WO2022028353A1 - 作为bcl-2抑制剂的杂环化合物 - Google Patents

作为bcl-2抑制剂的杂环化合物 Download PDF

Info

Publication number
WO2022028353A1
WO2022028353A1 PCT/CN2021/109991 CN2021109991W WO2022028353A1 WO 2022028353 A1 WO2022028353 A1 WO 2022028353A1 CN 2021109991 W CN2021109991 W CN 2021109991W WO 2022028353 A1 WO2022028353 A1 WO 2022028353A1
Authority
WO
WIPO (PCT)
Prior art keywords
hydrogen
compound
methyl
pharmaceutically acceptable
bcl
Prior art date
Application number
PCT/CN2021/109991
Other languages
English (en)
French (fr)
Inventor
孔祥龙
周超
郑之祥
Original Assignee
北京诺诚健华医药科技有限公司
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 北京诺诚健华医药科技有限公司 filed Critical 北京诺诚健华医药科技有限公司
Priority to US18/019,701 priority Critical patent/US20230271959A1/en
Priority to KR1020237007712A priority patent/KR20230065986A/ko
Priority to CA3190686A priority patent/CA3190686A1/en
Priority to EP21852552.5A priority patent/EP4194454A1/en
Priority to AU2021319597A priority patent/AU2021319597A1/en
Priority to CN202180057392.3A priority patent/CN116419753A/zh
Priority to JP2023508059A priority patent/JP2023537032A/ja
Priority to MX2023001618A priority patent/MX2023001618A/es
Publication of WO2022028353A1 publication Critical patent/WO2022028353A1/zh

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D471/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00
    • C07D471/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, at least one ring being a six-membered ring with one nitrogen atom, not provided for by groups C07D451/00 - C07D463/00 in which the condensed system contains two hetero rings
    • C07D471/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/4353Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems
    • A61K31/437Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom ortho- or peri-condensed with heterocyclic ring systems the heterocyclic ring system containing a five-membered ring having nitrogen as a ring hetero atom, e.g. indolizine, beta-carboline
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/496Non-condensed piperazines containing further heterocyclic rings, e.g. rifampin, thiothixene
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/02Antineoplastic agents specific for leukemia

Definitions

  • the present invention relates to compounds, pharmaceutical compositions containing them and their use as B-cell lymphoma-2 (BCL-2) inhibitors. More specifically, the present invention provides novel compounds as BCL-2 inhibitors, pharmaceutical compositions containing such compounds and the use of said compounds in the treatment or prevention of BCL-2-mediated related diseases and dysfunctions, Such related diseases are eg tumors.
  • BCL-2 B-cell lymphoma-2
  • the present invention also relates to methods of preparing said compounds.
  • the BCL-2 protein family is one of the core regulatory mechanisms of apoptosis (also called programmed cell death), which can receive and transmit internal intracellular signals or external environmental stress signals, such as nutritional or hypoxic stress, DNA damage, carcinogenesis Excessive activation of genes, endoplasmic reticulum stress, etc., mainly play a leading role in the intrinsic apoptosis pathway.
  • BCL-2 B-cell lymphoma-22
  • the BCL-2 gene is a proto-oncogene, and the proteins it expresses are called BCL-2 family proteins.
  • There are 27 BCL-2 family proteins in the human body which can be divided into 3 subclasses according to function and sequence analysis.
  • the first subclass is anti-apoptotic, including BCL-XL, BCL-2, BCL-W, MCL -1, BFL-1, they are mainly localized on mitochondria and protect mitochondria from adversity damage; the other two subclasses are apoptosis-promoting, and one subclass is the ultimate executor of mitochondrial damage, including BAX and BAK.
  • the rest belong to the BH3 subclass, which can directly sense various cellular stress signals.
  • BCL-2 protein which antagonizes apoptosis, is closely related to tumors.
  • BCL-2 family proteins bind to BCL-2 family proteins.
  • Multiple signaling pathways such as JAK-STAT, NFkB, and UPP (ubiquitin-proteasome) can cause the overexpression of BCL-2 protein that antagonizes apoptosis.
  • BCL-2 family anti-apoptotic proteins The high expression of BCL-2 family anti-apoptotic proteins is related to the drug resistance of various tumors.
  • the overexpression of BCL-2 anti-apoptotic proteins can enable tumor cells to escape apoptosis caused by anti-tumor drugs, thereby causing drug resistance. medicine.
  • inhibition of BCL-2 family proteins can inhibit the formation of tumor angiogenesis, thereby inhibiting tumor metastasis (Benjamin, D.; Isaac, J. et al. J. Clin. Oncol. 2008, 26(25), 4180 ). Therefore, the targeted inhibition of BCL-2 family anti-apoptotic proteins can inhibit the occurrence, development and drug resistance of tumors.
  • Venetoclax (ABT-199) jointly developed by AbbVie and Roche is a highly selective BCL-2 inhibitor (Andrew, J.; Joel, D. et al. Nature Medicine, 2013, 19(2), 202 ), in the treatment of relapsed/refractory chronic lymphocytic leukemia (CLL), mantle cell lymphoma (MCL), multiple myeloma (MM), etc., through combination therapy with Ibrutinib, etc., the objective response rate (ORR) and The complete remission rate (CR) has been greatly improved (Valentin, R.; Grablow, S. et al.
  • CLL chronic lymphocytic leukemia
  • MCL mantle cell lymphoma
  • MM multiple myeloma
  • ORR objective response rate
  • CR complete remission rate
  • the present invention provides a compound of formula (I), its isomer, prodrug, solvate, stable isotope derivative or pharmaceutically acceptable salt, useful as a selective inhibitor of BCL-2,
  • X 1 is selected from an optionally substituted 6-membered saturated heterocyclic group containing one or two heteroatoms selected from N, O, S, wherein the optional substituent is selected from a group containing one or two heteroatoms selected from N 4-membered saturated heterocyclic group of heteroatoms of , O, S; preferably, X 1 is selected from optionally substituted 6-membered saturated heterocyclic group containing one or two heteroatoms selected from N, O, wherein the The optional substituent is selected from oxetanyl group; further preferably, X 1 is selected from 1,4-dioxanyl group, tetrahydropyranyl group, N-oxetanyl piperidinyl group , N-oxetanyl morpholinyl; most preferably, X 1 is selected from (S)-1,4-dioxan-2-yl, (R)-1,4-dioxane -2-yl, tetrahydropyran-4-yl
  • X 2 is selected from a 5-6-membered heterocyclic group containing one or two N atoms; wherein, the ring can be optionally substituted by one or two C1-C4 alkyl groups; preferably, X 2 is selected from a group containing A 6-membered heterocyclic group with one or two N atoms; wherein, the ring can be optionally substituted by one or two C1-C4 alkyl groups; more preferably, X 2 is selected from Wherein, the ring can be optionally substituted by a C1-C4 alkyl group; most preferably, X 2 is selected from
  • R 0 is selected from hydrogen, halogen; preferably, R 0 is selected from hydrogen, fluorine, chlorine; most preferably, R 0 is selected from hydrogen, fluorine;
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen, C1-C6 alkyl, wherein R 1 and R 2 or R 3 and R 4 can form a 3-6 membered ring together with the attached carbon atoms alkyl; preferably, R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen, C1-C4 alkyl, wherein R 1 and R 2 or R 3 and R 4 may be taken together with the carbon atom to which they are attached form a 3-4 membered cycloalkyl; further preferably, R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen or methyl and R 1 , R 2 , R 3 , R 4 are not all hydrogen, wherein R 1 and R 2 or R 3 and R 4 may together with the carbon atom to which they are attached form a cyclopropyl group; most preferably, R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen or methyl and R 1
  • the present invention relates to a compound of formula (I) as described above, its isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts, wherein:
  • X 1 is selected from an optionally substituted 6-membered saturated heterocyclic group containing one or two heteroatoms selected from N, O, wherein the optional substituent is selected from an oxetanyl group;
  • X 2 is selected from a 6-membered heterocyclic group containing one or two N atoms; wherein, the ring can be optionally substituted by one or two C1-C4 alkyl groups;
  • R 0 is selected from hydrogen, halogen
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen, C1-C4 alkyl, wherein R 1 and R 2 or R 3 and R 4 can form a 3-4 membered ring together with the attached carbon atoms alkyl.
  • the present invention relates to the aforementioned compounds of formula (I), isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts thereof, wherein:
  • X 1 is selected from an optionally substituted 6-membered saturated heterocyclic group containing one or two heteroatoms selected from N, O, wherein the optional substituent is selected from an oxetanyl group;
  • X 2 is Wherein, the ring can be optionally substituted by a C1-C4 alkyl group
  • R 0 is selected from hydrogen, fluorine, chlorine
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen, C1-C4 alkyl, wherein R 1 and R 2 or R 3 and R 4 can form a 3-4 membered ring together with the attached carbon atoms alkyl.
  • the present invention relates to the aforementioned compounds of formula (I), isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts thereof, wherein:
  • X 1 is selected from 1,4-dioxanyl, tetrahydropyranyl, N-oxetanylpiperidinyl, N-oxetanylmorpholinyl;
  • X 2 is Wherein, the ring can be optionally substituted by a C1-C4 alkyl group
  • R 0 is selected from hydrogen, fluorine, chlorine
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen or methyl and R 1 , R 2 , R 3 , R 4 are not all hydrogen, wherein R 1 and R 2 or R 3 and R 4 may be combined with The attached carbon atoms together form a cyclopropyl group.
  • the present invention relates to the aforementioned compound of formula (I), its isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts, wherein:
  • X 1 is selected from 1,4-dioxanyl, tetrahydropyranyl, N-oxetanylpiperidinyl, N-oxetanylmorpholinyl;
  • X 2 is selected from
  • R 0 is selected from hydrogen, fluorine
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen or methyl and R 1 , R 2 , R 3 , R 4 are not all hydrogen, wherein R 1 and R 2 or R 3 and R 4 may be combined with The attached carbon atoms together form a cyclopropyl group.
  • the present invention relates to the aforementioned compounds of formula (I), isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts thereof, wherein:
  • X 1 is selected from (S)-1,4-dioxan-2-yl, (R)-1,4-dioxan-2-yl, tetrahydropyran-4-yl, 1-( oxetan-3-yl)piperidin-4-yl, (S)-4-(oxetan-3-yl)morpholin-2-yl;
  • X 2 is selected from
  • R 0 is selected from hydrogen, fluorine
  • R 1 , R 2 , R 3 , R 4 are each independently selected from hydrogen or methyl and R 1 , R 2 , R 3 , R 4 are not all hydrogen, wherein R 3 and R 4 may be taken together with the carbon atom to which they are attached Forms cyclopropyl.
  • the present invention relates to the aforementioned compounds of formula (I), isomers, prodrugs, solvates, stable isotopic derivatives or pharmaceutically acceptable salts thereof, selected from:
  • the present invention also relates to compounds of formula (I), isomers, prodrugs, solvates, stable isotope derivatives or pharmaceutically acceptable salts thereof according to any one of the embodiments of the present invention for use in the preparation of Use of a BCL-2 inhibitor in medicine.
  • the present invention also relates to a compound of formula (I) or an isomer, prodrug, solvate, stable isotope derivative or a pharmaceutically acceptable salt thereof according to any one of the embodiments of the present invention in the manufacture of a medicament
  • BCL-2-mediated related diseases such as tumors selected from hematological malignancies, lung cancer, breast cancer, ovarian cancer, especially acute lymphoblastic leukemia cancer, prostate cancer, rectal cancer, pancreatic cancer, glioma.
  • the present invention further relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or an isomer, prodrug, solvate, stable isotopic derivative or A pharmaceutically acceptable salt thereof, optionally one or more other BCL-2 inhibitors, and one or more pharmaceutically acceptable carriers, diluents and excipients.
  • the present invention also relates to the use of the pharmaceutical composition according to the present invention in the preparation of a medicament, wherein the medicament is used for the treatment or prevention of BCL-2-mediated related diseases, such as tumors, which are selected from From hematological malignancies, especially acute lymphoblastic leukemia, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreatic cancer, glioma.
  • BCL-2-mediated related diseases such as tumors, which are selected from From hematological malignancies, especially acute lymphoblastic leukemia, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreatic cancer, glioma.
  • the present invention also relates to a method of treating or preventing related diseases mediated by BCL-2, comprising administering to a patient in need thereof a therapeutically effective amount of the compound described in any one of the embodiments of the present invention or an isomer, pro- Drugs, solvates, stable isotope derivatives or pharmaceutically acceptable salts thereof; or pharmaceutical compositions according to the present invention, said related diseases such as tumors selected from malignant tumors, especially acute lymphoblastic leukemia Blood diseases, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreatic cancer, glioma.
  • related diseases such as tumors selected from malignant tumors, especially acute lymphoblastic leukemia Blood diseases, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreatic cancer, glioma.
  • Another aspect of the present invention pertains to a compound described in any one of the embodiments of the present invention, or an isomer, prodrug, solvate, stable isotope derivative or pharmaceutically acceptable salt thereof, for use in therapy or Prevention of related diseases mediated by BCL-2, such as tumors selected from hematological malignancies, especially acute lymphoblastic leukemia, lung, breast, ovarian, prostate, rectal, pancreatic cancer ,Glioma.
  • Another aspect of the present invention relates to a pharmaceutical composition
  • a pharmaceutical composition comprising a compound of formula (I) or an isomer, prodrug, solvate, stable isotope thereof, as described in any one of the embodiments of the present invention
  • Derivatives or pharmaceutically acceptable salts thereof optionally one or more other BCL-2 inhibitors, and one or more pharmaceutically acceptable carriers, diluents and excipients for use in therapy Or prevent related diseases mediated by BCL-2, such as tumors selected from hematological malignancies, especially acute lymphoblastic leukemia, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreas Cancer, glioma.
  • Another aspect of the present invention pertains to a compound of formula (I) or an isomer, prodrug, solvate thereof as described in any one of the embodiments of the present invention for the treatment and/or prophylaxis of BCL-2-mediated related diseases , a stable isotope derivative or a pharmaceutically acceptable salt thereof.
  • the BCL-2-mediated related disease is for example a tumor selected from hematological malignancies, especially acute lymphoblastic leukemia, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, pancreatic cancer, brain glioma .
  • the drug can be in any pharmaceutical dosage form, including but not limited to tablets, capsules, solutions, freeze-dried preparations, and injections.
  • the pharmaceutical formulations of the present invention may be administered in dosage unit form containing a predetermined amount of active ingredient per dosage unit.
  • a predetermined amount of active ingredient per dosage unit may contain, for example, from 0.5 mg to 1 gram, preferably from 1 mg to 700 mg, particularly preferably from 5 mg to 300 mg, of a compound of the invention, or drug, depending on the condition to be treated, the method of administration and the age, weight and condition of the patient.
  • the formulations can be administered in dosage unit form containing a predetermined quantity of active ingredient per dosage unit.
  • Preferred dosage unit formulations are those containing a daily dose or sub-dose, as indicated above, or a corresponding fraction thereof, of an active ingredient.
  • pharmaceutical formulations of this type can be prepared using methods well known in the pharmaceutical art.
  • the pharmaceutical formulations of the present invention may be suitable for administration by any desired suitable method, such as oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods of administration.
  • suitable method such as oral (including buccal or sublingual), rectal, nasal, topical (including buccal, sublingual or transdermal), vaginal or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) methods of administration.
  • Such formulations can be prepared, for example, by combining the active ingredient with one or more excipients or one or more adjuvants, using all methods known in the art of pharmacy.
  • the present invention also provides methods for preparing the compounds.
  • R 0 and X 1 are as described above;
  • R 1 , R 2 , R 3 and R 4 are as described above;
  • compound (V) Under nitrogen protection, compound (V), p-chlorobenzeneboronic acid, alkali such as potassium carbonate, phase-transfer catalyst such as tetra-n-butylammonium bromide and catalyst such as palladium acetate are added to a solvent such as water, the system is evacuated and replaced with nitrogen three times, and heated to 40 °C. React at ⁇ 100°C for 2 to 10 hours to obtain compound (VI);
  • R 0 , R 1 , R 2 , R 3 , R 4 and X 1 are as defined above;
  • R 0 , R 1 , R 2 , R 3 , R 4 and X 1 are as defined above;
  • Cx-Cy used in the present invention represents a range of carbon atoms, where x and y are both integers, eg C3-C8 cycloalkyl means a cycloalkyl group having 3-8 carbon atoms, -C0 -C2 alkyl represents an alkyl group having 0-2 carbon atoms, wherein -C0 alkyl refers to a chemical single bond.
  • alkyl refers to saturated aliphatic hydrocarbon groups, including straight and branched chain groups of 1 to 20 carbon atoms, for example, may be 1 to 18 carbon atoms, 1 to 12 carbon atoms , straight and branched chain groups of 1 to 8 carbon atoms, 1 to 6 carbon atoms or 1 to 4 carbon atoms.
  • Non-limiting examples include methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, tert-butyl, sec-butyl, n-pentyl, 1,1-dimethylpropyl, 1 , 2-dimethylpropyl, 2,2-dimethylpropyl, 1-ethylpropyl, 2-methylbutyl, 3-methylbutyl, n-hexyl, 1-ethyl-2- Methylpropyl, 1,1,2-trimethylpropyl, 1,1-dimethylbutyl, 1,2-dimethylbutyl, 2,2-dimethylbutyl, 1,3 -Dimethylbutyl, 2-ethylbutyl, and various branched isomers thereof, etc.
  • Alkyl groups can be optionally substituted or unsubstituted.
  • cycloalkyl refers to a saturated monocyclic or polycyclic cyclic hydrocarbon group comprising 3 to 12 ring atoms, such as may be 3 to 12, 3 to 10, 3 to 8 or 3 to 6 ring atoms, or can be a 3, 4, 5, or 6 membered ring.
  • monocyclic cyclyl groups include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cycloheptyl, cyclooctyl, and the like. Cycloalkyl groups can be optionally substituted or unsubstituted.
  • heterocyclyl refers to a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon group comprising 3 to 20 ring atoms, for example, may be 3 to 16, 3 to 12, 3 to 10, 3 to 8, or 3 to 6 ring atoms, one or more of which is selected from nitrogen, oxygen, or a heteroatom of S(O)m (where m is an integer from 0 to 2), excluding- The ring moiety of OO-, -OS- or -SS-, the remaining ring atoms being carbon.
  • the heterocyclyl ring contains 3 to 10 ring atoms, more preferably 3 to 8 ring atoms, most preferably a 5- or 6-membered ring A ring of which 1-4 are heteroatoms, more preferably 1-3 are heteroatoms, and most preferably 1-2 are heteroatoms.
  • Non-limiting examples of monocyclic heterocyclyl groups include oxetanyl, pyrrolidinyl, piperidinyl, 4-piperidinyl, piperazinyl, 1,4-dioxanyl, morpholinyl, 2-morpholinyl, 4-morpholinyl, thiomorpholinyl, pyranyl, tetrahydropyranyl, 4-tetrahydropyranyl, homopiperazinyl, dioxanyl, 2-dioxanyl Alkyl etc.
  • Polycyclic heterocyclyls include spiro, fused and bridged heterocyclyls. Heterocyclyl groups can be optionally substituted or unsubstituted.
  • heterocyclylene refers to a substituted or unsubstituted heterocyclyl having a core of two terminal monovalent groups obtained by removing one hydrogen atom from each of the two terminal atoms. produced; the heterocyclyl group has the meanings set forth above.
  • Non-limiting examples of “heterocyclylene” include pyrrolidylene, piperidinylene, piperazinylene, morpholinylene, and the like.
  • halogen refers to fluorine, chlorine, bromine or iodine.
  • a heterocyclic group optionally substituted with an alkyl group means that an alkyl group may, but need not, be present, and the description includes the case where the heterocyclic group is substituted with an alkyl group and the case where the heterocyclic group is not substituted with an alkyl group .
  • substituted means that one or more hydrogen atoms in a group, preferably up to 5, more preferably 1 to 3 hydrogen atoms, independently of each other, are substituted by the corresponding number of substituents. It goes without saying that the substituents are only in their possible chemical positions, and the person skilled in the art can determine (either experimentally or theoretically) possible or impossible substitutions without undue effort. For example, amino or hydroxyl groups with free hydrogens may be unstable when combined with carbon atoms with unsaturated (eg, olefinic) bonds.
  • the substituents include, but are not limited to, the various groups described above.
  • the compounds claimed in the present invention include not only the compounds themselves, but also isomers, prodrugs, solvates, stable isotope derivatives or pharmaceutically acceptable salts thereof.
  • the "pharmaceutical composition” of the present invention refers to containing one or more isomers, prodrugs, solvates, stable isotope derivatives or pharmaceutically acceptable salts thereof and other chemical components of the compounds of the present invention mixture. Other components such as pharmaceutically acceptable pharmaceutically acceptable carriers, diluents and excipients.
  • the purpose of the pharmaceutical composition is to facilitate the administration to the organism, facilitate the absorption of the active ingredient and then exert the biological activity.
  • room temperature in the present invention refers to 15-30°C.
  • stable isotopic derivatives include: isotopically substituted derivatives obtained by replacing any hydrogen atom in formula (I) with 1-5 deuterium atoms, and any carbon atom in formula (I) by 1-3 Isotopically substituted derivatives obtained by substituting one carbon 14 atom or isotopically substituted derivatives obtained by substituting 1-3 oxygen 18 atoms in formula (I) for any oxygen atom.
  • “Pharmaceutically acceptable salts” according to the present invention are discussed in Berge, et al., “Pharmaceutically acceptable salts", J. Pharm. It will be apparent that the salts are substantially non-toxic and provide the desired pharmacokinetic properties, palatability, absorption, distribution, metabolism or excretion and the like.
  • the pharmaceutically acceptable salts of the present invention can be synthesized by general chemical methods.
  • salts can be prepared by reacting the free base or acid with an equivalent or an excess of acid (inorganic or organic) or base in a suitable solvent or solvent composition.
  • prodrug in the present invention refers to the conversion of the compound into the original active compound after metabolism in the body.
  • a prodrug is an inactive substance, or is less active than the active parent compound, but can provide ease of manipulation, administration, or improved metabolic properties.
  • the "isomers” in the present invention refer to the tautomers, mesomers, racemates, enantiomers, diastereomers, and other isomers of the compound of formula (I) of the present invention. Mixture form, etc. All of these isomers, including stereoisomers and geometric isomers, are included in the present invention.
  • the geometric isomers include cis-trans isomers.
  • solvents refers to the association of one or more solvent molecules with a compound of the present invention or a salt thereof.
  • solvents that form pharmaceutically acceptable solvates include, but are not limited to, water, isopropanol, ethanol, methanol, ethyl acetate, acetic acid, and the like.
  • the present invention includes any polymorphs of the compounds or salts thereof, as well as any hydrates or other solvates.
  • the term "patient” generally refers to mammals, especially humans.
  • tumor includes benign tumors and malignant tumors, such as cancer.
  • cancer includes various tumors mediated by BCL-2 including, but not limited to, hematological malignancies, especially acute lymphoblastic leukemia, lung cancer, breast cancer, ovarian cancer, prostate cancer, rectal cancer, Pancreatic cancer, brain glioma.
  • the term "therapeutically effective amount” is meant to include an amount of the compound of the present invention effective to treat or prevent related diseases mediated by BCL-2.
  • the structures of all compounds of the present invention can be identified by nuclear magnetic resonance (1H NMR) and/or mass spectrometry (MS).
  • MS mass spectra
  • the thin layer silica gel plate is Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate.
  • Column chromatography generally uses Yantai Yellow Sea 100-200 or 200-300 mesh silica gel as a carrier.
  • Preparative liquid chromatography uses Waters SQD2 mass spectrometry-oriented high-pressure liquid chromatography, XBridge-C18; 30X 150mm preparative column, 5 ⁇ m;
  • Method 1 acetonitrile-water (0.2% formic acid), flow rate 25mL/min;
  • Method 2 acetonitrile-water (0.8% ammonium bicarbonate), flow rate 25mL/min;
  • the known starting materials of the present invention can be synthesized by adopting or according to methods known in the art, or can be purchased from Acros Organics, Aldrich Chemical Company, Accela ChemBio Inc, Shanghai Bide Medicine, Shanghai Accela Latin Chemical, Shanghai Merrill Chemical, Bailingwei Chemical, Anai Chemical and other companies. Venetoclax was purchased from WuXi Lab (Wuhan) Chemical Technology Co., Ltd.
  • the solvent used in the reaction is an anhydrous solvent, wherein the anhydrous tetrahydrofuran uses commercially available tetrahydrofuran, takes sodium block as water scavenger, takes benzophenone as indicator, and refluxes to solution under argon protection. It was blue-purple, collected by distillation, and stored at room temperature under argon protection.
  • Other anhydrous solvents were purchased from Annai Chemical and Bailingwei Chemicals. All the transfer and use of anhydrous solvents should be carried out under argon protection unless otherwise specified.
  • Argon or nitrogen atmosphere means that the reaction flask is connected to an argon or nitrogen balloon with a volume of about 1 L.
  • Hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon with a volume of about 1 L.
  • the hydrogenation reaction is usually evacuated and filled with hydrogen, and the operation is repeated 3 times.
  • reaction temperature is room temperature, and the temperature range is 15°C-30°C.
  • the monitoring of the reaction progress in the examples adopts thin layer chromatography (TLC), and the systems of the developing solvents used in the reaction are A: dichloromethane and methanol system; B: petroleum ether and ethyl acetate system.
  • TLC thin layer chromatography
  • A dichloromethane and methanol system
  • B petroleum ether and ethyl acetate system.
  • the volume ratio of the solvent is adjusted according to the polarity of the compound.
  • the eluent system for column chromatography and the developing solvent system for thin layer chromatography used to purify the compound include A: dichloromethane and methanol system; B: petroleum ether and ethyl acetate system.
  • the volume ratio of the solvent is adjusted according to the polarity of the compound, and can also be adjusted by adding a small amount of triethylamine and an acidic or basic reagent.
  • 1,2-Difluoro-3-nitrobenzene (10.00 g, 62.89 mmol) was dissolved in chlorosulfonic acid (21 mL), heated to 150° C. and stirred under reflux for 10 hours. After cooling to room temperature, a saturated aqueous sodium bicarbonate solution was added to the reaction solution under an ice bath to adjust the pH to about 7. Extract with dichloromethane (100 mL ⁇ 3), wash the organic phase with saturated brine (100 mL ⁇ 2), dry over anhydrous sodium sulfate, filter and concentrate to obtain the crude product 3,4-difluoro-5-nitrobenzenesulfonyl chloride.
  • intermediate 2 The synthetic steps of intermediate 2 refer to intermediate 1.
  • N,N-dimethylformamide (0.59 g, 8.00 mmol) was dissolved in dichloromethane (10 mL), cooled to 0 °C on an ice bath, phosphine trichloride (0.92 g, 6.00 mmol) was added dropwise, and the temperature was kept warm. After stirring for 30 minutes, the temperature was naturally raised to room temperature, and stirring was continued for 3 hours. Cool to 0°C, slowly add (R)-3-methylcyclohexyl-1-one (synthesis reference: Tetrahedron 73 (2017) 3202-3212) (0.45 g, 4.0 mmol) in dichloromethane (5 mL) solution , slowly warmed to room temperature and stirred the reaction for 18 hours.
  • reaction solution was quenched by adding saturated sodium bicarbonate solution (10 mL), concentrated to remove dichloromethane, ethyl acetate (25 mL) and water (15 mL) were added, the organic phase was separated, and the aqueous phase was extracted with ethyl acetate (25 mL ⁇ 2). , the combined organic phases were washed with saturated brine (20 mL ⁇ 2), dried over anhydrous sodium sulfate, filtered and concentrated to obtain the target compound (R)-2-chloro-4-methylcyclohex-1-ene-1-carbaldehyde ( 0.45g, light yellow liquid), crude product.
  • reaction was quenched by dropwise addition of saturated aqueous sodium bicarbonate solution (10 mL), extracted with dichloromethane (10 mL), the organic phase was washed with saturated brine (10 mL), dried, filtered, the filtrate was concentrated, and the reaction solution was spin-dried to obtain a crude product ( R)-4'-Chloro-6-(chloromethyl)-3-methyl-2,3,4,5-tetrahydro-1,1'-biphenyl (0.26 g, pale yellow oil), crude Used directly in the next step without purification.
  • reaction solution was quenched with saturated aqueous ammonium chloride solution (200 mL), the aqueous phase was extracted with ethyl acetate (100 mL ⁇ 2), the organic phases were combined and washed with saturated brine (200 mL), the organic phase was dried over anhydrous sodium sulfate and filtered. , the filtrate was concentrated under reduced pressure. The residue was separated by silica gel column chromatography (0-10% ethyl acetate/petroleum ether) to give the product 7,7-dimethyl-8-methylene-1,4-dioxaspiro[4.5]decane (3.80 g, colorless oil). Yield: 64%.
  • Phosphorus oxychloride (1.10 g, 6.90 mmol) was added dropwise to an ice-bath solution of N,N-dimethylformamide (0.67 g, 9.20 mmol) in dichloromethane (30 mL) under nitrogen protection. , stirred at room temperature for 30 minutes, cooled to 0 °C again, added dropwise a solution of 4,4-dimethylspiro[2.5]octan-6-one (0.70 g, 4.60 mmol) in dichloromethane (5 mL), and reacted at room temperature overnight .
  • 6-chloro-8,8-dimethylspiro[2.5]oct-5-ene-5-carbaldehyde (0.34 g, 1.70 mmol)
  • p-chlorophenylboronic acid (0.41 g, 2.60 mmol)
  • carbonic acid Potassium (0.70 g, 5.10 mmol
  • tetra-n-butylammonium bromide (0.55 g, 1.70 mmol)
  • palladium acetate 77 mg, 0.34 mmol
  • the mixture was quenched with 10 mL of water and extracted with ethyl acetate (10 mL x 2).
  • the combined organic phases were washed with saturated brine (10 mL ⁇ 2).
  • the organic phase was dried with anhydrous sodium sulfate, the drying agent was removed by filtration, and the crude product was obtained by desolvation under reduced pressure.
  • Example 57 Synthesis refers to the synthesis steps of Example 1, wherein the second step is replaced by (4aR, 8aR)-decahydroquinoxaline (synthesis reference: European Journal of Organic Chemistry, 2012(34), 6752-6759; 2012) (R)-2-methylpiperazine to give the target compound 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxo)-4-((4aR,8aR)-4- ((4'-Chloro-5,5-dimethyl-3,4,5,6-tetrahydro-[1,1'-biphenyl]-2-yl)methyl)octahydroquinoxaline- 1(2H)-yl)-N-((3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)phenyl)sulfonyl)benzamide 57.
  • Example 58 Synthesis refers to the synthetic steps of Example 1, wherein (2R,5R)-2,5-dimethylpiperazine is used in the second step (synthesis reference: Organic Chemistry Frontiers, 5(23), 3402-3405; 2018 ) instead of (R)-2-methylpiperazine to give the target compound 2-((1H-pyrrolo[2,3-b]pyridin-5-yl)oxo)-4-((2R,5R)- 4-((4'-Chloro-5,5-dimethyl-3,4,5,6-tetrahydro-[1,1'-biphenyl]-2-yl)methyl)-2,5 -Dimethylpiperazin-1-yl)-N-((3-nitro-4-(((tetrahydro-2H-pyran-4-yl)methyl)amino)phenyl)sulfonyl)benzene Formamide 58.
  • Example 59 Synthesis refers to the synthesis steps of Example 1, wherein in the second step, (2R,6R)-2,6-dimethylpiperazine is used to replace (R)-2-methylpiperazine to obtain the target compound N-( (4-((((S)-1,4-dioxan-2-yl)methyl)amino)-3-nitrophenyl)sulfonyl)-2-((1H-pyrrolo[2, 3-b]pyridin-5-yl)oxo)-4-((3R,5R)-4-((4'-chloro-5,5-dimethyl-3,4,5,6-tetrahydro -[1,1'-Biphenyl]-2-yl)methyl)-3,5-dimethylpiperazin-1-yl)benzamide 59.
  • the effect of compounds on the biological activity of BCL-2 was assessed by detecting the effect of the compounds on the binding activity of BCL-2 and leukemia pro-apoptotic protein (BIM) using an affinity assay method based on the principle of fluorescence polarization.
  • the reaction buffer contains the following components: PBS (pH 7.4, 3 mM Na 2 HPO 4 , 155 mM NaCl, 1 mM KH 2 PO 4 ), 1 mM DTT; human recombinant Bcl-2 protein (Cat. No.
  • Y is the percentage of inhibition
  • X is the logarithm of the concentration of the compound to be tested
  • Bottom is the maximum percentage of inhibition
  • Top is the minimum percentage of inhibition
  • slope factor is the slope coefficient of the curve.
  • the experimental method is outlined as follows:
  • the effect of compounds on the biological activity of BCL-XL was evaluated by detecting the effect of the compounds on the binding activity of BCL-XL and BIM using an affinity assay method based on the principle of fluorescence polarization.
  • the reaction buffer contains the following components: PBS (pH 7.4, 3 mM Na 2 HPO 4 , 155 mM NaCl, 1 mM KH 2 PO 4 ), 1 mM DTT; human recombinant Bcl-XL protein (Cat. No.
  • 10455-H08E was purchased from Beijing Yiqiao Shenzhou Biotechnology Co., Ltd., diluted to 10 nM with reaction buffer;
  • FITC-labeled BIM polypeptide was purchased from Nanjing GenScript Biotechnology Co., Ltd. and diluted to 10 nM with reaction buffer.
  • Y is the percentage of inhibition
  • X is the logarithm of the concentration of the compound to be tested
  • Bottom is the maximum percentage of inhibition
  • Top is the minimum percentage of inhibition
  • slope factor is the slope coefficient of the curve.
  • the effect of the compounds of the present invention on the proliferation of RS4;11 cells was evaluated using a luminescence cell viability assay.
  • the experimental method is outlined as follows:
  • CellTilter-Glo reagent (Promega, G7572) is composed of CellTilter-Glo lyophilized powder and CellTilter-Glo buffer. The lyophilized powder can be dissolved in the buffer when used.
  • RS4;11 cells (ATCC, CRL-1873) were cultured in RPMI1640 complete medium (Thermofisher, 72400-047) containing 10% FBS (GBICO, 10099-141) and 100 units/ml penicillin-streptomycin (Thermofisher, 15140122) ), when the cells covered 80-90% in the culture vessel, they were digested with 0.25% trypsin (containing EDTA) (Thermofisher, 25200056) and then planted in a white 384-well plate (Thermofisher, 164610), and then 384-well Plates were incubated overnight in a 37°C, 5% CO2 incubator.
  • the percent inhibition of RS4;11 cell proliferation by a compound can be calculated using the following formula:
  • Bottom is the bottom plateau of the curve (the bottom plateau value of the S-shaped curve)
  • Top is the top plateau of the curve (the top plateau value of the S-shaped curve)
  • X is the concentration of the compound to be tested. numerical value.
  • the compounds of the examples of the present invention can effectively and selectively inhibit the activity of BCL-2, and have weak inhibition on BCL-XL.
  • Some compounds can also effectively inhibit RS4;11 acute lymphocyte proliferation. It has a strong inhibitory effect on malignant blood diseases such as acute lymphoblastic leukemia.

Abstract

本发明涉及化合物、含有其的药物组合物、以及它们的制备方法,和其作为BCL-2抑制剂的用途。所述化合物为式(I)所示的化合物,或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐。本发明还涉及所述化合物用于治疗或预防由BCL-2介导的相关疾病例如肿瘤的用途以及应用其治疗所述疾病的方法。

Description

作为BCL-2抑制剂的杂环化合物 技术领域
本发明涉及化合物、含有其的药物组合物和其作为B细胞淋巴瘤-2(BCL-2)抑制剂的用途。更具体说,本发明提供了新的作为BCL-2抑制剂的化合物、含有这种化合物的药物组合物和应用所述化合物治疗或预防由BCL-2介导的相关疾病和功能障碍的用途,所述相关疾病例如肿瘤。本发明还涉及制备所述的化合物的方法。
背景技术
BCL-2蛋白家族是细胞凋亡(也叫程序性细胞死亡)的核心调节机制之一,可以接收和传递内在胞内信号或外部环境应激信号,如营养或缺氧胁迫、DNA损伤、致癌基因过量激活、内质网应激胁迫等,主要在内在凋亡途径(Intrinsic pathway)中起主导作用。BCL-2(B-cell lymphoma-2)蛋白在1986年首次被发现,由BCL-2基因表达。BCL-2基因是一种原癌基因,其表达的蛋白称为BCL-2家族蛋白。人体中共有27个BCL-2家族的蛋白,按照功能和序列分析可分为3个亚类,第一亚类是拮抗细胞凋亡的,包括BCL-XL、BCL-2、BCL-W、MCL-1、BFL-1,它们主要定位于线粒体上,保护线粒体免于逆境损伤;另两个亚类是促进细胞凋亡的,一亚类是线粒体损伤的最终执行者,包括BAX和BAK。其余都归于BH3亚类,可以直接感应各种不同细胞逆境胁迫信号。拮抗和促进细胞凋亡的蛋白之间相互作用的动态平衡以决定细胞的生死命运。拮抗细胞凋亡的BCL-2蛋白与肿瘤密切相关,约有50%的肿瘤(如白血病、直肠癌、前列腺癌等)存在BCL-2家族蛋白异常过量表达,其中BCL-2活性异常在血液肿瘤中广泛存在。JAK-STAT、NFkB、UPP(ubiquitin-proteasome)等多个信号通路可以引起拮抗凋亡的BCL-2蛋白的过表达。
BCL-2家族拮抗凋亡蛋白的高表达与多种肿瘤的药物耐药性相关,如BCL-2拮抗凋亡蛋白的过表达能使肿瘤细胞逃避抗肿瘤药物引起的凋亡,从而引起药物耐药。有研究表明,抑制BCL-2家族蛋白可以抑制肿瘤新生血管的形成,从而抑制肿瘤的转移(Benjamin,D.;Isaac,J.et al.J.Clin.Oncol.2008,26(25),4180)。因此,靶向抑制BCL-2家族 抗凋亡蛋白能抑制肿瘤的发生、发展和耐药。
尽管已经有20多种靶向BCL-2家族的小分子抑制剂的报导,但进入临床实验的寥寥无几,梯瓦的Obatoclax在26例接受治疗的慢性淋巴细胞白血病(CLL)中只有1例获得部分应答,且有较强的神经毒性,于2013年终止了开发;艾伯维研发的Navitoclax(ABT-263)尽管在复发或难治性淋巴恶性肿瘤患者的I期剂量爬坡实验中,表现出50%的良好反应率,但同时也表现出非常强的BCL-XL的靶向毒性:如血小板降低和严重贫血。艾伯维与罗氏联合开发的Venetoclax(ABT-199)是一种高选择性的BCL-2抑制剂(Andrew,J.;Joel,D.et al.Nature Medicine,2013,19(2),202),在复发/难治性慢性淋巴细胞白血病(CLL)、套细胞淋巴瘤(MCL)、多发性骨髓瘤(MM)等的治疗中通过与Ibrutinib等的联合用药,客观缓解率(ORR)与完全缓解率(CR)都得到了极大提升(Valentin,R.;Grablow,S.et al.Blood,2018,132(12),1248),但依然有白细胞与血小板减少、贫血、腹泻、头晕、疲劳、易感染等毒副作用,严重的毒副作用还包括肺炎、贫血、高烧等。因此有必要研发高活性,低毒副作用的BCL-2选择性抑制剂。
发明内容
本发明提供一种可用作BCL-2选择性抑制剂的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,
Figure PCTCN2021109991-appb-000001
其中:
X 1选自任选被取代的含有一或二个选自N、O、S的杂原子的6元饱和杂环基,其中所述任选的取代基选自含有一或二个选自N、O、S 的杂原子的4元饱和杂环基;优选地,X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;进一步优选地,X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;最优选地,X 1选自(S)-1,4-二氧六环-2-基、(R)-1,4-二氧六环-2-基、四氢吡喃-4-基、1-(氧杂环丁烷-3-基)哌啶-4-基、(S)-4-(氧杂环丁烷-3-基)吗啉-2-基;
X 2选自含有一或二个N原子的5-6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;优选地,X 2选自含有一或二个N原子的6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;进一步优选地,X 2选自
Figure PCTCN2021109991-appb-000002
其中,环上任选可被一个C1~C4的烷基所取代;最优选地,X 2选自
Figure PCTCN2021109991-appb-000003
R 0选自氢、卤素;优选地,R 0选自氢、氟、氯;最优选地,R 0选自氢、氟;
R 1、R 2、R 3、R 4各自独立地选自氢、C1-C6烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~6元环烷基;优选地,R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基;进一步优选地,R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基;最优选地,R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 3和R 4可以与所连接的碳原子一起组成环丙基。
优选地,本发明涉及上述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;
X 2选自含有一或二个N原子的6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;
R 0选自氢、卤素;
R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基。
进一步优选地,本发明涉及前述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;
X 2
Figure PCTCN2021109991-appb-000004
其中,环上任选可被一个C1~C4的烷基所取代;
R 0选自氢、氟、氯;
R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基。
进一步优选地,本发明涉及前述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;
X 2
Figure PCTCN2021109991-appb-000005
其中,环上任选可被一个C1~C4的烷基所取代;
R 0选自氢、氟、氯;
R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基。
再进一步优选地,本发明涉及前述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;
X 2选自
Figure PCTCN2021109991-appb-000006
R 0选自氢、氟;
R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基。
还进一步优选地,本发明涉及前述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
X 1选自(S)-1,4-二氧六环-2-基、(R)-1,4-二氧六环-2-基、四氢吡喃-4-基、1-(氧杂环丁烷-3-基)哌啶-4-基、(S)-4-(氧杂环丁烷-3-基)吗啉-2-基;
X 2选自
Figure PCTCN2021109991-appb-000007
R 0选自氢、氟;
R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 3和R 4可以与所连接的碳原子一起组成环丙基。
最优选地,本发明涉及前述的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其选自:
Figure PCTCN2021109991-appb-000008
Figure PCTCN2021109991-appb-000009
Figure PCTCN2021109991-appb-000010
Figure PCTCN2021109991-appb-000011
Figure PCTCN2021109991-appb-000012
本发明还涉及根据本发明任意一个实施方案中所述的式(I)化合物、其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐在制备用作BCL-2抑制剂的药物中的用途。
本发明还涉及根据本发明任意一个实施方案中所述的式(I)化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐在制备药物中的用途,所述药物用于治疗或预防由BCL-2介导的相关疾病,所述相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
本发明进一步涉及一种药物组合物,所述药物组合物包括本发明任意一个实施方案中所述的式(I)化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐,任选的一种或多种其它BCL-2抑制剂,以及一种或多种药学上可接受的载体、稀释剂和赋形剂。
本发明还涉及根据本发明所述的药物组合物在制备药物中的用 途,其中所述药物用于治疗或者预防由BCL-2介导的相关疾病,所述相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
本发明还涉及一种治疗或者预防由BCL-2介导的相关疾病的方法,其包括给予有需要的患者治疗有效量的本发明任意一个实施方案中所述的化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐;或本发明所述的药物组合物,所述相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
本发明的另一方面涉及本发明任意一个实施方案中所述的化合物、或其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其用于治疗或者预防由BCL-2介导的相关疾病,所述相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
本发明的另一方面涉及一种药物组合物,所述药物组合物包括本发明任意一个实施方案中所述的式(I)化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐,任选的一种或多种其它BCL-2抑制剂,以及一种或多种药学上可接受的载体、稀释剂和赋形剂,其用于治疗或者预防由BCL-2介导的相关疾病,所述相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
本发明的另一方面涉及作为治疗和/或预防由BCL-2介导的相关疾病的本发明任意一个实施方案中所述的式(I)化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐。所述BCL-2介导的相关疾病例如肿瘤,所述肿瘤选自尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
根据本发明,所述药物可以是任何药物剂型,包括但不限于片剂、胶囊剂、溶液剂、冻干制剂、注射剂。
本发明的药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。这种单位可根据治疗的病症、给药方法和患者的年 龄、体重和状况包含例如0.5毫克至1克,优选1毫克至700毫克,特别优选5毫克至300毫克的本发明的化合物,或药物制剂可以以每剂量单位包含预定量的活性成分的剂量单位形式给药。优选剂量单位制剂是包含如上指示的日剂量或分剂量或其相应分数的活性成分的那些。此外,可以使用制药领域中公知的方法制备这种类型的药物制剂。
本发明药物制剂可适于通过任何所需的合适方法给药,例如通过经口(包括口腔或舌下)、直肠、经鼻、局部(包括口腔、舌下或经皮)、阴道或肠胃外(包括皮下、肌内、静脉内或皮内)方法给药。可以使用制药领域中已知的所有方法通过例如将活性成分与一种或多种赋形剂或一种或多种辅助剂合并来制备这样的制剂。
制备流程
本发明还提供制备所述化合物的方法。
流程1
Figure PCTCN2021109991-appb-000013
R 0与X 1的定义如前文所述;
第一步:
化合物(I)溶解在氯磺酸中,在油浴加热(120~150℃)下反应10~20小时,冷却到室温,冰水淬灭,乙酸乙酯萃取,有机相干燥浓缩得到的粗品溶于无水四氢呋喃中,低温(-80~-60℃)下滴加氨水,并继续搅拌1~5小时,盐酸酸化,得到化合物(II);
第二步:
化合物(II)与相应的胺溶于溶剂(如乙腈)中,加入碱(如三乙胺或者二异丙基乙基胺等),在惰性气体(如氮气或氩气)保护下,在温度为25~60℃下搅拌10~20小时,得到化合物(III);
流程2
Figure PCTCN2021109991-appb-000014
R 1、R 2、R 3、R 4的定义如前文所述;
第一步:
氮气保护下将三氯氧磷滴加入冰浴的N,N-二甲基甲酰胺的二氯甲烷溶液,滴加完后,室温搅拌30分钟,再次降温至0℃,滴加入化合物(IV)的二氯甲烷溶液,室温~60℃反应10~24小时,得到化合物(V);
第二步:
氮气保护下将化合物(V),对氯苯硼酸,碱如碳酸钾,相转移催化剂如四正丁基溴化铵以及催化剂如醋酸钯加入溶剂如水中,体系抽真空氮气置换三次,加热至40~100℃反应2~10小时,得到化合物(VI);
第三步:
将化合物(VI)溶于溶剂如四氢呋喃与甲醇,加入还原剂如硼氢化钠,室温搅拌1~5小时,得到化合物(VII);
第四步:
将化合物(VII)溶于溶剂如二氯甲烷中,加入氯代试剂如氯化亚砜,室温搅拌10~24小时,得到化合物(VIII);
流程3
Figure PCTCN2021109991-appb-000015
R 0、R 1、R 2、R 3、R 4与X 1的定义如前文所述;
第一步:
将化合物(IX)(合成参考:WO 2017212431)(0.51g,2.00mmol)和(R)-3-甲基哌嗪-1-甲酸叔丁酯溶解在溶剂(如二甲基亚砜)中,室温下加入碱如磷酸氢二钾,在温度为90~120℃下搅拌12~48小时,得到化合物(X);
第二步:
将化合物(X)加入酸如氯化氢的1,4-二氧六环溶液,室温搅拌1~3小时,得到化合物(XI);
第三步:
将化合物(XI)和化合物(VIII)溶解在溶剂如乙腈中,加入碱如N,N-二异丙基乙胺,加热到60~90℃搅拌2~8小时,得到化合物(XII);
第四步:
将化合物(XII)溶解在溶剂如2-丁醇中,加入碱如氢氧化钠,加热到90~120℃搅拌12~36小时,再以酸如盐酸酸化,得到化合物(XIII);
第五步:
将化合物(XIII)、化合物(III),缩合剂如1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐、碱如4-二甲氨基吡啶溶解在溶剂如二氯甲烷中,室温搅拌12~36小时,得到化合物(XIV);
流程4
Figure PCTCN2021109991-appb-000016
R 0、R 1、R 2、R 3、R 4与X 1的定义如前文所述;
第一步:
将化合物(XV)(合成参考:Journal of Organic Chemistry,84(8),4814-4829;2019)(0.51g,2.00mmol)和(R)-2-甲基哌嗪溶解在溶剂(如二甲基亚砜)中,室温下加入碱如N,N-二异丙基乙胺,在温度为50~100℃下搅拌12~24小时,得到化合物(XVI);
第二步:
将化合物(XVI)和化合物(VIII)溶解在溶剂如乙腈中,加入碱如N,N-二异丙基乙胺,加热到50~90℃搅拌8~24小时,得到化合物(XVII);
第三步:
将化合物(XVII)溶解在溶剂如水、甲醇和四氢呋喃中,加入碱如氢氧化锂,加热到50~90℃搅拌1~5小时,再以酸如盐酸酸化,得到化合物(XVIII);
第四步:
将化合物(XVIII)、化合物(III),缩合剂如1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐、碱如4-二甲氨基吡啶溶解在溶剂如二氯甲烷中,室温搅拌12~36小时,得到化合物(XIX)。
具体实施方式
定义
除非有相反陈述,否则下列用在说明书和权利要求书中的术语具有下述含义。在本发明中未具体定义的基团具有本领域技术人员公知的本领域通常代表的含义。
在本发明中使用的表示方式“Cx-Cy”表示碳原子数的范围,其中x和y均为整数,例如C3-C8环烷基表示具有3-8个碳原子的环烷基,-C0-C2烷基表示具有0-2个碳原子的烷基,其中-C0烷基是指化学单键。
在本发明中,术语“烷基”指饱和的脂族烃基团,包括1至20个碳原子的直链和支链基团,例如可以是1至18个碳原子、1至12个碳原子、1至8个碳原子、1至6个碳原子或1至4个碳原子的直链和支链基团。非限制性实例包括甲基、乙基、正丙基、异丙基、正丁基、异丁基、叔丁基、仲丁基、正戊基、1,1-二甲基丙基、1,2-二甲基丙基、2,2-二甲基丙基、1-乙基丙基、2-甲基丁基、3-甲基丁基、正己基、1-乙基-2-甲基丙基、1,1,2-三甲基丙基、1,1-二甲基丁基、1,2-二甲基丁基、2,2-二甲基丁基、1,3-二甲基丁基、2-乙基丁基、及其各种支链异构体等。烷基可以是任选取代的或未取代的。
在本发明中,术语“环烷基”指饱和单环或多环环状烃基,其包括3至12个环原子,例如可以是3至12个、3至10个、3至8个或3至6个环原子,或者可以是3、4、5、6元环。单环环基的非限制性实例包含环丙基、环丁基、环戊基、环己基、环庚基、环辛基等。环烷基可以是任选取代的或未取代的。
在本发明中,术语“杂环基”指饱和或部分不饱和单环或多环环状烃基,其包括3至20个环原子,例如可以是3至16个、3至12个、3 至10个、3至8个或3至6个环原子,其中一个或多个环原子选自氮、氧或S(O)m(其中m是整数0至2)的杂原子,但不包括-O-O-、-O-S-或-S-S-的环部分,其余环原子为碳。优选包括3至12个环原子,其中1~4个是杂原子,更优选杂环基环包含3至10个环原子、更优选包括3至8个环原子,最优选5元环或6元环,其中1~4个是杂原子,更优选1~3个是杂原子,最优选1~2个是杂原子。单环杂环基的非限制性实例包含氧杂环丁烷基、吡咯烷基、哌啶基、4-哌啶基、哌嗪基、1,4-二氧六环基、吗啉基、2-吗啉基、4-吗啉基、硫代吗啉基、吡喃基、四氢吡喃基、4-四氢吡喃基、高哌嗪基、二噁烷基、2-二噁烷基等。多环杂环基包括螺环、稠环和桥环的杂环基。杂环基可以是任选取代的或未取代的。
在本发明中,术语“亚杂环基”指具有两个端部单价基团核心的取代或未取代的杂环基,其是从两个端部原子的每个原子上除去一个氢原子所产生的;所述杂环基具有前文所述的含义。“亚杂环基”的非限制性实例包含亚吡咯烷基、亚哌啶基、亚哌嗪基、亚吗啉基等。
在本发明中,术语“卤素”指氟、氯、溴或碘。
在本发明中,“任选”或“任选地”意味着随后所描述的事件或环境可以但不必发生,该说明包括该事件或环境发生或不发生的场合。例如,“任选被烷基取代的杂环基团”意味着烷基可以但不必须存在,该说明包括杂环基团被烷基取代的情形和杂环基团不被烷基取代的情形。
在本发明中,“取代的”指基团中的一个或多个氢原子,优选为最多5个,更优选为1~3个氢原子彼此独立地被相应数目的取代基取代。不言而喻,取代基仅处在它们的可能的化学位置,本领域技术人员能够在不付出过多努力的情况下确定(通过实验或理论)可能或不可能的取代。例如,具有游离氢的氨基或羟基与具有不饱和(如烯属)键的碳原子结合时可能是不稳定的。
所述取代基包括但不限于前文所述的各种基团。
本发明要求保护的化合物不仅包括所述化合物本身,还包括所述化合物的异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐。
本发明所述“药物组合物”表示含有一种或多种本发明所述化合 物异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐及其他化学组分的混合物。其他组分例如药学上可接受的药学可接受的载体、稀释剂和赋形剂。药物组合物的目的是促进对生物体的给药,利于活性成分的吸收进而发挥生物活性。
当在说明书中使用时,术语“包含”包括“由…组成”。
本发明所述“室温”是指15-30℃。
本发明所述“稳定的同位素衍生物”包括:式(I)中任意的氢原子被1-5个氘原子取代得到的同位素取代衍生物、式(I)中任意的碳原子被1-3个碳14原子取代得到的同位素取代衍生物或式(I)中任意的氧原子被1-3个氧18原子取代得到的同位素取代衍生物。
本发明所述“药学上可接受的盐”在Berge,et al.,“Pharmaceutically acceptable salts”,J.Pharm.Sci.,66,1-19(1977)中有讨论,并对药物化学家来说是显而易见,所述的盐是基本上无毒性的,并能提供所需的药代动力学性质、适口性、吸收、分布、代谢或排泄等。
本发明药学上可接受的盐可通过一般的化学方法合成。
一般情况下,盐的制备可以通过游离碱或酸与等化学当量或者过量酸(无机酸或有机酸)或碱在合适的溶剂或溶剂组合物中反应制得。
本发明所述“前药”是指化合物在体内代谢后转换成原始活性化合物。代表性地讲,前药为非活性物质,或者比活性母体化合物活性小,但可以提供方便的操作、给药或者改善代谢特性。
本发明所述“异构体”是指本发明的式(I)化合物的互变异构体、内消旋体、外消旋体、对映异构体、非对映异构体、其混合物形式等。所有这些异构体,包括立体异构体、几何异构体均包含在本发明中。所述几何异构体包括顺反异构体。
本发明所述“溶剂合物”指一种或多种溶剂分子与本发明的化合物或其盐的缔合。形成药学上可接受的溶剂化物的溶剂的实例包括但不限于水、异丙醇、乙醇、甲醇、乙酸乙酯、乙酸等。
本发明包括所述化合物或其盐的任何多晶型物以及任何水合物或其它溶剂合物。
在本发明中,术语“患者”通常指哺乳动物,尤其是人。
在本发明中,术语“肿瘤”包括良性肿瘤和恶性肿瘤,例如癌症。
在本发明中,术语“癌症”包括由BCL-2介导的各种肿瘤,包括但不限于尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、前列腺癌、直肠癌、胰腺癌、脑胶质瘤。
在本发明中,术语“治疗有效量”是指包括可有效治疗或预防由BCL-2介导的相关疾病的本发明化合物的量。
实施例
下面通过实施例的方式进一步说明本发明,但并不因此将本发明限制在所述的实施例范围之中。下列实施例中未注明具体条件的实验方法,按照常规方法和条件,或按照商品说明书选择。
本发明所有化合物的结构可通过核磁共振(1H NMR)和/或质谱检测(MS)鉴定。
1H NMR化学位移(δ)以PPM(parts per million,百万分之几)记录。NMR通过Bruker AVANCE III-400MHz光谱仪进行。合适的溶剂选自氘代氯仿(CDCl 3)、氘代甲醇(CD 3OD)、氘代二甲亚砜(DMSO-d 6)等,四甲基硅烷作为内标(TMS)。
低分辨率质谱(MS)由Agilent 1260 HPLC/6120质谱仪测定,使用Agilent ZORBAX XDB-C18,4.6×50mm,3.5μm。
梯度洗脱条件一:0∶95%溶剂A1和5%溶剂B1,1-2∶5%溶剂A1和95%溶剂B1;2.01-2.50∶95%溶剂A1和5%溶剂B1。百分数为某一溶剂占总溶剂体积的体积百分数。溶剂A1:0.01%甲酸水溶液;溶剂B1:0.01%甲酸的乙腈溶液;百分数为溶质占溶液的体积百分数。
薄层硅胶板是烟台黄海HSGF254或青岛GF254硅胶板。柱层析一般使用烟台黄海100-200或200-300目硅胶作为载体。
制备液相色谱(prep-HPLC)使用Waters SQD2质谱导向高压液相色谱分离仪,XBridge-C18;30X 150mm制备柱,5μm;
方法一:乙腈-水(0.2%甲酸),流速25mL/分钟;方法二:乙腈-水(0.8%碳酸氢铵),流速25mL/分钟;
本发明的已知的起始原料可以采用或按照本领域已知的方法来合成,或可购买自Acros Organics,Aldrich Chemical Company,韶远化学科技(Accela ChemBio Inc)、上海毕得医药、上海阿拉丁化学、上海迈瑞尔化学、百灵威化学、安耐及化学等公司。Venetoclax购自药明览博 (武汉)化学科技有限公司。
实施例中如无特殊说明,反应所用溶剂均为无水溶剂,其中无水四氢呋喃使用市售四氢呋喃,以钠块为除水剂,以二苯甲酮作为指示剂,氩气保护下回流至溶液呈蓝紫色,蒸馏收集,氩气保护下室温储存,其他无水溶剂购自安耐及化学及百灵威化学,所有无水溶剂的转移和使用如无特殊说明,均需在氩气保护下进行。
实施例中如无特殊说明,反应均在氩气氛或氮气氛下进行。
氩气氛或氮气氛是指反应瓶连接一个约1L容积的氩气或氮气气球。
氢气氛是指反应瓶连接一个约1L容积的氢气气球。
氢化反应通常抽真空,充入氢气,反复操作3次。
实施例中如无特殊说明,反应的温度为室温,温度范围是15℃-30℃。
实施例中的反应进程的监测采用薄层色谱法(TLC),反应所使用的展开剂的体系有A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系。溶剂的体积比根据化合物的极性不同而进行调节。
纯化化合物采用的柱层析的洗脱剂的体系和薄层色谱法的展开剂的体系包括A:二氯甲烷和甲醇体系;B:石油醚和乙酸乙酯体系。溶剂的体积比根据化合物的极性不同而进行调节,也可以加入少量的三乙胺和酸性或碱性试剂等进行调节。
中间体1
3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯磺酰胺
Figure PCTCN2021109991-appb-000017
第一步
3,4-二氟-5-硝基苯磺酰胺
将1,2-二氟-3-硝基苯(10.00g,62.89mmol)溶解在氯磺酸(21mL)中,加热至150℃回流搅拌10小时。冷却至室温,在冰浴下向反应液中加入饱和碳酸氢钠水溶液,调节pH值大约为7。用二氯甲烷(100mL ×3)萃取,有机相用饱和食盐水(100mL×2)洗,无水硫酸钠干燥,过滤浓缩得到粗产品3,4-二氟-5-硝基苯磺酰氯。在1000mL三口瓶中加入异丙醇(200mL)和氨水(5mL,37%)于-78℃下搅拌10分钟,将所得的粗产品3,4-二氟-5-硝基苯磺酰氯溶于异丙醇中(30mL),缓慢滴入到上面的异丙醇和氨水混合物中在-78℃,滴加完毕后在-78℃搅拌两小时,加入稀盐酸(1N)调制体系pH值大约为6。升至室温,减压浓缩去除大部分异丙醇溶剂,向其中加入纯水,固体析出,过滤得固体粗品。粗品用二氯甲烷打浆纯化得到目标产物3,4-二氟-5-硝基苯磺酰胺(4.90g,黄色固体)。产率:33%。
1H NMR(400MHz,DMSO-d 6)δ8.38-8.36(m,1H),8.29-8.26(m,1H),7.84(s,2H)。
第二步
3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯磺酰胺
将化合物3,4-二氟-5-硝基苯磺酰胺(1.55g,6.51mmol)、(四氢-2H-吡喃-4-基)甲胺(0.89g,7.73mmol)、N,N-二异丙基乙胺(3.91g,30.31mmol)和乙腈(20.0mL)混合。40℃下搅拌2小时。旋干溶剂,此混合物用50mL水淬灭,用乙酸乙酯(60mL×2)萃取。合并有机相用饱和食盐水(50mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品。柱层析纯化(石油醚/乙酸乙酯=1∶1)得到目标产物3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯磺酰胺(1.66g,黄色固体)。产率:76%。
MS m/z(ESI):334[M+1];
1H NMR(400MHz,DMSO-d 6)δ8.33-8.30(m,2H),7.76-7.74(m,1H),7.45(s,2H),3.86-3.84(m,2H),3.50-3.45(m,2H),3.29-3.23(m,3H),1.59-1.57(m,2H),1.25-1.20(m,2H)。
中间体2的合成步骤参考中间体1。
Figure PCTCN2021109991-appb-000018
中间体3
3-氟-5-硝基-4-(((1-(3-氧杂环丁烷基)哌啶-4-基)甲基)氨基)苯磺酰胺
Figure PCTCN2021109991-appb-000019
第一步
3-氟-5-硝基-4-((哌啶-4-基甲基)氨基)苯磺酰胺
将化合物4-(((2-氟-6-硝基-4-氨磺酰苯基)氨基)甲基)哌啶-1-甲酸叔丁酯(合成参考中间体1的合成步骤)(0.49g,1.12mmol)和三氟乙酸(3mL)、二氯甲烷(9mL)混合,常温下搅拌0.5小时。旋干溶剂,此混合物用三乙胺调节pH值为中性,再次旋干溶剂,得到粗品3-氟-5-硝基-4-((哌啶-4-基甲基)氨基)苯磺酰胺(0.35g,黄色油)。此混合物不经纯化直接用于下一步反应。MS m/z(ESI):333[M+1]。
第二步
3-氟-5-硝基-4-(((1-(3-氧杂环丁烷基)哌啶-4-基)甲基)氨基)苯磺酰胺
将化合物3-氟-5-硝基-4-((哌啶-4-基甲基)氨基)苯磺酰胺(0.35g,1.05mmol)和氧杂环丁烷基-3-酮(0.23g,3.15mmol)、氰基硼氢化钠(0.23g,5.23mmol)和甲醇(8mL)混合,常温下搅拌1.5小时。此混合物用10mL水淬灭,用乙酸乙酯(10mL×2)萃取。合并有机相用饱和食盐水(10mL)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,旋干,残余物用柱层析纯化(石油醚/乙酸乙酯=3∶1)纯化得到目标产物3-氟-5-硝基-4-(((1-(3-氧杂环丁烷基)哌啶-4-基)甲基)氨基)苯磺酰胺(0.32g,黄色固体)。产率:78%。
MS m/z(ESI):389[M+1];
1H NMR(400MHz,CD 3OD)δ8.47-8.46(m,1H),7.72-7.70(m,1H),4.68-4.65(m,2H),4.60-4.59(m,2H),3.57-3.55(m,2H),3.49-3.45(m,1H),2.83-2.80(m,2H),1.89-1.78(m,3H),1.42-1.20(m,4H)。
中间体4
(R)-4′-氯-6-(氯甲基)-3-甲基-2,3,4,5-四氢-1,1′-联苯
Figure PCTCN2021109991-appb-000020
第一步
(R)-2-氯-4-甲基环己-1-烯-1-甲醛
将N,N-二甲基甲酰胺(0.59g,8.00mmol)溶解在二氯甲烷(10mL)中,冰浴降温至0℃,逐滴加入三氯氧膦(0.92g,6.00mmol),保温搅拌30分钟后,自然升至室温,继续搅拌3小时。降温至0℃,缓慢加入(R)-3-甲基环己基-1-酮(合成参考文献:Tetrahedron 73(2017)3202-3212)(0.45g,4.0mmol)的二氯甲烷(5mL)溶液,缓慢升至室温搅拌反应18小时。反应液中加入饱和碳酸氢钠溶液(10mL)淬灭,浓缩除去二氯甲烷,加入乙酸乙酯(25mL)和水(15mL),分离有机相,水相用乙酸乙酯(25mL×2)萃取,合并有机相用饱和食盐水(20mL×2)洗涤,无水硫酸钠干燥,过滤浓缩,得到目标化合物(R)-2-氯-4-甲基环己-1-烯-1-甲醛(0.45g,浅黄色液体),粗品。
MS m/z(ESI):159&161[M+1];
1H NMR(400MHz,CDCl 3)δ10.20(s,0.8H),10.18(s,0.2H),2.63-2.47(m,2H),2.29-2.26(m,1H),2.24-2.15(m,1H),1.90-1.77(m,2H),1.27-1.21(m,1H),1.03(d,J=6.4Hz,3H)。
第二步
(R)-4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-甲醛
单口烧瓶中加入依次(R)-2-氯-4-甲基环己-1-烯-1-甲醛(0.45g,2.80mmol),对氯苯硼酸(0.44g,2.80mmol),四丁基溴化铵(0.91g,2.80mmol),碳酸钾(1.17g,8.50mmol),醋酸钯(0.16g,0.70mmol)和水(15mL),反应体系用氮气置换3次,加热至45℃反应4小时。反应液降至室温,加入乙酸乙酯(25mL),分离有机相,水相用乙酸乙酯萃取(25mL),合并有机相后饱和食盐水洗涤(15mL),无水硫酸钠干燥,过滤,滤液浓缩,残余物用柱层析(石油醚∶乙酸乙酯=96∶4)纯化得到目标化合物(R)-4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯 基]-2-甲醛(0.45g,浅黄色油状物),收率67.5%。
MS m/z(ESI):235&237[M+1];
1H NMR(400MHz,CDCl 3)δ9.41(s,0.9H),9.37(s,0.1H),7.30-7.24(m,2H),7.10-7.08(m,2H),2.54-2.47(m,2H),2.16-2.03(m,2H),1.82-1.75(m,2H),1.21-1.15(m,1H),0.96(d,J=6.4Hz,3H)。
第三步
(R)-(4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲醇
将(R)-4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-甲醛(0.23g,1.00mmol)溶解在四氢呋喃中(2mL),0℃下加入硼氢化钠(57mg,1.50mmol),室温搅拌2小时。反应液滴加饱和氯化铵水溶液(10mL)淬灭,以乙酸乙酯(10mL)萃取,有机相用饱和食盐水(10mL)洗涤,干燥、过滤,滤液浓缩,得到粗品(R)-(4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲醇(0.24g,浅黄色油状物),粗品直接用于下一步,无需纯化。MS m/z(ESI):237&239[M-17]。
第四步
(R)-4′-氯-6-(氯甲基)-3-甲基-2,3,4,5-四氢-1,1′-联苯
将(R)-(4′-氯-5-甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲醇(0.24g,1.00mmol)溶解在二氯甲烷溶液中(2mL),室温下加入二氯亚砜(0.24g,1.50mmol)搅拌1小时。反应液滴加饱和碳酸氢钠水溶液(10mL)淬灭,以二氯甲烷(10mL)萃取,有机相用饱和食盐水(10mL)洗涤,干燥、过滤,滤液浓缩,反应液旋干得到粗产品(R)-4′-氯-6-(氯甲基)-3-甲基-2,3,4,5-四氢-1,1′-联苯(0.26g,浅黄色油状物),粗品直接用于下一步,无需纯化。
中间体5-7的合成参考中间体4的合成步骤。
Figure PCTCN2021109991-appb-000021
中间体8
5-(氯甲基)-6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯
Figure PCTCN2021109991-appb-000022
第一步
7,7-二甲基-8-亚甲基-1,4-二氧杂螺[4.5]癸烷
将化合物甲基三苯基溴化膦(14.00g,39.20mmol)加入无水四氢呋喃(120mL),冷却至-40℃,氮气保护下滴加入正丁基锂的正己烷溶液(2.5M,39.2mmol,15.6mL),滴加完毕后,自然升温至室温搅拌1小时。然后滴加入7,7-二甲基-1,4-二氧杂螺[4.5]癸烷-8-酮(6.00g,32.60mmol),室温反应过夜。反应液用饱和氯化铵水溶液(200mL)淬灭,水相用乙酸乙酯(100mL×2)萃取,有机相合并后用饱和食盐水洗涤(200mL)洗涤,有机相无水硫酸钠干燥,过滤,滤液减压浓缩。残余物硅胶柱层析分离(0~10%乙酸乙酯/石油醚)得到产物7,7-二甲基-8-亚甲基-1,4-二氧杂螺[4.5]癸烷(3.80g,无色油状物)。产率:64%。
1H NMR(400MHz,CDCl 3)δ4.73(s,2H),3.98-3.94(m,4H), 2.43-2.36(m,2H),1.73-1.68(m,2H),1.62(s,2H),1.16(s,6H)。
第二步
4,4-二甲基-7,10-二氧杂二螺[2.2.4 6.2 3]十二烷
将化合物7,7-二甲基-8-亚甲基-1,4-二氧杂螺[4.5]癸烷(0.90g,4.94mmol)溶于二氯甲烷(20mL),在0℃氮气保护下滴加入二乙基锌的甲苯溶液(2M,14.80mmol,7.4mL),滴加完后搅拌0.5小时。随后滴加氯碘甲烷(5.20g,29.60mmol),室温搅拌过夜。加入饱和氯化铵水溶液(30mL)淬灭反应,分离有机相,水相用二氯甲烷萃取(30mL×2),合并有机相用水(30mL×2)和饱和食盐水(20mL)洗涤。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗产物4,4-二甲基-7,10-二氧杂二螺[2.2.4 6.2 3]十二烷(1.50g,黄色油状物)。粗品。
1H NMR(400MHz,CDCl 3)δ3.96-3.91(m,4H),1.69-1.62(m,2H),1.55(s,2H),1.44-1.40(m,2H),0.83(s,6H),0.50-0.46(m,2H),0.11-0.08(m,2H)。
第三步
4,4-二甲基螺[2.5]辛烷-6-酮
将化合物4,4-二甲基-7,10-二氧杂二螺[2.2.4 6.2 3]十二烷(1.50g,粗产物,4.94mmol)溶于盐酸四氢呋喃混合液(2M,14.00mmol,7mL),室温搅拌2小时。反应液加入饱和碳酸氢钠水溶液(30mL)淬灭。用乙酸乙酯(30mL)萃取,有机相分别用水(30mL×2)和饱和食盐水(30mL)洗涤。有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到粗产物4,4-二甲基螺[2.5]辛烷-6-酮(0.70g,黄色油状物)。严率:93.2%。MS m/z(ESI):153[M+1]。
第四步
6-氯-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛
在氮气保护下将三氯氧磷(1.10g,6.90mmol)滴加入冰浴的N,N-二甲基甲酰胺(0.67g,9.20mmol)的二氯甲烷溶液(30mL),滴加完后,室温搅拌30分钟,再次降温至0℃,滴加入4,4-二甲基螺[2.5]辛烷-6-酮(0.70g,4.60mmol)的二氯甲烷溶液(5mL),室温反应过夜。加水(30mL)淬灭反应,分离出有机相,有机相分别用饱和碳酸氢钠溶液(30mL)和饱和食盐水(30mL)洗涤。有机相无水硫酸钠干燥,过滤,滤液减压浓缩,残余物硅胶柱层析分离纯化(100%石 油醚)得到6-氯-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛(0.34g,淡黄色油状物)。产率:37.2%。MS m/z(ESI):199&201[M+1]。
第五步
6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛
在氮气保护下将6-氯-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛(0.34g,1.70mmol)、对氯苯硼酸(0.41g,2.60mmol)、碳酸钾(0.70g,5.10mmol)、四正丁基溴化铵(0.55g,1.70mmol)和醋酸钯(77mg,0.34mmol)加入水(15mL)中。体系抽真空氮气置换三次,升温至50℃反应4小时。冷却至室温,反应液用乙酸乙酯(20mL×2)萃取,合并的有机相用饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用硅胶柱层析分离纯化(0-5%乙酸乙酯/石油醚)得到6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛(0.18g,淡黄色油状物)。产率:38.2%。MS m/z(ESI):275&277[M+1]。
第六步
(6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲醇
将6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-甲醛(0.18g,0.66mmol)溶于四氢呋喃-甲醇混合溶液(v/v=6/1,7mL),加入硼氢化钠(75mg,1.78mmol),室温搅拌2小时。加入饱和氯化铵水溶液(20mL)淬灭反应,反应液用乙酸乙酯(20mL×2)萃取,合并的有机相用饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到(6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲醇(0.16g,无色油状物)。产率:88.2%。MS m/z(ESI):260&262[M-17]。
第七步
5-(氯甲基)-6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯
将(6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲醇(0.16g,0.58mmol)溶于二氯甲烷(10mL),加入氯化亚砜(0.21g,1.74mmol)在室温下搅拌过夜。减压浓缩得到5-(氯甲基)-6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯(0.17g,黄色油状物)。粗品。
1H NMR(400MHz,CDCl 3)δ7.36-7.29(m,2H),7.20-7.13(m,2H),3.88(s,2H),2.20-2.11(m,2H),1.64-1.53(m,2H),0.85(s,6H),0.63-0.57(m,2H),0.24-0.16(m,2H)。
中间体9
7,7,8,8-四甲基-1,4-二氧杂螺[4.5]癸烷
Figure PCTCN2021109991-appb-000023
7,7,8,8-四甲基-1,4-二氧杂螺[4.5]癸烷
将化合物4,4-二甲基-7,10-二氧杂二螺[2.2.4 6.2 3]十二烷(见中间体8第二步)(4.30g,21.9mmol)和二氧化铂(1.00g,4.40mmol)加入醋酸(30mL),反应体系抽真空氢气置换后升温至40℃反应过夜。冷却至室温,加入乙酸乙酯(30mL)稀释,反应液硅藻土过滤,滤饼用乙酸乙酯(20mL)洗涤,滤液减压浓缩得到粗产物7,7,8,8-四甲基-1,4-二氧杂螺[4.5]癸烷(4.50g,无色油状物)。粗品。MS m/z(ESI):199[M+1]。
中间体10的合成参考中间体8的合成步骤。
Figure PCTCN2021109991-appb-000024
实施例1
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)-N-((3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
Figure PCTCN2021109991-appb-000025
第一步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(3-甲基哌嗪-1-基)苯甲酸甲酯
将化合物2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-氟苯甲酸甲酯(合成参考:Journal of Organic Chemistry,84(8),4814-4829;2019)(2.86g,10.00mmol)、(R)-2-甲基哌嗪(3.00g,30.00mmol)、N,N-二异丙基乙胺(3.12g,24.18mmol)和二甲基亚砜(20mL)混合。60℃下搅拌16小时。此混合物用50mL水稀释,乙酸乙酯(60mL×2)萃取。合并有机相用饱和食盐水(50mL×3)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品。柱层析纯化(二氯甲烷/甲醇=90∶10)得到目标产物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(3-甲基哌嗪-1-基)苯甲酸甲酯(2.55g,黄色固体)。产率:70%。
MS m/z(ESI):367[M+1];
1H NMR(400MHz,CDCl 3)δ9.92(s,1H),8.19(d,J=2.4Hz,1H),7.91(d,J=9.2Hz,1H),7.52(d,J=2.4Hz,1H),7.36-7.35(m,1H),6.66-6.63(m,1H),6.43(d,J=1.6Hz,1H),6.34(d,J=2.4Hz,1H),3.79(s,3H),3.54-3.48(m,2H),3.05-3.02(m,1H),2.93-2.84(m,2H),2.77-2.71(m,1H),2.42-2.37(m,1H),1.08(d,J=6.4Hz,3H)。
第二步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯
将化合物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(3-甲基哌嗪 -1-基)苯甲酸甲酯(34mg,0.09mmol)、4′-氯-6-(氯甲基)-3,3-二甲基-2,3,4,5-四氢-1,1′-联苯(合成参考:US 20100298323)(30mg,0.11mmol)、N,N-二异丙基乙胺(36mg,0.28mmol)和乙腈(5.0mL)混合。60℃下搅拌16小时。此混合物用10mL水淬灭,用乙酸乙酯(10mL×2)萃取。合并有机相用饱和食盐水(10mL×2)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品。用制备板纯化(石油醚/乙酸乙酯=1∶1)得到目标产物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯(28mg,无色油状液体)。产率:50%。MS m/z(ESI):599&601[M+1]。
第三步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸
将化合物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯(28mg,0.05mmol)、氢氧化锂(20mg,0.83mmol)、甲醇(1mL)、四氢呋喃(1mL)和水(1mL)混合,60℃下搅拌1.5小时。此混合物用1N盐酸调节pH值为中性,用乙酸乙酯(10mL×2)萃取。合并有机相用饱和食盐水(10mL)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸(25.6mg,粗品)。此混合物不经纯化直接用于下一步反应。MS m/z(ESI):585&587[M+1]。
第四步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)-N-((3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
将化合物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)苯甲酸(26mg,0.04mmol)、3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯磺酰胺(中间体1)(15mg,0.04mmol)、4-二甲氨基吡啶(11mg,0.09mmol)、1-(3-二甲氨基丙基)-3-乙基碳二亚胺盐酸(17mg,0.09 mmol)和二氯甲烷(2.5mL)混合。常温下搅拌16小时。此混合物用10mL水淬灭,用二氯甲烷(10mL×2)萃取。合并有机相用饱和食盐水(10mL)洗涤。将有机相用无水硫酸钠干燥,过滤除去干燥剂,减压脱溶得粗品。制备硅胶板纯化(二氯甲烷/甲醇=15∶1)得到目标产物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3-甲基哌嗪-1-基)-N-((3-氟-5-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺1(15.7mg,黄色固体)。产率:38%。
MS m/z(ESI):900&902[M+1];
1H NMR(400MHz,CD 3OD)δ8.49(s,1H),8.08-8.05(m,1H),7.71-7.64(m,2H),7.50(s,1H),7.44(d,J=3.6Hz,1H),7.36-7.34(m,2H),7.09-7.07(m,2H),6.77(d,J=8.0Hz,1H),6.42-6.36(m,2H),4.04-3.93(m,3H),3.71-3.67(m,2H),3.52-3.38(m,5H),3.28-3.26(m,2H),3.08-3.02(m,1H),2.96-2.93(m,1H),2.65-2.63(m,1H),2.24-2.15(m,2H),2.06-2.04(m,1H),1.93-1.89(m,1H),1.85(d,J=4.0Hz,3H),1.70-1.64(m,3H),1.58-1.56(m,2H),1.39-1.28(m,2H),1.01(s,3H),0.90(s,3H)。
实施例2到7的合成步骤参考实施例1的过程。
Figure PCTCN2021109991-appb-000026
Figure PCTCN2021109991-appb-000027
实施例8
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基 -3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
Figure PCTCN2021109991-appb-000028
第一步
(R)-4-(3-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-氰基苯基)-3-甲基哌嗪-1-甲酸叔丁酯
将化合物2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-氟苯甲腈(合成参考:WO 2017212431)(0.51g,2.00mmol)和(R)-3-甲基哌嗪-1-甲酸叔丁酯(1.20g,6.00mmol)溶解在二甲基亚砜(15mL)中,室温下加入磷酸氢二钾(1.05g,6.00mmol),110℃搅拌36小时。冷却至室温,反应液加水稀释,用乙酸乙酯(30mL×2)萃取,合并有机相用饱和食盐水(30mL×2)洗涤,有机相干燥、浓缩,得粗产品,用硅胶柱层析纯化(石油醚∶乙酸乙酯=3∶1)得到(R)-4-(3-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-氰基苯基)-3-甲基哌嗪-1-甲酸叔丁酯(0.35g,白色固体),收率41%。
MS m/z(ESI):434[M+1];
1H NMR(400MHz,CDCl 3)δ9.29(s,1H),8.19(s,1H),7.69(s,1H),7.49(d,J=8.8Hz,1H),7.42-7.40(m,1H),6.53-6.49(m,2H),6.08(s,1H),40.5-3.83(m,3H),3.21-3.20(m,2H),3.04-2.95(m,2H),1.44(s,9H),1.02(d,J=6.4Hz,3H)。
第二步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(2-甲基哌嗪-1-基)苯甲腈
将(R)-4-(3-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-氰基苯基)-3-甲基哌嗪-1-甲酸叔丁酯(0.35g,0.81mmol)溶解在盐酸二氧六环溶液中 (4M,20.00mmol,5mL),室温搅拌1小时。反应液旋干,得到粗产品(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(2-甲基哌嗪-1-基)苯甲腈(0.38g,粗品),直接用于下一步,无需纯化。MS m/z(ESI):334[M+1]。
第三步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)苯甲腈
将(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(2-甲基哌嗪-1-基)苯甲腈(0.38g,粗品)和4′-氯-6-(氯甲基)-3,3-二甲基-2,3,4,5-四氢-1,1′-联苯基(0.21g,0.80mmol)(合成参考:US 20100298323)溶解在乙腈(10mL)中,加入N,N-二异丙基乙胺(0.31g,2.40mmol),80℃搅拌4小时。冷却至室温,反应液加水稀释,用乙酸乙酯(20mL×2)萃取,合并有机相用饱和食盐水(20mL)洗涤。有机相干燥、浓缩,得粗产品,用硅胶柱纯化(石油醚∶乙酸乙酯=2∶1)得到(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)苯甲腈(0.30g,白色固体),收率66%。
MS m/z(ESI):566&568[M+1];
1H NMR(400MHz,CDCl 3)δ9.40(s,1H),8.19(s,1H),7.68(s,1H),7.43(d,J=8.8Hz,1H),7.42-7.40(m,1H),7.23(d,J=8.4Hz,2H),6.92(d,J=8.4Hz,2H),6.52-6.45(m,2H),6.04(s,1H),3.81-3.78(m,1H),3.16-3.13(m,1H),2.98-2.95(m,1H),2.69-2.65(m,3H),2.54-2.51(m,1H),2.21-2.14(m,2H),2.04-1.97(m,2H),1.89-1.76(m,2H),1.43-1.40(m,2H),1.07(d,J=6.4Hz,3H),0.94(s,3H),0.93(s,3H)。
第四步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)苯甲酸
将化合物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)苯甲腈(0.11g,0.20mmol)溶于2-丁醇(3mL)中,90℃下加入氢氧化钠(160mg,4.00mmol),加热至105℃搅拌24小时。反应液减压脱溶,残余物加入1N盐酸水溶液(4mL),以乙酸乙酯(10mL×2)萃 取,合并有机相用饱和食盐水(10mL)洗涤。将有机相干燥、浓缩,得粗产品,用制备硅胶板纯化(二氯甲烷∶甲醇=10∶1)得到(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)苯甲酸(90mg,白色固体),收率77%。MS m/z(ESI):585&587[M+1]。
第五步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
合成参考实施例1的最后一步的操作步骤,得到目标产物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺8。
MS m/z(ESI):882&884[M+1];
1H NMR(400MHz,CDCl 3)δ10.13(s,1H),9.29(s,1H),8.88(s,1H),8.53-8.50(m,1H),8.22-8.16(m,2H),7.94(d,J=8.8Hz,1H),7.70(s,1H),7.47-7.45(m,1H),7.22(d,J=8.0Hz,2H),6.93-6.89(m,3H),6.56-6.49(m,2H),5.94(s,1H),4.05-4.01(m,2H),3.78-3.74(m,1H),3.45-3.39(m,2H),3.28-3.25(m,2H),3.15-3.12(m,1H),2.96-2.89(m,1H),2.73-2.62(m,3H),2.51-2.48(m,1H),2.20-2.11(m,2H),1.99-1.95(m,3H),1.86-1.83(m,1H),1.79-1.72(m,3H),1.45-1.39(m,4H),1.03(d,J=6.4Hz,3H),0.93(s,3H),0.92(s,3H)。
实施例9到13的合成步骤参考实施例8的过程。
Figure PCTCN2021109991-appb-000029
Figure PCTCN2021109991-appb-000030
实施例14到22的合成步骤参考实施例1的过程。
Figure PCTCN2021109991-appb-000031
Figure PCTCN2021109991-appb-000032
Figure PCTCN2021109991-appb-000033
实施例23到26的合成步骤参考实施例8的过程。
Figure PCTCN2021109991-appb-000034
Figure PCTCN2021109991-appb-000035
实施例27到30的合成步骤参考实施例1。
Figure PCTCN2021109991-appb-000036
Figure PCTCN2021109991-appb-000037
实施例31
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
Figure PCTCN2021109991-appb-000038
第一步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯
将5-(氯甲基)-6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯(中间体8)(0.17g,0.58mmol)、(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(3-甲基哌嗪-1-基)苯甲酸甲酯和N,N-二异丙基乙胺(0.37g,2.90mmol)溶于乙腈(15mL),加热回流3小时。冷却后减压浓缩,将残余物溶于乙酸乙酯(30mL),分别用水(30mL×3)和饱和食盐水(30mL)洗涤,有机相无水硫酸钠干燥,过滤,滤液减压浓缩。残余物硅胶柱 层析分离纯化(10%-40%乙酸乙酯/石油醚)得到(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯(0.10g,白色固体)。产率:27.7%。MS m/z(ESI):625&627[M+1]。
第二步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸
将(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸甲酯(0.10g,0.16mmol)溶于乙醇(3mL)和氢氧化钠水溶液(2N,6.00mmol,3mL),加热至70℃反应2小时。冷却后加水(20mL)稀释,加入1N稀盐酸调节pH至5左右,水相用乙酸乙酯(20mL×2)萃取。合并的有机相用饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩,得到(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸(80mg,白色固体)。产率:81.8%。MS m/z(ESI):611&613[M+1]。
第三步
(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
将化合物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)苯甲酸(20mg,0.03mmol)、3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯甲磺酰胺(合成参考:WO 2018041248A1)(10mg,0.03mmol)、1-乙基-(3-二甲基氨基丙基)碳酰二亚胺盐酸盐(31mg,0.17mmol)和4-二甲氨基吡啶(8mg,0.07mmol)溶于二氯甲烷(5mL),室温搅拌过夜。加入二氯甲烷(20mL)稀释,反应液分别用水(20mL×3)和饱和食盐水(20mL)洗涤,有机相用无水硫酸钠干燥,过滤,滤液减压浓缩。残余物用制备薄层色谱纯化(二氯甲烷/甲醇=15/1)得到目标产物(R)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-(4-((6-(4-氯苯基)-8,8-二甲基螺[2.5]辛-5-烯-5-基)甲基)-3-甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢 -2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺31(16.6mg,黄色固体),产率:55.7%
MS m/z(ESI):908&910[M+1];
1H NMR(400MHz,CDCl 3)δ10.14(brs,1H),9.70(s,1H),8.96-8.82(m,1H),8.57-8.48(m,1H),8.27-8.09(m,2H),8.00-7.90(m,1H),7.75-7.63(m,1H),7.50-7.43(m,1H),7.25-7.19(m,2H),7.01-6.85(m,3H),6.59-6.49(m,2H),5.98(d,J=2.0Hz,1H),4.10-3.95(m,2H),3.48-3.38(m,2H),3.32-3.17(m,4H),3.12-3.03(m,1H),2.87-2.73(m,1H),2.70-2.45(m,3H),2.18-2.04(m,3H),2.03-1.83(m,3H),1.50-1.19(m,6H),0.93-0.78(m,8H),0.60-0.48(m,2H),0.15-0.02(m,2H)。
实施例32到34的合成步骤参考实施例31的过程。
Figure PCTCN2021109991-appb-000039
实施例35到38的合成步骤参考实施例1的过程。
Figure PCTCN2021109991-appb-000040
Figure PCTCN2021109991-appb-000041
实施例39到42的合成步骤参考实施例31的过程。
Figure PCTCN2021109991-appb-000042
Figure PCTCN2021109991-appb-000043
实施例43到46的合成步骤参考实施例8的过程。
Figure PCTCN2021109991-appb-000044
Figure PCTCN2021109991-appb-000045
实施例47到56的合成步骤参考实施例1的过程。
Figure PCTCN2021109991-appb-000046
Figure PCTCN2021109991-appb-000047
Figure PCTCN2021109991-appb-000048
Figure PCTCN2021109991-appb-000049
实施例57
2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((4aR,8aR)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)八氢喹喔啉-1(2H)-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
Figure PCTCN2021109991-appb-000050
实施例57合成参考实施例1的合成步骤,其中第二步用(4aR,8aR)-十氢喹喔啉(合成参考:European Journal of Organic Chemistry,2012(34),6752-6759;2012)代替(R)-2-甲基哌嗪,得到目标化合物2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((4aR,8aR)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)八氢喹喔啉-1(2H)-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺57。
MS m/z(ESI):822&824[M+1];
1H NMR(400MHz,CDCl 3)δ10.21(s,1H),8.93-8.92(m,1H),8.88-8.87(m,1H),8.57-8.53(m,1H),8.20-8.18(m,2H),8.01(d,J=8.4Hz,1H),7.71(d,J=1.6Hz,1H),7.46-7.44(m,1H),7.20(d,J=8.0Hz,2H),6.93(m,1H),6.89(d,J=8.0Hz,2H),6.73-6.70(m,1H),6.57-6.56(m,1H),6.17(d,J=1.6Hz,1H),4.06-4.02(m,2H),3.45-3.41(m,3H),3.29-3.26(m,2H),3.19-3.16(m,1H),3.04-3.01(m,1H),2.81-2.69(m,2H),2.49-2.43(m,2H),2.22-2.19(m,1H),1.98-1.96(m,4H),1.96-1.92(m,4H),1.85-1.82(m,1H),1.76-1.73(m,3H),1.47-1.44(m,3H),1.41-1.35(m,4H),0.96(s,3H),0.93(s,3H)。
实施例58
2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((2R,5R)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2,5-二甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺
Figure PCTCN2021109991-appb-000051
实施例58合成参考实施例1的合成步骤,其中第二步用(2R,5R)-2,5-二甲基哌嗪(合成参考:Organic Chemistry Frontiers,5(23),3402-3405;2018)代替(R)-2-甲基哌嗪,得到目标化合物2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((2R,5R)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-2,5-二甲基哌嗪-1-基)-N-((3-硝基-4-(((四氢-2H-吡喃-4-基)甲基)氨基)苯基)磺酰基)苯甲酰胺58。
MS m/z(ESI):896&898[M+1];
1H NMR(400MHz,CDCl 3)δ10.10(s,1H),9.25(s,1H),8.88(d,J=2.2Hz,1H),8.53-8.51(m,1H),8.20(d,J=2.5Hz,1H),8.19-8.02(m,1H),7.94(d,J=9.2Hz,1H),7.70(d,J=2.4Hz,1H),7.48-7.42(m,1H),7.22(d,J=8.4Hz,2H),6.99-6.87(m,2H),6.59-6.52(m,1H),6.50-6.40(m,1H),5.95(d,J=2.2Hz,1H),4.10-4.01(m,2H),3.70-3.60(m,2H),3.47-3.37(m,2H),3.33-3.21(m,2H),3.19-3.03(m,2H),2.59-2.50(m,2H),2.39-2.31(m,2H),1.97-1.90(m,3H),1.82-1.71(m,2H),1.46-1.33(m,3H),1.28-1.20(m,3H),1.25-1.15(m,4H),1.02-1.00(m,2H),0.95(s,3H),0.93(s,3H)。
实施例59
N-((4-((((S)-1,4-二噁烷-2-基)甲基)氨基)-3-硝基苯基)磺酰基)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((3R,5R)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3,5-二甲基哌嗪-1-基)苯甲酰胺
Figure PCTCN2021109991-appb-000052
实施例59合成参考实施例1的合成步骤,其中第二步用(2R,6R)-2,6-二甲基哌嗪代替(R)-2-甲基哌嗪,得到目标化合物N-((4-((((S)-1,4-二噁烷-2-基)甲基)氨基)-3-硝基苯基)磺酰基)-2-((1H-吡咯并[2,3-b]吡啶-5-基)氧代)-4-((3R,5R)-4-((4′-氯-5,5-二甲基-3,4,5,6-四氢-[1,1′-联苯基]-2-基)甲基)-3,5-二甲基哌嗪-1-基)苯甲酰胺59。
MS m/z(ESI):898&890[M+1];
1H NMR(400MHz,CDCl 3)δ9.16(s,1H),8.86(d,J=2.2Hz,1H),8.61(s,1H),8.22-8.13(m,2H),7.91(d,J=9.1Hz,1H),7.67-7.60(m,1H),7.44(s,1H),7.23(d,J=8.3Hz,2H),6.93(d,J=8.3Hz,2H),6.88(d,J=9.2Hz,1H),6.60-6.48(m,2H),5.94-5.90(m,1H),3.97-3.85(m,2H),3.85-3.74(m,2H),3.67-3.60(m,2H),3.55-3.29(m,3H),3.13-3.02(m,2H),2.79-2.70(m,2H),2.69-2.59(m,2H),2.24-2.15(m,2H),2.12-2.05(m,4H),2.05-1.86(m,2H),0.93-0.91(m,6H),0.69(s,3H),0.67(s,3H)。
生物学实验
BCL-2生物活性抑制的测试
使用荧光偏振实验评估本发明的化合物对BCL-2生物活性的影响实验方法概述如下:
使用荧光偏振原理的亲和性测定方法,通过检测化合物对BCL-2和白血病促凋亡蛋白(BIM)的结合活性的影响,从而评估化合物对BCL-2生物活性的影响。反应缓冲液包含以下组分:PBS(pH 7.4,3mM Na 2HPO 4、155mM NaCl、1mM KH 2PO 4)、1mM DTT;人源重组Bcl-2蛋白(货号10195-H08E)购自北京义翘神州生物技术有限公司,用反应缓冲液稀释成5nM;FITC标记的BIM多肽购自南京金斯瑞生物科技 有限公司,用反应缓冲液稀释成5nM。
将化合物在100%DMSO中溶解稀释至0.1、1、10μM,然后用DMSO进行4倍的系列稀释至最低浓度为0.0061、0.061、0.61nM,每个浓度点再使用反应缓冲液稀释50倍。
向黑色384孔检测板中添加3μL化合物溶液和12μL BCL-2溶液,混合均匀后室温孵育15分钟。随后加入15μL FITC-BIM溶液,将反应混合物在室温避光孵育30分钟后,立即在Envision多功能酶标仪(Perkin Elmer)上进行荧光偏振的检测,480nm为激发波长,535nm为发射波长。该实验中,未加BCL-2蛋白组作为阴性对照(100%抑制),加BCL-2蛋白但未加化合物组作为阳性对照(0%抑制)。化合物对BCL-2亲和性的抑制百分比可以用以下公式计算:
化合物IC50值由8个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((logIC 50-X)*slope factor))
其中Y为抑制百分比,X为待测化合物浓度的对数值,Bottom为最大抑制百分比,Top为最小抑制百分比,slope factor为曲线斜率系数。
BCL-XL生物活性抑制的测试
使用荧光偏振实验评估本发明的化合物对BCL-XL生物活性的影响
实验方法概述如下:
使用荧光偏振原理的亲和性测定方法,通过检测化合物对BCL-XL和BIM的结合活性的影响,从而评估化合物对BCL-XL生物活性的影响。反应缓冲液包含以下组分:PBS(pH 7.4,3mM Na 2HPO 4、155mM NaCl、1mM KH 2PO 4)、1mM DTT;人源重组Bcl-XL蛋白(货号10455-H08E)购自北京义翘神州生物技术有限公司,用反应缓冲液稀释成10nM;FITC标记的BIM多肽购自南京金斯瑞生物科技有限公司,用反应缓冲液稀释成10nM。
将化合物在100%DMSO中溶解稀释至1μM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061nM,每个浓度点再使用反应缓冲液稀释50倍。
向黑色384孔检测板中添加3μl化合物溶液和12μL BCL-XL溶液,混合均匀后室温孵育15分钟。随后加入15μL FITC-BIM溶液, 将反应混合物在室温避光孵育30分钟后,立即在Envision多功能酶标仪(Perkin Elmer)上进行荧光偏振的检测,480nm为激发波长,535nm为发射波长。该实验中,未加BCL-XL蛋白组作为阴性对照(100%抑制),加BCL-XL蛋白但未加化合物组作为阳性对照(0%抑制)。化合物对BCL-XL亲和性的抑制百分比可以用以下公式计算:
化合物IC 50值由8个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((logIC 50-X)*slope factor))
其中Y为抑制百分比,X为待测化合物浓度的对数值,Bottom为最大抑制百分比,Top为最小抑制百分比,slope factor为曲线斜率系数。
RS4;11细胞(急性淋巴白血病细胞)半数有效抑制浓度IC 50的测定
使用发光法细胞活力检测实验评估本发明的化合物对RS4;11细胞增殖的影响。
实验方法概述如下:
使用CellTilter-Glo(CTG)检测试剂盒,通过采用一种独特的、高灵敏度且稳定性的荧光素酶检测活细胞新陈代谢的关键指标ATP,试验中产生的发光信号和培养基中的有活力细胞数呈正比,从而检测RS4;11的细胞增殖状况。
CellTilter-Glo试剂(Promega,G7572)由CellTilter-Glo冻干粉和CellTilter-Glo缓冲液组成,使用时将冻干粉溶解到缓冲液中即可。
RS4;11细胞(ATCC,CRL-1873)培养在RPMI1640完全培养基(Thermofisher,72400-047)中含10%FBS(GBICO,10099-141)和100units/ml青链霉素混合液(Thermofisher,15140122),当细胞在培养容器中覆盖率达80-90%时,用0.25%胰酶(含EDTA)(Thermofisher,25200056)消化吹散后种植于白色384孔板(Thermofisher,164610),然后384孔板置于37℃,5%CO 2的培养箱中培养过夜。将化合物在100%DMSO中溶解稀释至5mM,然后用DMSO进行4倍的系列稀释至最低浓度为0.061μM,每个浓度点再使用FBS-free的RPMI1640培养基稀释50倍。如果化合物IC 50值非常低,可以降低化合物的起始浓度。过夜后每孔加入3μL培养基稀释后的化合物,轻轻离心混匀,其中,不加细胞组作为阴性对照(100%抑制),加0.2%DMSO组作为阳性对照(0%抑制)。该384孔板置于37℃,5%CO 2的培养箱中继续 培养,48小时后取出平衡至室温,每孔加15μl CTG试剂,置于摇床上轻轻摇动3分钟以确保细胞裂解充分,放置10分钟使冷光信号稳定,然后用EnVision(Perkin Elmer)读取冷光信号。
化合物对RS4;11细胞增殖抑制的百分比可以用以下公式计算:
抑制百分比=100-100*(signal化合物-signal阴性对照)/(signal阳性对照-signal阴性对照)
化合物IC 50值由8个浓度点用XLfit(ID Business Solutions Ltd.,UK)软件通过以下公式计算:
Y=Bottom+(Top-Bottom)/(1+10^((LogIC 50-X)*slope factor))
其中Y为抑制百分比,Bottom为the bottom plateau of the curve(S型曲线的底部平台值),Top为the top plateau of the curve(S型曲线的顶部平台值),X为待测化合物浓度的对数值。
上述体外BCL-2和BCL-XL蛋白活性检测实验的结果见下表1,
细胞实验的结果见表2。
表1:BCL-2和BCL-XL蛋白活性检测结果
化合物编号 FP BCL-2IC 50(nM) FP BCL-XL IC 50(nM)
1 2.05 >1000
2 2.56 >1000
3 3.06 >1000
4 2.29 742.8
5 1.54 >1000
6 0.90 >1000
7 >100 >1000
8 4.55 >1000
9 5.03 >1000
10 8.21 >1000
11 12.8 >1000
12 4.89 >1000
13 >100 >1000
14 2.35 >1000
15 5.99 >1000
16 3.41 >1000
17 1.46 952.96
18 1.32 >1000
19 2.13 >1000
20 2.57 >1000
21 1.97 >1000
22 1.49 >1000
23 9.37 >1000
24 6.3 >1000
25 2.71 >1000
26 1.63 >1000
27 5.12 >1000
28 1.37 >1000
29 1.6 >1000
30 0.84 830.51
31 3.57 >1000
32 1.8 >1000
33 3.16 >1000
34 3.33 >1000
35 2.96 >1000
36 4.78 >1000
37 3.15 >1000
38 5.24 >1000
39 2.68 >1000
40 1.77 >1000
41 1.28 >1000
42 1.53 >1000
43 6.33 >1000
44 4.98 >1000
45 4.22 >1000
46 1.09 >1000
47 >100 >1000
48 63.79 >1000
49 >100 >1000
50 >100 >1000
51 >100 >1000
52 1.9 >1000
53 >100 >1000
54 11 >1000
55 2.3 >1000
56 82 >1000
57 >500 >1000
58 29.86 >1000
59 73.22 >1000
表2:RS4;11细胞活性检测结果
化合物编号 RS4;11IC 50(nM)
1 4.73
2 11.07
3 6.4
4 5.8
5 2.75
6 2.37
7 >500
8 6.2
9 26
10 68
11 42
12 16
13 >500
14 3.8
15 19
16 8.8
17 3.1
18 2.4
19 4.25
20 8.8
21 8.8
22 4.1
23 51
24 24
25 32
26 14
27 16
28 6.55
29 4.2
30 2.3
31 8.1
32 6.6
33 16
34 6.6
35 17
36 21
37 22
38 32
39 14.5
40 15
41 8
42 10
43 161
44 54
45 52
46 18
47 486
48 229
49 >500
50 >500
51 >500
52 5.1
53 >500
54 25
55 5.7
56 224
57 >1000
58 141.94
59 193
Venetoclax 5.5
由上述实验结果可知,本发明的实施例化合物能够有效选择性的抑制BCL-2的活性,对BCL-XL抑制较弱。可以用于治疗BCL-2家族蛋白异常过量表达导致的多种癌症:尤其是急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、直肠癌、前列腺癌、胰腺癌、脑胶质瘤等。并且避免由BCL-XL抑制引起的毒副作用,如血小板降低。部分化合物也能有效抑制RS4;11急性淋巴细胞增殖。对急性淋巴性白血病等恶性血液病有较强的抑制疗效。
对于本领域技术人员来说,很明显,本公开不局限于上述说明性的实施例,并且在不背离本公开实质特性的条件下,其可以通过其它具体形式来具体实施。因此,期望在各方面都认为这些实施例是说明性的和非限制性的,应参照的是附加的权利要求,而不是上述实施例,且由此在权利要求的等效含义和范围内的所有变化都被包括在其中。

Claims (10)

  1. 式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,
    Figure PCTCN2021109991-appb-100001
    其中:
    X 1选自任选被取代的含有一或二个选自N、O、S的杂原子的6元饱和杂环基,其中所述任选的取代基选自含有一或二个选自N、O、S的杂原子的4元饱和杂环基;优选地,X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;进一步优选地,X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;最优选地,X 1选自(S)-1,4-二氧六环-2-基、(R)-1,4-二氧六环-2-基、四氢吡喃-4-基、1-(氧杂环丁烷-3-基)哌啶-4-基、(S)-4-(氧杂环丁烷-3-基)吗啉-2-基;
    X 2选自含有一或二个N原子的5-6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;优选地,X 2选自含有一或二个N原子的6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;进一步优选地,X 2选自
    Figure PCTCN2021109991-appb-100002
    其中,环上任选可被一个C1~C4的烷基所取代;最优选地,X 2选自
    Figure PCTCN2021109991-appb-100003
    R 0选自氢、卤素;优选地,R 0选自氢、氟、氯;最优选地,R 0选 自氢、氟;
    R 1、R 2、R 3、R 4各自独立地选自氢、C1-C6烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~6元环烷基;优选地,R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基;进一步优选地,R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基;最优选地,R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 3和R 4可以与所连接的碳原子一起组成环丙基。
  2. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
    X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;
    X 2选自含有一或二个N原子的6元亚杂环基;其中,环上任选可被一或二个C1~C4的烷基所取代;
    R 0选自氢、卤素;
    R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基。
  3. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
    X 1选自任选被取代的含有一或二个选自N、O的杂原子的6元饱和杂环基,其中所述任选的取代基选自氧杂环丁烷基;
    X 2
    Figure PCTCN2021109991-appb-100004
    其中,环上任选可被一个C1~C4的烷基所取代;
    R 0选自氢、氟、氯;
    R 1、R 2、R 3、R 4各自独立地选自氢、C1-C4烷基,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成3~4元环烷基。
  4. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
    X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;
    X 2
    Figure PCTCN2021109991-appb-100005
    其中,环上任选可被一个C1~C4的烷基所取代;
    R 0选自氢、氟、氯;
    R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基。
  5. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
    X 1选自1,4-二氧六环基、四氢吡喃基、N-氧杂环丁烷基哌啶基、N-氧杂环丁烷基吗啉基;
    X 2选自
    Figure PCTCN2021109991-appb-100006
    R 0选自氢、氟;
    R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 1和R 2或R 3和R 4可以与所连接的碳原子一起组成环丙基。
  6. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,其中:
    X 1选自(S)-1,4-二氧六环-2-基、(R)-1,4-二氧六环-2-基、四氢吡喃-4-基、1-(氧杂环丁烷-3-基)哌啶-4-基、(S)-4-(氧杂环丁烷-3-基)吗啉-2-基;
    X 2选自
    Figure PCTCN2021109991-appb-100007
    R 0选自氢、氟;
    R 1、R 2、R 3、R 4各自独立地选自氢或甲基且R 1、R 2、R 3、R 4不全是氢,其中R 3和R 4可以与所连接的碳原子一起组成环丙基。
  7. 根据权利要求1的式(I)化合物,其异构体、前药、溶剂合物、稳定的同位素衍生物或其药学上可接受的盐,其选自:
    Figure PCTCN2021109991-appb-100008
    Figure PCTCN2021109991-appb-100009
    Figure PCTCN2021109991-appb-100010
    Figure PCTCN2021109991-appb-100011
    Figure PCTCN2021109991-appb-100012
  8. 根据权利要求1-7任一项的化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐在制备用作BCL-2抑制剂的药物中的用途。
  9. 根据权利要求1-7任一项的化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐在制备用于治疗或预防由BCL-2介导的相关疾病的药物中的用途,所述相关疾病例如肿瘤,所述肿瘤例如急性淋巴性白血病的恶性血液病、肺癌、乳腺癌、卵巢癌、直肠癌、前列腺癌、胰腺癌、脑胶质瘤。
  10. 一种药物组合物,其含有根据权利要求1-7任一项的化合物或其异构体、前药、溶剂合物、稳定的同位素衍生物或药学上可接受的盐,任选的一种或多种其它BCL-2抑制剂,以及一种或多种药学上可接受的载体、稀释剂和赋形剂。
PCT/CN2021/109991 2020-08-06 2021-08-02 作为bcl-2抑制剂的杂环化合物 WO2022028353A1 (zh)

Priority Applications (8)

Application Number Priority Date Filing Date Title
US18/019,701 US20230271959A1 (en) 2020-08-06 2021-08-02 Heterocyclic compound as bcl-2 inhibitor
KR1020237007712A KR20230065986A (ko) 2020-08-06 2021-08-02 Bcl-2 억제제로서의 헤테로시클릭 화합물
CA3190686A CA3190686A1 (en) 2020-08-06 2021-08-02 Heterocyclic compound as bcl-2 inhibitor
EP21852552.5A EP4194454A1 (en) 2020-08-06 2021-08-02 Heterocyclic compound as bcl-2 inhibitor
AU2021319597A AU2021319597A1 (en) 2020-08-06 2021-08-02 Heterocyclic compound as BCL-2 inhibitor
CN202180057392.3A CN116419753A (zh) 2020-08-06 2021-08-02 作为bcl-2抑制剂的杂环化合物
JP2023508059A JP2023537032A (ja) 2020-08-06 2021-08-02 Bcl-2阻害剤としてのヘテロ環化合物
MX2023001618A MX2023001618A (es) 2020-08-06 2021-08-02 Compuestos heterocíclicos como inhibidores de bcl-2.

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202010782972.6A CN114057728A (zh) 2020-08-06 2020-08-06 作为bcl-2抑制剂的杂环化合物
CN202010782972.6 2020-08-06

Publications (1)

Publication Number Publication Date
WO2022028353A1 true WO2022028353A1 (zh) 2022-02-10

Family

ID=80117013

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2021/109991 WO2022028353A1 (zh) 2020-08-06 2021-08-02 作为bcl-2抑制剂的杂环化合物

Country Status (9)

Country Link
US (1) US20230271959A1 (zh)
EP (1) EP4194454A1 (zh)
JP (1) JP2023537032A (zh)
KR (1) KR20230065986A (zh)
CN (2) CN114057728A (zh)
AU (1) AU2021319597A1 (zh)
CA (1) CA3190686A1 (zh)
MX (1) MX2023001618A (zh)
WO (1) WO2022028353A1 (zh)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN115260191B (zh) * 2022-09-29 2022-12-27 上海睿跃生物科技有限公司 哌啶类化合物及其制备方法和应用

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298323A1 (en) 2008-12-04 2010-11-25 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
CN103153993A (zh) * 2010-05-26 2013-06-12 Abbvie公司 治疗癌症和免疫与自身免疫疾病的细胞程序死亡诱导药剂
WO2014158528A1 (en) * 2013-03-14 2014-10-02 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2017212431A1 (en) 2016-06-09 2017-12-14 Dr. Reddy’S Laboratories Limited Solid forms of venetoclax and processes for the preparation of venetoclax
WO2018009444A1 (en) * 2016-07-06 2018-01-11 Concert Pharmaceuticals, Inc. Deuterated venetoclax
WO2018041248A1 (zh) 2016-09-01 2018-03-08 北京赛林泰医药技术有限公司 Bcl-2选择性抑制剂及其制备和用途
CN109311871A (zh) * 2016-08-05 2019-02-05 密歇根大学董事会 作为bcl-2抑制剂的n-(苯基磺酰基)苯甲酰胺及相关化合物
CN110177788A (zh) * 2017-01-07 2019-08-27 上海复尚慧创医药研究有限公司 作为bcl-2选择性凋亡诱导剂的化合物
WO2021116929A1 (en) * 2019-12-13 2021-06-17 Universita' Degli Studi Di Padova Composition comprising the bh3-mimetic abt-199 (venetoclax) and at least one inhibitor of the nadph production

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20100298323A1 (en) 2008-12-04 2010-11-25 Abbott Laboratories Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
CN103153993A (zh) * 2010-05-26 2013-06-12 Abbvie公司 治疗癌症和免疫与自身免疫疾病的细胞程序死亡诱导药剂
WO2014158528A1 (en) * 2013-03-14 2014-10-02 Abbvie Inc. Apoptosis-inducing agents for the treatment of cancer and immune and autoimmune diseases
WO2017212431A1 (en) 2016-06-09 2017-12-14 Dr. Reddy’S Laboratories Limited Solid forms of venetoclax and processes for the preparation of venetoclax
WO2018009444A1 (en) * 2016-07-06 2018-01-11 Concert Pharmaceuticals, Inc. Deuterated venetoclax
CN109311871A (zh) * 2016-08-05 2019-02-05 密歇根大学董事会 作为bcl-2抑制剂的n-(苯基磺酰基)苯甲酰胺及相关化合物
WO2018041248A1 (zh) 2016-09-01 2018-03-08 北京赛林泰医药技术有限公司 Bcl-2选择性抑制剂及其制备和用途
CN110177788A (zh) * 2017-01-07 2019-08-27 上海复尚慧创医药研究有限公司 作为bcl-2选择性凋亡诱导剂的化合物
WO2021116929A1 (en) * 2019-12-13 2021-06-17 Universita' Degli Studi Di Padova Composition comprising the bh3-mimetic abt-199 (venetoclax) and at least one inhibitor of the nadph production

Non-Patent Citations (8)

* Cited by examiner, † Cited by third party
Title
ANDREW, J.JOEL, D. ET AL., NATURE MEDICINE, vol. 19, no. 2, 2013, pages 202
BENJAMIN, D.ISAAC, J. ET AL., J. CLIN. ONCOL., vol. 26, no. 25, 2008, pages 4180
BERGE ET AL.: "Pharmaceutically acceptable salts", J. PHARM. SCI., vol. 66, 1977, pages 1 - 19
JOURNAL OF ORGANIC CHEMISTRY, vol. 84, no. 8, 2019, pages 4814 - 4829
ORGANIC CHEMISTRY FRONTIERS, vol. 5, no. 23, 2018, pages 3402 - 3405
SYNTHESIS CAN BE REFERRED TO: EUROPEAN JOURNAL OF ORGANIC CHEMISTRY, no. 34, 2012, pages 6752 - 6759
TETRAHEDRON, vol. 73, 2017, pages 3202 - 3212
VALENTIN, R.GRABLOW, S. ET AL., BLOOD, vol. 132, no. 12, 2018, pages 1248

Also Published As

Publication number Publication date
JP2023537032A (ja) 2023-08-30
KR20230065986A (ko) 2023-05-12
MX2023001618A (es) 2023-05-17
EP4194454A1 (en) 2023-06-14
US20230271959A1 (en) 2023-08-31
AU2021319597A1 (en) 2023-03-30
CA3190686A1 (en) 2022-02-10
CN116419753A (zh) 2023-07-11
CN114057728A (zh) 2022-02-18

Similar Documents

Publication Publication Date Title
CN110483501B (zh) 作为bcl-2抑制剂的n-(苯基磺酰基)苯甲酰胺及相关化合物
CN105228997B (zh) Carm1抑制剂及其用途
CA3177261A1 (en) Benzothiazolyl biaryl compound, and preparation method and use
WO2016131381A1 (en) Fused-ring compounds, pharmaceutical composition and uses thereof
JP5897566B2 (ja) 環式n,n’−ジアリールチオ尿素及びn,n’−ジアリール尿素−アンドロゲン受容体アンタゴニスト、抗癌剤、その調製のための方法及び使用
JP2022516469A (ja) ユビキチン特異的プロテアーゼ1を阻害するための組成物
EA029771B1 (ru) Химические соединения
WO2022135365A1 (en) Disubstituted cyclopentane kinase inhibitors
WO2015144021A1 (zh) 取代氮杂环类衍生物、含其的药物组合物及其在抗肿瘤中的应用
JP2022533740A (ja) メチル基及びトリフルオロメチル基を含む二置換スルファミド系選択的bcl-2阻害剤
CN115698023A (zh) 作为mcl-1抑制剂的大环2-氨基-3-氟-丁-3-烯酰胺
WO2014055938A1 (en) Novel compounds, their preparation and their uses
CN113045559B (zh) 一种二芳基脲类PI3Kα/mTOR双靶点抑制剂及其药物组合物和应用
CN114195814A (zh) 羟基萘酮-苯硼酸类化合物、制备方法和用途
WO2022028353A1 (zh) 作为bcl-2抑制剂的杂环化合物
WO2022089463A1 (zh) Bcl-2蛋白凋亡诱导剂及应用
WO2022148439A1 (zh) 作为bcl-2抑制剂的杂环化合物
WO2023131118A1 (zh) 基于bcl-2家族蛋白配体化合物开发的蛋白降解剂及它们的应用
WO2022184049A1 (zh) Plk4抑制剂及其用途
WO2016001077A1 (en) Compounds inhibiting the enzyme monopolar spindle 1 kinase,pharmaceutical compositions and uses thereof
CN114751899A (zh) 一种二芳基脲类mTOR激酶抑制剂及其药物组合物和应用
CN106488918B (zh) 三唑并嘧啶酮或三唑并吡啶酮衍生物及其用途
EP4310088A1 (en) Heterocyclic jak inhibitor
TW202214631A (zh) 作為Akt激酶抑制劑的化合物
CN114728986A (zh) 作为mcl-1抑制剂的大环磺酰基衍生物

Legal Events

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

Ref document number: 21852552

Country of ref document: EP

Kind code of ref document: A1

ENP Entry into the national phase

Ref document number: 3190686

Country of ref document: CA

ENP Entry into the national phase

Ref document number: 2023508059

Country of ref document: JP

Kind code of ref document: A

NENP Non-entry into the national phase

Ref country code: DE

ENP Entry into the national phase

Ref document number: 2021852552

Country of ref document: EP

Effective date: 20230306

ENP Entry into the national phase

Ref document number: 2021319597

Country of ref document: AU

Date of ref document: 20210802

Kind code of ref document: A