WO2022170947A1 - 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用 - Google Patents

一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用 Download PDF

Info

Publication number
WO2022170947A1
WO2022170947A1 PCT/CN2022/073358 CN2022073358W WO2022170947A1 WO 2022170947 A1 WO2022170947 A1 WO 2022170947A1 CN 2022073358 W CN2022073358 W CN 2022073358W WO 2022170947 A1 WO2022170947 A1 WO 2022170947A1
Authority
WO
WIPO (PCT)
Prior art keywords
membered
heterocyclyl
heterocyclic group
cycloalkyl
alkyl
Prior art date
Application number
PCT/CN2022/073358
Other languages
English (en)
French (fr)
Inventor
郑苏欣
谢成英
郑明月
陆晓杰
乔刚
Original Assignee
苏州阿尔脉生物科技有限公司
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 苏州阿尔脉生物科技有限公司 filed Critical 苏州阿尔脉生物科技有限公司
Priority to CN202280007411.6A priority Critical patent/CN116600808A/zh
Publication of WO2022170947A1 publication Critical patent/WO2022170947A1/zh

Links

Images

Classifications

    • 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
    • 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

Definitions

  • the invention belongs to the technical field of KRAS mutant G12C inhibitors, in particular to a tetrahydronaphthyridine derivative as a KRAS mutant G12C inhibitor, a preparation method and application thereof.
  • the RAS gene is the first discovered human tumor gene.
  • RAS proteins are activated by binding to GTP under the catalysis of guanine nucleotide exchange factor (GEF), while GTPase activating proteins (GAPs) catalyze the hydrolysis of RAS-bound GTP into a GDP-terminating active state to inhibit RAS activity.
  • GEF guanine nucleotide exchange factor
  • GAPs GTPase activating proteins
  • RAS proteins By cycling between active (GTP-bound) and inactive (GDP-bound) states, RAS proteins transduce upstream signals received from a variety of tyrosine kinases to downstream effectors to regulate cell proliferation, survival, migration, and processes such as apoptosis. Because RAS proteins are central to the axis of many important cellular signaling networks, and these signals are associated with multiple tumor markers, overactive RAS signaling may ultimately lead to tumorigenesis.
  • KRAS mutations are most common in KRAS (85%), while NRAS (12%) and HRAS (3%) are less common, and KRAS mutations are common in pancreatic cancer, colorectal cancer, lung cancer, gallbladder cancer, And thyroid cancer, etc.
  • the abnormal expression of KRAS accounts for up to 20% of all cancers, and 80% of KRAS mutations are missense mutations of a single amino acid substitution at codon 12.
  • KRAS G12C mutation accounts for 12% of all KRAS mutations, but it has a higher proportion in lung cancer, especially non-small cell lung cancer (14%).
  • Mutations in oncogenes, such as EGFR, ALK, and BRAF are mutually exclusive, suggesting that KRAS mutations are unique in lung cancer and that they may serve as an important indicator of tumor prognosis.
  • KRAS G12C mutant Due to the protein structure of KRAS without obvious binding sites, after more than 30 years of research efforts by scientists, there is still no KRAS inhibitor with sufficient safety and efficacy in clinical practice. In recent years, breakthroughs in the study of covalent inhibitors of KRAS mutants have made it possible to target KRAS mutants through allosteric sites.
  • the small molecule covalently bound to the cysteine replaced by the missense mutation is more likely to bind to the GDP-binding KRAS protein to reduce the affinity of GTP to KRAS, while preventing GEF from catalyzing the replacement of GDP by GTP, Locking the KRAS G12C mutant in an inactive state.
  • the object of the present invention is to provide a tetrahydronaphthyridine derivative, a pharmaceutically acceptable salt thereof, a tautomer or a stereoisomer thereof as a KRAS mutant G12C inhibitor isomers in order to screen out compounds with excellent properties in terms of efficacy, safety and selectivity as KRAS mutant G12C inhibitors.
  • Another object of the present invention is to provide a method for the preparation of the derivatives, their pharmaceutically acceptable salts, their tautomers or their stereoisomers.
  • the present invention provides a tetrahydronaphthyridine derivative, a pharmaceutically acceptable salt thereof, a tautomer or a stereoisomer thereof, wherein the structure of the tetrahydronaphthyridine derivative is as follows Formula (I) shows:
  • R 1 is selected from H or F
  • R 2 is selected from 6-12-membered aryl or 5-12-membered heteroaryl, wherein said aryl or heteroaryl is optionally substituted by 1-4 R c ;
  • R c is selected from halogen, hydroxy, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl or 3-8 membered heterocycle group, wherein said alkyl, alkenyl, cycloalkyl, R a , R b or heterocyclyl is optionally substituted with 1-3 R c1 ;
  • R a and R b are each independently selected from H, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclyl, or R a and R b together with the atoms to which they are attached may further form 4-7 membered heterocyclic group;
  • R c1 is selected from halogen, hydroxy, -NR a R b , cyano, C1-C3 alkyl or C1-C3 alkoxy.
  • L 1 is selected from bond, O, S, NH or NCH 3 ;
  • R 3 is selected from C1-C3 alkyl, C1-C3 alkenyl, C3-C7 cycloalkyl, 3-8-membered heterocyclic group, 4-10-membered fused heterocyclic group, 5-10-membered bridged heterocyclic group cyclic group, 5-10-membered spiro heterocyclic group, 6-12-membered aryl group or 5-12-membered heteroaryl group, wherein the alkyl group, alkenyl group, cycloalkyl group, heterocyclic group, fused heterocyclic group Cyclyl, bridged heterocyclyl, spiroheterocyclyl, aryl or heteroaryl may be optionally substituted with 1-4 R c2 .
  • R c2 is selected from halogen, hydroxyl, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl, 3-8 membered heterocyclyl , 4-10-membered fused heterocyclic group, 5-10-membered bridged heterocyclic group, 5-10-membered spiro heterocyclic group, -C(O)R c1 , -C(O)NR a R b , - NR a C(O)R b , 6-12-membered aryl group or 5-12-membered heteroaryl group, wherein the alkyl, alkenyl, cycloalkyl, heterocyclyl, fused heterocyclyl, bridge Heterocyclyl, spiroheterocyclyl, R a , R b , aryl or heteroaryl optionally substitute
  • L 2 is selected from C3-C8 cycloalkyl, C5-C10 spirocycloalkyl, C4-C10 fused cycloalkyl, C5-C10 bridged cycloalkyl, 3-8-membered heterocyclyl, 5-10-membered Condensed heterocyclic group, 5-10-membered bridged heterocyclic group, 5-10-membered spiro heterocyclic group, 6-12-membered aryl group, 5-12-membered heteroaryl group, wherein the cycloalkyl, spirocycloalkyl, fused cycloalkyl, bridged cycloalkyl, heterocyclyl, fused heterocyclyl, bridged heterocyclyl, spiroheterocyclyl, aryl, heteroaryl optionally surrounded by 1-4 R c replaced;
  • R 4 is selected from -C(O)R d or -S(O) 2 R d ;
  • R d is selected from C2-C4 alkenyl or C2-C4 alkynyl, wherein the alkenyl and alkynyl can be optionally further substituted by 1-3 R d1 ;
  • R d1 is selected from halogen, cyano, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclyl, wherein said alkyl, cycloalkyl or heterocyclyl is optionally replaced by 1- 3 R c substitutions;
  • heteroatoms in the heterocyclic group and the heteroaryl group in the formula (I) are 1-4 and are selected from one or more of oxygen, nitrogen and sulfur.
  • R e1 , R e2 or R e3 are independently selected from H, halogen, cyano, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclic group, and the above-mentioned alkyl, ring Alkyl or heterocyclyl is optionally substituted with 1-3 halogens, C1-C3 alkyl, C3-C7 cycloalkyl, 4-7 membered heterocyclyl, hydroxy or amino;
  • heteroatoms in the heterocyclic group are 1-3 one or more selected from oxygen, nitrogen and sulfur;
  • R 2 is selected from 6-12-membered aryl or 5-12-membered heteroaryl, wherein said aryl or heteroaryl is optionally substituted by 1-4 R; wherein, R c is selected from halogen, hydroxy, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl or 3-8 membered heterocycle group, wherein said alkyl, alkenyl, cycloalkyl, R a , R b or heterocyclyl is optionally substituted with 1-3 R c1 ;
  • R c1 is selected from halogen, hydroxyl, -NR a R b , cyano, C1-C3 alkyl or C1-C3 alkoxy;
  • R 3 is selected from C1-C3 alkyl, C1-C3 alkenyl, C3-C7 cycloalkyl, 3-8 membered heterocyclyl, 4-10 membered fused heterocyclyl, 5-10 membered heterocyclyl Bridged heterocyclyl with 5-10 members, spiro heterocyclyl with 5-10 members, aryl with 6-12 members or heteroaryl with 5-12 members, wherein the alkyl, alkenyl, cycloalkyl, heterocycle base, fused heterocyclyl, bridged heterocyclyl, spiroheterocyclyl, aryl or heteroaryl may be optionally substituted with 1-4 R c2 ;
  • R c2 is selected from halogen, hydroxyl, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl, 3-8 membered heterocyclyl , 4-10-membered fused heterocyclic group, 5-10-membered bridged heterocyclic group, 5-10-membered spiro heterocyclic group, -C(O)R c1 , -C(O)NR a R b , - NR a C(O)R b , 6-12-membered aryl group or 5-12-membered heteroaryl group, wherein the alkyl, alkenyl, cycloalkyl, heterocyclyl, fused heterocyclyl, bridge Heterocyclyl, spiroheterocyclyl, R a , R b , aryl or heteroaryl optionally substitute
  • R a and R b are each independently selected from H, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclyl, or R a and R b together with the atoms to which they are attached may further form 4-7 membered heterocyclic group;
  • R 3 is a 4-8-membered heterocyclic group optionally substituted by 1-4 R c 2
  • R c 2 when the R c 2 is 2-4, the R c 2 are the same or different;
  • the heterocyclic group contains 1-2 heteroatoms
  • the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
  • R 3 is a 4-8 membered heterocyclic group optionally substituted by 1-4 R c2
  • the heteroatoms of the heterocyclic group are two, the two hetero atoms are the same or different.
  • R 1 , R 3 , R e1 , R e2 , R e3 and L 1 have the same limited range as above;
  • R is selected from C1-C3 alkyl, C1-C3 alkenyl, C3-C7 cycloalkyl, 3-8 membered heterocyclyl, 4-10 membered fused Heterocyclyl, 5-10-membered bridged heterocyclyl, 5-10-membered spiro heterocyclyl, 6-12-membered aryl or 5-12-membered heteroaryl, wherein the alkyl, alkenyl , cycloalkyl, heterocyclyl, fused heterocyclyl, bridged heterocyclyl, spiroheterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl can be optionally replaced by 1-4 R replaced by c2 ;
  • R c2 is selected from halogen, hydroxyl, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl, 3-8 membered heterocyclyl , 4-10-membered fused heterocyclic group, 5-10-membered bridged heterocyclic group, 5-10-membered spiro heterocyclic group, -C(O)R c1 , -C(O)NR a R b , - NR a C(O)R b , 6-12-membered aryl group or 5-12-membered heteroaryl group, wherein the alkyl, alkenyl, cycloalkyl, heterocyclyl, fused heterocyclyl, bridge Heterocyclyl, spiroheterocyclyl, R a , R b , aryl or heteroaryl optionally substitute
  • R c1 is selected from halogen, hydroxyl, -NR a R b , cyano, C1-C3 alkyl or C1-C3 alkoxy;
  • R a and R b are each independently selected from H, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclyl, or R a and R b together with the atoms to which they are attached may further form 4-7 membered heterocyclic group;
  • R 3 is a 4-8-membered heterocyclic group optionally substituted by 1-4 R c 2
  • R c 2 when the R c 2 is 2-4, the R c 2 are the same or different;
  • the heterocyclic group contains 1-2 heteroatoms
  • the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
  • R 3 is a 4-8 membered heterocyclic group optionally substituted by 1-4 R c2
  • the heteroatoms of the heterocyclic group are two, the two hetero atoms are the same or different.
  • R 3 , R e1 , R e2 , R e3 and L 1 have the same limited range as above;
  • R is selected from C1-C3 alkyl, C1-C3 alkenyl, C3-C7 cycloalkyl, 3-8 membered heterocyclyl, 4-10 membered fused Heterocyclyl, 5-10-membered bridged heterocyclyl, 5-10-membered spiro heterocyclyl, 6-12-membered aryl or 5-12-membered heteroaryl, wherein the alkyl, alkenyl , cycloalkyl, heterocyclyl, fused heterocyclyl, bridged heterocyclyl, spiroheterocyclyl, 6-12 membered aryl or 5-12 membered heteroaryl can be optionally replaced by 1-4 R replaced by c2 ;
  • R c2 is selected from halogen, hydroxyl, C1-C3 alkyl, C1-C3 alkenyl, -NR a R b , cyano, C1-C3 alkoxy, C3-C7 cycloalkyl, 3-8 membered heterocyclyl , 4-10-membered fused heterocyclic group, 5-10-membered bridged heterocyclic group, 5-10-membered spiro heterocyclic group, -C(O)R c1 , -C(O)NR a R b , - NR a C(O)R b , 6-12-membered aryl group or 5-12-membered heteroaryl group, wherein the alkyl, alkenyl, cycloalkyl, heterocyclyl, fused heterocyclyl, bridge Heterocyclyl, spiroheterocyclyl, R a , R b , aryl or heteroaryl optionally substitute
  • R c1 is selected from halogen, hydroxyl, NR a R b , cyano, C1-C3 alkyl or C1-C3 alkoxy;
  • R a and R b are each independently selected from H, C1-C3 alkyl, C3-C7 cycloalkyl or 4-7 membered heterocyclyl, or R a and R b together with the atoms to which they are attached may further form 4-7 membered heterocyclic group;
  • R 3 is a 4-8-membered heterocyclic group optionally substituted by 1-4 R c 2
  • R c 2 when the R c 2 is 2-4, the R c 2 are the same or different;
  • the heterocyclic group contains 1-2 heteroatoms
  • the heteroatom of the heterocyclic group is nitrogen and/or oxygen;
  • R 3 is a 4-8 membered heterocyclic group optionally substituted by 1-4 R c2
  • the heteroatoms of the heterocyclic group are two, the two hetero atoms are the same or different.
  • tetrahydronaphthyridine derivatives are selected from any one of the following structures:
  • the present invention provides a preparation method of the tetrahydronaphthyridine derivatives, pharmaceutically acceptable salts thereof, tautomers or stereoisomers thereof as described in the first aspect, which are selected from From the following schemes:
  • the preparation method of the compound described in the general formula (I) of the present invention or its stereoisomer, tautomer or its pharmaceutically acceptable salt comprises the following steps:
  • the compound of general formula (I-1) is reacted with ammonia methanol to obtain the compound of general formula (I-2);
  • the compound of the general formula (I-2) and the compound of the general formula (I-3) are subjected to condensation reaction in the presence of a basic reagent and a condensing agent to obtain the compound of the general formula (I-4);
  • the compound of the general formula (I-8) is reacted with the compound of the general formula (I-9) to obtain the compound of the general formula (I-A).
  • PG is Boc or Cbz;
  • X is halogen, preferably bromine;
  • R 1 , R 3 , L 1 , L 2 have the same limited ranges as above;
  • R 1 is preferably cyano;
  • L 1 is preferably O.
  • the compound of the general formula (I-12) is de-esterified to obtain the compound of the general formula (I-13) under acidic conditions;
  • the compound of the general formula (I-13) is halogenated at the 2-position under basic conditions to obtain the compound of the general formula (I-14);
  • the compound of the general formula (I-16) and the compound of the general formula (I-7) are subjected to Mitsunobu reaction to obtain the compound of the general formula (I-17);
  • the compound of the general formula (I-17) and the compound of the general formula (I-9) are subjected to Buchwald reaction under basic conditions, in the presence of a metal catalyst and a ligand, to obtain the compound of the general formula (I-18) ;
  • the compound of the general formula (I-18) is subjected to a reduction reaction to obtain the compound of the general formula (I-A).
  • X is halogen, preferably bromine; R 1 , R 3 , L 1 , L 2 have the same limited ranges as above; R 1 is preferably fluorine; L 1 is preferably O.
  • the compound of the general formula (I-2) is protected by a protective group under acid conditions to obtain the compound of the general formula (I-19);
  • the compound of the general formula (I-19) is reacted with methyl malonate under basic conditions to obtain the compound of the general formula (I-20);
  • the fifth step the compound of the general formula (I-22) and the compound of the general formula (I-9) obtain the compound of the general formula (I-23) under basic conditions;
  • the compound of the general formula (I-23) is de-esterified and the protecting group is obtained to obtain the compound of the general formula (I-24);
  • the compound of general formula (I-25) and the compound of general formula (I-7) are subjected to Mitsunobu reaction to obtain the compound of general formula (I-A).
  • PG and PG 1 are protecting groups; PG is preferably Boc or Cbz; PG 1 is preferably PMB; X is halogen or OTs, preferably bromine; R 1 , R 3 and L 1 , L 2 have the same definitions as above range; R 1 is preferably hydrogen; L 1 is preferably O.
  • the compound of the general formula (I-A) and the compound of the general formula (I-B) are subjected to Buchwald reaction under basic conditions, in the presence of a metal catalyst and a ligand, to obtain the compound of the general formula (I-C);
  • R 1 , R 2 , R 3 , R 4 , L 1 , L 2 have the same limited range as the general formula (I);
  • X is halogen or OTs, preferably bromine.
  • the reagent for providing alkaline conditions is selected from organic bases or inorganic bases, and the organic bases are triethylamine, N,N-diisopropylethylamine, n-butyllithium, diisopropylamine
  • the organic bases are triethylamine, N,N-diisopropylethylamine, n-butyllithium, diisopropylamine
  • the inorganic bases are sodium hydride, potassium phosphate, sodium carbonate,
  • potassium acetate, cesium carbonate, sodium hydroxide, potassium hydroxide, sodium bicarbonate and lithium hydroxide sodium hydride, potassium phosphate, sodium carbonate, potassium acetate, cesium carbonate, sodium hydroxide, potassium hydroxide, sodium bicarbonate and lithium hydroxide;
  • the reagent for providing acidic conditions is one or more of hydrogen chloride, hydrogen chloride solution in 1,4-dioxane, hydrogen chloride solution in methanol, trifluoroacetic acid, formic acid, acetic acid, hydrochloric acid, sulfuric acid, methanesulfonic acid, nitric acid and phosphoric acid kind;
  • the metal catalyst is palladium/carbon, Raney nickel, tetrakis-triphenylphosphine palladium, palladium dichloride, palladium acetate, [1,1'-bis(diphenylphosphino)ferrocene]palladium dichloride ( Pd(dppf)Cl 2 ), [1,1'-bis(diphenylphosphino)ferrocene]dichloropalladium dichloromethane complex, bistriphenylphosphonium palladium dichloride (Pd(PPh) 3 ) one or more of Cl 2 ) and tris(dibenzylideneacetone)dipalladium (Pd 2 (dba) 3 );
  • the ligands are 2-dicyclohexylphosphine-2,6'-dimethoxybiphenyl (SPhos), 4,5-bisdiphenylphosphine-9,9-dimethylxanthene (XantPhos), 2- Dicyclohexylphosphino-2,4,6-triisopropylbiphenyl (XPhos), 2-dicyclohexylphosphino-2'-(N,N-dimethylamine)-biphenyl (DavePhos), 1, One or more of 1'-bis(diphenylphosphino)ferrocene (Dppf) and 1,1'-binaphthyl-2,2'-bisdiphenylphosphine (BINAP), preferably 1,1 '-Binaphthalene-2,2'-bisdiphenylphosphine (BINAP);
  • the reducing agent is one or more of sodium borohydride, potassium borohydride, sodium cyanoborohydride, sodium triacetoxyborohydride, lithium aluminum tetrahydrogen;
  • the oxidant is one or more of potassium permanganate, manganese dioxide, potassium dichromate, sodium dichromate and potassium osmate;
  • the above reaction is preferably carried out in a solvent, and the solvent used is N,N-dimethylformamide, N-methylpyrrolidone, dimethylsulfoxide, 1,4-dioxane, water, tetrahydrofuran, dichloromethane, One or more of 1,2-dichloroethane, methanol, ethanol, toluene, petroleum ether, ethyl acetate, n-hexane and acetone.
  • the solvent used is N,N-dimethylformamide, N-methylpyrrolidone, dimethylsulfoxide, 1,4-dioxane, water, tetrahydrofuran, dichloromethane, One or more of 1,2-dichloroethane, methanol, ethanol, toluene, petroleum ether, ethyl acetate, n-hexane and acetone.
  • the present invention provides a pharmaceutical composition
  • a pharmaceutical composition comprising the above-mentioned tetrahydronaphthyridine derivatives, pharmaceutically acceptable salts thereof, tautomers or stereoisomers thereof body;
  • the pharmaceutical composition further comprises a pharmaceutically acceptable carrier and/or excipient.
  • pharmaceutically acceptable carrier is a pharmaceutically acceptable solvent, suspending agent or excipient for delivering the active substance of the present invention or a physiologically acceptable salt thereof to animals or humans.
  • the carrier can be liquid or solid.
  • the pharmaceutical composition contains a safe and effective amount (such as 0.001-99.9 parts by weight, more preferably 0.01-99 parts by weight, more preferably 0.1-90 parts by weight) of the compound represented by formula (I) or a pharmaceutically acceptable salt thereof; and a pharmaceutically acceptable carrier or excipient, wherein the total weight of the composition is 100 parts by weight.
  • a safe and effective amount such as 0.001-99.9 parts by weight, more preferably 0.01-99 parts by weight, more preferably 0.1-90 parts by weight
  • a pharmaceutically acceptable carrier or excipient wherein the total weight of the composition is 100 parts by weight.
  • the pharmaceutical composition of the present invention contains 0.001-99.9 wt %, more preferably 0.01-99 wt %, more preferably 0.1-90 wt % of the total weight of the compound represented by formula (I) or its pharmacy an acceptable salt; and a pharmaceutically acceptable carrier or excipient, wherein the total weight of the composition is 100% by weight.
  • the preferred ratio of the compound of formula (I) to a pharmaceutically acceptable carrier, excipient or sustained-release agent is that formula (I) as an active ingredient accounts for more than 65% of the total weight, and the rest accounts for The total weight ratio is 0.5-40%, or more preferably 1-20%, or more preferably 1-10%.
  • the unit dose of each dose comprises 0.05mg-500mg, preferably 0.5mg-200mg, more preferably 0.1mg-100mg of the compound of formula (I), enantiomer, external Racemates, pharmaceutically acceptable salts or mixtures thereof.
  • the amount of the active ingredients can generally be the conventional amount or lower in the prior art.
  • compositions of the present invention may be in various forms, such as tablets, capsules, powders, syrups, solutions, suspensions and aerosols, etc., wherein the compound of formula (I) may be present in a suitable solid or liquid carrier or diluent middle.
  • the pharmaceutical compositions of the present invention may also be stored in a suitable sterile device for injection or instillation.
  • the pharmaceutical composition may also contain odorants, flavoring agents, and the like.
  • the compound of formula (I) or the pharmaceutical composition comprising the compound of formula (I) of the present invention can be clinically used in mammals (including humans) through oral, nasal, skin, lung or gastrointestinal routes of administration.
  • the preferred route of administration is oral.
  • the preferred daily dose is 0.5 mg to 200 mg/kg body weight, taken in one or divided doses. Regardless of the method of administration, the optimal dose for an individual should depend on the specific treatment. It is common to start with a small dose and gradually increase the dose until the most suitable dose is found.
  • the effective dose of active ingredient employed may vary with the compound employed, the mode of administration, and the severity of the disease to be treated. Generally, however, satisfactory results are obtained when the compounds of the present invention are administered in doses of about 1-300 mg/kg of animal body weight per day, preferably in 1-3 divided doses per day, or in sustained release form Dosing. For most large mammals, the total daily dose is about 5-1000 mg, preferably about 10-500 mg.
  • Dosage forms suitable for oral administration contain about 1-200 mg of the active compound in intimate admixture with a solid or liquid pharmaceutically acceptable carrier. This dosage regimen can be adjusted to provide optimal therapeutic response. For example, several divided doses may be administered daily, or the dose may be proportionally reduced, as dictated by the exigencies of the therapeutic situation.
  • the compounds or pharmaceutically acceptable salts and compositions thereof can be administered orally as well as intravenously, intramuscularly or subcutaneously.
  • the preferred pharmaceutical compositions are solid compositions, especially tablets and solid- or liquid-filled capsules. Oral administration of the pharmaceutical composition is preferred.
  • Solid carriers include: starch, lactose, dicalcium phosphate, microcrystalline cellulose, sucrose and kaolin, while liquid carriers include: sterile water, polyethylene glycols, nonionic surfactants and edible oils (eg corn oil, peanut oil and sesame oil) as appropriate to the characteristics of the active ingredient and the particular mode of administration desired.
  • Adjuvants commonly used in the preparation of pharmaceutical compositions may also advantageously be included, such as flavors, colors, preservatives and antioxidants such as vitamin E, vitamin C, BHT and BHA.
  • the active compounds or pharmaceutically acceptable salts thereof and compositions thereof may also be administered parenterally or intraperitoneally.
  • Solutions or suspensions of these active compounds can also be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose.
  • Dispersions can also be prepared in glycerol, liquids, polyethylene glycols, and mixtures thereof in oils. Under ordinary conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
  • the pharmaceutical forms suitable for injection include sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, these forms must be sterile and must be fluid for easy syringe expelling. It must be stable under the conditions of manufacture and storage and must be resistant to the contaminating influence of microorganisms such as bacteria and fungi.
  • the carrier can be a solvent or dispersion medium containing, for example, water, alcohol (for example, glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
  • the compound represented by formula (I) or its pharmaceutically acceptable salt and its composition can also be administered in combination with other active ingredients or drugs for treating or preventing chronic pain diseases.
  • other active ingredients or drugs for treating or preventing chronic pain diseases.
  • two or more drugs are administered in combination, they generally have better effects than when the two drugs are administered separately.
  • the present invention provides a tetrahydronaphthyridine derivative as described in the first aspect, a pharmaceutically acceptable salt thereof, a tautomer or a stereoisomer thereof, or a tetrahydronaphthyridine derivative as described in the third aspect.
  • a pharmaceutically acceptable salt thereof a tautomer or a stereoisomer thereof, or a tetrahydronaphthyridine derivative as described in the third aspect.
  • the cancers described therein are, but are not limited to, astrocytic carcinoma, breast cancer, cervical cancer, colorectal cancer, endometrial cancer, esophageal cancer, gastric cancer, head and neck cancer, hepatocellular carcinoma, laryngeal cancer, pancreatic cancer, Lung cancer, oral cancer, ovarian cancer, prostate cancer, thyroid cancer, sarcoma, kidney cancer and bile duct cancer; preferably lung cancer, pancreatic cancer or colorectal cancer.
  • the present invention provides a tetrahydronaphthyridine derivative as described in the first aspect, a pharmaceutically acceptable salt thereof, a tautomer or a stereoisomer thereof or as described in the third aspect Application of the pharmaceutical composition in the preparation of KRAS mutant G12C inhibitor.
  • the present invention provides a method for treating and/or preventing cancer, comprising administering to a human a therapeutically effective amount of the tetrahydronaphthyridine derivative as described in the first aspect, a pharmaceutically acceptable A salt, a tautomer or a stereoisomer thereof or a pharmaceutical composition as described in the third aspect.
  • Alkyl refers to a saturated aliphatic hydrocarbon group comprising 1-20 carbon atoms, or 1-10 carbon atoms, or 1-6 carbon atoms, or 1-4 carbon atoms, or 1-3 carbon atoms , or a saturated linear or branched monovalent hydrocarbon group of 1-2 carbon atoms, wherein the alkyl group may be independently optionally substituted by one or more substituents described herein.
  • alkyl groups include, but are not limited to, 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, 2-methylpentyl, 3-methylpentyl, 4-methylpentyl, 2,3-di Methylbutyl etc. Alkyl groups can be optionally substituted or unsubstituted.
  • Alkenyl refers to a linear or branched monovalent hydrocarbon group of 2-12 carbon atoms, or 2-8 carbon atoms, or 2-6 carbon atoms, or 2-4 carbon atoms, wherein at least one CC is sp 2 double bond, wherein the alkenyl group can be independently optionally substituted by one or more substituents described in the present invention, specific examples of which include, but are not limited to, vinyl, allyl and alkene Butyl and so on. Alkenyl groups can be optionally substituted or unsubstituted.
  • Cycloalkyl means a saturated or partially unsaturated monocyclic or polycyclic cyclic hydrocarbon substituent, the cycloalkyl ring comprising 3 to 20 carbon atoms, preferably 3 to 12 carbon atoms, more preferably 3 to 6 carbon atoms carbon atoms.
  • Non-limiting examples of monocyclic cycloalkyl groups include, but are not limited to, cyclopropyl, cyclobutyl, cyclopentyl, cyclopentenyl, cyclohexyl, cyclohexenyl, cyclohexadienyl, cycloheptyl , cycloheptatrienyl, cyclooctyl, etc.; polycyclic cycloalkyl groups include spiro, fused and bridged cycloalkyl groups. Cycloalkyl groups can be optionally substituted or unsubstituted.
  • “Spirocycloalkyl” refers to a polycyclic group with 5 to 18 members, two or more cyclic structures, and the single rings share one carbon atom (called spiro atom) with each other, and the ring contains one or more aromatic systems with double bonds but none of the rings have fully conjugated pi electrons.
  • spiro atom carbon atom
  • it is 6 to 14 yuan, more preferably 7 to 10 yuan.
  • spirocycloalkyl groups are divided into mono-spiro, double-spiro or poly-spirocycloalkyl groups, preferably mono-spiro and double-spirocycloalkyl groups, preferably 4-membered/5-membered, 4-membered Yuan/6 Yuan, 5 Yuan/5 Yuan or 5 Yuan/6 Yuan.
  • spirocycloalkyl include, but are not limited to:
  • “Fused cycloalkyl” refers to a 5- to 18-membered all-carbon polycyclic group containing two or more cyclic structures that share a pair of carbon atoms with each other, and one or more rings may contain one or more double bonds, But none of the rings have an aromatic system with fully conjugated pi electrons, preferably 6 to 12 membered, more preferably 7 to 10 membered. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic fused cycloalkyl, preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicycloalkyl.
  • Non-limiting examples of "fused cycloalkyl” include, but are not limited to:
  • “Bridged cycloalkyl” refers to an all-carbon polycyclic group of 5 to 18 members, containing two or more cyclic structures, sharing two carbon atoms that are not directly connected to each other, and one or more rings may contain one or more Aromatic systems in which multiple double bonds, but none of the rings have fully conjugated pi electrons, are preferably 6 to 12 membered, more preferably 7 to 10 membered. According to the number of constituent rings, it can be divided into bicyclic, tricyclic, tetracyclic or polycyclic bridged cycloalkyl, preferably bicyclic, tricyclic or tetracyclic, more preferably bicyclic or tricyclic.
  • Non-limiting examples of "bridged cycloalkyl” include, but are not limited to:
  • the cycloalkyl ring can be fused to an aryl, heteroaryl or heterocyclyl ring, wherein the ring connected to the parent structure is a cycloalkyl, non-limiting examples include indanyl, tetrahydronaphthalene base, benzocycloheptyl, etc.
  • Heterocyclyl “heterocycle” or “heterocyclic” are used interchangeably in this application, and are used interchangeably in this application, and all refer to a saturated or partially unsaturated monocyclic ring containing 3-12 ring atoms , bicyclic or tricyclic non-aromatic heterocyclic groups, wherein at least one ring atom atom is a heteroatom, such as oxygen, nitrogen, sulfur atom and the like. It preferably has a 5- to 7-membered monocyclic ring or a 7- to 10-membered bi- or tricyclic ring, which may contain 1, 2 or 3 atoms selected from nitrogen, oxygen and/or sulfur.
  • heterocyclyl examples include, but are not limited to, morpholinyl, oxetanyl, thiomorpholinyl, tetrahydropyranyl, 1,1-dioxo-thiomorpholinyl, piperidine base, 2-oxo-piperidinyl, pyrrolidinyl, 2-oxo-pyrrolidinyl, piperazin-2-one, 8-oxa-3-aza-bicyclo[3.2.1]octyl and Piperazinyl.
  • the heterocyclyl ring can be fused to an aryl, heteroaryl or cycloalkyl ring, wherein the ring attached to the parent structure is a heterocyclyl.
  • Heterocyclyl groups can be optionally substituted or unsubstituted.
  • spiroheterocyclyls are classified into mono-spiroheterocyclyl, bis-spiroheterocyclyl or poly-spiroheterocyclyl, preferably mono-spiroheterocyclyl and bis-spiroheterocyclyl. More preferably, it is a 4-membered/4-membered, 4-membered/5-membered, 4-membered/6-membered, 5-membered/5-membered or 5-membered/6-membered monospiroheterocyclyl group.
  • spiroheterocyclyl include, but are not limited to:
  • bicyclic, tricyclic, tetracyclic or polycyclic fused heterocyclic groups preferably bicyclic or tricyclic, more preferably 5-membered/5-membered or 5-membered/6-membered bicyclic fused heterocyclic groups.
  • fused heterocyclyl include, but are not limited to:
  • bridged heterocyclyl include, but are not limited to:
  • Aryl refers to a carbocyclic aromatic system containing one or two rings, wherein the rings may be joined together in a fused fashion.
  • aryl includes aromatic groups such as phenyl, naphthyl, tetrahydronaphthyl. Preferred aryl groups are C6 - C10 aryl groups, more preferred aryl groups are phenyl and naphthyl, and most preferred are phenyl groups.
  • Aryl groups can be substituted or unsubstituted.
  • the "aryl” can be fused with a heteroaryl, a heterocyclyl or a cycloalkyl, wherein the parent structure is linked together by an aryl ring, non-limiting examples include but are not limited to:
  • Heteroaryl refers to an aromatic 5- to 6-membered monocyclic or 9- to 10-membered bicyclic ring, which may contain 1 to 4 atoms selected from nitrogen, oxygen, and/or sulfur.
  • heteroaryl include, but are not limited to, furanyl, pyridyl, 2-oxo-1,2-dihydropyridyl, pyridazinyl, pyrimidinyl, pyrazinyl, thienyl, isoxazolyl , oxazolyl, oxadiazolyl, imidazolyl, pyrrolyl, pyrazolyl, triazolyl, tetrazolyl, thiazolyl, isothiazolyl, 1,2,3-thiadiazolyl, benzo-diazolyl Oxolyl, benzimidazolyl, indolyl, isoindolyl, 1,3-dioxo-isoindo
  • Heteroaryl groups can be optionally substituted or unsubstituted.
  • the heteroaryl ring can be fused to an aryl, heterocyclyl or cycloalkyl ring, wherein the ring connected to the parent structure is a heteroaryl ring, non-limiting examples include but are not limited to:
  • Alkoxy refers to a group (alkyl-O-). Wherein, alkyl is as defined herein. Ci - C6 alkoxy groups are preferred. Examples include, but are not limited to: methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy, isobutoxy, tert-butoxy, and the like.
  • Haloalkyl refers to an alkyl group having one or more halogen substituents, wherein the alkyl group has the meaning as described herein.
  • haloalkyl include, but are not limited to, fluoromethyl, difluoromethyl, trifluoromethyl, perfluoroethyl, 1,1-dichloroethyl, 1,2-dichloropropyl, and the like.
  • Halogen means fluorine, chlorine, bromine and iodine, preferably fluorine, chlorine and bromine.
  • Amino refers to -NH2 .
  • Cyano refers to -CN.
  • Niro refers to -NO2 .
  • Benzyl refers to -CH2 -phenyl.
  • Carboxyl refers to -C(O)OH.
  • Alcohol refers to -C(O) CH3 or Ac.
  • Carboxylate means -C(O)O(alkyl) or (cycloalkyl), wherein alkyl and cycloalkyl are as defined above.
  • 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 compounds of the present invention may contain asymmetric centers or chiral centers and therefore exist in different stereoisomers. All stereoisomeric forms of the compounds of the present invention, including, but not limited to, diastereomers, enantiomers, atropisomers, and mixtures thereof, such as racemic mixtures, constitute the part.
  • Diastereomers can be separated into individual diastereomers by methods such as chromatography, crystallization, distillation or sublimation on the basis of their physicochemical differences.
  • Enantiomers can be separated by converting a chiral mixture into a diastereomeric mixture by reaction with an appropriate optically active compound such as a chiral auxiliary such as a chiral alcohol or Mosher's acid chloride , the diastereomers are separated, and the individual diastereomers are converted to the corresponding pure enantiomers.
  • the intermediates and compounds of the present invention may also exist in different tautomeric forms, and all such forms are included within the scope of the present invention.
  • optically active compounds that is, they have the ability to rotate the plane of plane-polarized light.
  • the prefixes D, L or R, S are used to denote the absolute configuration of the chiral center of the molecule.
  • the prefixes d, l or (+), (-) are used to designate the sign of the plane-polarized light rotation of the compound, (-) or l means that the compound is levorotatory, and the prefix (+) or d means that the compound is dextrorotatory.
  • the atoms or groups of atoms of these stereoisomers are connected to each other in the same order, but their steric structures are different.
  • a specific stereoisomer may be an enantiomer, and a mixture of isomers is often referred to as an enantiomeric mixture.
  • a 50:50 mixture of enantiomers is called a racemic mixture or racemate, which can result in no stereoselectivity or stereospecificity during chemical reactions.
  • the terms “racemic mixture” and “racemate” refer to an equimolar mixture of two enantiomers, devoid of optical activity.
  • Tautomer or "tautomeric form” means that isomers of structures of different energies can be interconverted through a low energy barrier.
  • proton tautomers ie, prototropic tautomers
  • Valence (valence) tautomers include interconversions that recombine bond electrons.
  • the structural formulas described herein include all isomeric forms (such as enantiomers, diastereomers, and geometric isomers): such as R, S configurations containing asymmetric centers, (Z), (E) isomers of double bonds, and (Z), (E) conformational isomers. Accordingly, individual stereochemical isomers of the compounds of the present invention or mixtures of enantiomers, diastereomers, or geometric isomers thereof are within the scope of the present invention.
  • “Pharmaceutically acceptable salts” refer to salts of the compounds of the present invention which are safe and effective when used in humans or animals.
  • the salts of the compounds can be obtained by using a sufficient amount of base or acid in neat solution or in a suitable inert solution to obtain the corresponding addition salts.
  • Pharmaceutically acceptable base addition salts include sodium, potassium, calcium, ammonium, organic ammonia or magnesium salts, etc.
  • Pharmaceutically acceptable acid addition salts include inorganic acid salts and organic acid salts, and the inorganic and organic acids include Hydrochloric acid, hydrobromic acid, carbonic acid, bicarbonate, phosphoric acid, monohydrogen phosphate, dihydrogen phosphate, sulfuric acid, monohydrogen sulfate, acetic acid, maleic acid, malonic acid, succinic acid, rice butenedioic acid, Phthalic acid, benzenesulfonic acid, p-toluenesulfonic acid, citric acid, and methanesulfonic acid, among others (see Berge et al., "Pharmaceutical Salts", Journal of Pharmaceutical Science 66:1-19 (1977)).
  • the invention provides a KRAS mutant G12C inhibitor with a new structure, and the test results show that the tetrahydronaphthyridine derivative exhibits excellent KRAS mutant G12C inhibitory activity, and at the same time exhibits excellent safety and selectivity, It can be used to prepare medicines for treating cancer, especially lung cancer, colorectal cancer or pancreatic cancer.
  • Fig. 1 is a graph showing the effect of the compounds of the present invention on the KRAS/ERK1/2 signal transduction pathway of KRAS G12C mutant MIAPaCa-2 pancreatic cells.
  • Figure 2 is a graph showing the in vivo antitumor effect of the compounds of the present invention on KRAS G12C mutant MIAPaCa-2 pancreatic xenograft tumor nude mice model.
  • Figure 3 is a graph showing the effect of the compounds of the present invention on the body weight of mice in a nude mouse model of KRAS G12C mutant MIAPaCa-2 pancreatic xenograft tumor.
  • the mass spectrum was measured by LC/MS, and the ionization mode was ESI.
  • HPLC model Agilent 1260, Thermo Fisher U3000; Column model: Waters xbrige C18 (4.6*150mm, 3.5 ⁇ m); Mobile phase: A: ACN, B: Water (0.1% H 3 PO 4 ); Flow rate: 1.0mL/min; Gradient: 5%A for 1min, increase to 20%A within 4min, increase to 80%A within 8min, 80%A for 2min, back to 5%A within 0.1min; Wavelength: 220nm; Column oven: 35°C.
  • the thin layer chromatography silica gel plate uses Yantai Huanghai HSGF254 or Qingdao GF254 silica gel plate, the size of the silica gel plate used for thin layer chromatography (TLC) is 0.2mm-0.3mm, and the specification used for TLC separation and purification products is 0.4mm -0.5mm.
  • HATU 2-(7-benzotriazole oxide)-N,N,N',N'-tetramethylurea hexafluorophosphate
  • Hydrogen atmosphere means that the reaction flask is connected to a hydrogen balloon with a volume of about 1L.
  • reaction temperature is room temperature, which is 20°C-30°C.
  • the monitoring of the reaction progress in the embodiment adopts thin layer chromatography (TLC), the developing solvent used in the reaction, the eluent system of the column chromatography used for purifying the compound or the developing solvent system of the thin layer chromatography method includes: A: Petroleum ether and ethyl acetate system; B: dichloromethane and methanol system; C: n-hexane: ethyl acetate; the volume ratio of the solvent varies according to the polarity of the compound, and a small amount of acidic or basic reagent can also be added Adjustments such as acetic acid or triethylamine, etc.
  • TLC thin layer chromatography
  • Methyl 3-amino-4-pyridinecarboxylate IN-2a 1.0 g, 6.57 mmol
  • monoethyl malonate 1.0 g, 7.88 mmol
  • pyridine 1.6 g, 19.71 mmol
  • ethyl acetate 20 mL
  • 1-propyl phosphoric anhydride 6.4 g, 10.06 mmol, 50% N,N-dimethylformamide solution
  • reaction solution was quenched with water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to obtain the title compound IN-2b (1.3 g, yield 74%).
  • the sixth step 4-bromo-3-fluoro-1,7 naphthyridin-2-ol IN-2
  • reaction solution was quenched by adding saturated aqueous ammonium chloride solution (1.0 L), extracted with ethyl acetate (1.5 L), the organic phases were combined, washed with saturated brine (1.0 L), dried over anhydrous sodium sulfate, concentrated, and the crude product was filtered through a silica gel column Analysis and purification gave the title compound IN-4b (303.0 g, 65% yield) as a yellow solid.
  • reaction solution was concentrated to (4.0 L), quenched by adding saturated aqueous ammonium chloride solution (3 L), extracted with ethyl acetate (2.0 L), the organic phases were combined, washed with saturated brine (3 L), and concentrated to obtain the title compound IN- 4c (140.0 g, 54% yield).
  • reaction solution was filtered, water (2.5L) and dichloromethane (2.5L) were added to the filtrate, the layers were separated, the aqueous phase was extracted with dichloromethane (2.0L), the organic phases were combined, washed with saturated brine (2.5L), and anhydrous sulfuric acid It was dried over sodium, concentrated, and the crude product was purified by silica gel column chromatography to give the title compound IN-4d (68.6 g, 49% yield) as a brown oil.
  • the sixth step ((2R,7aS)-2-fluorotetrahydro-1H-pyrrolizin-7a(5H)-yl)methanol and ((2S,7aR)-2-fluorotetrahydro-1H-pyrrolizine- 7a(5H)-yl)methanol mixture IN-4
  • the ninth step 2-((S)-1-acryloyl-4-(7-(8-chloronaphthalen-1-yl)-2-(((S)-1-methylpyrrolidin-2-yl) Methoxy)-5,6,7,8-tetrahydro-1,7-naphthyridin-4-yl)piperazin-2-yl)acetonitrile 3
  • reaction solution was quenched by adding a small amount of water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to obtain the title compound 4 (4.0 mg, yield 10%) .
  • the first step 1- (tert-butyl)2-(3-chloropropyl)pyrrolidine-1,2-dicarboxylic acid 2-methyl ester 5b
  • Boc-proline methyl ester 5a (5.0 g, 21.81 mmol) was dissolved in dry tetrahydrofuran (20 mL), and under nitrogen protection, the temperature was lowered to -65 °C, and bistrimethylsilylamine was slowly added to a constant pressure dropping funnel Lithium (32 mL, 32 mmol, 1M tetrahydrofuran solution), after adding -65 ° C for 1 hour, slowly adding 1-bromo-3-chloropropane (17 g, 109.2 mmol), slowly warming to room temperature after the addition and reacting for 2 hours, TLC Shows that the reaction is complete.
  • reaction solution was poured into water, extracted with ethyl acetate, the organic phases were combined, washed with saturated brine, dried over anhydrous sodium sulfate, concentrated, and the crude product was purified by silica gel column chromatography to obtain the title compound 5b (5.1 g, yield 80%) as a yellow liquid.
  • Lithium aluminum hydride (595 mg, 15.6 mmol) was dispersed in dry tetrahydrofuran (30 mL), cooled to 0 °C, and a solution of 6b-1 (1.7 g, 7.8 mmol) in tetrahydrofuran (15 mL) was added dropwise under nitrogen protection. After completion, the temperature was raised to 50° C. to react for 2 hours, and the reaction of the raw materials was monitored by TLC.
  • reaction solution was cooled to room temperature, quenched by adding water (0.6 mL), 15% sodium hydroxide solution (0.6 mL) and water (1.8 mL) successively, filtered, and the filtrate was concentrated to give the title compound 6c-1 (1.0 g, crude product), used directly in the next step.
  • the third step 4-bromo-3-fluoro-2-(((3R,4R)-4-methoxy-1-methylpyrrolidin-3-yl)oxy)-1,7-naphthyridine 6d
  • reaction solution was cooled to room temperature, diluted with water (5 mL), extracted with ethyl acetate (10 mL), the organic phases were combined, dried over anhydrous sodium sulfate, filtered, and the filtrate was concentrated and purified by silica gel column chromatography to obtain the title compound 8c as a pale yellow solid ( 30 mg, crude product), used directly in the next step.
  • the reaction solution was adjusted to pH 8 with saturated aqueous sodium bicarbonate solution, extracted with dichloromethane (5 mL), the organic phases were combined, concentrated to remove about half of the dichloromethane, extracted with 1N dilute hydrochloric acid (5 mL), the aqueous phases were combined, and the aqueous phase was washed with sodium carbonate
  • the first step 4-bromo-3-fluoro-2-((tetrahydro-1H-pyrrolazin-7a(5H)-yl)methoxy)-1,7-naphthyridine 9a
  • Human pancreatic cancer MIAPaCa-2 cells (CRL-1420) used in the present invention were purchased from American Type Culture Collection (ATCC). Cells were grown in DMEM medium containing 2.5% horse serum, 10% fetal bovine serum, and 1% dual antibodies at 37°C in a 5% CO 2 environment.
  • the inhibitory effect of compounds on the proliferation of MIAPaCa-2 cells cultured in vitro was determined by the following methods:
  • the compounds of the examples of the present invention can inhibit the proliferation of MIAPaCa-2 cells, and the activity of compound 5 is comparable to that of MRTX849.
  • MIAPaCa-2 cells in good logarithmic growth phase were seeded into a six-well plate at 1*10 6 cells/well, and cultured overnight at 37° C. and 5% CO 2 .
  • Protein sample preparation trypsin digestion, collect cells, centrifuge at 500g for 5 minutes, discard the supernatant, wash 3 times with PBS, and use 1 ⁇ SDS gel loading buffer (50mM Tris-HCl (pH 6.8), 100mM DTT, 2% SDS, 10% glycerol, 0.1% bromophenol blue) 100 ⁇ L of lysed cells. Cell lysates were denatured by heating at 100°C for 10 minutes.
  • 1 ⁇ SDS gel loading buffer 50mM Tris-HCl (pH 6.8), 100mM DTT, 2% SDS, 10% glycerol, 0.1% bromophenol blue
  • Compound 5 in the examples of the present invention has a significant inhibitory effect on the phosphorylation of ERK1/2 in MIAPaCa-2 cells, and the inhibitory activity is concentration gradient dependent, and the activity is roughly equivalent to MRTX849.
  • Compound 5 and MRTX849 were administered intravenously at a dose of 5 mg/kg.
  • the mice tested were 7-8 week old ICR male mice.
  • the sampling time points were 0.083h, 0.5h, 1h, 2h, and the sampling tissues were plasma, brain, lung, colorectum, pancreas, gallbladder and bile duct.
  • the specific operation process is as follows:
  • Blood was collected from the mouse heart, thoroughly mixed, placed on ice, and centrifuged within 30 min to separate the plasma (4°C, 8000 rpm for 5 min), and the plasma was stored at -80°C until measurement.
  • the brain, lung, colorectum, pancreas, gallbladder and bile duct were collected from mice on ice, perfused with normal saline, rinsed, blotted dry with filter paper, and finally stored at -80°C until measurement.
  • Blood, brain, lung, colorectum, pancreas, gallbladder and bile duct were collected from blank animals without administration of carbon dioxide after euthanasia, as blank matrix.
  • Euthanasia The experimental animals were euthanized by inhaling excess CO2 after the experiment.
  • the blood-brain drug concentration was measured at different time points, and the pharmacokinetic software WinNonlin was used to fit and calculate the metabolic parameters of the sample, such as the area under the drug-time curve (AUC), peak concentration (Cmax), time to peak (Tmax), half-life ( T1/2) and other parameters.
  • AUC area under the drug-time curve
  • Cmax peak concentration
  • Tmax time to peak
  • T1/2 half-life
  • the present invention is to illustrate a class of tetrahydronaphthyridine derivatives as KRAS mutant G12C inhibitors of the present invention, the preparation method and the application thereof through the above-mentioned examples, but the present invention is not limited to the above-mentioned examples. , that is, it does not mean that the present invention must rely on the above-mentioned embodiments to be implemented.
  • Those skilled in the art should understand that any improvement of the present invention, the equivalent replacement of each raw material of the product of the present invention, the addition of auxiliary components, the selection of specific methods, etc., all fall within the protection scope and disclosure scope of the present invention.

Abstract

属于KRAS突变体G12C抑制剂技术领域,具体涉及一类作为KRAS突变体G12C抑制剂的四氢萘啶类衍生物、其制备方法及应用。具体而言,所述四氢萘啶类衍生物的结构如式(I)所示。一种全新的四氢萘啶类衍生物,其具有显著的抑制KRAS G12C活性和治疗癌症的作用,其中所述的癌症优选肺癌、结直肠癌或胰腺癌。

Description

一类作为KRAS突变体G12C抑制剂的四氢萘啶类衍生物、其制备方法及其应用 技术领域
本发明属于KRAS突变体G12C抑制剂技术领域,具体涉及一种作为KRAS突变体G12C抑制剂的四氢萘啶类衍生物、其制备方法及应用。
背景技术
RAS基因是首个被发现的人类肿瘤基因,其编码的RAS蛋白为一类分布于质膜胞质侧的球状单体GTP结合蛋白(MW=21kDa),结合GTP时为活化状态,而结合GDP时为失活状态。RAS蛋白在鸟嘌呤核苷酸交换因子(GEF)催化下结合GTP而激活,而GTP酶激活蛋白(GAPs)催化与RAS结合的GTP水解成为GDP终止活性状态以抑制RAS活性。RAS蛋白通过其在活性(GTP结合型)和非活性(GDP结合型)状态间循环,将从多种酪氨酸激酶接收的上游信号转导至下游效应物以调节细胞增殖、存活、迁移和凋亡等过程。由于RAS蛋白在许多重要细胞信号网络的轴上处于中心位置,且这些信号与多种肿瘤标志物相关联,因此过度活化的RAS信号转导可能最终导致肿瘤发生。
在RAS家族成员中,致癌突变最常见于KRAS(85%),而NRAS(12%)和HRAS(3%)则较为少见,且KRAS突变常见于胰腺癌、结直肠癌、肺癌、胆囊癌、及甲状腺癌等,KRAS的异常表达占所有癌症的比例高达20%,而80%的KRAS突变为第12位密码子单个氨基酸替换的错义突变。其中KRAS G12C突变占所有KRAS突变的12%,但其在肺癌种所占比例较高,尤其是非小细胞肺癌(14%),基因组学研究表明,肺癌KRAS突变(包括G12C)与其它已知的致癌基因突变相斥,如EGFR、ALK及BRAF,表明KRAS突变在肺癌中的独特性且其可作为肿瘤预后的一个重要指标。
由于KRAS其无明显结合位点的蛋白结构,经过科学工作者三十多年的研究努力,临床上仍没有KRAS抑制剂呈现出足够的安全性和有效性。近年来,针对KRAS突变体的共价抑制剂研究的突破让通过异构位点(allosteric)靶向KRAS突变体成为可能。在KRAS G12C突变体中,与错义突变替换的半胱氨酸共价结合的小分子更倾向与结合GDP的KRAS蛋白相结合以降低GTP与KRAS的亲和力,同时能够阻碍GEF催化GTP替换GDP,将KRAS G12C突变体锁死在失活状态。利用这种策略目前有AMG510和MRTX-849进入了临床研究 1-2。同时各药物研发公司继续发表了多篇专利,比如WO2019150305、WO2020106640、WO2020146613、WO2019215203、WO2020081282和WO2020085493等。因此继续开发新颖的KRAS G12C抑制剂,且能够表现出治疗KRAS G12C介导肿瘤的有效性、稳定性及安全性,在KRAS突变引起的肿瘤的治疗领域是迫切需要和极具意义的。
参考文献:
1、Lanman et al.Discovery of a Covalent Inhibitor of KRAS G12C(AMG510)for the Treatment of Solid Tumors.J.Med.Chem.63,52-65(2020).
2、Fell et al.Identification of the Clinical Development Candidate MRTX849,a Covalent KRAS  G12C Inhibitor for the Treatment of Cancer.J.Med.Chem.63,6679-6693(2020).
发明内容
为了解决现有技术的上述问题,本发明的目的在于提供一种作为KRAS突变体G12C抑制剂的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,以筛选出在有效性、安全性和选择性等性能方面均具有优异性能的用作KRAS突变体G12C抑制剂的化合物。
本发明的另一个目的是提供所述衍生物、其药学上可接受的盐、其互变异构体或其立体异构体的制备方法。
为达到此发明目的,本发明采用以下技术方案:
第一方面,本发明提供一种四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,所述四氢萘啶类衍生物的结构如式(I)所示:
Figure PCTCN2022073358-appb-000001
其中:
R 1选自H或F;
R 2选自6-12元的芳基或5-12元的杂芳基,其中所述的芳基或杂芳基任选地被1-4个R c所取代;
R c选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基或3-8元的杂环基,其中所述的烷基、烯基、环烷基、R a、R b或杂环基任选地被1-3个R c1所取代;
R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基。
L 1选自键、O、S、NH或NCH 3
R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基或杂芳基可以任选地被1-4个R c2所取代。
R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
L 2选自C3-C8的环烷基、C5-C10螺环烷基、C4-C10稠环烷基、C5-C10桥环烷基、3-8元的杂环基、5-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基、5-12元的杂芳基,其中,所述环烷基、螺环烷基、稠环烷基、桥环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基、杂芳基任选地被1-4个R c所取代;
R 4选自-C(O)R d或-S(O) 2R d
R d选自C2-C4烯基或C2-C4炔基,其中所述的烯基、炔基可以任选地进一步被1-3个R d1所取代;
R d1选自卤素、氰基、C1-C3烷基、C3-C7环烷基或4-7元杂环基,其中所述的烷基、环烷基或杂环基任选地被1-3个R c取代;
所述式(I)中的杂环基、杂芳基中的杂原子为1-4个并选自氧、氮、硫中的一种或多种。
优选地,所述四氢萘啶类衍生物的结构如式(II)所示:
Figure PCTCN2022073358-appb-000002
其中,R 1、R 2、R 3、R c和L 1具有与上述相同的限定范围;n=0-4(例如n=0、n=1、n=2、n=3、n=4);
R e1、R e2或R e3分别独立选自H、卤素、氰基、C1-C3的烷基、C3-C7的环烷基或4-7元的杂环基,而且上述的烷基、环烷基或杂环基任选地被1-3个卤素、C1-C3的烷基、C3-C7的环烷基、4-7元的杂环基、羟基或氨基所取代;
所述杂环基中的杂原子为1-3个选自氧、氮和硫中的一种或多种;
和/或,R 2选自6-12元的芳基或5-12元的杂芳基,其中所述的芳基或杂芳基任选地被1-4个R c所取代;其中,R c选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基或3-8元的杂环基,其中所述的烷基、烯基、环烷基、R a、R b或杂环基任选地被1-3个R c1所取代;
R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
和/或,R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基或杂芳基可以任选地被1-4个R c2所取代;
R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮和/或氧;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
优选地,所述四氢萘啶类衍生物的结构如式(III)所示:
Figure PCTCN2022073358-appb-000003
其中,R 1、R 3、R e1、R e2、R e3和L 1具有与上述相同的限定范围;
优选地,对于式(III)化合物,其中R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、6-12元的芳基或5-12元的杂芳基可以任选地被1-4个R c2所取代;
R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮和/或氧;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
优选地,所述四氢萘啶类衍生物的结构如式(IV)所示:
Figure PCTCN2022073358-appb-000004
其中,R 3、R e1、R e2、R e3和L 1具有与上述相同的限定范围;
优选地,对于式(IV)化合物,其中R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基, 其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、6-12元的芳基或5-12元的杂芳基可以任选地被1-4个R c2所取代;
R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
R c1选自卤素、羟基、NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮和/或氧;
和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
进一步优选地,所述四氢萘啶类衍生物选自如下结构的任意一种:
Figure PCTCN2022073358-appb-000005
Figure PCTCN2022073358-appb-000006
Figure PCTCN2022073358-appb-000007
Figure PCTCN2022073358-appb-000008
Figure PCTCN2022073358-appb-000009
第二方面,本发明提供一种如第一方面所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体的制备方法,其选自如下方案:
本发明通式(I)所述的化合物或其立体异构体、互变异构体或其药用盐的制备方法,包括以下步骤:
关键中间体(I-A)制备方法,方法一:
Figure PCTCN2022073358-appb-000010
第一步,通式(I-1)的化合物和氨甲醇反应得到通式(I-2)的化合物;
第二步,通式(I-2)的化合物和通式(I-3)的化合物在碱性试剂和缩合剂存在下,经缩合反应得到通式(I-4)的化合物;
第三步,通式(I-4)的化合物在碱性条件下,关环得到通式(I-5)的化合物;
第四步,通式(I-5)的化合物在碱性条件下,3位羟基经卤代得到通式(I-6)的化合物;
第五步,通式(I-6)的化合物和通式(I-7)的化合物,经Mitsunobu反应得到通式(I-8)的化合物;
第六步,通式(I-8)的化合物和通式(I-9)的化合物反应得到通式(I-A)的化合物。
其中,PG为Boc或Cbz;X为卤素,优选为溴;R 1、R 3和L 1、L 2具有与上述相同的限定范围;R 1优选为氰基;L 1优选为O。
关键中间体(I-A)制备方法,方法二:
Figure PCTCN2022073358-appb-000011
第一步,通式(I-10)的化合物和丙二酸单乙酯在碱性条件下反应得到通式(I-11)的化合物;
第二步,通式(I-11)的化合物碱性条件下,关环得到通式(I-12)的化合物;
第三步,通式(I-12)的化合物在酸性条件下,脱酯基得到通式(I-13)的化合物;
第四步,通式(I-13)的化合物在碱性条件下,2位卤代得到通式(I-14)的化合物;
第五步,通式(I-14)的化合物羟基经卤代得到通式(I-15)的化合物;
第六步,通式(I-15)的化合物酸性条件下,得到通式(I-16)的化合物;
第七步,通式(I-16)的化合物和通式(I-7)的化合物,经Mitsunobu反应得到通式(I-17)的化合物;
第八步,通式(I-17)的化合物和通式(I-9)的化合物在碱性条件下,金属催化剂和配体存在下,经Buchwald反应得到通式(I-18)的化合物;
第九步,通式(I-18)的化合物经还原反应得到通式(I-A)的化合物。
其中,X为卤素,优选为溴;R 1、R 3和L 1、L 2具有与上述相同的限定范围;R 1优选为氟;L 1优选为O。
关键中间体(I-A)制备方法,方法三:
Figure PCTCN2022073358-appb-000012
第一步,通式(I-2)的化合物在酸条件下上保护基得到通式(I-19)的化合物;
第二步,通式(I-19)的化合物在碱性条件下,和丙二酸酰氯甲酯反应得到通式(I-20)的化合物;
第三步,通式(I-20)的化合物在酸性条件下,关环得到通式(I-21)的化合物;
第四步,通式(I-21)的化合物在碱性条件下,和4-甲基苯磺酰氯反应得到通式(I-22)的化合物;
第五步,通式(I-22)的化合物和通式(I-9)的化合物在碱性条件下得到通式(I-23)的化合物;
第六步,通式(I-23)的化合物脱酯基和保护基得到通式(I-24)的化合物;
第七步,通式(I-24)的化合物脱保护基得到通式(I-25)的化合物;
第八步,通式(I-25)的化合物和通式(I-7)的化合物,经Mitsunobu反应得到通式(I-A)的化合物。
其中,PG和PG 1为保护基;PG优选为Boc或Cbz;PG 1优选为PMB;X为卤素或OTs,优选为溴;R 1、R 3和L 1、L 2具有与上述相同的限定范围;R 1优选为氢;L 1优选为O。
通式(I)制备方法:
Figure PCTCN2022073358-appb-000013
第一步,通式(I-A)的化合物和通式(I-B)的化合物在碱性条件下,金属催化剂和配体存在下,经Buchwald反应得到通式(I-C)的化合物;
第二步,通式(I-C)的化合物和通式(I-D)的化合物在碱性条件下反应得到通式(I)的化合物;
其中,R 1、R 2、R 3、R 4和L 1、L 2具有与通式(I)相同的限定范围;X为卤素或OTs,优选为溴。
对于上述制备方法中,提供碱性条件的试剂选自有机碱或无机碱,所述的有机碱类为三乙胺、N,N-二异丙基乙胺、正丁基锂、二异丙基氨基锂、双三甲基硅基胺基锂、叔丁醇钠、甲醇钠和叔丁醇钾中的一种或多种,所述的无机碱类为氢化钠、磷酸钾、碳酸钠、碳酸钾、醋酸钾、碳酸铯、氢氧化钠、氢氧化钾、碳酸氢钠和氢氧化锂中的一种或多种;
提供酸性条件的试剂为氯化氢、氯化氢的1,4-二氧六环溶液、氯化氢的甲醇溶液、三氟乙酸、甲酸、乙酸、盐酸、硫酸、甲磺酸、硝酸和磷酸中的一种或多种;
金属催化剂为钯/碳、雷尼镍、四-三苯基膦钯、二氯化钯、醋酸钯、[1,1'-双(二苯基膦基)二茂铁]二氯化钯(Pd(dppf)Cl 2)、[1,1'-双(二苯基膦基)二茂铁]二氯化钯二氯甲烷络合物、双三苯基磷二氯化钯(Pd(PPh 3)Cl 2)和三(二亚苄基丙酮)二钯(Pd 2(dba) 3)中的一种或多种;
配体为2-双环己基膦-2,6'-二甲氧基联苯(SPhos)、4,5-双二苯基膦-9,9-二甲基氧杂蒽(XantPhos)、2-二环己基磷-2,4,6-三异丙基联苯(XPhos)、2-二环己膦基-2'-(N,N-二甲胺)-联苯(DavePhos)、1,1'-双(二苯基膦)二茂铁(Dppf)和1,1'-联萘-2,2'-双二苯膦(BINAP)中的一种或多种,优选为1,1'-联萘-2,2'-双二苯膦(BINAP);
还原剂为硼氢化钠、硼氢化钾、氰基硼氢化钠、三乙酰氧基硼氢化钠、四氢铝锂中的一种或多种;
氧化剂为高锰酸钾、二氧化锰、重铬酸钾、重铬酸钠和锇酸钾中的一种或多种;
上述反应优选在溶剂中进行,所用溶剂为N,N-二甲基甲酰胺、N-甲基吡咯烷酮、二甲基亚砜、1,4-二氧六环、水、四氢呋喃、二氯甲烷、1,2-二氯乙烷、甲醇、乙醇、甲苯、石油醚、乙酸乙酯、正己烷和丙酮中的一种或多种。
第三方面,本发明提供一种药物组合物,所述药物组合物包括如上所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体;
优选地,所述药物组合物还包括可药用载体和/或赋形剂。
本发明中,“药学上可接受的载体”是用于将本发明的活性物质或其生理上可接受的盐传送给动物或人的药学上可接受的溶剂、悬浮剂或赋形剂。载体可以是液体或固体。
在本发明中,所述的药物组合物含有安全有效量(如0.001-99.9重量份,更佳地,0.01-99重量份,更优选0.1-90重量份)的式(I)所示的化合物或其药学上可接受的盐;以及药学上可接受的载体或赋形剂,其中组合物的总重量为100重量份。
或者,本发明所述的药物组合物含有0.001-99.9wt%,更佳地,0.01-99重量%,更优选占总重量0.1-90重量%的式(I)所示的化合物或其药学上可接受的盐;以及药学上可接受的载体或赋形剂,其中组合物的总重量为100重量%。
在另一优选例中,式(I)化合物与药学上可接受的载体、赋形剂或缓释剂的优选比例是,式(I)作为活性成分占总重量比65%以上,其余部分占总重量比0.5-40%,或更好为1-20%,或最好为1-10%。
本发明药物组合物的各种制剂形式,其单位剂量每剂包含0.05mg-500mg,优选0.5mg-200mg,更优选0.1mg-100mg所述的式(I)化合物、对映异构体、外消旋体、药学上可接受的盐或它们的混合物。
当所述的药物组合物中含有额外的治疗或预防癌症的药物活性成分时,该活性成分的用量通常可以是现有技术中的常规用量或更低。
本发明的药物组合物可以是多种形式,如片剂、胶囊、粉末、糖浆、溶液状、悬浮液和气雾剂等,其中式(I)化合物可以存在于适宜的固体或液体载体或稀释液中。本发明的药物组合物也可以储存在适宜的注射或滴注的消毒器具中。该药物组合物中还可包含气味剂、香味剂等。
本发明的式(I)化合物或包含式(I)化合物的药物组合物可通过口、鼻、皮肤、肺或胃肠道等给药途径对哺乳动物(包括人)临床使用。优选的给药途径为口服。优选的每日剂量为0.5mg-200mg/kg体重,一次或分次服用。不管用何种服用方法,个人的最佳剂量应根据具体治疗而定。通常情况下是从小剂量开始,逐渐增加剂量一直到找到最合适的剂量。
所用的活性成分的有效剂量可随所用的化合物、给药的模式和待治疗的疾病的严重程度而变化。然而,通常当本发明的化合物每天以约1-300mg/kg动物体重的剂量给予时,能得到令人满意的效果,较佳地每天以1-3次分开的剂量给予,或以缓释形式给药。对大部分大型哺乳动物而言,每天的总剂量约为5-1000mg,较佳地约为10-500mg。适用于内服的剂量形式,包含与固态或液态药学上可接受的载体密切混合的约1-200mg的活性化合物。可调节此剂量方案以提供最佳治疗应答。例如,由治疗状况的迫切要求,可每天给予若干次分开的剂量,或将剂量按比例地减少。
所述化合物或其药学上可接受的盐及其组合物可通过口服以及静脉内、肌内或皮下等途径给药。从易于制备和给药的立场看,优选的药物组合物是固态组合物,尤其是片剂和固体填充或液体填充的胶囊。药物组合物的口服给药是优选的。
固态载体包括:淀粉、乳糖、磷酸二钙、微晶纤维素、蔗糖和白陶土,而液态载体包括:无菌水、聚乙二醇、非离子型表面活性剂和食用油(如玉米油、花生油和芝麻油),只要适合活性成分的特性和所需的特定给药方式。在制备药物组合物中通常使用的佐剂也可有利地被包括,例如调味剂、色素、防腐剂和抗氧化剂如维生素E、维生素C、BHT和BHA。
所述活性化合物或其药学上可接受的盐及其组合物也可肠胃外或腹腔内给药。也可在适当混合有表面活性剂(如羟丙基纤维素)的水中制备这些活性化合物(作为游离碱或药学上可接受的盐)的溶液或悬浮液。还可在甘油、液体、聚乙二醇及其在油中的混合物中制备分散液。在常规储存和使用条件下,这些制剂中含有防腐剂以防止微生物的生长。
适应于注射的药物形式包括:无菌水溶液或分散液和无菌粉(用于临时制备无菌注射溶液或分散液)。在所有情况中,这些形式必须是无菌的且必须是流体以易于注射器排出流体。在制造和储存条件下必须是稳定的,且必须能防止微生物(如细菌和真菌)的污染影响。载体可以是溶剂或分散介质,其中含有如水、醇(如甘油、丙二醇和液态聚乙二醇)、它们的适当混合物和植物油。
式(I)所示的化合物或其药学上可接受的盐及其组合物还可与其它治疗或预防慢性疼痛类疾病的活性成分或药物联合给药。当两种或两种以上的药物联合给药时,一般具有优于两种药物分别单独给药的效果。
第四方面,本发明提供一种如第一方面所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体或如第三方面所述的药物组合物在制备用于治疗癌症的药物中的用途;
其中所述的癌症,为但不限于星形细胞癌、乳腺癌、***、结直肠癌、子宫内膜癌、食道癌、胃癌、头颈部癌、肝细胞癌、喉癌、胰腺癌、肺癌、口腔癌、卵巢癌、***癌、甲状腺癌、肉瘤、肾癌和胆管癌;优选肺癌、胰腺癌或结直肠癌。
第五方面,本发明提供一种如第一方面所述四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体或如第三方面所述的药物组合物在制备作为KRAS突变体G12C抑制剂中的应用。
第六方面,本发明提供一种用于治疗和/或预防癌症的方法,其包括向人类给予治疗有效量的如第一方面所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体或如第三方面所述的药物组合物。
术语解释
除非有相反陈述,否则本发明在说明书和权利要求书中所使用的部分术语定义如下:
“烷基”指饱和脂肪族烃基团,包括1-20个碳原子,或1-10个碳原子,或1-6个碳原子,或1-4个碳原子,或1-3个碳原子,或1-2个碳原子饱和直链或支链的单价烃基,其中烷基可以独立任选地被一个或多个本发明所描述地取代基所取代。烷基基团更近一步地实例包括,包括但不限于甲基、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、2-甲基戊基、3-甲基戊基、4-甲基戊基、2,3-二甲基丁基等。烷基可以是任选取代或未取代的。
“烯基”指2-12个碳原子,或2-8个碳原子,或2-6个碳原子,或2-4个碳原子直链或支链的一价烃基,其中至少一个C-C为sp 2双键,其中烯基的基团可以独立任选地被1个或多个本发明所描述的取代基所取代,其中具体的实例包括,但并不限于乙烯基、烯丙基和烯丁基等等。烯基可以是任选取代或未取代的。
“环烷基”是指饱和或部分不饱和单环或多环环状烃取代基,环烷基环包括3至20个碳原子,优选包括3至12个碳原子,更优选包含3至6个碳原子。单环环烷基的非限制性实施例包括,但不限于环丙基、环丁基、环戊基、环戊烯基、环己基、环己烯基、环己二烯基、环庚基、环庚三烯基、环辛基等;多环环烷基包括螺环、稠环和桥环的环烷基。环烷基可以是任选取代的或未取代的。
“螺环烷基”指5至18元,两个或两个以上环状结构,且单环之间彼此共用一个碳原子(称螺原子)的多环基团,环内含有1个或多个双键,但没有一个环具有完全共轭的π电子的芳香***。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺环烷基分为单螺、双螺或多螺环烷基,优选 为单螺和双螺环烷基,优选为4元/5元、4元/6元、5元/5元或5元/6元。“螺环烷基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000014
“稠环烷基”指5至18元,含有两个或两个以上环状结构彼此公用一对碳原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香***,优选为6至12元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠环烷基,优选为双环或三环,更优选为5元/5元或5元/6元双环烷基。“稠环烷基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000015
“桥环烷基”指5至18元,含有两个或两个以上环状结构,彼此共用两个不直接相连接碳原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香***,优选为6至12元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥环烷基,优选为双环、三环或四环,更有选为双环或三环。“桥环烷基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000016
所述环烷基环可以稠合于芳基、杂芳基或杂环基环上,其中与母体结构连接在一起的环为环烷基,非限制性实施例包括茚满基、四氢萘基、苯并环庚烷基等。
“杂环基”、“杂环”或“杂环的”在本申请中可交换使用,本申请中可交换使用,都是指包含3-12个环原子的饱和或部分不饱和的单环、双环或三环的非芳香性杂环基,其中至少一个环原子原子是杂原子,如氧、氮、硫原子等。优选具有5至7元单环或7至10元双-或三环,其可以包含1,2或3个选自氮、氧和/或硫中的原子。“杂环基”的实例包括但不限于吗啉基,氧杂环丁烷基,硫代吗啉基,四氢吡喃基,1,1-二氧代-硫代吗啉基,哌啶基,2-氧代-哌啶基,吡咯烷基,2-氧代-吡咯烷基,哌嗪-2-酮,8-氧杂-3-氮杂-双环[3.2.1]辛基和哌嗪基。所述杂环基环可以稠合于芳基、杂芳基或环烷基环上,其中与母体结构连接在一起的环为杂环基。杂环基可以是任选取代的或未取代的。
“螺杂环基”指5至18元,两个或两个以上环状结构,且单环之间彼此共用一个原子的多环基团,环内含有1个或多个双键,但没有一个环具有完全共轭的π电子的芳香***,其中一个或多个环原子选自氮、氧、硫或S(O) m的杂原子,其余环原子为碳,m=1或2。优选为6至14元,更优选为7至10元。根据环与环之间共用螺原子的数目将螺杂环基分为单螺杂环基、双螺杂环基或多螺杂环基,优选为单螺杂环基和双螺杂环基。更优选为4元/4元、4元/5元、4元/6元、5元/5元或5元/6元单螺杂环基。“螺杂环基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000017
“稠杂环基”指含有两个或两个以上环状结构彼此公用一对原子的全碳多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香***,其中一个或多个环原子选自氮、氧、硫或S(O) m的杂原子,其余环原子为碳,m=1或2。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环稠杂环基,优选为双环或三环,更优选为5元/5元或5元/6元双环稠杂环基。“稠杂环基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000018
“桥杂环基”指5至18元,含有两个或两个以上环状结构,彼此共用两个不直接相连接的原子的多环基团,一个或多个环可以含有一个或多个双键,但没有一个环具有完全共轭的π电子的芳香***,其中一 个或多个环原子选自氮、氧、硫或S(O) m的杂原子,其余环原子为碳,m=1或2。优选为6至14元,更优选为7至10元。根据组成环的数目可以分为双环、三环、四环或多环桥杂环基,优选为双环、三环或四环,更有选为双环或三环。“桥杂环基”的非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000019
“芳基”是指含有一个或者两个环的碳环芳香***,其中所述环可以以稠合的方式连接在一起。术语“芳基”包括比如苯基、萘基、四氢萘基的芳香基团。优选芳基为C 6-C 10芳基,更优选芳基为苯基和萘基,最优选为苯基。芳基可以是取代或未取代的。所述“芳基”可与杂芳基、杂环基或环烷基稠合,其中与母体结构连接在一起的为芳基环,非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000020
“杂芳基”是指芳香族5至6元单环或9至10元双环,其可以包含1至4个选自氮、氧和/或硫中的原子。“杂芳基”的实施例包括但不限于呋喃基,吡啶基,2-氧代-1,2-二氢吡啶基、哒嗪基、嘧啶基、吡嗪基、噻吩基、异噁唑基、噁唑基、噁二唑基、咪唑基、吡咯基、吡唑基、***基、四唑基、噻唑基、异噻唑基、1,2,3-噻二唑基、苯并间二氧杂环戊烯基、苯并咪唑基、吲哚基、异吲哚基、1,3-二氧代-异吲哚基、喹啉基、吲唑基、苯并异噻唑基、苯并噁唑基和苯并异噁唑基。杂芳基可以是任选取代或未取代的。所述杂芳基环可以稠合于芳基、杂环基或环烷基环上,其中与母体结构连接在一起的环为杂芳基环,非限制性实施例包括但不限于:
Figure PCTCN2022073358-appb-000021
“烷氧基”是指(烷基-O-)的基团。其中,烷基见本文有关定义。C 1-C 6的烷氧基为优先选择。其实例包括,但不限于:甲氧基、乙氧基、正丙氧基、异丙氧基、正丁氧基、异丁氧基、叔丁氧基等。
“卤代烷基”指具有一个或者多个卤素取代基的烷基,其中烷基基团具有如本发明所述的含义。卤代烷基的实例包括,但并不限于氟甲基、二氟甲基、三氟甲基、全氟乙基、1,1-二氯乙基、1,2-二氯丙基等。
“羟基”指-OH基团。
“卤素”是指氟、氯、溴和碘,优选氟、氯和溴。
“氨基”指-NH 2
“氰基”指-CN。
“硝基”指-NO 2
“苄基”指-CH 2-苯基。
“羧基”指-C(O)OH。
“乙酰基”指-C(O)CH 3或Ac。
“羧酸酯基”指-C(O)O(烷基)或(环烷基),其中烷基、环烷基的定义如上所述。
“任选”意味着其所描述的事件可以但不必发生。例如,“AR任选被1到多个R c取代”该说明包含着AR基团可以被1到多个R c取代或者不被R c取代的情形。
“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。
本说明书所述的“取代”或“取代的”,如无特别指出,均是指基团可被一个或多个选自以下的基团取代: 烷基、烯基、炔基、烷氧基、烷硫基、烷基氨基、卤素、疏基、羟基、硝基、氰基、环烷基、杂环基、芳基、杂芳基、环烷氧基、杂环烷氧基、环烷硫基、杂环烷硫基、氨基、卤代烷基、羟烷基、羧基、羧酸酯基、=O、-C(O)R b、-OC(O)R b、-NR bR b、-C(O)NR bR b、-NR bC(O)R b、-S(O)NR bR b或-S(O) 2NR bR b,其中,R b的定义如通式(I)中所述。
本发明中立体化学的定义和惯例的使用通常参考以下文献:
S.P.Parker,Ed.,McGraw-Hill Dictionary of Chemical Terms(1984)McGraw-HillBook Company,New York;and Eliel,E.and Wilen,S.,"Stereochemistry of Organic Compounds",John Wiley&Sons,Inc.,New York,1994.本发明的化合物可以包含不对称中心或手性中心,因此存在不同的立体异构体。本发明的化合物所有的立体异构形式,包括但绝不限于,非对映体,对映异构体,阻转异构体,和它们的混合物,如外消旋混合物,组成了本发明的一部分。非对映异构体可以以其物理化学差异为基础,通过层析、结晶、蒸馏或升华等方法被分离为个别非对映异构体。对映异构体可以通过分离,使手性异构混合物转化为非对映异构混合物,其方式是与适当光学活性化合物(例如手性辅助剂,譬如手性醇或Mosher氏酰氯)的反应,分离非对映异构体,且使个别非对映异构体转化为相应的纯对映异构体。本发明的中间体与化合物也可以不同互变异构形式存在,且所有此种形式被包含在本发明的范围内。很多有机化合物都以光学活性形式存在,即它们有能力旋转平面偏振光的平面。在描述光学活性化合物时,前缀D、L或R、S用来表示分子手性中心的绝对构型。前缀d、l或(+)、(-)用来命名化合物平面偏振光旋转的符号,(-)或l是指化合物是左旋的,前缀(+)或d是指化合物是右旋的。这些立体异构体的原子或原子团互相连接次序相同,但是它们的立体结构不一样。特定的立体异构体可以是对映体,异构体的混合物通常称为对映异构体混合物。50:50的对映体混合物被称为外消旋混合物或外消旋体,这可能导致化学反应过程中没有立体选择性或立体定向性。术语“外消旋混合物”和“外消旋体”是指等摩尔的两个对映异构体的混合物,缺乏光学活性。
“互变异构体”或“互变异构的形式”是指不同能量的结构的同分异构体可以通过低能垒互相转化。例如质子互变异构体(即质子移变的互变异构体)包括通过质子迁移的互变,如酮式-烯醇式和亚胺-烯胺的同分异构化作用。原子价(化合价)互变异构体包括重组成键电子的互变。除非其他方面表明,本发明所描述的结构式包括所有的同分异构形式(如对映异构,非对映异构,和几何异构):例如含有不对称中心的R、S构型,双键的(Z)、(E)异构体,和(Z)、(E)的构象异构体。因此,本发明的化合物的单个立体化学异构体或其对映异构体,非对映异构体,或几何异构体的混合物都属于本发明的范围。
“药学上可接受的盐”指本发明化合物的盐,这类盐用于人或动物体内时具有安全性和有效性。化合物的盐可以通过在纯的溶液或合适的惰性溶解中用足量的碱或酸获得相应的加成盐。可药用的碱加成盐包括钠、钾、钙、铵、有机氨或镁盐等,可药用的酸加成盐包括无机酸盐和有机酸盐,所述的无机酸和有机酸包括盐酸、氢溴酸、碳酸、碳酸氢根、磷酸、磷酸一氢根、磷酸二氢根、硫酸、硫酸一氢根、乙酸、马来酸、丙二酸、琥珀酸、饭丁烯二酸、邻苯二甲酸、苯磺酸、对甲苯磺酸、柠檬酸和甲磺酸等(参见Berge et al.,“Pharmaceutical Salts”,Journal of Pharmaceutical Science 66:1-19(1977))。
本发明提供了一种新结构的KRAS突变体G12C抑制剂,试验结果表明,该四氢萘啶类衍生物表现出优异的KRAS突变体G12C抑制活性,同时表现出优异的安全性和选择性,可用于制备治疗癌症,尤其是肺癌、结直肠癌或胰腺癌等疾病的药物。
附图说明
图1是本发明所涉及化合物对KRAS G12C突变体MIAPaCa-2胰腺细胞KRAS/ERK1/2信号转导通路的影响结果图。
图2是本发明所涉及化合物对KRAS G12C突变体MIAPaCa-2胰腺异种移植瘤裸小鼠模型的体内抗肿瘤作用图。
图3是本发明所涉及化合物对KRAS G12C突变体MIAPaCa-2胰腺异种移植瘤裸小鼠模型的小鼠体重影响图。
具体实施方式
下面通过具体实施例对本发明的方法进行说明,以使本发明技术方案更易于理解、掌握,但本发明并不局限于此。下述实施例中 1H NMR图谱是用Bruker仪器(400MHz)测定而得,化学位移用ppm表示。使用四甲基硅烷内标准(0.00ppm)。 1H NMR的表示方法:s=单峰,d=双重峰,t=三重峰,q=四重峰,m=多重峰,br=宽峰,dd=双重峰的双重峰,dt=三重峰的双重峰。若提供偶合常数时,其单位为Hz。
质谱是用LC/MS仪测定得到,离子化方式为ESI。
高效液相色谱仪型号:安捷伦1260、赛默飞U3000;色谱柱型号:Waters xbrige C18(4.6*150mm,3.5μm);流动相:A:ACN,B:Water(0.1%H 3PO 4);流速:1.0mL/min;梯度:5%A for 1min,increase to 20%A within 4min,increase to 80%A within 8min,80%A for 2min,back to 5%A within 0.1min;波长:220nm;柱温箱:35℃。
薄层层析硅胶板使用烟台黄海HSGF254或青岛GF254硅胶板,薄层色谱法(TLC)使用的硅胶板采用的规格是0.2mm-0.3mm,薄层层析分离纯化产品采用的规格是0.4mm-0.5mm。
柱层析一般使用烟台黄海硅胶200-300目硅胶为载体。
在下列实例中,除非另有指明,所有温度为摄氏温度,除非另有指明,各种起始原料和试剂来自市售或者是根据已知的方法合成,市售原料和试剂均不经进一步纯化直接使用,除非另有指明,市售厂家包括但不限于国药集团,百灵威科技有限公司,梯希爱(上海)化成工业发展有限公司,上海毕得医药科技有限公司和上海迈瑞尔化学科技有限公司等。
CD 3OD:氘代甲醇
CDCl 3:氘代氯仿
DMSO-d 6:氘代二甲基亚砜
Pd 2(dba) 3:三(二亚苄基丙酮)二钯
Pd(dppf)Cl 2:[1,1'-双(二苯基膦基)二茂铁]二氯化钯
XantPhos:4,5-双二苯基膦-9,9-二甲基氧杂蒽
XPhos:2-二环己基磷-2,4,6-三异丙基联苯
HATU:2-(7-氧化苯并三氮唑)-N,N,N',N'-四甲基脲六氟磷酸酯
TLC:薄层色谱法
HPLC:高效液相色谱法
purity:纯度
&:和
氢气氛围是指反应瓶连接一个约1L容积的氢气气球。
实施例中无特殊说明,反应中的溶液是指水溶液。
实施例中无特殊说明,反应的温度为室温,为20℃-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,纯化化合物采用的柱层析的洗脱剂的体系或薄层色谱法的展开剂体系包括:A:石油醚和乙酸乙酯体系;B:二氯甲烷和甲醇体系;C:正己烷:乙酸乙酯;其中溶剂的体积比根据化合物的极性不同而不同,也可以加入少量的酸性或碱性试剂进行调节,如醋酸或三乙胺等。
中间体的制备
中间体1
1-苄基4-(叔丁基)(S)-2-(氰基甲基)哌嗪-1,4-二甲酸酯IN-1
Figure PCTCN2022073358-appb-000022
第一步 1-苄基4-(叔丁基)(R)-2-(羟甲基)哌嗪-1,4-二甲酸酯IN-1b
(R)-3-(羟甲基)哌嗪-1-羧酸叔丁酯IN-1a(35g,0.16mol)溶于乙酸乙酯(300mL)中,室温下加入水(300mL)和碳酸氢钠(40.8g,0.49mol),冷却至0℃,加入氯甲酸苄酯(41.4g,0.24mol),加完升至室温反应过夜,TLC显示反应完全。反应液萃取分液,有机相无水硫酸钠干燥,浓缩,粗品经硅胶柱纯化得标题化合物IN-1b(57g,收率100%)。
LC-MS:m/z=251.2[M-Boc+H] +
第二步 1-苄基4-(叔丁基)(R)-2-(((甲基磺酰基)氧基)甲基)哌嗪-1,4-二甲酸酯IN-1c
化合物IN-1b(57g,0.16mol)溶于二氯甲烷(400mL)中,室温下加入三乙胺(24.6g,0.24mol),冷却至0℃,加入甲基磺酰氯(22.4g,0.2mol),加完升至室温反应1小时,TLC显示反应完全。反应液加水(200mL),萃取分液,有机相水洗,无水硫酸钠干燥,浓缩得标题化合物IN-1c(70g,粗品),直接用于下一步。
第三步 1-苄基4-(叔丁基)(S)-2-(氰基甲基)哌嗪-1,4-二甲酸酯IN-1
化合物IN-1c(70g,粗品)溶于N,N-二甲基甲酰胺(350mL)中,室温下加入碳酸钾(45g,0.33mol)和三甲基氰硅烷(32.3g,0.33mol),升温至80℃反应5小时,TLC显示反应完全。反应液加水(500mL),乙酸乙酯萃取,合并有机相,水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱纯化得标题化合物IN-1(29.3g,两步收率50%)。
LC-MS:m/z=304.2[M-tBu+H] +
1H NMR(400MHz,CDCl 3)δ7.42-7.31(m,5H),5.17(s,2H),4.55(br,1H),4.20-3.88(m,3H),3.20-3.09(m,2H),2.86(br,1H),2.72-2.54(m,2H),1.48(s,9H).
中间体2
4-溴-3-氟-1,7萘啶-2-醇IN-2
Figure PCTCN2022073358-appb-000023
第一步 3-(3-乙氧基-3-氧代丙酰胺基)异烟酸甲酯IN-2b
3-氨基-4-吡啶羧酸甲酯IN-2a(1.0g,6.57mmol),丙二酸单乙酯(1.0g,7.88mmol)和吡啶(1.6g,19.71mmol)溶于乙酸乙酯(20mL)中,加入1-丙基磷酸酐(6.4g,10.06mmol,50%N,N-二甲基甲酰胺溶液),室温反应过夜,原料还有少量剩余。反应液加水淬灭,乙酸乙酯萃取,合并有机相,饱和食盐水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物IN-2b(1.3g,收率74%)。
第二步 2,4-二羟基喹啉-3-甲酸乙酯IN-2c
化合物IN-2b(300mg,1.13mmol)溶于甲醇(4mL)中,室温下加入甲醇钠(122mg,2.25mmol),升温至70℃反应3小时,原料还有少量剩余,补加甲醇钠(122mg,2.25mmol),70℃继续反应过夜,TLC显示原料反应完全。反应液浓缩,加水溶解,滴加稀盐酸(0.5mL)调节pH至4-5,固体析出,过滤,滤饼洗涤干燥得标题化合物IN-2c(180mg,粗品),直接用于下一步。
第三步 1,7-萘啶-2,4-二醇IN-2d
化合物IN-2c(2.0g,粗品)溶于盐酸(10mL,6N)中,加热回流反应过夜,TLC显示反应完全。反应液浓缩除去溶剂,加饱和碳酸氢钠水溶液调节体系pH=7,过滤,滤饼洗涤干燥得标题化合物IN-2d(1.3g,粗品),直接用于下一步。
LC-MS:m/z=163.1[M+H] +
第四步 3-氟-1,7-萘啶-2,4-二醇IN-2e
化合物IN-2d(50mg,粗品)分散在乙腈(3mL)中,室温下加入1-氯甲基-4-氟-1,4-重氮化双环[2.2.2]辛烷二(四氟硼酸盐)(330mg,0.93mmol),升温至60℃反应过夜,TLC显示反应完全。反应液冷却至室温,加水淬灭,乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,浓缩得标题化合物IN-2e(25mg,粗品),直接用于下一步。
第五步 2,4-二溴-3-氟-1,7-萘啶IN-2f
化合物IN-2e(600mg,粗品)分散在三溴氧磷(5mL)中,升温至120℃反应过夜,TLC显示反应完全。反应液冷却至室温,加入冰水和氢氧化钠溶液(15%),调节体系pH=8,过滤,滤饼洗涤烘干得标题化合物IN-2f(800mg,粗品),直接用于下一步。
第六步 4-溴-3-氟-1,7萘啶-2-醇IN-2
化合物IN-2f(800mg,粗品)分散在水(4.5mL),1,4-二氧六环(3mL)和氢溴酸(4.5mL,48%)中,升温至80℃反应过夜,TLC显示反应完全。反应液冷却至室温,氢氧化钠水溶液(15%)调节体系pH=7,乙酸乙酯萃取,合并有机相,无水硫酸钠干燥,浓缩,粗品经硅胶柱纯化得标题化合物IN-2(580mg,五步收率21%)。
LC-MS:m/z=245.0[M+H] +
1H NMR(400MHz,DMSO-d 6)δ12.45(br,1H),8.70(s,1H),8.49(d,J=5.2Hz,2H),7.73(d,J=5.2Hz,1H)
中间体3
7-苄基-1-(4-甲氧基苄基)-2-氧代-4-(甲苯磺酰氧基)-1,2,5,6,7,8-六氢-1,7-萘啶-3-甲酸甲酯IN-3
Figure PCTCN2022073358-appb-000024
第一步 1-苄基-5-((4-甲氧基苄基)氨基)-1,2,3,6-四氢吡啶-4-甲酸乙酯IN-3b
1-苄基-3-氧哌啶-4-甲酸乙酯IN-3a(15.0g,57.40mmol)溶于甲醇(150mL)中,室温下加入乙酸(1.5g,24.98mmol)和4-甲氧基苄胺(6.9g,50.30mmol),升温至50℃反应过夜,TLC显示反应完全。反应液冷却至室温,浓缩,粗品经硅胶柱层析纯化得标题化合物IN-3b(16.0g,收率73%)。
LC-MS:m/z=381.3[M+H] +
第二步 1-苄基-5-(3-甲氧基-N-(4-甲氧基苄基)-3-氧代丙酰胺基)-1,2,3,6-四氢吡啶-4-甲酸乙酯IN-3c
化合物IN-3b(8.0g,21.03mmol)溶于乙腈(80mL)中,室温下加入碳酸钾(5.8g,41.97mmol)和丙二酸酰氯甲酯(4.3g,31.49mmol),升温至50℃反应过夜,TLC显示反应完全。反应液冷却至室温,过滤,滤饼洗涤,滤液浓缩,粗品经硅胶柱层析纯化得标题化合物IN-3c(9.7g,收率96%)。
LC-MS:m/z=481.3[M+H] +
第三步 7-苄基-4-羟基-1-(4-甲氧基苄基)-2-氧代-1,2,5,6,7,8-六氢-1,7-萘啶-3-甲酸甲酯IN-3d
化合物IN-3c(10.0g,20.81mmol)溶于甲醇中(50mL)中,加入甲醇钠(2.2g,40.73mmol),室温反应2小时,TLC显示反应完全。反应液浓缩除去溶剂,加稀盐酸(2N)调节体系pH=6,乙酸乙酯萃取,合并有机相,水洗,无水硫酸钠干燥,浓缩,粗品用石油醚打浆得标题化合物IN-3d(9.0g,粗品),直接用于下一步。
LC-MS:m/z=435.2[M+H] +
第四步 7-苄基-1-(4-甲氧基苄基)-2-氧代-4-(甲苯磺酰氧基)-1,2,5,6,7,8-六氢-1,7-萘啶-3-甲酸甲酯IN-3
化合物IN-3d(2.0g,4.60mmol)溶于二氯甲烷(20mL)中,加入N,N-二异丙基乙胺(1.2g,9.29mmol)和4-甲基苯磺酰氯(1.05g,5.51mmol),室温反应2小时,TLC显示反应完全。反应液加入少量水,分液,有机相无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物IN-3(2.1g,两步收率75%)。
1H NMR(400MHz,DMSO-d 6)δ7.85(d,J=8.4Hz,2H),7.55(d,J=8.0Hz,2H),7.31-7.26(m,3H),7.23-7.19(m,2H),6.97(d,J=8.8Hz,2H),6.82(d,J=8.8Hz,2H),5.08(s,2H),3.73(s,3H),3.57(s,2H),3.52(s,5H),2.51-2.49(m,2H),2.45(s,3H),2.38-2.28(m,2H).
中间体4
((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲醇和((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲醇混合物IN-4
Figure PCTCN2022073358-appb-000025
第一步 2-(2-(氯甲基)烯丙基)-5-氧代吡咯烷-2-甲酸乙酯IN-4b
将(S)-5-氧代吡咯烷-2-甲酸乙酯IN-4a(300.0g,1.91mol)和3-氯-2-氯甲基丙烯(716.0g,5.73mol)加入到四氢呋喃(2.0L)中,降温至-40℃,氮气保护下,缓慢滴加双三甲基硅基胺基锂(3.82L,3.82mol,1N),滴加完毕,-40℃条件下继续搅拌1.0小时,TLC监测原料反应完全。反应液加入饱和氯化铵水溶液(1.0L)淬灭,乙酸乙酯(1.5L)萃取,合并有机相,饱和食盐水(1.0L)洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得黄色固体标题化合物IN-4b(303.0g,收率65%)。
LC-MS:m/z=246.1[M+H] +.
第二步 2-亚甲基-5-氧代四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯IN-4c
氮气保护下,将氢化钠(74.0g,1.85mol,60%)分批加入冷却至0℃的四氢呋喃(18L)中,再滴加化合物IN-4b(303.0g,1.23mol)的四氢呋喃(3L)混合液,滴加完毕,升温至回流反应6.0小时,LCMS监测原料反应完全。反应液浓缩至(4.0L),加入饱和氯化铵水溶液(3L)淬灭,乙酸乙酯萃取(2.0L),合并有机相,饱和食盐水(3L)洗,浓缩得棕色油状标题化合物IN-4c(140.0g,收率54%)。
LC-MS:m/z=210.2[M+H] +.
第三步 2,5-二氧四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯IN-4d
化合物IN-4c(140.0g,0.67mol)溶于丙酮/水(v/v=2/1;4.0L)的混合液中,室温加入二水锇酸钾(1.2g,3.25mmol)的水溶液,继续搅拌30分钟,冷却至0℃,分批加入高碘酸钾(572.0g,2.67mol),加料完 毕,缓慢升至室温继续搅拌1小时,TLC监测原料反应完全。反应液过滤,滤液加入水(2.5L)和二氯甲烷(2.5L),分层,水相二氯甲烷萃取(2.0L),合并有机相,饱和食盐水(2.5L)洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得棕色油状标题化合物IN-4d(68.6g,收率49%)。
LC-MS:m/z=212.1[M+H] +.
第四步 (2R,7aR)-2-羟基-5-氧代四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯和(2S,7aS)-2-羟基-5-氧代四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯混合物IN-4e
化合物IN-4d(68.6g,0.32mol)溶于甲醇(1.0L)中,冰水浴冷却至0℃,分批加入硼氢化钠(3.08g,81.19mmol),0℃继续反应1.0小时。TLC监测原料反应完全。加水(20mL)搅拌1.0小时,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得黄色油状反式构型杂质和黄色油状标题化合物IN-4e(32.8g,收率47%)。
LC-MS:m/z=214.1[M+H] +.
第五步 (2R,7aS)-2-氟-5-氧代四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯和(2S,7aR)-2-氟-5-氧代四氢-1H-吡咯嗪-7a(5H)-甲酸乙酯混合物IN-4f
化合物IN-4e(32.8g,0.15mol)溶于二氯甲烷(800mL)中,氮气保护下,降温至-60℃缓慢滴加二乙胺基三氟化硫(37.2g,0.23mmol)的二氯甲烷(200mL)溶液,滴加完毕后升至室温反应过夜,TLC监测原料反应完全,加入甲醇(200mL)淬灭,浓缩,粗品经硅胶柱层析纯化得黄色油状标题化合物IN-4f(18.8g,收率57%)。
LC-MS:m/z=216.1[M+H] +.
第六步 ((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲醇和((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲醇混合物IN-4
氮气保护下,将氢化铝锂(9.95g,0.26mol)加入四氢呋喃(200ml)中,冰水浴冷却至0℃,滴加化合物IN-4f(18.8g,87.35mmol)溶于四氢呋喃(30mL)溶液,滴完升温至回流反应4.5小时,LCMS监测原料反应完全,冷却至0℃,依次缓慢滴加水(10mL),氢氧化钠(15%,10mL),水(30mL),搅拌30分钟,反应液垫硅藻土过滤,滤液浓缩,粗品经硅胶柱层析纯化得淡黄色油状标题化合物IN-4f(6.8g,收率49%)。
1H NMR(400MHz,CDCl 3)δ5.29-5.24(m,0.5H),5.15-5.10(m,0.5H),3.27(s,2H),3.27-3.10(m,1H),3.09-2.97(m,2H),2.96-2.88(m,1H),2.18-2.20(m,2H),1.96-1.82(m,2H),1.82-1.70(m,2H).
实施例1
2-((S)-1-丙烯酰基-4-(7-(8-氯萘-1-基)-3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈1
Figure PCTCN2022073358-appb-000026
第一步 (S)-2-(氰基甲基)哌嗪-1-甲酸苄酯1a
中间体IN-1(10g,27.82mmol)溶于二氯甲烷(15mL)中,加入三氟乙酸(15mL),室温反应2小时,TLC显示反应完全。反应液浓缩除去溶剂,加入饱和碳酸氢钠水溶液,二氯甲烷萃取,合并有机相,无水硫酸钠干燥,浓缩得目标化合物标题化合物1a(6.5g,粗品),直接用于下一步。
第二步 (S)-4-溴-3-氟-2-((1-甲基吡咯烷-2-基)甲氧基)-1,7-萘啶1b
中间体IN-2(300mg,1.24mmol),化合物(S)-(1-甲基吡咯烷-2-基)甲醇(284mg,2.47mmol)和三苯基膦(1.3g,4.96mmol)依次加入三口瓶中,氮气保护下,加入四氢呋喃(8mL),冷却至0℃,加入偶氮二甲酸二异丙酯(998mg,4.94mmol),加完升至室温反应过夜,TLC显示原料消耗完。反应液浓缩,粗品经硅胶柱纯化得标题化合物1b(220mg,收率52%)。
第三步 (S)-2-(氰基甲基)-4-(3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-1,7-萘啶-4-基)哌嗪-1-甲酸苄酯1c
化合物1b(500mg,1.47mmol),化合物1a(762mg,2.94mmol),Pd 2(dba) 3(296mg,0.29mmol),Xantphos(340mg,0.59mmol)和碳酸铯(1.2g,3.69mmol)依次加入三口瓶中,氮气保护下,加入甲苯(20mL),加热回流反应24小时。反应液冷却至室温,浓缩除去溶剂,粗品经硅胶柱纯化得标题化合物1c(150mg,收率20%)。
LC-MS:m/z=519.3[M+H] +
第四步 (S)-2-(氰基甲基)-4-(3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-甲酸苄酯1d
化合物1c(150mg,0.29mmol)溶于醋酸(2mL)中,加入硼氢化钠(150mg,3.95mmol),室温反应5小时。反应液浓缩,粗品中加入少量碳酸氢钠和二氯甲烷搅拌,过滤,滤液浓缩,粗品经硅胶柱纯化得标题化合物1d(100mg,收率66%)。
第五步 (S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸苄酯1e
化合物1d(100mg,0.19mmol),化合物1-溴-8-氯萘(94mg,0.38mmol),Pd 2(dba) 3(35mg,0.04mmol),Xantphos(44.3mg,0.08mmol)和碳酸铯(187mg,0.57mmol)依次加入三口瓶中,氮气保护下,加入甲苯(6mL),加热回流反应4小时,TLC显示反应完全。反应液冷却至室温,浓缩除去溶剂,粗品经硅胶柱纯化得标题化合物1e(60mg,收率46%)。
第六步 2-((S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈1f
化合物1e(60mg,0.09mmol)溶于二氯甲烷(3mL)中,冷却至0℃,加入三溴化硼(220mg,0.88mmol),0℃反应1小时。反应液过滤,滤饼二氯甲烷洗涤,干燥得标题化合物1f(60mg,粗品),直接用于下一步。
第七步 2-((S)-1-丙烯酰基-4-(7-(8-氯萘-1-基)-3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈1
化合物1f(30mg,粗品)溶于二氯甲烷(2mL)中,室温下加入碳酸氢钠(46mg,0.55mmol)和水(2mL),冷却至0℃,加入丙烯酰氯(2.5mg,0.03mmol),0℃反应30分钟。反应液萃取分液,有机相饱和食盐水洗涤,无水硫酸钠干燥,浓缩,粗品经Prep-HPLC纯化得标题化合物1(4.5mg,两步收率9%)。
LC-MS:m/z=603.3[M+H] +
1H NMR(400MHz,CD 3OD)δ7.82(d,J=8.0Hz,1H),7.69-7.64(m,1H),7.53-7.46(m,2H),7.39-7.31(m,2H),7.00-6.71(m,1H),6.28(d,J=16.8Hz,1H),5.82(d,J=10.0Hz,1H),5.14-5.11(m,0.5H),4.80-4.64(m,2H),4.59-4.38(m,1H),4.38-4.20(m,1H),4.15-3.98(m,0.5H),3.87-3.72(m,2H),3.70-3.51(m,4H),3.25-3.12(m,4H),3.01(s,3H),2.81-2.72(m,1H),2.45-2.24(m,1H),2.23-1.70(m,4H),1.52-1.45(m,1H),1.37-1.17(m,2H).(96.18%purity by HPLC)
实施例2
2-((S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈2
Figure PCTCN2022073358-appb-000027
化合物1f(30mg,0.056mmol)溶于N,N-二甲基甲酰胺(2mL)中,室温下依次加入2-氟丙烯酸(10mg,0.11mmol),N,N-二异丙基乙胺(21mg,0.16mmol)和HATU(31mg,0.08mmol),室温反应1.5小时,TLC显示反应完全。反应液加入少量水淬灭,乙酸乙酯萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,浓缩,粗品经硅胶柱纯化得标题化合物2(3.5mg,收率10%)。
LC-MS:m/z=621.3[M+H] +
1H NMR(400MHz,CD 3OD)δ7.74-7.70(m,1H),7.59-7.54(m,1H),7.43-7.36(m,2H),7.29-7.21(m,2H),5.31-5.08(m,4H),4.59-4.51(m,4H),4.39-4.25(m,1H),4.19(d,J=16.4Hz,1H),3.73-3.62(m,1H),3.57-3.49(m,3H),3.14-2.82(m,6H),2.79(s,3H),2.11-2.07(m,2H),1.95-1.90(m,4H).(96.78%purity by HPLC)
实施例3
2-((S)-1-丙烯酰基-4-(7-(8-氯萘-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈3
Figure PCTCN2022073358-appb-000028
第一步 (S)-7-苄基-4-(4-((苄氧基)羰基)-3-(氰基甲基)哌嗪-1-基)-1-(4-甲氧基苄基)-2-氧代-1,2,5,6,7,8-六氢-1,7-萘啶-3-甲酸甲酯3a
中间体IN-3(200mg,0.33mmol)溶于乙腈(3mL)中,室温下加入N,N-二异丙基乙胺(132mg,1.02mmol)和化合物1a(176mg,0.68mmol),升温至80℃反应过夜,LCMS显示反应完全。反应液冷却至室温,浓缩,粗品经硅胶柱层析纯化得标题化合物3a(137mg,收率60%)。
LC-MS:m/z=676.3[M+H] +
第二步 (S)-4-(7-苄基-2-羟基-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸叔丁酯3b
化合物3a(400mg,0.59mmol)溶于甲醇(2mL)中,室温下加入盐酸(6mL,4N),升温至100℃反应20小时,LCMS显示反应完全。反应液浓缩除去溶剂,加入二氯甲烷(10mL)溶解,加入三乙胺(1mL)和二碳酸二叔丁酯(1mL),室温搅拌过夜。反应液浓缩,粗品经硅胶柱层析纯化得标题化合物3b(105mg,收率38%)。
LC-MS:m/z=464.3[M+H] +
第三步 (S)-2-(氰甲基)-4-(2-羟基-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-甲酸叔丁酯3c
化合物3b(100mg,0.22mmol)分散在甲醇(10mL)中,室温下加入甲酸铵(82mg,1.30mmol)和钯/碳(35mg,10%),升温至60℃反应5小时,TLC显示反应完全。反应液垫硅藻土过滤,滤液浓缩,粗品经硅胶柱层析纯化得标题化合物3c(55mg,收率68%)。
第四步 (S)-4-(4-(叔丁氧羰基)-3-(氰基甲基)哌嗪-1-基)-2-羟基-5,8-二氢-1,7-萘啶-7(6H)-甲酸苄酯3d
化合物3c(55mg,0.15mmol)分散在二氯甲烷(2mL)中,加入三乙胺(30mg,0.30mmol)和氯甲酸苄酯(25.1mg,0.15mmol),室温反应3小时,TLC显示反应完全。反应液加入少量水,萃取分液,有机相水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物3d(48mg,收率64%)。
第五步 4-((S)-4-(叔丁氧羰基)-3-(氰基甲基)哌嗪-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,8-二氢-1,7-萘啶-7(6H)-甲酸苄酯3e
化合物3d(48mg,0.09mmol),化合物(S)-(1-甲基吡咯烷-2-基)甲醇(32.7mg,0.28mmol)和三苯基膦(99.3mg,0.38mmol)依次加入三口瓶中,氮气保护下,加入四氢呋喃(5mL),冷却至0℃,加入偶氮二甲酸二异 丙酯(76.6mg,0.38mmol),加完升至室温反应过夜,TLC显示原料被消耗。反应液浓缩,粗品经硅胶柱层析纯化得标题化合物3e(50mg,收率87%)。
LC-MS:m/z=605.4[M+H] +
第六步 (S)-2-(氰基甲基)-4-(2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-甲酸叔丁酯3f
化合物3e(50mg,0.08mmol)溶于甲醇(5mL)中,加入钯/碳(45mg,10%),氢气氛围下室温反应3小时,TLC显示反应完全。反应液垫硅藻土过滤,滤液浓缩得标题化合物3f(25mg,粗品),直接用于下一步。
第七步 (S)-4-(7-(8-氯萘-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸叔丁酯3g
化合物3f(25mg,粗品),1-溴-8-氯萘酚(26mg,0.11mmol),Pd 2(dba) 3(10mg,0.11mmol),Xantphos(12.3mg,0.02mmol)和碳酸铯(52mg,0.16mmol)依次加入三口瓶中,氮气保护下,加入甲苯(6mL),回流反应4小时,TLC显示反应完全。反应液浓缩除去溶剂,粗品经硅胶柱层析纯化得标题化合物3g(15mg,两步收率29%)。
LC-MS:m/z=631.3[M+H] +
第八步 2-((S)-4-(7-(8-氯萘-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈3h
化合物3g(15mg,0.02mmol)溶于二氯甲烷(4mL)中,加入三氟乙酸(2mL),室温搅拌30分钟,TLC显示反应完全。反应液浓缩除去溶剂,加入饱和碳酸氢钠水溶液,二氯甲烷萃取,合并有机相,无水硫酸钠干燥,浓缩得标题化合物3h(10mg,粗品),直接用于下一步。
第九步 2-((S)-1-丙烯酰基-4-(7-(8-氯萘-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈3
化合物3h(10mg,粗品)溶于二氯甲烷(3mL)中,室温下依次加入水(3mL),碳酸氢钠(24mg,0.28mmol)和丙烯酰氯(1.5mg,0.02mmol),室温搅拌10分钟,TLC显示反应完全。反应液萃取分液,有机相水洗,无水硫酸钠干燥,浓缩,粗品经Prep-HPLC纯化得标题化合物3的甲酸盐(5mg,两步收率24%)。
LC-MS:m/z=585.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ7.90(d,J=8.0Hz,1H),7.72(d,J=8.0Hz,1H),7.62-7.49(m,2H),7.46-7.42(m,1H),7.40-7.30(m,1H),6.98-6.72(m,1H),6.29-6.10(m,2H),5.78(dd,J=2.0,10.4Hz,1H),5.11-4.65(m,1H),4.54-4.35(m,1H),4.28-4.14(m,2H),4.09-3.97(m,1H),3.80(dd,J=4.4,15.6Hz,1H),3.60-3.48(m,4H),3.18-2.89(m,7H),2.83-2.65(m,2H),2.32(s,3H),2.20-2.11(m,1H),1.95-1.85(m,1H),1.74-1.49(m,3H).(86.78%purity by HPLC)
实施例4
2-((S)-4-(7-(8-氯萘-1-基)-2-(((S)-1-甲基吡咯烷-2-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈4
Figure PCTCN2022073358-appb-000029
化合物3h(35mg,0.066mmol)溶于N,N-二甲基甲酰胺(2mL),室温下依次加入2-氟丙烯酸(10mg,0.11mmol),N,N-二异丙基乙胺(21mg,0.16mmol)和HATU(38mg,0.10mmol),室温反应1.5小时,TLC显示反应完全。反应液加入少量水淬灭,乙酸乙酯萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物4(4.0mg,收率10%)。
LC-MS:m/z=603.3[M+H] +
1H NMR(400MHz,CD 3OD)δ7.85-7.78(m,1H),7.66(dd,J=2.4,8.0Hz,1H),7.53-7.45(m,2H),7.40-7.30(m,2H),6.40(d,J=16.8Hz,1H),5.42-5.19(m,2H),4.65-4.51(m,2H),4.46-4.33(m,2H),3.83-3.75(m,1H),3.70-3.43(m,6H),3.30-3.11(m,4H),3.10-2.97(m,2H),2.94-2.63(m,6H),2.33-2.23(m,1H),2.15-2.04(m,1H),2.04-1.88(m,2H).(91.98%purity by HPLC)
实施例5
(S)-2-(4-(7-(8-氯萘-1-基)-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈5
Figure PCTCN2022073358-appb-000030
第一步 1-(叔丁基)2-(3-氯丙基)吡咯烷-1,2-二甲酸2-甲酯5b
Boc-脯氨酸甲酯5a(5.0g,21.81mmol)溶于干燥的四氢呋喃(20mL)中,氮气保护下,降温至-65℃,恒压滴液漏斗缓缓加入双三甲基硅基胺基锂(32mL,32mmol,1M四氢呋喃溶液),加完-65℃反应1小时,缓缓加入1-溴-3-氯丙烷(17g,109.2mmol),加完缓慢升至室温反应2小时,TLC显示反应完成。反应液倒水中,乙酸乙酯萃取,合并有机相,饱和食盐水洗,无水硫酸钠干燥,浓缩,粗品硅胶柱层析纯化得黄色液体标题化合物5b(5.1g,收率80%)。
LC-MS:m/z=250.1[M-tBu+H] +
第二步 2-(3-氯丙基)吡咯烷-2-甲酸甲酯5c
化合物5b(4.1g,13.41mmol)溶于二氯甲烷(20mL)中,室温缓缓加入三氟醋酸(6mL),加完室温反应1小时,TLC显示反应完成。反应液浓缩得黄色粘稠液体标题化合物5c(3.5g,粗品),直接用于下一步。
第三步 四氢-1H-吡咯嗪-7a(5H)-甲酸甲酯5d
化合物5c(3.5g,粗品)溶于甲醇(20mL)中,依次加入碘化钾(232mg,1.40mmol)和碳酸钾(5.7g,41.23mmol),室温反应2小时,体系由黄色透明液体慢慢变为白色浑浊液,TLC显示反应完成。反应液浓缩,剩余物倒入水中,乙酸乙酯萃取,合并有机相,饱和食盐水洗,无水硫酸钠干燥,浓缩,粗品硅胶柱层析纯化得黄色粘稠液体标题化合物5d(2.0g,两步收率88%)。
LC-MS:m/z=170.2[M+H] +.
第四步 (四氢-1H-吡咯嗪-7a(5H)-基)甲醇5e
化合物5d(2.0g,11.82mmol)溶于四氢呋喃(10mL)中,降温至0℃,缓缓加入铝锂氢(1.3g,34.26mmol),室温反应3小时,TLC显示反应完成。反应液加水(1.3mL)淬灭,室温下搅拌0.5小时,过滤,滤液浓缩,粗品硅胶柱层析纯化得黄色粘稠液体标题化合物5e(1.3g,收率78%)。
LC-MS:m/z=142.2[M+H] +
1H NMR(400MHz,DMSO-d 6):δ4.37(br,1H),3.04(s,2H),2.86-2.81(m,2H),2.49-2.44(m,2H),1.81-1.69(m,4H),1.65-1.57(m,2H),1.40-1.35(m,2H).
第五步 (S)-4-(4-(叔丁氧羰基)-3-(氰基甲基)哌嗪-1-基)-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,8-二氢-1,7-萘啶-7(6H)-甲酸苄酯5f
化合物3d(100mg,0.20mmol),化合物5e(84mg,0.59mmol)和三苯基膦(207mg,0.79mmol)依次加入三口瓶中,氮气保护下,加入四氢呋喃(5mL),冷却至0℃,加入偶氮二甲酸二异丙酯(160mg,0.79mmol),加完升至室温反应过夜,TLC显示原料被耗尽。反应液浓缩,粗品经硅胶柱层析纯化得标题化合物5f(100mg,收率80%)。
第六步 (S)-2-(氰基甲基)-4-(2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1- 甲酸叔丁酯5g
化合物5f(100mg,0.16mmol)溶于甲醇(10mL)中,加入钯/碳(60mg,10%),氢气氛围下室温反应3小时,TLC显示反应完全。反应液垫硅藻土过滤,滤液浓缩得目标化合物标题化合物5g(90mg,粗品),直接用于下一步。
第七步 (S)-4-(7-(8-氯萘-1-基)-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸叔丁酯5h
化合物5g(90mg,粗品),1-溴-8-氯萘酚(133mg,0.54mmol),Pd 2(dba) 3(50mg,0.05mmol),Xantphos(63mg,0.11mmol)和碳酸铯(236mg,0.72mmol)依次加入三口瓶中,氮气保护下,加入甲苯(10mL),加热回流反应4小时,TLC显示反应完全。反应液浓缩除去溶剂,粗品过硅胶柱层析纯化得标题化合物5h(100mg,两步收率77%)。
LC-MS:m/z=657.4[M+H] +
第八步 (S)-2-(4-(7-(8-氯萘-1-基)-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈5i
化合物5h(100mg,0.15mmol)溶于二氯甲烷(4mL)中,加入三氟乙酸(2mL),室温搅拌30分钟,TLC显示反应完全。反应液浓缩除去溶剂,粗品加入饱和碳酸氢钠水溶液,二氯甲烷萃取,合并有机相,无水硫酸钠干燥,浓缩,粗品过硅胶柱层析纯化得标题化合物5i(30mg,收率35%)。
LC-MS:m/z=557.3[M+H] +
第九步 (S)-2-(4-(7-(8-氯萘-1-基)-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈5
化合物5i(30mg,0.05mmol)溶于N,N-二甲基甲酰胺(2mL)中,室温下依次加入2-氟丙烯酸(7.3mg,0.08mmol),N,N-二异丙基乙胺(21mg,0.16mmol)和HATU(31mg,0.08mmol),室温反应1.5小时,TLC显示反应完全。反应液加入少量水,乙酸乙酯萃取,合并有机相,水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物5(9.0mg,收率27%)。
LC-MS:m/z=629.3[M+H] +
1H NMR(400MHz,DMSO-d 6)δ7.93(d,J=8.0H Z,1H),7.74(d,J=8.0H Z,1H),7.62-7.50(m,2H),7.46(t,J=7.6H Z,1H),7.34(t,J=6.8H Z,1H),6.30(d,J=18.4H Z,1H),5.46-5.37(m,1H),5.37-5.20(m,1H),4.24(d,J=17.2H Z,1H),4.10(s,2H),3.91-3.65(m,2H),3.59-3.39(m,4H),3.25-2.94(m,7H),2.93-2.62(m,4H),2.05-1.67(m,9H).(92.58%purity by HPLC)
实施例6
2-((S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈6
Figure PCTCN2022073358-appb-000031
第一步 (3R,4R)-3-羟基-4-甲氧基吡咯烷-1-甲酸叔丁酯6b-1&(3S,4S)-3-羟基-4-甲氧基吡咯烷-1-甲酸叔丁酯6b-2
3-N-叔丁氧羰基-6-氧杂-3-氮杂二环[3.1.0]己烷6a(10.6g,57.2mmol)溶于甲醇(50mL)中,氮气保护下加入甲醇钠(3.71g,68.7mmol),升温至60℃反应5小时,TLC监测原料反应完全。反应液冷却至室温,加水稀释,乙酸乙酯萃取,合并有机相,水洗,饱和食盐水洗,无水硫酸钠干燥,浓缩,粗品硅胶柱层析纯化得白色固体标题化合物6b(8.0g,收率65%)。化合物6b(6.0g)手性拆分(LC016-1,21.2*250mm,5um,20mL/min,EtOH:Hexane=5:95)得化合物6b-2(1号峰,RT 10.62min)(1.8g,收率77%)和化合物6b-1(2号峰,RT 13.01min)(2.3g,收率60%)。
第二步 (3R,4R)-4-甲氧基-1-甲基吡咯烷-3-醇6c-1
将氢化铝锂(595mg,15.6mmol)分散至干燥的四氢呋喃(30mL)中,降温至0℃,氮气保护下,滴加6b-1(1.7g,7.8mmol)的四氢呋喃(15mL)溶液,滴加完毕,升温至50℃反应2小时,TLC监测原料反应完全。反应液降至室温,依次加入水(0.6mL)、15%氢氧化钠溶液(0.6mL)和水(1.8mL)淬灭,过滤,滤液浓缩得黄色油状物标题化合物6c-1(1.0g,粗品),直接用于下一步。
1H NMR(400MHz,DMSO-d 6)δ5.02(s,1H),3.93(s,1H),3.55-3.52(m,1H),3.22(s,3H),2.71-2.64(m,2H),2.34(dd,J=3.6,9.6Hz,1H),2.21(dd,J=4.8,9.6Hz,1H),2.16(s,3H).
第三步 4-溴-3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-1,7-萘啶6d
中间体IN-2(500mg,2.07mmol),化合物6c-1(405mg,3.09mmol)和三苯基膦(1.6g,6.1mmol)依次加入三口瓶中,氮气保护下,加入四氢呋喃(15mL),冷却至0℃,加入偶氮二甲酸二异丙酯(1.2g,5.93mmol),加完升至室温反应过夜,TLC显示反应完全。反应液浓缩,粗品经硅胶柱纯化得标题化合物6d(350mg,收率48%)。
LC-MS:m/z=358.1[M+H] +
第四步 (S)-2-(氰基甲基)-4-(3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-1,7-萘啶-4-基)哌嗪-1-甲酸苄酯6e
化合物6d(350mg,0.98mmol),化合物1a(510mg,1.97mmol),Pd 2(dba) 3(270mg,0.29mmol),Xantphos(341mg,0.59mmol)和碳酸铯(958mg,2.9mmol)依次加入三口瓶中,氮气保护下,加入甲苯(20mL),加热回流反应24小时。反应液冷却至室温,浓缩除去溶剂,粗品经硅胶柱纯化得标题化合物6e(150mg,收率29%)。
LC-MS:m/z=535.3[M+H] +
第五步 (S)-2-(氰基甲基)-4-(3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-甲酸苄酯6f
化合物6e(150mg,0.28mmol)溶于醋酸(2mL)中,加入硼氢化钠(53mg,1.39mmol),室温反应5小时。反应液浓缩,加入少量碳酸氢钠,加入二氯甲烷,过滤,滤饼二氯甲烷洗涤,滤液浓缩,粗品经硅胶柱纯化得标题化合物6f(80mg,收率53%)。
LC-MS:m/z=539.3[M+H] +
第六步 (S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸苄酯6g
化合物6f(80mg,0.15mmol),化合物1-溴-8-氯萘(110mg,0.45mmol),Pd 2(dba) 3(41mg,0.04mmol),Xantphos(51.6mg,0.09mmol)和碳酸铯(193mg,0.59mmol)依次加入三口瓶中,氮气保护下,加入甲苯(15mL),加热回流反应4小时,TLC显示反应完全。反应液浓缩除去溶剂,粗品经硅胶柱纯化得标题化合物6g(60mg,收率58%)。
LC-MS:m/z=699.3[M+H] +
第七步 2-((S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈6h
化合物6g(60mg,0.09mmol)溶于二氯甲烷(2mL)中,冷却至0℃,加入三溴化硼(215mg,0.86mmol),0℃反应1小时。反应液过滤,滤饼二氯甲烷洗涤干燥得标题化合物6h(25mg,粗品),直接用于下一步。
LC-MS:m/z=565.3[M+H] +
第八步 2-((S)-4-(7-(8-氯萘-1-基)-3-氟-2-(((3R,4R)-4-甲氧基-1-甲基吡咯烷-3-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈6
化合物6h(25mg,粗品)溶于N,N-二甲基甲酰胺(2mL)中,室温下依次加入2-氟丙烯酸(8.0mg,0.09mmol),N,N-二异丙基乙胺(17mg,0.13mmol)和HATU(25mg,0.066mmol),室温反应1.5小时,TLC显示反应完全。反应液加入少量水,乙酸乙酯萃取,合并有机相,水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱纯化得标题化合物6(5.0mg,收率18%)。
LC-MS:m/z=637.3[M+H] +
1H NMR(400MHz,CD 3OD)δ7.81(d,J=8.4Hz,1H),7.74-7.59(m,2H),7.56-7.42(m,2H),7.41-7.25(m,2H),5.40-5.22(m,3H),4.31-4.20(m,2H),4.05-4.00(m,1H),3.84-3.69(m,1H),3.66-3.57(m,2H),3.38(d,J=8.4Hz,3H),3.27-2.99(m,8H),2.95-2.90(m,1H),2.78-2.69(m,1H),2.56(dd,J=3.2,9.6Hz,1H),2.36(d,J=5.2Hz,3H),1.76-1.67(m,1H),1.49-1.41(m,1H).(92.20%purity by HPLC)
实施例7
(S)-2-(4-(7-(8-氯萘-1-基)-2-((1-甲基哌啶-4-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈7
Figure PCTCN2022073358-appb-000032
第一步 (S)-4-(4-(叔丁氧羰基)-3-(氰基甲基)哌嗪-1-基)-2-((1-甲基哌啶-4-基)氧基)-5,8-二氢-1,7-萘啶-7(6H)-甲酸苄酯7a
化合物3d(120mg,0.24mmol),1-甲基-4-哌啶醇(82mg,0.71mmol)和三苯基膦(248mg,0.95mmol)依次加入三口瓶中,氮气保护下,加入四氢呋喃(5mL),冷却至0℃,加入偶氮二甲酸二异丙酯(191.2mg,0.95mmol),加完升至室温反应过夜,TLC显示原料被耗尽。反应液浓缩,粗品经硅胶柱层析纯化得标题化合物7a(100mg,收率70%)。
LC-MS:m/z=605.4[M+H] +
第二步 (S)-2-(氰基甲基)-4-(2-((1-甲基哌啶-4-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-甲酸叔丁酯7b
化合物7a(100mg,0.17mmol)溶于甲醇(10mL)中,加入钯/碳(60mg,10%),氢气氛围下室温反应3小时,TLC显示反应完全。反应液垫硅藻土过滤,滤液浓缩得标题化合物7b(75mg,粗品),直接用于下一步。
第三步 (S)-4-(7-(8-氯萘-1-基)-2-((1-甲基哌啶-4-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-甲酸叔丁酯7c
化合物7b(75mg,粗品),化合物1-溴-8-氯萘(125mg,0.51mmol),Pd 2(dba) 3(47mg,0.05mmol),Xantphos(59mg,0.1mmol)和碳酸铯(221mg,0.68mmol)依次加入三口瓶中,氮气保护下,加入甲苯(10mL),加热回流反应4小时,TLC显示反应完全。反应液浓缩除去溶剂,粗品过硅胶柱层析纯化得标题化合物7c(40mg,两步收率38%)。
LC-MS:m/z=631.3[M+H] +
第四步 (S)-2-(4-(7-(8-氯萘-1-基)-2-((1-甲基哌啶-4-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈7d
化合物7c(40mg,0.06mmol)溶于二氯甲烷(4mL)中,加入三氟乙酸(2mL),室温搅拌30分钟,TLC显示反应完全。反应液浓缩除去溶剂,粗品加入饱和碳酸氢钠水溶液,二氯甲烷萃取,合并有机相,无水硫酸钠干燥,浓缩得标题化合物7d(20mg,粗品),直接用于下一步。
第五步 (S)-2-(4-(7-(8-氯萘-1-基)-2-((1-甲基哌啶-4-基)氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈7
化合物7d(20mg,粗品)溶于N,N-二甲基甲酰胺(2mL)中,室温下依次加入2-氟丙烯酸(5.1mg,0.06mmol),N,N-二异丙基乙胺(15mg,0.12mmol)和HATU(38mg,0.10mmol),室温反应1.5小时,TLC显示反应完全。反应液加入少量水,乙酸乙酯萃取,合并有机相,水洗,无水硫酸钠干燥,浓缩,粗品经硅胶柱层析纯化得标题化合物7(4.7mg,两步收率12%)。
LC-MS:m/z=603.3[M+H] +
1H NMR(400MHz,CD 3OD)δ7.82(d,J=8.0H Z,1H),7.66(dd,J=3.2,8.0H Z,1H),7.53-7.45(m,2H),7.39-7.29(m,2H),6.44-6.29(m,1H),5.43-5.21(m,3H),4.35(d,J=16.4H Z,1H),3.86-3.65(m,2H),3.65-3.47(m,4H),3.46-3.35(m,3H),3.24-2.95(m,6H),2.91-2.67(m,6H),2.45-2.35(m,1H),2.33-2.23(m,1H),2.12-1.99(m,2H).(97.99%purity by HPLC)
实施例8
2-((S)-4-(7-(8-氯萘-1-基)-2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈和2-((S)-4-(7-(8-氯萘-1-基)-2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈混合物8
Figure PCTCN2022073358-appb-000033
第一步 (S)-4-(7-苄基-2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰甲基)哌嗪-1-羧酸叔丁酯和(S)-4-(7-苄基-2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰甲基)哌嗪-1-羧酸叔丁酯混合物8a
称取化合物3b(250mg,0.55mmol),中间体IN-4(230mg,1.65mmol,反式异构体)和三正丁基膦(668mg,3.30mmol)溶于干燥的四氢呋喃(5mL)中,50℃下分批加入偶氮二甲酸二异丙酯(668mg,3.30mmol),保温反应1小时,TLC监测原料反应完全,LCMS显示有两个目标产物的分子量。反应液降至室温,加水(15mL)稀释,乙酸乙酯(30mL)萃取,合并有机相,饱和食盐水(20mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后经硅胶柱层析(石油醚/乙酸乙酯=1:1)纯化得到无色油状物标题化合物8a(94mg,收率28.8%)。
LC-MS:m/z=605.4[M+H] +
1HNMR(400MHz,CDCl 3)δ7.42-7.30(m,4H),7.29-7.26(m,1H),6.12(s,1H),5.30-5.28(s,0.5H),5.19-5.14(m,0.5H),5.01-4.86(m,1H),4.70-4.51(m,1H),4.09-3.99(m,2H),3.95-3.89(m,1H),3.77-3.48(m,4H),3.30-3.10(m,6H),2.99-2.82(m,4H),2.75-2.59(m,4H),2.16-2.06(m,2H),2.03-1.96(m,1H),1.94-1.81(m,3H),1.50(s,9H).
第二步 (S)-2-(氰基甲基)-4-(2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-羧酸叔丁酯和(S)-2-(氰基甲基)-4-(2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-羧酸叔丁酯混合物8b
称取化合物8a(92mg,0.14mmol)溶于甲醇(6mL)中,依次加入钯/碳(50mg,10%)和甲酸铵(91mg,1.44mmol),升温至60℃反应3小时,TLC监测原料反应完全。反应液降温至室温,垫硅藻土过滤,滤液浓缩,加乙酸乙酯(15mL)和水(5mL)稀释,萃取分液,有机层依次饱和碳酸氢钠溶液(5mL)和饱和食盐水(5mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到无色油状标题化合物8b(64mg,收率82%)。
LC-MS:m/z=515.3[M+H] +
第三步 (S)-4-(7-(8-氯萘-1-基)-2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰甲基)哌嗪-1-羧酸叔丁酯和(S)-4-(7-(8-氯萘-1-基)-2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰甲基)哌嗪-1-羧酸叔丁酯混合物8c
称取化合物8b(40mg,0.08mmol),1-溴-8-氯萘(51mg,0.21mmol)溶于甲苯(1mL)中,依次加入Pd 2(dba) 3(14mg,0.015mmol),Xantphos(18mg,0.03mmol)和碳酸铯(104mg,0.32mmol),置换氮气三次,升温至100℃反应16小时,TLC监测原料反应完全。反应液冷却至室温,加水(5mL)稀释,乙酸乙酯(10 mL)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液浓缩后经硅胶柱层析纯化得浅黄色固体标题化合物8c(30mg,粗品),直接用于下一步。
LC-MS:m/z=675.3[M+H] +
1HNMR(400MHz,CDCl 3)δ7.77-7.71(m,1H),7.61-7.54(m,1H),7.53-7.48(m,1H),7.48-7.38(m,1H),7.35-7.29(m,1H),7.29-7.26(m,0.55H),7.23-7.19(m,0.45H),6.22(s,0.45H),6.17(s,0.55H),5.32(s,0.45H),5.18(s,0.55H),4.64(s,1H),4.44-4.37(m,1H),4.21-3.81(m,4H),3.63-3.52(m,1H),3.42-3.03(m,8H),3.01-2.53(m,6H),2.22-1.97(m,3H),1.96-1.81(m,3H),1.51(s,9H).
第四步 2-((S)-4-(7-(8-氯萘-1-基)-2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈和2-((S)-4-(7-(8-氯萘-1-基)-2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈混合物8d
称取化合物8c(30mg,粗品)溶于二氯甲烷(0.5mL)中,加入三氟乙酸(0.2mL),室温反应1小时,TLC监测原料反应完全。反应液用饱和碳酸氢钠水溶液调pH至8,二氯甲烷(5mL)萃取,合并有机相,浓缩除去一半左右二氯甲烷,1N稀盐酸(5mL)萃取,合并水相,水相用碳酸钠调pH至9,二氯甲烷(5mL)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液浓缩得到30mg粗品,经Prep-TLC(二氯甲烷/甲醇=10:1)纯化得到类白色固体标题化合物8d(5mg,两步收率11%)。
LC-MS:m/z=575.3[M+H] +
第五步 2-((S)-4-(7-(8-氯萘-1-基)-2-(((2R,7aS)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈和2-((S)-4-(7-(8-氯萘-1-基)-2-(((2S,7aR)-2-氟四氢-1H-吡咯里嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈混合物8
称取化合物8d(18mg,0.031mmol),N,N-二异丙基乙胺(6mg,0.047mmol)和2-氟丙烯酸(4mg,0.044mmol)溶于N,N-二甲基甲酰胺(3mL)中,加入HATU(15mg,0.039mmol),室温反应2小时,TLC监测原料反应完全。反应液加水(6mL)淬灭,乙酸乙酯(10mL)萃取,合并有机相,饱和食盐水洗涤,无水硫酸钠干燥,过滤,滤液浓缩后经Prep-TLC(二氯甲烷/甲醇=20/1)纯化得到类白色固体标题化合物8(4mg,收率18.2%)。
LC-MS:m/z=647.3[M+H] +
1HNMR(400MHz,CDCl 3)δ7.78-7.71(m,1H),7.59(t,J=8.4Hz,1H),7.54-7.48(m,1H),7.48-7.38(m,1H),7.36-7.29(m,1H),7.29-7.26(m,0.55H),7.23-7.19(m,0.45H),6.25-6.16(m,1H),5.43-5.16(m,3H),4.46-4.35(m,1H),4.19-3.78(m,4H),3.63-3.55(m,1H),3.49-3.37(m,1H),3.34-2.81(m,12H),2.80-2.60(m,2H),2.19-2.07(m,2H),2.06-1.83(m,4H).(99.24%purity by HPLC)
实施例9
(S)-2-(4-(7-(8-氯萘-1-基)-3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈9
Figure PCTCN2022073358-appb-000034
第一步 4-溴-3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-1,7-萘啶9a
中间体IN-2(2.70g,17.7mmol),三苯基膦(5.86g,22.2mmol)和化合物5e(2.70g,19.1mmol)溶于无水四氢呋喃(72mL)中,冷却至0℃,氮气保护下加入偶氮二甲酸二异丙酯(6.69g,33.3mmol),加完搅拌0.5小时,TLC检测原料反应完全。反应液加水(50mL)淬灭,乙酸乙酯(50mL)萃取2次,合并有机层,饱和食 盐水(50mL)洗涤,无水硫酸钠干燥,浓缩,粗品硅胶柱层析(二氯甲烷/氨的甲醇溶液=40/1)纯化得淡黄色固体标题化合物9a(1.8g,收率45%)。
LC-MS:m/z=366.1[M+H] +
1H NMR(400MHz,CDCl 3)δ9.17-9.15(m,1H),8.60-8.58(m,1H),7.83-7.77(m,1H),5.00-4.93(m,1H),4.35-4.34(m,2H),3.20-3.12(m,2H),2.70-2.68(m,2H),2.05-1.99(m,2H),1.92-1.87(m,4H),1.73-1.72(m,1H).
第二步 (S)-2-(氰基甲基)-4-(3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-1,7-萘啶-4-基)哌嗪-1-羧酸苄酯9b
化合物9a(200mg,0.55mmol)和化合物1a(244mg,0.83mmol)溶于甲苯(8mL)中,依次加入Pd 2(dba) 3(101mg,0.11mmol),Xantphos(127mg,0.22mmol)和碳酸铯(538mg,1.65mmol),置换氮气三次,升温至100℃反应16小时,TLC监测原料反应完全。反应液冷却至室温,加水(10mL)稀释,乙酸乙酯(20mL)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液浓缩,粗品经硅胶柱层析(二氯甲烷/氨的甲醇溶液=30/1)纯化,Prep-TLC纯化得到白色固体标题化合物9b(40mg,收率13%)。
LC-MS:m/z=545.3[M+H] +
1HNMR(400MHz,CDCl 3)δ9.15(s,1H),8.52(d,J=6.4Hz,1H),7.68(d,J=6.0Hz,1H),7.45-7.33(m,5H),5.27-5.19(m,2H),4.75(br,1H),4.40-4.33(m,2H),4.21(br,1H),3.72-3.64(m,1H),3.49-3.30(m,3H),3.25-3.18(m,3H),3.05-2.94(m,2H),2.74-2.66(m,2H),2.09-1.84(m,7H),1.73-1.70(m,1H).
第三步 (S)-2-(氰基甲基)-4-(3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-1-羧酸苄酯9c
化合物9b(100mg,0.18mmol)溶于醋酸(2mL)中,冷却至0℃,分批加入硼氢化钠(56mg,1.47mmol),加完20℃反应30分钟,TLC监测原料反应完全。反应液加二氯甲烷(15mL)稀释,滴加饱和碳酸钠溶液调节pH=9,分液,有机层饱和食盐水(10mL)洗涤一次,无水硫酸钠干燥,过滤,滤液浓缩得到浅棕色固体标题化合物9c(76mg,收率75%)。
LC-MS:m/z=549.3[M+H] +
第四步 (S)-4-(7-(8-氯萘-1-基)-3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-2-(氰基甲基)哌嗪-1-羧酸苄酯9d
化合物9c(76mg,0.14mmol)和1-溴-8-氯萘(68mg,0.28mmol)溶于甲苯(8mL)中,依次加入Pd 2(dba) 3(13mg,0.014mmol),Xantphos(16mg,0.03mmol)和碳酸铯(91mg,0.28mmol),置换氮气三次,升温至100℃反应16小时,TLC监测原料反应完全。反应液冷却至室温,加水(10mL)稀释,乙酸乙酯(10mL)萃取,合并有机相,无水硫酸钠干燥,过滤,滤液浓缩后经硅胶柱层析(二氯甲烷/氨的甲醇溶液=30/1)纯化得浅棕色固体标题化合物9d(32mg,收率33%)。
LC-MS:m/z=709.3[M+H] +
第五步 (S)-2-(4-(7-(8-氯萘-1-基)-3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)哌嗪-2-基)乙腈9e
化合物9d(32mg,0.05mmol)溶于二氯甲烷(3mL)中,冰盐浴冷至-10℃,滴加三溴化硼的二氯甲烷溶液(0.45mL,0.45mmol,1M),在此温度下反应30分钟,TLC监测原料反应完全。反应液滴加到二氯甲烷和饱和碳酸钠的混合液(v/v=1/1,20mL)中,搅拌5分钟,分液,有机相用饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩得到黄色固体标题化合物9e(24mg,粗品),直接用于下一步。
LC-MS:m/z=575.3[M+H] +
第六步 (S)-2-(4-(7-(8-氯萘-1-基)-3-氟-2-((四氢-1H-吡咯嗪-7a(5H)-基)甲氧基)-5,6,7,8-四氢-1,7-萘啶-4-基)-1-(2-氟丙烯酰基)哌嗪-2-基)乙腈9
化合物9e(24mg,粗品),N,N-二异丙基乙胺(10mg,0.08mmol)和2-氟丙烯酸(4.5mg,0.05mmol)溶于N,N-二甲基甲酰胺(2mL)中,加入HATU(19.0mg,0.05mmol),室温反应30分钟,TLC监测原料反应完全。反应液加水(6mL)淬灭,乙酸乙酯(10mL)萃取,合并有机相,饱和食盐水(10mL)洗涤,无水硫酸钠干燥,过滤,滤液浓缩后经Prep-TLC(二氯甲烷/甲醇=20/1)纯化得到淡黄色固体标题化合物9(6mg,两步收率21%)。
LC-MS:m/z=647.3[M+H] +
1HNMR(400MHz,CDCl 3)δ7.76-7.73(m,1H),7.59(t,J=8.4Hz,1H),7.52-7.50(m,1H),7.48-7.40(m,1H),7.33(t,J=8.4Hz,1H),7.28-7.26(m,0.5H),7.21-7.20(m,0.5H),5.45-5.30(m,1.5H),5.26-2.21(m,1H),4.98(br,0.5H),4.34-4.24(m,3H),3.93-3.80(m,1H),3.70(s,1H),3.66-3.58(m,2H),3.48-2.95(m,10H),2.83-2.67(m,3H),2.17-1.86(m,8H).(96.02%purity by HPLC)
测试例1化合物对MIAPaCa-2细胞增殖抑制的IC 50测定
本发明中使用的人胰腺癌MIAPaCa-2细胞(CRL-1420)购自American Type Culture Collection(ATCC)。细胞在2.5%马血清、10%胎牛血清及1%双抗的DMEM培养基,于37℃,5%CO 2的环境中生长。
化合物对体外培养的MIAPaCa-2细胞增殖抑制作用通过以下方法进行测定:
1)细胞接种:取对数生长期状态良好的胰腺癌MIAPaCa-2细胞重悬于完全培养基,以3000个/孔接种到96孔板中,每孔180μL,在37℃、5%CO 2条件下培养过夜。
2)加药:将需要测试的化合物以完全培养基进行梯度稀释,取20μL稀释的化合物加入到180μL的细胞中,使化合物终浓度为10000、3000、1000、300、100、30、10、3、1nM,同时设相应的溶媒对照。置于37℃、5%CO 2细胞培养箱中培养120小时。
3)检测:以10%三氯乙酸固定细胞,去上清洗涤后,每孔加入100μL 4mg/mL SRB溶液染色15分钟(Sigma,S1402-25g),最后加入加150μL 10mM Tris溶液溶解SRB,TECAN SPARK多功能酶标仪读取OD510值。
4)计算:以下列公式计算细胞生长抑制率:抑制率=(OD值对照孔-OD值给药孔)/OD值对照孔×100%。用Graphpad prism 5.0软件根据化合物浓度与对应的抑制率计算IC 50值。试验结果见表1:
表1本发明中化合物对MIAPaCa-2细胞增殖抑制的IC 50(nM)
化合物编号 MIAPaCa-2 化合物编号 MIAPaCa-2
MRTX849 1.9 1 132.5
2 2640 3 30.7
4 92.4 5 2.7
6 412.7 7 3208
8 12.35 9 377.2
结论:本发明实施例化合物对MIAPaCa-2细胞具有增殖抑制作用,其中化合物5的活性与MRTX849相当。
测试例2化合物对MIAPaCa-2细胞中KRAS下游信号分子ERK1/2磷酸化水平的影响
本发明的化合物对MIAPaCa-2细胞中ERK1/2磷酸化水平的影响通过以下方法检测:
1)细胞接种:取对数生长期状态良好的MIAPaCa-2细胞以1*10 6个/孔接种到六孔板中,在37℃、5%CO2条件下培养过夜。
2)加药:将需要测试的化合物以完全培养基进行梯度稀释后,加入细胞中,使化合物的终浓度为1000、100、10、1nM。置于37℃、5%CO 2细胞培养箱中培养24小时。
3)蛋白样品制备:胰酶消化,收集细胞,500g离心5分钟,弃上清,PBS洗涤3次,以1×SDS凝胶上样缓冲液(50mM Tris-HCl(pH 6.8),100mM DTT,2%SDS,10%甘油,0.1%溴酚蓝)100μL裂解细胞。细胞裂解物在100℃中加热10分钟变性。
4)Western blot:将蛋白样品进行SDS-PAGE电泳,电泳结束后,用湿转***将蛋白转移至PVDF膜,将PVDF膜置于封闭液(5%脱脂奶粉稀释于TBS/T)中室温封闭1小时,然后I,II抗反应;洗膜后,用Immobilon Western HRP Substrate luminal reagent试剂发色,Western Blot成像仪(Tanon,4600)拍照。以下是所用抗体信息:p-ERK1/2(CST:4370);ERK1/2(CST:9102);β-tubulin(CST:2146)。
化合物对MIAPaCa-2细胞中ERK1/2磷酸化水平影响的结果见图1。
结论:本发明实施例中化合物5对MIAPaCa-2细胞ERK1/2的磷酸化具有明显抑制作用,抑制活性呈浓度梯度依赖,活性与MRTX849大致相当。
测试例3化合物的大鼠药代动力学性质
将实施例5的化合物(10mg/kg)给禁食过夜的SD大鼠(雄性,n=3/时间点)口服。给药2h后再给大鼠喂食。分别在给药前以及给药后1.5小时、4小时和8小时采集血样,在4℃离心(4500rpm)10min,获得血清。分别在给药前和给药后1.5小时、4小时和8小时采集肝脏。然后将肝脏样品快速冷冻并储存在-80℃。将100μL MeOH/ACN(1:1,v/v)添加到10μL血清中将混合物沉淀,之后涡旋1min,再离心(11000rpm)5min获得上清液。将20μL上清液溶解于20μL ACN/H2O(1:1,v/v)并用超高效液相色谱仪对其进行分析。肝脏样品中加入10倍重量的MeOH/ACN(1:1,v/v),然后用匀浆机在50Hz条件下匀浆120s后获得匀浆。将匀浆离心(11000rpm)5分钟并收集上清液。然后将20μL上清液重新溶解于20μL ACN/H2O(1:1,v/v)中,并用超高效液相色谱仪对其进行分析。结果见表2:
表2化合物5的大鼠药代性质
Figure PCTCN2022073358-appb-000035
由上表可知,化合物5在大鼠体内较好的血浆暴露量,较好的半衰期。
测试例4化合物在小鼠中的组织分布
将化合物5和MRTX849按照5mg/kg的给药剂量静脉给药。受试小鼠为7-8周龄的ICR雄性小鼠。采样时间点为0.083h,0.5h,1h,2h,采样组织为血浆、脑、肺、结直肠、胰腺、胆囊加胆管。具体操作流程如下:
采血
给药完成后记录时间,并按照设定的时间点收样。
小鼠心脏采血,充分混匀,置于冰上,于30min内离心分离血浆(4℃,8000rpm离心5min),血浆保存于-80℃直至测定。小鼠冰上取脑、肺、结直肠、胰腺、胆囊加胆管,用生理盐水心脏灌流,冲洗,滤纸吸干,最后保存于-80℃直至测定。未给药空白动物二氧化碳安乐死后取血、脑、肺、结直肠、胰腺、胆囊加胆管,作为空白基质。
实验结束
安乐死:实验动物在实验结束后采用吸入过量CO 2的方法安乐死。
留样
准确称取受试物1-2mg。留取100μL配制好的制剂冻存以备剂量复测。
样本分析方法
标准品制备
称量待测化合物制备待测物原液,得到浓度为1mg/mL的待测物原液。原液用乙腈稀释至50000ng/mL作为中间浓度稀释液。采用连续稀释法制备梯度浓度如:5000、1000、750、500、250、100、50、10、5、1ng/mL的校准标准样品。
LC-MS/MS分析血样
测定不同时间点血脑药物浓度,利用药代动力学软件WinNonlin对样品代谢参数进行拟合计算,如药时曲线下面积(AUC)、峰浓度(Cmax)、达峰时间(Tmax)、半衰期(T1/2)等参数。
表3化合物5在小鼠中的组织分布
Figure PCTCN2022073358-appb-000036
表4 MRTX849在小鼠中的组织分布
Figure PCTCN2022073358-appb-000037
通过两组数据的比较,出人意料的发现,化合物5的半衰期都较MRTX849有大幅度的延长,而且其在肺中的药物最大暴露量是MRTX849的近1.4倍,血药浓度-时间曲线下面积(AUC)分别比MRTX849高2.2倍和4.63倍。同时,化合物5在血浆暴露量和血药浓度-时间曲线下面积也比MRTX849有大幅度的提高。
测试例5化合物的体内抗肿瘤活性
在雌性BALB/c小鼠(6-8周)的右侧皮下注射MIAPaCa-2细胞(1.0E+07细胞)以及康宁的基质胶混合物。灌胃给药化合物5和MRTX849,剂量分别为3mg/kg和10mg/kg,连续给药36天。每天监测小鼠并且当肿瘤变得可见时开始卡尺测量。肿瘤的体积通过测量两个垂直直径使用如下公式计算得到:(L*W 2)/2,其中的L和W指的是肿瘤直径的长度和宽度。当肿瘤平均体积到达100mm 3时,将老鼠分组(n=5/组)并且施以化合物。在给药期间(36天),肿瘤体积及老鼠重量每3天测量1次。结果如图2-3所示。
实验结果表明,化合物5在3mg/kg剂量下的抗肿瘤活性明显好于MRTX849,二者在10mg/kg的剂量下表现出相当的抗肿瘤活性。而且,化合物5和MRTX849一样,两个剂量下,对小鼠的体重无明显的影响,表现出较好的安全性。综上可以看出,化合物5在3mg/kg的剂量下,胰腺中的药时曲线下面积(AUC)要好于MRTX849,而且其对胰腺癌的抗肿瘤活性也好于MRTX849。这些数据都表明,化合物5有更好的治疗前景。
申请人声明,本发明通过上述实施例来说明本发明的一类作为KRAS突变体G12C抑制剂的四氢萘啶类衍生物、其制备方法及其应用,但本发明并不局限于上述实施例,即不意味着本发明必须依赖上述实施例才能实施。所属技术领域的技术人员应该明了,对本发明的任何改进,对本发明产品各原料的等效替换及辅助成分的添加、具体方式的选择等,均落在本发明的保护范围和公开范围之内。
以上详细描述了本发明的优选实施方式,但是,本发明并不限于上述实施方式中的具体细节,在本发明的技术构思范围内,可以对本发明的技术方案进行多种简单变型,这些简单变型均属于本发明的保护范围。
另外需要说明的是,在上述具体实施方式中所描述的各个具体技术特征,在不矛盾的情况下,可以通过任何合适的方式进行组合,为了避免不必要的重复,本发明对各种可能的组合方式不再另行说明。

Claims (10)

  1. 一种四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,
    其特征在于,所述四氢萘啶类衍生物的结构如式(I)所示:
    Figure PCTCN2022073358-appb-100001
    其中:
    R 1选自H或F;
    R 2选自6-12元的芳基或5-12元的杂芳基,其中所述的芳基或杂芳基任选地被1-4个R c所取代;
    R c选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基或3-8元的杂环基,其中所述的烷基、烯基、环烷基、R a、R b或杂环基任选地被1-3个R c1所取代;
    R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
    R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
    L 1选自键、O、S、NH或NCH 3
    R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基或杂芳基可以任选地被1-4个R c2所取代;
    R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
    L 2选自C3-C8的环烷基、C5-C10螺环烷基、C4-C10稠环烷基、C5-C10桥环烷基、3-8元的杂环基、5-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基、 5-12元的杂芳基,其中,所述环烷基、螺环烷基、稠环烷基、桥环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基、杂芳基任选地被1-4个R c所取代;
    R 4选自-C(O)R d或-S(O) 2R d
    R d选自C2-C4烯基或C2-C4炔基,其中所述的烯基、炔基可以任选地进一步被1-3个R d1所取代;
    R d1选自卤素、氰基、C1-C3烷基、C3-C7环烷基或4-7元杂环基,其中所述的烷基、环烷基或杂环基任选地被1-3个R c取代;
    所述式(I)中的杂环基、杂芳基中的杂原子为1-4个并选自氧、氮和硫中的一种或多种。
  2. 如权利要求1所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,其特征在于,
    所述四氢萘啶类衍生物的结构如式(Ⅱ)所示:
    Figure PCTCN2022073358-appb-100002
    其中,R 1、R 2、R 3、R c和L 1具有与权利要求1相同的限定范围;n=0-4;
    R e1、R e2或R e3分别独立选自H、卤素、氰基、C1-C3的烷基、C3-C7的环烷基或4-7元的杂环基,而且上述的烷基、环烷基或杂环基任选地被1-3个卤素、C1-C3的烷基、C3-C7的环烷基、4-7元的杂环基、羟基或氨基所取代;
    所述杂环基中的杂原子为1-3个选自氧、氮和硫中的一种或多种;
    和/或,R 2选自6-12元的芳基或5-12元的杂芳基,其中所述的芳基或杂芳基任选地被1-4个R c所取代;其中,R c选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基或3-8元的杂环基,其中所述的烷基、烯基、环烷基、R a、R b或杂环基任选地被1-3个R c1所取代;
    R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
    和/或,R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其 中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、芳基或杂芳基可以任选地被1-4个R c2所取代;
    R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
    R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮或氧;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
  3. 如权利要求1或2所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,其特征在于,所述四氢萘啶类衍生物的结构如式(III)所示:
    Figure PCTCN2022073358-appb-100003
    其中,R 1、R 3和L 1具有与权利要求1相同的限定范围;R e1、R e2、R e3具有与权利要求2相同的限定范围;
    优选地,对于式(III)化合物,其中R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳 基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、6-12元的芳基或5-12元的杂芳基可以任选地被1-4个R c2所取代;
    R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
    R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
    R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮和/或氧;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
  4. 如权利要求1-3中任一项所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,其特征在于,所述四氢萘啶类衍生物的结构如式(IV)所示:
    Figure PCTCN2022073358-appb-100004
    其中,R 3和L 1具有与权利要求1相同的限定范围;R e1、R e2、R e3具有与权利要求2相同的限定范围;
    优选地,对于式(IV)化合物,其中R 3选自C1-C3烷基、C1-C3烯基、C3-C7的环烷基、 3-8元的杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、6-12元的芳基或5-12元的杂芳基,其中,所述烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、6-12元的芳基或5-12元的杂芳基可以任选地被1-4个R c2所取代;
    R c2选自卤素、羟基、C1-C3烷基、C1-C3烯基、-NR aR b、氰基、C1-C3烷氧基、C3-C7环烷基、3-8元杂环基、4-10元的稠杂环基、5-10元的桥杂环基、5-10元的螺杂环基、-C(O)R c1、-C(O)NR aR b、-NR aC(O)R b、6-12元的芳基或5-12元的杂芳基,其中所述的烷基、烯基、环烷基、杂环基、稠杂环基、桥杂环基、螺杂环基、R a、R b、芳基或杂芳基任选地被1-3个R c1所取代;
    R c1选自卤素、羟基、-NR aR b、氰基、C1-C3烷基或C1-C3烷氧基;
    R a和R b分别独立选自H、C1-C3烷基、C3-C7的环烷基或者4-7元的杂环基,或者R a和R b连同它们所附接的原子可以进一步形成4-7元的杂环基;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述R c2为2-4个时,R c2相同或不同;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基含有1-2个杂原子;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为氮和/或氧;
    和/或,所述R 3为任选地被1-4个R c2所取代的4-8元杂环基时,所述杂环基的杂原子为两个时,两个杂原子相同或不同。
  5. 如权利要求1所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体,其特征在于,所述四氢萘啶类衍生物选自如下结构的任意一种:
    Figure PCTCN2022073358-appb-100005
    Figure PCTCN2022073358-appb-100006
  6. 一种如权利要求1-5中任一项所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体的制备方法,其选自如下方法:
    Figure PCTCN2022073358-appb-100007
    第一步,通式(I-A)的化合物和通式(I-B)的化合物反应,得到通式(I-C)的化合物;
    第二步,通式(I-C)的化合物和通式(I-D)的化合物反应,得到通式(I)的化合物;
    其中,R 1、R 2、R 3、R 4和L 1、L 2具有与权利要求1相同的限定范围;X为卤素或OTs,优选为溴。
  7. 一种药物组合物,其特征在于,所述药物组合物包括如权利要求1-5中任一项所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体;优选地,所述药物组合物还包括可药用载体和/或赋形剂。
  8. 如权利要求1-5中任一项所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体或如权利要求7所述的药物组合物在制备用于治疗癌症的药物或在制备作为KRAS突变体G12C抑制剂中的用途。
  9. 根据权利要求8所述的用途,所述的癌症为但不限于星形细胞癌、乳腺癌、***、结直肠癌、子宫内膜癌、食道癌、胃癌、头颈部癌、肝细胞癌、喉癌、胰腺癌、肺癌、口腔癌、卵巢癌、***癌、甲状腺癌、肉瘤、肾癌和胆管癌;优选为肺癌、胰腺癌或结直肠癌。
  10. 一种用于治疗和/或预防癌症的方法,其包括向人类给予治疗有效量的如权利要求1-5中任一项所述的四氢萘啶类衍生物、其药学上可接受的盐、其互变异构体或其立体异构体或如权利要求7所述的药物组合物。
PCT/CN2022/073358 2021-02-09 2022-01-24 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用 WO2022170947A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN202280007411.6A CN116600808A (zh) 2021-02-09 2022-01-24 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN202110178535 2021-02-09
CN202110178535.8 2021-02-09

Publications (1)

Publication Number Publication Date
WO2022170947A1 true WO2022170947A1 (zh) 2022-08-18

Family

ID=82837432

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2022/073358 WO2022170947A1 (zh) 2021-02-09 2022-01-24 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用

Country Status (2)

Country Link
CN (1) CN116600808A (zh)
WO (1) WO2022170947A1 (zh)

Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109843856A (zh) * 2016-05-18 2019-06-04 米拉蒂治疗股份有限公司 Kras g12c抑制剂
WO2020239077A1 (zh) * 2019-05-29 2020-12-03 上海翰森生物医药科技有限公司 含氮杂环类衍生物调节剂、其制备方法和应用
CN112142735A (zh) * 2020-04-09 2020-12-29 上海凌达生物医药有限公司 一类稠和氰基吡啶类化合物、制备方法和用途
CN112300153A (zh) * 2019-07-26 2021-02-02 博瑞生物医药(苏州)股份有限公司 一种杂环化合物、药物组合物和用途
WO2021219072A1 (zh) * 2020-04-30 2021-11-04 上海科州药物研发有限公司 作为kras抑制剂的杂环化合物的制备及其应用方法

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
KR20160076519A (ko) * 2013-10-10 2016-06-30 아락세스 파마 엘엘씨 Kras g12c 억제제
US11059819B2 (en) * 2017-01-26 2021-07-13 Janssen Biotech, Inc. Fused hetero-hetero bicyclic compounds and methods of use thereof

Patent Citations (5)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN109843856A (zh) * 2016-05-18 2019-06-04 米拉蒂治疗股份有限公司 Kras g12c抑制剂
WO2020239077A1 (zh) * 2019-05-29 2020-12-03 上海翰森生物医药科技有限公司 含氮杂环类衍生物调节剂、其制备方法和应用
CN112300153A (zh) * 2019-07-26 2021-02-02 博瑞生物医药(苏州)股份有限公司 一种杂环化合物、药物组合物和用途
CN112142735A (zh) * 2020-04-09 2020-12-29 上海凌达生物医药有限公司 一类稠和氰基吡啶类化合物、制备方法和用途
WO2021219072A1 (zh) * 2020-04-30 2021-11-04 上海科州药物研发有限公司 作为kras抑制剂的杂环化合物的制备及其应用方法

Also Published As

Publication number Publication date
CN116600808A (zh) 2023-08-15

Similar Documents

Publication Publication Date Title
US11884661B2 (en) 3-substituted propionic acids as αV integrin inhibitors
TWI616436B (zh) 作為magl抑制劑的胺基甲酸1,1,1-三氟-3-羥基丙烷-2-基酯衍生物及胺基甲酸1,1,1-三氟-4-羥基丁烷-2-基酯衍生物
TWI593683B (zh) 雙環稠合之雜芳基或芳基化合物
KR102506327B1 (ko) αV 인테그린 길항제로서의 인다졸 유도체
US9890168B2 (en) 2,4-disubstituted 7H-pyrrolo[2,3-d]pyrimidine derivative, preparation method and medicinal use thereof
WO2015158310A1 (zh) 一种酪氨酸激酶抑制剂及其用途
WO2015127872A1 (zh) 2,4-二取代苯-1,5-二胺衍生物及其应用以及由其制备的药物组合物和药用组合物
TWI669300B (zh) 嘧啶類衍生物、其製備方法、其藥物組合物以及其在醫藥上的用途
WO2021143701A1 (zh) 嘧啶-4(3h)-酮类杂环化合物、其制备方法及其在医药学上的应用
EP3707142B1 (en) Pyrrolopyrazine derivatives as alpha v integrin inhibitors
TWI596095B (zh) Dihydropyridazine-3,5-dione derivatives
WO2022206723A1 (zh) 杂环类衍生物、其制备方法及其医药上的用途
WO2022170952A1 (zh) 一种作为sos1抑制剂的多环哒嗪酮类衍生物、其制备方法及用途
TW201422616A (zh) 作爲Syk抑制劑的取代吡啶並吡嗪類化合物
WO2021208918A1 (zh) 作为egfr抑制剂的三环化合物
WO2023025116A1 (zh) 杂环类衍生物、其制备方法及其医药上的用途
WO2020108500A1 (zh) 一类组蛋白乙酰化酶p300抑制剂及其用途
CN115515949A (zh) 新型氨基嘧啶类egfr抑制剂
WO2022170917A1 (zh) 一种作为sos1抑制剂的多环嘧啶类衍生物、其制备方法及用途
WO2022170802A1 (zh) 一种作为sos1抑制剂的嘧啶并吡啶酮类衍生物、其制备方法及用途
WO2019062657A1 (zh) 氮杂环类衍生物、其制备方法及其医药用途
CN112771032B (zh) 作为***egfr抑制剂的嘧啶吡唑类化合物
CN110582495B (zh) 作为tnf活性的调节剂的稠合五环咪唑衍生物
WO2022170947A1 (zh) 一类作为kras突变体g12c抑制剂的四氢萘啶类衍生物、其制备方法及其应用
WO2023274396A1 (zh) 苯并氮杂环类化合物及其在药物中的应用

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: 22752109

Country of ref document: EP

Kind code of ref document: A1

WWE Wipo information: entry into national phase

Ref document number: 202280007411.6

Country of ref document: CN

NENP Non-entry into the national phase

Ref country code: DE