically acceptable
stereoisomer
solvate
Prior art date
Application number
PCT/CN2022/135429
Other languages
English (en)
French (fr)
Inventor
丁会
柯天一
于海勇
许云雷
劳芳
刘岩
张西东
荣鹏飞
Original Assignee
昆山新蕴达生物科技有限公司
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 昆山新蕴达生物科技有限公司 filed Critical 昆山新蕴达生物科技有限公司
Publication of WO2023103854A1 publication Critical patent/WO2023103854A1/zh

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
    • A61K47/68Medicinal 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
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/18Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with hetero atoms or with carbon atoms having three bonds to hetero atoms with at the most one bond to halogen, e.g. ester or nitrile radicals, directly attached to ring carbon atoms
    • C07D307/20Oxygen atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K16/00Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies
    • C07K16/18Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans
    • C07K16/28Immunoglobulins [IGs], e.g. monoclonal or polyclonal antibodies against material from animals or humans against receptors, cell surface antigens or cell surface determinants

Definitions

  • the present disclosure relates to the field of medicine, in particular to antibody drug conjugates (ADCs) that bind human tumor antigen targets and/or provide anti-tubulin drug activity.
  • ADCs antibody drug conjugates
  • the present disclosure further relates to methods and compositions useful for the treatment and diagnosis of cancers that express tumor antigen targets and/or are treatable by disrupting tubulin.
  • MORAb-202 is Eisai's first ADC, and it is the first to combine Eisai's in-house developed anti-cancer agent eribulin (eribulin, or eribulin) with targeting folate receptor alpha (FR ⁇ )
  • eribulin eribulin, or eribulin
  • FR ⁇ folate receptor alpha
  • ADC conjugated with IgG1 monoclonal antibody is shown below.
  • An enzymatically cleavable linker (linker) is used between the antibody and the toxin, and the drug-antibody coupling ratio (DAR) is 4.0.
  • MORAb-202's payload eribulin (product name: Halaven, generic name: eribulin or eribulin) and paclitaxel belong to microtubule inhibitors, but it is the first soft spongin-like microtubule dynamics inhibitor agent with a novel mechanism of action.
  • eribulin is a simplified and synthetic version of halichondrin B (halichondrin B).
  • Halichondrin B is a natural product isolated from the sponge Halichondria okadai (Sponge okada). Eribulin is thought to prevent cell division by inhibiting the growth phase of microtubule dynamics.
  • Eribulin In cancer cells, this mechanism can lead to a prolonged and irreversible mitotic arrest, eventually leading to cell death. Eribulin was launched in China in January 2020 for the treatment of patients with locally recurrent or metastatic breast cancer who have previously received at least 2 chemotherapy regimens (including anthracyclines and taxanes), so Eribulin can effectively solve the problem caused by anthracene The problem of resistance to cyclic and taxane microtubule inhibitor therapy.
  • the linker of MORAb-202 adopts PEG2-val-cit-PAB structure, and the function of adding p-aminobenzyloxycarbonyl (PAB) group is to provide enough space for enzyme cleavage, so that ADC can be self-cleaved and released in the cell Eriblin small molecule.
  • the PEG2 structure provides better hydrophilicity for ADC molecules.
  • the linker As a bridge for ADC drugs, the linker is connected to the antibody through a cleavable or non-cleavable linker.
  • the linker needs to beakily designed, which not only needs to have stability and specific cleavage in tumor cells, but also has the convenience of drug making, such as Good hydrophilicity, suitable DAR value, and maintain good affinity of antibody. Therefore, there is still room and demand for optimization of ADC linkers.
  • the inventors of the present disclosure creatively found that the molecular structure of eribulin has a primary amine, and the primary amine structure is far from the main structure of eribulin, and there is enough space. Based on this, the inventors designed a PAB-free linker to directly connect the primary amine of eribulin with an amino acid or a short peptide (such as a Vc or Va structure) to form an amide bond; in addition, the inventors of the present disclosure also creatively found that , Using a short chain instead of a long chain to link with amino acids or short peptides (such as Vc or Va) after the maleimide group can increase the hydrophilicity of the ADC.
  • the present disclosure provides a compound represented by formula (I), its pharmaceutically acceptable salt, its stereoisomer, or its solvate,
  • X 1 , X 2 , X 3 , and X 4 are each independently selected from CH 2 , NH, O, and S, and at least one of X 1 , X 2 , X 3 , and X 4 is CH 2 ;
  • n 1 , n 2 , n 3 , n 4 are each independently selected from 0, 1, and at least one of n 1 , n 2 , n 3 and n 4 is 1;
  • L2 is selected from amino acid residues or short peptides consisting of 2-10 amino acid residues; preferably, the amino terminal of L2 is connected to L1 , and the carbonyl terminal of L2 is connected to D;
  • D is a drug linked to L2 through a chemical bond, and the drug is preferably eribulin or its derivatives.
  • Y is
  • X 1 , X 2 , X 3 , X 4 are all CH 2 .
  • n 1 is 0, n 2 , n 3 and n 4 are each independently selected from 0 and 1, and at least one of n 2 , n 3 and n 4 is 1.
  • n 1 , n 2 are 1, and n 3 , n 4 are 0.
  • n is selected from 1, 2, 3, 4.
  • n is selected from 1, 2, 3.
  • n is 2.
  • L is a short peptide consisting of 2-4 amino acid residues, preferably, the amino acid is selected from glycine, phenylalanine, valine, citrulline, alanine, and more Preferably, the amino acid is selected from valine, citrulline, alanine.
  • the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • L is selected from glycine-glycine-phenylalanine-glycine (GGFG), valine-citrulline (VC), valine-alanine (VA), preferably from valine Acid-Citrulline (VC), Valine-Alanine (VA).
  • GGFG glycine-glycine-phenylalanine-glycine
  • VC valine-citrulline
  • VA valine-alanine
  • VA valine Acid-Citrulline
  • VA Valine-Alanine
  • the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • L is selected from preferably selected from In some embodiments, the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • D is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • the linker is stable extracellularly such that the ADC remains intact when present in the extracellular environment, but is capable of cleavage when internalized in a cell, eg, a cancer cell.
  • the linker comprises a cleavable moiety such that said linker or said antibody portion remains bound to eribulin following cleavage.
  • the cleavable moiety in the linker is a cleavable peptide moiety.
  • ADCs comprising a cleavable peptide moiety exhibit lower levels of aggregation, improved antibody:drug ratios, increased targeted killing of cancer cells, reduced non- Off-target killing of cancer cells, and/or higher drug loading (p).
  • addition of a cleavable moiety increases cytotoxicity and/or potency relative to a non-cleavable linker.
  • the increased potency and/or cytotoxicity is in cancers expressing intermediate levels of an antigen targeted by the antibody portion of the ADC (eg, intermediate FRA expression).
  • the cleavable peptide moiety is capable of being cleaved by an enzyme
  • the linker is one that is cleavable by an enzyme.
  • the enzyme is a cathepsin and the linker is a linker that the cathepsin is capable of cleaving.
  • an enzymatically cleavable linker eg, a cathepsin-cleavable linker
  • the cleavable peptide moiety in the linker comprises amino acid units.
  • the amino acid unit comprises valine-citrulline (Val-Cit) or valine-alanine (Val-Ala).
  • ADCs comprising Val-Cit or Val-Ala exhibit increased stability, decreased off-target cell killing, increased on-target cell killing relative to ADCs comprising alternative amino acid units or alternative cleavable moieties , lower aggregation levels, and/or higher drug loading.
  • the linker comprises at least one spacer unit that joins the antibody moiety to the cleavable moiety.
  • the spacer unit in the linker may comprise at least one maleimidocaproyl (MC) moiety or a truncated fragment thereof, preferably a MC truncated fragment.
  • MC maleimidocaproyl
  • ADCs comprising shorter spacer units (e.g., truncated MC) relative to ADCs comprising longer spacer units (e.g., MC or (PEG) 2 - 8 ) despite shorter linker lengths Show lower aggregation level and stability and/or better efficacy.
  • the spacer unit of the linker in the antibody conjugates of the present disclosure does not comprise a spacer unit p-aminobenzyloxycarbonyl (PAB) attached to D.
  • PAB p-aminobenzyloxycarbonyl
  • the spacer unit in the linker is attached to the antibody portion of the ADC via a maleimide moiety (Mal).
  • the indicated L 1 -L 2 -D compounds are selected from:
  • the present disclosure provides an antibody-drug conjugate represented by formula (II), a pharmaceutically acceptable salt thereof, a stereoisomer thereof, or a solvate thereof,
  • Ab is an antibody or an antigen-binding fragment thereof
  • X 1 , X 2 , X 3 , and X 4 are each independently selected from CH 2 , NH, O, and S, and at least one of X 1 , X 2 , X 3 , and X 4 is CH 2 ;
  • n 1 , n 2 , n 3 , n 4 are each independently selected from 0, 1, and at least one of n 1 , n 2 , n 3 and n 4 is 1;
  • L2 is selected from amino acid residues or short peptides consisting of 2-10 amino acid residues; preferably, the amino terminal of L2 is connected to L1 , and the carbonyl terminal of L2 is connected to D;
  • D is a drug linked to L2 through a chemical bond, and the drug is preferably Eribulin or its derivatives;
  • p is any value between 1-20.
  • Y is
  • X 1 , X 2 , X 3 , X 4 are all CH 2 .
  • n 1 is 0, n 2 , n 3 and n 4 are each independently selected from 0 and 1, and at least one of n 2 , n 3 and n 4 is 1.
  • n 1 , n 2 are 1, and n 3 , n 4 are 0.
  • n is selected from 1, 2, 3, 4.
  • n is selected from 1, 2, 3.
  • n is 2.
  • L is a short peptide consisting of 2-4 amino acid residues, preferably, the amino acid is selected from glycine, phenylalanine, valine, citrulline, alanine, and more Preferably, the amino acid is selected from valine, citrulline, alanine.
  • the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • L is selected from glycine-glycine-phenylalanine-glycine (GGFG), valine-citrulline (VC), valine-alanine (VA), preferably from valine Acid-Citrulline (VC), Valine-Alanine (VA).
  • GGFG glycine-glycine-phenylalanine-glycine
  • VC valine-citrulline
  • VA valine-alanine
  • VA valine Acid-Citrulline
  • VA Valine-Alanine
  • the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • L is selected from preferably selected from In some embodiments, the amino terminus of L2 is linked to L1 and the carbonyl terminus of L2 is linked to D.
  • D is N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-(2-aminoethyl)-2-aminoethyl-N-phenyl
  • antibody-drug conjugate represented by the above formula (II) refers to a composition of ADC molecules with the same or different numbers of linked drug molecules.
  • compositions comprising a plurality of ADC molecules.
  • each ADC in the compositions described herein comprises the same number of one or more drug molecules.
  • each ADC in the compositions described herein comprises a different number of one or more drug molecules.
  • each antibody can be coupled with 1, 2, 3, 4, 5, 6, 7, 8 or more drug molecules, such as 1 to 5 (e.g. 1, 2, 3, 4 or 5) drug molecules, 5 to 8 (e.g. 5, 6, 7 or 8) drug molecules or 8 to 12 (e.g. 8, 9, 10, 11 or 12) drug molecules.
  • 1 to 5 e.g. 1, 2, 3, 4 or 5
  • 5 to 8 e.g. 5, 6, 7 or 8
  • 8 to 12 e.g. 8, 9, 10, 11 or 12
  • the Drug Antibody Ratio refers to the number of drug molecules conjugated to the antibody (eg, p in Formula II).
  • the number of drug molecules contained in the ADC described herein is generally an integer, and when the number of drug molecules contained in the ADC described herein (for example, p in formula II) is a fraction, the fraction refers to the number of drug molecules contained in the ADC described herein.
  • p is any value between 1-10.
  • p is any value between 3-5.
  • p is any value between 3.5-4.5, eg, p is 3.5, 3.9 or 4.1.
  • the antibody drug conjugate is selected from the following:
  • -S- is not an additional external sulfur atom, but the sulfhydryl group contained in the Ab itself after the disulfide bond is opened. -S- formed after making a ligation.
  • the antibody in the antibody-drug conjugate (ADC) of the present disclosure is selected from murine antibodies, chimeric antibodies, humanized antibodies, and fully human antibodies.
  • Suitable antibodies can bind any disease-associated antigen known in the art.
  • the disease state is cancer
  • a number of antigens expressed by or otherwise associated with tumor cells are known in the art, including but not limited to carbonic anhydrase IX, alpha-fetoprotein , AFP), ⁇ -actinin-4 ( ⁇ -actinin-4), A3, antigen specific to A33 antibody, ART-4, B7, Ba 733, BAGE, BrE3 antigen, CA125, CAMEL, CAP -1, CASP-8/m, CCL19, CCL21, CD1, CD1a, CD2, CD3, CD4, CD5, CD8, CD11A, CD14, CD15, CD16, CD18, CD19, CD20, CD21, CD22, CD23, CD25, CD29 , CD30, CD32b, CD33, CD37, CD38, CD40, CD40L, CD44, CD45, CD46, CD52, CD54, CD55, CD59, CD64, CD66a-e, CD67
  • the antibody binds her2, her3, claudin 18.2, ROR-1, dll-3, mucl, muc-17, EGP-1 (Trop-2).
  • Exemplary antibodies that may be utilized include, but are not limited to, hR1 (anti-IGF-1R, U.S. Patent No. 13/688,812), hPAM4 (anti-Mucin, U.S. Patent No. 7,282,567), hA20 (anti-CD20, U.S. Patent No. 7,151,164), hA19 ( Anti-CD19, U.S. Patent No. 7,109,304), hIMMU31 (anti-AFP, U.S. Patent No. 7,300,655), hLL1 (anti-CD74, U.S. Patent No. 7,312,318), hLL2 (anti-CD22, U.S. Patent No.
  • the antibody is IMMU-31 (anti-AFP), hRS7 (anti-Trop-2), hMN-14 (anti-CEACAM5), hMN-3 (anti-CEACAM6), hMN-15 (anti-CEACAM6), hLL1 (anti-CD74 ), hLL2 (anti-CD22), hL243 or IMMU-114 (anti-HLA-DR), hA19 (anti-CD19) or hA20 (anti-CD20).
  • the terms epratuzumab and hLL2 are interchangeable, as are the terms veltuzumab and hA20, hL243g4P, hL243 ⁇ 4P, and IMMU-114.
  • the antibody is anti-Trop-2 antibody such as hRS7; anti-ROR1 antibody such as 99961 (Chinese Patent No. 104662044); anti-HER-3 antibody such as patritumab (Chinese Patent No. 102174105); anti-HER2 antibody Herceptin (Trastuzumab), Pertuzumab.
  • Suitable alternative antibodies include, but are not limited to, abciximab (anti-glycoprotein IIb/IIIa), alemtuzumab (anti-CD52), bevacizumab (anti-VEGF), Western Cetuximab (anti-EGFR), gemtuzumab (anti-CD33), ibritumomab (anti-CD20), panitumumab (anti-EGFR), Rituximab (anti-CD20), tositumomab (anti-CD20), trastuzumab (anti-ErbB2), lambrolizumab (anti-PD1 receptor ), atezolizumab (anti-PD-L1), MEDI4736 (anti-PD-L1), nivolumab (anti-PD-1 receptor), ipilimumab (anti-PD-1 CTLA-4), abagovomab (anti-CA-125), adecatumumab (anti-EpCAM), atlizumab (anti
  • Patent 5,831,034 U.S. Patent 5,911,989, and Vcelar et al., AIDS 2007; 21(16):2161-2170 and Joos et al., Antimicrob. Agents Chemother. 2006; 50(5):1773-9, all by reference Incorporated into this article.
  • the drug moiety conjugated to the subject antibody is selected from eribulin or a derivative thereof, eg, the mesylate salt of eribulin.
  • eribulin refers to a synthetic analog of halichondrin B, a macrocyclic compound originally isolated from the sponge Halichondria okadais. Eribulin is a microtubule inhibitor that is thought to bind tubulin and cause cell cycle arrest in G2/M phase by inhibiting mitotic spindle assembly.
  • eribulin mesylate refers to the mesylate salt of eribulin, which is sold under the tradename Halaven TM .
  • the present disclosure provides a method of preparing the compound, pharmaceutically acceptable salt, stereoisomer or solvate of the first aspect.
  • the method includes: subjecting L 1 -L 2 -OH to amidation reaction with eribulin to obtain the compound represented by formula (I), its pharmaceutically acceptable salt, its stereoisomeric Construct, or a solvate thereof, wherein, L 1 and L 2 are as defined in the first aspect of the present disclosure.
  • the method comprises:
  • Step 1 Combine maleimide active ester with Dissolved with alkali in NMP, and stirred at room temperature for 4 hours, HPLC showed that a new peak was generated, and the target product was obtained after Pre-HPLC purification, and LCMS showed that it was the target product.
  • Step 2 Put Add eruibulin, HATU, and alkali to NMP, and stir at room temperature for 2 hours. HPLC shows that a new peak is formed. After Pre-HPLC purification, freeze-dry to obtain the target product, which is confirmed as the target product by LCMS.
  • the present disclosure provides a method for preparing the antibody-drug conjugate, pharmaceutically acceptable salt, stereoisomer or solvate described in the second aspect.
  • the method comprises: reacting the compound described in the aforementioned first aspect, its pharmaceutically acceptable salt, its stereoisomer, or its solvate with the aforementioned Ab to obtain the formula (II ), the antibody drug conjugate represented by ), its pharmaceutically acceptable salt, its stereoisomer, or its solvate.
  • the compound described in the first aspect is prepared by the method described in the aforementioned third aspect.
  • operations such as concentration, buffer exchange, and purification can be performed using the following common operations.
  • centrifuge to perform centrifugation (for example, centrifuge at 2000G-3800G for 5-20 minutes) to concentrate the antibody or antibody-drug conjugate solution.
  • Antibody concentration was measured using a UV analyzer according to the method specified by the manufacturer.
  • phosphate buffer e.g. PBS
  • sodium chloride e.g., 137 mM
  • EDTA ethylenediaminetetraacetic acid
  • a PD-10 column using Sephadex G-25 carrier was equilibrated.
  • 1 mL of an antibody aqueous solution was loaded, and then a fraction (3.5 mL) eluted with PBS/EDTA 2 mL was separated. This fraction was concentrated by common procedure A, and the antibody concentration was measured by common procedure B, and then the antibody concentration was adjusted to 10 mg/mL using PBS/EDTA.
  • Phosphate buffer eg, 50 mM, pH 6.5, also referred to as PBS6.5/EDTA in this specification
  • sodium chloride eg, 50 mM
  • EDTA eg, 2 mM
  • a PD-10 column using Sephadex G-25 carrier was equilibrated.
  • 1 mL of antibody aqueous solution was loaded, and then the fraction (3.5 mL) eluted with PBS6.5/EDTA 2 mL was separated and obtained. This fraction was concentrated by common procedure A, and the antibody concentration was measured by common procedure B, and then the antibody concentration was adjusted to 5 mg/mL using PBS6.5/EDTA.
  • phosphate buffer eg, PBS7.4
  • sodium phosphate buffer eg, 10 mM, pH6.5; also referred to as PBS6.5 in this specification
  • Acetic acid buffer e.g., 10 mM, pH 5.5; also referred to as ABS in this specification
  • sorbitol e.g., 5%
  • An antibody-drug conjugate reaction aqueous solution (for example, about 1 mL) was packed in this column, and the antibody fraction was separated and obtained by eluting with an amount of buffer solution specified by the manufacturer.
  • Drug linkers, antibody-drug conjugates of low molecular weight compounds tris(2-carboxyethyl)phosphine hydrochloride (TCEP), cysteine, dimethyl sulfoxide, etc.
  • Phosphate buffer eg, PBS7.4
  • sodium phosphate buffer eg, 10 mM, pH6.5; also referred to as PBS6.5 in this specification
  • sodium chloride eg, 137 mM
  • sorbitol-containing for example, 5%
  • acetate buffer solution for example, 10mM, pH5.5; also referred to as ABS in this specification
  • MES 25mM pH6.5 or any buffer solution in His10mM pH 5.5 AKTA column (filler : sephadex G 25) balance.
  • the injector is loaded with an antibody-drug conjugate reaction aqueous solution (for example, about 2 mL), and eluted with an amount of buffer solution specified by the manufacturer, thereby separating and obtaining antibody fractions.
  • an antibody-drug conjugate in which unlinked drug linkers and low-molecular compounds (tris(2-carboxyethyl)phosphine hydrochloride (TCEP), cysteine, dimethyl sulfoxide, etc.) were removed was obtained. United things.
  • the present disclosure provides a pharmaceutical composition
  • a pharmaceutical composition comprising the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the compound described in the second aspect
  • the pharmaceutical composition can be made into any pharmaceutically acceptable dosage form.
  • the pharmaceutical composition may also be administered to an individual in need of such treatment by any suitable means of administration, such as oral, parenteral, rectal or pulmonary administration.
  • the pharmaceutical composition can be made into conventional solid preparations, such as tablets, capsules, pills, granules, etc.; it can also be made into oral liquid preparations, such as oral solutions, oral suspensions , syrup, etc.
  • suitable fillers, binders, disintegrants, lubricants and the like can be added.
  • parenteral administration the pharmaceutical composition can be made into injections, including injections, sterile powders for injections and concentrated solutions for injections.
  • the pharmaceutical composition When making injections, it can be produced by conventional methods in the existing pharmaceutical field. When preparing injections, no additives can be added, and suitable additives can also be added according to the properties of the medicine.
  • the pharmaceutical composition For rectal administration, the pharmaceutical composition can be made into suppositories and the like.
  • the pharmaceutical composition When used for pulmonary administration, can be made into inhalants or sprays and the like.
  • the present disclosure provides the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the antibody drug conjugate described in the second aspect, Use of a pharmaceutically acceptable salt, stereoisomer, or solvate, or the pharmaceutical composition of the fifth aspect in the preparation of a medicament for treating and/or preventing a disease related to abnormal cell activity (such as cancer disease).
  • the present disclosure provides the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the antibody drug conjugate described in the second aspect, A pharmaceutically acceptable salt, stereoisomer, or solvate, or the pharmaceutical composition of the fifth aspect, which is used for treating and/or preventing a disease related to abnormal cell activity (such as cancer disease).
  • a disease related to abnormal cell activity such as cancer disease
  • the present disclosure provides a method for treating or preventing a disease (such as a cancer disease) related to abnormal cell activity, comprising administering an effective dose to an individual in need thereof.
  • a disease such as a cancer disease
  • the present disclosure provides the method described in the first aspect.
  • Dosage regimens may be adjusted to provide the optimum desired response. For example, a single bolus may be administered, several divided doses may be administered over time or the dose may be proportionally reduced or increased as indicated by the exigencies of the therapeutic situation.
  • Dosage values may vary with the type and severity of the condition to be alleviated and may comprise single or multiple doses. It is further understood that for any given individual, the particular dosing regimen may be adjusted over time according to the needs of the individual and the professional judgment of the person administering the composition or supervising the administration of the composition.
  • Various embodiments may relate to the use of the subject methods and compositions to treat cancer, including carcinoma in situ or metastases. Therefore, with respect to the sixth aspect, the seventh aspect, and the eighth aspect above, in some embodiments, the cancer is carcinoma in situ or metastatic carcinoma.
  • the cancer includes, but is not limited to, metastatic breast cancer, non-small cell lung cancer, Burkitt lymphoma, Hodgkin's lymphoma, acute myelogenous leukemia, chronic myelogenous leukemia , acute lymphoblastic leukemia, chronic lymphocytes, skin cancer, oral cancer, esophageal cancer, gastrointestinal cancer, lung cancer, lung cancer, stomach cancer, colon cancer, rectal cancer, triple negative breast cancer (TNBC), ovarian cancer, Prostate Cancer, Uterine Cancer, Endometrial Cancer, Cervical Cancer, Bladder Cancer, Pancreatic Cancer, Bone Cancer, Brain Cancer, Connective Tissue Cancer, Thyroid Cancer, Liver Cancer, Gallbladder Cancer, Bladder (Urothelial) Cancer, Kidney Cancer, Skin Cancer cancer, central nervous system cancer, testicular cancer, epithelial cancer, head and neck cancer.
  • TNBC triple negative breast cancer
  • the cancer is selected from metastatic breast cancer, TNBC, non-TNBC, cervical cancer, endometrial cancer, lung cancer, ovarian cancer, bladder (urothelial) cancer, colon cancer, pancreatic cancer.
  • the cancer is TNBC, ovarian cancer, cervical cancer, bladder (urothelial) cancer, or pancreatic cancer (eg, pancreatic carcinoma in situ).
  • the antibody or immunoconjugate can be combined with surgery, radiation therapy, chemotherapy, immunotherapy with naked antibodies including checkpoint inhibitory antibodies, radioimmunotherapy, immunomodulators, vaccines and other combinations.
  • the antibody or immunoconjugate is used in combination with a PARP inhibitor, microtubule inhibitor, Bruton kinase inhibitor and/or PI3K inhibitor.
  • combination therapies may allow lower doses of each therapeutic agent to be administered in the context of the combination, thus reducing certain serious side effects and potentially reducing the required course of treatment.
  • the full doses of each can also be administered in the absence or presence of minimal overlapping toxicity.
  • the antibody or immunoconjugate can be administered as a periodic bolus injection, in alternative embodiments, the antibody or immunoconjugate can be administered by continuous infusion.
  • continuous infusion can be administered, eg, through an indwelling catheter.
  • indwelling catheter Such devices are known in the art, such as or catheter (see, eg, Skolnik et al., The Drug Monit 32:741-48, 2010), and any such known indwelling catheter can be used.
  • a variety of continuous infusion pumps are also known in the art, and any such known infusion pump can be used. Dosages for continuous infusion may range between 0.1 and 3.0 mg/kg per day. More preferably, these immunoconjugates are administered by intravenous infusion over a relatively brief period of 2 to 5 hours, more preferably 2-3 hours.
  • the antibody or immunoconjugate and schedule of administration may be effective in patients resistant to standard therapy.
  • hRS7-eribulin immunoconjugates can be administered to patients who have not responded to prior therapy with eribulin.
  • irinotecan-resistant patients can show partial or even complete responses to hRS7-eribulin.
  • the ability of immunoconjugates to specifically target tumor tissue can overcome tumor resistance due to improved targeting and enhanced delivery of therapeutic agents.
  • a particularly preferred subject may be suffering from Trop-2 positive breast cancer, ovarian cancer, cervical cancer, endometrial cancer, lung cancer, prostate cancer, colon cancer, rectal cancer, gastric cancer, esophageal cancer, bladder (urothelial) cancer , kidney cancer, pancreatic cancer, brain cancer, thyroid cancer, epithelial cancer, or head and neck cancer.
  • the cancer is metastatic cancer. More preferably, the patient has previously failed treatment with at least one standard anticancer therapy.
  • the cancer is metastatic breast cancer, TNBC, non-TNBC, endometrial cancer, lung cancer, ovarian cancer or colon cancer.
  • the present disclosure provides a pharmaceutical preparation comprising the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the compound described in the second aspect Conjugates, pharmaceutically acceptable salts, stereoisomers, or solvates, or the pharmaceutical composition of the fifth aspect.
  • the present disclosure provides the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the conjugate described in the second aspect, pharmaceutically Acceptable salts, stereoisomers, or solvates, or uses of the pharmaceutical composition of the fifth aspect for the preparation of pharmaceutical preparations.
  • the present disclosure provides a kit comprising the compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in the first aspect, or the compound described in the second aspect
  • the aforementioned compounds, antibody drug conjugates, pharmaceutically acceptable salts, stereoisomers, or solvates, pharmaceutical preparations or kits provided in this disclosure can be used to inhibit the growth, proliferation or migration of cancer cells.
  • the compounds, antibody drug conjugates, pharmaceutically acceptable salts, stereoisomers, or solvates, pharmaceutical formulations or kits are for in vivo or in vitro administration; e.g., administered into a subject, or administered To cells in vitro (eg cell lines or cells from an individual such as cancer cells).
  • the aforementioned compounds, antibody drug conjugates, pharmaceutically acceptable salts, stereoisomers, or solvates, pharmaceutical preparations or kits are used for non-therapeutic purposes (such as in vitro scientific research, etc. ), for inhibiting the growth, proliferation or migration of cancer cells.
  • the present disclosure provides the compound described in the first aspect, its pharmaceutically acceptable salt, its stereoisomer, or a solvate thereof, or the compound described in the second aspect
  • the antibody drug conjugate, its pharmaceutically acceptable salt, its stereoisomer, or its solvate, or the pharmaceutical composition described in the fifth aspect, or the pharmaceutical preparation of the ninth aspect is used for inhibiting Use in reagents for growth, proliferation or migration of cancer cells, especially in the preparation of reagents for inhibiting growth, proliferation or migration of cancer cells in vitro.
  • the cancer in the cancer cells is cancer, and in some embodiments, the cancer is as described above. In some embodiments, the use is for non-therapeutic purposes.
  • the present disclosure provides the compound described in the first aspect, its pharmaceutically acceptable salt, its stereoisomer, or its solvate, or the antibody drug described in the second aspect
  • the conjugate, its pharmaceutically acceptable salt, its stereoisomer, or its solvate, or the pharmaceutical composition of the fifth aspect, or the pharmaceutical preparation of the ninth aspect which is used to inhibit the growth of cancer cells , proliferation or migration, especially for inhibiting cancer cell growth, proliferation or migration in vitro.
  • the cancer in the cancer cells is cancer, and in some embodiments, the cancer is as described above. In some embodiments, the use is for non-therapeutic purposes.
  • the present disclosure provides a method for inhibiting cancer cell growth, proliferation or migration (especially inhibiting cancer cell growth, proliferation or migration in vitro), which comprises administering an effective amount of a first The compound, pharmaceutically acceptable salt, stereoisomer, or solvate described in one aspect, or the antibody drug conjugate, pharmaceutically acceptable salt, stereoisomer, or solvent described in the second aspect compound, or the pharmaceutical composition of the fifth aspect, or the pharmaceutical preparation of the ninth aspect.
  • the cancer in the cancer cells is cancer, and in some embodiments, the cancer is as described above. In some embodiments, the methods are used for non-therapeutic purposes.
  • the cancer cells are cancer cells in situ or metastatic cancer cells.
  • the cancer cells are selected from metastatic breast cancer, non-small cell lung cancer, Burkitt lymphoma, Hodgkin's lymphoma, acute myelogenous leukemia, chronic myelogenous leukemia, Acute lymphocytic leukemia, chronic lymphocytic leukemia, skin cancer, oral cancer, esophagus cancer, gastrointestinal tract cancer, lung cancer, lung cancer, stomach cancer, colon cancer, rectal cancer, triple-negative breast cancer (TNBC), ovarian cancer, prostate cancer Cancer, Uterine Cancer, Endometrial Cancer, Cervical Cancer, Bladder Cancer, Pancreatic Cancer, Bone Cancer, Brain Cancer, Connective Tissue Cancer, Thyroid Cancer, Liver Cancer, Gallbladder Cancer, Bladder (Urothelial) Cancer, Kidney Cancer, Skin Cancer
  • TNBC triple-negative breast cancer
  • the cancer cells are selected from metastatic breast cancer, TNBC, non-TNBC, cervical cancer, endometrial cancer, lung cancer, ovarian cancer, bladder (urothelial) cancer, colon cancer, pancreatic cancer of cancer cells.
  • the cancer cell is TNBC, ovarian cancer, cervical cancer, bladder (urothelial) cancer, or pancreatic cancer cell (eg, pancreatic cancer cell in situ).
  • Y is n 1 , n 2 , n 3 and n 4 are each independently selected from 0 and 1, and at least one of n 1 , n 2 , n 3 and n 4 is 1.
  • Y is
  • Y is
  • Y is
  • n 1 and n 2 are 1 and n 3 and n 4 are 0, Y is
  • Other similar definitions can be understood with reference to the foregoing.
  • amino acid residue refers to the incomplete amino acid structure remaining after the amino group loses a hydrogen and the carboxyl group loses a hydroxyl group, having an amino terminal and a carbonyl terminal.
  • L is an amino acid residue or a short peptide consisting of 2-10 (preferably 2-4) amino acid residues, wherein the types of 2-10 (preferably 2-4) amino acids are mutually Can be the same or different.
  • L2 is a short peptide consisting of 4 amino acid residues, and the amino acid is selected from glycine, phenylalanine, then L2 can be the following short peptide or peptide residues: glycine-glycine-phenylalanine- Glycine (Gly-Gly-Phe-Gly), specifically can be
  • linker refers to a chemical structural fragment or bond that is connected to an antibody at one end and a drug (drug compound) at the other end, and other linkers can also be connected It is then linked to a drug compound.
  • linker structure of the present invention can be synthesized by methods known in the art, and can also be synthesized using the methods described in the present invention.
  • the "antibody-drug conjugate" in the present invention refers to a targeting moiety such as an antibody or an antigen-binding fragment thereof linked to a biologically active drug through a stable linker unit.
  • pharmaceutically acceptable salt refers to a relatively non-toxic acid addition salt or base addition salt of the compound or conjugate of the present invention.
  • the acid addition salt is a salt formed between the compound or conjugate of the present invention and a suitable inorganic acid or organic acid, and these salts can be prepared by making the compound or conjugate of the present invention and a suitable organic acid or inorganic acid prepared by reacting in a solvent.
  • Representative acid addition salts include hydrobromide, hydrochloride, sulfate, bisulfate, sulfite, acetate, oxalate, valerate, oleate, palmitate, stearate Salt, Luurosilicate, Borate, Benzoate, Lactate, Nitrate, Phosphate, Phosphate, Carbonate, Bicarbonate, Toluate, Citrate, Maleic Acid Salt, fumarate, succinate, malate, ascorbate, tannate, pamoate, alginate, naphthalenesulfonate, tartrate, benzoate, methanesulfonate, p-toluene Sulfonate, Gluconate, Lactobionate and Lauryl Sulfonate etc.
  • the base addition salt is a salt formed between the compound or conjugate of the present invention and a suitable inorganic base or organic base, and these salts can be obtained by making the compound or conjugate of the present invention and a suitable inorganic base or organic base prepared by reacting in a solvent.
  • Representative base addition salts include, for example, salts with alkali metal, alkaline earth metal, quaternary ammonium cations, such as sodium, lithium, potassium, calcium, magnesium, tetramethylquaternary ammonium, tetraethylquaternary ammonium Salts, etc.; amine salts, including salts formed with ammonia (NH3), primary amines, secondary amines or tertiary amines, such as methylamine salts, dimethylamine salts, trimethylamine salts, triethylamine salts, ethylamine salts, etc.
  • quaternary ammonium cations such as sodium, lithium, potassium, calcium, magnesium, tetramethylquaternary ammonium, tetraethylquaternary ammonium Salts, etc.
  • amine salts including salts formed with ammonia (NH3), primary amines, secondary amines or tertiary amines, such as methylamine salts
  • the compounds or conjugates of the present invention can exist in specific geometric or stereoisomer forms.
  • the chiral center can exist in the drug, in the linker structure, or in the Antibodies and their derivatives.
  • all such compounds including cis and trans isomers, (-)- and (+)-enantiomers, (R)- and (S)-enantiomers, diastereomers isomers, (D)-isomers, (L)-isomers, and racemic and other mixtures thereof, such as enantiomerically or diastereomerically enriched mixtures, are included in the present invention within the range.
  • Optically active (R)- and (S)-isomers as well as D and L-isomers can be prepared by chiral synthesis or chiral reagents or other conventional techniques. If one enantiomer of a compound or conjugate of the invention is desired, it can be prepared by asymmetric synthesis or derivatization with a chiral auxiliary, wherein the resulting diastereomeric mixture is separated and the auxiliary group is cleaved to provide the pure desired enantiomer.
  • a diastereoisomeric salt is formed with an appropriate optically active acid or base, and then a diastereomeric salt is formed by a conventional method known in the art. Diastereomeric resolution is performed and the pure enantiomers are recovered. Furthermore, the separation of enantiomers and diastereomers is usually accomplished by the use of chromatography using chiral stationary phases, optionally in combination with chemical derivatization methods (e.g. amines to amino groups formate).
  • solvates such as hydrates of the compounds, conjugates, pharmaceutically acceptable salts, and stereoisomers of the present invention are also within the scope of the present invention.
  • suitable solvates specifically, solvates formed between compounds or conjugates of the present invention and acetone, 2-butanol, 2-propanol, ethanol, ethyl acetate, tetrahydrofuran, diethyl ether, etc. . Hydrates or ethanolates are also mentioned.
  • treating means that the individual's symptoms are partially or completely relieved, or remain unchanged after treatment.
  • treatment includes prophylaxis, treatment and/or cure.
  • prevention means preventing an underlying disease and/or preventing worsening of symptoms or development of a disease. Treatment or prevention also includes any pharmaceutical use of the provided ADCs and the pharmaceutical compositions, pharmaceutical formulations provided herein.
  • the term “effective dose” refers to the amount of a compound which, when administered, alleviates to some extent one or more symptoms of the condition being treated.
  • the term “effective amount” refers to the amount of a compound which, when administered, inhibits the growth, proliferation or migration of cancer cells to some extent.
  • therapeutic effect means the effect resulting from the treatment of an individual, which alters, usually ameliorates or improves the symptoms of a disease or condition, or cures the disease or condition.
  • “individual” includes human or non-human animal.
  • exemplary human subjects include human subjects suffering from a disease (eg, a disease described herein) (referred to as a patient) or normal subjects.
  • non-human animals include all vertebrates, such as non-mammals (such as birds, amphibians, reptiles) and mammals, such as non-human primates, livestock and/or domesticated animals (such as sheep, dogs, cats, cows, pigs, etc.).
  • compositions and methods are not to be limited to the particular compositions and methods described and/or illustrated herein, and that the terminology used herein is for the purpose of describing particular embodiments by way of example only and is not intended to be limiting. Compositions and methods claimed.
  • compositions and methods of using the compositions refers to compositions and methods of using the compositions.
  • present disclosure describes or claims features or embodiments in relation to compositions, such features or embodiments apply equally to methods of using said compositions.
  • present disclosure describes or claims a feature or embodiment in relation to a method of using the composition, such feature or embodiment applies equally to said composition.
  • compositions and methods which are described herein in the context of separate embodiments, are, for clarity, also provided in combination in a single embodiment. Conversely, various features of the disclosed compositions and methods that are, for brevity, described in the context of a single embodiment, may also be provided separately or in any subcombination.
  • the ADC prepared by the linker designed in this disclosure not only has better drug efficacy than the ADC containing PAB structure, but also simplifies the structure of the linker, reduces the synthesis steps, and reduces the cost.
  • the length of the chain connecting the maleimide group and the amino acid residue or short peptide is shortened, for example, shortened to 3 methylene groups or even 2 methylene groups connected, and the hydrophobic part of PAB is removed.
  • the hydrophilicity of the ADC molecule is improved, the formation of ADC aggregates is reduced, the stability of the ADC is improved, and its PK in vivo can be improved, and the half-life of the drug in vivo can be increased.
  • the present disclosure unexpectedly finds that the ADC constructed by the truncated Linker of the present disclosure can significantly improve the affinity of the antibody to the target protein.
  • the ADC constructed with the truncated Linker of the present disclosure has improved drug efficacy in inhibiting tumors (such as pancreatic cancer or colon cancer).
  • the ADC of the present disclosure (such as ADC-1.1) has a side effect.
  • the ADC of the present disclosure (such as ADC-1.1) can not only kill the target cells with high expression of TROP-2 in tumor cells, but also kill the side The TROP-2 negative cells produced a certain degree of killing effect.
  • Fig. 1-1 is the SEC-HPLC detection spectrum of ADC-1.1 of the embodiment of the present invention.
  • Fig. 1-2 is the SEC-HPLC detection pattern of ADC-1.2 of the embodiment of the present invention.
  • Fig. 2-1 is the SEC-HPLC detection spectrum of ADC-2.1 of the embodiment of the present invention.
  • Fig. 2-2 is the SEC-HPLC detection spectrum of ADC-2.2 of the embodiment of the present invention.
  • Fig. 3 is the SEC-HPLC detection pattern of the control conjugate 2 (ie ADC-5) of the present invention.
  • Fig. 4 is the SEC-HPLC purity detection spectrum of the control conjugate ADC-217 of the present invention.
  • Fig. 5 is a graph showing the results of detecting the hydrophilicity and hydrophobicity of each ADC of the present invention by using a protein hydrophobic interaction chromatographic column (HIC).
  • HIC protein hydrophobic interaction chromatographic column
  • Fig. 6 is a graph showing the results of affinity detection between ADCs and TROP proteins of the present invention.
  • Fig. 7 is a statistical graph of the anti-tumor activity of each ADC of the present invention.
  • Fig. 8 is a graph showing body weight changes of experimental animals after treatment with various ADCs of the present invention.
  • Fig. 9 is a photograph taken of tumors taken from experimental animals after each ADC treatment of the present invention.
  • Fig. 10 is a graph showing the results of the bystander effect of ADC-1.1 of the present invention.
  • Fig. 11 is a statistical graph of the anti-tumor activity of each ADC of the present invention.
  • Fig. 12 is a graph showing body weight changes of experimental animals after treatment with various ADCs of the present invention.
  • Fig. 13 is a photograph taken of tumors taken from experimental animals after each ADC treatment of the present invention.
  • hRS7 antibody was produced in CHO cells.
  • the expression vectors containing the hRS7 antibody gene were constructed by conventional molecular biology methods.
  • the amino acid sequences of the hRS7 antibody light chain and heavy chain are shown in SEQ ID NO: 1 and SEQ ID NO: 2 respectively, and the corresponding nucleotide sequences Shown in SEQ ID NO:3 and SEQ ID NO:4 respectively.
  • Insert the above two sequences into the same expression vector extract a large number of transfection plasmids, and transfect them into CHO-K1 cells (ATCC CCL-61).
  • the specific transfection and antibody preparation processes are as follows:
  • Cell culture CHO-K1 cells were grown in suspension in ActiPro (GE HyClone) medium, and cultured at 37°C, 7% CO2, 140rpm, and 90% relative humidity;
  • SDS-PAGE and SEC-HPLC were used to analyze the molecular weight and purity of the purified antibody.
  • the results of SDS-PAGE showed that the molecular weight of the prepared hRS7 was as expected, and the purity of the antibody measured by SEC-HPLC was 99.1%.
  • Embodiment 2 prepares conjugate
  • Example 1 The antibody prepared in Example 1 was placed in a 10 mg/mL pH 7.4 PBS solution, using a 25°C water bath, while stirring and mixing, an equal volume of TCEP solution 2.4 times the amount of the substance was added, and the solution was placed in a 25°C water bath for 1.5 Hour. Use a 25°C water bath for the sample, then add 8 times the amount of compound A in DMSO solution (DMSO final concentration 10%) to the antibody solution while stirring and mixing, shake the reaction at 25°C for 90 minutes, and finally stir and mix the sample At the same time, 8 times the amount of cysteine was added, and placed in a water bath at 25°C for 10 minutes to react to obtain the coupling product ADC-1.1.
  • DMSO solution DMSO final concentration 10%
  • the purified ADC-1.1 is shown in Figure 1-1.
  • Example 1 The antibody prepared in Example 1 was placed in a 10 mg/mL pH 7.3 PBS solution, using a 22°C water bath, while stirring and mixing, an equal volume of TCEP solution 2.15 times the amount of the substance was added, and the solution was placed in a 22°C water bath to stand still for reaction 2 Hour.
  • Use a 22°C water bath for the sample then add 5.5 times the amount of compound A in DMSO solution (DMSO final concentration 10%) to the antibody solution while stirring and mixing, shake the reaction at 22°C for 60 minutes, and finally stir and mix the sample
  • 8 times the amount of cysteine was added, and placed in a water bath at 22°C for 10 minutes to react to obtain the coupling product ADC-1.2.
  • Example 1 The antibody prepared in Example 1 was placed in a 10 mg/mL pH 7.4 PBS solution, using a 25°C water bath, while stirring and mixing, an equal volume of TCEP solution 2.4 times the amount of the substance was added, and the solution was placed in a 25°C water bath for 1.5 Hour.
  • Use a 25°C water bath for the sample then add 8 times the amount of compound B in DMSO solution (DMSO final concentration 10%) to the antibody solution while stirring and mixing, shake the reaction at 25°C for 90 minutes, and finally stir and mix the sample
  • 8 times the amount of cysteine was added, and placed in a water bath at 25°C for 10 minutes to react to obtain the coupling product ADC-2.1.
  • the purified ADC-2.1 is shown in Figure 2-1.
  • Example 1 The antibody prepared in Example 1 was placed in a 10 mg/mL pH 7.3 PBS solution, using a 22°C water bath, while stirring and mixing, an equal volume of TCEP solution 2.15 times the amount of the substance was added, and the solution was placed in a 22°C water bath to stand still for reaction 2 Hour.
  • Use a 22°C water bath for the sample then add 5.5 times the amount of compound B in DMSO solution (DMSO final concentration 10%) to the antibody solution while stirring and mixing, shake the reaction at 22°C for 60 minutes, and finally stir and mix the sample
  • 8 times the amount of cysteine was added, and placed in a water bath at 22°C for 10 minutes to react to obtain the coupling product ADC-2.2.
  • Example 1 The antibody prepared in Example 1 was placed in an ice-water bath in a 10 mg/mL pH 7.4 PBS solution.
  • an equal volume of TCEP solution 3.5 times the amount of the substance was added while stirring and mixing, and the solution was placed at 25 °C water bath for 1 hour.
  • the temperature drops to 25°C then add 7 times the amount of compound C in 40% DMSO solution to the antibody solution while stirring and mixing, and shake the reaction at 25°C for 90 minutes.
  • cysteine was added in an amount 8 times that of the substance, and placed in a water bath at 25° C. for 10 minutes to react to obtain the coupling product ADC-4.
  • the detection, purification and DAR value determination methods of the coupling product are the same as ADC-1.1, and the DAR value of the obtained ADC-4 is 3.8.
  • Example 1 The antibody prepared in Example 1 was placed in a 5 mg/mL pH 7.4 PBS solution, while stirring and mixing, TCEP solution of 7 times the amount of the substance was added, and the solution was placed in a 37°C water bath for 1.5 hours to react. Use a 25°C water bath for the reaction solution. When the temperature drops to 25°C, then add 16 times the amount of compound C in 40% DMSO solution to the antibody solution while stirring and mixing, and shake the reaction at 25°C for 90 minutes. Finally, while the reaction solution was stirred and mixed, cysteine was added in an amount 16 times that of the substance, and placed in a water bath at 25° C. for 10 minutes to react to obtain the coupling product ADC-5.
  • DTT dithiothreitol
  • TROP-2 antibody (20mM acetic acid-sodium acetate, pH6.1) prepared in Example 1 was adjusted to pH7.5, and diluted with 50mM EPPS 5mM EDTA pH7.5buffer.
  • the product prepared in Section 5.3 was dissolved in DMSO. Put the protein solution (concentration 10mg/ml) (PH 7.5) in a water bath at 25°C. When the temperature of the protein solution reaches 25°C, add 1/10 volume of TCEP (soluble in water) (2.2989eq) while stirring and mixing, and the solution Place on a shaker at 25°C and shake at 30rmp for 90min.
  • the sample was bathed in a 25°C water bath, 5% DMSO was added while vortex mixing, and then 5% compound A' (7.5eq), 10% of the total DMSO was added, and the reaction was shaken on a shaker at 25°C at 30rmp for 90min. Cys (dissolved in water) (8eq) was added while the sample was vortex mixed, and placed on a shaker at 25°C at 30rmp for 10min to react. Centrifuge the sample at 5000rmp/min for 5min, use AKTA to desalt and change the medium in a solution of 25mM MES, pH5.5, and detect by SEC, the small molecules have been completely removed.
  • the purified ADC-217 is shown in Figure 4.
  • the bonding ratio DAR was estimated to be 3.80 by RP-HPLC, and the detection method was the same as that of ADC-1.1.
  • Embodiment 3 detects the hydrophilicity of each ADC
  • Example 2 The hydrophilicity and hydrophobicity of each ADC prepared in Example 2 were detected by using a protein hydrophobic interaction chromatographic column (HIC), and the specific experimental conditions were as follows:
  • Coating Trop2 protein Dilute Trop2 protein (Beijing Yiqiao Shenzhou Biotechnology Co., Ltd.) with coating solution (0.15M Na 2 CO 3 and 0.35M NaHCO 3 ) to 0.5 ⁇ g/mL, mix well and add to the microtiter plate medium, 100 ⁇ L/well, cover the plate with sealing film, and put it in a refrigerator at 4°C overnight.
  • Washing plate Take out the microplate plate Washing plate: Wash 3 times with 1*PBST.
  • Blocking Take the blocking solution (1% BSA), add 200 ⁇ L/well of the blocking solution, cover the plate with a sealing film, mix at 250 rpm and incubate at 25° C. for 1 h.
  • Plate washing wash 3 times with 1*PBST.
  • Antibody incubation After diluting the ADC samples 1, 2, 4, 5 prepared in Example 2 and the TROP-2 antibody prepared in Example 1 to 2000 ng/mL with 1% BSA, three-fold serial dilutions were made to 12 concentrations, 100 ⁇ L/ Add the wells to the ELISA plate, cover the plate with sealing film, mix at 250rpm and incubate at 25°C for 1h.
  • Plate washing Wash the microplate plate 3 times with 1*PBST.
  • Plate washing Wash the microplate plate 3 times with 1*PBST.
  • Color development add TMB (Huzhou Yingchuang) color development substrate: add 100ul per well to the microtiter plate, mix at 250rpm and place at 25°C for 15 minutes.
  • Stop reading add stop solution: add 1M H 2 SO 4 100ul to each well, and read at OD450nm on a microplate reader.
  • the test results are shown in Figure 6.
  • the ratio of the EC50 of the ADC prepared in Example 2 to the EC50 of the TROP-2 naked antibody is the vertical axis. The larger the ratio, the lower the affinity. Comparing the affinity of ADC products with different structures, the results show that ADC-1.1 The affinity with ADC-2.1 is the best, while the ADC with a similar structure to MORAb-202 has the worst affinity with TROP-2, that is, the ADC obtained by coupling the L-D structure of the present disclosure can better maintain the affinity of the antibody.
  • BxPC-3 cells human orthotopic pancreatic adenocarcinoma cells, ATCC CRL-1687
  • the culture conditions were 10% heat-inactivated fetal bovine serum and agar in 1640 medium, at 37°C, containing 5% Culture in an incubator with CO 2 air. Digested with 0.25% trypsin twice a week for passage. When the cells are in the exponential growth phase, the cells are collected, counted, and inoculated.
  • the P6 tumor tissue was used to evaluate the antitumor activity of the test product.
  • the tumor-bearing mice are anesthetized with CO 2 and killed, the tumor mass is removed, the surrounding necrotic tissue is removed, the tumor mass is cut into small tumor blocks of 20-30 mm 3 , and inoculated to the formal A total of 40 mice were inoculated at the right shoulder blade of the experimental mice.
  • Eighteen days after the tumor block was inoculated when the average tumor volume reached about 135 mm 3 , the mice with too small or too large tumor volume were excluded, and the remaining 25 mice were randomly grouped according to the tumor volume and started to be administered. In this experiment, only one administration was given, and no administration was given after the first administration on the grouping day. See the table below for the dosing regimen.
  • tumor volume (mm 3 ) 1/2 ⁇ (a ⁇ b 2 ) (where a represents the long diameter and b represents the short diameter).
  • the relative tumor proliferation rate, T/C% is the percentage value of the relative tumor volume or tumor weight between the treatment group and the control group at a certain time point.
  • Figures 7-9 The experimental results are shown in Figures 7-9, Figure 7 is the statistical curve of anti-tumor activity, Figure 8 is the curve of body weight change, and Figure 9 is the photograph taken of the tumor.
  • ADC-1.1 and ADC-2.1 have better anti-tumor effects than ADC-4 and ADC-5, among which ADC-1.1 and ADC-2.1 can completely make the tumor regress with only one administration, and have good safety .
  • Example 6 The bystander effect of ADC --- detection of the bystander effect of the ADC prepared in Example 2 (such as ADC-1.1)
  • fluorescent markers are loaded on A549, if the ADC-1.1 of the present disclosure has a side effect, then in the mixed cell culture group, eribulin small molecule toxin can be released by lysing BxPC-3 cells, thereby killing A549 cells, thereby making The fluorescence in the cells is quenched, and the cell inhibition rate of the bystander effect can be calculated by reading the absorbance value at 490nm from the microplate reader.
  • Co-cultivation A549-Luc cell suspension at a concentration of 2 ⁇ 10 4 cells/ml was mixed with BxPC-3 cells at a concentration of 8 ⁇ 10 4 cells/ml at a ratio of 1:1, mixed evenly and inoculated in each For the co-culture wells of the culture plate, 100 ⁇ l was inoculated in each well, and 3 wells were replicated.
  • Example 7 The tumor-inhibiting effect of the ADC prepared in Example 2 on colon cancer
  • Colo205 cells human colon cancer cells, ATCC CCL-222 were cultured in a single layer in vitro, and the culture conditions were 10% heat-inactivated fetal bovine serum and agar in 1640 medium, and cultured at 37°C with 5% CO2 air Cultivated in a box. Digested with 0.25% trypsin twice a week for passage. When the cells are in the exponential growth phase, the cells are collected, counted, and inoculated.
  • the P6 tumor tissue was used to evaluate the antitumor activity of the test product.
  • the tumor-bearing mice are anesthetized with CO 2 and killed, the tumor mass is removed, the surrounding necrotic tissue is removed, the tumor mass is cut into small tumor blocks of 20-30 mm 3 , and inoculated to the formal A total of 50 mice were inoculated at the right shoulder blade of the experimental mice. Seven days after tumor block inoculation, 30 mice with moderate tumor size were randomly divided into groups according to tumor volume and started to administer the drugs.
  • the dosing regimen is as follows:
  • tumor volume (mm 3 ) 1/2 ⁇ (a ⁇ b 2 ) (where a represents the long diameter and b represents the short diameter).
  • the relative tumor proliferation rate, T/C% is the percentage value of the relative tumor volume or tumor weight between the treatment group and the control group at a certain time point.
  • tumors were taken from all mice, weighed and photographed.
  • Figure 11 is the statistical curve of anti-tumor activity
  • Figure 12 is the curve of body weight change
  • Figure 13 is the photograph taken of the tumor. From the results, the anti-tumor effect ADC-1.2 ⁇ ADC-1.1>ADC-2.1>ADC-2.2>>ADC-217 (control). Among them, the tumors of all animals in the ADC-1.2 group completely regressed.

Abstract

一种亲和力改善的抗体-药物偶联物、其制备方法及应用,具体涉及了式(II)所示的抗体药物偶联物、其药物上可接受的盐、其立体异构体、或其溶剂合物。所述抗体药物偶联物不仅能够显著提高抗体与靶点蛋白的亲和力,而且能够提高ADC分子的亲水性,还能提高抑制肿瘤的药效。

Description

一种亲和力改善的抗体-药物偶联物、其制备方法及应用 技术领域
本公开涉及医药领域,具体涉及结合人类肿瘤学抗原标靶和/或提供抗微管蛋白药物活性的抗体药物偶联物(ADC)。本公开进一步涉及可用于治疗和诊断表达肿瘤抗原靶标和/或通过破坏微管蛋白可治疗的癌症的方法和组合物。
背景技术
MORAb-202是卫材的首个ADC,它是第一款将卫材内部开发的抗癌剂eribulin(艾立布林,或艾日布林)与靶向叶酸受体α(FRα)的人源化IgG1单抗偶联的ADC,其结构如下所示。抗体和毒素两者之间使用了一种可酶切割的连接子(linker)进行连接,药物抗体偶联比(DAR)为4.0。
Figure PCTCN2022135429-appb-000001
MORAb-202的有效载荷eribulin(产品名称:Halaven,通用名:艾立布林或艾日布林)与紫杉醇虽然同属于微管抑制剂,但其是首个软海绵素类微管动力学抑制剂,其具有一种新型作用机制。从结构上说,eribulin是一个简化和合成版本的软海绵素B(halichondrin B)。软海绵素B是从海绵Halichondria okadai(冈田软海绵)中分离出的一种天然产物。eribulin被认为通过抑制微管动力学的生长期,阻止细胞分裂。在癌细胞中,这种机制可导致长时间不可逆的有丝分裂阻滞,最终导致细胞死亡。Eribulin于2020年1月在中国上市,用于治疗既往接受过至少2种化疗方案(包括蒽环类和紫杉类)治疗的局部复发或转移性乳腺癌患者,因此Eribulin可有效的解决由蒽环类和紫衫类微管抑制剂治疗所带来的耐药性问题。
MORAb-202的连接子采用了PEG2-val-cit-PAB结构,加入对氨基苯甲氧基羰基(PAB)基团的作用是为了提供足够的酶切空间,使得ADC在胞内能够自断裂释放艾日布林小分子。而PEG2结构则为ADC分子提供更好的亲水性。
发明内容
虽然MORAb-202已经显示出良好的临床效果和应用前景,但其仍有改进的空间,特别是linker部分。linker作为ADC药物的桥梁,通过可切割或不可切割的连接物与抗体连接,linker需要精妙设计,既需要有稳定性和肿瘤细胞内的特异断裂性,同时也要有成药上的便利性,比如良好的亲水性,合适的DAR值,以及保持抗体良好的亲和力。因此对于ADC的连接子,仍旧有优化的空间和需求。
本公开发明人创造性的发现,艾日布林的分子结构带着一个伯胺,且该伯胺结构与艾日布林的主体结构较远,有足够的空间位置。基于此,发明人设计了不含PAB的连接子,直接将艾日布林的伯胺与氨基酸或短肽(例如Vc或者Va结构)连接形成酰胺键;此外,本公开发明人还创造性地发现,在马来酰亚胺基后采用短链而非长链与氨基酸或短肽(例如Vc或者Va)连接,可以增加ADC的亲水性。发明人基于前述两方面的“截短”理念的设计,获得了本公开的连接子、连接子-艾日布林小分子化合物以及ADC。本公开发明人意外地发现,本公开的截短型的Linker构建的ADC不仅能够显著提高ADC分子的亲水性,,而且能够提高抗体与靶点蛋白的亲和力,还能提高抑制肿瘤的药效。
为此,在本公开的第一方面,本公开提供了式(I)所示化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,
L 1-L 2-D
(I)
其中:
L 1
Figure PCTCN2022135429-appb-000002
Y为
Figure PCTCN2022135429-appb-000003
X 1、X 2、X 3、X 4各自独立地选自CH 2、NH、O、S,且X 1、X 2、X 3和X 4中至少有一个为CH 2
n 1、n 2、n 3、n 4各自独立地选自0、1,且n 1、n 2、n 3和n 4中至少有一个为1;
L 2选自氨基酸残基或由2-10个氨基酸残基组成的短肽;优选地,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接;
D为与L 2通过化学键相连的药物,所述药物优选为艾日布林或其衍生物。
在一些实施方案中,Y为
Figure PCTCN2022135429-appb-000004
在一些实施方案中,X 1、X 2、X 3、X 4均为CH 2
在一些实施方案中,n 1为0,n 2、n 3、n 4各自独立地选自0、1,且n 2、n 3和n 4中至少有一个为1。
在一些实施方案中,n 1、n 2为1,n 3、n 4为0。
在一些实施方案中,n选自1、2、3、4。
在一些实施方案中,n选自1、2、3。
在一些实施方案中,n为2。
在一些实施方案中,L 2为由2-4个氨基酸残基组成的短肽,优选地,所述氨基酸选自甘氨酸、苯丙氨酸、缬氨酸、瓜氨酸、丙氨酸,更优选地,所述氨基酸选自缬氨酸、瓜氨酸、丙氨酸。在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,L 2选自甘氨酸-甘氨酸-苯丙氨酸-甘氨酸(GGFG)、缬氨酸-瓜氨酸(VC)、缬氨酸-丙氨酸(VA),优选选自缬氨酸-瓜氨酸(VC)、缬氨酸-丙氨酸(VA)。在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,L 2选自
Figure PCTCN2022135429-appb-000005
Figure PCTCN2022135429-appb-000006
优选选自
Figure PCTCN2022135429-appb-000007
在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,D为
Figure PCTCN2022135429-appb-000008
在一些实施方案中,连接子在细胞外是稳定的,使得ADC在存在于细胞外环境中时保持完整,但在例如癌细胞的细胞中内化时能够裂解。在一些实施方案中,当ADC进入表达对ADC的抗体部分具有特异性的抗原的细胞时,艾日布林药物部分从抗体部 分裂解,且裂解释放艾日布林的未修饰形式。在一些实施方案中,连接子包含使得所述连接子或所述抗体部分在裂解后保持结合于艾日布林的可裂解部分。
在一些实施方案中,连接子中的可裂解部分为可裂解肽部分。在一些实施方案中,相对于包含替代可裂解部分的ADC,包含可裂解肽部分的ADC显示较低的聚集水平,改善的抗体:药物比率,增加的癌细胞的靶向杀死,减少的非癌细胞的脱靶杀死,和/或较高的药物负载(p)。在一些实施方案中,相对于不可裂解的连接子,添加可裂解部分增加细胞毒性和/或效力。在一些实施方案中,增加的效力和/或细胞毒性是在表达中等水平的由ADC的抗体部分所靶向的抗原(例如中等FRA表达)的癌症中。在一些实施方案中,可裂解肽部分能够由酶裂解,且连接子为酶能够裂解的连接子。在一些实施方案中,酶为组织蛋白酶,且连接子为组织蛋白酶能够裂解的连接子。在某些实施方案中,与替代分裂机制相比,酶能够裂解的连接子(例如组织蛋白酶能够裂解的连接子)显示上述改善特性中的一种或多种。
在一些实施方案中,连接子中的可裂解肽部分包含氨基酸单元。在一些实施方案中,氨基酸单元包含缬氨酸-瓜氨酸(Val-Cit)或缬氨酸-丙氨酸(Val-Ala)。在一些实施方案中,相对于包含替代氨基酸单元或替代可裂解部分的ADC,包含Val-Cit或Val-Ala的ADC显示增加的稳定性,减少的脱靶细胞杀死,增加的靶向细胞杀死,较低的聚集水平,和/或较高的药物负载。
在一些实施方案中,连接子包含至少一种将抗体部分接合于可裂解部分的间隔子单元。在一些实施方案中,连接子中的间隔子单元可以包含至少一种maleimidocaproyl(MC)部分或其截短短片段,优选MC截短片段。在一些实施方案中,尽管连接子长度较短,但相对于包含较长间隔子单元(例如MC或(PEG) 2- 8)的ADC,包含较短间隔子单元(例如截短MC)的ADC显示较低的聚集水平和稳定性和/或更好的药效。
在一些实施方案中,本公开抗体偶联物中连接子的间隔单元不包含连接于D的间隔单元对氨基苯甲氧基羰基(PAB)。
在一些实施方案中,连接子中的间隔子单元经由顺丁烯二酰亚胺部分(Mal)附接于ADC的抗体部分。
在某些实施方案中,所示L 1-L 2-D化合物选自:
化合物1
Figure PCTCN2022135429-appb-000009
化合物2
Figure PCTCN2022135429-appb-000010
化合物3
Figure PCTCN2022135429-appb-000011
另外,以下对照化合物1和对照化合物2仅作为本公开式(I)所示化合物的对照结构,并非本公开请求保护的化合物。
对照化合物1:
Figure PCTCN2022135429-appb-000012
对照化合物2:
Figure PCTCN2022135429-appb-000013
在本公开的第二方面,本公开提供了式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,
Ab-(L 1-L 2-D) p
(II)
其中:
Ab为抗体或其抗原结合片段;
L 1
Figure PCTCN2022135429-appb-000014
Y为
Figure PCTCN2022135429-appb-000015
X 1、X 2、X 3、X 4各自独立地选自CH 2、NH、O、S,且X 1、X 2、X 3和X 4中至少有一个为CH 2
n 1、n 2、n 3、n 4各自独立地选自0、1,且n 1、n 2、n 3和n 4中至少有一个为1;
L 2选自氨基酸残基或由2-10个氨基酸残基组成的短肽;优选地,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接;
D为与L 2通过化学键相连的药物,所述药物优选为艾日布林或其衍生物;
p为1-20之间的任意数值。
在一些实施方案中,Y为
Figure PCTCN2022135429-appb-000016
在一些实施方案中,X 1、X 2、X 3、X 4均为CH 2
在一些实施方案中,n 1为0,n 2、n 3、n 4各自独立地选自0、1,且n 2、n 3和n 4中至少有一个为1。
在一些实施方案中,n 1、n 2为1,n 3、n 4为0。
在一些实施方案中,n选自1、2、3、4。
在一些实施方案中,n选自1、2、3。
在一些实施方案中,n为2。
在一些实施方案中,L 2为由2-4个氨基酸残基组成的短肽,优选地,所述氨基酸选自甘氨酸、苯丙氨酸、缬氨酸、瓜氨酸、丙氨酸,更优选地,所述氨基酸选自缬氨酸、瓜氨酸、丙氨酸。在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,L 2选自甘氨酸-甘氨酸-苯丙氨酸-甘氨酸(GGFG)、缬氨酸-瓜氨酸(VC)、缬氨酸-丙氨酸(VA),优选选自缬氨酸-瓜氨酸(VC)、缬氨酸-丙氨酸(VA)。在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,L 2选自
Figure PCTCN2022135429-appb-000017
Figure PCTCN2022135429-appb-000018
优选选自
Figure PCTCN2022135429-appb-000019
在一些实施方案中,L 2的氨基端与L 1相连接,L 2的羰基端与D相连接。
在一些实施方案中,D为
Figure PCTCN2022135429-appb-000020
需要说明的是,上述式(II)所示的“抗体药物偶联物”指的是连接的药物分子的个 数相同或不同的ADC分子的组合物。
具体地,本公开提供了包含多个ADC分子的组合物。在某些情况下,本文所述的组合物中的每个ADC包含相同数目的一个或多个药物分子。在其他情况下,本文所述的组合物中的每个ADC包含不同数目的一个或多个药物分子。
本文所述的抗体药物偶联物中,每个抗体可以偶联有1个、2个、3个、4个、5个、6个、7个、8个或更多药物分子,例如是1至5个(例如1、2、3、4或5个)药物分子、5至8个(例如5、6、7或8个)药物分子或8至12个(例如8、9、10、11或12个)药物分子。
药物抗体比(DAR)是指偶联到抗体的药物分子的个数(例如,式II中的p)。本文所述的ADC中包含的药物分子的个数通常是整数,当本文所述的ADC中包含的药物分子的个数(例如,式II中的p)是分数时,该分数指的是包含多个ADC分子的组合物中,每个抗体偶联的药物分子的平均数量。
在一些实施方案中,p为1-10之间的任意数值。
在一些实施方案中,p为3-5之间的任意数值。
在一些实施方案中,p为3.5-4.5之间的任意数值,例如,p为3.5、3.9或4.1。
在一些实施方案中,所述抗体药物偶联物选自以下:
ADC-1
Figure PCTCN2022135429-appb-000021
ADC-2
Figure PCTCN2022135429-appb-000022
ADC-3
Figure PCTCN2022135429-appb-000023
需要说明的是,以上ADC-1、ADC-2或ADC-3中,-S-并非另外外接的硫原子,而是打开二硫键后的Ab自身所含有巯基与
Figure PCTCN2022135429-appb-000024
进行连接后形成的-S-。
相应地,本领域技术人员可以理解,上述式(II)中,L 1与Ab相连接后,L 1的结构式为
Figure PCTCN2022135429-appb-000025
其中,
Figure PCTCN2022135429-appb-000026
左侧的碳端与打开二硫键后的Ab自身所含有巯基相连接。
在一些实施方案中,本公开抗体-药物偶联物(ADC)中所述抗体选自鼠源抗体、嵌合抗体、人源化抗体和全人源抗体。
适用的抗体可结合本领域中已知的任何疾病相关的抗原。当疾病状态是癌症时,举例来说,由肿瘤细胞表达或另外与肿瘤细胞相关的许多抗原在本领域中是已知的,包括但不限于碳酸酐酶IX、α-胎蛋白(alpha-fetoprotein,AFP)、α-辅肌动蛋白-4(α-actinin-4)、A3、对A33抗体具有特异性的抗原、ART-4、B7、Ba 733、BAGE、BrE3抗原、CA125、CAMEL、CAP-1、CASP-8/m、CCL19、CCL21、CD1、CD1a、CD2、CD3、CD4、CD5、CD8、CD11A、CD14、CD15、CD16、CD18、CD19、CD20、CD21、CD22、CD23、CD25、CD29、CD30、CD32b、CD33、CD37、CD38、CD40、CD40L、CD44、CD45、CD46、CD52、CD54、CD55、CD59、CD64、CD66a-e、CD67、CD70、CD70L、CD74、CD79a、CD80、CD83、CD95、CD126、CD132、CD133、CD138、CD147、CD154、CDC27、CDK-4/m、CDKN2A、CTLA-4、CXCR4、CXCR7、CXCL12、HIF-1α、结肠特异性抗原p(CSAp)、CEA(CEACAM5)、CEACAM6、c-Met、DAM、EGFR、EGFRvIII、EGP-1(Trop-2)、EGP-2、ELF2-M、Ep-CAM、her2、her3、claudin 18.2、ROR1、ROR2、dll3、marc17、纤维母细胞生长因子(FGF)、Flt-1、Flt-3、叶酸受体、G250抗原、GAGE、gp100、GRO-β、HLA-DR、HM1.24、人绒毛膜促性腺激素(HCG)和它的亚基、HER2/neu、HMGB-1、缺氧诱导性因子(HIF-1)、 HSP70-2M、HST-2、Ia、IGF-1R、IFN-γ、IFN-α、IFN-β、IFN-λ、IL-4R、IL-6R、IL-13R、IL-15R、IL-17R、IL-18R、IL-2、IL-6、IL-8、IL-12、IL-15、IL-17、IL-18、IL-23、IL-25、胰岛素样生长因子1(IGF-1)、IGF-1R、KS1-4、Le-Y、LDR/FUT、巨噬细胞迁移抑制因子(MIF)、MAGE、MAGE-3、MART-1、MART-2、NY-ESO-1、TRAG-3、mCRP、MCP-1、MIP-1A、MIP-1B、MIF、MUC1、MUC2、MUC3、MUC4、MUC5ac、MUC13、MUC16、MUM-1/2、MUM-3、NCA66、NCA95、NCA90、胰腺癌粘蛋白、PD-1受体、PD-L1受体、胎盘生长因子、p53、PLAGL2、前列腺酸性磷酸酶、PSA、PRAME、PSMA、PlGF、ILGF、ILGF-1R、IL-6、IL-25、RS5、RANTES、T101、SAGE、S100、存活素(survivin)、存活素-2B、TAC、TAG-72、腱生蛋白(tenascin)、TRAIL受体、TNF-α、Tn抗原、汤姆逊-弗雷登里希抗原(Thomson-Friedenreich antigen)、肿瘤坏死抗原、VEGFR、ED-B纤维连接蛋白(fibronectin)、WT-1、17-1A抗原、补体因子C3、C3a、C3b、C5a、C5、血管生成标志物、bcl-2、bcl-6、Kras、致癌基因标志物和致癌基因产物(参见例如Sensi等,Clin Cancer Res2006,12:5023-32;Parmiani等,J Immunol 2007,178:1975-79;Novellino等Cancer Immunol Immunother 2005,54:187-207)。优选地,抗体结合her2、her3、claudin 18.2、ROR-1、dll-3、muc1、muc-17、EGP-1(Trop-2)。
可利用的示例性抗体包括但不限于hR1(抗IGF-1R,美国专利号13/688,812)、hPAM4(抗粘蛋白,美国专利号7,282,567)、hA20(抗CD20,美国专利号7,151,164)、hA19(抗CD19,美国专利号7,109,304)、hIMMU31(抗AFP,美国专利号7,300,655)、hLL1(抗CD74,美国专利号7,312,318)、hLL2(抗CD22,美国专利号5,789,554)、hMu-9(抗CSAp,美国专利号7,387,772)、hL243(抗HLA-DR,美国专利号7,612,180)、hMN-14(抗CEACAM5,美国专利号6,676,924)、hMN-15(抗CEACAM6,美国专利号8,287,865)、hRS7(抗EGP-1,美国专利号7,238,785)、hMN-3(抗CEACAM6,美国专利号7,541,440)、Ab124和Ab125(抗CXCR4,美国专利号7,138,496),各引用的专利或申请的实施例部分以引用的方式并入本文。更优选地,抗体是IMMU-31(抗AFP)、hRS7(抗Trop-2)、hMN-14(抗CEACAM5)、hMN-3(抗CEACAM6)、hMN-15(抗CEACAM6)、hLL1(抗CD74)、hLL2(抗CD22)、hL243或IMMU-114(抗HLA-DR)、hA19(抗CD19)或hA20(抗CD20)。如本文所用,术语依帕珠单抗(epratuzumab)和hLL2可互换,术语维妥珠单抗(veltuzumab)和hA20、hL243g4P、hL243γ4P和IMMU-114也是如此。在一最优选实施方案中,抗体是抗Trop-2抗体诸如hRS7;抗ROR1抗 体诸如99961(中国专利号104662044);抗HER-3抗体如patritumab(中国专利号102174105);抗HER2抗体赫赛汀(Trastuzumab),帕妥珠(Pertuzumab)。
适用的替代性抗体包括但不限于阿昔单抗(abciximab)(抗糖蛋白IIb/IIIa)、阿仑单抗(alemtuzumab)(抗CD52)、贝伐单抗(bevacizumab)(抗VEGF)、西妥昔单抗(cetuximab)(抗EGFR)、吉妥单抗(gemtuzumab)(抗CD33)、替伊莫单抗(ibritumomab)(抗CD20)、帕尼单抗(panitumumab)(抗EGFR)、利妥昔单抗(rituximab)(抗CD20)、托西莫单抗(tositumomab)(抗CD20)、曲妥珠单抗(trastuzumab)(抗ErbB2)、兰罗利珠单抗(lambrolizumab)(抗PD1受体)、阿特珠单抗(atezolizumab)(抗PD-L1)、MEDI4736(抗PD-L1)、纳武单抗(nivolumab)(抗PD-1受体)、伊匹单抗(ipilimumab)(抗CTLA-4)、阿巴伏单抗(abagovomab)(抗CA-125)、阿德木单抗(adecatumumab)(抗EpCAM)、阿利珠单抗(atlizumab)(抗IL-6受体)、贝那珠单抗(benralizumab)(抗CD125)、奥滨尤妥珠单抗(obinutuzumab)(GA101,抗CD20)、CC49(抗TAG-72)、AB-PG1-XG1-026(抗PSMA,美国专利申请11/983,372,以ATCC PTA-4405和PTA-4406保藏)、D2/B(抗PSMA,WO 2009/130575)、托珠单抗(tocilizumab)(抗IL-6受体)、巴利昔单抗(basiliximab)(抗CD25)、达利珠单抗(daclizumab)(抗CD25)、依法珠单抗(efalizumab)(抗CD11a)、GA101(抗CD20;GlycartRoche)、莫罗莫那(muromonab)-CD3(抗CD3受体)、那他珠单抗(natalizumab)(抗α4整联蛋白)、奥马珠单抗(omalizumab)(抗IgE);抗TNF-α抗体诸如CDP571(Ofei等,2011,Diabetes 45:881-85)、MTNFAI、M2TNFAI、M3TNFAI、M3TNFABI、M302B、M303(ThermoScientific,Rockford,IL)、英夫利昔单抗(infliximab)(Centocor,Malvern,PA)、聚乙二醇赛妥珠单抗(certolizumab pegol)(UCB,Brussels,Belgium)、抗CD40L(UCB,Brussels,Belgium)、阿达木单抗(adalimumab)(Abbott,Abbott Park,IL)、Benlysta(Human Genome Sciences);用于阿尔茨海默氏病(Alzheimer's disease)的治疗的抗体诸如Alz 50(Ksiezak-Reding等,1987,J Biol Chem 263:7943-47)、甘特如单抗(gantenerumab)、苏兰珠单抗(solanezumab)和英夫利昔单抗;抗纤维蛋白抗体如59D8、T2G1s、MH1;抗CD38抗体诸如MOR03087(MorphoSys AG)、MOR202(Celgene)、HuMax-CD38(Genmab)或达雷木单抗(daratumumab)(Johnson&Johnson);(抗HIV抗体诸如P4/D10(美国专利8,333,971)、Ab75、Ab76、Ab77(Paulik等,1999,Biochem Pharmacol 58:1781-90)以及由Polymun(Vienna,Austria)描述和销售的抗HIV抗体,也描述于美国专利5,831,034、美国专利5,911,989以及Vcelar等,AIDS 2007;21(16):2161-2170和Joos等,Antimicrob.Agents  Chemother.2006;50(5):1773-9中,全都以引用的方式并入本文。
在某些实施方案中,缀合于主题抗体的药物部分选自艾日布林或其衍生物,例如艾日布林的甲磺酸盐。如本文所用,术语“艾日布林”是指软海绵素B(最初从海绵冈田软海绵(Halichondria okadais)分离的大环化合物)的合成类似物。艾日布林为微管抑制剂,认为其结合微管蛋白并通过抑制有丝分裂纺锤体组件,在G2/M期引起细胞周期停滞。术语“艾日布林甲磺酸盐”是指艾日布林的甲磺酸盐,其以商品名Halaven TM出售。
在本公开的第三方面,本公开提供了制备第一方面的化合物、药学上可接受的盐、立体异构体或溶剂合物的方法。
在一些实施方案中,所述方法包括:将L 1-L 2-OH与艾日布林进行酰胺化反应,获得式(I)所示的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,其中,L 1、L 2如本公开第一方面所限定。
在一些具体实施方案中,所述方法包括:
步骤1:将马来酰亚胺活性酯与
Figure PCTCN2022135429-appb-000027
与碱溶解在NMP中,室温条件下搅拌4小时后,HPLC显示有新峰生成,Pre-HPLC纯化后得目标产物,LCMS显示为目标产物。
步骤2:将
Figure PCTCN2022135429-appb-000028
与eruibulin,HATU,碱加入到NMP中,室温搅拌2小时后,HPLC显示有新峰生成,Pre-HPLC纯化后,冻干得到目标产物,LCMS确认为目标产物。
在本公开的第四方面,本公开提供了制备第二方面所述的抗体-药物偶联物、药学上可接受的盐、立体异构体或溶剂合物的方法。
在一些实施方案中,所述方法包括:将前述第一方面所述的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物与前述的Ab反应,获得式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物。
在一些实施方案中,所述第一方面所述的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物是通过前述第三方面所述的方法制备得到的。
在一些实施方案中,对于制造的抗体-药物偶联物(II),可利用以下的共通操作,进 行浓缩、缓冲液交换、纯化等的操作。
共通操作A:抗体或抗体-药物偶联物水溶液的浓缩
在容器内,放入抗体或抗体-药物偶联物溶液,使用离心机进行离心操作(例如,以2000G~3800G离心5~20分钟),将抗体或抗体-药物偶联物溶液浓缩。
共通操作B:抗体的浓度测定
使用UV测定器,按照制造商规定的方法,进行抗体浓度的测定。
此时,使用随着抗体不同而显示不同的280nm吸光系数(1.3mLmg -1cm -1~1.8mLmg - 1cm -1)。
共通操作C-1:抗体的缓冲液交换
按照制造商说明书的方法,用含有氯化钠(例如,137mM)及乙二胺四乙酸(EDTA),例如,5mM)的磷酸缓冲液(例如PBS)(本说明书中也称为PBS/EDTA)。将使用了Sephadex G-25载体的PD-10柱平衡化。针对一根该PD-10柱,装填1mL抗体水溶液,然后分离用PBS/EDTA 2mL洗脱的级分(3.5mL)。利用共通操作A将该级分浓缩,使用共通操作B,进行抗体浓度的测定,然后使用PBS/EDTA,将抗体浓度调整为10mg/mL。
共通操作C-2:抗体的缓冲液交换
按照制造商规定的方法,用含有氯化钠(例如,50mM)及EDTA(例如,2mM)的磷酸缓冲液(例如,50mM,pH6.5,本说明书中也称为PBS6.5/EDTA)。将使用了Sephadex G-25载体的PD-10柱平衡化。针对一根该PD-10柱,装填1mL抗体水溶液,然后分离获取用PBS6.5/EDTA 2mL洗脱的级分(3.5mL)。利用共通操作A将该级分浓缩,使用共通操作B进行抗体浓度的测定,然后使用PBS6.5/EDTA,将抗体浓度调整为5mg/mL。
共通操作D-1:抗体-药物偶联物的纯化
使用市售的磷酸缓冲液(例如,PBS7.4)、含有氯化钠(例如,137mM)的磷酸钠缓冲液(例如,10mM,pH6.5;本说明书中也称为PBS6.5),或含有山梨糖醇(例如,5%)的乙酸缓冲液(例如,10mM,pH5.5;本说明书中也称为ABS)或MES 25mM pH6.5,或His 10mM pH 5.5中的任一种缓冲液将PD-10柱平衡化。在该柱中装填抗体-药物偶联物反应水溶液(例如,约1mL),用制造商规定的量的缓冲液洗脱,由此分离获取抗体级分,由此,得到了除去了未连接的药物接头、低分子化合物(三(2-羧基乙基)膦盐酸盐(TCEP),半胱氨酸,二甲基亚砜等)的抗体-药物偶联物。
共通操作D-2:抗体-药物偶联物的纯化
磷酸缓冲液(例如,PBS7.4)、含有氯化钠(例如,137mM)的磷酸钠缓冲液(例如,10mM,pH6.5;本说明书中也称为PBS6.5),或含有山梨糖醇(例如,5%)的乙酸缓冲液(例如,10mM,pH5.5;本说明书中也称为ABS)或MES 25mM pH6.5,或His10mM pH 5.5中的任一种缓冲液将AKTA柱(填料:sephadex G 25)平衡化。进样器装载抗体-药物偶联物反应水溶液(例如,约2mL),用制造商规定的量的缓冲液洗脱,由此分离获取抗体级分。由此,得到了除去了未连接的药物接头、低分子化合物(三(2-羧基乙基)膦盐酸盐(TCEP),半胱氨酸,二甲基亚砜等)的抗体-药物偶联物。
在本公开的第五方面,本公开提供了一种药物组合物,其包含第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物,以及一种或多种药用辅料,例如载体和/或赋形剂。
所述药物组合物可以制成药学上可接受的任一剂型。所述药物组合物还可以以任何合适的给药方式,例如口服、肠胃外、直肠或经肺给药等方式施用于需要这种治疗的个体。用于口服给药时,所述药物组合物可制成常规的固体制剂,如片剂、胶囊剂、丸剂、颗粒剂等;也可制成口服液体制剂,如口服溶液剂、口服混悬剂、糖浆剂等。制成口服制剂时,可以加入适宜的填充剂、粘合剂、崩解剂、润滑剂等。用于肠胃外给药时,所述药物组合物可制成注射剂,包括注射液、注射用无菌粉末与注射用浓溶液。制成注射剂时,可采用现有制药领域中的常规方法生产,配制注射剂时,可以不加入附加剂,也可根据药物的性质加入适宜的附加剂。用于直肠给药时,所述药物组合物可制成栓剂等。用于经肺给药时,所述药物组合物可制成吸入剂或喷雾剂等。
在本公开的第六方面,本公开提供了第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物在制备治疗和/或预防与细胞活动异常相关的疾病(例如癌症疾病)的药物中的用途。
在本公开的第七方面,本公开提供了第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物,其用于治疗和/或预防与细胞活动异常相关的疾病(例如癌症疾病)。
在本公开的第八方面,本公开提供了治疗或预防与细胞活动异常相关的疾病(例如癌症疾病)的方法,包括给有此需要的个体施用有效剂量的本公开提供第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物。可调整给药方案以提供最佳所需响应。例如,可给药单次推注,可随时间给药数个分剂量,或可如治疗情况的急需所表明而按比例减少或增加剂量。剂量值可随要减轻的病况的类型及严重性而变化,且可包括单次或多次剂量。要进一步理解,对于任何特定个体,具体的给药方案可根据个体需要及给药组合物或监督组合物的给药的人员的专业判断来随时间调整。
各种实施方案可涉及使用主题方法和组合物来治疗癌症,包括原位癌或转移癌。由此,针对上述第六方面、第七方面、第八方面,在一些实施方案中,所述癌症为原位癌或转移癌。在一些具体实施方案中,所述癌症包括但不限于转移性乳腺癌、非小细胞肺癌、伯基特淋巴瘤(Burkitt lymphoma)、霍奇金氏淋巴瘤、急性骨髓性白血病、慢性骨髓性白血病、急性淋巴细胞性白血病、慢性淋巴细、皮肤癌、口腔癌、食道癌、胃肠道癌、肺道癌、肺癌、胃癌、结肠癌、直肠癌、三阴性乳腺癌(TNBC)、卵巢癌、前列腺癌、子宫癌、子宫内膜癌、子宫颈癌、膀胱癌、胰腺癌、骨癌、脑癌、结缔组织癌、甲状腺癌、肝癌、胆囊癌、膀胱(尿道上皮)癌、肾癌、皮肤癌、中枢神经系统癌、睾丸癌、上皮癌、头颈部癌。在一些优选的实施方案中,所述癌症选自转移性乳腺癌、TNBC、非TNBC、子宫颈癌、子宫内膜癌、肺癌、卵巢癌、膀胱(尿道上皮)癌、结肠癌、胰腺癌。在一些更优选的实施方案中,所述癌症为TNBC、卵巢癌、子宫颈癌、膀胱(尿道上皮)癌或胰腺癌(如原位胰腺癌)。
在涉及治疗癌症的某些实施方案中,抗体或免疫缀合物可与手术、放射疗法、化学疗法、用包括检查点抑制抗体的裸抗体进行的免疫疗法、放射免疫疗法、免疫调节剂、疫苗等组合使用。最优选地,抗体或免疫缀合物与PARP抑制剂、微管抑制剂、布鲁顿激酶抑制剂和/或PI3K抑制剂组合使用。这些组合疗法可允许在所述组合的情况下给予较低剂量的各治疗剂,因此降低某些重度副作用,以及潜在缩减所需疗程。当不存在或存在最小重叠毒性时,也可给予各自的完全剂量。
尽管抗体或免疫缀合物可以定期弹丸注射液形式施用,但在替代性实施方案中,抗体或免疫缀合物可通过连续输注来施用。为使抗体或免疫缀合物在血液中的Cmax增加以及延长抗体或免疫缀合物在血液中的PK,可例如通过留置导管来施用连续输注。此 类装置在本领域中是已知的,诸如
Figure PCTCN2022135429-appb-000029
Figure PCTCN2022135429-appb-000030
导管(参见例如Skolnik等,Ther Drug Monit 32:741-48,2010),并且可使用任何所述已知留置导管。多种连续输注泵在本领域中也是已知的,并且可使用任何所述已知输注泵。连续输注的剂量范围可介于每天0.1与3.0mg/kg之间。更优选地,这些免疫缀合物可历经2至5小时,更优选2-3小时的相对短暂时期,通过静脉内输注来施用。
在特别优选实施方案中,在对标准疗法具有抗性的患者中,抗体或免疫缀合物和给药时程可为有效的。举例来说,可向尚未对用艾日布林进行的先前疗法响应的患者施用hRS7-艾日布林免疫缀合物。惊人的是,伊立替康抗性患者可显示对hRS7-艾日布林的部分或甚至完全响应。免疫缀合物特异性靶向肿瘤组织的能力可由于治疗剂的靶向性改进和递送增强而克服肿瘤抗性。一特定优选受试者可为患有Trop-2阳性乳腺癌、卵巢癌、子宫颈癌、子宫内膜癌、肺癌、前列腺癌、结肠癌、直肠癌、胃癌、食道癌、膀胱(尿道上皮)癌、肾癌、胰腺癌、脑癌、甲状腺癌、上皮癌、或头颈部癌的患者。优选地,癌症是转移性癌症。更优选地,患者用至少一种标准抗癌疗法进行的治疗先前已失败。在替代性优选实施方案中,癌症是转移性乳腺癌、TNBC、非TNBC、子宫内膜癌、肺癌、卵巢癌或结肠癌。
在本公开的第九方面,本公开提供了一种药物制剂,其包含第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物。
在本公开的第十方面,本公开提供了第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物用于制备药物制剂的用途。
在本公开的第十一方面,本公开提供了一种试剂盒,其包括第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物,或第九方面的药物制剂。
本公开提供的前述化合物、抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物、药物制剂或试剂盒可用于抑制癌细胞生长、增殖或迁移。所述化合物、抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物、药物制剂或试剂盒用于体内或者体外施用;例如,被施用至个体体内,或者被施用至体外细胞(例如细胞系或者来自个体的细胞例如癌症细胞)。在一些优选的实施方案中,前述化合物、抗体药物偶 联物、药学上可接受的盐、立体异构体、或溶剂合物、药物制剂或试剂盒用于非治疗目的(例如体外科学研究等),用于抑制癌细胞生长、增殖或迁移。
由此,在本公开的第十二方面,本公开提供了第一方面所述的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者第二方面所述的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者第五方面所述的药物组合物,或者第九方面的药物制剂在制备用于抑制癌细胞生长、增殖或迁移的试剂中的用途,尤其在制备用于体外抑制癌细胞生长、增殖或迁移的试剂中的用途。其中,所述癌细胞中的癌即癌症,在一些实施方案中,所述癌症如前所述。在一些实施方案中,所述用途用于非治疗目的。
在本公开的第十三方面,本公开提供了第一方面所述的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者第二方面所述的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者第五方面所述的药物组合物,或者第九方面的药物制剂,其用于抑制癌细胞生长、增殖或迁移,尤其用于体外抑制癌细胞生长、增殖或迁移。其中,所述癌细胞中的癌即癌症,在一些实施方案中,所述癌症如前所述。在一些实施方案中,所述用途用于非治疗目的。
在本公开的第十四方面,本公开提供了一种抑制癌细胞生长、增殖或迁移(尤其是体外抑制癌细胞生长、增殖或迁移)的方法,其包括给癌细胞施用有效量的第一方面所述的化合物、药学上可接受的盐、立体异构体、或溶剂合物,或第二方面所述的抗体药物偶联物、药学上可接受的盐、立体异构体、或溶剂合物,或第五方面的药物组合物,或第九方面的药物制剂。其中,所述癌细胞中的癌即癌症,在一些实施方案中,所述癌症如前所述。在一些实施方案中,所述方法用于非治疗目的。
具体地,针对上述第十二方面、第十三方面、第十四方面,在一些实施方案中,所述癌细胞为原位癌细胞或转移癌细胞。在一些具体实施方案中,所述癌细胞选自转移性乳腺癌、非小细胞肺癌、伯基特淋巴瘤(Burkitt lymphoma)、霍奇金氏淋巴瘤、急性骨髓性白血病、慢性骨髓性白血病、急性淋巴细胞性白血病、慢性淋巴细、皮肤癌、口腔癌、食道癌、胃肠道癌、肺道癌、肺癌、胃癌、结肠癌、直肠癌、三阴性乳腺癌(TNBC)、卵巢癌、前列腺癌、子宫癌、子宫内膜癌、子宫颈癌、膀胱癌、胰腺癌、骨癌、脑癌、结缔组织癌、甲状腺癌、肝癌、胆囊癌、膀胱(尿道上皮)癌、肾癌、皮肤癌、中枢神经系统癌、睾丸癌、上皮癌、头颈部癌的癌细胞。在一些优选的实施方案中,所述癌细胞选自转移性乳腺癌、TNBC、非TNBC、子宫颈癌、子宫内膜癌、肺癌、卵巢 癌、膀胱(尿道上皮)癌、结肠癌、胰腺癌的癌细胞。在一些更优选的实施方案中,所述癌细胞为TNBC、卵巢癌、子宫颈癌、膀胱(尿道上皮)癌或胰腺癌癌细胞(如原位胰腺癌细胞)。
除非另有定义,本文中所用的所有技术术语和科学术语的含义意图与本领域技术人员通常所理解的相同。提及本文中使用的技术意图指在本领域中通常所理解的技术,包括那些对本领域技术人员显而易见的技术的变化或等效技术的替换。虽然相信以下术语对于本领域技术人员很好理解,但仍然阐述以下定义以更好地解释本发明。
本发明中,Y为
Figure PCTCN2022135429-appb-000031
n 1、n 2、n 3、n 4各自独立地选自0、1,且n 1、n 2、n 3和n 4中至少有一个为1。其中,例如,n 1为0,n 2、n 3、n 4为1时,Y为
Figure PCTCN2022135429-appb-000032
再例如,n 2为0,n 1、n 3、n 4为1时,Y为
Figure PCTCN2022135429-appb-000033
或者例如,n 1、n 2为1,n 3、n 4为0时,Y为
Figure PCTCN2022135429-appb-000034
其余类似的定义可以参照前述内容进行理解。
本发明中,术语“氨基酸残基”指的是氨基酸的氨基失去一个氢、羧基失去一个羟基后,剩余的不完整的氨基酸结构,具有氨基端和羰基端。本发明中,L 2为氨基酸残基或由2-10个(优选2-4个)氨基酸残基组成的短肽,其中,2-10个(优选2-4个)氨基酸的种类相互之间可以相同,也可以不同。例如,若L 2为由4个氨基酸残基组成的短肽,且氨基酸选自甘氨酸、苯丙氨酸,则L 2可以为以下短肽或肽残基:甘氨酸-甘氨酸-苯丙氨酸-甘氨酸(Gly-Gly-Phe-Gly),具体可以为
Figure PCTCN2022135429-appb-000035
本发明中,除非以其他方式明确指出,在本文中通篇采用的描述方式“各…独立地选自”和“…各自独立地选自”可以互换,均应做广义理解,其既可以是指在不同基团中,相同或不同的符号之间所表达的具体选项之间互相不影响,也可以表示在相同的基团中,相同或不同的符号之间所表达的具体选项之间互相不影响。
本发明所述“接头”、“接头结构”或“连接子”或“连接单元”是指一端与抗体连接而另一端与药物(药物化合物)相连的化学结构片段或键,也可以连接其它接头后再与药物化合物相连。本发明的接头结构可以通过本领域已知方法合成,也可使用本发明所述的方法进行合成。
本发明所述“抗体-药物偶联物”,即ADC,指靶向部分如抗体或其抗原结合片段通过稳定的连接单元与具有生物活性的药物相连。
本发明中,“药学上可接受的盐”是指相对无毒的本发明的化合物或偶联物的酸加成盐或碱加成盐。所述酸加成盐为本发明的化合物或偶联物与合适的无机酸或者有机酸形成的盐,这些盐可通过使本发明的化合物或偶联物与适宜的有机酸或无机酸在适当的溶剂中进行反应来制备。代表性酸加成盐包括氢溴酸盐、盐酸盐、硫酸盐、硫酸氢盐、亚硫酸盐、乙酸盐、草酸盐、戊酸盐、油酸盐、棕榈酸盐、硬脂酸盐、月硅酸盐、硼酸盐、苯甲酸盐、乳酸盐、硝酸盐、磷酸盐、磷酸氢盐、碳酸盐、碳酸氢盐、甲苯甲酸盐、柠檬酸盐、马来酸盐、富马酸盐、琥珀酸盐、苹果酸盐、抗坏血酸盐、鞣酸盐、扑酸盐、藻酸盐、萘磺酸盐、酒石酸盐、苯甲酸盐、甲磺酸盐、对甲苯磺酸盐、葡萄糖酸盐、乳糖酸盐和月桂基磺酸盐等。所述碱加成盐为本发明的化合物或偶联物与合适的无机碱或者有机碱形成的盐,这些盐可通过使本发明的化合物或偶联物与适宜的无机碱或者有机碱在适当的溶剂中进行反应来制备。代表性碱加成盐包括例如与碱金属、碱土金属、季铵阳离子形成的盐,例如钠盐、锂盐、钾盐、钙盐、镁盐、四甲基季铵盐、四乙基季铵盐等;胺盐,包括与氨(NH3)、伯胺、仲胺或叔胺形成的盐,如甲胺盐、二甲胺盐、三甲胺盐、三乙胺盐、乙胺盐等。
本发明的化合物或偶联物可以存在特定的几何或立体异构体形式,本发明的化合物或偶联物中,手性中心可以存在于药物中,可以存在于接头结构中,还可以存在于抗体及其衍生物中。本发明中,所有的这类化合物,包括顺式和反式异构体、(-)-和(+)-对映体、(R)-和(S)-对映体、非对映异构体、(D)-异构体、(L)-异构体,及其外消旋混合物和其他混合物,例如对映异构体或非对映体富集的混合物,均包括在本发明的范围之内。
可以通过的手性合成或手性试剂或者其他常规技术制备光学活性的(R)-和(S)-异构体以及D和L异构体。如果想得到本发明的化合物或偶联物的一种对映体,可以通过不对称合成或者具有手性助剂的衍生作用来制备,其中将所得非对映体混合物分离,并且辅助基团裂开以提供纯的所需对映异构体。或者,当分子中含有碱性官能团(如氨基)或酸性官能团(如羧基)时,与适当的光学活性的酸或碱形成非对映异构体的盐,然后通过本领域所公知的常规方法进行非对映异构体拆分,然后回收得到纯的对映体。此外,对映异构体和非对映异构体的分离通常是通过使用色谱法完成的,所述色谱法采用手性固定相,并任选地与化学衍生法相结合(例如由胺生成氨基甲酸盐)。
本发明中,本发明的化合物、偶联物、药学上可接受的盐、立体异构体的溶剂合物(例如水合物)也在本发明的范围内。作为适当的溶剂合物,具体而言,可以列举本发 明的化合物或偶联物与丙酮、2-丁醇、2-丙醇、乙醇、乙酸乙酯、四氢呋喃、二乙醚等形成的溶剂合物。还可以列举出水合物或乙醇化物。
本发明中,“治疗”患有疾病或疾病状况的个体表示所述个体的症状部分或全部缓解,或者在治疗后保持不变。因此,治疗包括预防、治疗和/或治愈。“预防”指防止潜在疾病和/或防止症状恶化或疾病发展。治疗或预防还包括所提供的ADC以及本文所提供的药物组合物、药物制剂的任何药学用途。
本公开中,术语“有效剂量”指被给药后会在一定程度上缓解所治疗病症的一种或多种症状的化合物的量。类似地,术语“有效量”指被给药后会在一定程度上抑制癌细胞生长、增殖或迁移的化合物的量。
本发明中,“疗效”表示由个体的治疗所导致的效果,其改变、通常改良或改善疾病或疾病状况的症状,或者治愈疾病或疾病状况。
本发明中,“个体”包括人或非人动物。示例性人个体包括患有疾病(例如本文所述的疾病)的人个体(称为患者)或正常个体。本发明中,“非人动物”包括所有脊椎动物,例如非哺乳动物(例如鸟类、两栖动物、爬行动物)和哺乳动物,例如非人灵长类、家畜和/或驯化动物(例如绵羊、犬、猫、奶牛、猪等)。
应了解所公开的组合物和方法不局限于本文所述和/或所示的特定组合物和方法,且本文所用的术语仅仅出于借助于实例描述特定实施方案的目的,且不意图限制所要求的组合物和方法。
本文通篇,描述提及组合物和使用所述组合物的方法。在本公开描述或要求与组合物有关的特征或实施方案的情况下,这类特征或实施方案同等适用于使用所述组合物的方法。同样,在本公开描述或要求与使用组合物的方法有关的特征或实施方案的情况下,这类特征或实施方案同等适用于所述组合物。
当表述值的范围时,其包括使用所述范围内的任何特定值的实施方案。此外,提及范围内所述的值包括所述范围内的每个值。所有范围都包括其终点且可以组合。当通过利用前面的“约”,将值表述为近似值时,应了解特定值形成另一实施方案。除非上下文另外清楚规定,否则提及特定数值至少包括所述特定值。除非所使用的特定上下文另外规定,否则使用“或”将意指“和/或”。本文中引用的所有参考文献都以引用的方式并入以达成任何目的。在参考文献与本说明书冲突的情况下,将以本说明书为主。
应了解,为清楚起见,在本文中单独实施方案的上下文中描述的所公开组合物和方法的某些特征也可以组合提供于单个实施方案。相反,为简便起见,在单个实施方案的上下文中描述的所公开的组合物和方法的各种特征也可以分开或呈任何子组合提供。
本文中提及的文献均以其整体援引加入本文中。
有益效果:
1、本公开设计的linker制备获得的ADC不仅药效比含有PAB结构的ADC更好,且简化了linker的结构,减少了合成的步骤,降低了成本。
2、本公开linker中将连接马来酰亚胺基与氨基酸残基或短肽的链的长度缩短,例如缩短到3个亚甲基甚至2个亚甲基相连,以及去除了PAB这个疏水的基团后,提高了ADC分子的亲水性,降低了ADC聚体的形成,提高了ADC的稳定性,还能改善其在体内的PK,提高药物在体内的半衰期。
3、本公开意外发现,本公开截短型的Linker构建的ADC能够显著提高抗体与靶点蛋白的亲和力。
4、本公开截短型的Linker构建的ADC的抑制肿瘤(如胰腺癌或结肠癌)的药效提高。
5、本公开的ADC(如ADC-1.1)具有旁杀效应,例如,本公开的ADC(如ADC-1.1)能够在肿瘤细胞内杀伤高表达TROP-2的靶细胞之外,还能对旁边的TROP-2阴性细胞产生一定程度的杀伤作用。
附图说明
图1-1为本发明实施例的ADC-1.1的SEC-HPLC检测图谱。
图1-2为本发明实施例的ADC-1.2的SEC-HPLC检测图谱。
图2-1为本发明实施例的ADC-2.1的SEC-HPLC检测图谱。
图2-2为本发明实施例的ADC-2.2的SEC-HPLC检测图谱。
图3为本发明对照偶联物2(即ADC-5)的SEC-HPLC检测图谱。
图4为本发明对照偶联物ADC-217的SEC-HPLC纯度检测图谱。
图5为采用蛋白疏水作用色谱柱(HIC)检测本发明各ADC的亲疏水性的结果图。
图6为本发明各ADC与TROP蛋白的亲和力检测结果图。
图7为本发明各ADC的抑瘤活性统计曲线图。
图8为本发明各ADC治疗后实验动物体重变化曲线图。
图9为本发明各ADC治疗后实验动物的取瘤拍照图。
图10为本发明ADC-1.1的旁观者效应结果图。
图11为本发明各ADC的抑瘤活性统计曲线图。
图12为本发明各ADC治疗后实验动物体重变化曲线图。
图13为本发明各ADC治疗后实验动物的取瘤拍照图。
具体实施方式
下面将结合附图和实施例对本发明的实施方案进行详细描述,但是,本领域技术人员将理解,下列附图和实施例仅用于说明本发明,而不是对本发明的范围的限定。根据附图和优选实施方案的下列详细描述,本发明的各种目的和有利方面对于本领域技术人员来说将变得显然。以下实施例中,如果没有特别说明,各试剂均可以市购获得。
实施例1 hRS7抗体的制备和检测
1.基因合成、转染和抗体制备
hRS7抗体在CHO细胞产生。含hRS7抗体基因的表达载体分别用常规的分子生物学方法构建,hRS7抗体轻链和重链的氨基酸序列分别如SEQ ID NO:1和SEQ ID NO:2所示,其对应的核苷酸序列分别如SEQ ID NO:3和SEQ ID NO:4所示。将上述两序列插入到同一表达载体中,大量提取制备转染质粒,并转染到CHO-K1细胞(ATCC CCL-61)中,具体转染和抗体制备的过程如下:
(1)细胞培养:CHO-K1细胞悬浮生长于ActiPro(GE HyClone)培养基,于37℃,7%CO2,140rpm,90%相对湿度进行培养;
(2)转染:在进入对数生长期后,取细胞离心,重悬于新鲜的ActiPro培养基,计数并调节细胞密度到1.2×107个/毫升,转500μl细胞悬液到电击杯中,然后加入40μg构建好的质粒,将细胞与质粒混匀,然后用电转方式导入质粒(Bio-rad电转仪);
(3)亚克隆:电转后的细胞用37℃的ActiPro培养基重悬,每孔100μl分装于96孔板。测定细胞上清以测定抗体的表达水平。将表达水平较高的克隆从96孔板转移到24孔板培养,而后再转入6孔板培养,测定细胞的抗体产量和产率,选择表达量最高的4个克隆进行亚克隆,而后转入摇瓶,放在培养箱中继续培养。
2抗体的纯化
收集摇瓶培养的高表达的细胞液,用蛋白A亲和纯化(GE,Mab Select SuRe)和离 子交换纯化(GE,Capto S)。采用SDS-PAGE和SEC-HPLC对纯化后的抗体进行分子量和纯度分析。SDS-PAGE测定结果表明制备的hRS7分子量符合预期,用SEC-HPLC法测得抗体纯度为99.1%。
hRS7抗体轻链氨基酸序列
Figure PCTCN2022135429-appb-000036
hRS7抗体重链氨基酸序列
Figure PCTCN2022135429-appb-000037
hRS7抗体轻链核苷酸序列
Figure PCTCN2022135429-appb-000038
Figure PCTCN2022135429-appb-000039
hRS7抗体重链核苷酸序列
Figure PCTCN2022135429-appb-000040
实施例2制备偶联物
1-1.制备偶联物ADC-1.1
Figure PCTCN2022135429-appb-000041
1.1.1中间体A的合成
Figure PCTCN2022135429-appb-000042
1.1.1.1在A1(60mg,0.16mmol)的DCM(2mL)溶液中加入三氟乙酸(1mL)。将反应液在室温条件下反应3小时。将反应液旋干除去溶剂后得粗品目标产物A2大约42mg左右,产率:98.8%。
Figure PCTCN2022135429-appb-000043
1.1.1.2在A2(42mg,0.15mmol)的NMP(2mL)溶液中加入TEA(31mg,0.31mmol)和MC(3)-NHS(即
Figure PCTCN2022135429-appb-000044
21.14mg,0.18mmol)。将反应液在室温条件下反应16小时。将反应液直接送Pre-HPLC纯化后得目标产物A3大约21mg左右,产率:32.24%。LCMS:[M+1]+=426.4.
Figure PCTCN2022135429-appb-000045
1.1.1.3在A3(10mg,0.023mmol)的NMP(2mL)溶液中加入TEA(4.76mg,0.047mmol)和HATU(13.41mg,0.035mmol)。将反应液在室温条件下反应半小时后加入化合物eribulin(8.6mg,0.012mmol)并于室温条件下搅拌反应3小时。将反应液直接送Pre-HPLC纯化后得目标产物A大约3mg左右,产率:14.03%。LCMS:[M+1]+=1138.8.
1.1.2偶联粗产物ADC-1.1的合成
Figure PCTCN2022135429-appb-000046
实施例1制备的抗体在10mg/mL的pH 7.4PBS溶液中,使用25℃水浴锅,搅拌混合的同时加入2.4倍的物质的量的等体积TCEP溶液,溶液置25℃水浴锅静置反应1.5小时。样品使用25℃水浴锅,接着向抗体溶液中搅拌混合的同时加入8倍的物质的量的化合物A的DMSO溶液(DMSO终浓度10%),25℃摇床摇动反应90分钟,最后样品搅拌混合的同时加入8倍的物质的量的半胱氨酸,置于25℃水浴锅静置反应10min,得到偶联产物ADC-1.1。
1.1.3偶联粗产物ADC-1.1的检测
偶联反应粗产品用SEC检测,
SEC色谱条件如下:
色谱柱型号:TSKgel G4000SWxl 7.8mmI.D.*30cm,8μm
检测器波长:280nm/220nm
柱温:30℃
流速:1mL/min
洗脱方式:等度洗脱
进样体积:10μL
运行时间:55min
1.1.4偶联反应产品纯化:
通过PD-10脱盐柱(填料:sephadex G 25)脱盐纯化后,得偶联物,偶联物再换液到MES 25mM pH5.5海藻糖5%的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-1.1见图1-1。
1.1.5 DAR的测定
ADC-1.1和单抗溶液中加入等体积50mM DTT溶液,涡旋混匀,37℃水浴30min。还原后的ADC和抗体用RP-HPLC分析偶联和未偶联的抗体轻链和重链。通过分析其组成确定DAR值为3.9。
1-2制备偶联物ADC-1.2
1.2.1偶联粗产物ADC-1.2的合成
Figure PCTCN2022135429-appb-000047
实施例1制备的抗体在10mg/mL的pH 7.3PBS溶液中,使用22℃水浴锅,搅拌混合的同时加入2.15倍的物质的量的等体积TCEP溶液,溶液置22℃水浴锅静置反应2小时。样品使用22℃水浴锅,接着向抗体溶液中搅拌混合的同时加入5.5倍的物质的量的化合物A的DMSO溶液(DMSO终浓度10%),22℃摇床摇动反应60分钟,最后样品搅拌混合的同时加入8倍的物质的量的半胱氨酸,置于22℃水浴锅静置反应10min,得到偶联产物ADC-1.2。
1.2.2偶联粗产物ADC-1.2的检测
偶联反应粗产品用SEC检测,
SEC色谱条件如下:
色谱柱型号:TSKgel G4000SWxl 7.8mmI.D.*30cm,8μm
检测器波长:280nm/220nm
柱温:30℃
流速:1mL/min
洗脱方式:等度洗脱
进样体积:10μL
运行时间:55min
1.2.3偶联反应产品纯化:
通过PD-10脱盐柱(填料:sephadex G 25)脱盐纯化后,得偶联物,偶联物再换液到His 20mM pH6.0蔗糖5%的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-1.2见图1-2。
1.2.4 DAR的测定
ADC-1.2和单抗溶液中加入等体积50mM DTT溶液,涡旋混匀,37℃水浴30min。还原后的ADC和抗体用RP-HPLC分析偶联和未偶联的抗体轻链和重链。通过分析其组成确定DAR值为3.5。
2-1.制备偶联物ADC-2.1
Figure PCTCN2022135429-appb-000048
2.1.1中间体B的制备
Figure PCTCN2022135429-appb-000049
2.1.1.1将B1(50mg),B2(35mg)与吡啶(40μL)溶解在NMP(1.0mL)中,反应液在室温条件下搅拌过夜,Pre-HPLC纯化,冻干,得目标化合物B3约10mg.LCMS:[M+1]+=338.4
Figure PCTCN2022135429-appb-000050
2.1.1.2Mal-Val-Ala-eribulin的合成
将B3(10mg),eribulin(12mg),HATU(15mg)与吡啶(20μL)溶解在NMP(0.5mL)中,在室温条件下搅拌过夜,Pre-HPLC纯化后的目标产物B 8mg。LCMS:[M+1]+=1051.9
2.1.2偶联粗产物ADC-2.1的合成
Figure PCTCN2022135429-appb-000051
实施例1制备的抗体在10mg/mL的pH 7.4PBS溶液中,使用25℃水浴锅,搅拌混 合的同时加入2.4倍的物质的量的等体积TCEP溶液,溶液置25℃水浴锅静置反应1.5小时。样品使用25℃水浴锅,接着向抗体溶液中搅拌混合的同时加入8倍的物质的量的化合物B的DMSO溶液(DMSO终浓度10%),25℃摇床摇动反应90分钟,最后样品搅拌混合的同时加入8倍的物质的量的半胱氨酸,置于25℃水浴锅静置反应10min,得到偶联产物ADC-2.1。
2.1.3偶联粗产物ADC-2.1的检测
偶联反应粗产品用SEC检测,
SEC色谱条件如下:
色谱柱型号:TSKgel G4000SWxl 7.8mmI.D.*30cm,8μm
检测器波长:280nm/220nm
柱温:30℃
流速:1mL/min
洗脱方式:等度洗脱
进样体积:10μL
运行时间:55min
2.1.4偶联反应产品纯化:
通过PD-10脱盐柱(填料:sephadex G 25)脱盐纯化后,得偶联物,偶联物再换液到MES 25mM pH5.5海藻糖5%的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-2.1见图2-1。
2.1.5DAR的测定
ADC-2.1和单抗溶液中加入等体积50mM DTT溶液,涡旋混匀,37℃水浴30min。还原后的ADC和抗体用RP-HPLC分析偶联和未偶联的抗体轻链和重链。通过分析其组成确定DAR值为4.1。
2-2.制备偶联物ADC-2.2
2.2.1偶联粗产物ADC-2.2的合成
Figure PCTCN2022135429-appb-000052
实施例1制备的抗体在10mg/mL的pH 7.3PBS溶液中,使用22℃水浴锅,搅拌混合的同时加入2.15倍的物质的量的等体积TCEP溶液,溶液置22℃水浴锅静置反应2小时。样品使用22℃水浴锅,接着向抗体溶液中搅拌混合的同时加入5.5倍的物质的量的化合物B的DMSO溶液(DMSO终浓度10%),22℃摇床摇动反应60分钟,最后样品搅拌混合的同时加入8倍的物质的量的半胱氨酸,置于22℃水浴锅静置反应10min,得到偶联产物ADC-2.2。
2.2.2偶联粗产物ADC-2.2的检测
偶联反应粗产品用SEC检测,
SEC色谱条件如下:
色谱柱型号:TSKgel G4000SWxl 7.8mmI.D.*30cm,8μm
检测器波长:280nm/220nm
柱温:30℃
流速:1mL/min
洗脱方式:等度洗脱
进样体积:10μL
运行时间:55min
2.2.3偶联反应产品纯化:
通过PD-10脱盐柱(填料:sephadex G 25)脱盐纯化后,得偶联物,偶联物再换液到His 20mM pH6.0蔗糖5%的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-2.2见图2-2。
2.2.4 DAR的测定
ADC-2.2和单抗溶液中加入等体积50mM DTT溶液,涡旋混匀,37℃水浴30min。还原后的ADC和抗体用RP-HPLC分析偶联和未偶联的抗体轻链和重链。通过分析其组成确定DAR值为3.5。
3.制备对照偶联物1即ADC-4
Figure PCTCN2022135429-appb-000053
3.1中间体C的制备
Figure PCTCN2022135429-appb-000054
将C1(25mg)与eribulin A2(25mg)溶解在DMF(3mL)中,室温条件下加入DIEA(8.7mg),反应液在室温条件下搅拌2小时,HPLC显示有新峰生成,LCMS显示有目标产物生成,Pre-HPLC纯化冻干,得目标化合物约C 21mg.LCMS:[M+1]+=1329.4。
3.2偶联粗产物ADC-4的合成
Figure PCTCN2022135429-appb-000055
实施例1制备的抗体在10mg/mL的pH 7.4PBS溶液中置于冰水浴,当蛋白溶液温度达到4℃时,搅拌混合的同时加入3.5倍的物质的量的等体积TCEP溶液,溶液置25℃水浴锅静置反应1小时。反应液使用25℃水浴锅,当温度降低至25℃时, 接着向抗体溶液中搅拌混合的同时加入7倍的物质的量的化合物C的40%DMSO溶液,25℃摇床摇动反应90分钟,最后反应液搅拌混合的同时加入8倍的物质的量的半胱氨酸,置于25℃水浴锅静置反应10min,得到偶联产物ADC-4。
偶联产物的检测、纯化和DAR值测定方法同ADC-1.1,获得的ADC-4的DAR值为3.8。
4.制备对照偶联物2即ADC-5
Figure PCTCN2022135429-appb-000056
4.1中间体C的制备
Figure PCTCN2022135429-appb-000057
将C1(30mg),eribulin A2(30mg)与HATU溶解在DMF(0.5mL)中,室温条件下加入DIEA(21微升),反应液在室温条件下搅拌2小时,HPLC显示有新峰生成,LCMS显示有目标产物生成,Pre-HPLC纯化冻干,得目标化合物约C 25mg.LCMS:[M+1]+=1241.6。
4.2偶联粗产物ADC-5的合成
Figure PCTCN2022135429-appb-000058
实施例1制备的抗体在5mg/mL的pH 7.4PBS溶液中,搅拌混合的同时加入7倍的物质的量的TCEP溶液,溶液置37℃水浴锅静置反应1.5小时。反应液使用25℃水浴锅,当温度降低至25℃时,接着向抗体溶液中搅拌混合的同时加入16倍的物质的量的化合物C的40%DMSO溶液,25℃摇床摇动反应90分钟,最后反应液搅拌混合的同时加入16倍的物质的量的半胱氨酸,置于25℃水浴锅静置反应10min,得到偶联产物ADC-5。
4.3偶联粗产物ADC-5的检测
偶联反应粗产品用SEC检测,
SEC色谱条件如下:
色谱柱型号:TSKgel G4000SWxl 7.8mmI.D.*30cm,8μm
检测器波长:280nm/220nm
柱温:30℃
流速:1mL/min
洗脱方式:等度洗脱
进样体积:10μL
运行时间:55min
4.4偶联反应产品纯化:
通过PD-10脱盐柱(填料:sephadex G 25)脱盐纯化后,得偶联物,偶联物再换液到MES 25mM pH5.5海藻糖5%的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-5的SEC-HPLC结果见图3。
4.5 DAR的测定
ADC-5和单抗溶液中加入等体积50mM二硫苏糖醇(DTT)溶液,涡旋混匀,37℃水浴30min。还原后的ADC和抗体用RP-HPLC分析偶联和未偶联的抗体轻链和重链。通过分析其组成确定DAR值为7.2。
5.制备对照偶联物ADC-217
Figure PCTCN2022135429-appb-000059
5.1 Fmoc-VC-PABC-eribulin的合成
Figure PCTCN2022135429-appb-000060
在A1’(50mg,0.065mmol)的NMP(3mL)溶液中加入eribulin(19.04mg,0.026mmol)和DIEA(25.28mg,0.20mmol)。将反应液在室温条件下反应3小时。将反应液经Prep-HPLC纯化冻干后得目标产物A2’大约71mg左右。LCMS:[M+1]+=1357.1.
5.2 VC-PABC-eribulin的合成
Figure PCTCN2022135429-appb-000061
在A2’(71mg,0.052mmol)的THF(3mL)溶液中加入二乙胺(38.25mg,0.52mmol)。将反应液在室温条件下搅拌反应3小时。将反应液直接送Pre-HPLC纯化冻干后得目标产物A3’大约48mg左右。LCMS:[M+1]+=1135.4.
5.3 MP-VC-PABC-eribulin的合成
Figure PCTCN2022135429-appb-000062
在A3’(20mg,0.018mmol)的NMP(2mL)溶液中加入DIEA(6.83mg,0.053mmol)和MP-Osu(4.7mg,0.017mmol)。将反应液于室温条件下搅拌反应3小时。将反应液直接送Pre-HPLC纯化后得目标产物A’大约16mg左右。LCMS:[M+1]+=1386.4.
5.4偶联抗体:
Figure PCTCN2022135429-appb-000063
将实施例1制备的TROP-2抗体(20mM醋酸-醋酸钠,PH6.1)调节PH7.5,用50mM EPPS 5mM EDTA PH7.5buffer稀释。5.3节制备的产物使用DMSO溶解。蛋白溶液(浓度10mg/ml)(PH 7.5)置于25℃水浴锅水浴,当蛋白溶液温度达到25℃时,搅拌混合的同时加入1/10体积TCEP(溶于水)(2.2989eq),溶液置25℃摇床30rmp摇动反应90min。样品使用25℃水浴锅水浴,涡旋混合的同时加入5%DMSO,再加入5%化合物A’(7.5eq),总DMSO10%,25℃摇床30rmp摇动反应90min。样品涡旋混合的同时加入Cys(溶于水)(8eq),置于25℃摇床30rmp摇动反应10min。样品5000rmp/min离心5min,使用AKTA脱盐换液于25mM MES,PH5.5的溶液中,SEC检测,小分子已经完全除掉。纯化后的ADC-217见图4。
键合率DAR通过RP-HPLC估算为3.80,检测方法同ADC-1.1。
实施例3检测各ADC的亲水性
采用蛋白疏水作用色谱柱(HIC)检测实施例2中制备的各ADC的亲疏水性,具体实验条件如下:
取实施例2中制备的各ADC样本30ul进样到HIC(TSKgel Butyl-NPR 4.6mmI.D.×10cm,2.5um),样品盘温度4℃,柱温40℃,流动相A:1.5M Na 2SO 4&0.025M PB pH7.4&2%IPA(异丙醇)流动相B:0.025M PB pH7.4&11%IPA(异丙醇),洗脱梯度如下表:
Figure PCTCN2022135429-appb-000064
结果如图5所示,箭头指示的位置为各ADC出峰位置,峰位置越靠左说明亲水性越好。比较实施例2的4个ADC的亲水性,能看出亲水性的排序为ADC-1.1≈ADC-2.1>ADC-5>ADC-4。因此,本公开的ADC(如ADC-1.1或ADC-2.1)的L-D与MORAb-202的L-D相比具有更好的亲水性。
实施例4 ADC与TROP蛋白的亲和力实验
检测实施例2制备的各ADC与TROP蛋白的亲和力,以比较不同结构的ADC亲和力差别。
检测方法:
包被Trop2蛋白:将Trop2蛋白(北京义翘神州生物技术有限公司)用包被液(0.15M Na 2CO 3和0.35M NaHCO 3)稀释至0.5μg/mL,混匀后加入到酶标板中,100μL/孔,覆上封板膜,放至4℃冰箱,过夜。
洗板:拿出酶标板洗板:用1*PBST洗3次。
封闭:取封闭液(1%BSA),分别加入封闭液200μL/孔,覆上封板膜,250rpm混匀25℃孵育1h。
洗板:用1*PBST洗3次。
孵育抗体:用1%BSA将实施例2制备的ADC样品1、2、4、5,和实施例1制备的TROP-2抗体稀释成2000ng/mL后,三倍梯度稀释12个浓度,100μL/孔加至酶标板,覆上封板膜,250rpm混匀25℃孵育1h。
洗板:将酶标板用1*PBST洗3次。
孵育二抗:将Anti-Human HRP(Boster)用1%BSA稀释成1:10000,100μL/孔加至酶标板,覆上封板膜,250rpm混匀25℃孵育1h。
洗板:将酶标板用1*PBST洗3次。
显色:加入TMB(湖州英创)显色底物:每孔100ul加入酶标板,250rpm混匀25℃放置15分钟。
终止读数:加入终止液:每孔加入1M H 2SO 4 100ul,酶标仪OD450nm读数。
检测结果如图6,以实施例2制备的ADC的EC50与TROP-2裸抗的EC50的比值为纵坐标,比值越大说明亲和力越小,比较不同结构ADC产品的亲和力,结果表明ADC-1.1和ADC-2.1的亲和力最好,而MORAb-202结构类似的ADC与TROP-2的亲和力最差,即本公开的L-D结构偶联获得的ADC能够更好的保持抗体的亲和力。
实施例5实施例2制备的ADC对胰腺癌的抑瘤效果
1.实验方法
1.1细胞培养
BxPC-3细胞(人原位胰腺腺癌细胞,ATCC CRL-1687)体外单层培养,培养条件为1640培养基中加10%热灭活胎牛血清并加琼脂,于37℃、含5%CO 2空气的培养箱中培养。一周两次用0.25%胰酶进行消化处理传代。当细胞呈指数生长期时,收取细胞,计数,接种。
1.2肿瘤细胞接种及瘤块传代
将5.0×10 6BxPC-3肿瘤细胞悬浮于0.1ml PBS与Matrigel混合物(1:1),接种于5只裸鼠右侧肩胛处(P1代)。待肿瘤长至500-800mm 3时,将荷瘤小鼠用CO 2麻醉处死,取出瘤块,去除周围坏死的组织,将瘤块切成20-30mm 3的小瘤块,接种到新的一批裸鼠(P2代)。
1.3瘤块接种及分组给药
本试验使用P6代肿瘤组织进行受试品的抗肿瘤活性评价。待P5代肿瘤长至500-800mm 3时,将荷瘤小鼠用CO 2麻醉处死,取出瘤块,去除周围坏死的组织,将瘤块切 成20-30mm 3的小瘤块,接种到正式实验用鼠的右侧肩胛处,一共接种40只鼠。瘤块接种18天后肿瘤平均体积达到约135mm 3时,剔除瘤体积过小或过大的小鼠,将剩余的25只小鼠根据瘤体积随机分组并开始给药。本实验仅给药一次,分组当天一次给药后不再给药。给药方案见下表。
Figure PCTCN2022135429-appb-000065
1.5实验观察和数据收集
肿瘤细胞接种后,除了观察肿瘤生长情况,还对药物治疗对动物行为的影响进行监测:实验动物的活动性,摄食和饮水,体重变化(体重和瘤体积每周测量2次),眼睛、被毛及其它异常情况。实验过程中观察到的临床症状均记录在原始数据中。肿瘤体积计算方法为:肿瘤体积(mm 3)=1/2×(a×b 2)(其中a表示长径,b表示短径)。
当单只动物的体重下降超过15%时(BWL>15%),给予相应单只动物停药处理,体重下降恢复到10%以内,恢复给药。当单只小鼠体重下降>20%,按照动物福利对其实施安乐死。
1.6疗效评价标准
相对肿瘤增殖率,T/C%,即在某一时间点,治疗组和对照组的相对肿瘤体积或瘤重的百分比值。计算公式如下:T/C%=T RTV/C RTV×100%(T RTV:治疗组平均RTV;C RTV:溶媒对照组平均RTV;RTV=V t/V 0,V 0为分组时该动物的瘤体积,V t为治疗后该动物的瘤体积);或T/C%=T TW/C TW×100%(T TW:治疗组实验终结时平均瘤重;C TW:溶媒对照组实验终结时平均瘤重)。
1.7统计分析
本实验用one-way ANOVA进行各组间肿瘤均值的比较。方差齐性分析得出F值有显著性差异,在ANOVA分析之后用Dunnet’s T3(方差不齐)法再进行多重比较。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
2.实验结果
实验结果如图7-9所示,图7为抑瘤活性统计曲线,图8为体重变化曲线,图9为取瘤拍照图。
从结果来看,ADC-1.1、ADC-2.1比ADC-4、ADC-5具有更优异的抑瘤效果,其中ADC-1.1和ADC-2.1仅一次用药即可完全让肿瘤消退,且安全性好。
实施例6 ADC的旁杀效应---检测实施例2制备的ADC(如ADC-1.1)的旁观者效应
实验设计:设置单细胞培养组(A549-Luc)和混合细胞培养组(BxPC-3和A549-Luc)。实施例2制备的ADC-1.1作为检测组,实施例1制备的hRS7抗体作为对照组,设置不加药物培养A549-Luc细胞的空白组,另外设置溶剂对照孔。BxPC-3细胞高表达TROP-2,A549细胞低表达TROP-2。前者可更大程度内吞ADC-1.1,后者则内吞效率很低。而在A549上加载荧光标记,若本公开的ADC-1.1具有旁杀效应,则在混合细胞培养组可通过裂解BxPC-3细胞释放出艾日布林小分子毒素,从而杀伤A549细胞,从而使细胞中荧光淬灭,从酶标仪读取490nm的吸光值可计算旁杀效应的细胞抑制率。
实验方法:
1、选择TROP-2高表达的细胞BxPC-3(人原位胰腺腺癌细胞,ATCC CRL-1687)体外单层培养,将生长良好的细胞用胰蛋白酶进行消化、离心,通过细胞计数,用RPMI1640/F12-K+10%FBS重悬,将细胞密度调整为8×10 4cells/ml。
2、选择TROP-2低表达的荧光标记细胞A549-Luc(人非小细胞肺癌细胞,ATCC CCL-185细胞)体外单层培养,将生长良好的细胞用胰蛋白酶进行消化、离心,通过细胞计数,用RPMI1640/F12-K+10%FBS重悬,密度调整为1×10 4cells/ml及2×10 4cells/ml。将1×10 4cells/ml的细胞悬液接种于每板单细胞培养组,每孔100μl,设置3孔重复。
3、共培养:将2×10 4cells/ml浓度的A549-Luc细胞悬液,与8×10 4cells/ml浓度的BxPC-3的细胞按1:1比例混合,混匀后接种于各培养板的共培养孔,每孔接种100μl,设置3孔重复。
4、加药:将细胞置于二氧化碳培养箱中培养24h。次日,根据实验设计,将实施例2制备的ADC-1.1样品、实施例1制备的hRS7抗体进行相应的稀释,向细胞中加入100μL/well样品,放入到二氧化碳培养箱中继续培养96h。
5、培养结束后向细胞中加入100μl/well的ONE-GLO检测试剂,避光室温震荡3分 钟,利用Molecular SpectraMax M5型多功能酶标仪读取490nm处的吸光度。以空白组细胞活性为100%。利用样品信号值计算抑制率(%):杀伤率(%)=(1-样品孔信号值/溶剂对照孔信号平均值)×100。所得数据在GraphPad Prism9上进行作图分析查看旁杀效率。
检测结果如图10,ADC-1.1加入到细胞后,混合细胞产生了30%的细胞抑制率,而单细胞则未产生细胞抑制。TROP-2裸抗hRS7对混合细胞也有7%的抑制率,但没有ADC-1.1的抑制率高,该结果表明,ADC-1.1具有旁杀效应,能够在肿瘤细胞内杀伤高表达TROP-2的靶细胞之外,还能对旁边的TROP-2阴性细胞产生一定程度的杀伤作用。
实施例7实施例2制备的ADC对结肠癌的抑瘤效果
1.实验方法
1.1细胞培养
Colo205细胞(人结肠癌细胞,ATCC CCL-222)体外单层培养,培养条件为1640培养基中加10%热灭活胎牛血清并加琼脂,于37℃、含5%CO 2空气的培养箱中培养。一周两次用0.25%胰酶进行消化处理传代。当细胞呈指数生长期时,收取细胞,计数,接种。
1.2肿瘤细胞接种及瘤块传代
将5.0×10 6Colo205肿瘤细胞悬浮于0.1ml PBS与Matrigel混合物(1:1),接种于5只裸鼠右侧肩胛处(P1代)。待肿瘤长至500-800mm 3时,将荷瘤小鼠用CO 2麻醉处死,取出瘤块,去除周围坏死的组织,将瘤块切成20-30mm 3的小瘤块,接种到新的一批裸鼠(P2代)。
1.3瘤块接种及分组给药
本试验使用P6代肿瘤组织进行受试品的抗肿瘤活性评价。待P5代肿瘤长至500-800mm 3时,将荷瘤小鼠用CO 2麻醉处死,取出瘤块,去除周围坏死的组织,将瘤块切成20-30mm 3的小瘤块,接种到正式实验用鼠的右侧肩胛处,一共接种50只鼠。瘤块接种7天后肿瘤,选择肿瘤大小适中的30只小鼠根据瘤体积随机分组并开始给药。给药方案如下:
Figure PCTCN2022135429-appb-000066
Figure PCTCN2022135429-appb-000067
注:“仅一次”指分组当天一次给药后不再给药。
1.4实验观察和数据收集
肿瘤细胞接种后,除了观察肿瘤生长情况,还对药物治疗对动物行为的影响进行监测:实验动物的活动性,摄食和饮水,体重变化(体重每周测量2次),眼睛、被毛及其它异常情况。实验过程中观察到的临床症状均记录在原始数据中。肿瘤体积计算方法为:肿瘤体积(mm 3)=1/2×(a×b 2)(其中a表示长径,b表示短径)。
当单只动物的体重下降超过15%时(BWL>15%),给予相应单只动物停药处理,体重下降恢复到10%以内,恢复给药。当单只小鼠体重下降>20%,按照动物福利对其实施安乐死。
1.5疗效评价标准
相对肿瘤增殖率,T/C%,即在某一时间点,治疗组和对照组的相对肿瘤体积或瘤重的百分比值。计算公式如下:T/C%=T RTV/C RTV×100%(T RTV:治疗组平均RTV;C RTV:溶媒对照组平均RTV;RTV=V t/V 0,V 0为分组时该动物的瘤体积,V t为治疗后该动物的瘤体积);或T/C%=T TW/C TW×100%(T TW:治疗组实验终结时平均瘤重;C TW:溶媒对照组实验终结时平均瘤重)。
1.6实验终点
给药4周后,所有小鼠取肿瘤,并称重、拍照。
1.7统计分析
本实验用one-way ANOVA进行各组间肿瘤均值的比较。方差齐性分析得出F值有显著性差异,在ANOVA分析之后用Dunnet’s T3(方差不齐)法再进行多重比较。用SPSS 17.0进行所有数据分析。p<0.05认为有显著性差异。
2.实验结果
实验结果如下表和图11-13所示,图11为抑瘤活性统计曲线,图12为体重变化曲线,图13为取瘤拍照图。由结果来看,抑瘤效果ADC-1.2≥ADC-1.1>ADC-2.1>ADC-2.2>>ADC-217(对照)。其中,ADC-1.2组所有动物肿瘤完全消退。
Figure PCTCN2022135429-appb-000068

Claims (10)

  1. 式(I)所示化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,
    L 1-L 2-D
    (I)
    其中:
    L 1
    Figure PCTCN2022135429-appb-100001
    Y为
    Figure PCTCN2022135429-appb-100002
    优选地,Y为
    Figure PCTCN2022135429-appb-100003
    X 1、X 2、X 3、X 4各自独立地选自CH 2、NH、O、S,且X 1、X 2、X 3和X 4中至少有一个为CH 2
    优选地,X 1、X 2、X 3、X 4均为CH 2
    n 1、n 2、n 3、n 4各自独立地选自0、1,且n 1、n 2、n 3和n 4中至少有一个为1;
    优选地,n 1为0,n 2、n 3、n 4各自独立地选自0、1,且n 2、n 3和n 4中至少有一个为1;
    更优选地,n 1、n 2为1,n 3、n 4为0;
    n选自1、2、3、4;
    优选地,n选自1、2、3;
    更优选地,n为2;
    L 2选自氨基酸残基或由2-10个氨基酸残基组成的短肽;
    优选地,L 2为由2-4个氨基酸残基组成的短肽,优选地,所述氨基酸选自甘氨酸、苯丙氨酸、缬氨酸、瓜氨酸、丙氨酸,更优选地,所述氨基酸选自缬氨酸、瓜氨酸、丙氨酸;
    更优选地,L 2选自甘氨酸-甘氨酸-苯丙氨酸-甘氨酸、缬氨酸-瓜氨酸、缬氨酸-丙氨酸,优选选自缬氨酸-瓜氨酸、缬氨酸-丙氨酸;
    最优选地,L 2选自
    Figure PCTCN2022135429-appb-100004
    Figure PCTCN2022135429-appb-100005
    优选选自
    Figure PCTCN2022135429-appb-100006
    D为与L 2通过化学键相连的药物,所述药物优选为艾日布林或其衍生物;
    优选地,D为
    Figure PCTCN2022135429-appb-100007
  2. 权利要求1所述的化合物、其药学上可接受的盐、其立体异构体,或其溶剂合物、其中,所述化合物选自:
    Figure PCTCN2022135429-appb-100008
    Figure PCTCN2022135429-appb-100009
  3. 式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,
    Ab-(L 1-L 2-D) p
    (II)
    其中:
    Ab为抗体或其抗原结合片段;
    L 1、L 2、D如权利要求1所限定;
    p为1-20之间的任意数值;
    优选地,p为1-10之间的任意数值;
    更优选地,p为3-5之间的任意数值;
    最优选地,p为3.5-4.5之间的任意数值,例如,p为3.5、3.9或4.1。
  4. 权利要求3所述的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,其中,所述抗体选自抗TROP-2抗体、抗her2抗体、抗her3抗体、抗claudin 18.2抗体、抗ROR-1抗体、抗dll-3抗体、抗muc1抗体、抗muc-17抗体;优选地,所述抗体为鼠源抗体、嵌合抗体、人源化抗体;优选地,所述人源化抗体是全人源抗体;
    或者,所述抗原结合片段选自Fab、Fab'、F(ab') 2、单链Fv(scFv)、Fv和dsFv;
    优选地,所述抗体为抗TROP-2抗体;
    更优选地,所述抗体为抗TROP-2抗体,且所述抗Trop-2抗体的轻链可变区的互补决定区(CDR)包括由KASQDVSIAVA氨基酸序列组成的CDR1,由SASYRYT氨基酸序列组成的CDR2,和由QQHYITPLT氨基酸序列组成的CDR3;重链可变区的CDR包括由NYGMN氨基酸序列组成的CDR1,由WINTYTGEPTYTDDFKG氨基酸序列组成的CDR2,和由GGFGSSYWYFDV氨基酸序列组成的CDR3;优选地,所述抗Trop-2 抗体的轻链及重链的氨基酸序列分别如SEQ ID NO:1和SEQ ID NO:2所示;优选地,所述抗Trop-2抗体的轻链和重链的编码核苷酸序列分别如SEQ ID NO:3和SEQ ID NO:4所示。
  5. 权利要求3-4任一项所述的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,其中,所述抗体药物偶联物选自以下:
    Figure PCTCN2022135429-appb-100010
  6. 制备权利要求1-2任一项所述的式(I)所示的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物的方法,其包括:
    将L 1-L 2-OH与艾日布林进行酰胺化反应,获得式(I)所示的化合物、其药学上可 接受的盐、其立体异构体、或其溶剂合物,其中,L 1、L 2如权利要求1所限定。
  7. 制备权利要求3-5任一项所述的式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物的方法,其包括:
    将权利要求1-2任一项所述的式(I)所示的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物与权利要求3-4任一项所限定的Ab反应,获得式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物;
    优选地,所述权利要求1-2任一项所述的式(I)所示的化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物是通过权利要求6所述的方法制备得到的。
  8. 药物组合物,其包含权利要求1-2任一项所述的式(I)所示化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者包含权利要求3-5任一项所述的式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物;任选地,还包含一种或多种药用辅料,例如载体和/或赋形剂。
  9. 权利要求1-2任一项所述的式(I)所示化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者权利要求3-5任一项所述的式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者权利要求8所述的药物组合物在制备用于预防和/或治疗与细胞活动异常相关的疾病(例如癌症疾病)的药物中的用途;
    优选地,所述癌症为原位癌或转移癌;
    优选地,所述癌症选自转移性乳腺癌、非小细胞肺癌、伯基特淋巴瘤(Burkitt lymphoma)、霍奇金氏淋巴瘤、急性骨髓性白血病、慢性骨髓性白血病、急性淋巴细胞性白血病、慢性淋巴细、皮肤癌、口腔癌、食道癌、胃肠道癌、肺道癌、肺癌、胃癌、结肠癌、直肠癌、三阴性乳腺癌(TNBC)、卵巢癌、前列腺癌、子宫癌、子宫内膜癌、子宫颈癌、膀胱癌、胰腺癌、骨癌、脑癌、结缔组织癌、甲状腺癌、肝癌、胆囊癌、膀胱(尿道上皮)癌、肾癌、皮肤癌、中枢神经系统癌、睾丸癌、上皮癌、头颈部癌;
    更优选地,所述癌症选自转移性乳腺癌、TNBC、非TNBC、子宫颈癌、子宫内膜癌、肺癌、卵巢癌、膀胱(尿道上皮)癌、结肠癌、胰腺癌;
    最优选地,所述癌症为TNBC、卵巢癌、子宫颈癌、膀胱(尿道上皮)癌或胰腺癌(如原位胰腺癌)。
  10. 权利要求1-2任一项所述的式(I)所示化合物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者权利要求3-5任一项所述的式(II)所示的抗体药物偶联物、其药学上可接受的盐、其立体异构体、或其溶剂合物,或者权利要求8所述的药物组合物在制备用于抑制癌细胞生长、增殖或迁移的试剂中的用途;
    优选地,所述癌细胞为原位癌细胞或转移癌细胞;
    优选地,所述癌细胞选自转移性乳腺癌、非小细胞肺癌、伯基特淋巴瘤(Burkitt lymphoma)、霍奇金氏淋巴瘤、急性骨髓性白血病、慢性骨髓性白血病、急性淋巴细胞性白血病、慢性淋巴细、皮肤癌、口腔癌、食道癌、胃肠道癌、肺道癌、肺癌、胃癌、结肠癌、直肠癌、三阴性乳腺癌(TNBC)、卵巢癌、前列腺癌、子宫癌、子宫内膜癌、子宫颈癌、膀胱癌、胰腺癌、骨癌、脑癌、结缔组织癌、甲状腺癌、肝癌、胆囊癌、膀胱(尿道上皮)癌、肾癌、皮肤癌、中枢神经系统癌、睾丸癌、上皮癌、头颈部癌的癌细胞;
    更优选地,所述癌细胞选自转移性乳腺癌、TNBC、非TNBC、子宫颈癌、子宫内膜癌、肺癌、卵巢癌、膀胱(尿道上皮)癌、结肠癌、胰腺癌的癌细胞;
    最优选地,所述癌细胞为TNBC、卵巢癌、子宫颈癌、膀胱(尿道上皮)癌或胰腺癌癌细胞(如原位胰腺癌细胞)。
PCT/CN2022/135429 2021-12-09 2022-11-30 一种亲和力改善的抗体-药物偶联物、其制备方法及应用 WO2023103854A1 (zh)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202111496135 2021-12-09
CN202111496135.8 2021-12-09

Publications (1)

Publication Number Publication Date
WO2023103854A1 true WO2023103854A1 (zh) 2023-06-15

Family

ID=86729627

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/135429 WO2023103854A1 (zh) 2021-12-09 2022-11-30 一种亲和力改善的抗体-药物偶联物、其制备方法及应用

Country Status (1)

Country Link
WO (1) WO2023103854A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080131363A1 (en) * 2002-03-01 2008-06-05 Immunomedics, Inc. Rs7 antibodies
WO2017151979A1 (en) * 2016-03-02 2017-09-08 Eisai Inc. Eribulin-based antibody-drug conjugates and methods of use
WO2020259258A1 (zh) * 2019-06-28 2020-12-30 上海复旦张江生物医药股份有限公司 一种抗体偶联药物、其中间体、制备方法及应用
WO2021132166A1 (ja) * 2019-12-23 2021-07-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 エリブリンをベースとする抗体-薬物コンジュゲートの製造方法
WO2021148003A1 (zh) * 2020-01-22 2021-07-29 上海森辉医药有限公司 艾日布林衍生物的药物偶联物、其制备方法及其在医药上的应用

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20080131363A1 (en) * 2002-03-01 2008-06-05 Immunomedics, Inc. Rs7 antibodies
WO2017151979A1 (en) * 2016-03-02 2017-09-08 Eisai Inc. Eribulin-based antibody-drug conjugates and methods of use
WO2020259258A1 (zh) * 2019-06-28 2020-12-30 上海复旦张江生物医药股份有限公司 一种抗体偶联药物、其中间体、制备方法及应用
WO2021132166A1 (ja) * 2019-12-23 2021-07-01 エーザイ・アール・アンド・ディー・マネジメント株式会社 エリブリンをベースとする抗体-薬物コンジュゲートの製造方法
WO2021148003A1 (zh) * 2020-01-22 2021-07-29 上海森辉医药有限公司 艾日布林衍生物的药物偶联物、其制备方法及其在医药上的应用

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
CHENG XIN, LI JING, TANAKA KEIGO, MAJUMDER UTPAL, MILINICHIK ANDREW Z., VERDI ARIELLE C., MADDAGE CHRISTOPHER J., RYBINSKI KATHERI: "MORAb-202, an Antibody–Drug Conjugate Utilizing Humanized Anti-human FRα Farletuzumab and the Microtubule-targeting Agent Eribulin, has Potent Antitumor Activity", MOLECULAR CANCER THERAPEUTICS, AMERICAN ASSOCIATION FOR CANCER RESEARCH, US, vol. 17, no. 12, 1 December 2018 (2018-12-01), US , pages 2665 - 2675, XP055831595, ISSN: 1535-7163, DOI: 10.1158/1535-7163.MCT-17-1215 *

Similar Documents

Publication Publication Date Title
RU2758234C2 (ru) ЛЕЧЕНИЕ ТРИЖДЫ НЕГАТИВНОГО РАКА МОЛОЧНОЙ ЖЕЛЕЗЫ, ХАРАКТЕРИЗУЮЩЕГОСЯ ЭКСПРЕССИЕЙ Trop-2, С ПОМОЩЬЮ САЦИТУЗУМАБА ГОВИТЕКАНА И ИНГИБИТОРА Rad51
JP6980980B2 (ja) 抗hla‐drまたは抗trop‐2抗体と微小管阻害剤、parp阻害剤、ブルトンキナーゼ阻害剤またはホスホイノシチド3‐キナーゼ阻害剤との併用は癌の治療効果を有意に改善する
ES2909746T3 (es) Dosificaciones de inmunoconjugados de anticuerpos y SN-38 para una eficacia mejorada y una toxicidad disminuida
JP2022046678A (ja) チェックポイント阻害薬に再発/耐性を示す腫瘍を治療するための抗Trop-2-SN-38抗体薬物複合体の効果
JP2023109898A (ja) 治療抗体およびその使用
JP6427789B2 (ja) Cl2aリンカーを有する抗体−sn−38免疫複合体
BR112020005212A2 (pt) conjugado de anticorpo, kit, composição farmacêutica, e, métodos de tratamento ou prevenção e de diagnóstico de uma doença ou condição.
TWI793513B (zh) 對cd123具專一性之抗體和抗體-藥物共軛體及彼等之用途
TW201919693A (zh) 包含可裂解連接子之化合物及其用途
US20210393795A1 (en) Compounds comprising cleavable linker and uses thereof
TW201813671A (zh) 抗c-Met抗體-細胞毒性藥物偶聯物的醫藥用途
WO2021143741A1 (zh) 靶向多肽-药物缀合物及其用途
WO2023024949A1 (zh) 一种由可断裂连接子偶联的抗体偶联药物
RU2757395C2 (ru) Терапия метастатического уротелиального рака с использованием конъюгата антитело-лекарственное средство сацитузумаба говитекана (immu-132)
WO2023016488A1 (zh) 一种基于微管抑制剂的抗体偶联药物
JP2023533793A (ja) 治療用抗体およびそれらの使用
CN109562172B (zh) 抗HLA-DR抗体药物缀合物IMMU-140(hL243-CL2A-SN-38)在HLA-DR阳性癌症中的功效
TWI817190B (zh) 抗c-Met抗體藥物偶聯物及其應用
WO2023103854A1 (zh) 一种亲和力改善的抗体-药物偶联物、其制备方法及应用
US11497787B2 (en) Functionalized poly-ADP-ribose polymers for drug delivery
RU2789150C2 (ru) Антитела и конъюгаты антитела-лекарственного средства, специфичные к cd123, и их применения
CN116284054B (zh) 一种海鞘素类化合物、其抗体药物偶联物及其应用
JP2023100255A (ja) 治療用抗体およびそれらの使用

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: 22903276

Country of ref document: EP

Kind code of ref document: A1