[go: up one dir, main page]

CN118344386A - Substituted bridged ring inhibitors and preparation method and application thereof - Google Patents

Substituted bridged ring inhibitors and preparation method and application thereof Download PDF

Info

Publication number
CN118344386A
CN118344386A CN202310059893.6A CN202310059893A CN118344386A CN 118344386 A CN118344386 A CN 118344386A CN 202310059893 A CN202310059893 A CN 202310059893A CN 118344386 A CN118344386 A CN 118344386A
Authority
CN
China
Prior art keywords
alkyl
group
cycloalkyl
substitution
substituted
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202310059893.6A
Other languages
Chinese (zh)
Inventor
吕彬华
崔大为
张青
刘连军
柴传柯
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Zejing Pharmaceutical Zhejiang Co ltd
Suzhou Zelgen Biopharmaceutical Co Ltd
Shanghai Zelgen Pharmatech Co Ltd
Original Assignee
Zejing Pharmaceutical Zhejiang Co ltd
Suzhou Zelgen Biopharmaceutical Co Ltd
Shanghai Zelgen Pharmatech Co Ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Zejing Pharmaceutical Zhejiang Co ltd, Suzhou Zelgen Biopharmaceutical Co Ltd, Shanghai Zelgen Pharmatech Co Ltd filed Critical Zejing Pharmaceutical Zhejiang Co ltd
Priority to CN202310059893.6A priority Critical patent/CN118344386A/en
Priority to PCT/CN2023/082908 priority patent/WO2023179629A1/en
Priority to EP23773876.0A priority patent/EP4497751A1/en
Priority to CN202380029714.2A priority patent/CN119317628A/en
Priority to PCT/CN2024/072124 priority patent/WO2024149389A1/en
Publication of CN118344386A publication Critical patent/CN118344386A/en
Pending legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D519/00Heterocyclic compounds containing more than one system of two or more relevant hetero rings condensed among themselves or condensed with a common carbocyclic ring system not provided for in groups C07D453/00 or C07D455/00
    • 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
    • A61K31/4995Pyrazines or piperazines forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic 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
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • 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
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53861,4-Oxazines, e.g. morpholine spiro-condensed or forming part of bridged ring systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • A61K31/551Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole having two nitrogen atoms, e.g. dilazep
    • 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
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/08Bridged systems

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Epidemiology (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Acyclic And Carbocyclic Compounds In Medicinal Compositions (AREA)

Abstract

The invention relates to a substituted bridged ring inhibitor, a preparation method and application thereof. Specifically, the compound has a structure shown in a formula (A0), and the invention also discloses a preparation method of the compound and application of the compound as a KRAS G12D inhibitor, which has good selective inhibition effect on KRAS G12D, better pharmacodynamics, pharmacokinetics performance and lower toxic and side effects.

Description

Substituted bridged ring inhibitors and preparation method and application thereof
Technical Field
The invention belongs to the field of medicines, and particularly relates to a substituted bridged ring inhibitor, and a preparation method and application thereof.
Background
About one quarter of all human tumors are caused by RAS mutations. In the RAS family, KRAS mutations account for 85% of all RAS mutations. KRAS mutations are found in nearly 90% of pancreatic cancers, 30-40% of colon cancers, and 15-20% of lung cancers (mainly non-small cell lung cancers). The G12C and G12D mutations occur at the most predominant of KRAS mutations, with the G12C mutation occurring predominantly in HSCLC and the G12D mutation occurring predominantly in pancreatic cancer. Up to now, no drugs against KRAS G12D mutation have been approved for the market.
Current conventional treatment regimens for pancreatic cancer in clinic include gemcitabine monotherapy, gemcitabine in combination with albumin paclitaxel, the FOLFIRINOX regimen (oxaliplatin + irinotecan + 5-FU/LV), and the like. Wherein liposomal irinotecan is suitable for use in combination with fluorouracil and folinic acid in treating patients with advanced pancreatic cancer who are poorly treated with gemcitabine chemotherapy (second line therapy). However, in general, the current treatments for pancreatic cancer are limited and the overall survival time of patients is not more than 1 year. Although drug discovery for patients with advanced pancreatic cancer continues to be ongoing, research progress has been slow until now.
Since KRAS G12D target proteins are pathologically associated with a variety of diseases, particularly pancreatic cancer, there is a current need for novel KRAS G12D inhibitors for clinical treatment. High selectivity and high activity KRAS G12D inhibitors can be used for more effective treatment of diseases such as cancers caused by KRAS G12D mutation and reducing the potential of off-target effect, thus having more urgent clinical demands.
Disclosure of Invention
The invention aims to provide a novel compound with selective inhibition effect on KRAS G12D and/or better pharmacodynamic performance and application thereof.
In a first aspect of the invention, there is provided a compound of formula (A0), a stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof:
Ring C is selected from the group consisting of:
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
A group selected from the group consisting of: wherein X is selected from: n, CH, CD, CF, C (CN);
R 1 is selected from: -L 1-Q-L2-L3;
wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 0, 1,2,3, 4, 5 or 6; with the proviso that when W is monocyclic or bicyclic, n is not 0;
R 10 is selected from the group consisting of substituted or unsubstituted: c 6-C14 aryl, 5-14 membered heteroaryl; wherein the substitution refers to substitution with one or more Ra;
R 11 are each independently selected from the group consisting of substituted or unsubstituted: H. deuterium, halogen, cyano, ester, amine, amide, sulfone, ureido, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C6 alkyloxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
Ra are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C20 cycloalkyl, 4-20 membered heterocycle, C 3-C20 cycloalkyloxy, 4-20 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
m is an integer of 0, 1,2,3,4, 5 or 6;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, 4-20 membered heterocyclyl C (O), C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
In another preferred embodiment, the compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof has a structure according to formula (a):
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
A group selected from the group consisting of: wherein X is selected from: n, CH, CD, CF, C (CN);
R 1 is selected from: -L 1-Q-L2-L3;
wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 0, 1,2,3, 4, 5 or 6; with the proviso that when W is monocyclic or bicyclic, n is not 0;
R 10 is selected from the group consisting of substituted or unsubstituted: c 6-C14 aryl, 5-14 membered heteroaryl; wherein the substitution refers to substitution with one or more Ra;
R 11 are each independently selected from the group consisting of substituted or unsubstituted: H. deuterium, halogen, cyano, ester, amine, amide, sulfone, ureido, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C6 alkyloxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy;
Ra are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C20 cycloalkyl, 4-20 membered heterocyclyl, C 3-C20 cycloalkyloxy, 4-20 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
m is an integer of 0, 1,2,3,4, 5 or 6;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, 4-20 membered heterocyclyl C (O), C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
In another preferred embodiment, ra are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, OH, SONH 2、NHSO2CH3、C1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C20 cycloalkyl, 4-20 membered heterocyclyl; wherein the substitution refers to substitution with one or more R; r is as defined above.
In another preferred embodiment, the compound has the structure shown as A
Preferably, having the structure shown as A'
Wherein n, Y, Z, W, L 1、L2、L3、Q、R10、R11, m and ring A are as defined above.
In a further preferred embodiment of the present invention,A group selected from the group consisting of:
in another preferred embodiment, the compound has the structure of formula A' "
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
X, Y, Z, W, R 1, n and R 10 are as defined above.
In another preferred embodiment, R 3 is selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C6 cycloalkyl, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy; wherein, the substitution refers to substitution by one or more R.
In another preferred embodiment, R 10 is selected from: selected from the group consisting of substituted or unsubstituted: phenyl, naphthyl, 5-6 membered monocyclic heteroaryl (e.g. pyridinyl), 9-10 membered bicyclic heteroaryl (e.g. indazolyl, benzothiazole, benzothiophene, benzofuran), wherein said substitution is by one or more groups selected from the group consisting of: halogen, hydroxy, cyano, NH 2、C1-C6 alkyl, halo C 1-C6 alkyl, C 1-C6 alkoxy, halo C 1-C6 alkoxy, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 2-C6 alkenyl, C 2-C6 alkynyl: preferably, R 10 is selected from:
In another preferred embodiment, R 8 is selected from: NH 2、OH、SONH2、NHSO2CH3.
In another preferred embodiment, R 10 is
In another preferred embodiment, U is selected from: n, CH, CD, CF;
R 4 is selected from the group consisting of substituted or unsubstituted: halogen, C 1-C6 alkyl, C 2-C6 alkenyl, C 2-C6 alkynyl;
R 5、R6、R7、R9 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 8 is selected from: NH 2、OH、SONH2、NHSO2CH3.
In another preferred embodiment, R 10 is
In another preferred embodiment, U' is selected from: o, or S;
U' is selected from: n, or C (CN);
R 7'、R8'、R9' are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 5' is selected from: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy.
In another preferred embodiment, R 10 isWherein V 1、V2、V3、V4 and V 5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -C≡CH, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (= O).
In another preferred embodiment, R 10 isWherein R v1、Rv2、Rv3、Rv4 and R v5 are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl (e.g. CF 3、CF2CF3), Halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl), (NH 2)-C3-C6 cycloalkyl, halogen, CN, -C.ident.CH, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl, SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl) H, C 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 Cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -C.ident.CH, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), optionally substituted with one or more oxo (=o).
In another preferred embodiment, there is provided a compound of formula (IA) or (IB), a stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof:
in the method, in the process of the invention,
U is selected from: n, CH, CD, CF;
U' is selected from: o, or S;
U' is selected from: n, or C (CN);
X is selected from: n, CH, CD, CF, C (CN);
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
R 1 is selected from: -L 1-Q-L2-L3; wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 1, 2, 3, 4, 5 or 6;
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 4 is selected from the group consisting of substituted or unsubstituted: halogen, C 1-C6 alkyl, C 2-C6 alkenyl, C 2-C6 alkynyl; wherein the substitution refers to substitution with one or more R;
R 8 is selected from: OH, SONH 2、NHSO2CH3;
R 5、R6、R7、R9、R7'、R8'、R9 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 5' is selected from: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
In another preferred embodiment, each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, C 3-C20 cycloalkyl, a 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfone group or a urea group.
In another preferred embodiment, each R, which may be the same or different, is independently selected from: deuterium, C 1-C6 alkyl, deuterated C 1-C6 alkyl, halogenated C 1-C6 alkyl, (C 3-C10 cycloalkyl) C 1-C6 alkyl, (4-10 membered heterocyclyl) C 1-C6 alkyl, (C 1-C6 alkoxy) C 1-C6 alkyl, (C 3-C6 cycloalkyloxy) C 1-C6 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C10 cycloalkyl) ethynyl, and, (4-10 membered heterocyclyl) ethynyl, C 1-C6 alkoxy, deuterated C 1-C6 alkoxy, halogenated C 1-C6 alkoxy, C 3-C10 cycloalkyl, A 4-10 membered heterocyclic group, a C 6-C10 aryl group, a 5-10 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
In another preferred embodiment, the compound of formula I has a structure of formula (I 'A) or formula (I' B)
Preferably, the compound has a structure represented by the formula (I 'A) or the formula (I' B)
Wherein ,n、U、U'、U"、X、Y、Z、W、L1、L2、L3、Q、R2、R3、R4、R5、R6、R7、R8、R9、R5'、R7'、R8'、R9'、 is defined as described above.
In another preferred embodiment, the compound has a structure represented by formula (IIA) or formula (IIB):
wherein ,R1、R2、R3、R4、R5、R6、R7、R8、R9、R5'、R7'、R8'、R9'、U、U'、U"、X、Z、W and n are as defined above.
In another preferred embodiment, the compound has a structure of formula (IIIA) or formula (IIIB):
Wherein ,R1、R2、R3、R4、R5、R6、R7、R8、R9、R5'、R7'、R8'、R9'、U、U'、U"、Z、W and n are as defined above.
In another preferred embodiment, R 3 is selected from: H. d, F, cl, br, CN, methyl, ethyl, propyl, isopropyl, deuterated methyl, CH 2F、CHF2、CF3, methoxy, ethoxy, propoxy, OCH 2F、OCHF2、OCF3.
In another preferred embodiment, the compound has a structure of formula (IVA) or formula (IVB):
wherein ,R1、R4、R5、R6、R7、R8、R9、R5'、R7'、R8'、R9'、U、U'、U"、Z、W and n are as defined above.
In another preferred embodiment, the compound has a structure of formula (VA) or formula (VB):
Wherein ,R1、R4、R5、R6、R7、R9、R7'、R8'、R9'、U、U'、U"、Z、W and n are as defined above.
In another preferred embodiment, the compound has a structure of formula (VIA) or formula (VIB):
wherein ,R1、R4、R5、R6、R7、R9、R7'、R8'、R9'、U"、Z、W and n are as defined above.
In another preferred embodiment, the compound has a structure of formula (VIIA) or formula (VIIB):
Wherein R 1、R4、R5、R7'、R8'、R9', Z, W and n are as defined above.
In another preferred embodiment, the compound has a structure of formula (VIIIA) or formula (VIIIB):
Wherein R 1、R4、R5、R7'、R9', Z, W and n are as defined above.
In another preferred embodiment, W is a substituted or unsubstituted group of: c 3-C6 monocyclic cycloalkyl, C 7-C10 bicyclic or C 7-C11 tricyclic cycloalkyl, 4-6 membered saturated or unsaturated monocyclic heterocyclyl, 7-10 membered bicyclic or 7-11 membered tricyclic heterocyclyl; preferably, W is selected from: substituted or unsubstituted 7-10 membered saturated or unsaturated bridged heterocyclic group, substituted or unsubstituted 7-10 membered saturated or unsaturated condensed ring heterocyclic group.
In another preferred embodiment, W is selected from:
Wherein n' is an integer of 0,1, 2, 3,4, 5, or 6; r is as defined above, and R may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
In another preferred embodiment, when W is selected from:
When n' may be 1 or 2, more preferably n is 1, R is as defined above, and R may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
In another preferred embodiment, when W is selected from:
when n' may be 0,1 or 2, R is as defined above, and R may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
In another preferred embodiment, when W is selected from:
when n is 0, 1 or 2, R is as defined above, and R may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
In another preferred embodiment, R is selected from: deuterium, C 1-C6 alkyl, deuterated C 1-C6 alkyl, halogenated C 1-C6 alkyl, (C 3-C6 cycloalkyl) C 1-C18 alkyl, (4-6 membered heterocyclyl) C 1-C6 alkyl, (C 1-C6 alkoxy) C 1-C6 alkyl, (C 3-C6 cycloalkyloxy) C 1-C6 alkyl, (4-6 membered heterocyclyloxy) C 1-C6 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C6 cycloalkyl) ethynyl, and, (4-6 membered heterocyclyl) ethynyl, C 1-C6 alkoxy, deuterated C 1-C6 alkoxy, halogenated C 1-C6 alkoxy, 4-6 membered heterocyclyl C (O), C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 6-C10 aryl, 5-10 membered heteroaryl (preferably 5-6 membered), halogen, nitro, hydroxy, oxo, cyano, COOC 1-C6 alkyl, NH 2、NHC1-C6 alkyl, N (C 1-C6 alkyl) 2、CONH2、CONHC1-C6 alkyl, CON (C 1-C6 alkyl) 2、SO2C1-C6 alkyl, -NHCONH 2、-NHCO NHC1-C6 alkyl, -NHCON (C 1-C6 alkyl) 2.
In another preferred embodiment, Z is a substituted or unsubstituted C 1-C3 alkylene group, preferably methylene, ethylene, propylene.
In another preferred embodiment, Z is CD 2.
In another preferred embodiment, the compound has the structure of formula (IX):
Wherein n' is an integer of 0, 1, 2, 3, 4, 5, or 6;
R 5、R、L1、Q、L2、L3 and n are as defined above.
In another preferred embodiment, R 4 is selected from the group consisting of substituted or unsubstituted: halogen, methyl, ethyl, propyl, ethenyl, propenyl, allyl, butenyl, ethynyl, propynyl, butynyl.
In a further preferred embodiment of the present invention,Selected from:
in another preferred embodiment, the compound has a structure represented by formula (X):
V 1、V2、V3、V4 and V 5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
The definition of rings C, R, X, Z, W and n is described above.
In another preferred embodiment, the compound has the structure of formula (XI):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
Wherein R, R 1, X, Z, W and n are as defined above.
In another preferred embodiment, the compound has the structure of formula (XII):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 3 is selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
Wherein R, R 1, X, Z, W and n are as defined above.
In another preferred embodiment, the compound has the structure of formula (XIII):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 3 is selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
Wherein R, R 1, Z, W and n are as defined above.
In another preferred embodiment, each R v is independently selected from: H. halogen, CN, -C.ident.CH, or NH 2.
In another preferred embodiment, L 1 is selected from: a substituted or unsubstituted methylene group or a substituted or unsubstituted ethylene group; q is selected from: o; l 2 is selected from: an unsubstituted, or substituted or unsubstituted methylene group; l 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -a radical of formula (I) -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); Wherein said substitution means substitution with one or more groups selected from the group consisting of: deuterium, C 1-C6 alkyl, deuterated C 1-C6 alkyl, halogenated C 1-C6 alkyl, vinyl, ethynyl, C 1-C6 alkoxy, deuterated C 1-C6 alkoxy, halogenated C 1-C6 alkoxy, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 6-C14 aryl, A 5-to 14-membered heteroaryl group, halogen, nitro, hydroxy, oxo, cyano, ester, amino, amido, sulfone or urea group.
In another preferred embodiment, -L 1-Q-L2-L3 is selected from:
In a further preferred embodiment of the present invention, Is thatPreferably is
In a further preferred embodiment of the present invention,Is thatPreferably is
In a further preferred embodiment of the present invention,Selected from:
Or is selected from:
Wherein n' is an integer of 0,1, 2, 3, 4, 5 or 6; r, R 1 and n are as defined above, R, R 1 may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
Or is selected from:
Wherein n' is an integer of 0, 1, 2,3, 4, 5 or 6; r is as defined above, and R may be substituted on any one of the rings of the polycyclic ring (e.g., bridged or spiro ring).
In a further preferred embodiment of the present invention,Selected from: :
In another preferred embodiment, the prodrug of the compound has a structure represented by the following formula (XIV):
c' is selected from:
PG is selected from:
Y, Z, W, ring A, R 1、R10、R11, m and n are as defined above.
In another preferred embodiment, m, A ring 、n、U、X、Y、Z、W、L1、L2、L3、Q、R2、R3、R4、R5、R6、R7、R8、R9、R10 and R 11 have groups corresponding to each of the specific compounds in the examples.
In another preferred embodiment, V 1、V2、V3、V4、V5 and ring C, PG have groups corresponding to each of the specific compounds of the examples.
In another preferred embodiment, the compound is selected from the group consisting of:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
In another preferred embodiment, the compound is preferably the compound prepared in the examples.
In a second aspect of the present invention, there is provided a process for the preparation of a compound of formula (I), a stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, wherein the process comprises the steps of:
(i) Reacting a compound of formula V-1 with a compound of formula V-2 in an inert solvent, in the presence or absence of a Pd catalyst, and in the presence or absence of a condensing agent, to obtain a compound of formula V-3;
(ii) Reacting a compound of formula V-3 with a compound of formula V-4 in an inert solvent, in the presence of a base, with or without a Pd catalyst, to obtain a compound of formula V-5;
(iii) Reacting a compound of formula V-5 with a compound of formula V-6 in the presence of a Pd catalyst in an inert solvent in the presence of a base to obtain a compound of formula V-7;
(iv) Removing the protecting group PG1 of the compound of the formula V-7 under the action of acid (such as TFA, HCl and the like) or Pd catalytic hydrogenation condition to obtain a compound of the formula (I);
in the method, in the process of the invention,
X 1、X2 and X 3 are each independently selected from: OH, halogen, OTf, OTs or OMs;
PG1 is selected from: boc, cbz, or Bn;
LG1 is selected from: -B (OH) 2、-B(KBF3)、-Sn(nBu)3,
R1、R2、R3、R4、R5、R6、R7、R8、R9、U、X、Y、Z、W And n is as defined above.
In a third aspect of the invention, there is provided a pharmaceutical composition comprising one or more compounds of the first aspect, stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates, or prodrugs thereof; and a pharmaceutically acceptable carrier.
In another preferred embodiment, the pharmaceutical composition further comprises a drug selected from the group consisting of: PD-1 inhibitors (e.g., nivolumab、pembrolizumab、pidilizumab、cemiplimab、JS-001、SHR-120、BGB-A317、IBI-308、GLS-010、GB-226、STW204、HX008、HLX10,BAT 1306、AK105、LZM 009 or a biosimilar of the above, etc.), PD-L1 inhibitors (e.g., durvalumab、atezolizumab、avelumab、CS1001、KN035、HLX20、SHR-1316、BGB-A333、JS003、CS1003、KL-A167、F520、GR1405、MSB2311 or a biosimilar of the above, etc.), CD20 antibodies (e.g., rituximab、obinutuzumab、ofatumumab、veltuzumab、tositumomab、131I-tositumomab、ibritumomab、90Y-ibritumomab、90In-ibritumomab、ibritumomab tiuxetan, etc.), CD47 antibodies (e.g., hu5F9-G4, CC-90002, TTI-621, TTI-622, OSE-172, SRF-231, ALX-148, NI-1701, SHR-1603, IBI188, IMM 01), ALK inhibitors (e.g., ceritinib, alectinib, brigatinib, lorlatinib, oxatinib), PI3K inhibitors (e.g., idelalisib, duvelisib, dactolisib, taselisib, bimiralisib, omipalisib, buparlisib, etc.), BTK inhibitors (e.g., ibrutinib, tirabrutinib, acalabrutinib, zanubrutinib, vecabrutinib, etc.), EGFR inhibitors (e.g., Afatinib、Gefitinib、Erlotinib、Lapatinib、Dacomitinib、Icotinib、Canertinib、Sapitinib、Naquotinib、Pyrotinib、Rociletinib、Osimertinib, etc.), VEGFR inhibitors (e.g., sorafenib, pazopanib, regorafenib, sitravatinib, ningetinib, cabozantinib, sunitinib, duonafinil, etc.), HDAC inhibitors (e.g., Givinostat、Tucidinostat、Vorinostat、Fimepinostat、Droxinostat、Entinostat、Dacinostat、Quisinostat、Tacedinaline, etc.), CDK inhibitors (e.g., palbociclib, ribociclib, abemaciclib, milciclib, trilaciclib, lerociclib, etc.), MEK inhibitors (e.g., selumetinib (AZD 6244), trametinib (GSK 1120212), PD 032501, U0126, pimasertib (AS-703026), PD 1843352 (CI-etc.), mP inhibitors (e.g., mP-Vistusertib), P-46155, etc.), or combinations thereof.
In a fourth aspect, the present invention provides the use of a compound according to the first aspect, a stereoisomer, a tautomer, a crystal, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, or a pharmaceutical composition according to the third aspect, for the preparation of a medicament for the prophylaxis and/or treatment of a disease associated with the activity or expression of KRAS G12D.
In another preferred embodiment, the disease is a tumor or a disorder.
In another preferred embodiment, the disease is selected from the group consisting of: lung cancer, breast cancer, prostate cancer, esophageal cancer, colorectal cancer, bone cancer, kidney cancer, stomach cancer, liver cancer, colorectal cancer, melanoma, lymphoma, leukemia, brain tumor, myeloma, soft tissue sarcoma, pancreatic cancer, and skin cancer.
In a fourth aspect of the invention, there is provided a method of non-diagnostically, non-therapeutically inhibiting KRAS G12D comprising the steps of: administering to a subject in need thereof an effective amount of a compound as described in the first aspect, a stereoisomer, tautomer, crystal, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, or a pharmaceutical composition as described above.
In another preferred embodiment, the subject is a mammal, preferably a human.
In a fifth aspect of the invention, there is provided a method of inhibiting KRAS G12D activity in vitro comprising the steps of: contacting a compound according to the first aspect, a stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, or a pharmaceutical composition as described above, with a protein or a cell, thereby inhibiting the activity of KRAS G12D.
In another preferred embodiment, the cells are selected from the group consisting of: macrophages, intestinal cells (including intestinal stem cells, intestinal epithelial cells), or combinations thereof.
In another preferred embodiment, the cells are from a rodent (e.g., mouse, rat), or a primate (e.g., human).
It is understood that within the scope of the present invention, the above-described technical features of the present invention and technical features specifically described below (e.g., in the examples) may be combined with each other to constitute new or preferred technical solutions. And are limited to a space, and are not described in detail herein.
Detailed Description
The present inventors have conducted long and intensive studies to unexpectedly prepare a novel class of compounds having selective inhibitory effect and/or better pharmacodynamic properties on KRAS G12D. On this basis, the inventors completed the present invention.
Terminology
In the present invention, unless otherwise indicated, terms used have the ordinary meanings known to those skilled in the art.
The term "alkyl" refers to straight or branched chain alkanyl radicals containing 1 to 20 carbon atoms, such as 1 to 18 carbon atoms, especially 1 to 18 carbon atoms, preferably 1 to 10 carbon atoms (C1-C10), more preferably 1 to 6 carbon atoms (C1-C6). Typical "alkyl" groups include methyl, ethyl, propyl, isopropyl, n-butyl, t-butyl, isobutyl, and,Pentyl, isopentyl, heptyl, 4-dimethylpentyl, octyl, 2, 4-trimethylpentyl, nonyl, decyl, undecyl, dodecyl and the like. In the present invention, alkyl groups also include substituted alkyl groups. "substituted alkyl" means that one or more positions in the alkyl group are substituted, especially 1 to 4 substituents, and may be substituted at any position. Typical substitutions include, but are not limited to, one or more of the following groups: such as hydrogen, deuterium, halogen (e.g., a single halogen substituent or a multiple halogen substituent, the latter such as trifluoromethyl or an alkyl group containing Cl 3), cyano, nitro, oxygen (e.g., =o), trifluoromethyl, trifluoromethoxy, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, aromatic ring 、ORa、SRa、S(=O)Re、S(=O)2Re、P(=O)2Re、S(=O)2ORe,P(=O)2ORe、NRbRc、NRbS(=O)2Re、NRbP(=O)2Re、S(=O)2NRbRc、P(=O)2NRbRc、C(=O)ORd、C(=O)Ra、C(=O)NRbRc、OC(=O)Ra、OC(=O)NRbRc、NRbC(=O)ORe,NRdC(=O)NRbRc、NRdS(=O)2NRbRc、NRdP(=O)2NRbRc、NRbC(=O)Ra、, or NR bP(=O)2Re, where R a appearing herein may independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, 5-14 membered heterocycle, or C6-C14 aromatic ring, R b、Rc and R d may independently represent hydrogen, deuterium, C1-C6 alkyl, C3-C8 cycloalkyl, 5-14 membered heterocycle, or C6-C14 aromatic ring, or R b and R c together with the N atom may form a heterocycle; r e can independently represent hydrogen, C1-C6 alkyl, C3-C8 cycloalkyl, C2-C6 alkenyl, C3-C6 cycloalkenyl, C2-C6 alkynyl, a 5-to 14-membered heterocycle, or a C6-C14-aromatic ring. Typical substituents described above, such as alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, or aromatic ring, may be optionally substituted.
The term "alkylene" refers to a group formed by the removal of one more hydrogen atom from an "alkyl group or substituted alkyl group" such as methylene, ethylene, propylene, isopropylene (e.g) Butylene (e.g) Pentylene (e.g) Hexyl (e.g) Heptyl (e.g)、Etc. In addition, the term also includes the replacement of a methylene group of an alkylene group (e.g., a C1-C18 alkylene group) with a cycloalkylene group (e.g., a C3-C20 cycloalkylene group), such as "C1-C18 alkylene C3-C20 cycloalkylene" or "C3-C20 cycloalkylene C1-C18 alkylene".
The term "C1-C18 alkylene-C3-C20 cycloalkylene" or "C3-C20 cycloalkylene-C1-C18 alkylene" has the same meaning and refers to groups formed by the removal of two hydrogen atoms from a cycloalkylalkyl or alkylcycloalkyl group, e.g. Etc.
In the present invention, the term "alkenyl" means a straight or branched hydrocarbon group containing one or more double bonds and typically having a length of 2 to 20 carbon atoms. Alkenyl is preferably C2-C6 alkenyl, more preferably C2-C4 alkenyl. Alkenyl groups include, but are not limited to, for example, ethenyl, propenyl, butenyl, 1-methyl-2-buten-1-yl, and the like. In the present invention, alkenyl includes substituted alkenyl.
The term "alkynyl" denotes a straight or branched hydrocarbon radical containing one or more triple bonds and typically ranging in length from 2 to 20 carbon atoms. Alkynyl is preferably C2-C6 alkynyl, more preferably C2-C4 alkynyl. Alkynyl groups include, but are not limited to, ethynyl, propynyl, or the like. In the present invention, alkynyl also includes substituted alkynyl groups, and substituents may be halo, hydroxy, cyano, nitro, and the like.
In the present invention, the term "cycloalkyl" refers to a fully saturated cyclic hydrocarbon compound group comprising 1 to 4 rings, each ring containing 3 to 8 carbon atoms. The term "C 3-C20" refers to a cyclic alkyl group containing 3,4, 5, 6,7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 carbon atoms. Cycloalkyl is preferably C 3-C14 cycloalkyl, more preferably C 3-C10 cycloalkyl, more preferably C 3-C6 monocyclic cycloalkyl, C 7-C10 bicyclic or tricyclic cycloalkyl. "substituted cycloalkyl" means that one or more positions in the cycloalkyl group are substituted, especially 1-4 substituents, and can be substituted at any position. In the present invention, "cycloalkyl" includes substituted cycloalkyl groups, typical substitutions including, but not limited to, one or more of the following groups: such as hydrogen, deuterium, halogen (e.g., a single halogen substituent or a multiple halogen substituent, the latter such as trifluoromethyl or an alkyl group containing Cl 3), cyano, nitro, oxygen (e.g., =o), trifluoromethyl, trifluoromethoxy, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, aromatic ring 、ORa、SRa、S(=O)Re、S(=O)2Re、P(=O)2Re、S(=O)2ORe、P(=O)2ORe、NRbRc、NRbS(=O)2Re、NRbP(=O)2Re、S(=O)2NRbRc、P(=O)2NRbRc、C(=O)ORd、C(=O)Ra、C(=O)NRbRc、OC(=O)Ra、OC(=O)NRbRc、NRbC(=O)ORe、NRdC(=O)NRbRc、NRdS(=O)2NRbRc、NRdP(=O)2NRbRc、NRbC(=O)Ra、 or NR bP(=O)2Re, Wherein R a as herein presented may independently represent hydrogen, deuterium, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle or aromatic ring, R b、Rc and R d may independently represent hydrogen, deuterium, alkyl, cycloalkyl, heterocycle or aromatic ring, or R b and R c together with the N atom may form a heterocycle; r e can independently represent hydrogen, deuterium, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, or aromatic ring. Typical substituents described above may be optionally substituted. Typical substitutions also include spiro, bridged or fused ring substituents, especially spiro-cycloalkyl, spiro-alkenyl, spiro-heterocycle (excluding heteroaryl), bridged-cycloalkyl, bridged-cycloalkenyl, bridged-heterocycle (excluding heteroaryl), fused-ring alkyl, fused-ring alkenyl, fused-ring heterocyclyl or fused-ring aryl groups, which cycloalkyl, cycloalkenyl, heterocyclyl and heteroaryl groups may be optionally substituted. Examples of cycloalkyl groups include, but are not limited to: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, adamantyl, and the like.
The term "C3-C20 cycloalkylene" refers to a group formed by the removal of two hydrogen atoms from a cycloalkyl group, such as: Etc.
In the present invention, the term "heterocyclyl" refers to a fully saturated or partially unsaturated cyclic group (including, but not limited to, e.g., 3-7 membered monocyclic, 4-7 membered monocyclic, 6-11 membered bicyclic, or 8-16 membered tricyclic or polycyclic ring systems) wherein at least one heteroatom is present in a ring having at least one carbon atom. The term "4-20 membered heterocyclyl" refers to heterocyclyl containing 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19 or 20 ring atoms. "heterocyclyl" has the same meaning as "saturated or unsaturated heterocyclyl". The "heterocyclyl" is preferably a 4-14 membered heterocyclyl (including but not limited to, e.g., 4-6 membered monocyclic, 7-10 membered bicyclic or 8-14 membered tricyclic or polycyclic systems), more preferably a 4-12 membered heterocyclyl, more preferably a 4-10 membered heterocyclyl, such as a 4-6 membered monocyclic heterocyclyl, 7-10 membered bicyclic or tricyclic heterocyclyl, more preferably a 4-8 membered heterocyclyl, more preferably a 4-6 membered heterocyclyl. Each heteroatom-containing heterocycle may bear 1,2,3 or 4 heteroatoms selected from nitrogen atoms, oxygen atoms or sulfur atoms, where the nitrogen or sulfur atoms may be oxidized and the nitrogen atoms may also be quaternized. The heterocyclic group may be attached to any heteroatom or carbon atom residue of a ring or ring system molecule, preferably to an N or C atom of a ring or ring system molecule. Typical monocyclic heterocycles include, but are not limited to, azetidinyl, pyrrolidinyl, oxetanyl, pyrazolinyl, imidazolinyl, imidazolidinyl, oxazolidinyl, isoxazolidinyl, thiazolidinyl, isothiazolidinyl, tetrahydrofuranyl, piperidinyl, piperazinyl, 2-oxopiperazinyl, 2-oxopiperidinyl, 2-oxopyrrolidinyl, hexahydroazepinyl, 4-piperidonyl, tetrahydropyranyl, morpholinyl, thiomorpholinyl sulfoxide, thiomorpholinyl sulfone, 1, 3-dioxanyl, and tetrahydro-1, 1-dioxythiophene, and the like. Polycyclic heterocyclyl groups include spiro, fused and bridged heterocyclic groups; wherein the heterocyclic groups of the spiro ring, the condensed ring and the bridged ring are optionally connected with other groups through single bonds, or are further connected with other cycloalkyl groups, heterocyclic groups, aryl groups and heteroaryl groups through any two or more atoms on the ring in a parallel ring manner; the heterocyclic group may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, deuteroalkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, alkylthio, alkylamino, halogen, amino, nitro, hydroxy, mercapto, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylthio, oxo, carboxyl, and carboxylate.
The term "C4-C20 heterocyclylene" refers to a group formed by the removal of two hydrogen atoms from a heterocyclic group, such as: Etc.
In the present invention, the term "aryl" refers to aromatic cyclic hydrocarbon groups having 1 to 5 rings, especially monocyclic and bicyclic groups. Wherein "C 6-C14 aryl" refers to an aromatic cyclic hydrocarbon compound group containing 6, 7, 8, 9, 10, 11, 12, 13 or 14 ring carbon atoms. Aryl includes phenyl, biphenyl, or naphthyl. The aromatic ring of the aryl group may be linked by a single bond (e.g., biphenyl), or condensed (e.g., naphthalene, anthracene, etc.), where the aromatic ring contains two or more aromatic rings (bicyclic, etc.). "substituted aryl" means that one or more positions in the aryl group are substituted, especially 1 to 3 substituents, and can be substituted at any position. Typical substitutions include, but are not limited to, one or more of the following groups: such as hydrogen, deuterium, halogen (e.g., a single halogen substituent or a multiple halogen substituent, the latter such as trifluoromethyl or an alkyl group comprising Cl 3), cyano, nitro, oxo (e.g., =o), trifluoromethyl, trifluoromethoxy, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, aromatic ring 、ORa、SRa、S(=O)Re、S(=O)2Re、P(=O)2Re、S(=O)2ORe,P(=O)2ORe、NRbRc、NRbS(=O)2Re、NRbP(=O)2Re、S(=O)2NRbRc、P(=O)2NRbRc、C(=O)ORd、C(=O)Ra、C(=O)NRbRc、OC(=O)Ra、OC(=O)NRbRc、NRbC(=O)ORe,NRdC(=O)NRbRc、NRdS(=O)2NRbRc、NRdP(=O)2NRbRc、NRbC(=O)Ra、, or NR bP(=O)2Re, wherein R a appearing herein may independently represent hydrogen, deuterium, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, or aromatic ring, R b、Rc and R d may independently represent hydrogen, deuterium, alkyl, cycloalkyl, heterocycle, or aromatic ring, or R b and R c together with the N atom may form a heterocycle; r e can independently represent hydrogen, deuterium, alkyl, cycloalkyl, alkenyl, cycloalkenyl, alkynyl, heterocycle, or aromatic ring. Typical substituents described above may be optionally substituted. Typical substitutions also include fused ring substituents, especially fused ring alkyl, fused ring alkenyl, fused ring heterocyclyl or fused ring aryl groups, which cycloalkyl, cycloalkenyl, heterocyclyl and heteroaryl groups may be optionally substituted.
The term "heteroaryl" refers to an aromatic cyclic hydrocarbon group containing 1 to 4 heteroatoms selected from oxygen, nitrogen and sulfur. Wherein "5-14 membered heteroaryl" refers to a heteroaromatic system containing 1-4 heteroatoms, 5-14 ring atoms. Heteroaryl is preferably a 5-to 10-membered ring, more preferably 5-or 6-membered ring, such as pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, thiadiazolyl, isothiazolyl, furanyl, pyridyl, pyrazinyl, pyrimidinyl, pyridazinyl, triazolyl, tetrazolyl, and the like. "heteroaryl" may be substituted or unsubstituted, and when substituted, the substituent is preferably one or more groups independently selected from alkyl, deuteroalkyl, haloalkyl, alkoxy, haloalkoxy, alkenyl, alkynyl, alkylthio, alkylamino, halogen, amino, nitro, hydroxy, mercapto, cyano, cycloalkyl, heterocyclyl, aryl, heteroaryl, cycloalkylthio, oxo, carboxyl, and carboxylate.
In the present invention, the term "alkoxy" refers to straight or branched chain alkoxy groups including alkyl-O-, alkyl-O-alkyl groups, wherein "C 1-C18 alkoxy" refers to straight or branched chain alkoxy groups having 1 to 18 carbon atoms including C 1-C18 alkyl-O-, -C 1-C6 alkyl-O-C 1-C6 alkyl groups, including without limitation methoxy, ethoxy, propoxy, isopropoxy, butoxy and the like. Preferably C 1-C8 alkoxy, more preferably C1-C 6 alkoxy.
In the present invention, the term "cycloalkyloxy" refers to a cycloalkyl-O-, wherein "C 3-C20 cycloalkyloxy" refers to a C 3-C20 cycloalkyl-O-, wherein C 3-C20 cycloalkyl is as defined above.
In the present invention, the term "heterocyclyloxy" means a heterocyclic group-O-, in which "4-20 membered heterocyclyloxy" means a 4-20 membered heterocyclic group-O-, in which 4-20 membered heterocyclic group is defined as above.
In the present invention, the term "C 1-C18 alkyleneoxy" refers to a group obtained by removing one hydrogen atom from "C 1-C18 alkyleneoxy".
In the present invention, the term "halogen" or "halo" refers to chlorine, bromine, fluorine, iodine.
In the present invention, the term "halo" refers to substitution with halogen.
In the present invention, the term "deuterated" refers to substitution with deuterium.
In the present invention, the term "hydroxyl group" means a group having the structure OH.
In the present invention, the term "nitro" refers to a group with the structure NO 2.
In the present invention, the term "cyano" refers to a group with the structure CN.
In the present invention, the term "ester group" refers to a group having the structure-COOR, wherein R represents hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle. The ester group is preferably a-COO C 1-C6 alkyl group.
The term "amine group" refers to a group having the structure-NRR ', wherein R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R and R' may be the same or different in the dialkylamine fragment. The amine group is preferably NH 2、NH C1-C6 alkyl, N (C 1-C6 alkyl) 2.
The term "amide" refers to a group having the structure-CONRR ' or-NRCOR ', wherein R and R ' may each independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. The amide group is preferably CONH 2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl).
The term "sulfonamide" refers to a group having the structure-S 2 ONRR ' or-NRSO 2 R ', where R and R ' may each independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. Sulfonamide groups are preferably SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl).
The term "sulfone" refers to a group with the structure-SO 2 R, wherein R may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above.
The term "ureido" refers to a group having the structure-NRCONR 'R "wherein R, R' and R" may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R, R' and R "may be the same or different in the dialkylamine fragment.
The term "alkylaminoalkyl" refers to a group with the structure-RNHR ', wherein R and R' may independently represent hydrogen, alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R and R' may be the same or different.
The term "dialkylaminoalkyl" refers to a group with the structure-RNHR 'R ", wherein R, R' and R" may independently represent alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl, heterocycle or substituted heterocycle, as defined above. R, R' and R "may be the same or different in the dialkylamine fragment.
The term "heterocyclylalkyl" refers to a group bearing the structure-RR', wherein R may independently represent alkyl or substituted alkyl, cycloalkyl or substituted cycloalkyl, cycloalkenyl or substituted cycloalkenyl, aryl or substituted aryl; r' represents a heterocycle or a substituted heterocycle.
In the present invention, the term "substituted" means that one or more hydrogen atoms on a particular group are replaced with a particular substituent. The specific substituents are those described in the foregoing for each of the examples or are those found in each of the examples. Unless otherwise specified, a substituted group may have a substituent selected from a specific group at any substitutable site of the group, which may be the same or different at each position. Those skilled in the art will appreciate that combinations of substituents contemplated by the present invention are those that are stable or chemically achievable. Such as (but not limited to): halogen, hydroxy, cyano, carboxyl (-COOH), C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, C3-C8 cycloalkyl, 3-to 12-membered heterocyclyl, aryl, heteroaryl, C1-C8 aldehyde, C2-C10 acyl, C2-C10 ester, amine, C1-C6 alkoxy, C1-C10 sulfonyl, C1-C6 ureido, and the like.
Unless otherwise indicated, it is assumed that any heteroatom in an underfilling state has sufficient hydrogen atoms to complement its valence.
When the substituent is a non-terminal substituent, it is a subunit of the corresponding group, e.g., alkyl corresponds to alkylene, cycloalkyl corresponds to cycloalkylene, heterocyclyl corresponds to heterocyclylene, alkoxy corresponds to alkyleneoxy, and the like.
In the present invention, a plurality means 2, 3, 4, 5.
Active ingredient
As used herein, "compounds of the invention" refers to compounds of formula I, and also includes stereoisomers or optical isomers, pharmaceutically acceptable salts, prodrugs or solvates of the compounds of formula I.
Salts which may be formed with the compounds of the present invention are also within the scope of the present invention. Unless otherwise indicated, the compounds of the present invention are understood to include salts thereof. The term "salt" as used herein refers to salts formed with inorganic or organic acids and bases in the acid or base form. Furthermore, when the compound of the present invention contains a basic moiety, it includes, but is not limited to, pyridine or imidazole, and an acidic moiety, including, but not limited to, carboxylic acids, the possible formation of zwitterions ("inner salts") are included within the term "salts". Pharmaceutically acceptable (i.e., non-toxic, physiologically acceptable) salts are preferred, although other salts are also useful, for example, in isolation or purification steps during the preparation process. The compounds of the invention may form salts, for example, by reacting compound I with an amount of, for example, an equivalent of, an acid or base, salting out in a medium, or lyophilizing in aqueous solution.
The compounds of the present invention contain basic fragments, including but not limited to amine or pyridine or imidazole rings, which may form salts with organic or inorganic acids. Typical acids that may be salified include acetates (e.g., with acetic acid or trihaloacetic acid, such as trifluoroacetic acid), adipates, alginates, ascorbates, aspartate, benzoate, benzenesulfonate, bisulfate, borate, butyrate, citrate, camphorate, camphorsulfonate, cyclopentanepropionate, diglycolate, dodecyl sulfate, ethane sulfonate, fumarate, glucoheptonate, glycerophosphate, hemisulfate, heptanoate, caproate, hydrochloride, hydrobromide, hydroiodide, hydroxyethanesulfonate (e.g., 2-hydroxyethanesulfonate), lactate, maleate, mesylate, naphthalene sulfonate (e.g., 2-naphthalene sulfonate), nicotinate, nitrate, oxalate, pectate, persulfate, phenylpropionate (e.g., 3-phenylpropionate), phosphate, picrate, pivalate, propionate, salicylate, succinate, sulfate (e.g., formed with sulfuric acid), sulfonate, tartrate, thiocyanate, toluene sulfonate such as p-toluenesulfonate, dodecanoate, and the like.
Certain compounds of the present invention may contain acidic moieties, including but not limited to carboxylic acids, that may form salts with various organic or inorganic bases. Typical base-forming salts include ammonium salts, alkali metal salts such as sodium, lithium, potassium salts, alkaline earth metal salts such as calcium, magnesium salts, and salts with organic bases (e.g., organic amines), such as benzathine, dicyclohexylamine, sea-bamine (salts with N, N-bis (dehydroabietyl) ethylenediamine), N-methyl-D-glucamine, N-methyl-D-glucamide, t-butylamine, and salts with amino acids such as arginine, lysine, and the like. Basic nitrogen-containing groups can be combined with halide quaternary ammonium salts, such as small molecule alkyl halides (e.g., methyl, ethyl, propyl and butyl chlorides, bromides and iodides), dialkyl sulfates (e.g., dimethyl, diethyl, dibutyl and dipentyl sulfates), long chain halides (e.g., decyl, dodecyl, tetradecyl and tetradecyl chlorides, bromides and iodides), aralkyl halides (e.g., benzyl and phenyl bromides), and the like.
Prodrugs and solvates of the compounds of the invention are also within the scope of coverage. The term "prodrug" as used herein refers to a compound that undergoes chemical conversion by metabolic or chemical processes to produce a compound, salt, or solvate of the invention when used in the treatment of a related disorder. The compounds of the present invention include solvates, such as hydrates.
The compounds, salts or solvates of the present invention, may exist in tautomeric forms (e.g., amides and imine ethers). All of these tautomers are part of the present invention.
Stereoisomers of all compounds (e.g., those having asymmetric carbon atoms which may be present as a result of various substitutions), including enantiomeric and diastereoisomeric forms thereof, are contemplated as falling within the scope of the present invention. The individual stereoisomers of the compounds of the invention may not be present simultaneously with the other isomers (e.g., having particular activity as one pure or substantially pure optical isomer), or may be mixtures, such as racemates, or mixtures with all or a portion of the other stereoisomers. The chiral center of the present invention has two configurations, S or R, defined by the International Association of theory and application chemistry (IUPAC) 1974. The racemic forms can be resolved by physical methods, such as fractional crystallization, or by separation of crystals by derivatization into diastereomers, or by chiral column chromatography. Individual optical isomers may be obtained from the racemates by suitable methods, including but not limited to conventional methods, such as salt formation with an optically active acid followed by recrystallization.
The compounds of the present invention are prepared, isolated and purified in sequence to give the compounds in an amount of 90% by weight or more, for example 95% or more and 99% or more ("very pure" compounds), as listed in the text description. Such "very pure" compounds of the invention are also included herein as part of the invention.
All configurational isomers of the compounds of the present invention are within the scope of coverage, whether in mixtures, pure or very pure form. The definition of compounds in the present invention includes both the cis (Z) and the trans (E) olefin isomers, as well as the cis and trans isomers of carbocycles and heterocycles.
Throughout the specification, groups and substituents may be selected to provide stable fragments and compounds.
Specific functional groups and chemical term definitions are described in detail below. For the purposes of the present invention, chemical elements are in accordance with those defined in Periodic Table of THE ELEMENTS, CAS version, handbook of CHEMISTRY AND PHYSICS,75 th Ed.. The definition of specific functional groups is also described herein. Furthermore, the basic principles of organic chemistry and specific functional groups and reactivities are described in "Organic Chemistry", thomas Sorrell, university Science Books, sausalato 1999, which is incorporated by reference in its entirety.
Certain compounds of the invention may exist in specific geometric or stereoisomeric forms. The present invention encompasses all compounds, including cis and trans isomers, R and S enantiomers, diastereomers, (D) isomers, (L) isomers, racemic mixtures, and other mixtures thereof. In addition, an asymmetric carbon atom may represent a substituent such as an alkyl group. All isomers and mixtures thereof are encompassed by the present invention.
According to the invention, the mixture of isomers may contain various isomer ratios. For example, in a mixture of only two isomers, there may be a combination of: all ratios of 50:50, 60:40, 70:30, 80:20, 90:10, 95:5, 96:4, 97:3, 98:2, 99:1, or 100:0 isomers are within the scope of the invention. Similar ratios, as well as ratios for more complex mixtures of isomers, are within the scope of the present invention, as would be readily understood by one of ordinary skill in the art.
The present invention also includes isotopically-labeled compounds, equivalent to those disclosed herein as original compounds. In practice it will often occur that one or more atoms are replaced by an atom of a different atomic weight or mass number than it is. Examples of isotopes that can be listed as compounds of the invention include hydrogen, carbon, nitrogen, oxygen, phosphorus, sulfur, fluorine and chlorine isotopes, such as 2H、3H、13C、11C、14C、15N、18O、17O、31P、32P、35S、18F and 36 Cl, respectively. The compounds of the present invention, or enantiomers, diastereomers, isomers, or pharmaceutically acceptable salts or solvates thereof, wherein isotopes or other isotopic atoms containing such compounds are within the scope of the present invention. Certain isotopically-labeled compounds of the present invention, for example, the radioisotopes of 3 H and 14 C, are also useful in drug and substrate tissue distribution experiments. Tritium, 3 H and carbon-14, 14 C, are relatively easy to prepare and detect. Is the first choice in isotopes. In addition, heavier isotopic substitutions such as deuterium, 2 H, may be preferred in certain situations because of their good metabolic stability which may be advantageous in certain therapies, such as increasing half-life or reducing dosage in vivo. Isotopically-labeled compounds can be prepared by conventional methods by using readily available isotopically-labeled reagents in place of non-isotopically-labeled reagents using the protocols disclosed in the examples.
If one is to design the synthesis of a particular enantiomer of a compound of the invention, it may be prepared by asymmetric synthesis or by derivatization with chiral auxiliary, separating the resulting diastereomeric mixture and removing the chiral auxiliary to give the pure enantiomer. Alternatively, if the molecule contains a basic functional group, such as an amino acid, or an acidic functional group, such as a carboxyl group, diastereomeric salts can be formed therewith using an appropriate optically active acid or base, and then the resulting mixture can be separated by conventional means such as fractional crystallization or chromatography to give the pure enantiomer.
As described herein, the compounds of the present invention may be substituted with any number of substituents or functional groups to extend their inclusion. In general, the term "substituted", whether appearing before or after the term "optional", in the formulas of the present invention includes substituents, means that the specified structural substituent is substituted for the hydrogen radical. When multiple of a particular structure are substituted at a position with multiple particular substituents, the substituents may be the same or different at each position. The term "substitution" as used herein includes all permissible organic compound substitutions. In a broad sense, permissible substituents include acyclic, cyclic, branched, unbranched, carbocyclic and heterocyclic, aromatic and non-aromatic organic compounds. In the present invention, the heteroatom nitrogen may have a hydrogen substituent or any of the permissible organic compounds described hereinabove to supplement the valence state thereof. Furthermore, the present invention is not intended to be limited in any way to allow substitution of organic compounds. The present invention contemplates that the combination of substituents and variable groups is useful in the treatment of diseases, such as infectious or proliferative diseases, in the form of stable compounds. The term "stable" as used herein refers to a compound that is stable for a period of time sufficient to maintain structural integrity of the compound, preferably for a period of time sufficient to be effective, as used herein for the purposes described above.
Metabolites of the compounds and pharmaceutically acceptable salts thereof of the present application, as well as prodrugs that can be converted in vivo to structures of the compounds and pharmaceutically acceptable salts thereof of the present application are also encompassed by the claims of the present application. In the present application, the term "prodrug" refers to a compound which is inactive or less active in vitro, but releases an active drug to exert its efficacy in vivo through the conversion of an enzyme (e.g., hydrolase, oxidase, etc.) or non-enzyme (e.g., pH, light, radiation, etc.) after the chemical structure modification of the drug to introduce an easy-leaving group. Such as (but not limited to): carboxylic acid esters, phosphoric acid esters, carbamates, carbonic acid esters, and the like. Easy leaving group structures such as (but not limited to):
Preparation method
The following more specifically describes the preparation method of the compounds of the structures of the formulae (I-A) and (I-B) of the present invention, but these specific methods do not limit the present invention in any way. The compounds of the present invention may also be conveniently prepared by optionally combining the various synthetic methods described in this specification or known in the art, such combinations being readily apparent to those skilled in the art to which the present invention pertains.
Typically, the compounds of the present invention are prepared by the following process wherein the starting materials and reagents used, unless otherwise specified, are commercially available.
Preferably, the compounds of the present invention are prepared by the following method
(I) Reacting a compound of formula V-1 with a compound of formula V-2 in an inert solvent, in the presence or absence of a Pd catalyst, and in the presence or absence of a condensing agent, to obtain a compound of formula V-3;
(ii) Reacting a compound of formula V-3 with a compound of formula V-4 in an inert solvent, in the presence of a base, with or without a Pd catalyst, to obtain a compound of formula V-5;
(iii) Reacting a compound of formula V-5 with a compound of formula V-6 in the presence of a Pd catalyst in an inert solvent in the presence of a base to obtain a compound of formula V-7;
(iv) Removing the protecting group PG1 of the compound of the formula V-7 under the action of acid (such as TFA, HCl and the like) or Pd catalytic hydrogenation condition to obtain a compound of the formula (I);
in the method, in the process of the invention,
X 1、X2 and X 3 are each independently selected from: OH, halogen, OTf, OTs or OMs;
PG1 is selected from: boc, cbz, or Bn;
LG1 is selected from: -B (OH) 2、-B(KBF3)、-Sn(nBu)3,
R1、R2、R3、R4、R5、R6、R7、R8、R9、U、X、Y、Z、W And n is as defined above.
Pharmaceutical compositions and methods of administration
The pharmaceutical composition provided by the invention is used for preventing and/or treating the following diseases: inflammation, cancer, cardiovascular disease, infection, immune disease, metabolic disease.
The compounds of formula (I) may be used in combination with other drugs known to treat or ameliorate similar conditions. When administered in combination, the mode of administration and dosage of the original drug may remain unchanged, while the compound of formula I is administered simultaneously or subsequently. When the compound of formula I is administered simultaneously with one or more other drugs, it may be preferable to use a pharmaceutical composition containing one or more known drugs together with the compound of formula I. Drug combinations also include administration of the compound of formula I with one or more other known drugs over overlapping time periods. When a compound of formula I is administered in combination with one or more other drugs, the dosage of the compound of formula I or the known drug may be lower than the dosage of the compound of formula I alone.
Drugs or active ingredients that may be used in combination with the compounds of formula (I) include, but are not limited to: PD-1 inhibitors (e.g., nivolumab、pembrolizumab、pidilizumab、cemiplimab、JS-001、SHR-120、BGB-A317、IBI-308、GLS-010、GB-226、STW204、HX008、HLX10,BAT 1306、AK105、LZM 009 or a biosimilar of the above), PD-L1 inhibitors (e.g., durvalumab、atezolizumab、avelumab、CS1001、KN035、HLX20、SHR-1316、BGB-A333、JS003、CS1003、KL-A167、F 520、GR1405、MSB2311 or a biosimilar of the above), CD20 antibodies (e.g., rituximab、obinutuzumab、ofatumumab、veltuzumab、tositumomab、131I-tositumomab、ibritumomab、90Y-ibritumomab、90In-ibritumomab、ibritumomab tiuxetan, etc.), and the like, CD47 antibodies (e.g., hu5F9-G4, CC-90002, TTI-621, TTI-622, OSE-172, SRF-231, ALX-148, NI-1701, SHR-1603, IBI188, IMM 01), ALK inhibitors (e.g., ceritinib, alectinib, brigatinib, lorlatinib, okatinib), PI3K inhibitors (e.g., idelalisib, duvelisib, dactolisib, taselisib), Bimiralisib, omipalisib, buparlisib, etc.), BTK inhibitors (e.g., ibrutinib, tirabrutinib, acalabrutinib, zanubrutinib, vecabrutinib, etc.), EGFR inhibitors (e.g., Afatinib、Gefitinib、Erlotinib、Lapatinib、Dacomitinib、Icotinib、Canertinib、Sapitinib、Naquotinib、Pyrotinib、Rociletinib、Osimertinib, etc.), VEGFR inhibitors (e.g., sorafenib, pazopanib, regorafenib, Sitravatinib, ningetinib, cabozantinib, sunitinib, dorafinib, etc.), HDAC inhibitors (e.g., Givinostat、Tucidinostat、Vorinostat、Fimepinostat、Droxinostat、Entinostat、Dacinostat、Quisinostat、Tacedinaline, etc.), CDK inhibitors (e.g., palbociclib, ribociclib, abemaciclib, milciclib, trilaciclib, lerociclib, etc.), MEK inhibitors (e.g., selumetinib (AZD 6244), MEK inhibitors (e.g., selumetinib), Trametinib (GSK 1120212), PD0325901, U0126, pimasertib (AS-703026), PD184352 (CI-1040), etc.), mTOR inhibitors (e.g., vistusertib, etc.), SHP2 inhibitors (e.g., RMC-4630, JAB-3068, TNO155, etc.), or combinations thereof. Dosage forms of the pharmaceutical composition of the present invention include (but are not limited to): injection, tablet, capsule, aerosol, suppository, pellicle, dripping pill, external liniment, controlled release or sustained release preparation, or nanometer preparation.
The pharmaceutical compositions of the present invention comprise a safe and effective amount of a compound of the present invention or a pharmaceutically acceptable salt thereof, and a pharmaceutically acceptable excipient or carrier. Wherein "safe and effective amount" means: the amount of the compound is sufficient to significantly improve the condition without causing serious side effects. Typically, the pharmaceutical compositions contain 1-2000mg of the compound of the invention per dose, more preferably 10-1000mg of the compound of the invention per dose. Preferably, the "one dose" is a capsule or tablet.
"Pharmaceutically acceptable carrier" means: one or more compatible solid or liquid filler or gel materials which are suitable for human use and must be of sufficient purity and sufficiently low toxicity. "compatible" as used herein means that the components of the composition are capable of blending with and between the compounds of the present invention without significantly reducing the efficacy of the compounds. Examples of pharmaceutically acceptable carrier moieties are cellulose and its derivatives (e.g., sodium carboxymethylcellulose, sodium ethylcellulose, cellulose acetate, and the like), gelatin, talc, solid lubricants (e.g., stearic acid, magnesium stearate), calcium sulfate, vegetable oils (e.g., soybean oil, sesame oil, peanut oil, olive oil, and the like), polyols (e.g., propylene glycol, glycerol, mannitol, sorbitol, and the like), emulsifiers (e.g.) Wetting agents (such as sodium lauryl sulfate), coloring agents, flavoring agents, stabilizing agents, antioxidants, preservatives, pyrogen-free water and the like.
The mode of administration of the compounds or pharmaceutical compositions of the present invention is not particularly limited, and representative modes of administration include (but are not limited to): oral, intratumoral, rectal, parenteral (intravenous, intramuscular or subcutaneous), and topical administration.
Solid dosage forms for oral administration include capsules, tablets, pills, powders and granules. In these solid dosage forms, the active compound is admixed with at least one conventional inert excipient (or carrier), such as sodium citrate or dicalcium phosphate, or with the following ingredients: (a) Fillers or compatibilizers, for example, starch, lactose, sucrose, glucose, mannitol and silicic acid; (b) Binders, for example, hydroxymethyl cellulose, alginate, gelatin, polyvinylpyrrolidone, sucrose and acacia; (c) humectants, e.g., glycerin; (d) Disintegrants, for example, agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain complex silicates, and sodium carbonate; (e) a slow solvent, such as paraffin; (f) an absorption accelerator, e.g., a quaternary amine compound; (g) Wetting agents, such as cetyl alcohol and glycerol monostearate; (h) an adsorbent, for example, kaolin; and (i) a lubricant, for example, talc, calcium stearate, magnesium stearate, solid polyethylene glycol, sodium lauryl sulfate, or mixtures thereof. In capsules, tablets and pills, the dosage forms may also comprise buffering agents.
Solid dosage forms such as tablets, dragees, capsules, pills and granules can be prepared with coatings and shells, such as enteric coatings and other materials well known in the art. They may contain opacifying agents and the release of the active compound or compounds in such compositions may be released in a delayed manner in a certain part of the digestive tract. Examples of embedding components that can be used are polymeric substances and waxes. The active compound may also be in the form of microcapsules with one or more of the above excipients, if desired.
Liquid dosage forms for oral administration include pharmaceutically acceptable emulsions, solutions, suspensions, syrups or tinctures. In addition to the active compound, the liquid dosage forms may contain inert diluents commonly used in the art such as, for example, water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, propylene glycol, 1, 3-butylene glycol, dimethylformamide and oils, in particular, cottonseed, groundnut, corn germ, olive, castor and sesame oils or mixtures of these substances and the like.
In addition to these inert diluents, the compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, and perfuming agents.
Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum methoxide and agar-agar or mixtures of these substances, and the like.
Compositions for parenteral injection may comprise physiologically acceptable sterile aqueous or anhydrous solutions, dispersions, suspensions or emulsions, and sterile powders for reconstitution into sterile injectable solutions or dispersions. Suitable aqueous and nonaqueous carriers, diluents, solvents or excipients include water, ethanol, polyols and suitable mixtures thereof.
Dosage forms of the compounds of the present invention for topical administration include ointments, powders, patches, sprays and inhalants. The active ingredient is mixed under sterile conditions with a physiologically acceptable carrier and any preservatives, buffers, or propellants which may be required if necessary.
The methods of treatment of the present invention may be administered alone or in combination with other therapeutic means or therapeutic agents.
When a pharmaceutical composition is used, a safe and effective amount of the compound of the present invention is applied to a mammal (e.g., a human) in need of treatment, wherein the dose at the time of administration is a pharmaceutically effective dose, and the daily dose is usually 1 to 2000mg, preferably 50 to 1000mg, for a human having a body weight of 60 kg. Of course, the particular dosage should also take into account factors such as the route of administration, the health of the patient, etc., which are within the skill of the skilled practitioner.
The invention also provides a preparation method of the pharmaceutical composition, which comprises the following steps: mixing a pharmaceutically acceptable carrier with the compounds of general formula (I-A) and formula (I-B) or crystalline forms, pharmaceutically acceptable salts, hydrates or solvates thereof, thereby forming a pharmaceutical composition.
The invention also provides a treatment method, which comprises the following steps: administering to a subject in need thereof a compound of formula (I) as described herein, or a crystalline form, pharmaceutically acceptable salt, hydrate or solvate thereof, or a pharmaceutical composition as described herein, for selectively inhibiting KRAS G12D.
Compared with the prior art, the invention has the following main advantages:
(1) The compound has good selective inhibition effect on KRAS G12D;
(2) The compound has better pharmacodynamics, pharmacokinetics and lower toxic and side effects.
The invention will be further illustrated with reference to specific examples. It is to be understood that these examples are illustrative of the present invention and are not intended to limit the scope of the present invention. The experimental procedure, which does not address the specific conditions in the examples below, is generally followed by routine conditions such as Sambrook et al, molecular cloning: conditions described in the laboratory Manual (New York: cold Spring Harbor Laboratory Press, 1989) or as recommended by the manufacturer. Percentages and parts are by weight unless otherwise indicated.
Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art. In addition, any methods and materials similar or equivalent to those described herein can be used in the methods of the present invention. The preferred methods and materials described herein are presented for illustrative purposes only.
The structure of the compounds of the present invention is determined by Nuclear Magnetic Resonance (NMR) and liquid chromatography-mass spectrometry (LC-MS).
NMR was performed using a Bruker AVANCE-400 nuclear magnetic resonance apparatus, and the measurement solvents included deuterated dimethyl sulfoxide (DMSO-d 6), deuterated acetone (CD 3COCD3), deuterated chloroform (CDCl 3), deuterated methanol (CD 3 OD), etc., with the internal standard being Tetramethylsilane (TMS), and the chemical shifts being measured in parts per million (ppm).
Liquid chromatography (LC-MS) was performed using WATERS SQD2 mass spectrometers. HPLC was determined using an Agilent1100 high pressure chromatograph (Microsorb 5micron C18 100x 3.0mm column).
The thin layer chromatography silica gel plate is Qingdao GF254 silica gel plate, TLC is 0.15-0.20mm, and the preparation thin layer chromatography is 0.4-0.5 mm. Column chromatography generally uses Qingdao silica gel 200-300 mesh silica gel as carrier.
The starting materials in the examples of the present invention are known and commercially available, or may be synthesized using or according to literature reported in the art.
Except for the special descriptions, all reactions of the invention are carried out by continuous magnetic stirring under the protection of dry inert gas (such as nitrogen or argon), and the reaction temperature is in degrees centigrade.
Examples
Preparation of intermediate 1-1 (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
The first step: preparation of 1-benzyl 2-methyl 2- (but-3-en-1-yl) pyrroline-1, 2-dicarboxylic acid ester
A solution of 1-benzyl-2-methyl (S) -pyrroline-1, 2-dicarboxylic acid ester (50.0 g,190mmol,1.00 eq) in THF (100 mL) at-70℃under nitrogen was added dropwise to LiHMDS (1.00M, 284 mL,1.50 eq). The resulting reaction solution was reacted at-70℃for 2 hours, followed by dropwise addition of 4-bromobut-1-ene (51.2 g,380mmol,38.6mL,2.00 eq). The resulting reaction solution was warmed to 25 ℃ and reacted for 16h followed by quenching with saturated aqueous NH 4 Cl (200 mL) followed by extraction with EtOAc (100 mL x 3). The combined organic phases were washed with saturated brine (200 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to give the desired product (38.0 g,120mmol,63.1% yield).
1H NMR(400MHz,CDCl3)δ7.33(m,5H)5.77(m,1H)5.07(m,4H)3.72(m,3H)3.48(m,2H)2.01(m,8H)
And a second step of: preparation of 1-benzyl 2-methyl 2- (2- (propylene oxide-2-yl) ethyl) pyrroline-1, 2-dicarboxy late
To a solution of 1-benzyl 2-methyl-2- (but-3-en-1-yl) pyrroline-1, 2-dicarboxylic acid ester (37.5 g,118mmol,1.00 eq) in DCM (650 mL) at 0deg.C was added m-CPBA (60.0 g,295mmol,85.0% purity,2.50 eq) in portions. The reaction mixture obtained was reacted for 16h at 25 ℃ and then washed with saturated Na 2SO3 (300 ml x 3). The organic phase was dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (29.0 g,87.0mmol,73.6% yield).
1H NMR(400MHz,CDCl3)δ7.33(br d,J=7.88Hz,5H)5.11(m,2H)3.70(m,3H)3.48(m,2H)2.73(m,2H)2.36(m,2H)1.98(m,5H)1.50(m,2H)
And a third step of: preparation of methyl 3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylate
To a solution of 1-benzyl 2-methyl 2- (2- (propylene oxide-2-yl) ethyl) pyrroline-1, 2-dicarboxylic acid ester (28.0 g,84.0mmol,1.00 eq) in MeOH (600 mL) was added Pd/C (6.00 g,10.0% wt). The reaction was reacted at 25℃for 16h under a hydrogen atmosphere and then filtered. The filtrate was concentrated under reduced pressure to give the desired product (16.8 g). The reaction mixture was used in the next reaction without purification.
Chiral resolution of 3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester to obtain cis-3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester and trans-3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester.
Fourth step: preparation of methyl 3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylate
NaH (602 mg,15.1 mmol,60.0%wt,3.00 eq) was added to a solution of 3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester (1.00 g,5.02 mmol,1.00 eq) in THF (20 mL) at 0deg.C under nitrogen. The reaction mixture was reacted at 25℃for 0.5 h, followed by addition of MeI (1.42 g,10.0 mmol,625 uL,2.00 eq). The resulting reaction solution was reacted at 25 ℃ for 3H, then quenched with H 2 O (10 mL) at 0 ℃ and extracted with EtOAc (30 ml x 2). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (1.0 g,1.88 mmol,37.5% yield).
1H NMR(400 MHz,CDCl3)δ3.66-3.72(m,3H)3.30-3.33(m,3 H)2.22-2.43(m,1 H)1.83-2.12(m,6 H)1.56-1.72(m,1 H).
Cis-3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester and trans-3- (hydroxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester are used as starting materials to respectively obtain cis-3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester and trans-3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester.
Fifth step: preparation of (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LiAlH 4 (356 mg,9.38 mmol,2.00 eq) was added to a solution of 3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidine-7 a (5H) -carboxylic acid methyl ester (1.00g,4.69 mmol,1.00 eq) in THF (10 mL) at 0deg.C under nitrogen. The reaction mixture was reacted at 25 ℃ for 1H, then quenched by sequential addition of H 2 O (10 mL), 15% naoh (10 mL) and H 2 O (30 mL) at 0 ℃ and extracted with EtOAc (50 ml x 2). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (1.1 g).
LC-MS:m/z 186(M+H)+.1H NMR(400 MHz,CDCl3)δ4.39(dd,J=10.54,9.03 Hz,1 H)4.26(br d,J=6.53 Hz,1 H)3.97-4.06(m,1 H)3.80-3.96(m,2 H)3.68-3.76(m,2H)3.50(s,3 H)2.04-2.40(m,6 H)1.78-1.93(m,2 H)
The following compounds were synthesized from different starting materials according to the same synthesis method as intermediate 1-1:
intermediate 1-1A and intermediate 1-1B cis- (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol and trans- (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
Intermediate 1-1A
LC-MS:m/z 186(M+H)+
Intermediate 1-1B
LC-MS:m/z 186(M+H)+
Intermediate 1-2 (3- ((ethoxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 200(M+H)+
Intermediate 1-3 (3- ((isopropoxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 214(M+H)+
Intermediate 1-4 (3- ((cyclopropylmethoxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 226(M+H)+
Intermediate 1-5 (3- (cyclopropoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 212(M+H)+
Intermediate 1-6 ((2R) -2-fluoro-5- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 204(M+H)+
Intermediate 1-7 ((2R) -2-fluoro-5- (cyclopropoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol
LC-MS:m/z 230(M+H)+
Preparation of intermediate 1-8 (dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidinyl ] -7a ' (5 ' H) -yl) methanol
The first step: preparation of (1- ((benzyloxy) methyl) cyclopropyl) methanol
To a solution of cyclopropane-1, 1-dimethanol (25.0 g,245mmol,1.00 eq) in THF (500 mL) was added NaH (9.79 g,245mmol,60.0% wt,1.00 eq) at 0deg.C. The mixture was stirred at 25℃for 30min, followed by BnBr (41.9 g,245mmol,29.1mL,1.00 eq). The resulting reaction was reacted at 25 ℃ for 18h, then quenched with saturated aqueous NH 4 Cl (50 mL) and extracted with EtOAc (50 mL x 3). The combined organic phases were washed with saturated brine (20 ml×3), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (26.00 g,108mmol,44.1% yield).
1H NMR(400MHz,CDCl3)δ7.35(s,5H),4.56(s,2H),3.58(s,2H),3.47(s,2H),0.64-0.44(m,4H)
And a second step of: preparation of((1- (bromomethyl) cyclopropyl) methoxy) methyl) benzene
To a solution of (1- ((benzyloxy) methyl) cyclopropyl) methanol (26.0 g,135mmol,1.00 eq) in DCM (300 mL) was added PPh 3 (39.0 g,149mmol,1.10 eq) and NBS (26.5 g,148mmol,1.10 eq) at 0deg.C. The reaction solution was reacted at 25℃for 16 hours, and then concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (24.0 g,84.7mmol,62.6% yield).
1H NMR(400MHz,CDCl3)δ7.27(s,5H),4.47(s,2H),3.49(s,2H),3.38(s,2H),0.54-0.27(m,4H)
And a third step of: preparation of 1- (tert-butyl) 2-methyl 2- ((1- ((benzyloxy) methyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester
LiHMDS (1.00M, 78.5mL,1.20 eq) was added dropwise to 1- (tert-butyl) 2-methylpyrrolidine-1, 2-dicarboxylic acid ester (15.0 g,65.4mmol,1.00 eq) in THF (150 mL) at-70 ℃. The resulting reaction solution was reacted at the present temperature for 2 hours, followed by addition of (((1- (bromomethyl) cyclopropyl) methoxy) methyl) benzene (21.7 g,85.1mmol,1.30 eq) and HMPA (44.5 g, 247 mmol,43.7mL,3.80 eq). The resulting reaction was reacted at 25 ℃ for 16h, then quenched with water (100 mL) and extracted with EtOAc (100 mL x 2). The combined organic phases were washed with saturated brine (50 ml×2), dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (12.0 g,26.8mmol,40.9% yield).
1H NMR(400MHz,CD3OD)δ7.46-7.21(m,5H),4.68-4.35(m,2H),3.79-3.56(m,4H),3.45-3.26(m,3H),2.59-2.38(m,2H),2.09-1.82(m,4H),1.47-1.37(m,9H),0.54-0.42(m,4H)
Fourth step: preparation of 1- (tert-butyl) 2-methyl 2- ((1- (hydroxymethyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester
Pd/C (10% wt,1.50 g) was added to a solution of 1- (tert-butyl) 2-methyl 2- ((1- ((benzyloxy) methyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester (11.0 g,27.3mmol,1.00 eq) in MeOH (100 mL) under nitrogen. The resulting reaction solution was reacted at 25℃for 16 hours under a hydrogen atmosphere (15 psi). The reaction solution was filtered, the filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (10.0 g,25.5mmol,93.6% yield).
1H NMR(400MHz,CD3OD)δ3.73-3.60(m,4H),3.37(s,5H),2.08-1.90(m,4H),1.48-1.42(m,9H),0.59-0.36(m,4H)
Fifth step: preparation of 1- (tert-butyl) 2-methyl 2- ((1- (bromomethyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester
To a solution of 1- (tert-butyl) 2-methyl 2- ((1- (hydroxymethyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester (5.00 g,16.0mmol,1.00 eq) in DCM (50 mL) was added CBr 4 (7.94 g,23.9mmol,1.50 eq) and PPh 3 (6.11 g,23.3mmol,1.46 eq). The resulting mixture was reacted at 25℃for 16 hours, and then concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (0.50 g,1.20mmol,7.50% yield).
1H NMR(400MHz,CDCl3)δ3.80-3.70(m,5H),3.52-3.42(m,1H),3.19-3.02(m,1H),2.82(dd,J=7.0,15.6Hz,1H),2.45-2.20(m,1H),2.13-1.89(m,4H),1.49-1.44(m,9H),1.00-0.51(m,4H).
Sixth step: preparation of methyl 2- ((1- (bromomethyl) cyclopropyl) methyl) pyrrolidine-2-carboxylate
To a solution of 1- (tert-butyl) 2-methyl 2- ((1- (bromomethyl) cyclopropyl) methyl) pyrrolidine-1, 2-dicarboxylic acid ester (1.40 g,3.72mmol,1.00 eq) in DCM (14 mL) was added TFA (64.7 g,567.3mmol,42.0mL,152 eq). The reaction was reacted at 0℃for 2 hours and then concentrated under reduced pressure to give the objective product (0.60 g,1.74mmol,46.7% yield). The reaction mixture was used in the next reaction without purification.
Seventh step: preparation of dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -pyrrolidine ] -7a ' (5 ' H) -carboxylic acid methyl ester
To a solution of methyl 2- ((1- (bromomethyl) cyclopropyl) methyl) pyrrolidine-2-carboxylate (0.60 g,2.17mmol,1.00 eq) in DMF (10 mL) was added K 2CO3 (1.20 g,8.69mmol,4.00 eq). The obtained reaction solution was reacted at 25℃for 16 hours and then concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to obtain the objective product (0.60 g,1.84mmol,84.9% yield).
1H NMR(400MHz,CDCl3)δ3.75(s,3H),3.31-3.18(m,1H),3.01(d,J=10.0Hz,1H),2.81(s,1H),2.71(d,J=10.0Hz,1H),2.28-2.22(m,2H),1.94-1.84(m,4H),0.64-0.45(m,4H).
Eighth step: preparation of (dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bisbipyrrolidine ] -7a ' (5 ' H) -yl) methanol
To a solution of dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -pyrrolidine ] -7a ' (5 ' H) -carboxylic acid methyl ester (0.60 g,3.07mmol,1.00 eq) in THF (1 mL) under nitrogen was added LAH (174.9 mg,4.61mmol,1.50 eq). The resulting reaction solution was reacted at 0℃for 4 hours, then quenched by addition of water (0.3 mL), and then MgSO 4 (2 g) was added. The resulting reaction solution was stirred at room temperature for 30min and then filtered. The filtrate was concentrated under reduced pressure to give the desired product (0.200 g,1.08mmol,35.0% yield).
LC-MS:m/z 168(M+H)+.1H NMR(400MHz,CDCl3)δ3.50-3.43(m,1H),3.39-3.31(m,1H),3.10-3.03(m,1H),2.90(d,J=10.3Hz,1H),2.80(td,J=6.3,10.5Hz,1H),2.69(d,J=10.5Hz,1H),1.98-1.82(m,6H),0.62-0.46(m,4H).
The following compounds were synthesized from different starting materials in the same synthesis as intermediates 1 to 8:
Intermediate 1-9 ((6'R) -6' -fluoro dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methanol
LC-MS:m/z 186(M+H)+.1H NMR(400MHz,CDCl3)δ7.00(s,1H),5.44-4.99(m,1H),3.60-3.25(m,3H),3.23-2.95(m,3H),2.88-2.51(m,1H),2.36-2.03(m,4H),1.96-1.57(m,1H),1.43-1.25(m,1H),0.67-0.38(m,4H)
Intermediate 1-10 ((6'S) -6' -fluoro dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methanol
LC-MS:m/z 186(M+H)+.1H NMR(400MHz,CDCl3)δ5.15-5.43(m,1H)3.41-3.54(m,3H)3.00-3.14(m,3H)2.56(d,J=11.22Hz,1H)2.22-2.36(m,1H)2.00-2.17(m,1H)1.94(d,J=12.98Hz,1H)1.64(d,J=12.76Hz,1H)0.44-0.69(m,4H).
Intermediate 1-11A and 1-11B ((trans) -2, 2-difluorodihydro-1 'H,3' H-spiro [ cyclopropane-1, 2 '-bisfused pyrrolidinyl ] -7a' (5 'H) -yl) methanol ((cis) -2, 2-difluorodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidinyl ] -7a '(5' H) -yl) methanol preparation scheme one
The first step: preparation of ethyl 2, 2-difluoro-5 '-oxo-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisbipyrrolidine ] -7a '(5' H) -carboxylate (intermediate 1-11-I)
To a solution of ethyl 2-methylene-5-oxotetrahydro-1H-bis-fused-pyrrolidine-7 a (5H) -carboxylate (4.8 g,22.9mmol,1.00 eq) in toluene (75 mL) was added TBAB (222 mg,688 mol,0.03 eq) and [ bromo (difluoro) methyl ] -trimethyl-silicon (14.0 g,68.8mmol,3.00 eq). The reaction solution was reacted at 110℃for 16 hours, and then concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (5.2 g,87.6% yield).
LC-MS:m/z 260(M+H)+
And a second step of: preparation of ((trans) -2, 2-difluorodihydro-1 'H,3' H-spiro [ cyclopropane-1, 2 '-bisbipyrrolidine ] -7a' (5 'H) -yl) methanol and ((cis) -2, 2-difluorodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisbipyrrolidine ] -7a '(5' H) -yl) methanol (intermediates 1-11A and 1-11B)
LAH (2.1 g,57.6mmol,6.00 eq) was added to a solution of ethyl 2, 2-difluoro-5 '-oxo-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidine ] -7a '(5' H) -carboxylate (2.40 g,9.26mmol,1.00 eq) in THF (40 mL) at 0deg.C. The reaction mixture was reacted at 60℃for 2 hours and then quenched with Na 2SO4.10H2 O (20 g) at 0℃and then filtered. The filter cake was washed with THF (40 mL). The filtrate was concentrated under reduced pressure to give intermediate 1-11 ((2, 2-difluoro-dihydro-1 ' h,3' h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidine ] -7a ' (5 ' h) -yl) methanol).
LC-MS:m/z 204(M+H)+
The intermediates 1-11 were separated by silica gel column chromatography (DCM: meOH: NH 3.H2 o=20:1:0.05) to give ((trans) -2, 2-difluoro-1 'h,3' h-spiro [ cyclopropane-1, 2 '-bis-fused pyrrolidino ] -7a' (5 'h) -yl) methanol ((cis) -2, 2-difluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a '(5' h) -yl) methanol.
Isomers 1-11A (0.50 g):
LC-MS:m/z 204(M+H)+.1H NMR(400MHz,CDCl3)δ3.44-3.35(m,2H),3.19-3.13(m,2H),2.83(d,J=12.1Hz,1H),2.73-2.59(m,1H),2.73-2.59(m,1H),2.05(dd,J=6.3,13.3Hz,1H),1.93-1.84(m,3H),1.83-1.74(m,1H),1.74-1.63(m,1H),1.36(ddd,J=4.0,8.1,12.1Hz,1H),1.28(ddd,J=4.2,8.1,12.4Hz,1H).
Isomers 1-11B (0.64 g):
LC-MS:m/z 204(M+H)+.1H NMR(400MHz,CDCl3)δ3.32(d,J=2.0Hz,2H),
3.24(d,J=11.0Hz,1H),3.12-3.02(m,1H),2.83-2.71(m,2H),2.05(d,J=13.2Hz,1H),1.98-1.89(m,2H),1.89-1.81(m,1H),1.81-1.70(m,2H),1.36-1.28(m,2H).
Preparation of intermediate 1-11A and 1-11B ((trans) -2, 2-difluorodihydro-1 'H,3' H-spiro [ cyclopropane-1, 2 '-bisfused pyrrolidinyl ] -7a' (5 'H) -yl) methanol ((cis) -2, 2-difluorodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidinyl ] -7a '(5' H) -yl) methanol scheme II
The first step: preparation of (trans) -2, 2-difluoro-5 '-oxodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ] -7a '(5' H) -carboxylic acid ethyl ester and (cis) -2, 2-difluoro-5 '-oxodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ] -7a '(5' H) -carboxylic acid ethyl ester (intermediate 1-11-IA and intermediate 1-11-IB)
Separating intermediate 1-11-I (6 g) by silica gel column chromatography40gSILICA FLASH Column, eluent: intermediate I-11-IA (2.80 g,10.8mmol,47% yield) and intermediate I-11-IB (2.40 g,9.26mmol,40% yield) were obtained in sequence from 0-50% THF/petroleum ether 40 mL/min.
And a second step of: preparation of ((trans) -2, 2-difluorodihydro-1 'H,3' H-spiro [ cyclopropane-1, 2 '-bisbipyrrolidine ] -7a' (5 'H) -yl) methanol and ((cis) -2, 2-difluorodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisbipyrrolidine ] -7a '(5' H) -yl) methanol (intermediate 1-11-A and intermediate 1-11-B)
Intermediate 1-11A was synthesized as intermediate I-11-IA according to the same procedure as described.
Intermediate 1-11B was synthesized as the same procedure as the preparation route using intermediate I-11-IB.
Isomer 1-11A is chiral resolved to give isomer 1-11A-1 and isomer 1-11A-2.
The first step: preparation of intermediates 1-11A-I1 and intermediates 1-11A-I2
To a solution of intermediate 1-11A (9.00 g,44.3mmol,1.00 eq) in DMF (100 mL) was added imidazole (6.03 g,88.6mmol,2.00 eq) and TBDPSCl (14.6 g,53.1mmol,13.6mL,1.20 eq). The reaction solution was reacted at 25℃for 16 hours and then concentrated under reduced pressure. Silica gel column chromatography of the residue gave the desired product intermediate 1-11A-I (20.0 g,41.7mmol,94.1% yield).
LC-MS:m/z 442(M+H)+.1H NMR(400MHz,CDCl3)δ7.77-7.62(m,4H),7.53-7.34(m,6H),3.51(s,2H),3.14-3.02(m,2H),2.79(d,J=12.0Hz,1H),2.65-2.54(m,1H),2.13-1.99(m,2H),1.90(d,J=13.3Hz,1H),1.86-1.77(m,1H),1.72-1.61(m,2H),1.23-1.16(m,2H),1.10(s,9H).
Intermediate 1-11A-I was chiral separated (column:DAICEL CHIRALPAK IC(250mm*50mm,10um);mobile phase:[CO2-EtOH(0.1%NH3H2O)];15% B isocratic elution mode) by SFC to afford two isomers:
Intermediate 1-11A-I1 (4.50 g,10.2mmol,45.0% yield): RT 1.931min. Lc-MS: m/z 442 (M+H) +.
Intermediate 1-11A-I2 (4.20 g,9.51mmol,42.0% yield): RT 2.412min. Lc-MS: m/z 442 (M+H) +.
And a second step of: preparation of intermediate 1-11A-1 and intermediate 1-11A-2
To a solution of intermediate 1-11A-I1 (4.50 g,10.2mmol,1.00 eq) in MeOH (50 mL) was added KHF 2 (15.9 g,203mmol,6.72mL,20.0 eq). The reaction solution was reacted at 25℃for 16 hours and then concentrated under reduced pressure. Chromatography of the residue on silica gel column20gSILICA FLASH Column, eluent: 0-25% MeOH/DCM@40 mL/min) to give the desired product 1-11A-1 (1.71 g,7.98mmol,78.3% yield ).LC-MS:m/z 204(M+H)+.1H NMR(400MHz,CDCl3)δ3.40-3.16(m,2H),3.03(dd,J=7.4,12.1Hz,2H),2.73(d,J=12.1Hz,1H),2.66-2.43(m,1H),2.00-1.86(m,1H),1.84-1.73(m,3H),1.72-1.54(m,2H),1.33-1.10(m,2H)
Intermediate 1-11A-2 (1.79 g,8.38mmol,88.1% yield) was synthesized according to the same procedure:
LC-MS:m/z 204(M+H)+.1H NMR(400MHz,CDCl3)δ3.35-3.20(m,2H),3.10-2.96(m,2H),2.73(d,J=12.1Hz,1H),2.63-2.49(m,1H),1.97-1.87(m,1H),1.84-1.74(m,3H),1.72-1.53(m,2H),1.31-1.11(m,2H).
Intermediate D-1-11A-1 and intermediate D-1-11A-2 were synthesized in the same manner as intermediate 1-11A-1 and intermediate 1-11A-2.
Intermediate products D-1-11A-1.LC-MS:m/z 208(M+H)+.1H NMR(400MHz,CDCl3)δ2.81-3.08(m,2H)2.73(d,J=12.13Hz,1H)1.92(dd,J=13.26,6.13Hz,1H)1.73-1.83(m,3H)1.52-1.71(m,2H)1.08-1.33(m,2H).
Intermediate products D-1-11A-2.LC-MS:m/z 208(M+H)+.1H NMR(400MHz,CDCl3)δ3.03(dd,J=12.13,7.25Hz,2H)2.73(d,J=12.13Hz,1H)1.92(dd,J=13.26,6.25Hz,1H)1.74-1.83(m,3H)1.53-1.70(m,2H)1.13-1.32(m,2H).
Preparation of intermediate 1-12 (tetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a ' (5 ' H) -yl) methanol
The first step: preparation of (S) -5- (((tert-butylmethylsiloxy) methyl) pyrrolin-2-one
To a solution of (S) -5- (hydroxymethyl) pyrrolin-2-one (50.0 g,434mmol,1.00 eq) and imidazole (59.1 g,868mmol,2.00 eq) in DCM (1000 mL) was added TBSCl (78.6 g,521mmol,63.9mL,1.20 eq) in portions. The reaction mixture was reacted at 25℃for 2H, then H 2 O (1000 mL) was added dropwise at 0℃and DCM (1000 mL) was added to dilute. The mixture was washed with saturated brine, dried over anhydrous magnesium sulfate, and filtered. Concentrating the filtrate under reduced pressure to obtain the target product. The reaction mixture was used in the next reaction without purification.
1H NMR(400MHz,CDCl3)δ6.43(br s,1H)3.71(m,1H)3.57(m,1H)3.44(m,1H)2.30(m,2H)2.13(m,1H)1.74(m,1H)0.85(s,9H)0.02(m,6H).
And a second step of: preparation of (S) -1-benzyl-5- (((tert-butylmethylsiloxy) oxy) methyl) pyrrolin-2-one
To a solution of 0 ℃ (S) -5- (((tert-butylmethylsilyl) oxy) methyl) pyrrolin-2-one (100 g,436mmol,1.00 eq) in THF (1250 mL) under nitrogen was added NaH (72.7 g,1.82mol,60.0% wt,4.17 eq) in portions over 2 h. The mixture was reacted at 0℃for 0.5h, followed by the addition of BnBr (112 g, 254 mmol,77.7mL,1.50 eq). The resulting mixture was reacted at 25 ℃ for 16h, then quenched (1000 mL) by addition of saturated aqueous NH 4 Cl at 0-10 ℃ under nitrogen protection, and extracted with EtOAc (1000 mL x 2). The combined organic phases were washed with saturated brine (1000 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (122 g,382mmol,87.6% yield).
1H NMR(400MHz,CDCl3)δ7.29(m,5H)5.02(d,J=14.97Hz,1H)4.06(d,J=14.97Hz,1H)3.69(m,1H)3.54(m,2H)2.55(m,1H)2.38(m,1H)2.05(m,1H)1.91(m,1H)0.89(s,9H)0.04(s,6H)
And a third step of: preparation of (S) -4-benzyl-5- (((tert-butylmethylsilyl) oxy) methyl) -4-azaspiro [2.4] heptane
Ti (OiPr) 4 (224 g,789mmol,2.25 eq) was added to a solution of TiCl 4 (49.9 g,263mmol,0.750 eq) in toluene (263 mL) at 0deg.C under nitrogen. The reaction mixture was reacted at 25℃for 2 hours, followed by dropwise addition of MeLi (1.60M, 657mL,3.00 eq) at 0 ℃. The resulting compound was reacted at 25℃for 1 hour, followed by addition of a solution of (S) -1-benzyl-5- (((tert-butylmethylsilyl) oxy) methyl) pyrrolin-2-one (112 g,351mmol,1.00 eq) in THF (560 mL), followed by dropwise addition of EtMgBr (3.00M, 400mL,3.42 eq) at 0℃over 2 hours. The resulting mixture was reacted at 25℃for 2 hours, followed by quenching the reaction with water (700 mL) at 0℃and then filtration. The filter cake was washed with EtOAc (1000 mL). The combined organic phases were washed with saturated brine (500 ml×2), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (19.0 g,57.3mmol,16.4% yield).
1H NMR(400MHz,CDCl3)δ7.29(m,5H)3.75(d,J=13.82Hz,1H)3.54(d,J=13.82Hz,1H)3.40(m,2H)3.03(m,1H)2.07(m,1H)1.95(dt,J=11.80,8.22Hz,1H)1.78(m,1H)1.55(ddd,J=12.01,8.04,4.40Hz,1H)0.84(s,9H)0.79(m,1H)0.56(m,1H)0.48(m,1H)0.41(m,1H)-0.06(d,J=9.17Hz,6H)
Fourth step: preparation of (S) - (4-benzyl-4-azaspiro [2.4] heptane-5-yl) methanol
To a solution of (S) -4-benzyl-5- (((tert-butylmethylsilyl) oxy) methyl) -4-azaspiro [2.4] heptane (19.0 g,57.3mmol,1.00 eq) in THF (300 mL) was added TBAF (1.00M, 57.3mL,1.00 eq). The mixture was reacted at 25℃for 16h, followed by dilution with EtOAc (500 mL). The resulting mixture was dried over anhydrous MgSO 4 after being successively washed with H 2 O (100 mL) and saturated brine (100 mL), and then filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (5.30 g,24.4mmol,42.6% yield).
1H NMR(400MHz,CDCl3)δ7.28(m,4H)7.20(m,1H)4.14(br t,J=5.39Hz,1H)3.67(d,J=14.09Hz,1H)3.50(d,J=14.08Hz,1H)3.24(m,1H)3.14(m,1H)2.91(m,1H)1.96(m,2H)1.65(m,1H)1.49(ddd,J=11.50,8.20,3.41Hz,1H)0.80(m,1H)0.45(m,2H)0.37(m,1H).
Fifth step: preparation of (S) - (4-azaspiro [2.4] heptan-5-yl) methoxide hydrochloride
A solution of (S) - (4-benzyl-4-azaspiro [2.4] heptane-5-yl) methanol (5.00 g,23.0mmol,1.00 eq) in MeOH (50.0 mL) under nitrogen was added Pd/C (500 mg,10.0% wt) and HCl (12.0M, 8.00mL,4.17 eq). The mixture was reacted at 30℃under a hydrogen atmosphere for 3days and then filtered, and the filtrate was concentrated under reduced pressure to give the objective product (4.50 g, crude product). It was used in the next reaction without further purification.
1H NMR(400MHz,CDCl3)δ9.67(br s,1H)8.97(br s,1H)3.60(m,3H)2.07(m,1H)1.92(m,2H)1.79(m,1H)1.14(m,2H)0.77(m,2H).
Sixth step: preparation of (S) -5- (hydroxymethyl) -azaspiro [2.4] heptane-4-carboxylic acid tert-butyl ester
To a solution of (S) - (4-azaspiro [2.4] heptan-5-yl) methanolic hydrochloride (4.50 g,27.5mmol,1.00 eq) in DCM (90.0 mL) were added TEA (3.34 g,33.0mmol,4.59mL,1.20 eq) and Boc 2 O (7.20 g,33.0mmol,7.58mL,1.20 eq). The mixture was reacted at 25 ℃ for 16h, then quenched with water (50 mL) at 0 ℃ and extracted with DCM (50 mL x 2). The combined organic phases were washed with saturated brine (50 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure to give the desired product (5.90 g,26.0mmol,94.4% yield).
1H NMR(400MHz,CDCl3)δ3.85(br d,J=5.94Hz,1H)3.52(ddd,J=9.96,5.56,3.85Hz,1H)3.25(ddd,J=10.12,8.58,5.94Hz,1H)2.14(m,1H)1.85(m,2H)1.50(br s,1H)1.35(s,9H)1.15(m,1H)0.84(m,1H)0.39(m,2H).
Seventh step: preparation of (S) -4- (tert-butyloxycarbonyl) -4-azaspiro [2.4] heptane-5-carboxylic acid
To a solution of NaIO 4 (11.7 g,54.7mmol,3.03mL,2.54 eq) in H 2 O (18.0 mL) at 0deg.C was added a mixed solution of (S) -tert-butyl 5- (hydroxymethyl) -azaspiro [2.4] heptane-4-carboxylate (4.90 g,21.6mmol,1.00 eq) in ACN (12.0 mL) and CCl 4 (12.0 mL), followed by RuCl 3.H2 O (206 mg, 912. Mu. Mol,0.0423 eq). The mixture was reacted at 25 ℃ for 16h, then quenched with dropwise addition of water (30 mL) at 0 ℃ and extracted with DCM (50 mL x 2). The combined organic phases were washed with saturated brine (50 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure to give the desired product (4.70 g, crude). It was used in the next reaction without further purification.
LC-MS:m/z 242(M+H)+
Eighth step: preparation of methyl (S) -4- (tert-butyloxycarbonyl) -4-azaspiro [2.4] heptane-5-carboxylate
To a solution of (S) -4- (tert-butyloxycarbonyl) -4-azaspiro [2.4] heptane-5-carboxylic acid (4.70 g,19.5mmol,1.00 eq) in acetone (50.0 mL) at 0deg.C were added MeI (5.53 g,39.0mmol,2.43mL,2.00 eq) and K 2CO3 (8.08 g,58.4mmol,3.00 eq). The mixture was reacted at 25 ℃ for 16h, then quenched with dropwise addition of water (30 mL) at 0 ℃ and extracted with DCM (50 mL x 2). The combined organic phases were washed with saturated brine (50 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (1.10 g,4.31mmol,22.1% yield).
1H NMR(400MHz,CDCl3)δ4.33(br dd,J=8.44,3.79Hz,1H)3.65(m,3H)2.20(m,1H)1.81(m,3H)1.39(m,1H)1.30(br s,9H)0.86(m,1H)0.48(br s,2H).
Ninth step: preparation of 4- (tert-butyl) 5-methyl 5- (3-chloropropyl) -4-azaspiro [2.4] heptane-4, 5-dicarboxylic acid ester
LiHMDS (1.00M, 5.88mL,1.50 eq) was added to a solution of methyl-70 ℃ (S) -4- (tert-butyloxycarbonyl) -4-azaspiro [2.4] heptane-5-carboxylate (1.00 g,3.92mmol,1.00 eq) in THF (10.0 mL) under nitrogen. The reaction mixture was reacted at-70℃for 1 hour, followed by the addition of 1-chloro-3-iodo-propane (2.40 g,11.8mmol,1.26mL,3.00 eq). The resulting mixture was reacted at-70℃for 1 hour, followed by reaction at 25℃for 40 hours. The mixture was quenched with saturated NH 4 Cl (20 mL) at-0 ℃ and extracted with EtOAc (30 mL x 2). The combined organic phases were washed with saturated brine (50 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (767 mg,2.31mmol,59.0% yield).
1H NMR(400MHz,CDCl3)δ3.73(s,3H)3.59(br t,J=6.27Hz,2H)2.39(m,1H)2.07(m,4H)1.89(m,1H)1.74(m,2H)1.44(m,1H)1.39(m,9H)0.90(br dd,J=14.97,7.04Hz,1H)0.50(m,2H).
Tenth step: preparation of 5-methyl 5- (3-chloropropyl) -4-azaspiro [2.4] heptane-5-carboxylic acid ester hydrochloride
To a solution of 4- (tert-butyl) 5-methyl 5- (3-chloropropyl) -4-azaspiro [2.4] heptane-4, 5-dicarboxylic acid ester (700 mg,2.11mmol,1.00 eq) in DCM (8.00 mL) was added HCl/dioxane (4.00M, 4.00mL,7.58 eq). The mixture was reacted at 25℃for 1 hour, and then concentrated under reduced pressure to give the objective product (698 mg, crude). It was used in the next reaction without further purification.
LC-MS:m/z 232(M+H)+
Eleventh step: preparation of tetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a ' (5 ' H) -methyl formate
To a solution of 5-methyl-5- (3-chloropropyl) -4-azaspiro [2.4] heptane-5-carboxylic acid ester hydrochloride (698 mg,2.60mmol,1.00 eq) in ACN (8.00 mL) was added K 2CO3 (1.08 g,7.81mmol,3.00 eq). The mixture was reacted at 25℃for 16 hours and filtered, and the filtrate was concentrated under reduced pressure to give the objective product (412 mg,2.11mmol,81.1% yield). It was used in the next reaction without further purification.
1H NMR(400MHz,CDCl3)δ3.74(m,3H)3.15(dt,J=9.24,4.62Hz,1H)2.73(m,1H)2.45(m,2H)2.13(td,J=11.55,7.92Hz,1H)1.94(m,1H)1.77(m,4H)1.31(m,2H)0.97(m,1H)0.66(m,2H)0.37(m,1H)
Twelfth step: preparation of (tetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a ' (5 ' H) -yl) methanol
To a solution of tetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a ' (5 ' H) -carboxylic acid methyl ester (412 mg,1.78mmol,1.00 eq) in THF (4.00 mL) at 0deg.C under nitrogen was added LiAlH 4 (135 mg,3.56mmol,2.00 eq). The reaction was reacted at 25℃for 1 hour, then quenched by the sequential addition of H 2 O (0.04 mL), aqueous NaOH (15% wt,0.04 mL) and H 2 O (0.12 mL). The mixture was dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the desired product (263 mg,1.57 mmol).
LC-MS:m/z 168(M+H)+.1H NMR(400MHz,CDCl3)δ3.40(d,J=10.15Hz,1H)
3.28(d,J=10.15Hz,1H)2.99(m,1H)2.73(m,1H)2.19(m,1H)1.91(m,2H)1.69(m,4H)1.26(m,2H)0.86(m,1H)0.61(dt,J=9.81,5.73Hz,1H)0.50(dt,J=10.18,5.00Hz,1H)0.38(ddd,J=9.90,6.05,4.10Hz,1H).
Intermediates 1-13A and 13B were synthesized in the same manner as intermediate 1-12: ((cis) -6 '-fluorotetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a '(5' H) -yl) methanol and ((trans) -6 '-fluorotetrahydrospiro [ cyclopropane-1, 3' -bisbipyrrolidine ] -7a '(5' H) -yl) methanol
Intermediate 1-13A
LC-MS:m/z 186(M+H)+.1H NMR(400MHz,CDCl3)δ5.26-5.02(m,1H),3.48(d,J=10.8Hz,1H),3.42-3.13(m,3H),2.96-2.57(m,1H),2.52-2.29(m,2H),2.18-1.94(m,2H),1.87-1.74(m,1H),1.40-1.29(m,1H),0.95-0.82(m,1H),0.76-0.59(m,2H),0.53-0.38(m,1H),0.53-0.38(m,1H).
Intermediates 1-13B
LC-MS:m/z 186(M+H)+.1H NMR(400MHz,CDCl3)δ5.30-5.06(m,1H),3.57-3.41(m,2H),3.33(br dd,J=12.4,18.8Hz,1H),3.07-2.86(m,1H),2.82-2.42(m,1H),2.40-2.27(m,1H),2.25-2.12(m,1H),2.11-1.93(m,2H),1.73(ddd,J=2.0,8.8,12.5Hz,1H),1.34(ddd,J=1.8,7.8,12.2Hz,1H),0.92-0.81(m,1H),0.77-0.57(m,2H),0.44(ddd,J=3.8,5.9,9.6Hz,1H).
Intermediates 1-14 were synthesized in the same manner as intermediates 1-8, starting from different starting materials: (7 ',7' -Difluorodihydro-3 'H-spiro [ cyclopropane-1, 2' -indolizine ] -8a '(1' H) -yl) methanol
LC-MS:m/z 218(M+H)+.1H NMR(400MHz,CDCl3)δ3.66(dd,J=10.88,2.32Hz,
1H)3.24(d,J=10.88Hz,1H)2.97(m,4H)2.26(d,J=11.62Hz,1H)1.95(m,6H)0.58(m,4H)。
Intermediates 1-15 were synthesized in the same manner as intermediates 1-8, starting from different starting materials: (6 ',6' -Difluorodihydro-3 'H-spiro [ cyclopropane-1, 2' -indolizine ] -8a '(1' H) -yl) methanol
LC-MS:m/z 218(M+H)+.1H NMR(400MHz,CDCl3)δ3.68(d,J=10.39Hz,1H)
3.29(m,2H)2.96(m,4H)2.04(m,6H)0.55(m,4H)。
Preparation of intermediates 1-16A and 1-16B ((1S, 6'R,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol and ((1R, 6'R,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol
The first step: preparation of (3R, 6R,7 aS) -3- (tert-butyl) -6-fluorotetrahydro-1H, 3H-pyrrolo [1,2-c ] oxazol-1-one
To a solution of (2S, 4R) -4-fluoropyrrolidine-2-carboxylic acid (13.3 g,100mmol,1.00 eq) in THF (40 mL) was added 2, 2-dimethylpropionaldehyde (17.3 g,200mmol,22.1mL,2.00 eq), trimethoxymethane (31.9 g,300mmol,32.9mL,3.00 eq) and TFA (1.14 g,10.0mmol, 744. Mu.L, 0.100 eq). The mixture was reacted at 57℃for 16 hours and then concentrated under reduced pressure to give the objective product (22.0 g, crude product). The reaction mixture was used in the next reaction without purification.
1H NMR(400MHz,CDCl3)δppm 5.27(m,1H)4.52(s,1H)4.09(t,J=8.14Hz,1H)3.56(m,1H)2.94(m,1H)2.54(m,1H)2.20(m,1H)0.96(m,9H).
And a second step of: preparation of (3R, 6R,7 aS) -3- (tert-butyl) -7a- (2- (chloromethyl) propenyl) -6-fluorotetrahydro-1H, 3H-pyrrolo [1,2-c ] oxazol-1-one
LDA (2.00M, 82.0mL,1.50 eq) and DMPU (28.0 g,219mmol,26.3mL,2.00 eq) were added dropwise under nitrogen to a solution of-70 ℃ (3R, 6R,7 aS) -3- (tert-butyl) -6-fluorotetrahydro-1H, 3H-pyrrolo [1,2-c ] oxazol-1-one (22.0 g,109mmol,1.00 eq) in THF (220 mL). The mixture was reacted at-70℃for 2 hours, followed by addition of 3-chloro-2- (chloromethyl) prop-1-ene (17.8 g,142mmol,16.5mL,1.30 eq). The resulting mixture was reacted at 25 ℃ for 16h, then quenched with saturated aqueous NH 4 Cl (250 mL) at 0 ℃ and extracted with EtOAc (250 mL x 2). The combined organic phases were washed with saturated brine (250 mL), dried over anhydrous MgSO 4, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (16.7 g,57.6mmol,52.7% yield).
1H NMR(400MHz,CDCl3)δ5.37(m,1H)5.33(s,1H)5.24(m,1H)5.17(s,1H)4.21(m,3H)3.43(m,1H)3.23(m,1H)2.72(s,2H)2.37(m,1H)2.12(m,1H)0.91(s,9H).
And a third step of: preparation of (2R, 7 aS) -2-fluoro-6-methylalkenyl tetrahydro-1H-bisbipyrrolidine-7 a (5H) -carboxylic acid methyl ester
A solution of (3R, 6R,7 aS) -3- (tert-butyl) -7a- (2- (chloromethyl) propenyl) -6-fluorotetrahydro-1H, 3H-pyrrolo [1,2-c ] oxazol-1-one (11.2 g,38.7mmol,1.00 eq) in MeOH (56 mL) was added NaI (6.95 g,46.4mmol,1.20 eq). The mixture was reacted at 68℃for 16h and then filtered. The filtrate was concentrated under reduced pressure. The residue was slurried with acetone (12 mL) at 25℃for 30min and filtered. ACN (70 mL) and K 2CO3 (12 g) were added to the solid, which was then filtered after 3h reaction at 25 ℃. The filtrate was concentrated under reduced pressure, and the residue was added with THF (70 mL), and then filtered, and the filtrate was concentrated under reduced pressure to give the objective product (3.50 g,17.6mmol,45.5% yield). The reaction mixture was used in the next reaction without purification.
1H NMR(400MHz,CDCl3)δ5.29(m,1H)4.97(dt,J=11.44,1.87Hz,2H)3.82(br d,J=13.86Hz,1H)3.72(m,3H)3.59(br d,J=13.86Hz,1H)3.26(m,2H)3.04(br d,J=16.07Hz,1H)2.81(br d,J=16.07Hz,1H)2.56(m,1H)2.30(m,1H).
Fourth step: preparation of (1S, 6'R,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ] -7a '(5' H) -carboxylic acid methyl ester and (1R, 6'R,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ] -7a '(5' H) -carboxylic acid methyl ester
To a solution of (2R, 7 aS) -2-fluoro-6-alkenyltetrahydro-1H-bisbipyrrolidine-7 a (5H) -carboxylic acid methyl ester (2.00 g,10.0mmol,1.00 eq) in THF (20 mL) under nitrogen was added NaI (752 mg,5.02mmol,0.500 eq) and TMSCF 3 (3.57 g,25.1mmol,2.50 eq). The reaction solution was reacted at 80℃for 16 hours, and then concentrated under reduced pressure. Chromatography of the residue on silica gel column20gSILICA FLASH Column, eluent: isomer A (279 mg,1.08mmol,10.8% yield) and isomer B (372 mg,1.49mmol,14.9% yield) were obtained in this order at 10.3% THF/PE@60 mL/min.
Isomers of A:1H NMR(400MHz,CDCl3)δ5.33(m,1H)3.78(s,3H)3.49(d,J=9.90Hz,1H)3.27(m,3H)2.45(m,4H)1.37(m,2H).
Isomers of B:1H NMR(400MHz,CDCl3)δ5.26(m,1H)3.78(m,3H)3.31(m,4H)2.54(m,2H)2.35(m,2H)1.36(m,2H).
Fifth step: preparation of intermediates 1-16A and 1-16B ((1S, 6'R,7a' S) -2,6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol and ((1R, 6'R,7a' S) -2,6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol
LAH (81.9 mg,2.16mmol,2.00 eq) was added to a solution of isomer A (399 mg,1.08mmol,1.00 eq) obtained in one step at 0deg.C in THF (3 mL) under nitrogen. The mixture was reacted at 25℃for 1H, then H 2 O (0.03 mL), aqueous NaOH (15% wt, 0.03 mL) and H 2 O (0.09 mL) were added sequentially at 0 ℃. The resulting mixture was dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the desired product intermediate 1-16A (193 mg, 871. Mu. Mol,80.7% yield). It was used in the next reaction without further purification.
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.34(br s,1H)5.21(br d,J=2.64Hz,1H)3.24(m,5H)2.77(br s,1H)2.19(m,3H)1.88(d,J=12.98Hz,1H)1.33(m,2H).
Intermediates 1-16B were synthesized in the same manner using isomer B obtained in the above step.
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.28(m,1H)5.14(m,1H)3.27(m,5H)2.80(m,1H)2.20(m,3H)1.82(br dd,J=13.31,4.73Hz,1H)1.30(m,2H).
The following compounds were synthesized in the same manner as intermediates 1 to 16A and 1 to 16B, starting from different starting materials:
Intermediates 1-16C and 1-16D: ((1S, 6'R,7a' R) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol and ((1R, 6'R,7a' R) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol
Intermediates 1 to 16C
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.31(m,1H)5.17(m,1H)3.56(m,1H)3.43(m,2H)3.21(dd,J=12.53,7.52Hz,1H)2.78(d,J=12.59Hz,2H)2.10(m,4H)1.29(s,2H).
Intermediates 1-16D
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.38(br t,J=4.95Hz,1H)
5.24(m,1H)3.75(m,1H)3.42(m,2H)3.06(m,1H)2.68(m,2H)2.26(m,3H)1.82(m,1H)1.34(m,2H)。
Intermediates 1-16E and 1-16F: ((1S, 6'S,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol and ((1R, 6'S,7a' S) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol
Intermediates 1 to 16E
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.31(m,1H)5.18(t,J=4.29
Hz,1H)3.57(d,J=11.00Hz,1H)3.45(m,2H)3.21(dd,J=12.54,7.48Hz,1H)2.83(m,2H)2.10(m,4H)1.34(m,2H)。
Intermediates 1 to 16F
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.36(m,1H)5.22(br d,J=4.40Hz,1H)3.74(m,1H)3.42(m,2H)3.03(m,1H)2.72(m,2H)2.35(m,1H)2.13(m,2H)1.78(dd,J=13.53,6.05Hz,1H)1.33(m,2H).
Intermediates 1-16G and 1-16H: ((1S, 6'S,7a' R) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol and ((1R, 6'S,7a' R) -2, 6 '-trifluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' H) -yl) methanol
Intermediates 1-16G
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.35(br s,1H)5.18(m,1H)3.73(m,1H)3.28(m,5H)2.25(m,3H)1.87(m,1H)1.32(m,2H).
Intermediates 1 to 16H
LC-MS:m/z 222(M+H)+.1H NMR(400MHz,CDCl3)δ5.05(m,1H)3.48(m,1H)3.06(m,5H)2.03(m,3H)1.62(m,1H)1.08(m,2H).
Preparation of intermediate 1-17 (1-methyltetrahydro-1H-spiro [ cyclopentane [ b ] pyrrole-2, 1' -cyclopropane ] -3a (3H) -yl) methanol
The first step: preparation of methyl 1- (2-ethoxy-2-oxoethyl) -2-oxocyclopropane-1-carboxylate
To a solution of methyl 2-oxolane-1-carboxylate (20.0 g,141mmol,17.5mL,1.00 eq) and ethyl 2-bromoacetate (25.9 g,15.5mmol,17.1mL,1.10 eq) in ACN (200 mL) was added K 2CO3 (29.2 g,211mmol,1.50 eq). The reaction solution was reacted at 70℃for 16 hours, then filtered, and the filtrate was concentrated under reduced pressure to give the objective product (32.0 g). It was used in the next reaction without further purification.
1H NMR(400MHz,CDCl3)δ4.16-4.06(m,2H),3.71(s,3H),3.03-2.76(m,2H),2.65-2.53(m,1H),2.51-2.35(m,1H),2.51-2.35(m,1H),2.17-1.95(m,1H),2.17-1.95(m,2H),1.26-1.20(m,1H),1.26-1.20(m,1H),1.24(t,J=7.2Hz,1H).
And a second step of: preparation of methyl 1-methyl-2-oxahexahydrocyclopentane [ b ] pyrrole-3 a (1H) -carboxylate
To a solution of methyl 1- (2-ethoxy-2-oxoethyl) -2-oxocyclopropane-1-carboxylate (5.00 g,21.9mmol,1.00 eq) in MeOH (50 mL) was added methylamine methanol solution (2.0M, 16.4mL,1.50 eq), acOH (1.32 g,21.9mmol,1.25mL,1.00 eq) and NaBH 3 CN (1.79 g,28.5mmol,1.30 eq). The reaction solution was reacted at 80℃for 24 hours, and then concentrated under reduced pressure. The residue was washed with 3.0M HCl (50 mL) and extracted with DCM (500 mL. Times.3). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (700 mg,3.02mmol,13.8% yield).
1H NMR(400MHz,CDCl3)δ4.15(d,J=5.6Hz,1H),3.76-3.74(m,3H),3.10(d,J=17.9Hz,1H),2.81(s,3H),2.39(d,J=17.9Hz,1H),2.23-2.11(m,1H),1.91-1.73(m,4H),1.70-1.57(m,1H).
And a third step of: preparation of 3a- (hydroxymethyl) -1-methyl hexahydrocyclopenta [ b ] pyrrol-2 (1H) -one
To MeOH (10 mL) of 1-methyl-2-oxohexahydrocyclopentane [ b ] pyrrole-3 a (1H) -carboxylic acid methyl ester (650 mg,3.30mmol,1.00 eq) at 0deg.C was added NaBH 4 (277 mg,3.29mmol,2.20 eq) in MeOH (10 mL). The reaction was reacted at 25℃for 4h, then quenched with 2mL of 1M HCl and concentrated under reduced pressure. The residue was diluted with DCM (200 mL), dried over anhydrous magnesium sulfate, filtered, and the filtrate was concentrated under reduced pressure to give the desired product (390 mg). It was used in the next reaction without further purification.
LC-MS:m/z 170(M+H)+
Fourth step: preparation of 3a- (((tert-butyldiphenylsilyl) oxy) methyl) -1-methylhexahydrocyclopenta [ b ] pyrrol-2 (1H) -one
TBDPSCl (702 mg,2.55mmol,1.20 eq) and imidazole (290 mg,4.25mmol,2.00 eq) were added to a solution of 3a- (hydroxymethyl) -1-methyl hexahydrocyclopenta [ b ] pyrrol-2 (1H) -one (360 mg,2.13mmol,1.00 eq) in DCM (5 mL) at room temperature. The reaction solution was reacted at room temperature for 2 hours, and then concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (540 mg,1.19mmol,56.04% yield).
1H NMR(400MHz,CDCl3)δ7.70-7.59(m,4H),7.52-7.37(m,6H),3.64-3.46(m,2H),2.80(s,3H),2.61(d,J=17.5Hz,1H),2.25(d,J=17.5Hz,1H),2.08-1.84(m,1H),1.83-1.75(m,1H),1.74-1.65(m,2H),1.65-1.51(m,3H),1.08(s,9H).
Fifth step: preparation of 3a- (((tert-butyldiphenylsilyl) oxy) methyl) -1-methylhexahydro-1H-spiro [ cyclopentane [ b ] pyrrole-2, 1' -cyclopropane ]
To a solution of 3a- (((tert-butyldiphenylsilyl) oxy) methyl) -1-methylhexahydrocyclopenta [ b ] pyrrol-2 (1H) -one (500 mg,1.23mmol,1.00 eq) in THF (10 mL) at 0deg.C under nitrogen was added MeTi (OiPr) 3 (1.0M, 3.07mL,2.50 eq) and EtMgBr (3.0M, 2.45mL,6.00 eq). The reaction was reacted at 25 ℃ for 16h, then diluted with water (10 mL) and extracted with EtOAc (100 mL x 2). The combined organic phases were washed with saturated brine (50 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (190 mg, 385. Mu. Mol,31.4% yield).
1H NMR(400MHz,CDCl3)δ7.68(br t,J=5.7Hz,4H),7.54-7.35(m,6H),3.73-3.42(m,2H),2.77-2.50(m,1H),2.12-1.39(m,11H),1.09(s,9H),0.96--0.27(m,1H),0.96--0.31(m,1H).
Sixth step: preparation of (1-methyltetrahydro-1H-spiro [ cyclopentane [ b ] pyrrole-2, 1' -cyclopropane ] -3a (3H) -yl) methanol
To a solution of 3a- (((tert-butyldiphenylsilyl) oxy) methyl) -1-methylhexahydro-1H-spiro [ cyclopentane [ b ] pyrrole-2, 1' -cyclopropane ] (190 mg, 453. Mu. Mol,1.00 eq) in MeOH (5 mL) was added KHF 2 (707 mg,9.05mmol, 298. Mu.L, 20.0 eq). The reaction solution was reacted at 70℃for 16 hours and then concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (72.6 mg, 360. Mu. Mol,79.6% yield).
1H NMR(400MHz,CDCl3)δ3.70-3.44(m,3H),3.70-3.44(m,1H),2.97-2.70(m,2H),2.21-1.94(m,3H),1.87(br d,J=13.4Hz,3H),1.73-1.50(m,3H),1.37(t,J=7.6Hz,1H),1.11-1.00(m,1H),0.99-0.73(m,1H),0.58-0.13(m,1H).
Preparation of intermediate 2-1 ((2-fluoro-6- (methoxymethoxy) -8- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) naphthalen-1-yl) ethynyl) triisopropylsilane
The first step: preparation of 7-fluoro-8- ((triisopropylsilyl) ethynyl) naphthalene-1, 3-diol
To a solution of 7-fluoronaphthalene-1, 3-diol (5 g,28.1mmol,1 eq), (bromoacetylene) triisopropylsilane (7.33 g,28.1mmol,1 eq) and potassium acetate (5.51 g,56.1mmol,2 eq) in dioxane (120 mL) under nitrogen at room temperature was added ruthenium dichloride 1-isopropyl-4-methyl-benzene (1.20 g,1.96mmol,0.07 eq). The reaction solution was reacted at 110℃for 16 hours and then filtered. The filter cake was washed with EtOAc (200 ml x 2). The combined organic phases were concentrated under reduced pressure. The residue was chromatographed on a column of silica gel to give the desired product (8 g,22.3mmol,79.5% yield).
1H NMR(400MHz,CDCl3)δ9.19(m,1H)7.60(dd,J=9.17,5.62Hz,1H)7.18(m,1H)6.73(m,2H)5.29(br s,1H)1.22(m,21H)
And a second step of: preparation of 7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-ol
To a solution of 7-fluoro-8- ((triisopropylsilyl) ethynyl) naphthalene-1, 3-diol (7 g,19.5mmol,1 eq) and DIEA (5.05 g,39.0mmol,6.80mL,2 eq) in DCM (100 mL) at 0 ℃ was added MOMCl (1.66 g,20.6mmol,1.57mL,1.06 eq). The resulting reaction solution was reacted at 25℃for 1.5H and then quenched with H 2 O (200 mL), pH 6-7 was adjusted with 1N HCl, and extracted with DCM (80.0 mL. Times.2). The combined organic phases were washed with saturated brine (200 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (4.2 g,10.4mmol,53.4% yield).
1H NMR(400MHz,DMSO-d6)δ8.94-9.20(m,1H)8.93-9.13(m,1H)7.58(dd,J=9.07,5.69Hz,1H)7.10(t,J=8.82Hz,1H)6.89(d,J=2.50Hz,1H)6.73(d,J=1.88Hz,1H)5.12-5.24(m,2H)3.43(s,3H)1.09-1.13(m,21H)
And a third step of: preparation of 7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl trifluoromethane sulfonate
To a solution of 7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-ol (4 g,9.94mmol,1 eq) in DCM (40 mL) at-40℃were added DIEA (3.85 g,29.8mmol,5.19mL,3 eq) and Tf 2 O (4.21 g,14.9mmol,2.46mL,1.5 eq). The resulting reaction was reacted at-40 ℃ for 0.5H, then diluted with H 2 O (20 mL) and extracted with DCM (50 mL x 2). The combined organic phases were washed with saturated brine (100 mL), dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the desired product (5 g,8.88mmol,89.4% yield, 95% purity) as a yellow oil.
1H NMR(400MHz,CDCl3)δ7.72(dd,J=9.03,5.27Hz,1H)7.44(d,J=2.26Hz,1H)7.36-7.39(m,1H)7.31-7.35(m,1H)5.20-5.37(m,2H)3.46-3.63(m,3H)1.17-1.33(m,17H)
Fourth step: preparation of ((2-fluoro-6- (methoxymethoxy) -8- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) naphthalen-1-yl) ethynyl) triisopropylsilane
Pd (dppf) Cl 2 (684 mg, 935. Mu. Mol,0.1 eq) was added to a solution of 7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl trifluoromethanesulfonate (5 g,9.35mmol,1 eq), 4, 5-tetramethyl-2- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) -1,3, 2-dioxaborolan (4.75 g,18.70mmol,2 eq), acOK (2.75 g,28.06mmol,3 eq) in tolene (100 mL) under nitrogen. The resulting reaction solution was reacted at 130℃for 8 hours and then filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (3.5 g,6.49mmol,69.3% yield).
1H NMR(400MHz,CDCl3)δ7.68(dd,J=8.93,5.62Hz,1H)7.52(d,J=2.45Hz,1H)7.39(d,J=2.57Hz,1H)7.24(t,J=8.80Hz,1H)5.18-5.39(m,2H)3.43-3.58(m,3H)1.45(s,12H)1.15-1.21(m,20H)
Preparation of intermediate 3-1 (1R, 5S) -3- (2, 7-dichloro-8-fluoropyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
The first step: preparation of 2-chloro-3-fluoro-5-iodopyridin-4-amine
To a solution of 2-chloro-3-fluoropyridin-4-amine (10 g,68.5 mmol) and NIS (18.5 g,82.2mmol,1.2 eq) in acetonitrile (50 mL) was added p-toluenesulfonic acid monohydrate (0.65 g,3.43mmol,0.05 eq). The reaction was stirred at 70℃for 16 hours, then diluted with water (30 mL) and EtOAc (200 mL). The separated organic phase was washed with s.aq.na 2CO3、S.aq.Na2SO3 and saturated brine, dried over anhydrous sodium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the desired product (16.6 g,61mmol, yield: 89%). The reaction mixture was used in the next reaction without purification.
LC-MS:m/z 273(M+H)+
And a second step of: preparation of 4-amino-6-chloro-5-fluoronicotinic acid ethyl ester
Pd (PPh 3)2Cl2 (2.1 g,3mmol,0.1 eq) and TEA (11.1 g,0.11mmol,3.6 eq) were added to a solution of 2-chloro-3-fluoro-5-iodopyridin-4-amine (8.2 g,30mmol,1.0 eq) in EtOH (150 mL) under nitrogen, and the resulting reaction mixture was reacted at 80℃for 15 hours under CO 2, then filtered, the filtrate concentrated under reduced pressure to 70-80% of the original volume and then filtered again, the filter cakes were collected and dried in vacuo to give the desired product (quantitative yield) which was used directly in the next reaction without purification.
LC-MS:m/z 219(M+H)+
And a third step of: preparation of 7-chloro-8-fluoropyridine [4,3-d ] pyrimidine-2, 4-diol
To a solution of ethyl 4-amino-6-chloro-5-fluoronicotinate (657 mg,3 mmol) obtained above in THF (7 mL) was added trichloroacetyl isocyanate (673 mg,3.6mmol,1.2 eq) at 0deg.C. The reaction solution was reacted at rt for 30min and then concentrated under reduced pressure. MeOH (15 mL) was added to the residue and cooled to 0 ℃, followed by NH 3 in methanol (7M in MeOH,15mL,105mmol). The resulting reaction solution was reacted at rt for 16 hours and then filtered. The filter cake was washed with methanol and then dried in vacuo to give the desired product (quantitative yield). The reaction mixture was used in the next reaction without purification.
LC-MS:m/z 216(M+H)+
Fourth step: preparation of 2,4, 7-trichloro-8-fluoropyridine [4,3-d ] pyrimidine
A solution of 7-chloro-8-fluoropyridine [4,3-d ] pyrimidine-2, 4-diol (500 mg,2.4 mmol) and DIPEA (1.55 g,12mmol,5.0 eq) in POCl 3 (5 mL) was reacted at 100℃for 1 hour, then concentrated under reduced pressure to give the title compound (quantitative yield). The reaction mixture was used in the next reaction without purification.
LC-MS:m/z 252(M+H)+
Fifth step: preparation of (1R, 5S) -3- (2, 7-dichloro-8-fluoropyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
To a solution of the above-obtained 2,4, 7-trichloro-8-fluoropyridine [4,3-d ] pyrimidine and DIPEA (2 g,15.5 mmol) in DCM (2 mL) was added 3, 8-diazabicyclo [3.2.l ] octane-8-carboxylic acid tert-butyl ester (500 mg,2.4 mmol) at-40 ℃. The resulting reaction solution was reacted at the current temperature for 0.5h, then diluted with water (2 mL) and extracted with DCM (2X 2 mL). The organic phase was separated, washed with saturated brine, dried over anhydrous sodium sulfate, and filtered. The filtrate was concentrated under reduced pressure to give the desired product (400 mg, crude yield: 38.9%). The reaction mixture was used in the next reaction without purification.
LC-MS:m/z 428(M+H)+
Preparation of intermediate 4-1 (1R, 5S) -3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
The first step: preparation of (1R, 5S) -3- (7-chloro-8-fluoro-2- (2, 2-trifluoroethoxy) pyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
(1R, 5S) -3- (2, 7-dichloro-8-fluoropyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester (8.00 g,18.7mmol,1.00 eq) and 2, 2-trifluoroethanol (56.1 g,560mmol,40.3mL,30.0 eq) were added DIEA (7.24 g,56.0mmol,9.76mL,3.00 eq). The resulting reaction solution was reacted at 70℃for 2 hours and then concentrated under reduced pressure. The residue was diluted with H 2 O (100 mL) and extracted with EtOAc (100 mL x 3). The combined organic phases were washed with saturated test water (100 mL), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure to give the desired product (9 g,18.3mmol,97.9% yield).
LC-MS:m/z 492(M+H)+.1H NMR(400MHz,DMSO-d6)δ8.71(s,1H),4.82(q,J=8.4Hz,2H),4.44(br d,J=12.0Hz,2H),4.31(br s,2H),3.63(br s,2H),1.95-1.83(m,2H),1.68-1.58(m,2H),1.45(s,9H)
And a second step of: preparation of (1R, 5S) -3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
To a mixture of tert-butyl (1R, 5S) -3- (7-chloro-8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate (9.00 g,18.3mmol,1.00 eq), ((2-fluoro-6- (methoxymethoxy) -8- (4, 5-tetramethyl-1, 3, 2-dioxaborolan-2-yl) naphthalen-1-yl) ethynyl) triisopropylsilane (9.85 g,19.2mmol,1.05 eq) and Cs 2CO3 (11.9 g,36.6mmol,2.00 eq) H 2 O (20.0 mL) and dioxane (100 mL) under nitrogen atmosphere was added CataCXium Pd G (611 mg,915 mmol, 0.055 eq). The resulting reaction solution was reacted at 100 ℃ for 1H, then quenched with H 2 O (50 mL) and extracted with EtOAc (100 ml×3). The combined organic phases were dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (14.8 g,17.6mmol,96.10% yield).
LC-MS:m/z 842(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.25(s,1H),8.11(dd,J=5.9,9.2Hz,1H),7.75(d,J=2.5Hz,1H),7.57(t,J=8.9Hz,1H),7.35(d,J=2.4Hz,1H),5.76(s,1H),5.46-5.27(m,2H),5.21-4.91(m,2H),4.81(d,J=12.4Hz,1H),4.38-4.17(m,3H),3.81(d,J=12.0Hz,1H),3.60-3.52(m,1H),3.43(s,3H),1.63(d,J=7.4Hz,1H),1.46(s,9H),0.80(t,J=7.8Hz,20H),0.57-0.38(m,3H).
According to the synthesis method of the intermediate 4-1, the following compounds are synthesized from different starting materials:
intermediate 4-2 (1R, 5S) -3- (7- (6- (bis (4-methoxybenzyl) amino) -4-methyl-3- (trifluoromethyl) pyridin-2-yl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 872(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.15(s,1H),7.17(br d,J=
8.4Hz,4H),6.88(br d,J=8.4Hz,4H),6.82(s,1H),5.08(q,J=8.9Hz,2H),4.69(br s,4H),4.55(br d,J=12.1Hz,2H),4.27(br s,2H),3.73(s,6H),3.70-3.62(m,2H),2.39(br s,3H),1.85-1.76(m,2H),1.68(br d,J=7.5Hz,2H),1.47(s,9H).
Intermediate 4-3 (1R, 5S) -3- (7- (2- ((tert-Butoxycarbonyl) amino) -7-fluorobenzo [ d ] thiazol-4-yl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 724(M+H)+
Intermediate 4-4 (1R, 5S) -3- (7- (2- ((tert-Butoxycarbonyl) amino) -3-cyano-7-fluorobenzo [ b ] thiophen-4-yl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 748(M+H)+.1H NMR(400MHz,DMSO-d6)δ11.79(s,1H),9.21(s,1H),7.61(dd,J=5.3,8.4Hz,1H),7.42(t,J=8.8Hz,1H),5.11(q,J=9.1Hz,2H),4.57(br d,J=12.5Hz,2H),4.28(br s,2H),3.78-3.67(m,2H),1.82(br d,J=4.8Hz,2H),1.61(br d,J=7.5Hz,2H),1.52(s,9H),1.48(s,9H).
Intermediate 4-5 (1R, 5S) -3- (8-fluoro-7- (6-fluoro-5-methyl-1- (tetrahydro-2H-pyran-2-yl) -1H-indazol-4-
Phenyl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 690(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.29(s,1H)7.75-7.81(m,2H)5.86(dd,J=9.68,1.98Hz,1H)5.12(q,J=9.17Hz,2H)4.51-4.72(m,2H)4.29(br s,2H)3.91(br d,J=11.88Hz,1H)3.76-3.84(m,1H)3.62-3.75(m,2H)2.32-2.47(m,1H)2.16-2.24(m,1H)2.19(s,2H)1.94-2.08(m,2H)1.83(br d,J=3.30Hz,2H)1.70(br d,J=7.48Hz,3H)1.59(br s,2H)1.48(s,9H).
Intermediate 4-6 (1R, 5S) -3- (8-fluoro-7- (7-fluoro-8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 782(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.26(s,1H)8.24(m,2H)7.66(m,3H)5.08(m,2H)4.83(br d,J=12.59Hz,1H)4.34(m,3H)3.83(br d,J=11.86Hz,1H)3.41(m,1H)1.91(br d,J=8.44Hz,2H)1.82(m,1H)1.65(m,1H)1.48(s,9H)0.82(m,18H)0.50(m,3H).
Intermediate 4-7 (1R, 5S) -3- (7- (8-ethyl-7-fluoro-3- (methoxymethoxy) naphthalen-1-yl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 690(M+H)+.1H NMR(400MHz,CDCl3)δ9.08(s,1H)7.71(dd,J=
9.01,5.88Hz,1H)7.55(d,J=2.63Hz,1H)7.28(s,1H)7.23(d,J=2.50Hz,1H)5.31(d,J=2.13Hz,2H)4.94(q,J=8.38Hz,2H)4.54-4.67(m,2H)4.44(br s,2H)3.60-3.90(m,2H)3.53(s,3H)2.51(br dd,J=13.95,7.07Hz,1H)2.23(ddd,J=14.32,7.32,2.75Hz,1H)2.00-2.06(m,2H)1.81(br d,J=8.38Hz,2H)1.26(s,9H)0.85(t,J=7.44Hz,3H).
Intermediate 4-8 3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 9-diazabicyclo [4.2.1] nonane-9-carboxylic acid tert-butyl ester
LC-MS:m/z 856(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.15-9.32(m,1H)8.05
-8.19(m,1H)7.76(br d,J=2.32Hz,1H)7.50-7.64(m,1H)7.14-7.46(m,1H)5.28
-5.43(m,2H)4.94-5.20(m,2H)4.08-4.51(m,4H)3.49-3.80(m,1H)3.39-3.48(m,3H)3.29-3.34(m,3H)2.01-2.21(m,2H)1.50-1.77(m,2H)1.44(s,3H)1.20(s,3H)0.94-1.06(m,3H)0.81(t,J=6.48Hz,18H)0.40-0.61(m,3H).
Intermediate 4-9 (1- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3-methylpiperidin-3-yl) carbamic acid tert-butyl ester
LC-MS:m/z 842(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.01-9.27(m,1H)8.12(dd,J=9.17,5.87Hz,1H)7.73-7.80(m,1H)7.58(t,J=8.93Hz,1H)7.35-7.41(m,1H)7.12-7.20(m,1H)6.41-6.65(m,1H)5.32-5.42(m,2H)5.08-5.19(m,1H)4.92-5.05(m,1H)4.40(br d,J=12.35Hz,1H)3.63-3.74(m,1H)3.44(s,3H)3.35-3.41(m,1H)3.28-3.34(m,2H)2.01-2.14(m,1H)1.84-1.95(m,1H)1.50-1.83(m,2H)1.30-1.41(m,3H)1.21-1.27(m,2H)1.12(s,6H)0.72-0.88(m,18H)0.34-0.58(m,3H)
Intermediate 4-10 (1R, 5S) -3- (7- (3- ((bis-tert-butyloxycarbonyl) amino) -2-fluoro-5-methyl-6- (trifluoromethyl) phenyl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 849(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.24(s,1H)7.82(d,J=9.26Hz,1H)5.11(q,J=9.01Hz,2H)4.57(br d,J=12.38Hz,2H)4.29(br s,2H)3.69(br d,J=12.26Hz,2H)2.28(br s,3H)1.82(br d,J=4.13Hz,2H)1.66(br d,J=7.50Hz,2H)1.47(s,9H)1.38(s,18H).
Intermediate 4-11 (1R, 5S) -3- (7- (2- ((di-t-butoxycarbonyl) amino) -3, 5-dichloro-6-fluorophenyl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 835(M+H)+.1H NMR(400MHz,CDCl3)δ9.00(s,1H)7.68(d,J=7.09
Hz,1H)4.81-5.01(m,2H)4.64-4.77(m,1H)4.34-4.49(m,3H)3.59-3.89(m,2H)2.02(br d,J=1.47Hz,2H)1.79-1.86(m,4H)1.78-1.86(m,1H)1.53-1.59(m,10H)1.43-1.51(m,9H)1.16-1.27(m,8H).
Intermediate 4-12 (1R, 5S) -3- (7- (5- ((bis-t-butoxycarbonyl) amino) -3-chloro-2- (trifluoromethyl) phenyl) -8-fluoro-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 851(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.06(br s,1H)7.52(s,1H)7.20(br s,1H)4.92(br d,J=7.46Hz,2H)4.44(br s,4H)3.74(br s,2H)2.02(br s,2H)1.78(br s,2H)1.55(s,9H)1.48(s,18H).
Intermediate 4-13 (1R, 5S) -3- (7- (3- (bis (4-methoxybenzyl) amino) -2-fluoro-5-methyl-6- (trifluoromethyl) phenyl) -8-methoxy-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 901(M+H)+.1H NMR(400MHz,DMSO-d6)δ8.99-9.10(m,1H)7.26-7.45(m,1H)7.02-7.23(m,4H)6.75-6.96(m,4H)4.98-5.21(m,2H)4.47-4.55(m,2H)4.21-4.29(m,2H)4.04-4.13(m,4H)3.81-3.88(m,3H)3.71-3.78(m,6H)3.60-3.69(m,2H)2.00-2.12(m,3H)1.77-1.87(m,2H)1.62-1.71(m,2H)1.41-1.51(m,9H).
Intermediate 4-14 (1R, 5S) -3- (7- (5- (bis (tert-butylcarbonyl) amino) -3-chloro-2- (trifluoromethyl) phenyl) -8-yloxy-2- (2, 2-trifluoroethoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
LC-MS:m/z 863(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),7.88(s,1H),7.37-7.27(m,1H),5.09(br d,J=9.0Hz,2H),4.65-4.39(m,2H),4.28(br s,2H),3.95-3.87(m,3H),3.76-3.53(m,2H),1.82(br s,2H),1.74-1.57(m,2H),1.47(s,9H),1.42-1.39(m,18H)
Example 14 preparation of- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
The first step: preparation of (1R, 5S) -3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester
To a solution of (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methanol (1.01 g,5.45mmol,4.59 eq) and (1R, 5S) -3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- (2, 2-trifluoroethoxy) pyridine [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate (1.00 g,1.19mmol,1.00 eq) in THF (20 mL) was added t-Buona (913 mg,9.50mmol,8.00 eq). The reaction was reacted at 25℃for 16H, then quenched with H 2 O (10 mL) and extracted with EtOAc (30 mL. Times.3). The combined organic phases were washed with saturated test water (100 mL), dried over anhydrous magnesium sulfate and filtered. The filtrate was concentrated under reduced pressure, and the residue was separated by silica gel column chromatography to give the objective product (723 mg, 255 umol,21.7% yield, 33.0% purity). The reaction mixture was used in the next reaction without purification.
And a second step of: preparation of 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxymethyl) pyridin [4,3-d ] pyrimidin-7-yl) -6-fluoro-5- ((triisopropylsilyl) ethynyl) naphthalen-2-ol
(1R, 5S) -3- (8-fluoro-7- (7-fluoro-3- (methoxymethoxy) -8- ((triisopropylsilyl) ethynyl) naphthalen-1-yl) -2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid tert-butyl ester (640 mg,238umol,33.0% purity, 1.00 eq) in DCM (6 mL) was added HCl/dioxane (4.00M, 945uL,15.8 eq). The reaction solution was reacted at 25℃for 2 hours, and then concentrated under reduced pressure to give the objective product (430 mg, crude). The reaction mixture was used in the next reaction without purification.
And a third step of: preparation of 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
To a solution of 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxymethyl) pyridin [4,3-d ] pyrimidin-7-yl) -6-fluoro-5- ((triisopropylsilyl) ethynyl) naphthalen-2-ol (400 mg, 400 umol,32.0% purity,1.00 eq) in DMF (6 mL) was added CsF (497 mg,3.27mmol,121ul,20.0 eq). The reaction solution was reacted at 25℃for 16 hours, followed by filtration. The filtrate was concentrated under reduced pressure, and the residue was separated by prep-HPLC to give the desired product (63.0 mg).
LC-MS:m/z 627(M+H)+
Chiral SFC separation gave example 1 isomer 1 (11.3 mg,17.6umol,10.8% yield) and example 1 isomer 2 (19.2 mg,29.9umol,18.3% yield).
Example 1 isomer 1
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(br s,1H)9.03(s,1H)7.98(dd,J=9.16,5.90Hz,1H)7.47(t,J=8.91Hz,1H)7.39(d,J=2.51Hz,1H)7.18(d,J=2.26Hz,1H)4.48(br d,J=11.80Hz,1H)4.31(br d,J=12.30Hz,1H)3.95-4.08(m,2H)3.94(s,1H)3.64(br d,J=12.05Hz,1H)3.57(br s,3H)3.17-3.28(m,5H)2.79-2.94(m,2H)2.65-2.70(m,1H)1.90-2.06(m,2H)1.78(br d,J=4.52Hz,3H)1.51-1.69(m,8H).
Example 1 isomer 2
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(br s,1H)9.04(s,1H)7.98(dd,J=9.16,5.90Hz,1H)7.47(t,J=9.03Hz,1H)7.40(d,J=2.51Hz,1H)7.18(d,J=2.26Hz,1H)4.49(br d,J=12.05Hz,1H)4.32(br d,J=12.30Hz,1H)3.96-4.10(m,2H)3.94(s,1H)3.65(br d,J=14.31Hz,1H)3.56-3.62(br s,3H)3.17-3.28(m,5H)2.80-2.95(m,2H)2.68(br d,J=1.76Hz,1H)1.90-2.03(m,2H)1.78(br s,3H)1.51-1.72(m,8H).
Example 1 isomer 1 and example 1 isomer 2 were separated again by SFC to give example 1 isomer 1A, example 1 isomer 1B, example 1 isomer 2A and example 1 isomer 2B:4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3 s,7 ar) - (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol, 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3 r,7 as) - (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol, 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2-oxa-yl) -8-fluoro-2- ((3, 2.1] oct-3-yl) -8-fluoro-2- ((3 r, 7-bis- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -methoxy) pyridin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol, 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3 s,7 as) - (3- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Example 1 isomer 1A
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),9.03(s,1H),7.98(dd,J=9.20,5.92Hz,1H),7.46(t,J=9.00Hz,1H),7.39(d,J=2.64Hz,1H),7.17(d,J=2.56Hz,1H),4.47(d,J=12.44Hz,1H),4.30(d,J=12.32Hz,1H),3.97-4.07(m,2H),3.92(s,1H),3.56-3.66(m,4H),3.17-3.27(m,5H),2.79-2.92(m,2H),2.63-2.69(m,1H),1.51–2.03(m,13H).
Example 1 isomer 1B
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),9.03(s,1H),7.98(dd,J=9.24,5.92Hz,1H),7.46(t,J=9.00Hz,1H),7.39(d,J=2.56Hz,1H),7.17(d,J=2.60Hz,1H),4.47(d,J=12.32Hz,1H),4.30(d,J=12.24Hz,1H),3.97-4.07(m,2H),3.92(s,1H),3.56-3.66(m,4H),3.17-3.27(m,5H),2.80-2.92(m,2H),2.63-2.68(m,1H),1.51–2.04(m,13H).
Example 1 isomer 2A
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.03(s,1H),7.99-7.95(m,1H),7.46(t,J=8Hz,1H),7.39(s,1H),7.18(s,1H),4.47(d,J=8Hz,1H),4.30(d,J=8Hz,1H),4.11(d,J=4Hz,1H),4.01(d,J=4Hz,1H),3.93(s,1H),3.63(d,J=8Hz,1H),3.57-3.53(m,3H),3.44-3.40(m,1H),3.25(s,3H),3.19(s,2H),2.72-2.64(m,2H),2.05-1.97(m,2H),1.71-1.56(m,9H),1.52-1.44(m,1H).
Example 1 isomer 2B
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.04(s,1H),7.96(m,1H),7.46(t,J=8Hz,1H),7.39(m,1H),7.18(m,1H),4.65-4.56(m,1H),4.48-4.39(m,1H),4.30(d,J=8Hz,1H),4.15-4.06(m,1H),4.02(t,J=4Hz,1H),3.93(s,1H),3.85-3.75(m,1H),3.63(d,J=4Hz,1H),3.58-3.54(m,3H),3.44-3.40(m,2H),3.25(s,3H),3.18(s,2H),2.72-2.66(m,2H),2.07-2.02(m,3H),1.71-1.57(m,7H),1.51-1.44(m,1H).
The following examples were synthesized according to the procedure of example 1 starting from different starting materials:
Example 24 preparation of- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- ((ethoxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 641(M+H)+.
Example 34 preparation of- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- ((isopropoxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 655(M+H)+.
Example 44 preparation of- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- ((cyclopropylmethyloxy) methyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 667(M+H)+.
Example 5 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((3- (cyclopropoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 653(M+H)+.
Example 6 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((2R) -2-fluoro-5- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 645(M+H)+.
Chiral separation to obtain isomer 6A and isomer 6B: cis-4- (4- ((1R, 5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((2R) -2-fluoro-5- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and trans-4- (4- ((1R, 5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((2R) -2-fluoro-5- (methoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Isomer 6A
LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(s,1H),9.03(s,1H),7.97(q,J=8Hz,1H),7.47(t,J=8Hz,1H),7.39(s,1H),7.18(d,J=4Hz,1H),5.47-5.32(m,1H),4.50(d,J=8Hz,1H),4.31(d,J=8Hz,1H),4.07(s,2H),3.96(s,1H),3.66(d,J=12Hz,1H),3.56(s,3H),3.28-3.21(m,6H),3.00-2.85(m,3H),2.36-2.24(m,1H),2.087-1.92(m,3H),1.86-1.82(m,1H),1.73-1.58(m,6H).
Isomer 6B
LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),9.03(s,1H),7.98(dd,J=5.92Hz,9.20Hz,1H),7.46(t,J=9.00Hz,1H),7.38(d,J=2.52Hz,1H),7.17(d,J=2.48Hz,1H),5.34(m,0.5H),5.20(m,0.5H),4.47(m,1H),4.29(m,1H),4.18(m,1H),4.10(m,1H),3.94(d,J=5.28Hz,1H),3.63(m,1H),3.48(m,5H),3.26(s,3H),3.00(m,2H),2.29(m,1H),1.72(m,9H).
Example 7 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((2R) -5- (cyclopropoxymethyl) -2-fluorotetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 671(M+H)+.
Example 8A and example 8B cis 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (trifluoromethoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and trans 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((3- (trifluoromethoxymethyl) tetrahydro-1H-bis-fused pyrrolidin-7 a (5H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Example 8A
LCMS:m/z 681(M+H)+.
Example 8B
LCMS:m/z 681(M+H)+.
Example 9 4- (4- (1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (dihydro-1 ' H,3' H-spiro [1,2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoro-naphthalen-2-ol
LCMS:m/z 609(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.03(m,1H),7.97(t,J=8Hz,1H),7.46(t,J=8Hz,1H),7.39(d,J=4Hz,1H),7.18-7.17(m,1H),4.47(t,J=8Hz,1H),4.34-4.21(m,2H),4.08-4.04(m,1H),3.94(d,J=4Hz,1H),3.63(t,J=8Hz,1H),3.55(s,3H),2.99-2.94(m,1H),2.74-2.61(m,3H),2.03-1.85(m,3H),1.83-1.76(m,3H),1.71-1.66(m,4H),0.49-0.45(m,4H).
Example 10 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((6'R) -6' -fluoro-dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a ' (5 ' H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 627(M+H)+.
Example 10 chiral resolution gives isomers 10A and 10B:4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((6 'r,7 a's) -6 '-fluoro dihydro-1' h,3 'h-spiro [ cyclopropan-1, 2' -bis-fused pyrrolidino ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((6 'r,7a' r) -6 '-fluoro dihydro-1' h,3 'h-spiro [ cyclopropan-1, 2' -bis-fused pyrrolidino ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Isomer 10A:
LCMS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.18(s,1H),9.06(s,1H),7.99(dd,J=5.92,9.24Hz,1H),7.48(t,J=9.00Hz,1H),7.40(d,J=2.56Hz,1H),7.19(m,1H),5.41(m,0.5H),5.28(m,0.5H),4.49(m,1H),4.35(m,2H),4.09(d,J=10.24Hz,1H),3.95(m,1H),3.58(m,4H),3.27(m,2H),3.11(m,3H),2.70(m,1H),2.16(m,3H),1.68(m,4H),0.51(m,4H).
isomer 10B:
LCMS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.02(s,1H),7.99(dd,J=5.96,9.16Hz,1H),7.48(t,J=9.04Hz,1H),7.40(d,J=2.52Hz,1H),7.16(m,1H),5.48(m,0.5H),5.35(m,0.5H),4.48(m,1H),4.30(m,2H),4.12(m,1H),3.93(s,1H),3.58(m,4H),3.26(m,2H),3.01(m,0.5H),2.89(m,0.5H),2.80(d,J=9.92Hz,1H),2.62(d,J=9.92Hz,1H),2.32(m,1H),2.03(m,2H),1.82(m,3H),1.65(m,3H),0.48(m,4H).
Example 11 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((tetrahydrospiro [ cyclopropane-1, 3' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) pyrido [4,3d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 609(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.03(m,1H),7.95(dd,J=6.00,9.28Hz,1H),7.45(t,J=9.00Hz,1H),7.37(d,J=2.56Hz,1H),7.18(m,1H),4.49(m,2H),4.32(m,1H),4.14(m,2H),3.92(m,1H),3.76(m,1H),3.62(m,1H),3.58(m,1H),2.90(m,1H),2.63(m,1H),2.04(m,7H),1.63(m,6H),1.24(m,1H),0.75(m,1H),0.58(m,1H),0.43(m,1H),0.33(m,1H).
Examples 12A and 12B were synthesized starting from intermediates 1-13A and 1-13B, respectively: 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((trans) -6 '-fluorotetrahydrospiro [ cyclopropan-1, 3' -bis-fused-pyrrolidin-7 a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((cis) -6 '-fluorotetrahydrospiro [ cyclopropan-1, 3' -bis-fused-pyrrolidin ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Example 12A
LCMS:m/z 627(M+H)+.
Example 12B
LCMS:m/z 627(M+H)+.
Example 13 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((7 ',7' -difluorotetrahydro-3 '-H-spiro [ cyclopropa-1, 2' -bisfused pyrrolidino ] -8a '(1' H) -yl) methoxy) -8-fluoropyridine [4,3d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol trifluoroacetate
LCMS:m/z 659(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.25(br,1H),9.42(m,1H),9.15(m,2H),8.00(dd,J=6.00,9.20Hz,1H),7.47(t,J=9.20Hz,1H),7.42(d,J=2.40Hz,1H),7.19(m,1H),4.63(m,5H),4.23(s,2H),3.91(m,4H),3.52(m,1H),3.39(m,4H),3.52(m,1H),3.39(m,1H),2.53(m,1H),1.97(m,5H),0.74(m,4H).
Example 14 4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((6 ',6' -difluorotetrahydro-3 '-H-spiro [ cyclopropa-1, 2' -bisfused pyrrolidino ] -8a '(1' H) -yl) methoxy) -8-fluoropyridine [4,3d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.05(s,1H),7.97(dd,J=5.92,9.24Hz,1H),7.46(t,J=9.00Hz,1H),7.39(d,J=2.60Hz,1H),7.18(s,1H),4.64(dd,J=2.92,10.88Hz,1H),4.47(d,J=12.64Hz,1H),4.35(m,2H),3.93(m,1H),3.60(m,4H),3.10(m,4H),2.81(dd,J=3.96,8.68Hz,1H),2.84(m,1H),2.02(m,1H),1.91(m,3H),1.68(m,5H),0.52(m,4H).
Examples 15A, 15B were synthesized from intermediates 1-16A and 1-16B according to the procedure of example 1:
4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 s,6'r,7 a's) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 r,6'r,7 a's) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidin ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Example 15A
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.16(s,1H),9.05(s,1H),7.98(dd,J=5.92,9.16Hz,1H),7.47(t,J=9.04Hz,1H),7.40(d,J=2.64Hz,1H),7.18(m,1H),5.43(m,0.5H),5.29(m,0.5H),4.51(t,J=12.16Hz,1H),4.31(m,1H),4.05(m,2H),3.94(d,J=4.16Hz,1H),3.61(m,4H),3.12(m,4H),2.22(m,5H),1.69(s,4H),1.54(m,2H).
Example 15B
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(s,1H),9.06(s,1H),7.98(dd,J=5.92,9.12Hz,1H),7.47(t,J=9.00Hz,1H),7.40(d,J=2.68Hz,1H),7.18(m,1H),5.35(m,0.5H),521(m,0.5H),4.48(m,1H),4.31(m,2H),4.18(m,1H),3.94(s,1H),3.61(m,4H),3.21(m,2H),3.01(m,2H),2.16(m,5H),1.69(s,4H),1.54(m,2H)
Examples 15C, 15D were synthesized from intermediates 1 to 16C and 1 to 16D according to the procedure of example 1
4- (4- ((1 R,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 s,6'r,7a' r) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 r,6'r,7a' r) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidin ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Example 15C
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.20(s,1H),9.05(s,1H),7.98(dd,J=5.92,9.28Hz,1H),7.48(t,J=8.96Hz,1H),7.40(d,J=2.52Hz,1H),7.19(d,J=2.52Hz,1H),5.45(m,0.5H),5.32(m,0.5H),4.48(d,J=11.56Hz,1H),4.30(m,3H),3.96(d,J=2.88Hz,1H),3.59(m,4H),3.31(m,2H),3.01(m,2H),2.16(m,5H),1.69(s,4H),1.54(m,2H).
Example 15D
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.16(s,1H),9.04(s,1H),7.98(dd,J=6.00,9.28Hz,1H),7.48(t,J=9.04Hz,1H),7.40(d,J=2.44Hz,1H),7.19(m,1H),5.51(m,0.5H),5.37(m,0.5H),4.50(m,1H),4.30(m,1H),4.16(m,1H),3.98(m,2H),3.61(m,4H),3.28(m,1H),3.07(m,1H),2.79(m,1H),2.21(m,5H),1.59(s,6H).
Examples 15E, 15F were synthesized from intermediates 1 to 16E and 1 to 16F as in example 1
4- (4- ((1 R,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 s,6's,7 a's) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 r,6's,7 a's) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidin ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Example 15E
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.05(s,1H),7.97(dd,J=5.92,9.16Hz,1H),7.46(t,J=9.00Hz,1H),7.39(d,J=2.52Hz,1H),7.17(d,J=2.52Hz,1H),5.44(m,0.5H),5.30(m,0.5H),4.48(d,J=11.72Hz,1H),4.30(m,3H),3.93(d,J=2.84Hz,1H),3.61(m,4H),3.16(m,2H),2.77(m,2H),2.14(m,5H),1.61(s,6H).
Example 15F
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.16(s,1H),9.04(s,1H),7.97(dd,J=5.92,9.16Hz,1H),7.46(t,J=8.96Hz,1H),7.39(d,J=2.60Hz,1H),7.18(m,1H),5.50(m,0.5H),5.36(m,0.5H),4.50(m,1H),4.29(m,1H),4.14(m,1H),4.01(m,1H),3.94(s,1H),3.60(m,4H),3.28(m,1H),3.03(m,2H),2.78(m,1H),2.16(m,5H),1.65(s,6H).
Examples 15G, 15H were synthesized from intermediates 1-16G and 1-16H according to the procedure of example 1:
4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 s,6's,7a' r) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- (((1 r,6's,7a' r) -2, 6 '-trifluoro-dihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidin ] -7a '(5' h) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-fluoronaphthalen-2-ol
Example 15G
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(s,1H),9.04(s,1H),7.97(dd,J=5.96,9.20Hz,1H),7.46(t,J=9.00Hz,1H),7.39(d,J=2.56Hz,1H),7.17(m,1H),5.42(m,0.5H),5.28(m,0.5H),4.49(m,1H),4.29(m,1H),4.04(m,2H),3.93(m,1H),3.62(m,4H),3.09(m,4H),2.11(m,5H),1.56(s,6H).
Example 15H
LCMS:m/z 663(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.01(s,1H),7.66(m,1H),7.21(m,1H),7.01(m,2H),5.34(m,0.5H),5.20(m,0.5H),4.28(m,4H),3.75(m,1H),3.58(m,3H),3.17(m,3H),2.99(m,2H),2.19(m,5H),1.56(s,6H).
The following examples were synthesized following the procedure of example 1, starting from intermediates 1-11:
Examples 16A and 16b 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((cis) -2, 2-difluoro-dihydro-1 'h,3' h-spiro [ cyclopropan-1, 2 '-bis-fused-pyrrolidin ] -7a' (5 'h) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol and 4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- (((trans) -2, 2-difluoro-dihydro-1' h,3 'h-spiro [ cyclopropan-1, 2' -bis-fused-pyrrolidin ] -7a '(5' h) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
Example 16A
LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.14(s,1H),9.03(s,1H),7.99-7.95(m,1H),7.46(t,J=8Hz,1H),7.39(s,1H),7.18(t,J=4Hz,1H),4.49(t,J=12Hz,1H),4.31(t,J=12Hz,1H),4.09(d,J=12Hz,1H),3.98(d,J=4Hz,1H),3.95-3.93(m,1H),3.65(t,J=8Hz,1H),3.59-3.56(m,3H),3.06(d,J=12Hz,1H),3.00-2.95(m,1H),2.80-2.76(m,1H),2.71-2.65(m,1H),2.14-2.10(m,1H),1.96-1.87(m,3H),1.84-1.75(m,2H),1.66(s,4H),1.56-1.51(m,2H).
Example 16B
LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.17(s,1H),9.04(s,1H),7.99-7.95(m,1H),7.46(t,J=8Hz,1H),7.39(s,1H),7.18(d,J=4Hz,1H),4.49(t,J=12Hz,1H),4.33(d,J=12Hz,1H),4.22-4.15(m,2H),3.92(s,1H),3.95-3.93(m,1H),3.65(d,J=12Hz,1H),3.58-3.56(m,3H),3.12-3.08(m,1H),3.04-3.00(m,1H),2.73(d,J=12Hz,1H),2.56-2.54(m,1H),2.10-2.05(m,1H),2.01-1.97(m,1H),1.92(d,J=16Hz,1H),1.84-1.75(m,2H),1.66-1.44(m,7H).
Example 16B was resolved by chiral HPLC (SFC-150 (Waters), column: IG 20 x 250mm,10um (Daicel), column temperature: 35 ℃, mobile phase: CO 2/EtOH[0.5%NH3 (7M in MeOH) ]=120/40 to give isomers example 16B-1 and example 16B-2.
Example 16B-1
RT:15.079min.LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),7.93-7.89(m,1H),7.41(d,J=8Hz,1H),7.32(s,1H),7.17(s,1H),4.49(d,J=12Hz,1H),4.35(d,J=12Hz,1H),4.20-4.16(m,2H),3.89(s,1H),3.64-3.55(m,5H),3.13-3.09(m,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-2.06(m,1H),2.02-1.98(m,1H),1.92(d,J=12Hz,1H),1.84-1.75(m,3H),1.66-1.47(m,8H).
Example 16B-2
RT:20.572min.LCMS:m/z 645(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.04(s,1H),7.95-7.99(m,1H),7.46(t,J=8Hz,1H),7.39(s,1H),7.18(s,1H),4.49(d,J=12Hz,1H),4.35(d,J=12Hz,1H),4.20-4.18(m,2H),392(s,1H),3.65-3.56(m,5H),3.13-3.09(m,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-2.06(m,1H),2.02-1.98(m,1H),1.92(d,J=12Hz,1H),1.84-1.74(m,3H),1.67-1.45(m,8H).
The following examples were synthesized following the procedure of example 1, starting from intermediates 1-11A:
Example 17:4- (4- (1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((-2, 2-difluoro-1 ' h,3' h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' h) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethyl-6-fluoronaphthalen-2-ol
LCMS:m/z 649(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.01(s,1H),9.10(s,1H),7.77(q,J=4Hz,1H),7.38(d,J=8Hz,1H),7.34(d,J=4Hz,1H),7.03(d,J=4Hz,1H),4.44(d,J=12Hz,2H),4.26-4.17(m,2H),3.62(t,J=12Hz,2H),3.55(s,2H),3.13(q,J=8Hz,1H),3.04-2.99(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.39-2.34(m,1H),2.15(t,J=8Hz,1H),2.12-2.06(m,1H),2.02-1.98(m,1H),1.92(d,J=12Hz,1H),1.80-1.73(m,2H),1.63-1.44(m,7H),7.38(t,J=8Hz,3H).
Resolution by chiral HPLC gives two isomers:
Example 17A:
RT:14.093min (U3000 (ThermoFisher); column: CHIRALPAK IF-3 4.6mm*150mm,3um; column temperature: 40 ℃ C.; mobile phase: n-hexane: ethanol-diethylamine) =85/15-0.1%).LCMS:m/z 649(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.94(s,1H),9.11(s,1H),7.77(q,J=4Hz,1H),7.38(d,J=8Hz,1H),7.34(s,1H),7.04(d,J=4Hz,1H),4.44(d,J=12Hz,2H),4.26-4.17(m,2H),3.65-3.56(m,4H),3.13(q,J=8Hz,1H),3.04-2.99(m,1H),2.74(d,J=12Hz,1H),2.57-2.54(m,1H),2.39-2.34(m,1H),2.17-2.00(m,2H),2.00-1.98(m,1H),1.92(d,J=12Hz,1H),1.82-1.44(m,10H),0.73(t,J=8Hz,3H).
Examples: 17B
RT:14.840min (U3000 (ThermoFisher); column: CHIRALPAK IF-3 4.6mm*150mm,3um; column temperature: 40 ℃ C.; mobile phase: n-hexane: ethanol-diethylamine) =85/15-0.1%).LCMS:m/z 649(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.96(s,1H),9.11(s,1H),7.77(q,J=4Hz,1H),7.38(d,J=8Hz,1H),7.34(s,1H),7.03(d,J=4Hz,1H),4.45(d,J=16Hz,2H),4.26-4.17(m,2H),3.65-3.55(m,4H),3.13(q,J=8Hz,1H),3.04-2.99(m,1H),2.74(d,J=12Hz,1H),2.57-2.54(m,1H),2.39-2.34(m,1H),2.17-2.00(m,2H),2.00-1.98(m,1H),1.92(d,J=12Hz,1H),1.80-1.44(m,10H),0.73(t,J=8Hz,3H).
Example 18:6- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyrido [4,3d ] pyrimidin-7-yl) -4-methyl-5- (trifluoromethyl) pyridin-2-amine
LCMS:m/z 635(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.01(s,1H),6.78(s,1H),4.36(d,J=9.20Hz,1H),4.17(m,2H),3.55(m,4H),3.09(m,1H),2.99(m,1H),2.70(m,2H),2.54(m,1H),2.36(m,3H),2.05(m,1H),1.97(m,1H),1.99(m,1H),1.79(m,2H),1.56(m,7H).
Example 19:6- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyrido [4,3d ] pyrimidin-7-yl) -4-methyl-5- (pentafluoroethyl) pyridin-2-amine
LCMS:m/z 685(M+H)+.
The following examples were synthesized following the procedure of example 1, starting from intermediate 1-11A-1:
Example 20:4- (2- ((2, 2-difluoro-1 ' h,3' h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' h) -yl) methoxy) -8-fluoro-4- (1-methyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoro naphthol-2-ol
LCMS:m/z 659(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.20(br,1H),9.06(s,1H),7.98(dd,J=5.88Hz,J=9.20Hz,1H),7.48(t,J=9.00Hz,1H),7.40(d,J=2.64Hz,1H),7.19(s,1H),4.30(m,4H),3.94(s,1H),3.58(m,2H),3.11(m,1H),3.02(m,1H),2.71(d,J=11.96Hz,1H),2.55(m,1H),2.06(m,2H),1.71(m,10H),1.25(m,5H).
Isomers 20A and 20B were obtained by chiral resolution (SFC-150 (Waters); column: OD 20X 250mm,10um (Daicel)): 4- (2- ((2, 2-difluoro-1 'h,3' h-spiro [ cyclopropan-1, 2 '-bis-fused pyrrolidino ] -7a' (5 'h) -yl) methoxy) -8-fluoro-4- ((1 s,5 r) -1-methyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthol-2-ol and 4- (2- ((2, 2-difluoro-dihydro-1' h,3 'h-spiro [ cyclopropan-1, 2' -bis-fused pyrrolidino ] -7a '(5' h) -yl) methoxy) -8-fluoro-4- ((1 r,5 s) -1-methyl-3, 8-diazabicyclo [3.2.1] oct-3-yl) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthol-2-ol
Isomer 20A
RT:0.905min.LCMS:m/z 659(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.18(s,1H),9.07(s,1H),8.01-7.97(m,1H),7.48(t,J=8Hz,1H),7.40(d,J=4Hz,1H),7.19(s,1H),4.44(t,J=12Hz,1H),4.35-4.15(m,3H),3.94(s,1H),3.60-3.57(m,2H),3.38(s,1H),3.13-3.09(m,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-1.99(m,2H),1.93(d,J=16Hz,1H),1.84-1.46(m,9H),1.38-1.32(m,1H),1.26(s,3H).
Isomer 20B
RT:1.419min.LCMS:m/z 659(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.20(s,1H),9.07(s,1H),8.01-7.97(m,1H),7.48(t,J=8Hz,1H),7.40(d,J=4Hz,1H),7.20(s,1H),4.49-4.35(m,2H),4.31(d,J=12Hz,1H),4.20(s,2H),3.95(s,1H),3.64-3.56(m,2H),3.43(d,J=16Hz,1H),3.13-3.09(m,1H),3.04-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-2.07(m,1H),2.02-1.99(m,1H),1.82-1.35(m,10H),1.29-1.24(m,3H).
Example 21:7a '- ((4- (1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -7- (8-ethynyl-7-fluoronaphthol-1-yl) -8-fluoropyridin [4,3-d ] pyrimidin-2-yl) oxy) methyl) -2, 2-difluorodihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ]
LC-MS:m/z 609(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),8.25(d,J=8Hz,1H),8.22(d,J=8Hz,1H),7.79-7.60(m,3H),4.51(d,J=12Hz,1H),4.38(d,J=12Hz,1H),4.24-4.19(m,2H),4.02(s,1H),3.67-3.62(m,4H),3.14-3.09(m,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-2.06(m,1H),2.02-1.90(m,2H),1.82-1.45(m,9H).
Example 22:7a '- ((4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-7- (6-fluoro-5-methyl-1H-indazol-4-yl) pyridin [4,3-d ] pyrimidin-2-yl) oxy) methyl) -2, 2-difluoro-dihydro-1' H,3 'H-spiro [ cyclopropane-1, 2' -bisfused pyrrolidine ]
LC-MS:m/z 609(M+H)+.1H NMR(400MHz,DMSO-d6)δ13.21(s,1H),9.20(s,1H),7.74(s,1H),7.50(d,J=8Hz,1H),4.47(s,2H),4.25-4.19(m,2H),3.64-3.58(m,3H),3.14(q,J=7Hz,J=5Hz,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,4H),2.19(s,2H),2.12-1.45(m,8H).
Example 23:4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -7-fluorobenzo [ d ] thiazol-2-amine
LC-MS:m/z 627(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),7.91(s,1H),7.42(dd,J=5.80,8.48Hz,1H),7.07(t,J=8.88Hz,1H),4.38(d,J=12.12Hz,2H),4.19(m,2H),3.57(m,4H),3.09(m,1H),3.01(m,1H),2.70(d,J=11.84Hz,1H),2.54(m,1H),2.08(m,1H),1.98(m,1H),1.88(d,J=13.32Hz,1H),1.63(m,9H).
Example 24:4- (4- ((1R, 5S) -diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidinyl ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -2-amino-7-fluorobenzene [ b ] thiophene-3-carbonitrile
LC-MS:m/z 651(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),8.10(s,2H),7.44-7.40(m,1H),7.15(t,J=8Hz,1H),4.43(d,J=12Hz,2H),4.23-4.17(m,2H),3.63(d,J=12Hz,2H),3.53(s,2H),3.13-3.09(m,1H),3.05-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.12-2.07(m,1H),2.03-1.97(m,1H),1.92-1.89(m,2H),1.86-1.71(m,2H),1.64-1.44(m,7H).
Example 25:2- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -4, 6-dichloro-3-fluoroaniline
LC-MS:m/z 638(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.18(s,1H),9.59(s,1H),9.36(s,1H),9.25(s,1H),7.71(d,J=7.56Hz,1H),5.76(s,2H),4.67(m,4H),4.21(m,2H),3.64(m,2H),3.13(m,1H),2.47(m,1H),2.28(m,4H),1.88(m,9H).
Example 26:3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -2-fluoro-5-methyl-4- (trifluoromethyl) aniline
LC-MS:m/z 652(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.09(s,1H),7.31(d,J=11.76Hz,1H),5.86(s,1H),4.42(d,J=11.40Hz,2H),4.19(d,J=3.24Hz,2H),3.61(m,4H),3.11(m,1H),3.02(m,1H),2.71(d,J=11.84Hz,1H),2.55(m,1H),2.17(s,3H),2.09(m,1H),2.00(m,1H),1.88(d,J=13.32Hz,1H),1.64(m,10H).
Example 27:3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((difluoro-dihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-chloro-4- (trifluoromethyl) aniline
LC-MS:m/z 654(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.06(s,1H),6.89(s,1H),6.48(s,1H),6.34(s,1H),4.39(d,J=12.08Hz,2H),4.20(m,2H),3.59(m,4H),3.10(m,1H),3.02(m,1H),2.71(d,J=11.88Hz,1H),2.56(m,1H),2.07(m,1H),1.99(m,1H),1.88(d,J=13.32Hz,1H),1.62(m,10H).
Example 28:4- (4- (3, 9-diazabicyclo [4.2.1] non-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LC-MS:m/z 659(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.20(s,1H),9.09(d,J=
16Hz,1H),8.00-7.95(m,1H),7.47(t,J=8Hz,1H),7.40(s,1H),7.22-7.19(m,1H),4.71(d,J=12Hz,1H),4.42(d,J=12Hz,1H),4.25-4.15(m,2H),4.09-3.99(m,2H),3.92-3.82(m,2H),3.73-3.67(m,2H),3.13-3.08(m,1H),3.04-3.00(m,1H),2.74(d,J=12Hz,1H),2.57-2.55(m,1H),2.11-1.95(m,3H),1.92-1.89(m,3H),1.84-1.72(m,3H),1.65-1.45(m,5H).
Example 29:4- (4- (3-amino-3-methylpiperidin-1-yl) -2- ((2, 2-difluorodihydro-1 ' h,3' h-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' h) -yl) methoxy) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LC-MS:m/z 647(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.16(s,2H),9.25(d,J=
56Hz,1H),7.99-7.96(m,1H),7.46(t,J=8Hz,1H),7.40(d,J=4Hz,1H),7.22-7.20(m,1H),4.24-4.16(m,2H),4.10(m,1H),4.03-4.00(d,J=12Hz,1H),3.95(s,1H),3.89(d,J=12Hz,1H),3.73-3.65(m,1H),3.60-3.47(m,2H),3.13-2.99(m,2H),2.73(d,J=12Hz,1H),2.56-2.54(m,1H),2.10-2.05(m,1H),2.02-1.97(m,2H),1.92(s,1H),1.89(s,1H),1.83-1.74(m,3H),1.64-1.44(m,5H),1.10(d,J=4Hz,3H).
Example 30 3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-methoxypyridine [4,3-d ] pyrimidin-7-yl) -2-fluoro-5-methyl-4- (trifluoromethyl) aniline
LC-MS:m/z 664(M+H)+.1H NMR(400MHz,DMSO-d6)δ8.96(s,1H),7.18(d,J=11.68Hz,1H),5.67(s,2H),4.35(d,J=12.40Hz,2H),4.20(m,2H),3.85(s,3H),3.53(m,4H),3.11(m,1H),3.02(m,1H),2.71(m,1H),1.73(m,17H).
Example 31:3- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -8-methoxypyridine [4,3-d ] pyrimidin-7-yl) -5-chloro-4- (trifluoromethyl) aniline
LC-MS:m/z 666(M+H)+
The following examples were synthesized following the procedure of example 1, starting from intermediate D-1-11A-1:
Example D01:4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl-5 ',5' -d 2) methoxy-d 2) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LC-MS:m/z 649(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.05(s,1H),7.98(dd,J=5.92,9.24Hz,1H),7.47(t,J=9.04Hz,1H),7.40(d,J=2.60Hz,1H),7.18(d,J=2.64Hz,1H),4.47(d,J=12.36Hz,1H),4.31(m,1H),3.94(s,1H),3.63(m,4H),3.29(m,1H),3.10(m,1H),2.71(d,J=11.72Hz,1H),2.07(m,1H),1.99(m,1H),1.89(m,1H),1.67(m,1H).
The following examples were synthesized following the procedure of example 1, starting from intermediate D-1-11A-2:
Example D02:4- (4- ((1R, 5S) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl-5 ',5' -d 2) methoxy-d 2) -8-fluoropyridine [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LC-MS:m/z 649(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.15(s,1H),9.05(s,1H),7.98(dd,J=5.96,9.20Hz,1H),7.47(t,J=9.04Hz,1H),7.40(d,J=2.60Hz,1H),7.18(d,J=2.56Hz,1H),4.47(d,J=12.20Hz,1H),4.32(m,1H),3.94(s,1H),3.63(m,4H),3.29(m,1H),3.10(m,1H),2.71(d,J=11.72Hz,1H),2.07(m,1H),1.99(m,1H),1.89(m,1H),1.67(m,1H).
The following examples were synthesized following the procedure of example 1, starting from intermediates 1-17:
Example E01:4- (4- ((1 r,5 s) -3, 8-diazabicyclo [3.2.1] oct-3-yl) -8-fluoro-2- ((1-methyltetrahydro-1H-spiro [ cyclopentane [ b ] pyrrole-2, 1' -cyclopropane ] -3a (3H) -yl) methoxy) pyridin [4,3-d ] pyrimidin-7-yl) -5-ethynyl-6-fluoronaphthalen-2-ol
LC-MS:m/z 623(M+H)+
Example P01: preparation of((1R, 5S) -3- (2- ((2, 2-difluoro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -bis-fused pyrrolidino ] -7a ' (5 ' H) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] oct-8-yl) methyl isobutyrate trifluoroacetate
Example 16B-1 (100 mg,0.155 mmol) was dissolved in THF (5 mL) and 2, 6-lutidine (83 mg,0.775 mmol) and iodomethyl isobutyrate (189 mg,0.775 mmol) were added sequentially. After the addition was completed, the mixture was stirred at room temperature for 16 hours, and then acetonitrile was added to dissolve the mixture, followed by direct separation by preparative HPLC to obtain the objective product (28 mg).
LC-MS:m/z 745(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.28(s,1H),9.45(d,J=9.64Hz,1H),9.19(m,2H),8.00(dd,J=5.96,9.20Hz,1H),7.49(t,J=9.04Hz,1H),7.43(d,J=2.52Hz,1H),7.18(d,J=2.60Hz,1H),5.59(m,2H),4.95(s,2H),4.72(m,1H),4.56(m,1H),4.25(s,2H),4.12(m,1H),3.88(m,5H),3.73(m,2H),2.70(m,2H),2.40(m,2H),2.22(m,3H),1.98(m,7H),1.15(m,6H).
The following compounds were synthesized in the same manner as in example P01:
Example P02: (1R, 5S) -3- (2- ((2, 2-Difluorodihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -pyrrolizine ] -7a ' (5 ' H) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylic acid isopropyl ester
LC-MS:m/z 731(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.07(s,1H),7.97(dd,J=5.96,
9.24Hz,1H),7.46(t,J=9.04Hz,1H),7.39(d,J=2.56Hz,1H),7.18(d,J=2.48Hz,1H),4.87(m,1H),4.56(m,1H),4.41(m,3H),4.20(m,2H),3.92(s,1H),3.64(m,2H),3.10(m,1H),3.01(m,1H),2.70(d,J=11.84Hz,1H),2.54(m,1H),2.06(m,1H),1.99(m,1H),1.81(m,7H),1.61(m,3H),1.24(d,J=6.48Hz,6H).
Example P03: ((1R, 5S) -3- (2- ((2, 2-Difluorodihydro-1 ' H,3' H-spiro [ cyclopropan-1, 2' -pyrrolizine ] -7a ' (5 ' H) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] oct-8-yl) methyl isopropyl carbonate
LC-MS:m/z 761(M+H)+.1H NMR(400MHz,DMSO-d6)δ10.28(s,1H),9.57(s,1H),9.29(s,1H),9.19(s,1H),8.01(t,J=8Hz,1H),7.52-7.44(m,2H),7.19(s,1H),5.65-5.56(m,2H),4.95(s,2H),4.87-4.74(m,2H),4.60(d,J=12Hz,1H),4.25(s,2H),4.18(d,J=12Hz,1H),3.93-3.86(m,5H),3.77-3.74(m,2H),2.70-2.66(m,2H),2.60(s,1H),2.43-2.34(m,2H),2.25-2.18(m,3H),1.99-1.90(m,6H),1.25-1.24(m,6H).
Example P04 and example P05: preparation of 1- (isobutyryloxy) ethyl (1 r,5 s) -3- (2- ((2, 2-difluorodihydro-1 'h,3' h-spiro [ cyclopropane-1, 2 '-pyrrolizine ] -7a' (5 'h) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3- (((1- (isobutyryloxy) ethoxy) carbonyl) oxy) naphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate and 1- (isobutyryloxy) ethyl (1 r,5 s) -3- (2- ((2, 2-difluorodihydro-1' h,3 'h-spiro [ cyclopropane-1, 2' -pyrrolizine ] -7a '(5' h) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate
The first step: preparation of 1- (isobutyryloxy) ethyl (1R, 5S) -3- (2- ((2, 2-difluorodihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -pyrrolizine ] -7a ' (5 ' H) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3- (((1- (isobutyryloxy) ethoxy) carbonyl) oxy) naphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate
Example 16B-1 (100 mg,0.155 mmol) was dissolved in DCM (4 mL), DIPEA (100 mg,0.775 mmol) was added, ethyl 1- (((4-nitrophenoxy) carbonyl) oxy) isobutyrate (184 mg,0.62 mmol) was added and after the addition was complete stirring at room temperature for 16h. After the reaction, the reaction mixture was concentrated to dryness, and after addition of acetonitrile, the mixture was directly separated by preparative HPLC to give the objective product (65 mg).
LC-MS:m/z 961(M+H)+.1H NMR(400MHz,DMSO-d6)δ9.08(s,1H),8.25(dd,J=5.92Hz,9.28Hz,1H),8.15(d,J=2.52Hz,1H),7.68(t,J=9.00Hz,1H),7.62(m,1H),6.75(m,2H),4.46(m,4H),4.20(m,2H),4.08(s,1H),3.68(m,2H),3.09(m,1H),3.00(m,1H),2.07(d,J=11.84Hz,1H),2.61(m,3H),2.09(m,1H),1.99(m,1H),1.85(m,7H),1.57(m,9H),1.09(m,12H).
The first step: preparation of 1- (isobutyryloxy) ethyl (1R, 5S) -3- (2- ((2, 2-difluorodihydro-1 ' H,3' H-spiro [ cyclopropane-1, 2' -pyrrolizine ] -7a ' (5 ' H) -yl) methoxy) -7- (8-ethynyl-7-fluoro-3-hydroxynaphthalen-1-yl) -8-fluoropyrido [4,3-d ] pyrimidin-4-yl) -3, 8-diazabicyclo [3.2.1] octane-8-carboxylate
Example P04 (105 mg,0.109 mmol) was dissolved in acetonitrile (4 mL), and aqueous ammonia (10 drops) was added thereto, followed by stirring at room temperature for 2 hours. After the reaction, the reaction mixture was concentrated to dryness, and after addition of acetonitrile, the mixture was directly separated by preparative HPLC to give the objective product (65 mg).
LC-MS:m/z 803(M+H)+.1H NMR(400MHz,DMSO-d6):10.16(s,1H),9.06(s,1H),7.98(dd,J=5.92,9.24Hz,1H),7.47(t,J=9.00Hz,1H),7.40(d,J=2.60Hz,1H),7.18(d,J=2.52Hz,1H),6.74(m,1H),4.59(m,1H),4.39(m,3H),4.22(m,2H),3.93(s,1H),3.67(m,2H),3.11(m,2H),2.71(d,J=11.84Hz,1H),2.55(m,1H),1.98(m,10H),1.56(m,6H),1.08(d,J=7.16Hz,6H).
Biological test case
The following biological test examples further illustrate the invention, but these examples are not meant to limit the scope of the invention.
KRAS Gl2D binding inhibition assay
Experimental procedure
The binding capacity of the compounds to KRAS G12D protein was tested using TR-FRET technology. The biotin-labeled GDP-loaded KRAS G12D recombinant human protein was co-incubated with Cy 5-labeled tracer, europium-labeled streptavidin, compound (2% DMSO final concentration) in buffer (HEPES (pH 7.5), mgCl 2, tween-20 and DTT). After incubation at 22℃for 60min, the reactivity was measured by the EnVision multifunctional microplate reader dual wavelength technique and the protein binding capacity (POC) was calculated using ratiometric emission factor.
100POC represents no compound; 0POC indicates that the control compound completely inhibited the binding of the tracer to KRAS G12D at this concentration. POC values were curve fitted using a four parameter Logistic model, IC 50 representing a 50POC concentration value.
The results show that the compounds of the examples of the present invention show good inhibitory activity against KRAS G12D.
ERK phosphorylation inhibition assay
Experimental procedure
KRAS G12D mutant cells (e.g., GP2D, AGS, etc.) were seeded in 384 well plates and incubated overnight in a 5% CO2 incubator at 37 ℃.
200NL of diluted compound was added to Echo 500, and the mixture was incubated in a 5% CO2 incubator at 37℃for 1 hour at a final DMSO concentration of 0.5%. hEGF was added for 10 min.
Removing the culture medium, adding cell fixing solution, and fixing cells
PBS was washed 1 time, incubated with cold 100% methanol,
Methanol was removed and washed 1 time with PBS.
PBS was removed, li-Cor blocking buffer was added to each well and blocked at room temperature for 1hr.
The blocking solution was removed, primary antibody mix was added to each well and incubated overnight at 4℃at room temperature.
The primary antibody mixture was removed and washed 3 times with PBST.
Adding the secondary antibody mixed solution, and incubating for 45min at room temperature in a dark place.
And removing the secondary antibody mixture, adding PBST for washing for 3 times, finally sucking out the PBST, reversely buckling and centrifuging, and centrifuging at 1000rpm for 1min.
Odyssey CLx readings. The test results are shown in tables 1 and 2 below.
Reference compound MRTX1133 had the following structure:
TABLE 1 GP2D ERK phosphorylation inhibition assay results
Examples IC50(nM)
Example 1 isomer 1A 2.6
Example 1 isomer 1B 2.1
TABLE 2 AGS ERK phosphorylation inhibition test results
Examples IC50(nM)
Example 1 isomer 1A 9.67
Example 1 isomer 1B 3.76
Example 1 isomer 2A 19
Example 1 isomer 2B 139
Example 9 8.2
Example 16A 10.6
Example 16B 3.34
The results show that the compound of the embodiment of the invention shows good inhibition activity on ERK phosphorylation of KRAS G12D mutant cells.
Inhibition experiments of compounds on proliferation of KRAS G12D mutant cells
Experimental procedure
1. Cell culture
(A) Resuscitates cells in T75 cell culture flasks:
(b) When the cell fusion degree reaches 80-90%, the cells are passaged.
2. Cell proliferation assay
Experimental procedure
And adding the diluted compound to be tested into a 384-well cell culture plate by utilizing a nanoliter pipetting system, and arranging multiple wells. Adding the positive control group into an equal volume of culture medium; the negative control group was added with an equal volume of DMSO and centrifuged at 1000rpm for 1min at room temperature.
Cells were inoculated into a) 384 plates, negative control groups were added with equal volumes of cells, positive control groups were added with equal volumes of medium only. Centrifugation at 1000rpm at room temperature for 1min, final compound DMSO concentration of 0.5%, and incubation in a 5% CO2 incubator at 37℃for 7 days.
By adding 20. Mu.L/well3D to b) 384-well cell culture plates, shake for 20min at 320rpm in the dark and incubate for 2hrs at room temperature in the dark.
And (5) reading the luminescence value by using an Envision multifunctional enzyme-labeled instrument.
3. Data analysis
The Inhibition Rate (IR) of the test compound was calculated using the following formula: IR (%) = (1- (RLU compound-RLU placebo)/(RLU vehicle control-RLU placebo)) × 100%. Inhibition rates of compounds at different concentrations were calculated in Excel, and then GRAPHPAD PRISM software was used for inhibition graphs and to calculate relevant parameters, including minimum inhibition, maximum inhibition and IC 50. The experimental results are shown in table 3.
TABLE 3 cell proliferation inhibitory Activity of the inventive example Compounds
Pharmacokinetic test evaluation
Male ICR mice, weighing 22-24g, were orally administered 30mg/kg of a solution of the compound of the invention or control compound [10% DMSO+60% PEG400+30% in water ] after overnight fast. Blood was collected 0.25,0.5,1.0,2.0,4.0,6.0,8.0, 12.0 and 24 hours after administration of the compounds of the invention, respectively, and the concentration of the compounds of the invention or control compounds in the plasma was determined by LC/MS.
TABLE 4 principal pharmacokinetic parameters in plasma
BLOQ below the minimum detection limit; NA: not obtained
From the detection results, the compound has good oral pharmacokinetic properties.
Pharmacokinetic test evaluation
Male SD rats weighing around 220g were orally administered 30mg/kg of a solution of the compound of the invention or a control compound [10% DMSO+60% PEG400+30% in water ] after overnight fast. Blood was collected 0.25,0.5,1.0,2.0,4.0,6.0,8.0, 12.0 and 24 hours after administration of the compounds of the invention, respectively, and the concentration of the compounds of the invention or control compounds in the plasma was determined by LC/MS.
From the detection results, the compound has good pharmacokinetic properties.
Evaluation of antitumor Activity pharmacodynamics test (AsPC-1 CDX tumor model)
100UL of 5x10 6 AsPC-1 tumor cell-containing suspension was inoculated subcutaneously into the right posterior flank of nude mice. Mice were monitored daily for health and measurements were started when tumors grew to be accessible. The calculation formula of the tumor volume adopts 0.5xLxW 2, wherein L, W respectively represents the length and width of the tumor. Tumors were grown to 200mm 3 and mice were randomly grouped. Mice were given daily intraperitoneal injections or orally with the corresponding dose of compound, while monitoring their general status. Tumors were measured 3 times per week and body weights were measured twice per week.
The detection result shows that the compound has good anti-tumor effect.
All documents mentioned in this disclosure are incorporated by reference in this disclosure as if each were individually incorporated by reference. Further, it will be appreciated that various changes and modifications may be made by those skilled in the art after reading the above teachings, and such equivalents are intended to fall within the scope of the application as defined in the appended claims.

Claims (13)

1. A compound of formula (A0), a stereoisomer, tautomer, crystalline form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof:
Ring C is selected from the group consisting of:
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
A group selected from the group consisting of: wherein X is selected from: n, CH, CD, CF, C (CN);
R 1 is selected from: -L 1-Q-L2-L3;
wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 0, 1,2,3, 4, 5 or 6; with the proviso that when W is monocyclic or bicyclic, n is not 0;
R 10 is selected from the group consisting of substituted or unsubstituted: c 6-C14 aryl, 5-14 membered heteroaryl; wherein the substitution refers to substitution with one or more Ra;
R 11 are each independently selected from the group consisting of substituted or unsubstituted: H. deuterium, halogen, cyano, ester, amine, amide, sulfone, ureido, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C6 alkyloxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
Ra are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C20 cycloalkyl, 4-20 membered heterocyclyl, C 3-C20 cycloalkyloxy, 4-20 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
m is an integer of 0, 1,2,3,4, 5 or 6;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, 4-20 membered heterocyclyl C (O), C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
2. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to claim 1, having a structure according to formula (a):
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
A group selected from the group consisting of: wherein X is selected from: n, CH, CD, CF, C (CN);
R 1 is selected from: -L 1-Q-L2-L3;
wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 0, 1,2,3, 4, 5 or 6; with the proviso that when W is monocyclic or bicyclic, n is not 0;
R 10 is selected from the group consisting of substituted or unsubstituted: c 6-C14 aryl, 5-14 membered heteroaryl; wherein the substitution refers to substitution with one or more Ra;
R 11 are each independently selected from the group consisting of substituted or unsubstituted: H. deuterium, halogen, cyano, ester, amine, amide, sulfone, ureido, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C6 alkyloxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy;
Ra are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy, C 2-C6 alkenyl, C 2-C6 alkynyl, C 3-C20 cycloalkyl, 4-20 membered heterocyclyl, C 3-C20 cycloalkyloxy, 4-20 membered heterocyclyloxy; wherein the substitution refers to substitution with one or more R;
m is an integer of 0, 1,2,3,4, 5 or 6;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, 4-20 membered heterocyclyl C (O), C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
3. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 2, having a structure according to formula (IA) or formula (IB):
in the method, in the process of the invention,
U is selected from: n, CH, CD, CF;
U' is selected from: o, or S;
U' is selected from: n, or C (CN);
X is selected from: n, CH, CD, CF, C (CN);
y is selected from: bond, O, NH, N (C 1-C3 alkyl);
Z is a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
W is selected from: a substituted or unsubstituted C 3-C14 cycloalkyl group, or a substituted or unsubstituted 4-14 membered saturated or unsaturated heterocyclyl group; wherein the substitution refers to substitution with one or more R;
R 1 is selected from: -L 1-Q-L2-L3; wherein:
l 1 is selected from: a substituted or unsubstituted C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
Q is selected from: o, S, SO 2, NH, or N (C 1-C3 alkyl);
L 2 is selected from: an unsubstituted, or substituted or unsubstituted, C 1-C6 alkylene group; wherein the substitution refers to substitution with one or more R;
L 3 is selected from the group consisting of substituted or unsubstituted: -C 1-C6 alkyl, -C 3-C6 cycloalkyl, -C 4-C6 heterocyclyl, -C 1-C6 alkylene (C 3-C6 cycloalkyl), -C 1-C6 alkylene (C 4-C6 heterocyclyl), -C 1-C6 alkylene (C 1-C6 alkoxy), -C 1-C6 alkylene (C 3-C6 cycloalkyloxy), or-C 1-C6 alkylene (C 4-C6 heterocyclyloxy); wherein the substitution refers to substitution with one or more R;
n is an integer of 1, 2, 3, 4, 5 or 6;
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 4 is selected from the group consisting of substituted or unsubstituted: halogen, C 1-C6 alkyl, C 2-C6 alkenyl, C 2-C6 alkynyl; wherein the substitution refers to substitution with one or more R;
R 8 is selected from: OH, SONH 2、NHSO2CH3;
R 5、R6、R7、R9、R7'、R8'、R9' are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
R 5' is selected from: H. d, halogen, CN, NH 2、OH、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SONH 2、SO2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), C 1-C6 alkyl, C 1-C6 deuteroalkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy;
Each R, which may be the same or different, is independently selected from: deuterium, C 1-C18 alkyl, deuterated C 1-C18 alkyl, halogenated C 1-C18 alkyl, (C 3-C18 cycloalkyl) C 1-C18 alkyl, (4-20 membered heterocyclyl) C 1-C18 alkyl, (C 1-C18 alkoxy) C 1-C18 alkyl, (C 3-C18 cycloalkyloxy) C 1-C18 alkyl, (4-20 membered heterocyclyloxy) C 1-C18 alkyl, vinyl, ethynyl, (C 1-C6 alkyl) vinyl, deuterated (C 1-C6 alkyl) vinyl, halogenated (C 1-C6 alkyl) vinyl, (C 1-C6 alkyl) ethynyl, deuterated (C 1-C6 alkyl) ethynyl, halogenated (C 1-C6 alkyl) ethynyl, (C 3-C14 cycloalkyl) ethynyl, and, (4-14 membered heterocyclyl) ethynyl, C 1-C18 alkoxy, deuterated C 1-C18 alkoxy, halogenated C 1-C18 alkoxy, C 3-C20 cycloalkyl, A 4-20 membered heterocyclic group, a C 6-C14 aryl group, a 5-14 membered heteroaryl group, a halogen, a nitro group, a hydroxyl group, an oxo group, a cyano group, an ester group, an amine group, an amide group, a sulfonamide group, a sulfone group or a urea group.
4. A compound according to any one of claims 1 to 3, a stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof, having a structure according to formula (IIA) or formula (IIB):
Wherein ,R11、R2、R3、R4、R5、R6、R7、R8、R9、R5'、R7'、R8'、R9'、U、U'、U"、X、Z、W and n are as defined in claim 3.
5. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 4, having a structure according to formula (VA) or formula (VB):
wherein ,R1、R4、R5、R6、R7、R9、R7'、R8'、R9'、U、U'、U"、Z、W and n are as defined in claim 3.
6. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 5, having a structure according to formula (VIA) or formula (VIB):
Wherein ,R1、R4、R5、R6、R7、R9、R7'、R8'、R9'、U"、Z、W and n are as defined in claim 3.
7. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 6, having a structure according to formula (VIIA) or formula (VIIB):
Wherein R 1、R4、R5、R7'、R8'、R9', Z, W and n are as defined in claim 3.
8. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 7, having a structure according to formula (VIIIA) or formula (VIIIB):
Wherein R 1、R4、R5、R7'、R9', Z, W and n are as defined in claim 3.
9. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to claim 1, having a structure according to formula (IXA) or formula (IXB):
Wherein n' is an integer of 0, 1, 2, 3, 4, 5, or 6;
R 5、R、R7、R9、R7'、R9'、L1、Q、L2、L3 and n are as defined in claim 3.
10. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to claim 1, having a structure according to formula (X):
Wherein,
V 1、V2、V3、V4 and V 5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
The definition of rings C, R, R 1, X, Z, W and n is as defined in claim 1;
preferably, the compound has a structure represented by formula (XI):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 2 and R 3 are identical or different and are each independently selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
wherein R, R 1, X, Z, W and n are as defined in claim 1;
preferably, the compound has a structure represented by formula (XII):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 3 is selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
wherein R, R 1, X, Z, W and n are as defined in claim 1;
preferably, the compound has a structure represented by formula (XIII):
V 1、V2、V3、V4、V5 are each independently selected from: n, or CR v;Rv are the same or different and are each independently selected from: H. c 1-C3 alkyl, C 3-C6 cycloalkyl, 4-6 membered heterocyclyl, C 1-C3 alkoxy, C 3-C6 cycloalkyloxy, 4-6 membered heterocyclyloxy, halogenated C 1-C3 alkyl, halogenated C 3-C6 cycloalkyl, halogenated 4-6 membered heterocyclyl, (HO) -C 1-C3 alkyl, (HO) -C 3-C6 cycloalkyl, (NH 2)-C1-C3 alkyl, (NH 2)-C3-C6 cycloalkyl, halogen, CN, -c≡ch, OH, NH 2、CONH2、NHCO(C1-C6 alkyl), NHCO (C 3-C6 cycloalkyl), SO 2NH2、NHSO2(C1-C6 alkyl), NHSO 2(C3-C6 cycloalkyl), wherein the heterocyclyl is optionally substituted with one or more oxo (=o);
R 3 is selected from the group consisting of substituted or unsubstituted: H. d, halogen, CN, C 1-C6 alkyl, C 1-C6 deuterated alkyl, C 1-C6 haloalkyl, C 1-C6 alkoxy; wherein the substitution refers to substitution with one or more R;
Wherein R, R 1, Z, W and n are as defined in claim 1.
11. The compound, stereoisomer, tautomer, crystal form, pharmaceutically acceptable salt, hydrate, solvate, or prodrug thereof according to any one of claims 1 to 10, wherein the compound is selected from the group consisting of:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
Or is selected from:
12. a pharmaceutical composition comprising one or more compounds according to any one of claims 1-11, stereoisomers, tautomers, crystalline forms, pharmaceutically acceptable salts, hydrates, solvates, or prodrugs thereof; and a pharmaceutically acceptable carrier.
13. Use of a compound according to any one of claims 1-11, a stereoisomer, a tautomer, a crystal, a pharmaceutically acceptable salt, a hydrate, a solvate, or a prodrug thereof, or a pharmaceutical composition according to claim 12, for the preparation of a medicament for the prophylaxis and/or treatment of a disease associated with the activity or expression of KRAS G12D.
CN202310059893.6A 2022-03-22 2023-01-13 Substituted bridged ring inhibitors and preparation method and application thereof Pending CN118344386A (en)

Priority Applications (5)

Application Number Priority Date Filing Date Title
CN202310059893.6A CN118344386A (en) 2023-01-13 2023-01-13 Substituted bridged ring inhibitors and preparation method and application thereof
PCT/CN2023/082908 WO2023179629A1 (en) 2022-03-22 2023-03-21 Substituted bridged ring inhibitor, preparation method therefor and application thereof
EP23773876.0A EP4497751A1 (en) 2022-03-22 2023-03-21 Substituted bridged ring inhibitor, preparation method therefor and application thereof
CN202380029714.2A CN119317628A (en) 2022-03-22 2023-03-21 Substituted bridge ring inhibitors and preparation method and application thereof
PCT/CN2024/072124 WO2024149389A1 (en) 2023-01-13 2024-01-12 Substituted bridge ring inhibitor, preparation method therefor, and application thereof

Applications Claiming Priority (1)

Application Number Priority Date Filing Date Title
CN202310059893.6A CN118344386A (en) 2023-01-13 2023-01-13 Substituted bridged ring inhibitors and preparation method and application thereof

Publications (1)

Publication Number Publication Date
CN118344386A true CN118344386A (en) 2024-07-16

Family

ID=91816605

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202310059893.6A Pending CN118344386A (en) 2022-03-22 2023-01-13 Substituted bridged ring inhibitors and preparation method and application thereof

Country Status (2)

Country Link
CN (1) CN118344386A (en)
WO (1) WO2024149389A1 (en)

Family Cites Families (7)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
AU2022232460A1 (en) * 2021-03-12 2023-09-14 Bristol-Myers Squibb Company Kras inhibitors
BR112023017763A2 (en) * 2021-03-12 2023-10-03 Bristol Myers Squibb Co KRAS G12D INHIBITORS
EP4332105A1 (en) * 2021-04-30 2024-03-06 Genfleet Therapeutics (Shanghai) Inc. Pyridino- or pyrimido-cyclic compound, preparation method therefor and medical use thereof
WO2023072188A1 (en) * 2021-10-29 2023-05-04 贝达药业股份有限公司 Kras g12d inhibitors and use thereof in medicine
WO2023125989A1 (en) * 2021-12-31 2023-07-06 上海医药集团股份有限公司 Quinazoline compound and application thereof
CN118556063A (en) * 2022-01-21 2024-08-27 上海湃隆生物科技有限公司 Heterocyclic compound, pharmaceutical composition and application thereof
EP4497751A1 (en) * 2022-03-22 2025-01-29 Suzhou Zelgen Biopharmaceuticals Co., Ltd. Substituted bridged ring inhibitor, preparation method therefor and application thereof

Also Published As

Publication number Publication date
WO2024149389A1 (en) 2024-07-18

Similar Documents

Publication Publication Date Title
CN115192577B (en) KRAS mutein inhibitors
JP7626772B2 (en) KRAS G12C inhibitors
JP7073359B2 (en) Piperidine derivative as an inhibitor of ubiquitin-specific protease 7
CN119317628A (en) Substituted bridge ring inhibitors and preparation method and application thereof
ES2744636T3 (en) 4,5,6,7-Tetrahydropyrazolo [1,5-a] pyrazine derivatives substituted as casein kinase 1 D / E inhibitors
WO2021248090A1 (en) Heterocyclic compounds and methods of use thereof
IL262345A (en) Inhibitors of activin receptor-like kinase
JP2021523104A (en) Pyridadinone as a PARP7 inhibitor
AU2009257434B2 (en) Diamino-pyridine, pyrimidine, and pyridazine modulators of the histamine H4 receptor
AU2013290054A1 (en) Imidazotriazinecarbonitriles useful as kinase inhibitors
US11098060B2 (en) Macrocycle containing aminopyrazole and pyrimidine and pharmaceutical composition and use thereof
JP6110787B2 (en) Pyrimidodiazepinone compounds
CN117327103A (en) Substituted pyrimido ring inhibitor and preparation method and application thereof
JP2020510083A (en) CDK4 / 6 inhibitor
CN117917416A (en) KRAS G12D degradation agent and preparation method and application thereof
IL266312A (en) Pyrido[3, 4-d]pyrimidine derivative and pharmaceutically acceptable salt thereof
CN118344386A (en) Substituted bridged ring inhibitors and preparation method and application thereof
CA3214088A1 (en) Pkc-theta modulators
WO2021167495A1 (en) Pyrimidine-based bicycles as antiviral agents for the treatment and prevention of hiv infection
JP2022553261A (en) Compounds and compositions for the treatment of parasitic diseases
CN117659049A (en) Substituted bridged ring inhibitors and preparation method and application thereof
CN119119085A (en) Substituted pyrimidocyclic inhibitors and preparation method and application thereof
RU2779497C2 (en) Macrocycle containing aminopyrazole and pyrimidine, and its pharmaceutical composition and use
US20240262821A1 (en) PKC-Theta Modulators
WO2024238221A1 (en) Substituted quinoxalines

Legal Events

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