WO2018082567A1 - 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂 - Google Patents

一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂 Download PDF

Info

Publication number
WO2018082567A1
WO2018082567A1 PCT/CN2017/108941 CN2017108941W WO2018082567A1 WO 2018082567 A1 WO2018082567 A1 WO 2018082567A1 CN 2017108941 W CN2017108941 W CN 2017108941W WO 2018082567 A1 WO2018082567 A1 WO 2018082567A1
Authority
WO
WIPO (PCT)
Prior art keywords
spiro
imidazo
cyclobutane
isoindole
trans
Prior art date
Application number
PCT/CN2017/108941
Other languages
English (en)
French (fr)
Inventor
王召印
郭巍
朱继东
胡新波
Original Assignee
中国科学院上海有机化学研究所
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 中国科学院上海有机化学研究所 filed Critical 中国科学院上海有机化学研究所
Priority to CN201780064706.6A priority Critical patent/CN109890826A/zh
Publication of WO2018082567A1 publication Critical patent/WO2018082567A1/zh

Links

Classifications

    • 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
    • C07D487/10Spiro-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41641,3-Diazoles
    • A61K31/41881,3-Diazoles condensed with other heterocyclic ring systems, e.g. biotin, sorbinil
    • 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
    • A61K31/41Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having five-membered rings with two or more ring hetero atoms, at least one of which being nitrogen, e.g. tetrazole
    • A61K31/41921,2,3-Triazoles
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/44Non condensed pyridines; Hydrogenated derivatives thereof
    • A61K31/4427Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems
    • A61K31/4439Non condensed pyridines; Hydrogenated derivatives thereof containing further heterocyclic ring systems containing a five-membered ring with nitrogen as a ring hetero atom, e.g. omeprazole
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07BGENERAL METHODS OF ORGANIC CHEMISTRY; APPARATUS THEREFOR
    • C07B2200/00Indexing scheme relating to specific properties of organic compounds
    • C07B2200/07Optical isomers

Definitions

  • the invention belongs to the technical field of medicinal chemistry, and particularly relates to a high-efficiency IDO/TDO inhibitor containing a nitrogen heterocyclic helix structure and a preparation method thereof.
  • Indoleamine-2,3-dioxygenase is a monomeric enzyme containing heme found in the cell for the first time in 1967 by the Hayaishi group.
  • the cDNA encodes a protein. 403 amino acid composition with a molecular weight of 45 kDa, which is the rate-limiting enzyme of the leucine-kynurenine pathway catabolism and has extensive expression in a variety of mammalian tissues (Hayaishi O. et al Science, 1969, 164, 389-396).
  • IDO In tumor cells, IDO often plays an important physiological role in inducing tumor microenvironmental immune tolerance, and its mediated tryptophan (Trp)-kynurenine (Kyn) metabolic pathway is involved in tumors. Immune escape, and IDO also plays an important role as an immune tolerance to induce tumor microenvironment.
  • Tryptophan is one of the important essential amino acids in mammals. It needs to be ingested in large quantities from food, maintain cell activation and proliferation, and synthesize proteins and some neurotransmitters. Therefore, its lack of dysfunction can lead to some important cells. IDO can catalyze the conversion of tryptophan to N-formyl kynurenine in vivo, reduce the content of tryptophan and cause deficiency of tryptophan in vivo, leading to tumor formation. Immunohistological studies have shown that the kynurenine pathway can lead to an increase in the excitotoxic quinolinic acid, as well as many serious human diseases such as Alzheimer's and other neurological diseases (Guillemin GJet al Neuropathol. and Appl. Neurobiol. 2005, 31, 395–404).
  • IDO can inhibit local T cell immune responses in the tumor microenvironment by: tryptophan depletion, toxic metabolism, and induction of regulatory T cell proliferation. In many cases, it is overexpressed in tumors, thereby consuming local tryptophan and producing a large amount of metabolites such as kynurenine. In fact, in the absence of tryptophan or kynurenine culture conditions, T cells undergo proliferation inhibition, decreased activity, and even apoptosis. There is a regulatory point in T cells that is very sensitive to tryptophan levels. IDO can consume tryptophan, which leads to T cell arrest in the middle of G1 phase, which inhibits T cell proliferation and inhibits T cell immunity. Answer.
  • T cells Once T cells stop proliferating, they may not be stimulated any more. This is the mechanism of IDO immunity in vivo (Mellor A. et al Biochem. Biophys. Res. Commun. 2005, 338(1): 20-24). LeRond S. et al J. Exp. Med. 2002, 196(4): 447-457).
  • TDO is another tryptophan metabolizing enzyme that acts in the same way as IDO, except that it was originally thought to be endogenous, especially in normal liver and brain cells, especially in the liver. It has been demonstrated that TDO is overexpressed in a variety of cancer patient tissues, and that TDO inhibitors have been validated by immunological rejection of tumor cell growth in a mouse tumor model (Pilotte, l. et al. Proc. Natl. Acad. Sci.USA; Published online, Jan. 30, 2012).
  • IDO, TDO inhibitors or IDO/TDO double inhibitors are important targets for tumor immunotherapy, and there is a need in the art to develop highly active novel IDO, TDO or IDO/TDO inhibitors.
  • the object of the present invention is to provide a novel nitrogen-heterocyclic helix-containing compound which can be used as a highly efficient IDO/TDO double inhibitor, and a preparation method thereof, and the compound of the formula (I) of the present invention has antitumor and therapeutic nerves.
  • Degenerative diseases Alzheimer's disease
  • anti-inflammatory and other pharmacological activities are examples of pharmacological activities.
  • the first aspect of the invention provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, or a tautomer thereof, or a prodrug thereof:
  • A is selected from the group consisting of S, O, -C(R 1 ) 2 - or none;
  • B is selected from the group consisting of -C(R 1a ) 2 - or none;
  • E and M are each independently selected from: N or CR 1 ;
  • Each R 1 is independently selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 8 cycloalkyl, OH, substituted or unsubstituted OC 1 -C a 6 alkyl group, a substituted or unsubstituted OC 3 -C 8 cycloalkyl group;
  • Each R 1a is independently selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 8 cycloalkyl;
  • the substitution means having one or more (e.g., 1, 2, 3, 4) substituents selected from the group A: halogen, hydroxy, -NH 2 , nitro, - CN, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, OC 1 -C 4 alkyl, C 3 -C 8 cycloalkyl, OC 3 -C 8 cycloalkyl, C 2 -C 4 chain Alkenyl, C 2 -C 4 alkynyl, phenyl, benzyl;
  • substituents selected from the group A: halogen, hydroxy, -NH 2 , nitro, - CN, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, OC 1 -C 4 alkyl, C 3 -C 8 cycloalkyl, OC 3 -C 8 cycloalkyl, C 2 -C 4 chain Alkenyl, C 2 -C 4
  • substitution means having one or more (eg 1, 2, 3, 4) substituents selected from Group B: halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted Or unsubstituted C 1 -C 6 alkoxy, hydroxy, amino, nitro, substituted or unsubstituted C 1 -C 6 aldehyde, -CF 3 , -CN, -SF 5 , NR a R b , carboxyl -COR a , -CO 2 C 1 -C 6 alkyl, -CONR a R b , -SO 2 R b , -SO 2 NR a R b ;
  • X is selected from the group consisting of H, substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 6 -C 20 aryl, or substituted Or unsubstituted C 3 -C 14 heteroaryl, OH, substituted or unsubstituted OC 1 -C 6 alkyl, substituted or unsubstituted OC 3 -C 8 cycloalkyl, NR a R b or NR a C (O) R 3 ;
  • X is selected from: H, a substituted or unsubstituted C 1 -C 10 alkyl group, a substituted or unsubstituted C 3 -C 10 cycloalkyl group, a substituted or unsubstituted C 6 -C 20 aryl group, or
  • Y is selected from the group consisting of substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or Unsubstituted C 1 -C 6 alkylthio, substituted or unsubstituted C 6 -C 20 aryl, or substituted or unsubstituted C 3 -C 14 heteroaryl, -(CR 1 2 ) n C(O NR 2 R 3 , -C(CR 1 2 ) n C(S)NR 2 R 3 , -O(CR
  • Y is selected from the group consisting of substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 6 -C 20 aryl, or substituted or unsubstituted C 3 -C 14 heteroaryl, substituted or unsubstituted C 3 -C 10 cycloalkyl, Substituted or unsubstituted C 3 -C 10 heterocycloalkyl, -(CR 1 2 ) n C(O)NR 2 R 3 , -(CR 1 2 ) n C(S)NR 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3
  • each R a and each R b are independently selected from the group consisting of hydrogen, substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 2 -C 10 alkenyl, substituted or unsubstituted C 6 -C 20 aryl, or substituted or unsubstituted C 3 -C 14 heteroaryl, or R a and R b and the N atom attached thereto Forming a substituted or unsubstituted 4-8 membered heterocyclic ring, and the heterocyclic ring has at least one N and 0-2 heteroatoms selected from S, O, wherein N is NH in the nitrogen-containing heterocycle Or the NR g form, wherein R g is independently C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 6 -C 20 aryl, or C 3 -C 14
  • Each R 2 and each R 3 are independently selected from the group consisting of hydrogen, substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 2 -C 10 alkenyl group, a substituted or unsubstituted C 6 -C 20 aryl group, a substituted or unsubstituted C 3 -C 14 heteroaryl group, a substituted or unsubstituted C 3 -C 10 heterocyclyl, substituted Or unsubstituted amino group;
  • substitution means having one or more selected from Group G (eg 1, 2, 3) , 4) substituents: halogen, C 1 -C 6 alkyl, -OC 1 -C 6 alkyl, -CN, -CF 3 , -O-CF 3 , C 3 -C 10 cycloalkyl, C 2 -C 8 ester group, C 6 -C 10 aryl group;
  • Z is a carbon atom or N; when Z is N, X does not exist;
  • n is an integer from 0 to 8.
  • the compound of formula (I) is as shown in formula (II):
  • R c is one or more groups selected from the group consisting of halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 10 cycloalkoxy, hydroxy, amino, nitro, substituted or unsubstituted C 1 -C 6 aldehyde group, -CF 3 ,- CN, -SF 5 , NR a R b , carboxyl group, -COR a , -CO 2 C 1 -C 6 alkyl group, -CONR a R b , -SO 2 R 3 , -SO 2 NR a R b ; R 3 , R a and each R b are as defined above;
  • n is an integer from 1 to 4.
  • q 1, 2 or 3;
  • r is 1 or 2;
  • Z, X, and Y are as defined above.
  • the substituted or unsubstituted C 6 -C 10 aryl group is preferably a substituted or unsubstituted benzene ring.
  • Preferred is a substituted or unsubstituted thienyl or furyl group, preferably, From the following group:
  • Is a substituted or unsubstituted pyridyl group preferably, said From the following group:
  • X is selected from the group consisting of: H, a substituted or unsubstituted C 1 -C 10 alkyl group, a substituted or unsubstituted C 3 -C 10 cycloalkyl group, a substituted or unsubstituted C 6 -C
  • Y is selected from the group consisting of substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 6 -C 20 aryl a substituted or unsubstituted C 3 -C 14 heteroaryl group, -(CR 1 2 ) n C(O)NR 2 R 3 , -C(CR 1 2 ) n C(S)NR 2 R 3 , -O(CR 1 2 ) n C(O)NR 2 R 3 , -S(CR 1 2 ) n C(O)NR
  • Y is selected From the following group: substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 6 -C 20 aryl, or substituted or unsubstituted C 3 -C 14 heteroaryl, -(CR 1 2 n C(O)NR 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3 , -(CR 1 2 ) n S(O) 2 NR 2 R 3 ,- (CR 1 2 ) n NR 2 S(O) 2 NR 2 R 3 ,- (CR 1 2 ) n NR 2 S(O) 2 NR 2 R 3 , -(CR 1 2 ) n NR 2 C(O)NR 1 S(O) 2 R 3 , -(CR 1 2 ) n NR 2 C(O)NR 1 S(O) 2 R 3 , -(CR 1 2 )
  • each of R 2 and each R 3 is independently selected from the group consisting of hydrogen, substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 3 -C 10 ring An alkyl group, a substituted or unsubstituted C 2 -C 10 alkenyl group, a substituted or unsubstituted C 6 -C 20 aryl group, or a substituted or unsubstituted C 3 -C 14 heteroaryl group.
  • each R a and each R b and the N atom to which they are attached form a substituted or unsubstituted 4-8 membered heterocyclic ring
  • said heterocyclic ring has at least one N and 0-2 a hetero atom selected from the group consisting of S, O, wherein N in the nitrogen-containing heterocyclic ring is in the form of NH or NR g , wherein R g is independently C 1 -C 10 alkyl, C 3 -C 10 cycloalkyl, C 6- C 20 aryl, or C 3 -C 14 heteroaryl.
  • the compound of formula (I) is as shown in formula (III),
  • X, Y, Z, R c are as defined above, and m is an integer of 1 to 4.
  • the A is none.
  • the B is none.
  • the R 1 is H.
  • the E and M are each independently CR 1 .
  • said R 1 is independently selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 8 cycloalkyl, preferably H.
  • the X is selected from the group consisting of H, a substituted or unsubstituted C 1 -C 10 alkyl group, a substituted or unsubstituted C 3 -C 10 cycloalkyl group, preferably, X is H.
  • Y is selected from the group consisting of substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 1 -C 6 alkylthio, substituted or unsubstituted Substituted C 3 -C 14 heteroaryl, -NR 2 (CR 1 2 ) n C(O)NR 2 R 3 , -NR 2 (CR 1 2 ) n C(O)R 3 , -NR 2 C( O) (CR 1 2 ) n C(O)R 3 , -NR 2 (CR 1 2 ) n C(O)O(CR 1 2 ) n R 3 , -NR 2 (CR 1 2 ) n C(S NR 2 R 3 , -(CR 1 2 ) n C(O)NR 2 S(O) 2 R 3 , -NR 2 (CR 1 2 ) n S(O) 2 NR 2 R 3 , -(CR 1 2 (CR 1 2 ) n
  • X is OH
  • the Y is selected from the group consisting of substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 6 -C 20 aryl, or substituted or Unsubstituted C 3 -C 14 heteroaryl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted or unsubstituted C 3 -C 10 heterocycloalkyl.
  • the D group substituent is selected from the group consisting of a substituted or unsubstituted C 6 -C 20 aryl group, a hydroxyl group, a C 3 -C 6 heterocyclic group, -CH 2 -C 6 -C 10 aryl.
  • the compound of formula (I) is as shown in formula (IV):
  • R c is a group selected from the group consisting of halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3- C 8 cycloalkyl, substituted or unsubstituted C 3 -C 8 cycloalkoxy, hydroxy, -CF 3 , -CN, -SF 5 , NR a R b , carboxy, -COR a , -CONR a R b , -SO 2 R 3 , -SO 2 NR a R b ;
  • each R 3 , R a and each R b are as defined above;
  • n is an integer from 1 to 4.
  • X is selected from the group consisting of OH, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 3 -C 6 cycloalkyl, alkyl OC 1 -C 6 alkyl, OC 3 -C 8 Cycloalkyl, NR a R b or NR a C(O)R 3 ;
  • the compound is as shown in formula (V):
  • R c is a group selected from the group consisting of halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, hydroxy, -CF 3 , -CN, -SF 5 , NR a R b , carboxyl group, -COR a , -CONR a R b , -SO 2 R 3 , -SO 2 NR a R b ; wherein each R 3 , R a and each R b are as defined above;
  • n is an integer from 1 to 4.
  • R d is selected from the group consisting of H, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3 -C 8 cycloalkyl, Substituted or unsubstituted C 3 -C 8 cycloalkoxy, substituted or unsubstituted C 1 -C 6 haloalkyl, substituted or unsubstituted C 6 -C 20 aryl, substituted or unsubstituted C 3 -C C14 heteroaryl group; the substituent means a group having one or more (e.g., 3, 4) selected from substituent group H: halogen, C 1 -C 6 alkyl, C 3 -C 8 cycloalkyl , C 1 -C 6 haloalkyl, C 1 -C 6 alkoxy, C 3 -C 10 cycloalkyl, substituted or
  • the compound is as shown in formula (VI),
  • R c is one or more groups selected from the group consisting of halogen, substituted or unsubstituted C 1 -C 6 alkyl, substituted or unsubstituted C 1 -C 6 alkoxy, substituted or unsubstituted C 3- C 8 cycloalkyl, substituted or unsubstituted C 3 -C 8 cycloalkoxy, hydroxy, -CF 3 , -CN, -SF 5 , NR a R b , carboxy, -COR a , -CONR a R b , -SO 2 R 3 , -SO 2 NR a R b ; wherein each R a and each R b are as defined above;
  • n is an integer from 1 to 4.
  • R f is selected from the group consisting of H, SO 2 NH 2 , C 1 -C 6 haloalkyl, substituted or unsubstituted C 1 -C 10 alkyl, substituted or unsubstituted C 3 -C 10 cycloalkyl, substituted Or an unsubstituted C 2 -C 10 alkenyl group, a substituted or unsubstituted C 6 -C 20 aryl group, or a substituted or unsubstituted C 3 -C 14 heteroarylamino group; said substitution means having a group selected from Group I One or more (eg 1, 2, 3, 4) substituents: C 1 -C 6 alkyl, C 1 -C 6 alkoxy, C 1 -C 6 cycloalkyl, C 1 -C 6 Haloalkyl, alkoxy, heterocyclic, aryl, heteroaryl, -CF 3 , -CN, -SF 5 , NR a R b
  • the compound of formula (I) is selected from the group consisting of
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • R H, OH, NH 2 , alkyl, aryl, heteroaryl, O-alkyl, O-aryl, O-heteroaryl, NH-alkyl, NH-aryl, NH-heteroaryl ,N(alkyl) 2
  • the pharmaceutically acceptable salt is selected from the group consisting of hydrochloride, hydrobromide, sulfate, phosphate, methanesulfonate, triflate, benzenesulfonic acid Salt, p-toluenesulfonate (tosylate), 1-naphthalenesulfonate, 2-naphthalenesulfonate, acetate, trifluoroacetate, malate, tartrate, citrate, milk Acid, oxalate, succinate, fumarate, maleate, benzoate, salicylate, phenylacetate, mandelate.
  • the compound is the compound 1-117 prepared in the examples.
  • the compound is a cis-trans isomer.
  • the compound is a cis isomer.
  • the compound is a trans isomer.
  • the compound is a racemate.
  • the compound is an enantiomer.
  • a pharmaceutical composition comprising:
  • the anti-tumor drug is selected from the group consisting of a checkpoint antibody, such as a PD-1 antibody, a PD-L1 antibody, or a CTLA4 antibody.
  • a checkpoint antibody such as a PD-1 antibody, a PD-L1 antibody, or a CTLA4 antibody.
  • the anti-tumor drug is an immunotherapeutic drug for cancer.
  • the immunotherapeutic agent for the cancer is selected from the group consisting of a PD-1 antibody, a CTLA-4 antibody, a PD-L1 antibody, a PD-L2 antibody, a chemotherapeutic drug, or a targeted therapeutic drug.
  • the targeted therapeutic agent is selected from the group consisting of an HDAC inhibitor, a kinase inhibitor, an EP4 antagonist, or a combination thereof.
  • a third aspect of the invention provides a compound of formula (I), or a pharmaceutically acceptable salt thereof, a stereoisomer thereof, or a tautomer thereof, or a prodrug thereof, according to the first aspect of the invention Use for:
  • the IDO/TDO mediated disease is a disease characterized by the pathology of an IDO/TDO mediated tryptophan metabolic pathway.
  • the IDO/TDO mediated disease is selected from the group consisting of cancer, neurodegenerative disease, ocular disease, psychological disorder, depression, anxiety, Alzheimer's disease, or autoimmune disease.
  • the cancer includes, but is not limited to, colon cancer, breast cancer, gastric cancer, lung cancer, colon cancer, pancreatic cancer, ovarian cancer, prostate cancer, kidney cancer, liver cancer, brain cancer, melanoma, multiple Myeloma, chronic myeloid leukemia, hematological tumors, lymphoid tumors, including metastases in other tissues or organs away from the primary site of the tumor Lesion.
  • a method of preventing and/or treating an IDO/TDO mediated disease comprising administering to a patient a compound of the formula (I) according to the first aspect or the pharmaceutical composition of the second aspect step.
  • the IDO/TDO mediated disease is cancer, the method further comprising administering to the patient an additional anticancer agent (also referred to as an antitumor drug, as described above) step.
  • an additional anticancer agent also referred to as an antitumor drug, as described above
  • the inventors have extensively and intensively studied and developed for the first time a novel compound containing a nitrogen heterocyclic helix structure which can be used as a highly effective nitrogen-containing heterocyclic helix structure inhibitor for the prevention and/or treatment of IDO/ TDO-mediated diseases can also be used as anti-inflammatory drugs.
  • the present invention has been completed.
  • alkyl refers to a monovalent saturated aliphatic hydrocarbon group having from 1 to 10 carbon atoms, including straight-chain and branched hydrocarbon groups such as methyl (ie, CH 3 -), ethyl (ie, CH 3 CH 2 -).
  • n-propyl ie CH 3 CH 2 CH 2 -
  • isopropyl ie (CH 3 ) 2 CH-
  • n-butyl ie CH 3 CH 2 CH 2 CH 2 -
  • isobutyl ie (CH 3 ) 2 CHCH 2 -
  • sec-butyl ie (CH 3 )(CH 3 CH 2 )CH-
  • tert-butyl ie (CH 3 ) 3 C-
  • n-pentyl ie CH 3 ) CH 2 CH 2 CH 2 CH 2 -
  • neopentyl ie (CH 3 ) 3 CCH 2 -
  • the term includes a substituted or unsubstituted alkyl group.
  • substituted or unsubstituted means that the group may be unsubstituted or that H in the group is one or more (preferably 1-6, more preferably 1- Replaced by three substituents.
  • substituted or “substituted” means that the group has one or more (preferably 1-6, more preferably 1-3) substituents selected from the group consisting of: Halogen, hydroxy, -NH 2 , nitro, -CN, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, C 1 -C 4 alkoxy, C 3 -C 6 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, phenyl, benzyl.
  • substituents selected from the group consisting of: Halogen, hydroxy, -NH 2 , nitro, -CN, C 1 -C 4 alkyl, C 1 -C 4 haloalkyl, C 1 -C 4 alkoxy, C 3 -C 6 cycloalkyl, C 2 -C 4 alkenyl, C 2 -C 4 alkynyl, phenyl, benzyl.
  • cycloalkyl refers to a substituted or unsubstituted C 3 -C 12 cycloalkyl.
  • alkoxy refers to -O-alkyl, wherein the alkyl group can be saturated or unsaturated, can be branched, straight chain, or cyclic.
  • the alkoxy group has 1 to 10 carbon atoms, preferably 1 to 6 carbon atoms. Representative examples include, but are not limited to, methoxy, ethoxy, propoxy.
  • aryl refers to a monovalent aromatic carbocyclic group of 6 to 20 (preferably 6 to 14) carbon atoms which has a monocyclic (eg phenyl) or fused ring (eg naphthyl) Or sulfhydryl), if the point of attachment is on the aromatic carbon, the fused ring may be non-aromatic (eg 2-benzoxazolone, 2H-1,4-benzoxazine-3(4H)-one-7 - base, etc.).
  • Preferred aryl include phenyl and naphthyl.
  • the term includes substituted or unsubstituted forms wherein the substituents are as defined above.
  • cycloalkyl refers to a cyclic alkyl group having from 3 to 10 carbon atoms having a single or multiple ring (including fused systems, bridged ring systems, and spiro ring systems).
  • one or more of the rings may be a cycloalkyl, heterocyclic, aryl or heteroaryl group as long as the attachment site is a ring through a cycloalkyl group.
  • suitable cycloalkyl groups include, for example, adamantyl, cyclopropyl, cyclobutyl, cyclopentyl and cyclooctyl.
  • halo or halogen refers to fluoro, chloro, bromo and iodo.
  • heteroaryl refers to an aromatic group having from 1 to 10 carbon atoms and from 1 to 4 heteroatoms selected from the group consisting of oxygen, nitrogen and sulfur, such heteroaryl groups may be monocyclic (such as pyridyl or furyl) or a fused ring (such as indolizinyl or benzothienyl), wherein the fused ring may be non-aromatic and/or contain a hetero atom as long as the point of attachment is passed Atoms of aromatic heteroaryl groups.
  • the ring atom nitrogen and/or sulfur of the heteroaryl group is optionally oxidized to an N-oxide (N-O), a sulfinyl group or a sulfonyl group.
  • the heteroaryl group includes pyridinyl, pyrrolyl, indolyl, benzoxoxadiazole, imidazolyl, quinolyl, oxazolyl, isoxazolyl, thiazolyl, tetrazolyl, 1,2 , 3-triazolyl, 1,2,4-triazolyl, 1,3,4-oxadiazole, 1,2,4-oxadiazole, 1,2,5-oxadiazole, thiophene Base and furanyl.
  • the term includes substituted or unsubstituted heteroaryl.
  • substituted heteroaryl refers to a heteroaryl group substituted with 1 to 5, preferably 1 to 3, more preferably 1 to 2 substituents selected from the group consisting of The same substituent as defined by the aryl group.
  • heterocycle or “heterocycle” or “heterocycloalkyl” or “heterocyclyl” refers to a saturated, partially saturated or unsaturated group (but not aromatic), Has a single ring or a fused ring (including a bridged ring system and a spiro ring system having from 1 to 10 carbon atoms and from 1 to 4 heteroatoms selected from nitrogen, sulfur or oxygen in the ring, in a fused ring system, one or more
  • the ring may be a cycloalkyl, aryl or heteroaryl group as long as the point of attachment passes through the non-aromatic ring.
  • the nitrogen and/or sulfur atom of the heterocyclic group is optionally oxidized to provide N-oxide, sulfinyl and sulfonyl moieties.
  • substituted heterocyclic or “substituted heterocycloalkyl” or “substituted heterocyclic” as used herein refers to a heterocyclic group substituted by 1 to 5 (eg, 1 to 3) substituents.
  • the substituent is the same as the substituent defined by the substituted cycloalkyl group.
  • stereoisomer refers to a chiralally different compound of one or more stereocenters. Stereoisomers include enantiomers and diastereomers.
  • tautomer refers to an alternative form of a compound having a different proton position, such as an enol-ketone and an imine-enamine tautomer, or a tautomeric form of a heteroaryl group.
  • Prodrug refers to any derivative of an embodiment compound that, when administered to a subject, is capable of providing, directly or indirectly, a compound of the Examples, or an active metabolite or residue thereof. Particularly preferred derivatives and prodrugs are those, When administered to a subject, increasing the bioavailability of the example compound (eg, the compound administered orally is more readily absorbed into the blood) or increasing the parent compound to the bioregion (eg, brain or lymphatic system) relative to the parent species Transport of derivatives and prodrugs.
  • Prodrugs include the ester form of the compounds of the invention.
  • compound of the invention refers to a compound of formula (I), a racemate thereof, a stereoisomer thereof or a tautomer thereof, or a pharmaceutically acceptable salt thereof.
  • the present invention relates to a racemic mixture of these compounds, enriched in a mixture of any one of the enantiomers, and any of the separated enantiomers.
  • the racemic mixture refers to a mixture of two R and S enantiomers of 50%: 50%.
  • the isolated enantiomer is understood to be the pure enantiomer (ie 100%) or highly enriched in an enantiomer (purity ⁇ 98%, ⁇ 95%, ⁇ 93%, ⁇ 90%, ⁇ 88%) , ⁇ 85%, ⁇ 80%) of the mixture.
  • the invention includes all stereoisomers of the compound.
  • the invention includes all tautomers of the compounds.
  • the invention also includes deuterated compounds resulting from the substitution of any one or more of the hydrogen atoms of the compound by its stable isotope oxime.
  • the compound of the formula (I) of the present invention can be obtained by the following production method:
  • the cis isomer N can be synthesized in the following steps:
  • the compound of the formula (I) of the present invention can be obtained by the following production method:
  • the invention also provides a further preparation method of the compound Q of the formula (I), comprising the following steps:
  • the cis isomer T can be synthesized in the following steps:
  • Compound M can be obtained by the same reaction procedure as in the synthesis of compound Q to obtain compound T of formula (I).
  • the invention also provides a further preparation method of the compound X of the formula (I), comprising the following steps:
  • the invention also provides a further preparation method of the compound Z of the formula (I), comprising the following steps:
  • the invention also provides a further preparation method of the compound Z of the formula (I), comprising the following steps:
  • the invention also provides a preparation method of another compound (F) of formula (I), comprising the following steps:
  • the invention also provides a preparation method of another compound (H) of formula (I), comprising the following steps:
  • the invention also provides a process for the preparation of another compound of formula (I), comprising the steps of:
  • the invention also provides a process for the preparation of another compound of formula (I), comprising the steps of:
  • the invention also provides a process for the preparation of another compound of formula (I), comprising the steps of:
  • the invention also provides a process for the preparation of another compound of formula (I), comprising the steps of:
  • the invention also provides a pharmaceutical composition
  • a pharmaceutical composition comprising an active ingredient in a safe and effective amount, together with a pharmaceutically acceptable carrier.
  • the "active ingredient” as used herein means a compound of the formula (I), or a pharmaceutically acceptable salt thereof, a stereoisomer thereof or a tautomer thereof, or a prodrug thereof.
  • active ingredients and pharmaceutical compositions described herein are useful as IDO or TDO inhibitors.
  • Another preferred example In the preparation of a medicament for preventing and/or treating a tumor.
  • a medicament for the prevention and/or treatment of an IDO or TDO mediated disease is prepared.
  • the pharmaceutical compositions contain from 1 to 2000 mg of active ingredient per dose, more preferably from 10 to 200 mg of active ingredient per dose.
  • the "one dose” is a tablet.
  • “Pharmaceutically acceptable carrier” means: one or more compatible solid or liquid fillers or gel materials which are suitable for human use and which must be of sufficient purity and of sufficiently low toxicity.
  • “compatibility” it is meant herein that the components of the composition are capable of intermingling with the active ingredients of the present invention and with respect to each other without significantly reducing the efficacy of the active ingredients.
  • the compounds of the preferred embodiments of the invention may be administered as separate active agents or in combination with one or more other agents useful in the treatment of cancer.
  • the use of the compounds of the preferred embodiments of the invention in combination with known therapeutic agents and anticancer agents is also effective, and combinations of currently known compounds with other anticancer or chemotherapeutic agents are within the scope of the preferred embodiments. Examples of such agents can be found in "Cancer Principles and Practice of Oncology", VT Devita and S. Hellman (editor), 6th edition (February 15, 2001), Lippincott Williams & Wilkins Press .
  • One of ordinary skill in the art will be able to discern effective combinations of agents based on the particular nature of the drug and the cancer involved.
  • anticancer agents include, but are not limited to, the following: estrogen receptor modulators, androgen receptor modulators, retinol receptor modulators, cytotoxic/cytostatic agents, antiproliferative agents, iseswagen Alkenyl protein transferase inhibitors, acetylase (HDAC) inhibitors, HMG-CoA reductase inhibitors and other angiogenesis inhibitors, cell proliferation and survival signal inhibitors, apoptosis inducers, and interfering cell cycle checkpoints ( Cell cycle checkpoint), CTLA4 antibody, PD-1 antibody, PD-L1 antibody, and the like.
  • the compounds of the preferred embodiments are also effective when administered concurrently with radiation therapy.
  • the therapeutically effective dose can generally be the total daily dose administered to the patient in a single or divided dose, for example, from about 0.001 to about 1000 mg/kg body weight per day, preferably from about 1.0 to about daily. 30 mg / kg body weight.
  • a Dosage unit composition can include its dosage factor to form a daily dose. The choice of dosage form will depend on various factors such as the mode of administration and the bioavailability of the drug substance.
  • the compounds of the preferred embodiments can be administered as a pharmaceutical composition by any of the following routes: oral, systemic (e.g., transdermal, intranasal or by suppository), or parenteral (e.g., intramuscular, intravenous). Or subcutaneous).
  • the preferred mode of administration is oral, and a convenient daily dose can be adjusted depending on the degree of bitterness.
  • the compositions may take the form of tablets, pills, capsules, semi-solids, powders, sustained release formulations, solutions, suspensions, elixirs, aerosols or any other suitable compositions.
  • Another preferred mode of administration of the preferred embodiment compounds is inhalation. This is an effective method of delivering a therapeutic agent directly to the respiratory tract (see, e.g., U.S. Patent No. 5,607,915).
  • Suitable pharmaceutically acceptable carriers or excipients include, for example, treatment and drug delivery modifiers and accelerators such as calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starches, gelatin, cellulose , sodium methyl cellulose, carboxymethyl cellulose, glucose, hydroxypropyl-B-cyclodextrin, polyvinylpyrrolidone, low melting wax, ion exchange resin Etc., and any combination of two or more thereof.
  • treatment and drug delivery modifiers and accelerators such as calcium phosphate, magnesium stearate, talc, monosaccharides, disaccharides, starches, gelatin, cellulose , sodium methyl cellulose, carboxymethyl cellulose, glucose, hydroxypropyl-B-cyclodextrin, polyvinylpyrrolidone, low melting wax, ion exchange resin Etc., and any combination of two or more thereof.
  • the liquid and semi-solid excipients may be selected from the group consisting of glycerin, propylene glycol, water, ethanol, and various oils, including petroleum, animal, vegetable, or synthetic sources such as peanut oil, soybean oil, mineral oil, sesame oil, and the like.
  • Preferred liquid carriers, particularly carriers for injectable solutions include water, saline, aqueous dextrose and ethylene glycol.
  • Other suitable pharmaceutically acceptable excipients are described in Remington's Pharmaceutical Sciences, Mack Pub. Co., New Jersey (1991), incorporated herein by reference.
  • pharmaceutically acceptable salt refers to a non-toxic acid or alkaline earth metal salt of a compound of formula I. These salts can be prepared in situ by final isolation and purification of the compound of formula I, or by separately reacting a suitable organic or inorganic acid or base with a basic or acidic functional group.
  • Representative salts include, but are not limited to, acetate, adipate, alginate, citrate, aspartate, benzoate, besylate, hydrogen sulfate, butyrate , camphorate, camphor sulfonate, digluconate, cyclopentane propionate, lauryl sulfate, ethanesulfonate, glucose heptanoate, glycerol phosphate, hemisulfate, heptanoic acid Salt, hexanoate, fumarate, hydrochloride, hydrobromide, hydroiodide, 2-hydroxyethanesulfonate, lactate, maleate, methanesulfonate, nicotinate , 2-naphthyl sulfonate, oxalate, pamoate, pectate, thiocyanate, 3-phenylpropionate, picrate, pivalate, propionate, Succinate, sul
  • the nitrogen-containing basic group can be quaternized by the following reagents: alkyl halides such as methyl, ethyl, propyl, butyl chloride, bromide and iodide; dialkyl sulfate Such as dimethyl, diethyl, dibutyl and dipentyl sulfate; long chain halides such as sulfhydryl, lauryl, myristyl and stearyl chlorides, bromides and iodides; aralkyl Base halides such as benzyl and phenethyl bromide. A water soluble or oil soluble or dispersible product is thus obtained.
  • alkyl halides such as methyl, ethyl, propyl, butyl chloride, bromide and iodide
  • dialkyl sulfate Such as dimethyl, diethyl, dibutyl and dipentyl sulfate
  • long chain halides
  • Examples of the acid which can be used to form a pharmaceutically acceptable acid addition salt include inorganic acids such as hydrochloric acid, sulfuric acid, phosphoric acid, and organic acids such as oxalic acid, maleic acid, methanesulfonic acid, succinic acid, and citric acid.
  • the base addition salt can be prepared in situ upon final isolation and purification of the compound of formula I, or by separately reacting the carboxylic acid moiety with a suitable base such as a hydroxide, carbonate or carbonate of a pharmaceutically acceptable metal cation. Hydrogen salt) or ammonia, or organic primary, secondary or tertiary amine reaction.
  • Pharmaceutically acceptable salts include, but are not limited to, alkali metal and alkaline earth metal based cations such as sodium, lithium, potassium, calcium, magnesium, aluminum, and the like, as well as non-toxic ammonium, quaternary ammonium, and amine cations, including However, it is not limited to: ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine and the like.
  • Other representative organic amines useful for the formation of base addition salts include diethylamine, ethylenediamine, ethanolamine, diethanolamine, piperazine, and the like.
  • the term "pharmaceutically acceptable prodrug” refers to a prodrug of a compound of those preferred embodiments, which is rapidly converted in vivo to a parent compound of the above formula, for example, hydrolyzed in blood.
  • pharmaceutically acceptable prodrug refers to a prodrug of a compound of those preferred embodiments, which is rapidly converted in vivo to a parent compound of the above formula, for example, hydrolyzed in blood.
  • the compound of the present invention can be used as a highly potent IDO/TDO enzyme inhibitor
  • the synthesis method is mild, the operation is simple and easy, the yield is high, and the derivatization is easy, and it is suitable for industrial scale production;
  • the compound 2-bromobenzyl cyanide (10.2 g, 52.0 mmol) was added to the reaction flask, cooled to minus 78 ° C, and methyl lithium (44 mL, 52.0 mmol) was added dropwise. After the addition was completed, the mixture was stirred at this temperature for 1 hour. Epichlorohydrin (5.3 g, 57.2 mmol), stirred at this temperature for 3 hours, TLC detection of the disappearance of the starting material, then the addition of methylmagnesium bromide format reagent (19 ml, 57.2 mmol), after the completion of the addition, naturally returned to room temperature Then slowly warm to 60 degrees and stir overnight. The TLC monitoring reaction was complete.
  • the product of the first step (24.0 g, 197.0 mmol) was dissolved in 230 mL of ethanol, and then 230 mL of water, potassium hydroxide (53.3 g, 952.0 mmol), heated to 105 ° C, and reacted for 24 hours, the material disappeared completely.
  • the product of the second step (10.6 g, 39.1 mmol) was dissolved in 100 mL of dioxane, diphenyl azide (13.0 g, 46.9 mmol) was added to the reaction mixture, then diisopropylethylamine (10.1 g) was added. 78.2 mmol) and tert-butanol (50 mL) were replaced with argon several times. After stirring for ten minutes, the mixture was heated to 80 ° C and reacted for 4 hours. After cooling to room temperature, a white solid precipitated in the reaction system, and 7.7 g of the target compound was obtained by filtration, yield 73%.
  • Step 4 (cis) 3-amino-3-(2-bromophenyl)cyclobutanol
  • the product of the third step (7.7 g, 28.7 mol) was added to 100 mL of water and 100 mL of isopropanol, and potassium hydroxide (80.0 g, 1.4 mol) was added thereto, and the reaction was refluxed for 24 hours. After cooling to room temperature, the mixture was extracted with EtOAc (EtOAc m.
  • Step 5 (cis) 3-(2-bromophenyl)-3-(1H-imidazol-1-yl)cyclobutanol
  • the product of the fourth step (5.0 g, 20.7 mmol) was dissolved in 50 mL of methanol, and then ethyldialdehyde (6.0 g, 41.3 mmol) of formaldehyde (3.4 g, 41.3 mmol) and ammonium acetate (3.2 g, 41.3 mmol) were added. After the addition, the reaction was slowly warmed to 70 °C, and the reaction was carried out for 4 hours. After the reaction was completed, the mixture was concentrated, and the mixture was combined and purified by column chromatography to give the title compound (5.5 g, yield: 91%).
  • Step 6 (cis) snail [cyclobutane 1,5'-imidazo[5,1-a]isoindole]-3-ol
  • Step 7 Spiro [cyclobutane 1,5'-imidazo[5,1-a]isoindole-3-one
  • the product of the sixth step (0.5 g, 2.4 mmol) was placed in a bottle, 5 mL dichloromethane was added, stirred under ice-cooling, sodium bicarbonate (1.9 g, 23.4 mmol) was added, and Dess-Martin oxidant (1.5 g) was added. , 3.5 mmol), stirring at room temperature, knowing that the raw materials disappeared. It was extracted with 20 mL of dichloromethane and 10 mL of water to give an organic phase, dried, filtered, concentrated and purified by column chromatography. The target compound (400 mg, yield 80%) was obtained.
  • Step 8 (cis/trans) 3-phenylspiro[cyclobutane 1,5'-imidazo[5,1-a]isoindole]-3-ol
  • Step 2 Spirulina [cyclobutane 1,5'-imidazo[5,1-a] is ⁇ ]-3-carbonitrile
  • the first step product (300 mg, 1.0 mmol) was dissolved in 10 mL of DMF and sodium cyanide (150 mg, 3.0 mmol). The raw materials disappeared. After cooling to room temperature, 30 mL of ethyl acetate was added, and the mixture was washed with 10 mL of water, and the organic phase was dried over magnesium sulfate, filtered, concentrated, and purified to give the title compound (220 mg, yield 100%).
  • the second step product (220 mg, 1.0 mmol) was added to ethanol (10 mL), then water (10 mL) was added, and potassium hydroxide (1.1 g, 20.0 mmol) was added and heated to 100 deg. TLC showed that the starting material had disappeared. After cooling to room temperature, 2N hydrochloric acid was added to adjust the pH to 6-7, the solution was spun, and the product was dissolved in 10% dichloromethane in methanol. The solid was filtered and collected. Combine, dry, filter and concentrate. The crude title compound was obtained (200 mg, yield: 83%).
  • the third step product (48 mg, 0.2 mmol) was added to dichloromethane, triethylamine (44 mg, 0.4 mmol) was added, and N,O- dimethyl hydrochloride (20 mg, 0.2 mmol) was added, and EDCI (76 mg) was added. , 0.4 mmol), HOBT (27 mg, 0.2 mmol) was reacted at room temperature overnight, and the starting material disappeared. The reaction mixture was dried and purified by column chromatography toield (yield:
  • Step 5 (trans) 3-fluorophenyl) (spiro[cyclobutane 1,5'-imidazo[5,1-a]isoindole]-3-yl)methanone
  • the product of the fourth step (60 mg, 0.2 mmol) was placed in a reaction flask, and tetrahydrofuran (2 mL) was added, and the mixture was cooled in an ice bath, and 3-fluorophenylmagnesium bromide (0.9 mL, 0.90 mmol) was added dropwise. The reaction was quenched with EtOAc (2 mL). Purification by column chromatography gave 50 mg of EtOAc (EtOAc:EtOAc.
  • Step 6 (cis/trans)(3-fluorophenyl)(spiro[cyclobutane 1,5'-imidazo[5,1-a]isoindole-3-yl)methanone
  • Step 7 (cis/trans)-(3-fluorophenyl)(spiro[cyclobutane 1,5'-imidazo[5,1-a]isoindole]-3-yl)methanol
  • 1,4-dioxaspiro[4.5]indole-7-ene-8-lithium was used instead of phenylmagnesium bromide as the starting material to give the title compound as a white solid (10 mg, Yield 29%).
  • Triphenylphosphine (393 mg, 1.5 mmol), benzoic acid (92 mg, 0.8 mmol) and (cis) spiro[cyclobutane-1,5'-imidazo[5,1-a]isoindole at room temperature ⁇ ]-3-ol (159 mg, 0.8 mmol) was added to a 50 mL two-necked flask, and Ar 2 was exchanged three times, and 4 mL of tetrahydrofuran was injected. Then, a solution of DIAD (273 mg, 1.4 mmol) in THF (2 mL) was added dropwise to the above system, and the mixture was allowed to react to room temperature for 4 h, and the reaction was stopped. The reaction mixture was concentrated and purified EtOAcjjjjjjjjjjjj
  • the third step product (300 mg, 1.0 mmol) was dissolved in 10 mL DMF and sodium azide (200 mg, 3.0 mmol) was added and heated to 100 deg. After cooling to room temperature, 30 mL of ethyl acetate was added, and the mixture was washed with 10 mL of water, and the organic phase was dried over magnesium sulfate, filtered, concentrated, and purified to give the title compound (220 mg, yield 92%).
  • Step 5 (cis) (1s, 3s)-spiro [cyclobutane 1,5'-imidazo[5,1-a]isoindole]-3-amine
  • the fourth step product (220 mg, 0.9 mmol) was dissolved in ethanol, Pd/C (20 mg, 10%) was added and hydrogen was applied. The reaction was overnight. The Pd/C was filtered off, and the filtrate was concentrated to give crude title compound (190 mg, yield: 96%).
  • the target compound can also be obtained by the following conditions: 5% Pd/C (20 mg) is added to the fourth step product cis-3-azidospiro[cyclobutane-1,5'- at room temperature under an argon atmosphere. Imidazo[5,1- ⁇ ]isoindole] (209 mg, 0.9 mmol) in EtOAc (4 mL). 1atm H 2 at room temperature the reaction after 24h, was added Celite and the filter cake was washed with 5mL EtOH, and the combined organic phases were dried to give a pale yellow solid spin title compound 170 mg, 87% yield.
  • Step 6 (cis) 4-methyl-N-((1s,3s)-spiro[cyclobutane 1,5'-imidazo[5,1-a]isoindole-3-yl) Benzenesulfonamide
  • the target compound was obtained according to the reaction conditions described in the sixth step of Example 4.
  • the intermediate (21 mg, 0.1 mmol) prepared in the first step of Example 5 was added to the reaction flask with 2-bromo-1-phenylethanone (30 mg, 0.2 mmol), triethylamine (20 mg, 0.2 mmol).
  • Dichloromethane (3 mL) was added and the reaction was heated to 40 °C for 2 hours. TLC showed the end of the reaction.
  • Dichloromethane (10 mL) and water (10 mL) were added to the mixture and the mixture was evaporated. It was filtered, concentrated, and purified by column chromatography to give 20 mg (yield: 60.0%).
  • Example 80 (48 mg, 0.2 mmol) was added to 5 mL of dichloromethane, MMPP (98 mg, 0.2 mmol) was added under ice-cooling, and the reaction was carried out at this temperature until the starting material disappeared, and the reaction was quenched by the addition of sodium thiosulfate solution. Extract (3 x 10 mL), combine the organic phases, dry, filtered, concentrated The title compound (20 mg, yield 37%) was obtained.
  • Example 80 (48 mg, 0.2 mmol) was added to 5 mL of dichloromethane, and MMPP (74 mg, 0.2 mmol) was added under ice-cooling. The reaction was continued at this temperature until the starting material disappeared, and the reaction was quenched by the addition of sodium thiosulfate solution. The mixture was extracted (3 ⁇ 10 mL).
  • Step 2 3-(1-Benzyl-1H-1,2,3-triazol-4-yl)spiro[cyclohexane 1,5'-imidazo[5,1-a]isoindole] 3-ol
  • the first step product (20 mg, 0.076 mmol), benzyl azide (20 mg, 0.2 mmol), copper sulfate pentahydrate (14 mg, 0.08 mmol) and sodium ascorbate (15 mg, 0.08 mmol) were added to dichloromethane, water, methanol (2 mL, 1 mL, 1 mL), stirring at room temperature for 24 hours, TLC showed the disappearance of the starting material, methylene chloride (10 ml) was added to the reaction mixture, filtered over Celite, and the filtrate was concentrated, The yield was 36%.
  • chlorosulfonyl isocyanate (9 uL, 0.1 mmol) was dissolved in 2 mL of dichloromethane, and 2-bromoethanol (7 uL, 0.1 mmol) was dissolved in 1 mL of dichloromethane, and the mixture was stirred for 10 minutes.

Abstract

本发明公开了一种含有氮杂环螺旋结构的高效IDO/TDO双抑制剂,具体地,涉及一种式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药。本发明的式(I)化合物,可以作为吲哚胺-2,3-双加氧酶抑制剂和色氨酸-2,3-双加氧酶,用于制备预防和/或治疗吲哚胺-2,3-双加氧酶和色氨酸-2,3-双加氧酶介导的疾病的药物,

Description

一种含有氮杂环螺旋结构的高效IDO/TDO双抑制剂 技术领域
本发明属于药物化学技术领域,具体涉及一种含有氮杂环螺旋结构的高效IDO/TDO抑制剂及其制备方法。
背景技术
吲哚胺-2,3-双加氧酶(Indoleamine-2,3-dioxygenase,IDO)是1967年Hayaishi小组首次在细胞内发现的一种含有亚铁血红素的单体酶,cDNA编码蛋白由403氨基酸组成,分子量为45kDa,它是延着色氨酸-犬尿氨酸途径分解代谢的限速酶,并且在多种哺乳动物的组织中具有广泛的表达(Hayaishi O.et al Science,1969,164,389-396)。在肿瘤患者的细胞中,IDO常作为诱导肿瘤微环境免疫耐受产生重要的生理作用,其介导的色氨酸(Tryptophan,Trp)-犬尿氨酸(Kynurenine,Kyn)代谢途径参与了肿瘤免疫逃逸,而IDO作为诱导肿瘤微环境免疫耐受也产生重要的作用。
色氨酸为哺乳动物体内重要的必须氨基酸之一,需要从食物中大量摄取,维持细胞活化和增殖,以及蛋白质以及一些神经递质的合成。因此,它的缺乏会导致一些重要的细胞的功能失常。IDO在体内能够催化色氨酸转化为N-甲酰犬尿氨酸,降低色氨酸的含量而造成色氨酸体内的不足,导致肿瘤的发生。而免疫组织学研究显示,犬尿氨酸途径能够导致兴奋毒素喹啉酸的增多,还会导致阿兹海默等神经系统疾病等多种严重的人类疾病(Guillemin G.J.et al Neuropathol.and Appl.Neurobiol.2005,31,395–404)。
研究表明,IDO在肿瘤微环境中可以通过以下几种方式来抑制局部T细胞免疫反应:色氨酸耗竭、毒性代谢和诱导调节性T细胞增殖。很多情况是在肿瘤中过度表达,从而消耗局部的色氨酸,产生大量的犬尿氨酸等代谢产物。事实上,在无色氨酸或犬尿氨酸的培养条件下,T细胞会发生增殖抑制、活性降低甚至凋亡。而T细胞中存在一个对色氨酸水平非常敏感的调节点,IDO作用能够消耗色氨酸,从而导致T细胞停滞于G1期中期,从而抑制了T细胞的增殖,也抑制了T细胞的免疫应答。而T细胞一旦停止增殖,可能就不会再被刺激作用,这是IDO在体内免疫作用机制(Mellor A.et al Biochem.Biophys.Res.Commun.2005,338(1):20-24)(LeRond S.et al J.Exp.Med.2002,196(4):447-457)。
TDO是另外一种色氨酸代谢酶,它的作用机制与IDO一样,只是它起初被认为是内源的,特别是在正常肝脏和脑细胞都有表达,特别是在肝脏中有高表达。现已验证了TDO在多种癌症病人组织中具有过度表达,而且TDO抑制剂在小鼠肿瘤模型中通过免疫排斥肿瘤细胞生长的功效得以验证(Pilotte,l.et al.Proc.Natl.Acad.Sci.USA; published online,Jan.30,2012)。
因此IDO、TDO抑制剂或IDO/TDO双抑制剂是肿瘤免疫治疗的重要靶点,本领域尚需研发高活性的新型IDO、TDO或IDO/TDO抑制剂。
发明内容
本发明的目的在于提供一种新型的可作为高效的IDO/TDO双抑制剂的含有氮杂环螺旋结构的化合物,及其制备方法,本发明的式(I)化合物,具有抗肿瘤、治疗神经退行疾病(阿尔茨海默病)、抗炎等多种药理活性。
本发明第一方面,提供一种式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药:
Figure PCTCN2017108941-appb-000001
式中,
A选自下组:S、O、-C(R1)2-或无;
B选自下组:-C(R1a)2-或无;
E和M分别独立地选自:N或CR1
各R1独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基、OH、取代或未取代的O-C1-C6烷基、取代或未取代的O-C3-C8环烷基;
各R1a独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基;
所述R1、R1a中,所述取代是指具有选自A组的一个或多个(如1、2、3、4个)取代基:卤素、羟基、-NH2、硝基、-CN、C1-C4烷基、C1-C4卤代烷基、O-C1-C4烷基、C3-C8环烷基、O-C3-C8环烷基、C2-C4链烯基、C2-C4炔基、苯基、苄基;
Figure PCTCN2017108941-appb-000002
选自下组:取代或未取代的C6-C10芳基、取代或未取代的C3-C10杂芳基且所述杂芳基具有1-3个选自S、O或N的杂原子;其中,所述取代是指具有选自B组的一个或多个(如1、2、3、4个)取代基:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、氨基、硝基、取代或未取代的C1-C6醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2Rb、-SO2NRaRb
X选自下组:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、OH、取代或未取代的OC1-C6烷基、取代或未取代的OC3-C8环烷基、NRaRb或NRaC(O)R3
并且,当X选自:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷硫基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-C(CR1 2)nC(S)NR2R3、-O(CR1 2)nC(O)NR2R3、-S(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)R3、-NR2C(O)(CR1 2)nC(O)R3、-NR2(CR1 2)nC(O)O(CR1 2)nR3、-NR2(CR1 2)nC(S)NR2R3、-(CR1 2)nNR2C(S)NR2R3、-(CR1 2)nNR2C(O)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-O(CR1 2)nC(O)NR2S(O)2R3、-S(CR1 2)nC(O)NR2S(O)2R3、-NR2(CR1 2)nS(O)2NR2R3、-NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-NR2S(O)2R3、-(CR1 2)nS(O)2R3、NR2C(=N-CN)NR2R3;所述取代是指具有选自C组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基、OSO2NH2、NHSO2NH2、氨基、硝基、醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2
当X选自:OH、OC1-C6烷基、O-C3-C8环烷基、NRaRb或NRaC(O)R3时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、取代或未取代的C3-C10环烷基、取代或未取代的C3-C10杂环烷基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3;所述取代是指具有选自D组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、取代或未取代的C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CH2-C6-C10芳基、-CH2-C6-C10芳基、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2、羟基、OSO2NH2、NHSO2NH2、C3-C6杂环基;
其中,各Ra和各Rb分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、或Ra和Rb和与之相连的N原子共同形成取代或未取代的4-8元杂环,且所述的杂环具有至少一个N和0-2个选自S、O的杂原子,其中所述的含氮 杂环中N为NH或NRg形式,其中Rg独立地为C1-C10烷基、C3-C10环烷基、C6-C20芳基、或C3-C14杂芳基;其中,所述取代指具有选自E组的一个或多个(如1、2、3、4个)取代基:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、OSO2NH2、NHSO2NH2、氨基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2Rb、-SO2NRaRb
各R2和各R3分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、取代或未取代的C3-C10杂环基、取代或未取代的氨基;
Figure PCTCN2017108941-appb-000003
为取代或未取代的3-10元碳环或杂环,所述杂环具有至少一个碳原子被选自下组的杂原子取代:O、S、S(O)、S(O)2、C(O)、C(S)和NRg;其中,所述取代指具有选自F组的一个或多个(如1、2、3、4个)取代基:OH、卤素、C1-6烷基;
在A组、B组、C组、D组、E组、F组、R2、R3的取代基中,所述取代指具有选自G组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、-O-C1-C6烷基、-CN、-CF3、-O-CF3、C3-C10环烷基、C2-C8酯基、C6-C10芳基;
Z是碳原子或N;当Z是N时,X不存在;
n为0至8的整数。
在另一优选例中,所述式(I)化合物如式(II)所示:
Figure PCTCN2017108941-appb-000004
式中,
Figure PCTCN2017108941-appb-000005
选自下组:
Figure PCTCN2017108941-appb-000006
Figure PCTCN2017108941-appb-000007
其中,Rc为一个或多个选自下组的基团:卤素、取代或未取代的C1-C6烷基、取代或未取代的C3-C10环烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C10环烷氧基、羟基、氨基、硝基、取代或未取代的C1-C6醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb;其中各R3、Ra和各Rb如上所定义;
m是1至4的整数;
q是1、2或3;
r是1或2;
Figure PCTCN2017108941-appb-000008
Z、X、Y如上所定义。
在另一优选例中,所述
Figure PCTCN2017108941-appb-000009
为取代或未取代的C6-C10芳基,较佳地为取代或未取代的苯环。
在另一优选例中,
Figure PCTCN2017108941-appb-000010
为取代或未取代的噻吩基或呋喃基,较佳地,
Figure PCTCN2017108941-appb-000011
选自下组:
Figure PCTCN2017108941-appb-000012
在另一优选例中,
Figure PCTCN2017108941-appb-000013
是取代或未取代的吡啶基,较佳地,所述
Figure PCTCN2017108941-appb-000014
选自下组:
Figure PCTCN2017108941-appb-000015
在另一优选例中,
Figure PCTCN2017108941-appb-000016
选自下组:
Figure PCTCN2017108941-appb-000017
在另一优选例中,当X选自:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10 环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-C(CR1 2)nC(S)NR2R3、-O(CR1 2)nC(O)NR2R3、-S(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)R3、-NR2C(O)(CR1 2)nC(O)R3、-NR2(CR1 2)nC(O)OR3、-NR2(CR1 2)nC(S)NR2R3、-(CR1 2)nNR2C(O)NR2R3、-(CR1 2)nNR2C(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-NR2(CR1 2)nC(S)NR2R3、-NR2C(O)NR2R3、-NR2C(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-O(CR1 2)nC(O)NR2S(O)2R3、-S(CR1 2)nC(O)NR2S(O)2R3、-NR2(CR1 2)nS(O)2NR2R3、-NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3、NR2C(=N-CN)NR2R3;所述取代是指具有选自C组的一个或多个(如1、2、3、4个)取代基。
在另一优选例中,当X选自下组:OH、OC1-C6烷基、OC3-C8环烷基、NRaRb或NRaC(O)R3时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3;所述取代是指具有选自D组的一个或多个(如1、2、3、4个)取代基。
在另一优选例中,所述各R2和各R3分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基。
在另一优选例中,各Ra和各Rb和与之相连的N原子共同形成取代或未取代的4-8元杂环,且所述的杂环具有至少一个N和0-2个选自S、O的杂原子,其中所述的含氮杂环中N为NH或NRg形式,其中Rg独立地为C1-C10烷基、C3-C10环烷基、C6-C20芳基、或C3-C14杂芳基。
在另一优选例中,所述式(I)化合物如式(III)所示,
Figure PCTCN2017108941-appb-000018
式中,X、Y、Z、Rc如上所定义,m是1至4的整数。
在另一优选例中,所述A为无。
在另一优选例中,所述B为无。
在另一优选例中,所述R1为H。
在另一优选例中,所述E和M分别独立地为CR1
在另一优选例中,所述R1独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基,较佳地为H。
在另一优选例中,所述X选自下组:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基,较佳地,X为H。
在另一优选例中,当X为H时,Y选自下组:取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷硫基、取代或未取代的C3-C14杂芳基、-NR2(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)R3、-NR2C(O)(CR1 2)nC(O)R3、-NR2(CR1 2)nC(O)O(CR1 2)nR3、-NR2(CR1 2)nC(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-NR2(CR1 2)nS(O)2NR2R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3或NR2C(=N-CN)NR2R3
在另一优选例中,X为OH。
在另一优选例中,当X为OH时,所述Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、取代或未取代的C3-C10环烷基、取代或未取代的C3-C10杂环烷基。
在另一优选例中,所述D组取代基选自下组:取代或未取代的C6-C20芳基、羟基、C3-C6杂环基、-CH2-C6-C10芳基。
在另一优选例中,所述式(I)化合物如式(IV)所示:
Figure PCTCN2017108941-appb-000019
式中,
式中,Rc为一个选自下组的基团:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb
其中各R3、Ra和各Rb如上所定义;
m是1至4的整数;
Figure PCTCN2017108941-appb-000020
为选自下组的取代或未取代的3-7元杂环:
Figure PCTCN2017108941-appb-000021
X选自下组:OH、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、烷基OC1-C6烷基、OC3-C8环烷基、NRaRb或NRaC(O)R3
Figure PCTCN2017108941-appb-000022
选自下组:取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基,其中芳环和杂芳环可被一个或多个选自下组的基团E取代:卤素、C1-C6烷基、C3-C8环烷基、C1-C6卤代烷基、C1-C6烷氧基、或未取代的C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、OH、OSO2NH2、NHSO2NH2、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2;其中各Ra和各Rb各自独立为氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基;Ra和Rb可以一起可以形成四至八元杂环,其中杂原子可以是硫、氧、NH或NRg
在另一优选例中,所述化合物如式(V)所示:
Figure PCTCN2017108941-appb-000023
式中,
Rc为一个选自下组的基团:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其 中各R3、Ra和各Rb如上所定义;
m是1至4的整数;
Figure PCTCN2017108941-appb-000024
选自下组:
Figure PCTCN2017108941-appb-000025
Rd选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、取代或未取代的C1-C6卤代烷基、取代或未取代的C6-C20芳基、取代或未取代的C3-C14杂芳基;所述取代指具有选自H组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C3-C8环烷基、C1-C6卤代烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、OH、OSO2NH2、NHSO2NH2、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2
在另一优选例中,所述化合物如式(VI)所示,
Figure PCTCN2017108941-appb-000026
式中,
Rc为一个或多个选自下组的基团:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其中各Ra和各Rb如上所定义;
m是1至4的整数;
Figure PCTCN2017108941-appb-000027
选自下组:
Figure PCTCN2017108941-appb-000028
Rf选自下组:H、SO2NH2、C1-C6卤代烷基、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基氨基;所述取代指具有选自I组的一个或多个(如1、2、3、4个)取代基:C1-C6烷基、C1-C6烷氧基、C1-C6环烷基、C1-C6卤代烷基、烷氧基、杂环基,芳基、杂芳基、-CF3、-CN、-SF5、NRaRb、羧基、羟基、OSO2NH2、NHSO2NH2、氨基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其中,所述Ra和Rb和与之相连的N原子共同形成取代或未取代的4-8元杂环,且所述的杂环具有至少一个N和0-2个选自S、O的杂原子,其中所述的含氮杂环中N为NH或NRg形式,其中Rg独立地为C1-C10烷基、C3-C10环烷基、C6-C20芳基、或C3-C14杂芳基。
在另一优选例中,所述式(I)化合物选自下组:
(顺/反)3-苯基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
(顺/反)3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲醇
(顺/反)3-(1,4-二氧杂螺[4.5]癸-7-烯-8-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
(顺式)4-甲基-N-((1s,3s)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
(顺式)4-甲基-N-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺/反)3-氟-N-(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺式)4-氟-N-(1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
(反式)4-氟-N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
(反式)N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己烷甲酰胺
(顺式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己基磺酰胺
(顺式)3-氯-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3-溴-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷磺酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷甲酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙烷-2-磺酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-2-磺酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-1-磺酰胺
(反式)4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2,4-二氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)N((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-2-萘甲酰胺
(反式)3-氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-氰基-N-(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3-溴-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-3-(三氟甲基)苯甲酰胺
(反式)苄基(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸叔丁酯
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)异丁酰胺
(反式)((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸苯酯
(顺式)N((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙烯酰胺
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)甲基磺酰胺
(顺/反)3-甲氧基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3,4-二氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺/反)2-甲基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3-甲基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2,4,6-三甲基-N-((1r,3r)-螺[环丁基烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2-氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3-氟-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)3-三氟甲氧基-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-溴-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-氟-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)4-氧代-4-苯基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丁酰胺
(反式)2-([1,1’-联苯]-4-基)-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环戊烷甲酰胺
(反式)3-环戊基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
(反式)2-环丙基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
(反式)叔丁基-2-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)吡咯烷-1-甲酸叔丁酯
(反式)4-羟基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺/反)N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-[1,1’-二苯基]-4-甲酰胺
(顺式)2-甲基-2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
(顺式)2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
(反式)2-溴-5-甲氧基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)2-溴-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)烟酰胺
(顺/反)3-氟-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)四氢呋喃-3-甲酰胺
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)呋喃-3-甲酰胺
(反式)(1r,3r)-N-(萘乙酰胺-2-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-甲酰胺
3-(苄氧基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
(顺/反)3-丁氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
(顺/反)3-乙氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
(顺/反)3-甲氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
(反式)1-苯基-2-((1r,3r)-螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基)乙醇
(反式)(1r,3r)-N-(3-溴-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-(对甲苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-(3-氯-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰
(反式)(1r,3r)-N-(3-氟苯基)螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-(3-氯苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)--N-(3-溴苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(顺式)(1r,3S,8a'S)-N-(3-溴苯基)-8a'H-螺[环丁基-1,8'-茚并[1,2-c]吡咯]-3-甲酰胺
N-(4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-(萘-2-基磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-((4-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-((3-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-N-((3-氯苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
(反式)(1r,3r)-3-(甲硫基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
(反式)(1r,3r)-3-(甲基磺酰基)螺[环丁烷,5’-咪唑并[5,1-a]异吲哚]
(反式)(1R,3r)-3-((R)-甲基亚磺酰基)螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]
(顺/反)3-(1-苄基-1H-1,2,3-三唑-4-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲 哚]-3-醇
(反式)(1r,3r)-3-(4-环己基-1氢-1,2,3三唑1-基)螺[c环丁烷-1,5’-咪唑[5,1-a]异吲哚]
(反式)(1r,3r)-3-(4-苯基-1H-1,2,3三唑-1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
(反式)(1r,3r)-3-(4-环丙基1H-1,2,3三唑1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
(反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
(反式)3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)磺酰脲
(反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
(顺式)1-(3-氟苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
(顺式)1-(4-氯苯基)-3-(1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
(顺式)1-(4-三氟甲基苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
(顺式)1-((1s,3s)-螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]-3-基)-3-(4-(三氟甲基)苯基)硫脲
(顺式)1-(3-氯-4-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
(顺式)1-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-3-(3-(三氟甲基)苯基)脲
(顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
(顺式)1-(4-氯-3-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
(顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
(顺式)1-(3,4-二氯苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
(顺式)1-苄基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-基)脲
(顺式)1-苯乙基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)脲
(反式)1-(3-三氟甲基苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚] -3-基)硫脲
(反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
(反式)1-(4-氯苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
(反式)4-氯-氮-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)苯磺酰胺
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)苯磺酰胺
(反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
(顺式)1-甲基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
(顺式)2-氰基-1-甲基-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)胍
(顺/反)1-(螺[环己基-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(3-(三氟甲基)苯基)硫脲
(顺/反)1-苯基-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
(顺/反)1-(4-氯苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
(顺/反)1-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(4-(三氟甲基)苯基)硫脲
(顺/反)1-(4-氯-3-(三氟甲基)苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)脲
(顺/反)N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)萘基-2-磺酰胺
(顺/反)3-溴-N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)苯甲酰胺
(顺/反)4-(1-苄基-1H-1,2,3-三唑-4-基)螺[环己烷1,5’-咪唑并[5,1-a]异吲哚]-4-醇
(顺式)(1s,3s)-螺[环丁烷-1,5’-咪唑并[5,1-a]异吲哚]-3-胺
(顺式)(1r,3r)-3-(1-苯基-1H-1,2,3-三唑-4-基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-醇
1'-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
(顺式)(1s,3s)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
(顺式)(1r,3r)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
(顺/反)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-胺
(顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)苯甲酰胺
(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)(螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-基)甲酮
(反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
(顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)脲
(顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-氨甲酰
(顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)苯磺酰胺
(反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
(顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)硫脲
N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-硫代酰胺
(反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
(顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
1'-((3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)磺酰)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
(顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
(反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺。
Figure PCTCN2017108941-appb-000029
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000030
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000031
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000032
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000033
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000034
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000035
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000036
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000037
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000038
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000039
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
Figure PCTCN2017108941-appb-000040
R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
在另一优选例中,所述药学上可接受的盐选自下组:盐酸盐、氢溴酸盐、硫酸盐、磷酸盐、甲磺酸盐、三氟甲磺酸盐、苯磺酸盐、对甲苯磺酸盐(甲苯磺酸盐)、1-萘磺酸盐、2-萘磺酸盐、乙酸盐、三氟乙酸盐、苹果酸盐、酒石酸盐、柠檬酸盐、乳酸盐、草酸盐、琥珀酸盐、富马酸盐、马来酸盐、苯甲酸盐、水杨酸盐、苯乙酸盐、扁桃酸盐。
在另一优选例中,所述的化合物是实施例中所制备的化合物1-117。
在另一优选例中,所述的化合物是顺反异构体。
在另一优选例中,所述的化合物是顺式异构体。
在另一优选例中,所述的化合物是反式异构体。
在另一优选例中,所述的化合物是外消旋体。
在另一优选例中,所述的化合物是对映异构体。
本发明第二方面,提供一种药物组合物,所述药物组合物包括:
(1)如本发明第一方面所述的式(I)化合物、或其药学上可接受的盐、其立体异构体或其互变异构体、或其前药;
(2)药学上可接受的载体和/或抗肿瘤药物。
在另一优选例中,所述抗肿瘤药物选自下组:检查点蛋白抗体,如PD-1抗体、PD-L1抗体或CTLA4抗体。
在另一优选例中,所述抗肿瘤药物为癌症的免疫治疗药物。
在另一优选例中,所述癌症的免疫治疗药物选自下组:PD-1抗体、CTLA-4抗体、PD-L1抗体、PD-L2抗体、化疗药物或靶向治疗药物。
在另一优选例中,所述靶向治疗药物选自下组:HDAC抑制剂、激酶抑制剂、EP4拮抗剂、或其组合。
本发明第三方面,提供一种如本发明第一方面所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药的用途,用于:
(i)制备IDO/TDO抑制剂;
(ii)制备预防和/或治疗IDO和TDO介导的疾病的药物。
在另一优选例中,所述IDO/TDO介导的疾病为IDO/TDO介导的色氨酸代谢途径的病理学特征的疾病。
在另一优选例中,所述IDO/TDO介导的疾病选自下组:癌症、神经退行疾病、眼疾、心里障碍、抑郁症、焦虑症、老年痴呆症、或自身免疫性疾病。
在另一优选例中,所述癌症包括但不限于:结肠癌、乳腺癌、胃癌、肺癌、大肠癌、胰腺癌、卵巢癌、前列腺癌、肾癌、肝癌、脑癌、黑色素瘤、多发性骨髓瘤、慢性粒细胞性白血病、血液肿瘤、淋巴肿瘤,包括在其他远离肿瘤原发部位的组织或器官的转移 病变。
本发明第四方面,提供一种预防和/或治疗IDO/TDO介导的疾病的方法,包括对患者给予第一方面所述的式(I)化合物或第二方面所述的药物组合物的步骤。
在另一优选例中,所述IDO/TDO介导的疾病为癌症,所述方法进一步包含对患者施用额外的抗癌剂(也称为抗肿瘤药物,所述抗肿瘤药物如上所述)的步骤。
应理解,在本发明范围内中,本发明的上述各技术特征和在下文(如实施例)中具体描述的各技术特征之间都可以互相组合,从而构成新的或优选的技术方案。限于篇幅,在此不再一一赘述。
具体实施方案
本发明人经过广泛而深入地研究,首次研发出一种新型的含有氮杂环螺旋结构的化合物,该化合物可作为高效的含有氮杂环螺旋结构抑制剂,用于预防和/或治疗IDO/TDO介导的疾病,也可作为抗炎药物使用。在此基础上,完成了本发明。
定义
术语“烷基”是指一价饱和脂族烃基,具有1至10个碳原子,包括直链和支链烃基,如甲基(即CH3-)、乙基(即CH3CH2-)、正丙基(即CH3CH2CH2-)、异丙基(即(CH3)2CH-)、正丁基(即CH3CH2CH2CH2-)、异丁基(即(CH3)2CHCH2-)、仲丁基(即(CH3)(CH3CH2)CH-)、叔丁基(即(CH3)3C-)、正戊基(即CH3CH2CH2CH2CH2-)、新戊基(即(CH3)3CCH2-)。在本发明中,该术语包括取代或未取代的烷基。
如本文所用,术语“取代或未取代的”指所述基团可以是未取代的,或者所述基团中的H被一个或多个(较佳地1-6个,更佳地1-3个)取代基所取代。
如本文所用,所述的“取代”或“取代的”指所述基团具有一个或多个(较佳地1-6个,更佳地1-3个)选自下组的取代基:卤素、羟基、-NH2、硝基、-CN、C1-C4烷基、C1-C4卤代烷基、C1-C4烷氧基、C3-C6环烷基、C2-C4链烯基、C2-C4炔基、苯基、苄基。
如本文所用,术语“环烷基”指取代或未取代的C3-C12环烷基。
如本文所用,术语“烷氧基”指-O-烷基,其中所述烷基可以是饱和或不饱和的、可以是支链、直链的、或环状的。优选地,烷氧基具有1-10个碳原子,较佳地1-6个碳原子。代表性的例子包括(但并不限于):甲氧基、乙氧基、丙氧基。
如本文所用,术语“芳基”是指6至20个(较佳6-14个)碳原子的单价芳香族碳环基团,它具有单环(如苯基)或稠环(如萘基或蒽基),如果连接点在芳香碳原上,稠环可能是非芳香性的(如2-苯并噁唑酮,2H-1,4-苯并噁嗪-3(4H)-酮-7-基等)。优选的芳基 包括苯基和萘基。该术语包括取代或未取代的形式,其中取代基的定义如上。
如本文所用,术语“烯基”是指具有2至10(如2至6或2至4)个碳原子的烯基,且具有至少1(如1至2)个不饱和烯键(>C=C<)。这类基团的例如有乙烯基、烯丙基、丁-3-烯基。如本文所用,术语“环烷基”是指具有3至10个碳原子的、具有单环或多环(包括稠合体系,桥环体系和螺环体系)的环状烷基。在稠环体系中,一个或多个环可以是环烷基、杂环的、芳基或杂芳基,只要连接位点是通过环烷基的环。合适的环烷基的例子包括:例如,金刚烷基、环丙基、环丁基、环戊基和环辛基。
如本文所用,术语“卤代”或“卤素”是指氟、氯、溴和碘。
如本文所用,术语“杂芳基”是指环内具有1至10个碳原子和1至4个选自氧、氮和硫的杂原子的芳香基团,这样的杂芳基可以是单环的(如吡啶基或呋喃基)或稠环(如吲嗪基(indolizinyl)或苯并噻吩基),其中,所述稠环可以是非芳香性的和/或含有一个杂原子,只要连接点是通过芳香性杂芳基的原子。在一实施例中,杂芳基的环原子氮和/或硫任选地被氧化为N-氧化物(N-O),亚磺酰基或磺酰基。优选地杂芳基包括吡啶基、吡咯基、吲哚基、苯并恶二唑、咪唑基、喹啉基、、恶唑基、异恶唑基、噻唑基、四氮唑基、1,2,3-三氮唑基、1,2,4-三氮唑基、1,3,4-噁二唑、1,2,4-噁二唑、1,2,5-噁二唑、噻吩基和呋喃基。该术语包括取代或未取代的杂芳基。
如本文所用,术语“取代的杂芳基”是指被1至5个、优选1至3个、更优选1至2个的取代基所取代的杂芳基,所述取代基选自与取代的芳基所定义的相同取代基。
如本文所用,术语“杂环”或“杂环的”或“杂环烷基”或“杂环基”是指饱和的、部分饱和的或不饱和的基团(但不是芳香性的),具有单环或稠环(包括桥环体系和螺环体系,环内具有1至10个碳原子和1至4个选自氮、硫或氧的杂原子,在稠环体系中,一个或多个环可以是环烷基、芳基或杂芳基,只要连接点通过非芳香性环。在一实施例中,杂环基团的氮原子和/或硫原子任选地被氧化,以提供N-氧化物、亚磺酰基和磺酰基部分。
如本文所用,术语“取代的杂环的”或“取代的杂环烷基”或“取代的杂环基”是指被1到5(如1至3)个取代基所取代的杂环基团,所述取代基与取代的环烷基所定义的取代基相同。
如本文所用,术语“立体异构体”是指一个或多个立体中心的手性不同的化合物。立体异构体包括对映异构体和非对映异构体。
如本文所用,术语“互变异构体”是指质子位置不同的化合物的替代形式,如烯醇-酮和亚胺-烯胺互变异构体,或杂芳基的互变异构形式,所述杂芳基包含与环的-NH-部分和环的=N-部分连接的环原子,如吡唑、咪唑、苯并咪唑、三唑和四唑。
“前药”是指实施例化合物的任何衍生物,当被施用于受试者时,其能够直接或间接地提供实施例的化合物或其活性代谢物或残余物。特别优选的衍生物和前药是那些, 当被施用于受试者时,提高实施例化合物的生物利用度(如口服给药的化合物更容易被吸收进入血液)或相对于母体种类提高母体化合物到生物区室(如脑或淋巴系统)的运送的衍生物和前药。前药包括本发明化合物的酯类形式。
本发明化合物
如本文所用,术语“本发明化合物”指通式(I)化合物、其外消旋体、其立体异构体或其互变异构体、或其药学,上可以接受的盐。
本发明涉及:这些化合物的外消旋混合物,富集任一种对映体的混合物,以及任一种分离的对映体。对于本发明的范围,应当理解为,所述外消旋混合物指两种R和S对映体50%:50%的混合物。所述分离的对映体应理解为纯的对映体(即100%)或者高度富集某种对映体(纯度≥98%、≥95%、≥93%、≥90%、≥88%、≥85%、≥80%)的混合物。
在本发明所述的化合物有立体异构体存在的情况下,本发明包括化合物的所有立体异构体。
在本发明所述的化合物有互变异构体存在的情况下,本发明包括化合物的所有互变异构体。
本发明还包括所述化合物中的任何一个或多个氢原子被其稳定同位素氘取代而产生的氘代化合物。
制备方法
以下提供的制备方法仅作示例性说明,不作为对本发明化合物制备方法的进一步概括。并且,以下制备方法中提供了各新颖的中间体及其在制备本发明所述化合物中的应用。
本发明的式(I)化合物可由以下的制备方法获得:
Figure PCTCN2017108941-appb-000041
(a)化合物A在上述条件下与表氯醇反应得到化合物B;(b)化合物B用过量的碱水解得到化合物C;(c)化合物C与(PhO)2P(O)Cl反应得到重排产物化合物D;(d)化合物D用碱水解得到化合物E;(e)化合物E在上述反应条件下得到咪唑化合物F;(f)化合物F在钯催化剂作用下关环生成化合物G;(g)化合物G与MsCl反应生成化合物H;(h)化合物H与NaN3进行取代反应得到化合物I;(i)化合物I在Pd/C催化下氢化得到中间体化合物J。
顺式异构体N可用一下步骤合成:
Figure PCTCN2017108941-appb-000042
(a)化合物B部分水解得到酰胺化合物K;(b)化合物K氧化重排产生化合物L;(c)化合物L通过上述生成化合物J的同样反应步骤得到中间体化合物M和化合物N。
本发明的式(I)化合物可由以下的制备方法获得:
Figure PCTCN2017108941-appb-000043
本发明还提供另一式(I)化合物Q的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000044
(a)化合物H与NaCN反应得到化合物O;(b)化合物O用用碱水解得到化合物羧酸P;(c)化合物P与合适的胺在酰胺的合成条件下反应产生式(I)化合物Q。
顺式异构体T可用一下步骤合成:
Figure PCTCN2017108941-appb-000045
化合物M可以通过与合成化合物Q相同的反应步骤得到式(I)化合物T。
本发明还提供另一式(I)化合物X的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000046
(a)羧酸化合物S转化为甲酯;(b)甲酯化合物U氧化得到化合物V;(c)碱性条件下水解化合物V得到化合物W;(d)化合物W与合适的胺在酰胺的合成条件下反应产生化合物X。
本发明还提供另一式(I)化合物Z的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000047
(a)化合物G氧化得到化合物Y;(b)化合物Y通过金属试剂亲核加成反应得到式(I)化合物Z。
本发明还提供另一式(I)化合物Z的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000048
(a)化合物Y通过乙炔格氏试剂加成反应得到得到化合物AA;(b)化合物AA通过与与叠氮化合物的点击反应得到式(I)化合物BB。
本发明还提供另一式(I)化合物FF的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000049
(a)化合物H与NaO2SCH2CH2O2Me反应得到化合物CC;(b)化合物CC在碱性条件下进行消除反应得到化合物DD;(c)化合物DD与H2NOSO3H反应得到化合物EE;(d)化合物EE与羧酸缩合得到式(I)化合物FF。
本发明还提供另一式(I)化合物HH的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000050
(a)化合物EE与ClC(O)OEt反应得到化合物GG;(b)化合物GG与相应的胺反应得到式(I)化合物HH。
本发明还提供另一式(I)化合物的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000051
(a)化合物O与Ti(Oi-Pr)4和EtMgBr反应得到化合物II;(b)化合物II与相应的试剂反应得到以上式(I)化合物。
本发明还提供另一式(I)化合物的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000052
(a)化合物JJ通过2-溴氰苄与3-氯丙酸甲酯在碱性条件下反应得到化合物JJ;(b)化合物JJ脱羧得到化合物KK;(c)化合物KK在酸催化条件下与乙二醇反应得到化合物LL;(d)化合物LL用碱部分水解得到化合物MM;(e)化合物MM在氧化反应条件下重排得到化合物NN;(f)化合物NN在上述反应条件下得到咪唑化合物OO;(g)化合物OO在钯催化剂作用下关环生成化合物PP;(h)化合物PP酸水解生成化合物QQ;(i)化合物QQ还原胺化得到化合物RR;
Figure PCTCN2017108941-appb-000053
与化合物J和化合物N类似,化合物RR与相应的试剂反应得到以上式(I)化合物。
本发明还提供另一式(I)化合物的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000054
(a)化合物JJ通过2-溴氰苄与双(2-氯乙基)氨基甲酸叔丁酯在碱性相催化条件下反应得到化合物SS;(b)化合物SS用碱部分水解得到化合物TT;(c)化合物TT在氧化反应条件下重排得到化合物UU;(d)化合物UU在上述反应条件下得到咪唑化合物VV;(e) 化合物VV在钯催化剂作用下关环生成化合物WW(i)化合物WW酸性条件下去保护得到化合物XX。
Figure PCTCN2017108941-appb-000055
与化合物RR类似,化合物XX与相应的试剂反应得到以上式(I)化合物。
本发明还提供另一式(I)化合物的制备方法,包括以下步骤:
Figure PCTCN2017108941-appb-000056
(a)化合物YY在碱性条件下与甲醛水溶液反应得到化合物ZZ;(b)化合物ZZ通过三氟甲磺酸基化和苄胺的取代关环得到化合物AAA;(c)化合物AAA水解后重排降解得到化合物BBB;(d)化合物BBB在上述反应条件下得到咪唑化合物CCC;(e)化合物CCC在钯催化剂作用下关环生成化合物DDD;(f)化合物DDD在钯催化氢化条件下脱苄基生成化合物EEE。
Figure PCTCN2017108941-appb-000057
与化合物XX类似,化合物EEE与相应的试剂反应得到以上式(I)化合物。
药物组合物
本发明还提供了一种药物组合物,它包含安全有效量范围内的活性成分,以及药学上可接受的载体。
本发明所述的“活性成分”是指本发明所述的式(I)化合物或其药学上可接受的盐、其立体异构体或其互变异构体、或其前药。
本发明所述的“活性成分”和药物组合物可用作IDO或TDO抑制剂。在另一优选例 中,用于制备预防和/或治疗肿瘤的药物。在另一优选例中,用于制备预防和/或治疗IDO或TDO介导的疾病的药物。
“安全有效量”指的是:活性成分的量足以明显改善病情,而不至于产生严重的副作用。通常,药物组合物含有1-2000mg活性成分/剂,更佳地,含有10-200mg活性成分/剂。较佳地,所述的“一剂”为一个药片。
“药学上可接受的载体”指的是:一种或多种相容性固体或液体填料或凝胶物质,它们适合于人使用,而且必须有足够的纯度和足够低的毒性。“相容性”在此指的是组合物中各组份能和本发明的活性成分以及它们之间相互掺和,而不明显降低活性成分的药效。
本发明优选实施例的化合物可以作为单独活性药剂给药,也可以与一个或多个其它用于治疗癌症的试剂组合使用。本发明优选实施例的化合物与已知的治疗剂和抗癌剂组合使用也是有效的,目前已知的化合物和其它抗癌剂或化疗剂的组合在优选实施例范围之内。这类药剂的例子可参见《癌症原理和实践肿瘤学》(Cancer Principles and Practice of Oncology),V.T.Devita和S.Hellman(编者),第6版(2001年2月15日),Lippincott Williams&Wilkins出版社。基于药物的特殊性质和所涉及的癌症,本领域普通技术人员能够辨别有效的药剂组合。这种抗癌剂包括(但不限于)如下:雌激素受体调节剂、雄激素受体调节剂、视黄醇类受体调节剂、细胞毒性/细胞生长抑制剂、抗增殖剂、异戊烯基蛋白转移酶抑制剂、取乙酰酶(HDAC)抑制剂、HMG-CoA还原酶抑制剂和其他血管生成抑制剂、细胞增殖和生存信号抑制剂、凋亡诱导剂和干扰细胞周期检查点(cell cycle checkpoint)的试剂、CTLA4抗体、PD-1抗体、PD-L1抗体等。优选实施例的化合物与放射治疗同时施用时也有效。
优选实施例的目的,治疗有效剂量通常可以是对患者一次性施用或分次施用的每日总剂量,例如,每日约0.001至约1000毫克/公斤体重,优选地,每日约1.0至约30毫克/千克体重。单位剂量组合物(Dosage unit composition)可包含其剂量因数以形成每日剂量。剂型的选择取决于各种因素,例如给药模式和药物物质的生物利用度。通常,优选实施例的化合物可作为药物组合物通过以下任意一种路线给药:口服、全身给药(如透皮、鼻内或通过栓剂)、或肠外给药(如肌内、静脉内或皮下)。优选的给药方式为口服,可根据苦的程度调节方便的日剂量。组合物可采取的形式为片剂、丸剂、胶囊、半固体、粉剂、缓释制剂、溶液、悬浮液、酏剂、气雾剂或任何其他适当的组合物。另一种优选的施用优选实施例化合物的方式为吸入。这是一种将治疗剂直接运送给呼吸道的有效方法(参见,如美国专利号5,607,915)。
合适的药学上可接受的载体或赋形剂包括:如处理剂和药物运送改性剂和促进剂,诸如磷酸钙、硬脂酸镁、滑石、单糖、二糖、淀粉、明胶、纤维素、甲基纤维素钠、羧甲基纤维素、葡萄糖、羟丙基-B-环糊精、聚乙烯吡咯烷酮、低熔点蜡、离子交换树脂 等,及其任意两种或多种的组合。液体和半固体的赋形剂可以选自甘油、丙二醇、水、乙醇和各种油,包括石油、动物油、植物油或合成来源,如花生油、豆油、矿物油、芝麻油等。优选的液体载体,特别是用于可注射溶液的载体,包括水、盐水、葡萄糖水性溶液和乙二醇。其它适宜的药学上可接受的赋形剂在《雷明顿药物科学》(Remington’’s Pharmaceutical Sciences),Mack Pub.Co.,新泽西(1991)有描述,通过引用纳入本文。
如本文所用,术语“药学上可接受的盐”是指通式I化合物的非毒性酸或碱土金属盐。这些盐可在最终分离和纯化通式I化合物时原位制得、或分别将合适的有机或无机酸或碱与碱性或酸性官能团反应制得。代表性的盐包括,但不限于:乙酸盐、己二酸盐、藻酸盐、柠檬酸盐、天冬氨酸盐、苯甲酸盐、苯磺酸盐、硫酸氢盐、丁酸盐、樟脑酸盐、樟脑磺酸盐、二葡糖酸盐、环戊烷丙酸盐、十二烷基硫酸盐、乙磺酸盐、葡萄糖庚酸盐、甘油磷酸盐、半硫酸盐、庚酸盐、己酸盐、富马酸盐、盐酸盐、氢溴酸盐、氢碘酸盐、2-羟基乙磺酸盐、乳酸盐、马来酸盐、甲磺酸盐、烟酸盐、2-萘基磺酸盐、草酸盐、双羟萘酸盐、果胶酸盐、硫氰酸盐、3-苯基丙酸盐、苦味酸盐、新戊酸盐、丙酸盐、琥珀酸盐、硫酸盐、酒石酸盐、硫氰酸盐、对甲苯磺酸盐和十一烷酸盐。此外,含氮的碱性基团可被如下试剂季铵盐化:烷基卤化物,如甲基、乙基、丙基、丁基的氯化物、溴化物和碘化物;二烷基硫酸盐,如二甲基、二乙基、二丁基和二戊基硫酸酯;长链卤化物如癸基、月桂基、肉豆蔻基和硬脂基的氯化物、溴化物和碘化物;芳烷基卤化物如苄基和苯乙基溴化物等。由此得到水溶性或油溶性或可分散产品。可被用于形成药学上可接受的酸加成盐的酸的例子包括如盐酸、硫酸、磷酸的无机酸,和如草酸、马来酸、甲磺酸、琥珀酸、柠檬酸的有机酸。碱加成盐可在最终分离和纯化通式I的化合物时原位制得、或使羧酸部分分别与合适的碱(如药学上可接受的金属阳离子的氢氧化物,碳酸盐或碳酸氢盐)或氨、或有机伯、仲或叔胺反应制得。药学上可接受的盐包括,但不限于,基于碱金属和碱土金属的阳离子,如钠、锂、钾、钙、镁、铝的盐等,以及无毒的铵、季铵和胺阳离子,包括,但不限于:铵、四甲基铵、四乙基铵、甲胺、二甲胺、三甲胺、三乙胺、乙胺等。其它代表性的用于形成碱加成盐的有机胺包括二乙胺、乙二胺、乙醇胺、二乙醇胺、哌嗪等。
如本文所用,术语“药学上可接受的前药”是指那些优选实施例的化合物的前药,在体内迅速转化为上述通式所示的母体化合物的化合物,例如在血液中水解。在“T.Higuchi和V.Stella,作为新型运送系统的前药(Pro-drugs as Novel Delivery Systems),A.C.S.15 Symposium Series的14卷”和“Edward B.Roche编,药物设计中的生物可逆载体(Bioreversible Carriers in Drug Design),美国药学协会和Pergamon出版社,1987年”中提供了完整的讨论,这两者都引入本文作为参考。
下面结合具体实施例,进一步阐述本发明。应理解,这些实施例仅用于说明本发明而不用于限制本发明的范围。下列实施例中未注明具体条件的实验方法,通常按照常规条件如Sambrook等人,分子克隆:实验室手册(New York:Cold Spring Harbor Laboratory Press,1989)中所述的条件,或按照制造厂商所建议的条件。除非另外说明,否则百分比和份数按重量计算。
本发明的有益之处在于:
(1)提供一种结构新颖的式(I)化合物;
(2)本发明的化合物可以作为高效的IDO/TDO酶抑制剂;
(3)合成方法温和,操作简单易行,收率较高,易于衍生化,适合工业放大量生产;
(4)具有抗肿瘤、神经退行性疾病(阿尔茨海默病)、抗炎等多种药理活性。
除非另行定义,文中所使用的所有专业与科学用语与本领域熟练人员所熟悉的意义相同。此外,任何与所记载内容相似或均等的方法及材料皆可应用于本发明方法中。文中所述的较佳实施方法与材料仅作示范之用。
实施例1
(顺/反)3-苯基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇的制备
Figure PCTCN2017108941-appb-000058
第一步:(顺/反)1-(2-溴苯基)-3-羟基环丁烷腈
Figure PCTCN2017108941-appb-000059
将化合物2-溴苄氰(10.2g,52.0mmol)加入反应瓶中,冷却到零下78度,滴加甲基锂(44mL,52.0mmol),滴加完毕后,此温度下搅拌1小时,加入环氧氯丙烷(5.3g,57.2mmol),此温度下搅拌3小时,TLC检测原料消失,然后滴加甲基溴化镁格式试剂(19ml,57.2mmol),滴加完毕后,自然回到室温,然后缓慢升温到60度,搅拌过夜。TLC监测反应已完全。冰浴冷却下,滴加2N的盐酸溶液,反应体系pH调整到7左右。加入乙酸乙酯(150mL)和水(150mL),萃取,水相再用乙酸乙酯(100mLx2)萃取,合并有机相,用硫酸钠固体干燥。过滤浓缩,柱层析纯化分离,得到黄色液体目标化合物11.5g(顺/反=2:1.4),收率88%。
MS ESI:m/z=253.1,[M+H]+
第二步:(顺/反)1-(2-溴苯基)-3-羟基环丁烷甲酸
Figure PCTCN2017108941-appb-000060
将第一步的产物(24.0g,197.0mmol)溶于230mL乙醇中,分别加入水230mL,氢氧化钾(53.3g,952.0mmol),加热到105度,反应24小时,原料完全消失。冷却到冰浴,用6N盐酸调节PH到4-5,加入200mL乙酸乙酯,萃取,再用100mL乙酸乙酯萃取2次,合并有机相,干燥,过滤,浓缩,得到白色固体目标化合物20.8g(顺/反=2:1.4),收率81%。
MS ESI:m/z=272.1,[M+H]+
第三步:(顺式)5-(2-溴苯基)-2-氧代-4-氮杂双环[3.1.1]庚-3-酮
Figure PCTCN2017108941-appb-000061
将第二步的产物(10.6g,39.1mmol)溶于100mL二氧六环,叠氮磷酸二苯酯(13.0g,46.9mmol)加入反应液中,再加入二异丙基乙胺(10.1g,78.2mmol),以及叔丁醇(50mL),置换氩气数次,搅拌十分钟后,加热到80度,反应4小时。冷却到室温,反应体系中有白色固体析出,过滤得到目标化合物7.7g,收率73%。
1H NMR(500MHz,DMSO-d6):δ8.22(s,1H),7.60(d,1H),7.40(t 1H),7.26-7.32(m,2H),4.88(s,1H),2.51(d,2H),2.28(d,2H).
MS ESI:m/z=267.1,[M+H]+
第四步:(顺式)3-氨基-3-(2-溴苯基)环丁醇
Figure PCTCN2017108941-appb-000062
将第三步的产物(7.7g,28.7mol)加入100mL水和100mL异丙醇,加入氢氧化钾(80.0g,1.4mol),回流反应24小时。冷却到室温,用二氯甲烷150mL萃取3次,合并有机层,干燥,旋干溶剂,干燥,得呈黄色油状物的目标化合物(7.5g,收率100%)。
1H NMR(500MHz,DMSO-d6):δ8.22(s,1H),7.60(d,1H),7.40(t 1H),7.26-7.32(m,2H),4.88(s,1H),2.51(d,2H),2.28(d,2H).
MS ESI:m/z=243.1,[M+H]+
第五步:(顺式)3-(2-溴苯基)-3-(1H-咪唑-1-基)环丁醇
Figure PCTCN2017108941-appb-000063
将第四步的产物(5.0g,20.7mmol)溶于50mL甲醇中,分别加乙二醛(6.0g,41.3mmol)甲醛(3.4g,41.3mmol),乙酸铵(3.2g,41.3mmol)。加毕,反应缓慢升温到70度,反应4小时,反应结束后,直接浓缩,拌样,柱层析分离,得黄色固体目标化合物(5.5g,收率91%)。
1H NMR(500MHz,甲醇-d4):δ7.88-7.86(m,1H),7.68-7.67(m,1H),7.59(s,1H),7.59-7.53(m,1H),7.34-7.30(m,1H),7.04(s,1H),6.94(s,1H),4.05-4.03(m,2H),3.50-3.46(m,2H),2.84-2.80(m,2H).
MS ESI:m/z=294.1,[M+H]+
第六步:(顺式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000064
将第五步的产物(2.5g,8.5mmol)放入反应瓶中,加入二甲亚砜(50mL),置换氩气数次,加入二氯二茂铁氯化钯(624mg,0.853mmol),加入碳酸铯(5.6g,17.1mmol),再次置换氩气数次后,加热到120度,反应5小时。反应用LCMS监测,原料消失后,冷却至室温,减压蒸馏出去二甲亚砜,然后加入乙酸乙酯(100mL)和水(50mL),萃取,水相再用乙酸乙酯(50mL)萃取,合并有机相。柱层析纯化分离,得到目标化合物(1.3g,收率70%)。
1H NMR(500MHz,甲醇-d4):δ8.11(s,1H),7.56-7.52(m,2H),7.35-7.33(m,2H),7.08(s,1H),4.80-4.77(m,1H),3.08-3.03(m,2H),2.4-2.69(m,2H),MS ESI:m/z=213.1,[M+H]+
第七步:螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-酮
Figure PCTCN2017108941-appb-000065
将第六步的产物(0.5g,2.4mmol),放入瓶中,加入5mL二氯甲烷,冰浴下搅拌,加入碳酸氢钠(1.9g,23.4mmol),加入Dess-Martin氧化剂(1.5g,3.5mmol),室温搅拌,知道原料消失。加入20mL二氯甲烷和10mL水萃取,得到有机相,干燥,过滤,浓缩,柱层析纯化。得到目标化合物(400mg,收率80%)。
1H NMR(500MHz,甲醇-d4):δ7.86(s,1H),7.59(d,1H),7.47-7.42(m,2H),7.37-7.35(m,1H),7.25(s,1H),3.872.82(m,4H),
MS ESI:m/z=211.1,[M+H]+
第八步:(顺/反)3-苯基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000066
在反应瓶中加入1mL无水四氢呋喃,加入无水氯化铈(450mg,1.5mmol)加入苯基溴化镁(1.5mL 1.5mmol),室温下搅拌1小时,冰浴下,将第七步的产物(100mg,0.5mmol)的四氢呋喃溶液滴加到上述反应液中,滴毕,室温反应2小时,TLC显示原料消失。滴加饱和氯化铵淬灭反应。加入乙酸乙酯20mL和水10mL,萃取,有机相干燥,过滤,浓缩,得到粗品,柱层析纯化分离,得到目标化合物(100mg,收率70%)。
1H NMR(500MHz,甲醇-d4):δ8.43(s,1H),7.62(d,2H),7.48-7.44(m,2H),7.37-7.33(m,2H),7.27(d,1H),7.14(s,1H),3.49(d,2H),2.92(d,2H).
MS ESI:m/z=289.3,[M+H]+
实施例2
(顺/反)(3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲醇
Figure PCTCN2017108941-appb-000067
第一步:(顺式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基甲磺酸酯
Figure PCTCN2017108941-appb-000068
(顺式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇(212mg,1.0mmol)溶于5mL二氯甲烷中,加入三乙胺(202mg,2.0mmol),滴加甲烷磺酰氯(216mg,1.5mmol),滴毕,自然回到室温,共计反应1小时。加入饱和食盐水10mL,分别加入二氯甲烷2x20mL,进行萃取,合并有机相,得到目标化合物(300mg,收率100%)。
MS ESI:m/z=291.0,[M+H]+.
第二步:螺[环丁烷1,5’-咪唑并[5,1-a]是吲哚]-3-甲腈
Figure PCTCN2017108941-appb-000069
第一步产物(300mg,1.0mmol),溶于10mLDMF中,加入氰化钠(150mg,3.0mmol),加热到100度,反应10小时。原料消失。冷却到室温后,加入30mL乙酸乙酯,用10mL水洗5次,有机相用硫酸镁干燥,过滤,浓缩,柱层析,得到目标化合物粗品(220mg,收率100%)。
MS ESI:m/z=222.1,[M+H]+
第三步:螺[环丁烷1,5’-咪唑并[5,1-a]是吲哚]-3-羧酸
Figure PCTCN2017108941-appb-000070
第二步产物(220mg,1.0mmol)加入乙醇中(10mL),再加入水(10mL),加入氢氧化钾(1.1g,20.0mmol),加热到100度,反应10小时。TLC显示原料已消失。冷却到室温,加入2N盐酸调节PH到6-7,旋干溶液,用10%的二氯甲烷甲醇溶液溶解产物,过滤掉里面的固体,收集滤液。合并,干燥,过滤,浓缩。得到目标化合物粗品(200mg,收率83%)。
MS ESI:m/z=241.1,[M+H]+
第四步:N-甲氧基-氮-甲基-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000071
将第三步产物(48mg,0.2mmol)加入二氯甲烷中,加入三乙胺(44mg,0.4mmol),加入N,O-二甲基盐酸盐(20mg,0.2mmol),加入EDCI(76mg,0.4mmol),HOBT(27mg,0.2mmol)室温反应过夜,原料消失。反应液旋干,直接柱层析纯化得到目标化合物(56mg,收率100%)。
MS ESI:m/z=284.2,[M+H]+
第五步:(反式)3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲酮
Figure PCTCN2017108941-appb-000072
将第四步产物(60mg,0.2mmol)放入反应瓶中,加入四氢呋喃(2mL),在冰浴下冷却,滴加3-氟苯基溴化镁(0.9mL,0.90mmol),滴毕,直到原料消失,加入饱和氯化铵(2mL)淬灭反应,加入乙酸乙酯(2x10mL)萃取,和并有机相,用硫酸钠干燥,过滤,浓缩得到粗品。柱层析纯化,得到50mg反式产物,将其(50mg,0.16mmol)溶于四氢呋喃(2mL)中,冰浴下,加入DBU(62mg,0.3mmol),搅拌1小时。TLC显示有2个很近的点,LCMS显示分子量一样,用饱和氯化铵(1mL)淬灭反应,乙酸乙酯萃取(2x5mL),和并有机相,硫酸钠干燥,过滤,弄得,得到顺反目标化合物50mg,收率100%。
第六步:(顺/反)(3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲酮
Figure PCTCN2017108941-appb-000073
将第五步产物(50mg,0.2mmol)溶于四氢呋喃(2mL)中,冰浴下,加入DBU(62mg,0.3mmol),搅拌1小时。TLC显示有2个很近的点,LCMS显示分子量一样,用饱和氯化铵(1mL)淬灭反应,乙酸乙酯萃取(2x5mL),和并有机相,硫酸钠干燥,过滤,弄得,得到粗品50mg,收率100%。
第七步:(顺/反)-(3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲醇
Figure PCTCN2017108941-appb-000074
将第六步产物(50mg,0.2mmol)加入反应瓶中,加入四氢呋喃(2mL),冰浴下,加入硼氢化钠(12mg,0.3mmol),直到反应完全。直接拌样,旋干,柱层析,得到化合物2,白色固体21mg,收率63%。
1H NMR(400MHz,CDCl3):δ7.89(s,1H),7.58-7.48(m,2H),7.35-7.31(m,3H),7.24-7.14(m,3H),7.03-6.96(m,1H),5.30(s,1H),4.95-4.90(m,1H),3.29-2.12(m,2H),2.99-2.95(m,2H).
MS ESI:m/z=321.1,[M+H]+
实施例3
(顺/反)3-(1,4-二氧杂螺[4.5]癸-7-烯-8-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000075
根据实施例1的第八步中的条件用1,4-二氧杂螺[4.5]癸-7-烯-8-锂代替苯基溴化镁作为原料,得到目标化合物,白色固体(10mg,收率29%)。
1H NMR(400MHz,CDCl3):δ8.35(s,1H),7.42-7.41(m,1H),7.37-7.35(m,1H),7.34-7.31(m,1H),7.27-7.25(m,1H),7.18(s,1H),5.75(s,1H),3.86(s,4H),3.15(d,2H),2.64(d,2H),2.40-2.36(m,4H),1.83-1.80(m,2H).
MS ESI:m/z=351.2,[M+H]+
实施例4
(顺式)4-甲基-N-((1s,3s)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000076
第一步:(反式)(1r,3r)--螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基苯甲酸酯
Figure PCTCN2017108941-appb-000077
室温下将三苯基膦(393mg,1.5mmol)、苯甲酸(92mg,0.8mmol)和(順式)螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-醇(159mg,0.8mmol)加入到50mL两口瓶中,抽换Ar2三次,注入4mL四氢呋喃。然后冰浴下将DIAD(273mg,1.4mmol)的THF(2mL)溶液滴加到上述体系中,升至室温反应4h,停止反应。反应液浓缩,经柱纯化(流动相DCM:MeOH=20:1)得到浅红色固体目标化合物187mg,收率79%。
1H NMR(400MHz,CDCl3):δ8.01(s,1H),7.70-7.65(m,4H),7.57-7.53(m,3H),7.39(t,1H),7.34(t,1H),7.27(s,1H),5.79(quint,1H),3.16(m,4H).
MS ESI:m/z=317.1,[M+H]+.(1r,3r)-spiro[cyclobutane-1,5′-imidazo[5,1-a]isoindol]-3-ol
第二步:反-(1r,3r)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000078
室温下将LiOH-H2O(243mg,5.8mmol)/H2O(2mL)水溶液滴加到第一步产物(610mg, 1.9mmol)/MeOH(15.0mL)溶液中。室温下反应10分钟后,将反应液浓缩并用3x5mLEA萃取上述溶液,合并有机相用无水Na2SO4干燥。反应液浓缩,经柱纯化(流动相DCM:MeOH=20:1)得到浅褐色固体目标化合物316mg,收率77%。
1H NMR(400MHz,CDCl3):δ7.76(s,1H),7.66(dd,1H),7.49(dd,1H),7.37-7.31(m,2H),7.16(s,1H),4.99(q,1H),2.90(m,4H).
MS ESI:m/z=213.1,[M+H]+.
第三步:(反式)-(1r,3r)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基甲磺酸酯
Figure PCTCN2017108941-appb-000079
冰浴下将MsCl(339mg,3.0mmol)和Et3N(599mg,5.9mmol)依次滴加到第二步产物(315mg,1.5mmol)/DCM(10mL)溶液中。0℃下反应10分钟后,往反应液中加入2mL饱和食盐水并用3x5mL DCM萃取上述溶液,合并有机相并用无水Na2SO4干燥旋干得到浅褐色固体目标化合物412mg,收率96%。
1H NMR(400MHz,CDCl3):δ7.79(s,1H),7.62(dd,1H),7.50(dd,1H),7.40-7.35(m,2H),7.18(s,1H),5.57(quint,1H),3.25-3.20(m,2H),3.14(s,3H),3.13-3.09(m,2H).
MS ESI:m/z=291.0,[M+H]+.
第四步:(顺式)-(1s,3s)-3-叠氮基-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000080
将第三步产物(300mg,1.0mmol),溶于10mL DMF中,加入叠氮化钠(200mg,3.0mmol),加热到100度,反应3小时。冷却到室温后,加入30mL乙酸乙酯,用10mL水洗5次,有机相用硫酸镁干燥,过滤,浓缩,柱层析,得到目标化合物(220mg,收率92%)。
MS ESI:m/z=238.1,[M+H]+.
第五步:(顺式)(1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-胺
Figure PCTCN2017108941-appb-000081
将第四步产物(220mg,0.9mmol)溶于乙醇中,加入Pd/C(20mg,10%),通入氢气, 反应过夜。滤掉Pd/C,浓缩滤液,得到目标化合物粗品(190mg,收率96%)。
目标化合物也可通过下面的条件得到:氩气氛围中,室温下将5%Pd/C(20mg)加入到第四步产物顺-3-叠氮螺[环丁烷基-1,5’-咪唑并[5,1-α]异吲哚](209mg,0.9mmol)的EtOH(4mL)溶液中。1atm H2中室温反应24h后,加入硅藻土过滤,滤饼用5mL EtOH洗涤,合并有机相,旋干得到浅黄色固体目标化合物170mg,收率87%。
1H NMR(400MHz,CDCl3):δ7.92(s,1H),7.49(t,2H),7.35(t,1H),7.28(t,1H),7.15(s,1H),3.95(q,1H),3.12-3.04(m,2H),2.52-2.47(m,2H).
MS ESI:m/z=212.1,[M+H]+
第六步:(顺式)4-甲基-N-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000082
将第五步产物(21mg,0.1mmol)置于烧瓶中,依次加入二氯甲烷(5mL)中,三乙胺(22mg,0.2mmol),对甲基苯磺酰氯(19mg,0.1mmol)。反应2小时后,反应液旋干,直接柱层析,纯化得到目标化合物(20mg,收率54%)。
1H NMR(500MHz,CDCl3):δ7.85(d,3H),7.55(d,1H),7.45(d,1H),7.35-7.32(m,3H),7.27-7.24(m,1H)7.16(s,1H),6.52(s,1H),4.35(s,1H),2.89-2.83(m,2H),2.76-2.73(m,2H),2.47(s,3H).
MS ESI:m/z=366.1,[M+H]+
实施例5
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000083
第一步:(反式)(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-胺
Figure PCTCN2017108941-appb-000084
以实施例2第二步产物(順式)螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-基甲磺酸酯为原料,根据实施例4第四步和第五步所描述的反应条件得到目标化合物。
1H NMR(500MHz,CDCl3):δ7.81(s,1H),7.59(d,1H),7.48(d,1H),7.31-7.36(dd,2H),7.16(s,1H),4.14-4.18(m,1H),2.84-2.89(m,2H),2.59-2.65(m,2H)。
MS ESI:m/z=212.0,[M+H]+.
第二步(反式)4-甲基-N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000085
以第一步产物为原料,根据实施例4第六步所描述的反应条件得到目标化合物。
1H NMR(500MHz,CDCl3):δ7.90(s,1H),7.83(d,2H),7.48(d,1H),7.44(d,1H),7.38-7.30(m,4H),7.14(s,1H),5.61(s,1H),4.24-4.23(m,1H),2.98-2.94(m,2H),2.67-2.70(m,2H),2.42(s,3H).
MS ESI:m/z=366.1,[M+H]+
实施例6
(顺式)4-甲基-N-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000086
1H NMR(500MHz,CDCl3):δ8.00(s,1H),7.76(d,2H),7.71(d,1H),7.63(d,1H),7.53-7.51(m,2H),7.39-7.35(m,3H),7.16(s,1H),7.09(d,1H),4.99-4.93(m,1H),3.23-3.18(m,2H),3.04-2.99(m,2H).
MS ESI:m/z=330.2,[M+H]+
实施例7
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000087
1H NMR(500MHz,CDCl3):δ7.98(s,1H),7.76(d,2H),7.65(d,1H),7.51(d,1H),7.39-7.36(m,1H),7.33-7.28(m,2H),6.75(s,1H),5.09-5.07(m,1H),3.20-3.14(m,2H),3.03-2.98(m,2H).
MS ESI:m/z=330.2,[M+H]+
实施例8
(顺/反)3-氟-N-(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000088
MS ESI:m/z=334.1,[M+H]+.
实施例9
(顺式)4-氟-N-(1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000089
MS ESI:m/z=370.1,[M+H]+.
实施例10
(反式)4-氟-N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000090
MS ESI:m/z=370.1,[M+H]+.
实施例11
(反式)N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己烷甲酰胺
Figure PCTCN2017108941-appb-000091
1H NMR(500MHz,CDCl3):δ7.94(s,1H),7.60(d,1H),7.49(d,1H),7.36-7.28(m,2H),7.18(s,1H),4.91-4.85(m,1H),3.05-3.00(m,2H),2.93-2.88(m,2H),2.18-1.13(m,1H),1.93-1.90(m,2H),1.84-1.81(m,2H),1.51-1.48(m,2H),1.48-1.43(m,2H),1.31-1.28(m,2H).
MS ESI:m/z=322.2,[M+H]+
实施例12
(顺式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己基磺酰胺
Figure PCTCN2017108941-appb-000092
1H NMR(500MHz,CDCl3):δ8.03(s,1H),7.67(d,1H),7.50(d,1H),7.44-7.37(m,3H),7.13(s,1H),5.05-5.01(m,1H),3.67-6.61(m,1H),3.53-3.47(m,2H),3.20-3.17(m,2H),2.24-2.27(m,2H),1.75-1.78(m,2H),1.58-1.55(m,2H),1.52-1.49(m,2H),1.25-1.30(m,2H).
MS ESI:m/z=358.2,[M+H]+
实施例13
(顺式)3-氯-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000093
1H NMR(500MHz,CDCl3):δ8.18(s,1H),8.16(d,1H),7.84(d,2H),7.64(d,1H),7.49(d,1H),7.38-7.35(m,1H),7.30-7.27(m,1H),7.22-7.15(m,2H),5.29(s,1H),3.29-3.23(m,2h),2.98-2.95(m,2H).
MS ESI:m/z=368.8,[M+H]+
实施例14
(反式)3-溴-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000094
1H NMR(400MHz,CDCl3):δ8.36(d,1H),8.15-8.12(m,2H),7.86-7.83(m,1H),7.67(d,1H),7.52(d,1H),7.41-7.31(m,3H),7.23-7.14(m,2H),5.08-5.02(m,1H),3.28-3.23(m,2H),2.99-2.96(m,2H).
MS ESI:m/z=413.7,[M+H]+
实施例15
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷磺酰胺
Figure PCTCN2017108941-appb-000095
1H NMR(500MHz,CDCl3):δ7.75(s,1H),7.48(d,1H),7.37-7.34(m,1H),7.32-7.28(m,2H),7.10-7.07(m,1H),5.10-5.07(m,1H),3.77(s,1H),3.30-3.26(m,2H),2.88-2.85(m,2H),1.38-1.39(m,2H),1.02-1.00(m,2H).
MS ESI:m/z=316.2,[M+H]+
实施例16
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷甲酰胺
Figure PCTCN2017108941-appb-000096
1H NMR(500MHz,CDCl3):δ7.93(s,1H),7.61(d,1H),7.49(d,1H),7.38-7.35(m,2H),7.19(s,1H),6.52(s,1H),4.95-4.89(m,1H),3.14-3.09(m,2H),2.94-2.89(m,2H),1.04-1.02(m,2H),0.83-0.78(m,2H).
MS ESI:m/z=280.1,[M+H]+.
实施例17
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙烷-2-磺酰胺
Figure PCTCN2017108941-appb-000097
1H NMR(500MHz,CDCl3):δ8.03(s,1H),7.64-7.62(m,1H),7.51-7.49(m,1H)),7.39-7.34(m,3H),7.11(s,1H),5.87(s,1H),4.59-4.54(m,1H),3.27-3.22(m,1H),3.09-2.98(m,4H),1.45-1.43(d,6H).
MS ESI:m/z=318.2,[M+H]+
实施例18
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-2-磺酰胺
Figure PCTCN2017108941-appb-000098
1H NMR(500MHz,CDCl3):δ8.52(s,1H),7.94-7.92(m,3H),7.86-7.84(m,2H),7.62-7.52(m,3H),7.41-7.39(m,1H),7.29-7.26(m,1H),7.23-7.20(m,1H),7.16(s,1H),4.46-4.42(m,1H),2.91-2.86(m,2H),2.74-2.69(m,2H).
MS ESI:m/z=402.2,[M+H]+
实施例19
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-1-磺酰胺
Figure PCTCN2017108941-appb-000099
1H NMR(400MHz,CDCl3):δ8.80(d,1H),8.36(d,1H),8.07(d,1H),7.95(d,1H),7.72-7.68(m,2H),7.61-7.55(m,2h),7.40(d,1H),7.35(d,1H),7.27(d,1H),7.16-7.12(m,1H),7.10(s,1H),6.74(s,1H),4.33(d,1H),2.77-2.72(m,2H),2.65-2.59(m,2H).
MS ESI:m/z=480.1,[M+H]+
实施例20
(反式)4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000100
1H NMR(500MHz,CDCl3):δ8.32(s,1H),8.12-8.09(m,2H),7.92-7.90(m,1H),7.91-7.89(m,1H),7.69-7.67(m,1H),4.48(d,1H),7.38-7.34(m,1H),7.36-7.32(m,1H),7.27-7.22(m,1H),7.10(s,1H),5.10-5.03(m,1H),3.34-3.24(m,2H),2.95-2.87(m,2H).
MS ESI:m/z=334.1,[M+H]+
实施例21
(反式)2-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000101
1H NMR(500MHz,CDCl3):δ8.22(s,1H),8.14-8.10(m,1H),7.73-7.71(m,1H),7.55-7.45(m,2H),7.39-7.28(m,4H),7.20-7.14(m,2H),5.10-5.04(m,1H),3.24-3.19(m,2H),3.07-3.01(m,2H).
MS ESI:m/z=334.1,[M+H]+
实施例22
(反式)2,4-二氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000102
1H NMR(500MHz,CDCl3):δ8.16(s,1H),7.70-7.64(m,2H),7.55-7.51(m,1H),7.44-7.36(m,3H),7.34-7.26(m,2H),7.20(s,1H),5.08-5.02(m,1H),3.27-3.21(m,2H),3.05-2.98(m,2H).
MS ESI:m/z=385.1,[M+H]+
实施例23
(反式)4-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰 胺
Figure PCTCN2017108941-appb-000103
1H NMR(500MHz,CDCl3):δ8.13(s,1H),8.08(d,1H),7.88(d,2H),7.72(d,1H),7.48(d,1H),7.37-7.33(m,2H),7.30-7.27(m,1H),6.92(d,2H),5.10-5.04(m,1H),3.83(s,3H),3.26-3.22(m,2H),2.96-2.90(m,2H).
MS ESI:m/z 346.1,[M+H]+
实施例24
(反式)N((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-2-萘甲酰胺
Figure PCTCN2017108941-appb-000104
1H NMR(500MHz,CDCl3):δ8.41(s,1H),8.03(s,1H),7.95-7.91(m,1H),7.87-7.77(m,4H),7.67(d,1H),7.55-7.45(m,3H),7.35-7.31(m,1H),7.26-7.22(m,1H),7.19(s,1H),5.15-5.06(m,1H),3.34-3.23(m,2H),2.95-2.90(m,2H).
MS ESI:m/z=366.1,[M+H]+.
实施例25
(反式)3-氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000105
1H NMR(500MHz,CDCl3):δ8.29(s,1H),8.09(s,1H),8.00(d,1H),7.88(s,1H),7.78-7.76(m,1H),7.72-7.66(m,2H),7.50-7.45(m,2H),7.37-7.31(m,3H),7.24-7.22(m,1H),5.03-4.98(m,1H),3.33-3.27(m,2H),2.97-2.91(m,2H)
MS ESI:m/z=350.1,[M+H]+
实施例26
(反式)4-氰基-N-(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000106
1H NMR(500MHz,CDCl3):δ9.21(s,1H),8.30(s,1H),8.10-8.03(m,4H),8.87(d,1H),7.57(d,1H),7.41-7.40(m,2H),7.15(s,1H),5.19-5.14(m,1H),3.11-3.08(m,2H),2.90-2.85(m,2H).
MS ESI:m/z=341.1,[M+H]+
实施例27
(反式)3-溴-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000107
1H NMR(500MHz,CDCl3):δ8.26(s,1H),8.21(s,1H),8.04(d,1H),7.84-7.70(m,1H),7.67(d,1H),7.62-7.60(m,1H),7.49(d,1H),7.38-7.35(m,1H),7.32-7.27(m,2H),7.21(s,1H),5.09-5.03(m,1H),3.32-3.26(m,2H),2.97-2.91(m,2H).
MS ESI:m/z=395.1[M+H]+
实施例28
(反式)2-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000108
1H NMR(500MHz,CDCl3):δ8.35(s,1H),8.24(d,1H),8.03(s,1H),7.70(d,1H),7.52-7.48(m,2H),7.40-7.32(m,2H),7.21(s,1H),7.15-7.13(m,1H),7.11-7.05(m,1H),5.04-4.99(m,1H),3.17-3.11(m,2H),3.07-3.01(m,2H).
MS ESI:m/z=346.1,[M+H]+
实施例29
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-3-(三氟甲基)苯甲酰胺
Figure PCTCN2017108941-appb-000109
1H NMR(500MHz,CDCl3):δ8.38(s,1H),8.29(d,1H),8.18(s,1H),8.11(d,1H),1.96(d,1H),7.77-7.73(m,2H),7.57-7.49(m,2H),7.39-7.29(m,2H),5.14-5.08(m,1H),3.39-3.33(m,2H),2.99-2.93(m,2H).
MS ESI:m/z=384.1,[M+H]+.
实施例30
(反式)苄基(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸叔丁酯
Figure PCTCN2017108941-appb-000110
1H NMR(500MHz,CDCl3):δ7.90(s,1H),7.58(s,1H),7.49(d,1H),7.39-7.31(m,7H),7.17(s,1H),5.51(s,1H),5.17-5.10(m,2H),4.69-4.67(m,1H),3.06-3.02(m,2H),2.93-2.88(m,2H).
MS ESI:m/z=346.1,[M+H]+
实施例31
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
Figure PCTCN2017108941-appb-000111
1H NMR(500MHz,CDCl3):δ7.96(s,1H),7.60(d,1H),7.49(d,1H),7.36-7.29(m,2H),7.18(s,1H),6.77(s,1H),4.95-4.89(m,1H),3.10-3.05(m,2H),2.93-2.89(m,2H),2.32-2.28(m,2H),1.26-1.20(m,3H).
MS ESI:m/z=268.1,[M+H]+
实施例32
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)异丁酰胺
Figure PCTCN2017108941-appb-000112
1H NMR(400MHz,CDCl3):δ7.97(s,1H),7.61(d,1H),7.49(d,1H),7.38-7.29(m,2H),7.19(s,1H),6.51(d,1H),4.93-4.88(m,1H),3.10-3.04(m,2H),2.93-2.88(m,2H),2.47-2.43(m,1H),1.23-1.21(m,6H).
MS ESI:m/z=282.1,[M+H]+
实施例33
(反式)((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸苯酯
Figure PCTCN2017108941-appb-000113
1H NMR(400MHz,CDCl3):δ8.50(d,1H),8.28(s,1H),7.87(d,1H),7.56(s,1H),7.55-7.43(m,3H),7.38-8.13(m,5H),4.82-4.78(m,1H),3.07-3.01(m,2H),2.86-2.81(m,2H);
MS ESI:m/z=332.1,[M+H]+
实施例34
(顺式)N((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙烯酰胺
Figure PCTCN2017108941-appb-000114
1H NMR(500MHz,CDCl3):δ8.05(s,1H),7.65(d,1H),7.49(d,1H),7.32-7.36(m,2H),6.34-6.39(d,1H),6.12-6.19(m,2H),5.71(d,1H),4.92(s,2H);
MS ESI:m/z=266.1,[M+H]+
实施例35
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000115
1H NMR(500MHz,CDCl3):δ7.85(d,2H),7.66(d,1H),7.44-7.55(m,5H),7.30-7.40(m,3H),6.85(s,1H),5.00-5.85(m,1H),3.15-3.23(m,2H),2.93-3.02(m,2H);
MS ESI:m/z=316.0,[M+H]+
实施例36
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)甲基磺酰胺
Figure PCTCN2017108941-appb-000116
1H NMR(500MHz,CDCl3):δ7.91(s,1H),7.53(d,1H),7.46(d,1H),7.35(t,1H),7.30(t,1H),7.18(s,1H),6.21(s,1H),4.55(s,1H),3.06(s,3H),2.97-3.01(m,4H).
MS ESI:m/z=290.1,[M+H]+
实施例37
(顺/反)3-甲氧基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000117
(顺:反=2:1)
MS ESI:m/z=347.1,[M+H]+
实施例38
(反式)3,4-二氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000118
1H NMR(500MHz,丙酮-d6):δ8.39(s,1H),8.09(s,1H),7.81-7.91(m,2H),7.59-7.70(m,2H),7.36-7.40(m,2H),7,21(s,1H),5.17-5.21(m,1H),3.19-3.25(m,2H),2.95-3.01(m,2H);
MS ESI:m/z=386.1,[M+H]+
实施例39
(顺/反)2-甲基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺的制备
Figure PCTCN2017108941-appb-000119
(顺:反=2:1)
MS ESI:m/z=330.1,[M+H]+
实施例40
(反式)3-甲基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺的制备
Figure PCTCN2017108941-appb-000120
1H NMR(500MHz,丙酮-d6):δ8.23(s,1H),7.85(d,1H),7.76-7.77(m,2H),7.57-7.59(d,1H),7.32-7.39(m,4H),7.15(s,1H),5.18-5.24(m,1H),3.24-3.29(t,2H)2.92-2.98(t,3H),2.38(s,3H).
MS ESI:m/z=330.1,[M+H]+
实施例41
(反式)2,4,6-三甲基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000121
1H NMR(500MHz,丙酮-d6):δ8.49(s,1H),7.89-7.90(d,1H),7.70-7.72(d,1H),7.30-7.58(m,6H),4.98-5.00(m,1H),3.39-3.44(m,2H)3.08-3.12(m,2H);2.60-2.70(m,9H).
MS ESI:m/z=358.2,[M+H]+
实施例42
(反式)2-氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000122
1H NMR(500MHz,丙酮-d6):δ8.34(s,1H),7.79-7.81(m,1H),7.53-7.57(m,2H),7.32-7.48(m,5H),7.16(s,1H),5.17-5.21(m,1H),3.16-3.22(m,2H)2.98-3.04(m,2H);
MS ESI:m/z=351.1,[M+H]+
实施例43
(反式)3-氟-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000123
1H NMR(500MHz,丙酮-d6):δ8.42(s,1H),8.40(s,1H),7.79-7.88(m,2H),7.68-7.71(d,1H),7.51-7.58(m,2H),7.25-7.40(m,2H),7.14(s,1H),5.18-5.24(m,1H),3.23-3.29(t,2H);2.86-2.91(m,2H);
MS ESI:m/z=334.1,[M+H]+
实施例44
(反式)3-三氟甲氧基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000124
1H NMR(500MHz,丙酮-d6):δ8.52(s,1H),8.24(s,1H),8.09(d,2H),7.86(d,1H),7.57(d,1H),7.25-7.45(m,3H),7.15(s,1H),5.20-5.26(m,1H),3.25-3.30(m,2H);2.86-2.91(m,2H);
MS ESI:m/z=400.1,[M+H]+
实施例45
(反式)4-溴-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000125
1H NMR(500MHz,丙酮-d6):δ8.32(s,1H),7.81-7.87(m,3H),7.66(d,2H),7.58(d,1H),7.34-7.41(m,2H),7.17(s,1H),5.17-5.22(m,1H),3.19-3.24(m,2H),2.94-3.00(m,2H);
MS ESI:m/z=396.0,[M+H]+
实施例46
(反式)4-氟-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000126
1H NMR(500MHz,丙酮-d6):δ8.12(s,1H),8.00-8.04(m,2H),7.72(d,1H),7.51(d,2H),7.28-7.37(m,4H),7.08(s,1H),4.57-4.64(m,1H),2.90-2.91(m,2H),2.74-2.79(m,2H);
MS ESI:m/z=370.1,[M+H]+
实施例47
(反式)4-氧代-4-苯基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丁酰胺
Figure PCTCN2017108941-appb-000127
(反式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-胺(21mg,0.1mmol)和4-氧代-4-苯基丁酸(18mg,0.1mmol)加入反应瓶中,加入二氯甲烷(2mL)水浴搅拌。加入三乙胺(51mg,0.5mmol)和HOBT(14mg,0.1mmol),以及EDCI(38mg,0.2mmol),搅拌中自然回到室温。TLC监测反应,完毕后,加入饱和碳酸氢钠水溶液(5mL)用二氯甲烷10mL进行萃取,重复2次,合并有机相,用无水硫酸钠固体干燥。过滤,浓缩,得到粗品。柱层析分离纯化(石油醚/乙酸乙酯),得呈固体的目标化合物(20mg,54%收率)。
1H NMR(500MHz,CDCl3):δ8.01(d,2H),7.93(s,1H),7.61-7.57(m,2H),7.49-7.46(m,2H),7.37-7.27(m,3H).7.17(s,1H),6.93(d,1H),4.92-4.86(m,1H),3.45-3.42(m,2H),3.04-2.98(m,2H),2.94-2.88(m,2H),2.73-2.70(m,2H).
MS ESI:m/z=372.2,[M+H]+
实施例48
(反式)2-([1,1’-联苯]-4-基)-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
Figure PCTCN2017108941-appb-000128
1H NMR(500MHz,CDCl3):δ7.86(s,1H),7.66-7.61(m,4H),7.52-7.45(m,3H),7.41-7.38(m,2H),7.36-7.32(m,1H),7.28-7.24(m,3H),7.16(s,1H),4.83-4.77(m,1H),3.70(s,2H),3.00-2.98(m,2H),2.91-2.89(m,2H).
MS ESI:m/z=406.2,[M+H]+
实施例49
(反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环戊烷甲酰胺
Figure PCTCN2017108941-appb-000129
1H NMR(500MHz,CDCl3):δ7.98(s,1H),7.61(d,1H),7.49(d,1H),7.38-7.34(m,2H),7.19(s,1H),4.93-4.86(m,1H),3.09-3.03(m,2H),2.93-2.88(m,2H),2.61-2.58(m,1H),1.92-1.76(m,6H),1.62-1.59(m,2H).
MS ESI:m/z=308.1,[M+H]+
实施例50
(反式)3-环戊基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
Figure PCTCN2017108941-appb-000130
1H NMR(500MHz,CDCl3):δ7.96(s,1H),7.62(d,1H),7.50(d,1H),7.37-7.34(m,2H),7.18(s,1H),6.02(d,1H),4.89-4.83(m,1H),3.08-3.03(m,2H),2.95-2.89(m,2H),2.29-2.25(m,2H),1.80-1.53(m,9H),1.15-1.13(m,3H).
MS ESI:m/z=336.2,[M+H]+
实施例51
(反式)2-环丙基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
Figure PCTCN2017108941-appb-000131
1H NMR(500MHz,CDCl3):δ8.03(s,1H),7.65(d,1H),7.50(d,1H),7.37-7.32(m,2H),7.19(s,1H),6.64(d,1H),4.93-4.87(m,1H),3.14-3.08(m,2H),2.94-2.90(m,2H),2.24-2.23(m,2H),1.07-1.03(m,1H),0.68-0.66(m,2H),0.27-0.26(m,2H).
MS ESI:m/z=294.1,[M+H]+
实施例52
(反式)叔丁基-2-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)吡咯烷-1-甲酸叔丁酯
Figure PCTCN2017108941-appb-000132
1H NMR(500MHz,CDCl3):δ7.96(s,1H),7.94(s,1H),7.62(d,1H),7.49(d,1H),7.39-7.34(m,2H),7.18(s,1H),4.90-4.87(m,1H),4.35-4.34(m,1H),3.65-3.28(m,2H),3.05-2.93(m,2H),2.21(s,4H),1.93(s,2H),1.51(s,9H).
MS ESI:m/z=409.2,[M+H]+
实施例53
(反式)4-羟基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000133
1H NMR(500MHz,丙酮-d6):δ8.49(s,1H),7.86-7.92(m,3H),7.72(d,1H),7.50(d,1H),7.42(d,1H),7.25-7.26(m,2H),6.92-6.93(m,1H),5.63(s,1H),5.20-5.25(m,1H),3.26-3.31(m,2H),2.98-3.03(m,2H);
MS ESI:m/z=332.1,[M+H]+
实施例54
(顺/反)N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-[1,1’-二苯基]-4-甲酰胺
Figure PCTCN2017108941-appb-000134
(顺/反=9:1)
MS ESI:m/z=392.2,[M+H]+
实施例55
(顺式)2-甲基-2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
Figure PCTCN2017108941-appb-000135
1H NMR(400MHz,CDCl3):δ7.73(s,1H),7.48(d,1H),7.39-7.42(m,2H),7.34-7.36(m,3H),7.20-7.31(m,3H),7.01(s,1H),5.80(d,1H),4.53-4.59(m,1H),2.93-2.99(m,2H),2.61-2.66(m,2H);1.55(s,6H);
MS ESI:m/z=358.1[M+H]+
实施例56
(顺式)2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
Figure PCTCN2017108941-appb-000136
1H NMR(400MHz,CDCL3-d6):δ7.96(s,1H),7.46-7.52(m,2H),7.36-7.40(m,3H),7.23-7.36(m,4H),7.01(s,1H),6.67(d,1H),4.64-6.70(m,1H),3.61(q,1H),2.93-3.06(m,2H),2.67-2.80(m,2H);1.54-1.57(m,3H);
MS ESI:m/z=344.1,[M+H]+
实施例57
(反式)2-溴-5-甲氧基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000137
1H NMR(500MHz,丙酮-d6):δ8.22(s,1H),8.13(s,1H),7.83(d,1H),7.51-7,57(m,2H),7.30-7.39(m,2H),7.13(s,1H),7.07(d,2H),6.94-6.98(dd,1H),5.14-5.20(m,1H),3.83(s,3H),3.21-3.27(m,2H),2.96-3.01(m,2H);
MS ESI:m/z=424.1,[M+H]+
实施例58
(反式)2-溴-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)烟酰胺
Figure PCTCN2017108941-appb-000138
1H NMR(500MHz,丙酮-d6):δ8.40-8.41(m,1H),7.88(d,1H),7.74(d,1H),7.56(d,1H),7.48-7.51(m,2H),7.33-7.39(m,2H),7.18(s,1H),5.10-5.1(m,1H),3.12-3,17(m,2H),2.98-3.03(m,2H);
MS ESI:m/z=397.0,[M+H]+
实施例59
(顺/反)3-氟-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000139
(顺:反=4:1)
MS ESI:m/z=335.0,[M+H]+
实施例60
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)四氢呋喃-3-甲酰胺
Figure PCTCN2017108941-appb-000140
1H NMR(500MHz,丙酮-d6):δ8.28(s,1H),7.88(t,1H),7.57(d,1H),7.33-7.40(d,2H),7.16(s,1H),5.01-5.04(m,1H),4.28(t,1H),3.91-3.96(m, 1H),3.79-3.85(m,1H),3.10-3.15(m,2H),2.82-2.87(m,2H),2.18-2.23(m,1H),1.97-2.00(m,1H),1.85-1.90(m,2H);
MS ESI:m/z=310.1,[M+H]+
实施例61
(反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)呋喃-3-甲酰胺
Figure PCTCN2017108941-appb-000141
1H NMR(500MHz,丙酮-d6):δ8.29(s,1H),8.14(s,1H),7.77(d,1H),7.56-7.62(m,2H),7.35-7.38(m,2H),7.16(s,1H),6.86(s,1H),5.11-5.15(m,1H),3.14-3,20(m,2H),2.90-2.95(m,2H);
MS ESI:m/z=307.1[M+H]+
实施例62
(反式)(1r,3r)-N-(萘乙酰胺-2-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000142
1H NMR(400MHz,CDCl3):δ8.23(s,1H),8.20(s,1H),7.79-7.89(m,4H),7.36-7.54(m,6H),9.31-7.33(m,2H),7.17(s,1H),3.46-3.50(m,1H),3.25-3.31(m,2H),3.01-3.06(m,2H).
MS ESI:m/z=366.1,[M+H]+
实施例63
3-(苄氧基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000143
(順/反)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇(22mg,0.1mmol)溶于THF中,冰浴下,加入氢化钠(4mg,60%,0.1mmol),搅拌15分钟后,加入溴苄(20mg,1.2mmol),搅拌直到原料消失。反应液加入水5mL,乙酸乙酯10mL萃取,有机相用硫酸钠干燥,过滤,浓缩,柱层析得到目标化合物(25mg,收率80%,顺/反=2:1)
MS ESI:m/z=303.1,[M+H]+
实施例64
(顺/反)3-丁氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000144
(顺/反=2:1)
MS ESI:m/z=269.1,[M+H]+
实施例65
(顺/反)3-乙氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000145
(顺/反=2:1)
MS ESI:m/z=240.1,[M+H]+
实施例66
(顺/反)3-甲氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000146
(顺/反=2:1)
MS ESI:m/z=227.1,[M+H]+
实施例67
(反式)1-苯基-2-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基)乙醇
Figure PCTCN2017108941-appb-000147
将实施例5中的第一步制备的中间体(21mg,0.1mmol)与2-溴-1-苯基乙酮(30mg,0.2mmol),三乙胺(20mg,0.2mmol)加入到反应瓶中,加入二氯甲烷(3mL),反应加热到40度,反应2小时。TLC显示反应结束。反应液中加入二氯甲烷(10mL)和水(10mL),萃取,收集有机相,硫酸钠干燥。过滤,浓缩,柱层析分离纯化,得到20mg,收率60.0%。
1H NMR(500MHz,CDCl3):δ7.94(s,1H),7.51-7.46(m,2H),7.41-7.38(m,2H),7.35-7.32(m,4H),7.27-7.25(m,2H),4.81-4.76(m,1H),3.65(s,2H),3.03-3.00(m,2H),2.87-2.83(m,2H).
MS ESI:m/z=330.2,[M+H]+
实施例68
(反式)(1r,3r)-N-(3-溴-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000148
(反式)(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]是吲哚]-3-羧酸(24mg,0.1mmol),4-氟-3溴-苯胺(19mg,0.1mmol)加入反应瓶中,加入3ml二氯甲烷,加入HOBT(14mg,0.1mmol)和三乙胺(50mg,0.5mmol),冷却到冰浴,加入EDCI(39mg,2mmol),自然回到室温,反应过夜。反应液浓缩后,直接柱层析分离纯化得到呈浅黄色固体的目标化合物(5mg,收率15%)。
H NMR(500MHz,CDCl3):δ8.15(s,1H),7.94-7.93(m,1H),7.56-7.52(m,2H),7.50-7.46(m,1H),7.40-7.37(m,1H),7.33-7.29(m,1H),7.13-7.07(m,1h),3.40-3.37(m,1H),3.25-3.20(m,2H),3.04-2.99(m,2H).
MS ESI:m/z=413.1,[M+H]+.
实施例69
(反式)(1r,3r)-N-(对甲苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000149
1H NMR(500MHz,CDCl3):δ8.13(s,1H),7.92-7.90(m,1H),7.54-7.50(m,2H),7.48-7.46(m,1H),7.40-7.37(m,1H),7.33-7.29(m,2H),7.13-7.07(m,1H),3.40-3.37(m,1H),3.25-3.20(m,2H),3.04-2.99(m,2H),2.48(s,3H).
MS ESI:m/z=330.2,[M+H]+
实施例70
(反式)(1r,3r)-N-(3-氯-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰
Figure PCTCN2017108941-appb-000150
1H NMR(400MHz,CDCl3):δ8.25(s,1H),7.88(s,1H),7.70-7.68(m,1H),7.62-7.51(m,2H),7.48-7.45(m,1H),7.43-7.38(m,1H),7.31-7.29(m,1H),7.20(s,1H),7.15-7.12(m,1H),5.4-5.35(m,1H),3.50-3.45(m,1H),3.28-3.23(m,2H),3.10-3.02(m,2H).
MS ESI:m/z=368.1,[M+H]+
实施例71
(反式)(1r,3r)-N-(3-氟苯基)螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000151
1H NMR(400MHz,CDCl3):δ9.12(s,1H),8.63(s,1H),7.80-7.78(m,1H),7.73-7.68(m,1H),7.62-7.49(m,2H),7.45-7.30(m,6H),6.79-7.78(m,1H),4.18-4.15(m,1H),3.5-3.40(m,2H),2.80-7.70(m,2H).
MS ESI:m/z=334.1,[M+H]+
实施例72
(反式)(1r,3r)-N-(3-氯苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000152
1H NMR(400MHz,CDCl3):δ8.48(s,1H),8.20(s,1H),7.79(s,1H),7.54-7.49(m,3H),7.46-7.38(m,2H),7.27-7.24(m,1H),7.18-7.18(s,1H),7.16(d,1H),3.51-3.46(m,1H),3.26-2.21(m,2H),3.05-2.99(m,2H).
MS ESI:m/z=350.1[M+H]+.
实施例73
(反式)(1r,3r)--N-(3-溴苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000153
1H NMR(400MHz,CDCl3):δ8.23(s,2H),7.91(s,1H),7.55-7.51(m,3H),7.40-7.36(m,1H),7.31-7.26(m,1H),7.22-7.18(m,1H),3.49-3.45(m,1H),3.24-3.18(m,2H),3.03-2.97(m,2H).
MS ESI:m/z=395.1[M+H]+
实施例74
(顺式)(1r,3S,8a'S)-N-(3-溴苯基)-8a'H-螺[环丁基-1,8'-茚并[1,2-c]吡咯]-3-甲酰胺
Figure PCTCN2017108941-appb-000154
1H NMR(400MHz,CDCl3):δ8.80(s,1H),8.27(s,1H),8.03(s,1H),7.80-7.78(m,1H),7.65-7.51(m,2H),7.43-7.41(m,2H),7.25-7.20(m,2H),3.88-3.86(m,1H),3.46-3.38(m,2H),2.33-2.25(m,2H).
MS ESI:m/z=395.1[M+H]+
实施例75
N-(4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000155
(顺/反2:1.7).
MS ESI:m/z=334.1,[M+H]+
实施例76
(反式)(1r,3r)-N-(萘-2-基磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000156
(反式)螺[环丁烷1,5’-咪唑并[5,1-a]是吲哚]-3-羧酸(24mg,0.1mmol),加入反应瓶中,加入HATU(76mg,0.20mmol),加入二氯甲烷(3mL),搅拌5分钟,加入DIPEA(64mg,0.5mmol,搅拌一小时,加入2-萘磺酰胺(47mg,0.3mmol),和DMAP(48mg,0.40mmol),反应过夜,原料消失,反应液中加入二氯甲烷(10mL),加入水(10mL),萃取,水相再用二氯甲烷(10mL)萃取,合并有机相,硫酸钠干燥。过滤,浓缩,拌样,柱层析得到呈浅黄色固体的目标化合物(10mg,)。
1H NMR(400MHz,MeOH-d4):δ8.53(s,1H),8.46(s,1H),8.00-7.94(m,2H),7.92-7.91(m,2H),7.87(d,1H),7.67-7.65(m,1H),7.58-7.1(m,3H),7.35-7.33(m,2H),7.18(s,1H),3.37-3.33(m,1H),2.91-2.78(m,4H).
MS ESI:m/z=430.1,[M+H]+
实施例77
(反式)(1r,3r)-N-((4-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000157
1H NMR(400MHz,CDCl3):δ8.21-8.20(m,2H),8.19-8.17(m,1H),7.74-7.73(m,1H),7.58-7.56(m,1H),7.49-7.30(m,4H),7.12(s,1H),3.68-3.67(m,1H),3.00-2.93(m,2H),2.90-2.83(m,2H).
MS ESI:m/z=398.1,[M+H]+
实施例78
(反式)(1r,3r)-N-((3-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000158
1H NMR(400MHz,CDCl3):δ7.85-7.83(d,1H),7.75-7.73(d,1H),7.63-7.57(m,3H),7.48-7.39(m,4H),7.18-7.13(m,1H),3.66-3.64(m,1H),3.50-3.48(m,1H),3.21-3.18(m,3H).
MS ESI:m/z=398.1,[M+H]+
实施例79
(反式)(1r,3r)-N-((3-氯苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
Figure PCTCN2017108941-appb-000159
1H NMR(400MHz,CDCl3):δ8.02(s,1H),7.88-7.85(d,1H),7.69-7.64(m,3H),7.60-7.48(m,5H),3.51-3.48(m,1H),3.45-3.40(m,4H).
MS ESI:m/z=414.5,[M+H]+
实施例80
(反式)(1r,3r)-3-(甲硫基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000160
(顺式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基甲磺酸酯(290mg,1.0mmol)溶于5mL DMF中,加入甲硫醇钠(105mg,1.5mmol),室温搅拌过夜。反应完毕后。加入乙酸乙酯20mL,加入水10mL,萃取,再用10mL的水洗,有机相干燥,过滤,浓缩。柱层析,得到目标化合物(240mg,收率99%)。
1H NMR(500MHz,CDCl3):δ7.88(s,1H),7.74(d,1H),7.48(d,1H),7.41-7.30(m,2H),7.16(s,1H),3.93-3.84(m,1H),3.09-2.97(m,2H),2.89-2.84(m,2H),2.20(s,3H).
MS ESI:m/z=243.1,[M+H]+
实施例81
(反式)(1r,3r)-3-(甲基磺酰基)螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000161
实施例80(48mg,0.2mmol)加入5mL二氯甲烷中,冰浴下加入MMPP(98mg,0.2mmol),此温度下反应,直到原料消失,加入硫代硫酸钠溶液淬灭反应,二氯甲烷萃取(3x10mL),合并有机相,干燥,过滤,浓缩,柱层析。得到目标化合物(20mg,收率37%)。
1H NMR(500MHz,CDCl3):δ7.91(s,1H),7.84(d,1H),7.49(d,1H),7.41-38(m,2H),7.18(s,1H),4.33-4.29(m,1H),3.55-3.49(m,2H),2.97-2.95(m,2H).
MS ESI:m/z=275.1,[M+H]+
实施例82
(反式)(1R,3r)-3-((R)-甲基亚磺酰基)螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000162
实施例80(48mg,0.2mmol)加入5mL二氯甲烷中,冰浴下加入MMPP(74mg,0.2mmol),此温度下反应,直到原料消失,加入硫代硫酸钠溶液淬灭反应,二氯甲烷萃取(3x10mL),合并有机相,干燥,过滤,浓缩,柱层析得到目标化合物(20mg,收率39%)。
1H NMR(500MHz,CDCl3):δ7.94(s,1H),7.81(d,1H),7.48(d,1H),7.38-7.32(m,2H),7.17(s,1H),3.85-3.80(m,1H),3.78-3.70(m,1H),3.39-3.34(m,1H),2.52(s,3H).
MS ESI:m/z=259.1,[M+H]+
实施例83
(顺/反)3-(1-苄基-1H-1,2,3-三唑-4-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000163
第一步:(顺/反)3-乙炔基螺[环丁烷-1,5'-咪唑并[5,1-a]isoindol]-3-醇
Figure PCTCN2017108941-appb-000164
在反应瓶中加入5mL无水四氢呋喃,加入乙炔基溴化镁(1.7mL,0.8mmol),冰浴搅拌,把螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-酮(20mg,0.08mmol)溶于四氢呋喃(1mL)滴入乙炔基溴化镁中,滴毕,自然回到室温反应1小时,TLC显示原料消失。滴加饱和氯化铵淬灭反应。加入乙酸乙酯20mL和水10mL,萃取,有机相干燥,过滤,浓缩,得到粗品,柱层析纯化分离,得到目标化合物(20mg,收率100%)。
MS ESI:m/z=237.1,[M+H]+.
第二步:3-(1-苄基-1H-1,2,3-三唑-4-基)螺[环己烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000165
将第一步产物(20mg,0.076mmol)、苄基叠氮(20mg,0.2mmol)、五水硫酸铜(14mg,0.08mmol)和抗坏血酸钠(15mg,0.08mmol)加入二氯甲烷,水,甲醇(2mL,1mL,1mL),室温搅拌24小时,TLC显示原料消失,反应液中加入二氯甲烷(10ml),用硅藻土过滤,滤液浓缩,拌样,柱层析,得到目标化合物10mg,收率36%。
MS ESI:m/z=370.1[M+H]+
实施例84
(反式)(1r,3r)-3-(4-环己基-1氢-1,2,3三唑1-基)螺[c环丁烷-1,5’-咪唑[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000166
1H NMR(500MHz,丙酮-d6):δ8.63(s,1H),8.53(s,1H),7.98(d,2H),7.89(d,1H),7.72(s,1H),7.35-7.49(m,6H),6.20(s,1H),3.76-3.80(m,2H),3.48(s,2H).
MS ESI:m/z=346.1,[M+H]+
实施例85
(反式)(1r,3r)-3-(4-苯基-1H-1,2,3三唑-1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000167
1H NMR(500MHz,CDCl3):δ8.00(s,1H),7.82(d,1H),7.50(d,1H),7.35-7.39(m,3H),5.46-5.48(m,1H),3.71-3.75(m,2H),3.14-3.22(m,2H);2.80-2.81(m,1H),2.08-2.09(m,2H),1.75-1.83(m,2H),1.43-1.44(m,2H),1.22-1.23(m,2H).
MS ESI:m/z=340.0,[M+H]+
实施例86
(反式)(1r,3r)-3-(4-环丙基1H-1,2,3三唑1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000168
1H NMR(300MHz,CDCl3):δ7.77(s,1H),7.64(s,1H),7.33-7.41(m,3H),7.0(s,1H),5.54(s,1H),3.89-3.95(m,2H),3.70(s,1H),3.33-3.42(m,2H),1.96-1.99(m,1H),0.96-1.00(m,2H),0.84-0.90(m,2H);
MS ESI:m/z=304.1,[M+H]+
实施例87
(反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000169
将(順式)螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-胺(40mg,0.2mol),三乙胺(27uL,0.2mmol)溶于5mL DMF,加入羰基二咪唑(31mg,0.2mmol)室温搅拌1.5小时,加入苯胺(20mg,0.2mmol)搅拌过夜.加入饱和碳酸氢溶液20mL,二氯甲烷萃取(10mL*3),和并有机层,盐水水洗,无水硫酸钠干燥,过滤。溶剂浓缩,柱色谱层析(DCM:MeOH=100:5)得到目标化合物为呈黄色固体的(60mg,96%).
1H NMR(500MHz,丙酮-d6):δ8.11(s,1H),7.80(d,1H),7.63(s,2H),7.33-7.37(m,1H),7.21-7.25(m,1H),7.11(s,1H),6.93(s,1H),6.50(d,1H),4.87-4.93(m,1H),3.09-3.15(m,2H),2.86-2.91(m,2H);
MS ESI:m/z=331.1,[M+H]+
实施例88
(反式)3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)磺酰脲
Figure PCTCN2017108941-appb-000170
冰浴下将氯磺酰异氰酸酯(37mg,0.2mmol)溶于1mL of二氯甲烷,加入25uL叔丁醇,室温下搅拌十分钟,加入化合物(反式)螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-胺(25mg,0.2mmol)和三乙胺(68uL),反应继续搅拌十分钟,然后用水20mL淬灭,二氯甲烷萃取(10mL*3),合并有机层,饱和氯化钠洗,无水硫酸钠干燥,过滤,柱层析(二氯甲烷:甲醇=20:1),得到黄色固体(45mg,50%).将得到的化合物溶于1mL二氯甲烷和1mL三氟乙酸,室温搅拌2小时,旋干溶剂,柱层析(二氯甲烷:甲醇=20:1),,得到目标化合物为黄色固体(14mg,52%).
1H NMR(500MHz,甲醇-d4):δ8.22(s,1H),7.73-7.75(m,1H),7.74-7.56(m,1H),7.38-7,41(m,2H),7.15(s,1H),4.53-4,58(m,1H),2.88-3.01(m,4H);
MS ESI:m/z=291.1[M+H]+
实施例89
(反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000171
冰浴下,将(順式)螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-胺(30mg,0.1mmol)和三乙胺(40uL,0.3mmol)溶于5mL二氯甲烷,加入苯基异硫氰酸酯(17uL,0.1mmol)。室温搅拌十分钟,然后水淬灭反应(20mL),二氯甲烷萃取(10mL*3),合并有机层,饱和氯化钠洗,无水硫酸钠干燥,过滤,浓缩,柱色谱层析(二氯甲烷:甲醇=20:1),干燥,得到目标化合物为呈黄色固体(40mg,81%).
1H NMR(500MHz,CDCl3):δ8.10(s,1H),7.86(s,1H),7.31-7.40(m,3H),7.14-7.27(m,2H),7.02-7.06(m,2H),6.96(d,1H),6.82(s,1H),5.30-5.32(m,1H),2.92-2.95(m,2H),2.79-2.81(m,2H);
MS ESI:m/z=347.1,[M+H]+
实施例90
(顺式)1-(3-氟苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
Figure PCTCN2017108941-appb-000172
1H NMR(400MHz,CDCl3):δ8.05(s,1H),7.83(s,1H),7.55(d,1H),7.44(d,1H),7.27-7.37(m,3H),7.04-7.11(m,2H),6.95-6.98(m,1H),5.13-5.15(m,1H),3.07-3.24(m,2H),2.63-2.67(m,2H).
MS ESI:m/z=365.1,[M+H]+
实施例91
(顺式)1-(4-氯苯基)-3-(1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
Figure PCTCN2017108941-appb-000173
1H NMR(400MHz,CDCl3):δ7.82-7.86(m,2H),7.62(d,1H),7.46-7.52(m,3H),7.35-7.39(m,2H),7.15(s,1H),6.42(s,1H),5.19-5.21(m,1H),3.24-3.27(m,2H),2.68-2.71(m,2H).
MS ESI:m/z=381.0,[M+H]+
实施例92
(顺式)1-(4-三氟甲基苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
Figure PCTCN2017108941-appb-000174
1H NMR(400MHz,CDCl3):δ8.35(s,1H),7.86(s,1H),7.49-7.60(m,6H),7.29-7.33(m,2H),7.09(s,1H),6.89(s,1H),5.10(s,1H),3.23-3.25(m,2H),2.64-2.66(m,2H);
MS ESI:m/z=415.1,[M+H]+
实施例93
(顺式)1-((1s,3s)-螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]-3-基)-3-(4-(三氟甲基)苯基)硫脲
Figure PCTCN2017108941-appb-000175
1H NMR(400MHz,CDCl3):δ9.14(s,1H),8.00(s,1H),7.66(s,1H),7.44(s,2H),7.31(s,2H),7.19(s,2H),7.09(s,1H),5.09(s,1H),3.23-3.25(m,2H),2.64-2.66(m,2H);
MS ESI:m/z=415.1,[M+H]+
实施例94
(顺式)1-(3-氯-4-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000176
1H NMR(400MHz,甲醇-d4):δ8.18(s,1H),7.99(s,1H),7.74-7.76(m,1H),7.58-7.64(m,2H),7.48-7.50(m,1H),7.40-7.42(s,2H),7.13(s,1H),4.66-4.70(m,1H),3.09-3.12(m,2H),2.97-3.02(m,2H);
MS ESI:m/z=432.9,[M+H]+
实施例95
(顺式)1-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-3-(3-(三氟甲基)苯基)脲
Figure PCTCN2017108941-appb-000177
1H NMR(400MHz,甲醇-d4):δ8.18(s,1H),7.89(s,1H),7.75-7.77(m,1H),7.57-7.61(m,2H),7.40-7.48(m,2H),7.27-7.29(m,1H),7.14(s,1H),4.65-4.71(m,1H),3.10-3.13(m,2H),2.97-3.02(m,2H);
MS ESI:m/z=398.9,[M+H]+
实施例96
(顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000178
1H NMR(400MHz,甲醇-d4):δ8.22(s,1H),8.09(s,1H),7.73-7.75(m,1H),7.67(s,1H),7.58-7.60(m,1H),7.40-7.44(m,2H),7.13(s,1H),5.16-5.20(m,1H),3.17-3.22(m,2H),2.89-2.94(m,2H);
MS ESI:m/z=483.1,[M+H]+
实施例97
(顺式)1-(4-氯-3-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000179
1H NMR(400MHz,甲醇-d4):δ8.01(s,1H),7.93(s,1H),7.60-7.65(m,2H),7.46-7.50(m,2H),7.31-7.34(m,2H),7.03(s,1H),5.12(s,1H),3..04-3.09(m,2H),2.79-2.85(m,2H);
MS ESI:m/z=449.0,[M+H]+
实施例98
(顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000180
1H NMR(400MHz,甲醇-d4):δ8.08(s,1H),7.98(s,1H),7.64-7.66(m,1H),7.47-7.50(m,1H),7.42(s,1H),7.30-7.42(m,2H),7.03(s,1H),4.57-4.61(m,1H),2.97-3.03(m,2H),2.88-2.94(m,2H);
MS ESI:m/z=467.1,[M+H]+
实施例99
(顺式)1-(3,4-二氯苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000181
1H NMR(400MHz,甲醇-d4):δ8.06(s,1H),7.62-7.67(m,1H),7.62-7.65(m,1H),7.47-7.49(m,1H),7.27-7.33(m,3H),7.17-7.20(dd,2H),7.02(s,1H),4.52-4.60(m,1H),2.95-3.00(m,2H),2.85-2.90(m,2H);
MS ESI:m/z=401.0[M+H]+
实施例100
(顺式)1-苄基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000182
1H NMR(400MHz,甲醇-d4):δ8.01(s,1H),7.60-7.62(m,1H),7.46-7.48(m,1H),7.27-7.30(m,2H),7.19-7.22(m,4H),7.01(s,1H),2.92-2.97(m,2H),2.75-2.80(m,2H);
MS ESI:m/z=345.2[M+H]+
实施例101
(顺式)1-苯乙基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000183
1H NMR(400MHz,甲醇-d4):δ7.99(s,1H),7.59-7.61(m,1H),7.46-7.48(m,1H),7.25-7.31(m,2H),7.07-7.20(m,7H),7.01(s,1H),4.43-4.52(m,1H),3.29-3.31(t,2H),2.89-2.94(m,2H),2.69-2.75(m,4H);
MS ESI:m/z=359.2[M+H]+
实施例102
(反式)1-(3-三氟甲基苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000184
1H NMR(400MHz,甲醇-d4):δ8.08(s,1H),7.79(s,1H),7.57-7.62(m,2H),7.41-7.45(m,2H),7.26-7.36(m,3H),7.03(s,1H),5.25(s,1H),2.87-2.89(d,4H);
MS ESI:m/z=415.0[M+H]+
实施例103
(反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000185
1H NMR(400MHz,甲醇-d4):δ8.09(s,1H),7.58(d,1H),7.45(d,1H),7.26-7.31(m,4H),7.04-7.11(m,2H),5.28(m,1H),2.87(d,4H);
MS ESI:m/z=365.1[M+H]+
实施例104
(反式)1-(4-氯苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000186
1H NMR(400MHz,甲醇-d4):δ8.09(s,1H),7.58(d,1H),7.45(d,1H),7.22-7.34(m,6H),7.04(m,1H),5.29(s,1H),2.87(d,4H);
MS ESI:m/z=381.0[M+H]+
实施例105
(反式)4-氯-氮-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲 酰基)苯磺酰胺
Figure PCTCN2017108941-appb-000187
1H NMR(400MHz,DMSO-d6):δ8.15(s,1H),7.88(d,2H),7.82(d,1H),7.61(d,2H),7.53(d,1H),7.36-7.40(m,2H),7.12(s,1H),4.67-4.73(m,1H),3.06-3.12(m,2H),2.86(t,2H),2.66-2.67(m,2H);
MS ESI:m/z=429.0[M+H]+
实施例106
(反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)苯磺酰胺
Figure PCTCN2017108941-appb-000188
1H NMR(400MHz,DMSO-d6):δ8.11(s,1H),7.79-7.81(m,3H),7.54-7.55(m1H),7.37-7.39(m,3H),7.20(s,1H),7.13(d,1H),4.70-4.72(m,1H),2.86-2.91(m,2H),2.67(s,2H),2.39(s,3H);
MS ESI:m/z=409.0[M+H]+
实施例107
(反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
Figure PCTCN2017108941-appb-000189
氮气保护冰浴下,将氯磺酰异氰酸酯(9uL,0.1mmol)溶于2mL二氯甲烷,将2-溴乙醇(7uL,0.1mmol)溶于1mL二氯甲烷地加入反应液,搅拌10分钟,化合物(反式)螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-胺(20mg,0.1mmol)和三乙胺(35uL)溶于1mL二氯甲烷,室温反应,LC-MS检测反应完全,将粗产物旋干,加入3mL乙腈, 三乙胺(35uL),3-氟苯胺(20uL),回流反应10h,然后水淬灭反应(20mL),二氯甲烷萃取(10mL*3),合并有机层,饱和氯化钠洗,无水硫酸钠干燥,过滤,浓缩,柱色谱层析(二氯甲烷:甲醇=20:1),干燥,得到呈黄色固体的目标化合物(10mg,28%).
1H NMR(400MHz,甲醇-d4):δ8.11(s,1H),7.52-7.57(m,1H),7.28-7.31(m,3H),7.12(s,1H),7.03-7.07(m,1H),6.95-6.98(m,1H),6.75-6.79(m,1H),4.58-4.52(m,1H),2.78(d,4H);
MS ESI:m/z=385.0[M+H]+
实施例108
(顺式)1-甲基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
Figure PCTCN2017108941-appb-000190
1H NMR(400MHz,CDCl3):δ8.02(s,1H),7.58-7.60(m,1H),7.47-7.49(m,1H),7.28-7.33(m,2H),7.03(s,1H),4.11-4.16(m,1H),2.98-3.06(m,2H),2.77-2.82(m,2H);
MS ESI:m/z=305.1,[M+H]+
实施例109
(顺式)2-氰基-1-甲基-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)胍
Figure PCTCN2017108941-appb-000191
将(順式)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-胺(25mg,0.1mmol)溶于2mL四氢呋喃,加入N-氰基碳亚胺二苯基酯(41mg,0.2mmol),三乙胺(66uL),室温搅拌2小时后,加入甲胺溶液0.6mL,继续搅拌两小时,滴加入1N氢氧化钠水溶液2mL,室温搅拌1小时,然后水淬灭反应(20mL),二氯甲烷萃取(10mLx3),合并有机层,饱和氯化钠洗,无水硫酸钠干燥,过滤,浓缩,柱色谱层析(二氯甲烷:甲醇=20:1),干 燥,得到黄色固体(40mg,81%).
1H NMR(400MHz,CDCl3):δ8.03(s,1H),7.63(d,1H),7.47(d,1H),7.29-7.32(m,2H),7.02(s,1H),4.57-4.61(m,1H),2.97-3.02(m,2H),2.80-2.91(m,2H);2.74(s,3H);
MS ESI:m/z=293.1,[M+H]+
实施例110
(顺/反)1-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(3-(三氟甲基)苯基)硫脲
Figure PCTCN2017108941-appb-000192
第一步:5-(2-溴苯基)-5-氰基-2-氧代环己烷-1-羧酸甲酯
Figure PCTCN2017108941-appb-000193
室温下将2-溴苯乙腈(10.0g,51mmol)及3-氯丙酸甲酯(13.4g,107mmol)加入到KOtBu(28.6g,255mmol)/THF(100mL)乳浊液中。室温下反应6h后,将反应液浓缩并用2M HCl酸化至pH=6~7。用3*50mL DCM萃取上述溶液,合并有机相用无水Na2SO4干燥,浓缩。经柱纯化(流动相PE:EA=4:1)得到浅黄色固体目标化合物9.8g,收率57%。
1H NMR(400MHz,CDCl3):δ12.24(s,1H),7.70(dd,1H),7.43(dd,1H),7.36(t,1H),7.23(t,1H),3.81(s,3H),3.39(d,1H),2.85–2.78(m,2H),2.56–2.49(m,2H),2.44–2.38(m,1H).
MS ESI:m/z=336.0,338.0,[M+H]+
第二步:1-(2-溴苯基)-4-氧代环己烷-1-腈的制备
Figure PCTCN2017108941-appb-000194
室温下将氯化钠(1.9g,31.9mmol)及第一步产物5-(2-溴苯基)-5-氰基-2-氧代环己烷-1-羧酸甲酯(9.76g,29.03mmol)加入到50mL两口瓶中,抽换氩气三次,注入12mL二甲基亚砜。150℃下反应6h后,冷至室温。将反应液用200mL饱和食盐 水稀释并用3x100mL DCM萃取上述溶液,合并有机相用无水Na2SO4干燥,浓缩。经柱纯化(流动相PE:DCM=1:1)得到白色固体目标化合物4.6g,收率57%。
1H NMR(400MHz,CDCl3):δ7.72(dd,1H),7.44(dd,1H),7.39(t,1H),7.25(t,1H),3.01–2.88(m,4H),2.63–2.58(m,2H),2.34-2.27(t,2H).
MS ESI:m/z=278.0,280.0,[M+H]+
第三步:8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-腈
Figure PCTCN2017108941-appb-000195
室温下将乙二醇(1.1g,18.0mmol)及一水合对甲苯磺酸(1.6g,8.2mmol)加入到第二步产物1-(2-溴苯基)-4-氧代环己烷-1-腈(4.6g,16.4mmol)/Toluene(100mL)溶液中。120℃下反应6h后,冷至室温。将反应液用300mL饱和食盐水稀释并用3*50mL EA萃取上述溶液。合并有机相,依次使用饱和NaHCO3,饱和食盐水洗涤,并用无水Na2SO4干燥,旋干得浅黄色固体目标化合物5.2g,收率98%。
1H NMR(400MHz,CDCl3):δ7.68(dd,1H),7.45(dd,1H),7.34(t,1H),7.20(t,1H),4.02(t,2H),3.95(t,2H),2.60–2.58(m,2H),2.24–2.13(m,4H),1.91–1.89(m,2H).
MS ESI:m/z=322.0,324.0,[M+H]+.
第四步:8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-羧酰胺
Figure PCTCN2017108941-appb-000196
室温下将KOH(18.0g,322mmol)加入到第三步产物8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-腈(5.2g,16.1mmol)/tBuOH(50mL)溶液中。100℃下反应6h后,冷至室温。往体系中加入20m水并用3*50mL DCM萃取上述溶液,合并有机相,经饱和食盐水洗涤后用无水Na2SO4干燥,旋干得浅黄色固体目标化合物4.5g,收率82%。
1H NMR(400MHz,CDCl3):δ7.63(dd,1H),7.59(dd,1H),7.35(t,1H),7.16(t,1H),5.18-5.04(br,2H),3.97(t,2H),3.92(t,2H),2.58–2.52(m,2H),2.36–2.29(m,2H),2.10–2.04(m,2H),1.69–1.62(m,2H).
MS ESI:m/z=340.1,342.1,[M+H]+.
第五步:8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-胺
Figure PCTCN2017108941-appb-000197
室温下将5.2%NaClO水溶液(48.7mL,33.2mmol)缓慢滴加入到第四步产物8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-羧酰胺(4.5g,13.3mmol)/1,4-二氧六环(100mL)溶液中。室温下反应过夜后,往反应液中加入20mL水并用3x50mL DCM萃取上述溶液。合并有机相,经饱和食盐水洗涤后用无水Na2SO4干燥,旋干得浅黄色固体目标化合物3.8g,收率90%。
1H NMR(400MHz,CDCl3):δ7.59(dd,1H),7.52(dd,1H),7.26(t,1H),7.06(td,1H),3.98(t,2H),3.94(t,2H),2.22–2.12(m,6H),1.67–1.64(m,2H).
MS ESI:m/z=312.0,314.0,[M+H]+.
第六步:1-(8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-基)-1H-咪唑
Figure PCTCN2017108941-appb-000198
室温下将40%乙二醛水溶液(4.4g,30.0mmol)、37%甲醛水溶液(2.4g,30.0mmol)及乙酸铵(2.3g,30.0mmol)缓慢滴加入到第五步产物8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-胺(3.75g,12.01mmol)/MeOH(60mL)溶液中。70℃反应6h后,冷至室温。将反应液浓缩,经柱纯化(流动相DCM:MeOH=30:1)得到浅黄色固体目标化合物3.4g,收率77%。
1H NMR(400MHz,CDCl3):δ7.83(s,1H),7.51(dd,1H),7.27(t,1H),7.15(dd,1H),7.13(s,1H),6.95(d,1H),6.89(s,1H),3.98(t,4H),3.22–3.17(m,2H),2.53–2.46(m,2H),1.76(t,4H).
MS ESI:m/z=363.0,365.0,[M+H]+.
第七步:二螺[咪唑并[5,1-a]异吲哚-5,1′-环己烷-4′,2″-[1,3]二氧杂环戊烷]
Figure PCTCN2017108941-appb-000199
室温下将Cs2CO3(2.1g,6.4mmol)、Pd(dppf)Cl2(471mg,0.6mmol)及第六步产物1-(8-(2-溴苯基)-1,4-二氧杂螺[4,5]癸烷-8-基)-1H-咪唑(1.2g,3.2mmol)加入到50mL两口瓶中,抽换氩气三次,注入10mL二甲基亚砜。120℃下反应4h后,冷至 室温。将反应液用200mL饱和食盐水稀释并用3x50mL EA萃取上述溶液。合并有机相用无水Na2SO4干燥,浓缩。经柱纯化(流动相PE:EA=1:2)得到浅褐色固体目标化合物476mg,收率52%。
1H NMR(400MHz,CDCl3):δ7.88(s,1H),7.54(d,1H),7.39(d,1H),7.35(dd,1H),7.26(t,1H),7.21(s,1H),4.07(t,4H),2.36(t,2H),2.11(t,2H),1.99(t,1H),1.96(t,1H),1.87-1.82(m,2H).
MS ESI:m/z=283.1,[M+H]+.
第八步:螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-酮
Figure PCTCN2017108941-appb-000200
室温下将2 N HCl水溶液(0.5mL)滴加到第七步产物二螺[咪唑并[5,1-α]异吲哚-5,1′-环己烷-4′,2″-[1,3]二氧杂环戊烷](28mg,0.1mmol)/THF(1mL)溶液中。50℃下反应1.5h后,冷至室温。将反应液浓缩并用5N NaOH调至pH=10。用3x5mL DCM萃取上述溶液,合并有机相用无水Na2SO4干燥,旋干得浅褐色固体化合物23mg,收率97%。
1H NMR(400MHz,CDCl3):δ7.90(s,1H),7.60(d,1H),7.42(t,1H),7.34(dd,1H),7.32(dd,1H),7.28(s,1H),2.93-2.86(m,2H),2.73(t,1H),2.69(t,1H),2.53(t,2H),2.22-2.16(m,2H).
MS ESI:m/z=239.1,[M+H]+.
第九步:(顺/反)螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-胺
Figure PCTCN2017108941-appb-000201
室温下将甲酸铵(127mg,2.0mmol)、水(0.1mL)及10%Pd/C(20mg)及第八步产物螺[环己烷-1,5’-咪唑并[5,1-α]异吲哚]-4-酮(24mg,0.1mmol)加入到50mL两口瓶中,抽换Ar三次,注入1mL甲醇.室温反应24h和48h后分别补加甲酸铵(39mg,0.6mmol).室温反应60h后,补加5mL甲醇,然后过滤出去Pd/C。将滤液用40%NaOH水溶液调至pH=12并用3*5mL DCM萃取上述溶液,合并有机相用无水Na2SO4干燥,浓缩。经柱纯化(流动相DCM:MeOH=15:1,0.5%NH3-H2O)得到淡黄色固体目标化合物16mg,收率67%(顺:反=1:1)。
MS ESI:m/z=240.1,[M+H]+.
第十步1-(螺[环己基-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(3-(三氟甲基)苯 基)硫脲
Figure PCTCN2017108941-appb-000202
(顺:反=1:1)
在0℃将3-(三氟甲基)苯基异硫氰酸酯(20mg,0.1mmol)加入到第九步产物(顺/反)螺[环己烷-1,5’-咪唑并[5,1-α]异吲哚]-4-胺(20mg,0.1mmol)及Et3N(17mg,0.2mmol)的1mL二氯甲烷混合溶液中。升至室温,反应10分钟后,用0.5mL淬灭反应,浓缩。经柱纯化(流动相DCM:MeOH=15:1)得到淡黄色固体化合物24mg,收率63%(顺:反=1:1)。
MS ESI:m/z=442.8,[M+H]+
实施例111
(顺/反)1-苯基-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
Figure PCTCN2017108941-appb-000203
(顺:反=1:1)
MS ESI:m/z=375.0,[M+H]+
实施例112
(顺/反)1-(4-氯苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
Figure PCTCN2017108941-appb-000204
(顺:反=1:1)
MS ESI:m/z=409.0,[M+H]+
实施例113
(顺/反)1-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(4-(三氟甲基)苯 基)硫脲
Figure PCTCN2017108941-appb-000205
(顺:反=1:1)
MS ESI:m/z=443.0,[M+H]+
实施例114
(顺/反)1-(4-氯-3-(三氟甲基)苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)脲
Figure PCTCN2017108941-appb-000206
(顺:反=1:1)
MS ESI:m/z=461.0,[M+H]+
实施例115
(顺/反)N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)萘基-2-磺酰胺
Figure PCTCN2017108941-appb-000207
(顺:反=1:1)
依据实例例2的方法,用2-萘磺酰氯替代3-(三氟甲基)苯基异硫氰酸酯作为原料,得到白色固体化合物17mg,收率47%。
MS ESI:m/z=430.0,[M+H]+
实施例116
(顺/反)3-溴-N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)苯甲酰胺
Figure PCTCN2017108941-appb-000208
(顺:反=1:1)
MS ESI:m/z=422.0,424.0[M+H]+
实施例117
(顺/反)4-(1-苄基-1H-1,2,3-三唑-4-基)螺[环己烷1,5’-咪唑并[5,1-a]异吲哚]-4-醇
Figure PCTCN2017108941-appb-000209
第一步:(顺/反)4-乙炔基-螺[环己烷1,5’-咪唑并[5,1-α]异吲哚]-4-醇的制备
Figure PCTCN2017108941-appb-000210
在反应瓶中加入5mL无水四氢呋喃,加入乙炔基溴化镁(1.7mL,0.8mmol),冰浴搅拌,把原料螺[环己烷-1,5’-咪唑并[5,1-α]异吲哚]-4-酮(20mg,0.1mmol)溶于四氢呋喃(1mL)滴入乙炔基溴化镁中,滴毕,自然回到室温反应1小时,TLC显示原料消失。滴加饱和氯化铵淬灭反应。加入乙酸乙酯20mL和水10mL,萃取,有机相干燥,过滤,浓缩,得到粗品,柱层析纯化分离,得到目标化合物(20mg,收率90%)。
MS ESI:m/z=265.1,[M+H]+.
第二步:4-(1-苄基-1H-1,2,3-三唑-4-基)螺[环己烷1,5’-咪唑并[5,1-a]异吲哚]-4-醇
Figure PCTCN2017108941-appb-000211
(顺∶反=2∶1)
将第一步产物(20mg,0.076mmol),和苄基叠氮(20mg,0.2mmol)五水硫酸铜(14mg,0.1mmol)和抗坏血酸钠(15.00mg,0.1mmol),加入二氯甲烷,水,甲醇(2mL,1mL,1mL),室温搅拌24小时,TLC显示原料消失,反应液中加入二氯甲烷(10mL),用硅藻土过滤,滤液浓缩,拌样,柱层析,得到目标化合物10mg,收率33%
MS ESI:m/z=398.1[M+H]+
实施例118A
(顺式)(1s,3s)-螺[环丁烷-1,5’-咪唑并[5,1-a]异吲哚]-3-胺
Figure PCTCN2017108941-appb-000212
第一步:顺-(1s,3s)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000213
室温下将Cs2CO3(7.9g,24.3mmol)、Pd(dppf)Cl2(1.8g,2.4mmol)及顺式3-(2-溴苯基)-3-(1H-咪唑-1-基)环丁基-1-醇(3.6g,12.1mmol)加入到50mL两口瓶中,抽换Ar三次,注入8mL二甲基亚砜。120℃下反应8h后,冷至室温。浓缩溶剂,将残余液用200mL饱和食盐水稀释并用3x50mL EA及3x50mL DCM萃取上述溶液,合并有机相用无水Na2SO4干燥,浓缩。经柱纯化(流动相DCM:MeOH=25:1)得到红褐色固体目标化合物1.8g,收率70%。
1H NMR(400MHz,CDCl3):δ7.95(s,1H),7.50(d,1H),7.40(d,1H),7.35(t,1H),7.30(t,1H),7.14(s,1H),4.79(quint,1H),3.17-3.09(m,2H),2.81-2.76(m,2H).
MS ESI:m/z=213.1,[M+H]+.
第二步:反-(1r,3r)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基苯甲酸酯
Figure PCTCN2017108941-appb-000214
室温下将三苯基膦(393mg,1.5mmol)、苯甲酸(92mg,0.8mmol)及第一步产物顺-螺[环丁烷基-1,5’-咪唑并[5,1-α]异吲哚]-3-醇(159mg,0.8mmol)加入到50mL两口瓶中,抽换Ar三次,注入4mL四氢呋喃。然后冰浴下将DIAD(273mg,1.4mmol)的THF/2mL溶液滴加到上述体系中,升至室温反应4h,停止反应。反应液浓缩,经柱纯化(流动相DCM:MeOH=20:1)得到浅红色固目标体化合物187mg,收率79%。
1H NMR(400MHz,CDCl3):δ8.01(s,1H),7.70-7.65(m,4H),7.57-7.53(m, 3H),7.39(t,1H),7.34(t,1H),7.27(s,1H),5.79(quint,1H),3.16(m,4H).
MS ESI:m/z=317.1,[M+H]+.
第三步:反-(1r,3r)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-醇
Figure PCTCN2017108941-appb-000215
室温下将LiOH-H2O(243mg,5.8mmol)/H2O(2mL)水溶液滴加到第二步产物反-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基苯甲酸酯(610mg,1.9mmol)/MeOH(15mL)溶液中。室温下反应10分钟后,将反应液浓缩并用3x5mL EA萃取上述溶液,合并有机相用无水Na2SO4干燥。反应液浓缩,经柱纯化(流动相DCM:MeOH=20:1)得到浅褐色固目标体化合物316mg,收率77%。
1H NMR(400MHz,CDCl3):δ7.76(s,1H),7.66(dd,1H),7.49(dd,1H),7.37-7.31(m,2H),7.16(s,1H),4.99(quint,1H),2.90(m,4H).
MS ESI:m/z=213.1,[M+H]+.
第四步:反-(1r,3r)-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基甲磺酸酯
Figure PCTCN2017108941-appb-000216
冰浴下将MsCl(339mg,2.96mmol)和Et3N(599mg,5.9mmol)依次滴加到第三步产物反-螺[环丁烷基-1,5’-咪唑并[5,1-α]异吲哚]-3-醇(315mg,1.5mmol)/DCM(10mL)溶液中。0℃下反应10分钟后,往反应液中加入2mL饱和食盐水并用3x5mL DCM萃取上述溶液,合并有机相并用无水Na2SO4干燥旋干得到浅褐色固体目标化合物412mg,收率96%。
1H NMR(400MHz,CDCl3):δ7.79(s,1H),7.62(dd,1H),7.50(dd,1H),7.40-7.35(m,2H),7.18(s,1H),5.57(quint,1H),3.25-3.20(m,2H),3.14(s,3H),3.13-3.09(m,2H).
MS ESI:m/z=291.0,[M+H]+.
第五步:顺(1s,3s)-3-叠氮螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]
Figure PCTCN2017108941-appb-000217
冰浴下将NaN3(277mg,4.3mmol)加入到第四步产物反-螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚]-3-基甲磺酸酯(412mg,1.4mmol)/DMF(6mL)溶液中。75℃下反应 5h后,往反应液中加入50mL饱和食盐水并用3x15mL EA萃取上述溶液,合并有机相并用无水Na2SO4干燥。反应液浓缩,经柱纯化(流动相DCM:MeOH=25:1)得到浅褐色油状化合物209mg,收率67%。
1H NMR(400MHz,CDCl3):δ7.90(s,1H),7.51(d,1H),7.44(d,1H),7.38(t,1H),7.31(t,1H),7.17(s,1H),4.41(quint,1H),3.18-3.12(m,2H),2.88-2.81(m,2H).
MS ESI:m/z=238.1,[M+H]+.
第六步:顺(1s,3s)--螺[环丁烷基-1,5’-咪唑并[5,1-α]异吲哚]-3-胺的制备
Figure PCTCN2017108941-appb-000218
氩气氛围中,室温下将5%Pd/C(20mg)加入到第五步产物顺-3-叠氮螺[环丁烷基-1,5’-咪唑并[5,1-a]异吲哚](209mg,0.9mmol)/EtOH(4mL)溶液中。1atm H2中室温反应24h后,加入硅藻土过滤,滤饼用5mLEtOH洗涤,合并有机相,旋干得到浅黄色固体化合物170mg,收率87%。
1H NMR(400MHz,CDCl3):δ7.92(s,1H),7.49(t,2H),7.35(t,1H),7.28(t,1H),7.15(s,1H),3.95(quint,1H),3.12-3.04(m,2H),2.52-2.47(m,2H).
MS ESI:m/z=212.1,[M+H]+
实施例118(顺式)(1r,3r)-3-(1-苯基-1H-1,2,3-三唑-4-基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-醇的制备
Figure PCTCN2017108941-appb-000219
步骤1:螺[环丁烷1,5'-咪唑并[5,1-a]异吲哚]-3-酮的制备
Figure PCTCN2017108941-appb-000220
将螺[环丁烷1,5'-咪唑并[5,1-a]异吲哚]-3-醇(0.5g,2.35mmol),放入瓶中,加入5mL二氯甲烷,冰浴下搅拌,加入碳酸氢钠(1.90g,23.55mmol),加入Dess-Martin氧化剂(1.50g,3.53mmol),室温搅拌,直到原料消失。加入20ml二氯甲烷和10ml水萃取,得到有机相,干燥,过滤,浓缩,柱层析纯化,得到目标化合物(400.00mg,收率80%)。
1H NMR(500MHz,甲醇-d4):δ7.86(s,1H),7.59(d,1H),7.47-7.42(m,2H),7.37-7.35(m,1H),7.25(s,1H),3.872.82(m,4H),
MS ESI:m/z=211.1,[M+H]+
步骤2:(顺式)(1r,3r)--3-乙炔基螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-醇的制备
Figure PCTCN2017108941-appb-000221
冰浴下,原料螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-酮(20mg,0.095mmol)加入到含有乙炔基溴化镁的反应瓶中,自然回到室温,搅拌直到反应完全。加入饱和氯化铵溶液(5mL)淬灭反应,加入乙酸乙酯(10ML),萃取,有机相用硫酸钠干燥。过滤,FLASH纯化(0-5%甲醇/二氯甲烷),得到目标产物(20mg,收率100%)
MS ESI:m/z=237.0,[M+H]+
步骤3:(顺式)(1r,3r)-3-(1-苯基-1H-1,2,3-三唑-4-基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-醇的制备
Figure PCTCN2017108941-appb-000222
原料3-乙炔基螺[环丁烷-1,5'-咪唑并[5,1-α]异吲哚]-3-醇(14.0mg,0.063mmol)和苯基叠氮(15.0mg,0.063mmol),胆矾(12.0mg,0.063mmol)以及抗坏 血酸钠(13mg,0.063mmol)均匀分散在水-二氯甲烷-甲醇(体积比1:1:1)的溶剂中,室温搅拌24小时,用二氯甲烷萃取(10mLx3),有机相用硫酸钠干燥。FLASH纯化,0-5%的甲醇/二氯甲烷,得到化合物(4.0mg,收率17%)
1H NMR(500MHz,甲醇-d4):δ8.23(s,1H),8.11(s,1H),8.04-8.02(m,1H),7.77(d,2H),7.58-7.46(m,4H),7.42-7.37(m,2H),3.58(d,2H),3.30(d,2H).
MS ESI:m/z=336.0,[M+H]+
实施例119
1'-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
Figure PCTCN2017108941-appb-000223
MS ESI:m/z=400.1,[M+H]+
实施例120
(顺式)(1s,3s)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
Figure PCTCN2017108941-appb-000224
ESI:m/z=386.1,[M+H]+
实施例121
(顺式)(1r,3r)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
Figure PCTCN2017108941-appb-000225
MS ESI:m/z=386.1,[M+H]+
实施例122
(顺/反)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-胺
Figure PCTCN2017108941-appb-000226
MS ESI:m/z=414.1,[M+H]+
实施例123
(顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)苯甲酰胺
Figure PCTCN2017108941-appb-000227
MS ESI:m/z=442.2,[M+H]+
实施例124
(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)(螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-基)甲酮
Figure PCTCN2017108941-appb-000228
MS ESI:m/z=428.1,[M+H]+
实施例125
(反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000229
MS ESI:m/z=414.1,[M+H]+
实施例126
(顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
Figure PCTCN2017108941-appb-000230
MS ESI:m/z=414.1,[M+H]+
实施例127
(顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并 [5,1-a]异吲哚]-4-基)脲
Figure PCTCN2017108941-appb-000231
MS ESI:m/z=457.1,[M+H]+
实施例128
(顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000232
MS ESI:m/z=429.1,[M+H]+
实施例129
N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-氨甲酰
Figure PCTCN2017108941-appb-000233
MS ESI:m/z=443.1,[M+H]+
实施例130
(顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)苯磺酰胺
Figure PCTCN2017108941-appb-000234
MS ESI:m/z=478.1,[M+H]+
实施例131
(反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
Figure PCTCN2017108941-appb-000235
Step 14-(2-氟-4-异氰酰苯基)-1-甲基-1H-吡唑
Figure PCTCN2017108941-appb-000236
把三光气(67mg,0.22mmol)溶于2mL甲苯中,室温下慢慢滴入3-氟-4-(1-(甲基-1H-吡唑-4-基))苯胺(60mg,0.32mol)和三乙胺(0.05mL)的混合甲苯1.5mL溶液。70℃加热两个小时,冷至室温。滤去三乙胺盐酸盐沉淀,滤液浓缩得到红色产品(36mg,53%)。
Step 21-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
冰水浴下(0℃),把4-(2-氟-4-异氰酰苯基)-1-甲基-1H-吡唑(10mg,0.04mmol)加入到(1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺(10mg,0.04mmol)和三乙胺(5mg,0.05mmol)的二氯甲烷(1mL)溶液中。室温下搅拌10min,反应用水淬 灭,浓缩。用二氯甲烷/甲醇(20:1,Rf=0.25)硅胶柱层析纯化得到呈浅黄色固体产物(3mg,15%)。
MS ESI:m/z=429.1,[M+H]+
实施例132
(顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)硫脲
Figure PCTCN2017108941-appb-000237
MS ESI:m/z=473.2,[M+H]+
实施例133
N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-硫代酰胺
Figure PCTCN2017108941-appb-000238
MS ESI:m/z=459.1,[M+H]+
实施例134
(反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000239
Step 14-(2-氟-4-异氰硫酰苯基)-1-甲基-1H-吡唑
Figure PCTCN2017108941-appb-000240
3-氟-4-(1-(甲基-1H-吡唑-4-基))苯胺(150mg,0.78mmol)和三乙胺(269mg,2.4mmol)的3mL丙酮溶液在室温下搅拌10分钟。滴加1.5mL二硫化碳,产生沉淀,继续室温过夜搅拌。过滤得到滤饼,70℃干燥1小时。将干燥的产物溶解到3mL氯仿中,加入三光气(2.70mmol)的3mL氯仿溶液。搅拌过夜,过滤,得到淡黄色溶液。用石油醚/乙酸乙酯(2:1,Rf=0.25)硅胶柱层析纯化得到白色固体(146mg,80%)。
Step 21-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
冰水浴下,把4-(2-氟-4-异氰硫酰苯基)-1-甲基-1H-吡唑(13mg,0.05mmol)加入到(1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺(10mg,0.04mmol)的二氯甲烷(1mL)溶液中。室温搅拌10min。用二氯甲烷/甲醇(15:1)硅胶柱层析纯化得到浅黄色固体(8mg,38%)。
MS ESI:m/z=445.1,[M+H]+
实施例135
(顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
Figure PCTCN2017108941-appb-000241
MS ESI:m/z=445.1,[M+H]+
实施例136
1'-((3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)磺酰)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
Figure PCTCN2017108941-appb-000242
MS ESI:m/z=464.1,[M+H]+
实施例137
(顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000243
MS ESI:m/z=450.0,[M+H]+
实施例138
(反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
Figure PCTCN2017108941-appb-000244
MS ESI:m/z=450.1,[M+H]+
上述实施例制备的化合物如下表1。
表1.本发明化合物列表
Figure PCTCN2017108941-appb-000245
Figure PCTCN2017108941-appb-000246
Figure PCTCN2017108941-appb-000247
Figure PCTCN2017108941-appb-000248
Figure PCTCN2017108941-appb-000249
Figure PCTCN2017108941-appb-000250
Figure PCTCN2017108941-appb-000251
Figure PCTCN2017108941-appb-000252
Figure PCTCN2017108941-appb-000253
Figure PCTCN2017108941-appb-000254
Figure PCTCN2017108941-appb-000255
Figure PCTCN2017108941-appb-000256
Figure PCTCN2017108941-appb-000257
Figure PCTCN2017108941-appb-000258
Figure PCTCN2017108941-appb-000259
Figure PCTCN2017108941-appb-000260
Figure PCTCN2017108941-appb-000261
Figure PCTCN2017108941-appb-000262
实施例139
活性测试
(1)IDO或TDO蛋白的诱导表达及纯化方法
首先PCR扩增IDO基因,扩增的PCR产物回收,然后将pET28a质粒(购自上海宝曼生物科技有限公司)和IDO胶回收产物用EcoR I和Xho I两种限制性内切酶进行酶切(37℃,酶切2h),跑胶,回收,T4连接酶链接过夜连接产物加入到DH5α感受态,冰上放置30min,42℃热击90s,摇菌涂板,挑取单克隆,PCR鉴定,测序鉴定,全部正确,即pET28a-IDO质粒构建成功。
将构建好的含有pET28a-IDO质粒的BL21,接种于含有终浓度为50μg/ml的kana抗生素LB培养基中37℃大摇至OD600为0.6-0.8,加入至终浓度为40μM的氯高铁血红素、50μg/ml的L-色氨酸和0.5mM的IPTG(异丙基-β-D-硫代半乳糖苷),16℃诱导20h;诱导后,4℃,6000rpm离心收集菌体,收集的菌体用50mM PBS(pH7.5)清洗一次,再离心收集菌体。
将收集的菌体用裂解液(50mM PBS pH7.5)重悬,并加入终浓度为1mM的L-色氨酸、10μM氯高铁血红素和PMSF高压破碎或超声裂解(功率40%裂解20min,冰上放置),将裂解后的细菌4℃、18000×g离心45min,弃去沉淀,保留上清并置于冰上0.22μm膜过滤;将镍柱用裂解液(50mM PBS pH7.5)平衡3个柱体积,然后将裂解上清上样到镍柱上,上样之后用漂洗液(50mM PBS pH7.5,20mM咪唑)清洗4个柱体积,最后用洗脱液(50mM PBS pH7.5,250mM咪唑)洗脱蛋白;将洗脱的蛋白溶液进行透析8h,透析溶液为50mM PBS pH7.5,透析之后蛋白样品浓缩,分装,液氮速冻,放入-80℃保存备用。
TDO用上述同样方法表达和纯化。
(2)IDO酶抑制活性测试方法
首先将化合物进行3倍梯度稀释,各个浓度及DMSO对照组各取1μL加入到96孔板中;加入50μL配好的IDO酶溶液(终浓度40nM):加入25μL底物1(甲基蓝3.5μM,0.2μg/μL过氧化氢酶,50mM PBS(pH7.5),均为终浓度)混合溶液,加入25μL的底物2(1.5mM L-Trp,20mM抗坏血酸钠,20mMPBS(pH7.5),均为终浓度)混合溶液起始反应。最后OD321nm读数40min。
(3)细胞活性测试方法
Hela细胞(100μL)接种在96孔板上,接种量为每个孔5×103,37℃,5%CO2恒温培养箱内培养24h。第二天,化合物3倍梯度稀释后,各取1μL加入到96孔板内,然后将含有人的干扰素γ(IFN-γ,终浓度50ng/mL)的培养基(RPMI-1640,含有10%胎牛血清)100μL加入到96孔板中,使最终体积为200μL。48小时孵化后,每个孔取140μL上清液转移到一个新的96孔板上并且每孔加入10μL 6.1N三氯乙酸混合,50℃孵育30分钟,IDO催化N甲酰犬尿氨酸为犬尿氨酸。孵育结束后反应混合物2500转离心10分钟去掉沉淀物。每个孔100μL上清液转移到另一个新的96孔板与100μL 2%(w/v)二甲氨基苯甲醛乙酸溶液混合。犬尿氨酸分离后,用酶标仪SPECTRAmax i3 reader在480nm测定数值。
各化合物的IDO酶抑制活性和细胞抑制活性的测试结果如表2所示。
表2.IDO酶、IDO细胞和TDO酶抑制活性测试结果(IC50,uM)
Figure PCTCN2017108941-appb-000263
Figure PCTCN2017108941-appb-000264
Figure PCTCN2017108941-appb-000265
上述结果表明,本发明化合物(包括所有异构体)均具有针对IDO酶和TDO酶的抑制活性。
在本发明提及的所有文献都在本申请中引用作为参考,就如同每一篇文献被单独引用作为参考那样。此外应理解,在阅读了本发明的上述讲授内容之后,本领域技术人员可以对本发明作各种改动或修改,这些等价形式同样落于本申请所附权利要求书所限定的范围。

Claims (10)

  1. 一种式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药:
    Figure PCTCN2017108941-appb-100001
    式中,
    A选自下组:S、O、-C(R1)2-或无;
    B选自下组:-C(R1a)2-或无;
    E和M分别独立地选自:N或CR1
    各R1独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基、OH、取代或未取代的O-C1-C6烷基、取代或未取代的O-C3-C8环烷基;
    各R1a独立地选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C3-C8环烷基;
    所述R1、R1a中,所述取代是指具有选自A组的一个或多个(如1、2、3、4个)取代基:卤素、羟基、-NH2、硝基、-CN、C1-C4烷基、C1-C4卤代烷基、O-C1-C4烷基、C3-C8环烷基、O-C3-C8环烷基、C2-C4链烯基、C2-C4炔基、苯基、苄基;
    Figure PCTCN2017108941-appb-100002
    选自下组:取代或未取代的C6-C10芳基、取代或未取代的C3-C10杂芳基且所述杂芳基具有1-3个选自S、O或N的杂原子;其中,所述取代是指具有选自B组的一个或多个(如1、2、3、4个)取代基:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、氨基、硝基、取代或未取代的C1-C6醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2Rb、-SO2NRaRb
    X选自下组:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、OH、取代或未取代的OC1-C6烷基、取代或未取代的OC3-C8环烷基、NRaRb或NRaC(O)R3
    并且,当X选自:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C1-C6烷硫基、取代 或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(S)NR2R3、-O(CR1 2)nC(O)NR2R3、-S(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)R3、-NR2C(O)(CR1 2)nC(O)R3、-NR2(CR1 2)nC(O)O(CR1 2)nR3、-NR2(CR1 2)nC(S)NR2R3、-(CR1 2)nNR2C(S)NR2R3、-(CR1 2)nNR2C(O)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-O(CR1 2)nC(O)NR2S(O)2R3、-S(CR1 2)nC(O)NR2S(O)2R3、-NR2(CR1 2)nS(O)2NR2R3、-NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-NR2S(O)2R3、-(CR1 2)nS(O)2R3、NR2C(=N-CN)NR2R3;所述取代是指具有选自C组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基、OSO2NH2、NHSO2NH2、氨基、硝基、醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2
    当X选自:OH、OC1-C6烷基、O-C3-C8环烷基、NRaRb或NRaC(O)R3时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、取代或未取代的C3-C10环烷基、取代或未取代的C3-C10杂环烷基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3;所述取代是指具有选自D组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、取代或未取代的C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CH2-C6-C10芳基、-CH2-C6-C10芳基、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2、羟基、OSO2NH2、NHSO2NH2、C3-C6杂环基;
    其中,各Ra和各Rb分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、或Ra和Rb和与之相连的N原子共同形成取代或未取代的4-8元杂环,且所述的杂环具有至少一个N和0-2个选自S、O的杂原子,其中所述的含氮杂环中N为NH或NRg形式,其中Rg独立地为C1-C10烷基、C3-C10环烷基、C6-C20芳基、或C3-C14杂芳基;其中,所述取代指具有选自E组的一个或多个(如1、2、3、4个)取代基:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、OSO2NH2、NHSO2NH2、氨基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2Rb、-SO2NRaRb
    各R2和各R3分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未 取代的C3-C14杂芳基、取代或未取代的C3-C10杂环基、取代或未取代的氨基;
    Figure PCTCN2017108941-appb-100003
    为取代或未取代的3-10元碳环或杂环,所述杂环具有至少一个碳原子被选自下组的杂原子取代:O、S、S(O)、S(O)2、C(O)、C(S)和NRg;其中,所述取代指具有选自F组的一个或多个(如1、2、3、4个)取代基:OH、卤素、C1-6烷基;
    在A组、B组、C组、D组、E组、F组、R2、R3的取代基中,所述取代指具有选自G组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、-O-C1-C6烷基、-CN、-CF3、-O-CF3、C3-C10环烷基、C2-C8酯基、C6-C10芳基;
    Z是碳原子或N;当Z是N时,X不存在;
    n为0至8的整数。
  2. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,所述式(I)化合物选自下组所示的式(II)-式(IV)化合物:
    式(II)化合物:
    Figure PCTCN2017108941-appb-100004
    式中,
    Figure PCTCN2017108941-appb-100005
    选自下组:
    Figure PCTCN2017108941-appb-100006
    Figure PCTCN2017108941-appb-100007
    其中,Rc为一个选自下组的基团:H、卤素、取代或未取代的C1-C6烷基、取代或未取代的C3-C10环烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C10环烷氧基、羟基、氨基、硝基、取代或未取代的C1-C6醛基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb;其中各R3、Ra和各Rb如上所定义;
    m是1至4的整数;
    q是1、2或3;
    r是1或2;
    Figure PCTCN2017108941-appb-100008
    Z、X、Y如上所定义;或者
    式(III)化合物:
    Figure PCTCN2017108941-appb-100009
    式中,X、Y、Z、Rc如上所定义,m是1至4的整数;或者
    式(IV)化合物:
    Figure PCTCN2017108941-appb-100010
    式中,
    式中,Rc为一个选自下组的基团:H、卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb
    其中各R3、Ra和各Rb如上所定义;
    m是1至4的整数;
    Figure PCTCN2017108941-appb-100011
    为选自下组的取代或未取代的3-7元杂环:
    Figure PCTCN2017108941-appb-100012
    X选自下组:OH、取代或未取代的C1-C6烷基、取代或未取代的C3-C6环烷基、烷基OC1-C6烷基、OC3-C8环烷基、NRaRb或NRaC(O)R3
    Figure PCTCN2017108941-appb-100013
    选自下组:取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基,其中芳环和杂芳环可被一个或多个选自下组的基团E取代:卤素、C1-C6烷基、C3-C8环烷基、C1-C6卤代烷基、C1-C6烷氧基、或未取代的C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、OH、OSO2NH2、NHSO2NH2、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2;其中各Ra和各Rb各自独立为氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基;Ra和Rb可以一起可以形成四至八元杂环,其中杂原子可以是硫、氧、NH或NRg;或者
    式(V)化合物:
    Figure PCTCN2017108941-appb-100014
    式中,
    Rc为一个选自下组的基团:H、卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其中各R3、Ra和各Rb如上所定义;
    m是1至4的整数;
    Figure PCTCN2017108941-appb-100015
    选自下组:
    Figure PCTCN2017108941-appb-100016
    Rd选自下组:H、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、取代或未取代的C1-C6卤代烷基、 取代或未取代的C6-C20芳基、取代或未取代的C3-C14杂芳基;所述取代指具有选自H组的一个或多个(如1、2、3、4个)取代基:卤素、C1-C6烷基、C3-C8环烷基、C1-C6卤代烷基、C1-C6烷氧基、C3-C10环烷基、取代或未取代的C3-C10杂环烷基、C6-C20芳基、C3-C14杂芳基羟基、OC6-C20芳基、OC3-C14杂芳基、羟基氨基、硝基、醛基、OH、OSO2NH2、NHSO2NH2、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CO2C1-C6烷基、-CONRaRb、-SO2R3、-SO2NRaRb、-P(O)Me2、-P(O)(OMe)2;或者
    式(VI)化合物:
    Figure PCTCN2017108941-appb-100017
    式中,
    Rc为一个选自下组的基团:卤素、取代或未取代的C1-C6烷基、取代或未取代的C1-C6烷氧基、取代或未取代的C3-C8环烷基、取代或未取代的C3-C8环烷氧基、羟基、-CF3、-CN、-SF5、NRaRb、羧基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其中各R3、Ra和各Rb如上所定义;
    m是1至4的整数;
    Figure PCTCN2017108941-appb-100018
    选自下组:
    Figure PCTCN2017108941-appb-100019
    Rf选自下组:H、SO2NH2、C1-C6卤代烷基、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基氨基;所述取代指具有选自I组的一个或多个(如1、2、3、4个) 取代基:C1-C6烷基、C1-C6烷氧基、C1-C6环烷基、C1-C6卤代烷基、烷氧基、杂环基,芳基、杂芳基、-CF3、-CN、-SF5、NRaRb、羧基、羟基、OSO2NH2、NHSO2NH2、氨基、-CORa、-CONRaRb、-SO2R3、-SO2NRaRb;其中,所述Ra和Rb和与之相连的N原子共同形成取代或未取代的4-8元杂环,且所述的杂环具有至少一个N和0-2个选自S、O的杂原子,其中所述的含氮杂环中N为NH或NRg形式,其中Rg独立地为C1-C10烷基、C3-C10环烷基、C6-C20芳基、或C3-C14杂芳基。
  3. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,所述
    Figure PCTCN2017108941-appb-100020
    选自下组:取代或未取代的噻吩基、取代或未取代的呋喃基、或取代或未取代的吡啶基。
  4. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,所述
    Figure PCTCN2017108941-appb-100021
    选自下组:
    Figure PCTCN2017108941-appb-100022
    Z如上所定义。
  5. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,当X选自:H、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(S)NR2R3、-O(CR1 2)nC(O)NR2R3、-S(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)NR2R3、-NR2(CR1 2)nC(O)R3、-NR2C(O)(CR1 2)nC(O)R3、-NR2(CR1 2)nC(O)OR3、-NR2(CR1 2)nC(S)NR2R3、-(CR1 2)nNR2C(O)NR2R3、-(CR1 2)nNR2C(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-NR2(CR1 2)nC(S)NR2R3、-NR2C(O)NR2R3、-NR2C(S)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-O(CR1 2)nC(O)NR2S(O)2R3、-S(CR1 2)nC(O)NR2S(O)2R3、-NR2(CR1 2)nS(O)2NR2R3、-NR2S(O)2NR2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3、NR2C(=N-CN)NR2R3;所述取代是指具有选自C组的一个或多个(如1、2、3、4个)取代基。
  6. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,当X选自下组:OH、OC1-C6烷基、OC3-C8环烷基、NRaRb或NRaC(O)R3时,Y选自下组:取代或未取代的C1-C10烷基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基、-(CR1 2)nC(O)NR2R3、-(CR1 2)nC(O)NR2S(O)2R3、-(CR1 2)nS(O)2NR2R3、-(CR1 2)nNR2S(O)2NR2R3、-(CR1 2)nNR2C(O)NR1S(O)2R3、-(CR1 2)nC(O)NR2S(O)2NR2R3、-(CR1 2)nS(O)(=NR2)R3、-(CR1 2)nS(O)(=NCN)R3、-(CR1 2)nS(O)R3、-(CR1 2)nS(O)2R3;所述取代是指具有选自D组的一个或多个(如1、2、3、4个)取代基。
  7. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,所述各R2和各R3分别独立地选自下组:氢、取代或未取代的C1-C10烷基、取代或未取代的C3-C10环烷基、取代或未取代的C2-C10烯基、取代或未取代的C6-C20芳基、或取代或未取代的C3-C14杂芳基。
  8. 如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药,所述式(I)化合物选自下组:
    (顺/反)3-苯基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
    (顺/反)3-氟苯基)(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)甲醇
    (顺/反)3-(1,4-二氧杂螺[4.5]癸-7-烯-8-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
    (顺式)4-甲基-N-((1s,3s)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
    (反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
    (顺式)4-甲基-N-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺/反)3-氟-N-(螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺式)4-氟-N-(1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
    (反式)4-氟-N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
    (反式)N-((1r,3r)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己烷甲酰胺
    (顺式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环己基磺酰胺
    (顺式)3-氯-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3-溴-4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷磺酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环丙烷甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙烷-2-磺酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-2-磺酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)萘-1-磺酰胺
    (反式)4-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2-氟-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2,4-二氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)N((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-2-萘甲酰胺
    (反式)3-氯-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-氰基-N-(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3-溴-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2-甲氧基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)-3-(三氟甲基)苯甲酰胺
    (反式)苄基(1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸叔丁酯
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)异丁酰胺
    (反式)((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酸苯酯
    (顺式)N((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙烯酰胺
    (反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)甲基磺酰胺
    (顺/反)3-甲氧基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3,4-二氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺/反)2-甲基-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3-甲基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2,4,6-三甲基-N-((1r,3r)-螺[环丁基烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2-氯-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3-氟-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)3-三氟甲氧基-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-溴-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-氟-N-((1r,3r)-螺[环丁烷,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)4-氧代-4-苯基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丁酰胺
    (反式)2-([1,1’-联苯]-4-基)-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
    (反式)N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)环戊烷甲酰胺
    (反式)3-环戊基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)丙酰胺
    (反式)2-环丙基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)乙酰胺
    (反式)叔丁基-2-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)吡咯烷-1-甲酸叔丁酯
    (反式)4-羟基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺/反)N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-[1,1’-二苯基]-4-甲酰胺
    (顺式)2-甲基-2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
    (顺式)2-苯基-N-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)丙酰胺
    (反式)2-溴-5-甲氧基-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)2-溴-N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)烟酰胺
    (顺/反)3-氟-N-(螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)苯甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)四氢呋喃-3-甲酰胺
    (反式)N-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)呋喃-3-甲酰胺
    (反式)(1r,3r)-N-(萘乙酰胺-2-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-甲酰 胺
    3-(苄氧基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
    (顺/反)3-丁氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
    (顺/反)3-乙氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
    (顺/反)3-甲氧基螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
    (反式)1-苯基-2-((1r,3r)-螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基)乙醇
    (反式)(1r,3r)-N-(3-溴-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-(对甲苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-(3-氯-4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰
    (反式)(1r,3r)-N-(3-氟苯基)螺[环丁基1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-(3-氯苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)--N-(3-溴苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (顺式)(1r,3S,8a'S)-N-(3-溴苯基)-8a'H-螺[环丁基-1,8'-茚并[1,2-c]吡咯]-3-甲酰胺
    N-(4-氟苯基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-(萘-2-基磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-((4-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-((3-氟苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-N-((3-氯苯基)磺酰基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-甲酰胺
    (反式)(1r,3r)-3-(甲硫基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]
    (反式)(1r,3r)-3-(甲基磺酰基)螺[环丁烷,5’-咪唑并[5,1-a]异吲哚]
    (反式)(1R,3r)-3-((R)-甲基亚磺酰基)螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]
    (顺/反)3-(1-苄基-1H-1,2,3-三唑-4-基)螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-醇
    (反式)(1r,3r)-3-(4-环己基-1氢-1,2,3三唑1-基)螺[c环丁烷-1,5’-咪唑[5,1-a]异吲哚]
    (反式)(1r,3r)-3-(4-苯基-1H-1,2,3三唑-1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
    (反式)(1r,3r)-3-(4-环丙基1H-1,2,3三唑1-基)螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]
    (反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
    (反式)3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)磺酰脲
    (反式)1-苯基-3-((1r,3r)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
    (顺式)1-(3-氟苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
    (顺式)1-(4-氯苯基)-3-(1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
    (顺式)1-(4-三氟甲基苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑5,1-a]异吲哚l]-3-基)硫脲
    (顺式)1-((1s,3s)-螺[环丁烷-1,5'-咪唑[5,1-a]异吲哚]-3-基)-3-(4-(三氟甲基)苯基)硫脲
    (顺式)1-(3-氯-4-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
    (顺式)1-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)-3-(3-(三氟甲基)苯基)脲
    (顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
    (顺式)1-(4-氯-3-(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷1,5’-咪唑[5,1-a]异吲哚]-3-基)硫脲
    (顺式)1-(3,5-二(三氟甲基)苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
    (顺式)1-(3,4-二氯苯基)-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)脲
    (顺式)1-苄基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-α]异吲哚]-3-基)脲
    (顺式)1-苯乙基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)脲
    (反式)1-(3-三氟甲基苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
    (反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
    (反式)1-(4-氯苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)硫脲
    (反式)4-氯-氮-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)苯磺酰胺
    (反式)4-甲基-N-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基氨基甲酰基)苯磺酰胺
    (反式)1-(3-氟苯基)-3-((1r,3r)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
    (顺式)1-甲基-3-((1s,3s)-螺[环丁烷1,5’-咪唑并[5,1-a]异吲哚]-3-基)磺酰脲
    (顺式)2-氰基-1-甲基-3-((1s,3s)-螺[环丁烷-1,5’-咪唑[5,1-a]异吲哚]-3-基)胍
    (顺/反)1-(螺[环己基-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(3-(三氟甲基)苯基)硫脲
    (顺/反)1-苯基-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
    (顺/反)1-(4-氯苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)硫脲
    (顺/反)1-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)-3-(4-(三氟甲基)苯基)硫脲
    (顺/反)1-(4-氯-3-(三氟甲基)苯基)-3-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)脲
    (顺/反)N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)萘基-2-磺酰胺
    (顺/反)3-溴-N-(螺[环己烷-1,5’-咪唑并[5,1-a]异吲哚]-4-基)苯甲酰胺
    (顺/反)4-(1-苄基-1H-1,2,3-三唑-4-基)螺[环己烷1,5’-咪唑并[5,1-a]异吲哚]-4-醇
    (顺式)(1s,3s)-螺[环丁烷-1,5’-咪唑并[5,1-a]异吲哚]-3-胺
    (顺式)(1r,3r)-3-(1-苯基-1H-1,2,3-三唑-4-基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-醇
    1'-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
    (顺式)(1s,3s)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
    (顺式)(1r,3r)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-胺
    (顺/反)-N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-胺
    (顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异 吲哚]-4-基)苯甲酰胺
    (3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)(螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-基)甲酮
    (反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯甲酰胺
    (顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)脲
    (顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
    N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-氨甲酰
    (顺/反)-3-氟-4-(1-甲基-1H-吡唑-4-基)-N-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)苯磺酰胺
    (反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)脲
    (顺/反)-1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-(螺[环己烷-1,5'-咪唑并[5,1-a]异吲哚]-4-基)硫脲
    N-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]-1'-硫代酰胺
    (反式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
    (顺式)1-(3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)-3-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)硫脲
    1'-((3-氟-4-(1-甲基-1H-吡唑-4-基)苯基)磺酰)螺[咪唑并[5,1-a]异吲哚-5,4'-哌啶]
    (顺式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1s,3s)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺
    (反式)3-氟-4-(1-甲基-1H-吡唑-4-基)-N-((1r,3r)-螺[环丁烷-1,5'-咪唑并[5,1-a]异吲哚]-3-基)苯磺酰胺。
    Figure PCTCN2017108941-appb-100023
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100024
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100025
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100026
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100027
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100028
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100029
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100030
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100031
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100032
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100033
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
    Figure PCTCN2017108941-appb-100034
    R=H,OH,NH2,烷基,芳基,杂芳基,O-烷基,O-芳基,O-杂芳基,NH-烷基,NH-芳基,NH-杂芳基,N(烷基)2
  9. 一种药物组合物,所述药物组合物包括:
    (1)如权利要求1所述的式(I)化合物、或其药学上可接受的盐、其立体异构体或其互变异构体、或其前药;
    (2)药学上可接受的载体和/或抗肿瘤药物。
  10. 一种如权利要求1所述的式(I)化合物或其药学上可接受的盐、其立体异构体、或其互变异构体、或其前药的用途,用于:
    (i)制备IDO/TDO抑制剂;
    (ii)制备预防和/或治疗IDO和TDO介导的疾病的药物。
PCT/CN2017/108941 2016-11-01 2017-11-01 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂 WO2018082567A1 (zh)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CN201780064706.6A CN109890826A (zh) 2016-11-01 2017-11-01 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
CN201610937361.8 2016-11-01
CN201610937361.8A CN108017643A (zh) 2016-11-01 2016-11-01 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂

Publications (1)

Publication Number Publication Date
WO2018082567A1 true WO2018082567A1 (zh) 2018-05-11

Family

ID=62070572

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/CN2017/108941 WO2018082567A1 (zh) 2016-11-01 2017-11-01 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂

Country Status (2)

Country Link
CN (2) CN108017643A (zh)
WO (1) WO2018082567A1 (zh)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020092183A1 (en) 2018-11-01 2020-05-07 Merck Sharp & Dohme Corp. Novel substituted pyrazole compounds as indoleamine 2,3-dioxygenase inhibitors

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN110628736B (zh) * 2019-10-09 2021-05-11 扬州大学 色氨酸2,3双加氧酶突变体及其制备方法和应用

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3835138A (en) * 1972-10-13 1974-09-10 Dow Chemical Co Bronchodilator 5,6-dihydro-tetrazolo(1,5-c) quinazolines

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2008046084A2 (en) * 2006-10-12 2008-04-17 Xenon Pharmaceuticals Inc. Spiroheterocyclic compounds and their uses as therapeutic agents

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3835138A (en) * 1972-10-13 1974-09-10 Dow Chemical Co Bronchodilator 5,6-dihydro-tetrazolo(1,5-c) quinazolines

Non-Patent Citations (2)

* Cited by examiner, † Cited by third party
Title
BOWIE, R. A. ET AL.: "Ring transformations involving chloroheterocycles.III. Reaction of 4-chloroquinazolines with hydrazines", JOURNAL OF THE CHEMICAL SOCIETY, PERKIN TRANSACTIONS 1: ORGANIC AND BIO-OR- GANIC CHEMISTRY, 31 December 1972 (1972-12-31), pages 1842 - 1848, XP055606012 *
MOUSTAFA, H. M.: "Synthesis and reactions of new fused heterocycles derived from 3-(o-aminophenyl)-4-amino-5-mercapto-1, 2, 4-triazole", SYNTHETIC COMMUNICATIONS, vol. 31, no. 1, 31 December 2001 (2001-12-31), pages 97 - 109, XP055606019 *

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2020092183A1 (en) 2018-11-01 2020-05-07 Merck Sharp & Dohme Corp. Novel substituted pyrazole compounds as indoleamine 2,3-dioxygenase inhibitors
EP3873464A4 (en) * 2018-11-01 2022-06-08 Merck Sharp & Dohme Corp. NOVEL SUBSTITUTED PYRAZOLE COMPOUNDS AS INDOLAMINE-2,3-DIOXYGENASE INHIBITORS

Also Published As

Publication number Publication date
CN109890826A (zh) 2019-06-14
CN108017643A (zh) 2018-05-11

Similar Documents

Publication Publication Date Title
JP7094456B2 (ja) Cdk2阻害剤
US20230113085A1 (en) Novel small molecule inhibitors of tead transcription factors
AU2015237788B2 (en) ROR-gamma modulators and uses thereof
CA3069720A1 (en) Carboxamides as modulators of sodium channels
WO2016041489A1 (zh) 一种吲哚胺-2,3-双加氧酶抑制剂及其制备方法
CA2716320C (en) Selective androgen receptor modulators
US7550481B2 (en) Phenylamide and pyridylamide beta-secretase inhibitors for the treatment of Alzheimer&#39;s disease
US7449599B2 (en) Phenyl carboxamide compounds useful as beta-secretase inhibitors for the treatment of alzheimer&#39;s disease
CN110267962B (zh) Magl抑制剂
EP1689713B1 (en) Benzylether and benzylamino beta-secretase inhibitors for the treatment of alzheimer s disease
US11851428B2 (en) Activator of TREK (TWIK RElated K+channels) channels
TW201734001A (zh) ROR-γ調節劑
TW202229285A (zh) Kras g12d抑制劑
WO2017129139A1 (zh) 一种作为吲哚胺-2,3-双加氧酶抑制剂的氨基磺酸脂及其制备方法和用途
EP1809275A1 (en) Chemical compounds
WO2017152857A1 (zh) 一种含氮烷基化和芳基化亚砜亚胺的吲哚胺-2,3-双加氧酶抑制剂
JP2008520670A (ja) アルツハイマー病の治療のための大環状第三アミンβ−セクレターゼ阻害剤
TW202227397A (zh) 雙環的-雜環衍生物及相關用途
CA3170664A1 (en) P2x3 modulators
TWI582080B (zh) 礦皮質素受體拮抗劑
ES2471919T3 (es) Derivados de ácido fenilac�tico como moduladores de inflamación
WO2018082567A1 (zh) 一种含有氮杂环螺旋结构的高效ido/tdo双抑制剂
TW201821404A (zh) 作為ROR γ調節劑之三環碸
WO2017008681A1 (zh) 酰胺类衍生物、其制备方法及其在医药上的用途
JP2001525398A (ja) 選択的β3アドレナリン作動性アゴニスト

Legal Events

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

Ref document number: 17866430

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 17866430

Country of ref document: EP

Kind code of ref document: A1