CN116761803A - Modulators of integrated stress response pathways - Google Patents

Modulators of integrated stress response pathways Download PDF

Info

Publication number
CN116761803A
CN116761803A CN202180086795.0A CN202180086795A CN116761803A CN 116761803 A CN116761803 A CN 116761803A CN 202180086795 A CN202180086795 A CN 202180086795A CN 116761803 A CN116761803 A CN 116761803A
Authority
CN
China
Prior art keywords
trifluoromethoxy
oxadiazol
ethoxy
piperidine
carboxylic acid
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
CN202180086795.0A
Other languages
Chinese (zh)
Inventor
H·V·阿顿
C·J·布朗
S·康佛斯-雷尼耶
C·F·帕默
M·萨巴
D·S·沃尔特
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Evertec International Co ltd
Original Assignee
Evertec International Co ltd
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Evertec International Co ltd filed Critical Evertec International Co ltd
Priority claimed from PCT/EP2021/079209 external-priority patent/WO2022084447A1/en
Publication of CN116761803A publication Critical patent/CN116761803A/en
Pending legal-status Critical Current

Links

Landscapes

  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

The present invention relates to compounds of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, wherein R 1 ,R 2 ,R 3 ,R 4a ,R 4b ,R 4c ,R 4d ,R 4f ,X 1 ,X 2 Having the meaning as indicated in the description and in the claims. The invention also relates to pharmaceutical compositions comprising said compounds, their use as medicaments and in methods of treatment or prevention of one or more diseases or disorders associated with an integrated stress response

Description

Modulators of integrated stress response pathways
The present invention relates to compounds of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof
Wherein R is 1 ,R 2 ,R 3 ,R 4a ,R 4b ,R 4c ,R 4d ,R 4f ,X 1 ,X 2 Having the meaning as indicated in the description and in the claims. The invention further relates to pharmaceutical compositions comprising said compounds, their use as medicaments and in methods for the treatment or prevention of one or more diseases or disorders associated with an integrated stress response.
The Integrated Stress Response (ISR) is a cellular stress response common to all eukaryotes (1). Dysregulation of ISR signaling has important pathological consequences, especially those associated with inflammation, viral infection, diabetes, cancer and neurodegenerative diseases.
ISR is a common feature of different types of cellular stress, resulting in phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eif2α) on serine 51, thereby inhibiting normal protein synthesis and expression of stress response genes (2). In mammalian cells, the phosphorylation is performed by a family of four eif2α kinases, namely: PKR-like ER kinase (PERK), double-stranded RNA-dependent Protein Kinase (PKR), heme regulated eif2α kinase (HRI), and general regulatory repressor kinase 2 (GCN 2), each respond to different environmental and physiological stresses (3).
eif2α forms an eIF2 complex with eif2β and eif2γ, which are key participants in normal mRNA translation initiation (4). eIF2 complex binds GTP and Met-tRNA i Thereby forming a ternary complex (eIF 2-GTP-Met-tRNA i ) Which is recruited by ribosomes for translation initiation (5, 6).
eIF2B is an isodecamer complex consisting of 5 subunits (α, β, γ, δ, epsilon) that form GEF-active decamers in duplicate (7).
In response to ISR activation, phosphorylated eif2α inhibits eIF 2B-mediated exchange of GDP with GTP, resulting in reduced ternary complex formation and thus inhibits translation of normal mRNA characterized by ribosome binding to the 5' aug start codon (8). Under these conditions of reduced ternary complex abundance, translation of several specific mRNAs, including the mRNA encoding the transcription factor ATF4, is activated by a mechanism involving translational changes in the upstream ORF (uORF) (7, 9, 10). These mrnas typically comprise one or more uofs that typically function in unstressed cells to limit the flow of ribosomes to the primary encoding ORF. For example, under normal conditions, the uORF in the 5' utr of ATF occupies ribosomes and prevents translation of the coding sequence of ATF 4. However, under stress conditions, i.e., conditions where ternary complex formation is reduced, the likelihood of ribosomal scanning past these upstream ORFs and translation begins at the ATF4 encoding ORF increases. ATF4 and other stress factors expressed in this manner then control the expression of a range of other stress genes. The acute phase consists in the expression of proteins intended to resume steady state, while the chronic phase leads to the expression of pro-apoptotic factors (1,11,12,13).
Upregulation of markers of ISR signaling has been demonstrated in a variety of disorders, including cancer and neurodegenerative diseases. ER stress mediated translation increases tolerance to hypoxic conditions and promotes tumor growth in cancer (14, 15, 16), and it has been demonstrated that deletion of PERK by gene targeting can slow down PE from transformationRK -/- Growth of tumors derived from mouse embryonic fibroblasts (14, 17). Furthermore, a recent report has provided the following conceptual evidence: activators of eIF2B are effective in treating an aggressive metastatic prostate cancer using a patient-derived xenograft model in mice (28). In summary, the prevention of cytoprotective ISR signaling may represent an effective antiproliferative strategy for the treatment of at least some forms of cancer.
Furthermore, modulation of ISR signaling may prove to be effective in maintaining synaptic function and reducing neuronal decline, as well as in neurodegenerative diseases characterized by activation of misfolded and Unfolded Protein Responses (UPR), such as Amyotrophic Lateral Sclerosis (ALS), frontotemporal dementia (FTD), alzheimer's Disease (AD), parkinson's Disease (PD), and Jakob Creutzfeld (prion) disease (18, 19, 20). For prion diseases (an example of an existing neurodegenerative disease), pharmacology and genetic inhibition of ISR signaling have been shown to normalize protein translation levels, rescue synaptic function, and prevent neuronal loss (21). Specifically, decreasing levels of phosphorylated eif2α by controlling overexpression of phosphatases that phosphorylate levels of eif2α increases survival in prion-infected mice, while sustained eif2α phosphorylation decreases survival (22).
Furthermore, direct evidence of the importance of control protein expression levels for proper brain function exists in the form of rare genetic diseases affecting the function of eIF2 and eIF 2B. Mutations in eif2γ that disrupt the complex integrity of eIF2 and thus lead to reduced levels of normal protein expression are associated with dysnoesia syndrome (ID) (23). Partial loss of function mutations in subunits of eIF2B have been demonstrated to be responsible for rare white matter dystrophy white matter ablative disease (VWMD) (24, 25). In particular, stabilization of eIF2B partial loss of function by small molecules associated with ISRIB in VWMD mouse models has been shown to reduce ISR markers and improve function and pathological endpoints (26, 27).
Modulators of the eif2α pathway are described in WO 2014/144952 A2. WO 2017/193030 A1, WO 2017/193034 A1, WO 2017/193041 A1 and WO 2017/193063 A1 describe modulators that integrate the stress pathway. WO 2017/212423 A1, WO 2017/212425 A1, WO 2018/225093 A1, WO 2019/008506 A1 and WO 2019/008507 A1 describe inhibitors of the ATF4 pathway. WO 2019/032743 A1, WO 2019/046779 A1, WO 2020/167994 A1, WO 2020/168411 A1 and WO 2020/181247 A1 relate to eukaryotic initiation factor 2B modulators. In WO 2020/77217 A1, compounds, compositions and methods useful for modulating Integrated Stress Response (ISR) and for treating related diseases, disorders and conditions are described.
Other documents describing modulators of the integrated stress pathway are WO 2019/090069 A1, WO 2019/090074 A1, WO 2019/090076 A1, WO 2019/090078 A1, WO 2019/090081 A1, WO 2019/090085 A1, WO 2019/090088 A1, WO 2019/090090 A1, WO 2020/223536 A1, WO 2020/223538 A1, WO 2020/252207 A1, european patent applications 20203309.8 and 20203312.2, WO 2021/180774 A1, WO 2021/151865 A1, WO 2020/216764 A1 and WO 2020/216766 A1.
Modulators of eukaryotic initiation factors are described in WO 2019/183589 A1. WO 2019/118785 A2, WO 2019/236710 A1, WO 2020/176728 A1 and WO 2020/252205 A1 describe inhibitors that integrate the stress response pathway. Heteroaryl derivatives as ATF4 inhibitors are described in WO 2019/193540 A1. Bicyclic aromatic ring derivatives as ATF4 inhibitors are described in WO 2019/193541 A1. WO 2020/031107 A1 and WO 2020/012339 A1 describe inhibitors of the ATF4 pathway.
However, there remains a need for new compounds with good pharmacokinetic properties that can be used as modulators of the integrated stress response pathway.
It is therefore an object of the present invention to provide a new class of compounds as modulators of the integrated stress pathway which may be effective in the treatment of diseases associated with the integrated stress pathway and which may exhibit improved pharmaceutically relevant properties including activity, solubility, selectivity, ADMET performance and/or reduced side effects.
Accordingly, the present invention provides a compound of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof
Wherein the method comprises the steps of
X 1 Is N (R) 4 ) And X is 2 Is CH (R) 4e ) The method comprises the steps of carrying out a first treatment on the surface of the Or X 1 And X 2 Is O.
R 1 Is H or C 1-4 Alkyl, preferably H, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
R 2 is phenyl, naphthyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl or 7-to 12-membered heterobicyclo, wherein R 2 Optionally by one or more of the same or different R 5 With the proviso that if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atom attached to the carbon atom of the amide group is not substituted with H or F;
R 5 independently halogen, CN, C (O) OR 6 、OR 6 、C(O)R 6 、C(O)N(R 6 R 6a )、S(O) 2 N(R 6 R 6a )、S(O)N(R 6 R 6a )、S(O) 2 R 6 、S(O)R 6 、N(R 6 )S(O) 2 N(R 6a R 6b )、SR 6 、N(R 6 R 6a )、NO 2 、OC(O)R 6 、N(R 6 )C(O)R 6a 、N(R 6 )S(O) 2 R 6a 、N(R 6 )S(O)R 6a 、N(R 6 )C(O)OR 6a 、N(R 6 )C(O)N(R 6a R 6b )、OC(O)N(R 6 R 6a ) Oxo (=o), wherein the ring is at least partially saturated, C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 7 Substitution;
R 6 、R 6a 、R 6b independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 7 is halogen, CN, C (O) OR 8 、OR 8 、C(O)R 8 、C(O)N(R 8 R 8a )、S(O) 2 N(R 8 R 8a )、S(O)N(R 8 R 8a )、S(O) 2 R 8 、S(O)R 8 、N(R 8 )S(O) 2 N(R 8a R 8b )、SR 8 、N(R 8 R 8a )、NO 2 、OC(O)R 8 、N(R 8 )C(O)R 8a 、N(R 8 )SO 2 R 8a 、N(R 8 )S(O)R 8a 、N(R 8 )C(O)N(R 8a R 8b )、N(R 8 )C(O)OR 8a Or OC (O) N (R) 8 R 8a );
R 8 、R 8a 、R 8b Independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 3 is OR (OR) 9 、SR 9a 、N(R 9 R 9a )、A 1 、C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 10 Substitution;
R 9 、R 9a independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl and A 1 Wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 11 Substitution;
R 10 is halogen OR 12 CN or A 1
R 11 Is halogen, CN, OR 12 、OA 1 Or A 1
R 12 Is H or C 1-4 Alkyl, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
A 1 is phenyl, C 3-7 Cycloalkyl, C 4-12 Bicycloalkyl or 3-7 membered heterocyclyl, wherein A 1 Optionally by one or more of the same or different R 13 Substitution;
R 13 is R 14 、OH、OR 14 Halogen or CN; and
R 14 is cyclopropyl, C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl, wherein R is 14 Optionally by one or more of the same or different R 15 Substitution; or alternatively
Two R 13 Are linked to form, together with the atoms to which they are attached, a ring A 2
R 15 Is halogen, CN OR OR 16
R 16 Is H or C 1-4 Alkyl, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
A 2 is phenyl, C 3-7 Cycloalkyl or 3-7 membered heterocyclyl, wherein A 2 Optionally by one or more of the same or different R 17 Substitution;
R 17 is C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 4 is H, C (O) OC 1-4 Alkyl or C 1-4 Alkyl, wherein C (O) OC 1-4 Alkyl and C 1-4 The alkyl group is optionally substituted with one or more substituents selected from the group consisting of: halogen, OH and OC 1-3 Alkyl, wherein the substituents are the same or different;
R 4a 、R 4b 、R 4c 、R 4f independently selected from H, halogen and C 1-4 An alkyl group; and
R 4d 、R 4e independently selected from H, OH, OC 1-4 Alkyl, halogen and C 1-4 An alkyl group;
or R is 4d And R is 4e One and R 4 Forming a methylene or ethylene group;
or R is 4 And R is R 4c Forming an ethylene group;
or R is 4b And R is R 4d Covalent single bonds are formed.
Surprisingly, the compounds according to the disclosed embodiments of the present invention possess advantageous physicochemical properties and/or selectivities, which in combination help achieve beneficial therapeutic efficacy while limiting unexpected consequences.
Where a variable or substituent may be selected from a group of different variants and such variable or substituent occurs more than once, the individual variants may be the same or different.
Within the meaning of the invention, the terms are used as follows:
the term "optionally substituted" refers to unsubstituted or substituted. Generally, but not limited to, "one or more substituents" refers to one, two, or three substituents, preferably one or two substituents, and more preferably one substituent. In general, these substituents may be the same or different. The term "one or more substituents" also means, for example, one, two, three, four or five, preferably, for example, one, two, three or four.
"alkyl" refers to a straight or branched hydrocarbon chain. Each hydrogen of the alkyl carbon may be replaced with a further specified substituent.
"alkenyl" refers to a straight or branched hydrocarbon chain containing at least one carbon-carbon double bond. Each hydrogen of the alkenyl carbon may be replaced with a further specified substituent.
"alkynyl" refers to a straight or branched hydrocarbon chain containing at least one carbon-carbon triple bond. Each hydrogen of the alkynyl carbon may be replaced with a further specified substituent.
“C 1-4 Alkyl "means an alkyl chain having 1 to 4 carbon atomsIf present at the end of the molecule, for example: methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, or when the two moieties of the molecule are linked by an alkyl group, e.g. -CH 2 -、-CH 2 -CH 2 -、-CH(CH 3 )-、-CH 2 -CH 2 -CH 2 -、-CH(C 2 H 5 )-、-C(CH 3 ) 2 -。C 1-4 Each hydrogen of the alkyl carbon may be replaced with a further specified substituent. The term "C" is defined accordingly 1-3 An alkyl group.
“C 1-6 Alkyl "refers to an alkyl chain having 1 to 6 carbon atoms, if present at the end of a molecule, such as: c (C) 1-4 Alkyl, methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n-pentyl, n-hexyl, or, when two parts of the molecule are linked by an alkyl group, e.g. -CH 2 -、-CH 2 -CH 2 -、-CH(CH 3 )-、-CH 2 -CH 2 -CH 2 -、-CH(C 2 H 5 )-、-C(CH 3 ) 2 -。C 1-6 Each hydrogen of the alkyl carbon may be replaced with a further specified substituent.
“C 2-6 Alkenyl "refers to alkenyl chains having 2 to 6 carbon atoms, if present at the end of a molecule, such as: -ch=ch 2 、-CH=CH-CH 3 、-CH 2 -CH=CH 2 、-CH=CH-CH 2 -CH 3 、-CH=CH-CH=CH 2 Alternatively, when two parts of the molecule are linked by an alkenyl group, for example, -ch=ch-. C (C) 2-6 Each hydrogen of the alkenyl carbon may be replaced with a further specified substituent.
“C 2-6 Alkynyl "refers to an alkynyl chain having 2 to 6 carbon atoms, if present at the end of a molecule, for example: -C.ident.CH, -CH 2 -C≡CH、CH 2 -CH 2 -C≡CH、CH 2 -C≡C-CH 3 Alternatively, when two parts of the molecule are linked by an alkynyl group, e.g. -c≡c-. C (C) 2-6 Each hydrogen of the alkynyl carbon may be replaced with a further specified substituent.
“C 3-7 Cycloalkyl "or" C 3-7 Cycloalkyl ring "means a cycloalkyl chain having 3 to 7 carbon atoms, such as cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclohexenyl, cycloheptyl. Preferably, cycloalkyl means cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl or cycloheptyl. Each hydrogen of the cycloalkyl carbon may be replaced by a substituent as further specified herein. The term "C" is defined accordingly 3-5 Cycloalkyl "or" C 3-5 Cycloalkyl rings).
“C 5 Cycloalkyl "refers to a divalent cycloalkyl group having 5 carbon atoms, i.e., a divalent cyclopentyl ring.
“C 5 The term "cycloalkenylene" refers to a divalent cycloalkenylene group, i.e., a divalent cyclopentene or cyclopentadiene.
“C 4-12 Bicycloalkyl "or" C 4-12 By a bicycloalkyl ring "is meant a bicyclofused, bridged or spiro alkyl chain having 4 to 12 carbon atoms, e.g. hexahydroindane, octahydropentalene, bicyclo [2.2.1 ]]Heptane or spiro (3.2) hexane. Each hydrogen of the bicycloalkyl carbon may be replaced by a substituent as further specified herein.
"halogen" means fluorine, chlorine, bromine or iodine. It is generally preferred that the halogen is fluorine or chlorine.
"3-7 membered heterocyclyl" or "3-7 membered heterocycle" refers to a ring having 3, 4, 5, 6 or 7 ring atoms which may contain up to a maximum number of double bonds (aromatic rings or fully saturated, partially saturated or unsaturated non-aromatic rings) wherein at least one ring atom up to 4 ring atoms are selected from sulfur (including-S (O) -, -S (O)) 2 (-), oxygen and nitrogen (including = N (O) -), and wherein the ring is attached to the rest of the molecule via a carbon or nitrogen atom. Examples of 3-to 7-membered heterocycles are aziridine, azetidine, oxetane, thietane, furan, thiophene, pyrrole, pyrroline, imidazole, imidazoline, pyrazole, pyrazoline, oxazole, oxazoline, isoxazole, isoxazoline, thiazole, thiazoline, isothiazole, isothiazoline, thiadiazole, tetrahydrofuran, tetrahydrothiophene, pyrrolidine, imidazolidine, pyrazolidine, oxazolidine, isoxazole An alkane, thiazolidine, isothiazolidine, thiadiazolidine, sulfolane, pyran, dihydropyran, tetrahydropyran, imidazolidine, pyridine, pyridazine, pyrazine, pyrimidine, piperazine, piperidine, morpholine, tetrazole, triazole, triazolidine, tetrazolidine, diazepane, azepine or homopiperazine. The term "5-6 membered heterocyclyl" or "5-6 membered heterocycle" is correspondingly defined and includes 5-6 membered aromatic heterocyclyl or heterocycle. The term "5 membered heterocyclyl" or "5 membered heterocycle" is correspondingly defined and includes 5 membered aromatic heterocyclyl or heterocycle.
The term "5-membered heterocyclylene containing a nitrogen ring atom" refers to a divalent 5-membered heterocyclic ring in which at least one of the five ring atoms is a nitrogen atom and in which the ring is attached to the remainder of the molecule via a carbon or nitrogen atom.
"saturated 4-7 membered heterocyclyl" or "saturated 4-7 membered heterocycle" refers to a fully saturated "4-7 membered heterocyclyl" or "4-7 membered heterocycle".
"4-7 membered at least partially saturated heterocyclyl" or "4-7 membered at least partially saturated heterocycle" refers to an at least partially saturated "4-7 membered heterocyclyl" or "4-7 membered heterocycle".
"5-6 membered aromatic heterocyclic group" or "5-6 membered aromatic heterocyclic ring" means a heterocyclic ring derived from cyclopentadienyl or benzene in which at least one carbon atom is selected from sulfur (including-S (O) -, -S (O)) 2 (-), oxygen and nitrogen (including = N (O) -). Examples of such heterocycles are furan, thiophene, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, thiadiazole, triazole, tetrazole, pyridine, pyrimidine, pyridazine, pyrazine, triazine.
"5-membered aromatic heterocyclic group" or "5-membered aromatic heterocyclic ring" refers to a heterocyclic ring derived from a cyclopentadienyl group in which at least one carbon atom is selected from sulfur (including-S (O) -, -S (O)) 2 (-), oxygen and nitrogen (including = N (O) -). Examples of such heterocycles are furan, thiophene, pyrrole, imidazole, pyrazole, oxazole, isoxazole, thiazole, isothiazole, thiadiazole, triazole, tetrazole.
"6-membered aromatic heterocyclic group" or "6-membered aromatic heterocyclic ring" refers to a heterocyclic ring derived from benzene, in which at least one carbon atomThe sub-quilt is selected from sulfur (including-S (O) -, -S (O) 2 (-), oxygen and nitrogen (including = N (O) -). Examples of such heterocycles are pyridine, pyrimidine, pyridazine, pyrazine, triazine.
"7-12 membered heterobicyclic group" or "7-12 membered heterobicyclic group" refers to a heterocyclic ring system having two rings of 7-12 ring atoms, wherein the two rings share at least one ring atom, and which may contain up to a maximum number of double bonds (aromatic rings or fully saturated, partially saturated or unsaturated non-aromatic rings), wherein at least one ring atom up to 6 ring atoms are selected from sulfur (including-S (O) -, -S (O)) 2 (-), oxygen and nitrogen (including = N (O) -), and wherein the ring is attached to the rest of the molecule via a carbon or nitrogen atom. Examples of 7-12 membered heterobicyclic rings are indole, indoline, benzofuran, benzothiophene, benzoxazole, benzisoxazole, benzothiazole, benzisothiazole, benzimidazole, benzimidazoline, quinoline, quinazoline, dihydroquinazoline, quinoline, dihydroquinoline, tetrahydroquinoline, decahydroquinoline, isoquinoline, decahydroisoquinoline, tetrahydroisoquinoline, dihydroisoquinoline, benzoazepine, purine or pteridine. The term 7-12 membered heterobicyclic also includes spiro structures of two rings such as 6-oxa-2-azaspiro [3,4 ]]Octane, 2-oxa-6-azaspiro [3.3 ]]Heptan-6-yl or 2, 6-diazaspiro [3.3 ]]Heptane-6-yl, or bridged heterocycles such as 8-aza-bicyclo [3.2.1]Octane or 2, 5-diazabicyclo [2.2.2]Octane-2-yl or 3, 8-diazabicyclo [3.2.1]Octane.
"saturated 7-12 membered heterobicyclic group" or "saturated 7-12 membered heterobicyclic group" refers to a fully saturated "7-12 membered heterobicyclic group" or "7-12 membered heterobicyclic group".
"7-12 membered at least partially saturated heterobicyclic group" or "7-12 membered at least partially saturated heterobicyclic group" means an at least partially saturated "7-12 membered heterobicyclic group" or "7-12 membered heterobicyclic group".
"9-11 membered aromatic heterobicyclo group" or "9-11 membered aromatic heterobicyclo" refers to a heterocyclic ring system of two rings, wherein at least one ring is aromatic, and wherein the heterocyclic ring system has 9-11 ring atoms, wherein the two rings share two ring atoms, and which may contain up to a maximum numberDouble bonds of interest (wholly or partly aromatic) in which at least one ring atom up to 6 ring atoms are selected from sulfur (including-S (O) -, -S (O)) 2 (-), oxygen and nitrogen (including = N (O) -), and wherein the ring is attached to the rest of the molecule via a carbon or nitrogen atom. Examples of 9-11 membered aromatic heterobicyclic rings are indole, indoline, benzofuran, benzothiophene, benzoxazole, benzisoxazole, benzothiazole, benzisothiazole, benzimidazole, benzimidazoline, quinoline, quinazoline, dihydroquinazoline, dihydroquinoline, tetrahydroquinoline, isoquinoline, tetrahydroisoquinoline, dihydroisoquinoline, benzoazepine, purine or pteridine. The term "9-10 membered aromatic heterobicyclo" or "9-10 membered aromatic heterobicyclo" is correspondingly defined.
Preferred compounds of formula (I) are those in which one or more residues contained therein have the meanings given above or below, wherein all combinations of preferred substituent definitions are the subject of the present invention. With respect to all preferred compounds of formula (I), the invention also includes all tautomeric and stereoisomeric forms and mixtures thereof in all ratios, as well as pharmaceutically acceptable salts thereof.
In a preferred embodiment of the invention, the substituents mentioned below independently have the following meanings. Thus, one or more of these substituents may have the preferred or more preferred meanings given below.
In a preferred embodiment of the invention, for a compound of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, X 1 Is N (R) 4 ),X 2 Is CH (R) 4e ) Obtaining the formula (I-1)
In another preferred embodiment of the invention, for a compound of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, X 1 And X 2 For O, give the formula (I-2)
Preferably, R 4 H, CH of a shape of H, CH 3 、CH 2 CH 3 Or CH (CH) 2 CH 2 OCH 3 The method comprises the steps of carrying out a first treatment on the surface of the More preferably H or CH 3 The method comprises the steps of carrying out a first treatment on the surface of the Even more preferably H.
Preferably, R 4a 、R 4b 、R 4c 、R 4f Independently selected from H, halogen and C 1-4 Alkyl and R 4d 、R 4e Independently selected from H, OH, OC 1-4 Alkyl, halogen and C 1-4 An alkyl group; more preferably R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e Independently selected from H, F and CH 3 The method comprises the steps of carrying out a first treatment on the surface of the Even more preferably, R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e Is H.
Preferably, R 1 Is H or CH 3 The method comprises the steps of carrying out a first treatment on the surface of the More preferably H.
Preferably, R in formula (I-1) 1 、R 4 、R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e For H, a compound of formula (Ia-1)
Also preferred is R in formula (I-2) 1 、R 4a 、R 4b 、R 4c 、R 4f 、R 4d H to give the formula (Ia-2)
R 2 Is phenyl, naphthyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl or 7-to 12-membered heterobicyclo, wherein R 2 Optionally by one or more of the same or different R 5 Substitution ofProvided that if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atom attached to the carbon atom of the amide group is not substituted with H or F.
Thus, if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atoms bound to the carbon atoms of the amide groups being unsubstituted by H or F, as in formula (I) of WO2020/216766A1 and PCT/EP2021/056023 for R 2a As defined. Preferably, R is 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Not all ring atoms of (a) are oxygen. More preferably, R 2 Not all ring atoms of (a) are oxygen.
Preferably, R 2 Is phenyl, pyridyl, thienyl, 1H-indolyl, quinolinyl, isoquinolinyl, quinazolinyl, pyrazolo [1,5-a ]]Pyridinyl, pyrrolo [1,2-a ]]Pyrazinyl, indolizinyl, chromeneyl, benzofuranyl or 2H-1, 3-benzodioxolyl; more preferably phenyl, pyridin-2-yl, pyridin-3-yl, thiophen-2-yl, 1H-indol-2-yl, quinolin-3-yl, quinolin-6-yl, quinolin-7-yl, isoquinolin-3-yl, quinazolin-2-yl, pyrazolo [1,5-a ]Pyridin-2-yl, pyrrolo [1,2-a ]]Pyrazin-3-yl, indolizin-2-yl, chromen-3-yl, benzofuran-2-yl or 2H-1, 3-benzodioxol-5-yl; and wherein R is 2 Optionally by one or more of the same or different R 5 With the proviso that if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atom attached to the carbon atom of the amide group is not substituted with H or F. More preferably, R 2 Is quinolinyl, especially quinolin-2-yl, quinolin-3-yl, quinolin-6-yl, quinolin-7-yl, wherein R 2 Optionally by one or more of the same or different R 5 And (3) substitution.
Preferably, R 2 By one, two or three identical or different R 5 And (3) substitution.
PreferablyGround, R 5 Is F, cl, CH 3 、CF 3 、OCF 3 Or OCH (optical wavelength) 2 CF 3
Preferably, R 3 Is OR (OR) 9 And R is 9 Is C 1-6 Alkyl or C 2-6 Alkenyl group, wherein C 1-6 Alkyl and C 2-6 Alkenyl groups being substituted by one or more identical or different R' s 11 And (3) substitution.
Preferably, R 3 Is OR (OR) 9 And R is 9 Is C 1-6 Alkyl, preferably ethyl, wherein C 1-6 Alkyl is substituted with one R 11 And (3) substitution.
Preferably, R 3 Is OCH 2 CH 2 OCF 3
Preferably, R 3 Is OR (OR) 9 ,R 9 Is C 1-6 Alkyl or C 2-6 Alkenyl, preferably but-2-enyl, wherein C 1-6 Alkyl and C 2-6 Alkenyl groups are each substituted with three F; more preferably R 3 Is OCH 2 CH=CHCF 3
Preferably, R 3 Is A 1 Preferably phenyl or cyclobutyl, wherein A is 1 Optionally by one or more of the same or different R 13 And (3) substitution.
Preferably, A 1 Is one or two, preferably one R 13 And (3) substitution.
Preferably, R 13 Is CH 3 、CHF 2 、CF 3 、CH 2 CF 3 、OCHF 2 、OCH 2 CF 3 、OCF 3 、OCH 3 F or Cl, more preferably Cl or OCF 3
Compounds of formula (I) in which some or all of the abovementioned radicals have the preferred or more preferred meanings are also an object of the invention.
For a preferred specific compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, R in formula (I) is selected 1 、R 2 、R 3 、R 4a 、R 4b 、R 4c 、R 4d 、R 4f 、X 1 、X 2 Obtaining
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethyl) quinoline-2-amid-yl ] piperidine-1-carboxylic acid tert-butyl ester;
n- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -6- (trifluoromethyl) quinoline-2-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2H-chromen-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] isoquinoline-3-carboxamide;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinazoline-2-carboxamide;
(2 r,5 s) -5- (6-chloroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
5-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -1-benzofuran-2-carboxamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-7-carboxamide;
7-chloro-6-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
5-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyrazolo [1,5-a ] pyridine-2-carboxamide;
6- (2, 2-trifluoroethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-3-carboxamide;
n- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2- (trifluoromethyl) quinoline-6-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyrrolo [1,2-a ] pyrazine-3-carboxamide;
6- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-3-carboxamide;
3, 4-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
7-chloro-8-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] indolizine-2-carboxamide;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] indolizine-2-carboxamide;
1-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -5- (trifluoromethyl) -1H-indole-2-carboxamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3, 5-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3, 4-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4, 5-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] thiophene-2-carboxamide;
4-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
(2 r,5 s) -5- [4- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
4- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
2, 2-difluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2H-1, 3-benzodioxol-5-carboxamide
4-chloro-3-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-chloro-3, 5-difluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -4- (trifluoromethyl) benzamide;
3-chloro-4- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -3- (trifluoromethyl) benzamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -4- (trifluoromethyl) benzamide;
4-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -3- (trifluoromethyl) benzamide;
4, 5-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-2-carboxamide;
5, 6-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-2-carboxamide;
4-chloro-3- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
(2 r,5 s) -5- [ [ 1-methyl-6- (trifluoromethyl) indole-2-carbonyl ] amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
1-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] -6- (trifluoromethyl) indole-2-carboxamide;
(2 r,5 s) -5- [ (3-chloro-4-methyl-benzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
3-chloro-4-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide;
(2 r,5 s) -2- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -5- [ (7-chloroquinoline-3-carbonyl) amino ] piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
(2 r,5 s) -5- [ (7-chloroquinoline-3-carbonyl) amino ] -2- [5- [ (E) -4, 4-trifluoro-but-2-yloxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- [5- [ (E) -4, 4-trifluoro-but-2-enoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 r,6 s) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
(2 r,5 s) -5- (7-chloroquinoxaline-3-amido) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 r,6 s) -6- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
(2 s,5 r) -5- [ (3, 4-dichlorobenzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
3, 4-dichloro-N- [ (3 r,6 s) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide;
(2 s,5 r) -5- (7-chloroquinoxaline-3-amido) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 s,5 r) -5- (7-chloroquinoxaline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-chloro-3- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (5, 6-dichloropyridine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4, 5-dichloropyridine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-chloro-3- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-chloro-4- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-fluoro-3- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-chloro-4- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-fluoro-4- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chloro-3, 5-difluorobenzamide) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chloro-3-methylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (2, 2-difluoro-2H-1, 3-benzodioxol-5-ylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-methylbenzamide) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4, 5-dimethylthiophene-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 4-dimethylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 5-dimethylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3-chlorobenzoylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 1-methyl-5- (trifluoromethyl) -1H-indole-2-amido ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (6-chloroindolizine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloroindolizine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-8-fluoroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chlorobenzoylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 4-dichlorobenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethoxy) pyridin-3-ylamino ] piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- { 7-chloropyrrolo [1,2-a ] pyrazine-3-amido } -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [2- (trifluoromethyl) quinoline-6-carboxamide ] piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [6- (2, 2-trifluoroethoxy) pyridin-3-ylamino ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- { 5-chloropyrazolo [1,5-a ] pyridine-2-amido } -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-6-fluoroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3-chloroquinoxaline-7-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (5-chloro-1-benzofuran-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (6-chloroquinazolin-2-ylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloroisoquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-2H-chromen-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2R, 5S) -5- (7-chloroquinoxaline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester or
7-chloro-N- [ trans-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -1, 3-dioxane-5-yl ] quinoline-3-carboxamide.
Where tautomerism of the compounds of formula (I) can occur (such as, for example, keto-enol tautomerism), each is included separately and as a mixture in any ratio, including various forms such as, for example, keto and enol forms. The same applies to stereoisomers, such as, for example, enantiomers, cis/trans isomers, conformational isomers and the like.
In particular, when enantiomeric or diastereoisomeric forms are given in the compounds according to formula (I), any mixture of each pure form alone and any ratio of at least two pure forms is encompassed by formula (I) and is the subject of the present invention.
One preferred compound is a compound of formula (I) having the relative configuration as shown in formula (Ib) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof
More preferably, the compound of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof has the configuration shown in formulas (Ib-1), (Ib-2), (Ib-3) and (Ib-4)
Isotopically-labeled compounds of formula (I) are also within the scope of the present invention. Methods for isotopic labeling are known in the art. Preferred isotopes are those of elements H, C, N, O and S. Solvates and hydrates of the compounds of formula (I) are also within the scope of the invention.
If desired, the isomers may be separated by methods well known in the art, for example, by liquid chromatography. The same applies to enantiomers by using, for example, chiral stationary phases. In addition, the enantiomers can be separated as follows: they are converted into diastereomers, i.e. coupled with enantiomerically pure auxiliary compounds, followed by separation of the resulting diastereomers and cleavage of the auxiliary residues. Alternatively, any enantiomer of a compound of formula (I) may be obtained from stereoselective synthesis using optically pure starting materials, reagents and/or catalysts.
In the case of compounds according to formula (I) containing one or more acidic or basic groups, the invention also comprises their corresponding pharmaceutically or toxicologically acceptable salts, in particular their pharmaceutically usable salts. Thus, according to the invention, compounds of the formula (I) containing acidic groups can be used, for example, as alkali metal salts, alkaline earth metal salts or as ammonium salts. More specific examples of such salts include sodium, potassium, calcium, magnesium salts or salts with ammonia or organic amines such as ethylamine, ethanolamine, triethanolamine or amino acids. Compounds of formula (I) containing one or more basic groups (i.e. groups which can be protonated) may be present and may be used according to the invention in the form of their addition salts with inorganic or organic acids. Examples of suitable acids include: hydrochloric acid, hydrobromic acid, phosphoric acid, sulfuric acid, nitric acid, methanesulfonic acid, p-toluenesulfonic acid, naphthalenedisulfonic acid, oxalic acid, acetic acid, tartaric acid, lactic acid, salicylic acid, benzoic acid, formic acid, propionic acid, pivalic acid, diethyl acetic acid, malonic acid, succinic acid, pimelic acid, fumaric acid, maleic acid, malic acid, sulfamic acid, phenylpropionic acid, gluconic acid, ascorbic acid, isonicotinic acid, citric acid, adipic acid, and other acids known to those skilled in the art. If the compounds of formula (I) contain both acidic and basic groups in the molecule, the invention includes, in addition to the salt forms mentioned, also internal salts or betaines (zwitterionic). The various salts according to formula (I) can be obtained by conventional methods known to the person skilled in the art, such as, for example, by contacting these with organic or inorganic acids or bases in solvents or dispersants, or by anion exchange or cation exchange with other salts. The invention also includes all salts of the compounds of formula (I) which, due to their low physiological compatibility, are not directly suitable for use in medicine, but which are useful, for example, as intermediates of chemical reactions or for the preparation of pharmaceutically acceptable salts.
As shown below, the compounds of the invention are believed to be useful in modulating the integrated stress response pathway.
The Integrated Stress Response (ISR) is a cellular stress response common to all eukaryotes (1). Dysregulation of ISR signaling has important pathological consequences, especially those associated with inflammation, viral infection, diabetes, cancer and neurodegenerative diseases.
ISR is a common feature of different types of cellular stress, resulting in phosphorylation of the α subunit of eukaryotic translation initiation factor 2 (eif2α) on serine 51, thereby inhibiting normal protein synthesis and expression of stress response genes (2). In mammalian cells, the phosphorylation is performed by a family of four eif2α kinases, namely: PKR-like ER kinase (PERK), double-stranded RNA-dependent Protein Kinase (PKR), heme regulated eif2α kinase (HRI), and general regulatory repressor kinase 2 (GCN 2), each respond to different environmental and physiological stresses (3).
eif2α forms an eIF2 complex with eif2β and eif2γ, which are key participants in normal mRNA translation initiation (4). eIF2 complex binds GTP and Met-tRNA i Thereby forming a ternary complex (eIF 2-GTP-Met-tRNA i ) Which is recruited by ribosomes for translation initiation (5, 6).
eIF2B is an isodecamer complex consisting of 5 subunits (α, β, γ, δ, epsilon) that form GEF-active decamers in duplicate (7).
In response to ISR activation, phosphorylated eif2α inhibits eIF 2B-mediated exchange of GDP with GTP, resulting in reduced ternary complex formation and thus inhibits translation of normal mRNA characterized by ribosome binding to the 5' aug start codon (8). Under these conditions of reduced ternary complex abundance, translation of several specific mRNAs, including the mRNA encoding the transcription factor ATF4, is activated by a mechanism involving translational changes in the upstream ORF (uORF) (7, 9, 10). These mrnas typically comprise one or more uofs that typically function in unstressed cells to limit the flow of ribosomes to the primary encoding ORF. For example, under normal conditions, the uORF in the 5' utr of ATF occupies ribosomes and prevents translation of the coding sequence of ATF 4. However, under stress conditions, i.e., conditions where ternary complex formation is reduced, the likelihood of ribosomal scanning past these upstream ORFs and translation begins at the ATF4 encoding ORF increases. ATF4 and other stress factors expressed in this manner then control the expression of a range of other stress genes. The acute phase consists in the expression of proteins intended to resume steady state, while the chronic phase leads to the expression of pro-apoptotic factors (1,11,12,13).
Upregulation of markers of ISR signaling has been demonstrated in a variety of disorders, including cancer and neurodegenerative diseases. ER stress mediated translation increases tolerance to hypoxic conditions and promotes tumor growth in cancer (14, 15, 16), and it has been demonstrated that deletion of PERK by gene targeting can slow down PERK from transformation -/- Growth of tumors derived from mouse embryonic fibroblasts (14, 17). Furthermore, a recent report has provided the following conceptual evidence: activators of eIF2B are effective in treating an aggressive metastatic prostate cancer using a patient-derived xenograft model in mice (28). In summary, the prevention of cytoprotective ISR signaling may represent an effective antiproliferative strategy for the treatment of at least some forms of cancer.
Furthermore, modulation of ISR signaling may prove to be effective in maintaining synaptic function and reducing neuronal decline, as well as in neurodegenerative diseases characterized by activation of misfolded and Unfolded Protein Responses (UPR), such as Amyotrophic Lateral Sclerosis (ALS), frontotemporal dementia (FTD), alzheimer's Disease (AD), parkinson's Disease (PD), and Jakob Creutzfeld (prion) disease (18, 19, 20). For prion diseases (an example of an existing neurodegenerative disease), pharmacology and genetic inhibition of ISR signaling have been shown to normalize protein translation levels, rescue synaptic function, and prevent neuronal loss (21). Specifically, decreasing levels of phosphorylated eif2α by controlling overexpression of phosphatases that phosphorylate levels of eif2α increases survival in prion-infected mice, while sustained eif2α phosphorylation decreases survival (22).
Furthermore, direct evidence of the importance of control protein expression levels for proper brain function exists in the form of rare genetic diseases affecting the function of eIF2 and eIF 2B. Mutations in eif2γ that disrupt the complex integrity of eIF2 and thus lead to reduced levels of normal protein expression are associated with dysnoesia syndrome (ID) (23). Partial loss of function mutations in subunits of eIF2B have been demonstrated to be responsible for rare white matter dystrophy white matter ablative disease (VWMD) (24, 25). In particular, stabilization of eIF2B partial loss of function by small molecules associated with ISRIB in VWMD mouse models has been shown to reduce ISR markers and improve function and pathological endpoints (26, 27).
The present invention provides compounds of the invention in free or pharmaceutically acceptable salt form or in solvate, hydrate, tautomer or stereoisomer form, for use in the treatment of a disease or disorder referred to herein. The same applies to the pharmaceutical compositions of the present invention.
Accordingly, one aspect of the present invention is a compound of the present invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, for use as a medicament. The same applies to the pharmaceutical compositions of the present invention.
The described methods of treatment may be applied to mammals such as dogs, cats, cattle, horses, rabbits, monkeys, and humans. Preferably, the mammalian patient is a human patient.
Accordingly, the present invention provides a compound of the present invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, or a pharmaceutical composition thereof, to be used in the treatment or prevention of one or more diseases or disorders associated with an integrated stress reaction.
Another aspect of the invention is a compound of the invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer or pharmaceutical composition thereof, for use in a method of treatment or prophylaxis of one or more disorders or diseases associated with an integrated stress reaction.
Another aspect of the invention is the use of a compound of the invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, or a pharmaceutical composition for the manufacture of a medicament for the treatment or prevention of one or more disorders or diseases associated with an integrated stress response.
Yet another aspect of the invention is a method for treating, controlling, delaying or preventing one or more diseases or disorders associated with an integrated stress response in a mammalian patient in need of such treatment, wherein the method comprises administering to the patient a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof or a pharmaceutical composition.
The present invention provides a compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof or a pharmaceutical composition thereof to be used in the treatment or prevention of one or more of the following diseases or disorders.
Another aspect of the invention is a compound of the invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer or pharmaceutical composition thereof, for use in a method of treatment or prophylaxis of one or more of the disorders or diseases described below.
Another aspect of the invention is the use of a compound of the invention, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, or a pharmaceutical composition for the manufacture of a medicament for the treatment or prevention of one or more of the following disorders or diseases.
Yet another aspect of the invention is a method for treating, controlling, delaying or preventing one or more of the following diseases or disorders in a mammalian patient in need of such treatment, wherein the method comprises administering to the patient a therapeutically effective amount of a compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof or a pharmaceutical composition.
Diseases or disorders include, but are not limited to, leukodystrophy, intellectual disability syndrome, neurodegenerative diseases and disorders, neoplastic diseases, infectious diseases, inflammatory diseases, musculoskeletal diseases, metabolic diseases, eye diseases, diseases selected from the group consisting of organ fibrosis, chronic and acute liver diseases, chronic and acute lung diseases, chronic and acute kidney diseases, myocardial infarction, cardiovascular diseases, cardiac arrhythmias, atherosclerosis, spinal cord injury, ischemic stroke and neuropathic pain.
White matter malnutrition
Examples of leukodystrophies include, but are not limited to, white matter ablative disease (VWMD) and childhood ataxia with insufficient CNS myelination (e.g., associated with impaired function of eIF2 or components in the signaling or signaling pathways that include eIF 2).
Mental disorder syndrome
Intellectual disabilities are especially those conditions: wherein a person has certain limitations in mental functions such as communicating, caring for himself, and/or has impaired social skills. Dysnoesia syndromes include, but are not limited to, dysnoesia disorders associated with impaired function of eIF2 or of components in the signaling or signaling pathway including eIF 2.
Neurodegenerative diseases/disorders
Examples of neurodegenerative diseases and disorders include, but are not limited to, alexander's disease, alzheimer's disease, amyotrophic lateral sclerosis, ataxia telangiectasia, barberry disease (also known as Spielmeeyer-Vogt-Sjogren-Batten disease), bovine Spongiform Encephalopathy (BSE), canavaliate's disease, cookjen's syndrome, corticobasal degeneration, creutzfeldt-Jakob disease, frontotemporal dementia, gerstmann-Straussler-Scheinker syndrome, huntington's disease, HIV-related dementia, kennedy's disease, kerber's disease, kuru disease, lewy body dementia Machado-Joseph's disease (spinocerebellar ataxia 3), multiple sclerosis, multiple system atrophy, narcolepsy, neurophobia, parkinson's disease, paMey's disease, pick's disease, primary lateral sclerosis, prion disease, progressive supranuclear palsy, raffsu's disease, dehoff's disease, sheffield's disease, subacute spinal cord mixed degeneration secondary to pernicious anemia, schizophrenia, spinocerebellar ataxia (multiple types with different characteristics), spinal muscular atrophy, steele-Richardson-Olszewski disease, spinal tuberculosis and Tau lesions.
The neurodegenerative disease or disorder is selected from among, inter alia, alzheimer's disease, parkinson's disease and amyotrophic lateral sclerosis.
Neoplastic disease
Neoplastic disease can be understood in its broadest sense as any tissue resulting from uncontrolled cell growth. In many cases, the tumor results in at least a large tissue mass, which is optionally innervated by blood vessels. Which may or may not comprise the formation of one or more metastasis/s. The neoplastic disease of the present invention can be any neoplasm classified by the international statistical classification of disease and related health problems, revision 10 (International Statistical Classification of Diseases and Related Health Problems th review) (ICD-10) category C00-D48.
Illustratively, a neoplastic disease according to the present invention may be the presence of one or more malignant tumors (tumors) (ICD-10 class C00-C97), the presence of one or more in situ tumors (ICD-10 class D00-D09), the presence of one or more benign tumors (ICD-10 class D10-D36), or the presence of one or more tumors of uncertain or unknown behaviour (ICD-10 class D37-D48). Preferably, a neoplastic disease according to the present invention refers to the presence of one or more malignant tumors, i.e. is malignant tumor formation (ICD-10 class C00-C97).
In a more preferred embodiment, the neoplastic disease is cancer.
Cancer can be understood in its broadest sense as any malignant disease, i.e. the presence of one or more malignant tumors in a patient. The cancer may be a solid or hematological malignancy. This paper covers but is not limited to leukemia, lymphoma, carcinoma and sarcoma.
In particular, neoplastic diseases, such as cancer, characterized by upregulated ISR markers are included herein.
Exemplary cancers include, but are not limited to, thyroid cancer, endocrine system cancer, pancreatic cancer, brain cancer (e.g., glioblastoma multiforme, glioma), breast cancer (e.g., ER positive, ER negative, chemotherapy resistant, herceptin resistant, HER2 positive, doxorubicin resistant, tamoxifen resistant, ductal carcinoma, lobular carcinoma, primary, metastatic), cervical cancer, ovarian cancer, uterine cancer, colon cancer, head and neck cancer, liver cancer (e.g., hepatocellular carcinoma), kidney cancer, lung cancer (e.g., non-small cell lung cancer, squamous cell lung cancer, adenocarcinoma, large cell lung cancer, small cell lung cancer, carcinoid, sarcoma), colon cancer, esophageal cancer, stomach cancer, bladder cancer, bone cancer, stomach cancer, prostate cancer, and skin cancer (e.g., melanoma).
Other examples include, but are not limited to, myeloma, leukemia, mesothelioma, and sarcoma.
Additional examples include, but are not limited to, medulloblastoma, hodgkin's disease, non-hodgkin's lymphoma, multiple myeloma, neuroblastoma, glioma, glioblastoma multiforme, rhabdomyosarcoma, primary thrombocytosis, primary macroglobulinemia, primary brain tumor, malignant pancreatic islet tumor, malignant carcinoid, bladder cancer, premalignant skin lesions, testicular cancer, lymphoma, genitourinary tract cancer, malignant hypercalcemia, endometrial cancer, adrenocortical cancer, endocrine or exocrine pancreatic tumors, medullary thyroid cancer, thyroid medullary carcinoma, melanoma, colorectal cancer, papillary thyroid cancer, hepatocellular carcinoma, paget's papilloma, phylloma, lobular carcinoma, ductal carcinoma, pancreatic astrocytocarcinoma, and hepatic astrocytoma.
Exemplary leukemias include, but are not limited to, acute non-lymphoblastic leukemia, chronic lymphocytic leukemia, acute myelogenous leukemia, chronic myelogenous leukemia, acute promyelocytic leukemia, adult T-cell leukemia, leukopenia leukemia, leukocytosis leukemia, basophilic leukemia, embryogenic leukemia, bovine leukemia, chronic myelogenous leukemia, skin leukemia, embryogenic leukemia, eosinophilic leukemia, gross's leukemia, hairy cell leukemia, hemangioblastic leukemia (hemoblastic leukemia), hemangioblastic leukemia (hemocytoblastic leukemia), histiocytic leukemia, stem cell leukemia, acute monocytic leukemia, leukemia leukopenia, lymphoblastic leukemia, lymphogenic leukemia, lymphoid leukemia, lymphosarcoma cell leukemia, mast cell leukemia, megakaryoblastic leukemia, mini-myeloblastic leukemia (micromyeloblastic leukemia), monocytic leukemia, myeloblastic leukemia, myelogenous leukemia, myelomonocytic leukemia, naegeli leukemia, plasma cell leukemia, multiple myeloma, plasma cell leukemia, promyelocytic leukemia, rieder cell leukemia, schlins leukemia, stem cell leukemia, sub-Bai Xiexing leukemia, and undifferentiated cell leukemia.
Exemplary sarcomas include, but are not limited to, chondrosarcoma, fibrosarcoma, lymphosarcoma, melanomas, myxosarcomas, osteosarcomas, eboloni's sarcoma, liposarcomas, soft tissue aciniform sarcomas, ameloblastic sarcomas, botulism sarcomas, green sarcomas, choriocarcinomas, embryonal sarcomas, wilms' sarcoma, endometrial sarcomas, interstitial sarcomas, ewing's sarcoma, fascia sarcomas, fibroblast sarcomas, giant cell sarcomas, granulocytosarcomas, hodgkin's sarcoma, idiopathic multiple-pigment hemorrhagic sarcomas, B cell immunoblastic sarcomas, lymphomas, T cell immunoblastic sarcomas, jensen sarcomas, kaposi's sarcoma, cookifugen's sarcoma, angiosarcomas, leukemia sarcomas, malignant mesenchymal sarcomas, periosteoexternal sarcomas, reticuloma, rous sarcoma, serous sarcoma, synovial sarcomas, and telangiectasia sarcomas.
Exemplary melanomas include, but are not limited to, acro-freckle nevus melanoma, melanotic melanoma, benign juvenile melanoma, claudeman' S melanoma, S91 melanoma, haemagglutinin melanoma, juvenile melanoma, malignant freckle-like melanoma, malignant melanoma, nodular melanoma, subungual melanoma, and superficial diffuse melanoma.
Exemplary cancers include, but are not limited to, medullary thyroid carcinoma, familial medullary thyroid carcinoma, acinar carcinoma, cystic adenoid carcinoma, adenoid cystic carcinoma, adenocarcinoma, adrenocortical carcinoma, alveolar cell carcinoma, basal epithelial cell carcinoma, basal squamous cell carcinoma, bronchioloalveolar carcinoma, bronchiolar carcinoma, and bronchi cancer, brain-like cancer, cholangiocellular carcinoma, choriocarcinoma, colloid cancer, acne-like cancer, uterine body cancer, sieve-like cancer, armor-like cancer, skin cancer, columnar cell cancer, tube cancer, duct cancer, hard cancer, embryonal cancer, brain-like cancer, epidermoid cancer, adenoid cancer, exogenous cancer, ulcerative cancer, fibrous cancer, colloid-like cancer, colloidal cancer, tumor-like cancer, and tumor-like cancer giant cell cancer (giant cell carcinoma), giant cell cancer (carcinoma gigantocellulare), adenocarcinoma, granulosa cell cancer, stroma cancer, blood sample cancer, hepatocellular cancer, hurthle cell cancer, mucinous cancer (hyaline carcinoma), adrenal gland-like cancer, infant embryo cancer, carcinoma in situ, epidermoid cancer, intraepithelial cancer, krompcher cancer, kulchitzky cell cancer, large cell cancer, bean-like cancer (lenticular carcinoma), bean-like cancer (carcinoma lenticulare), lipoma-like cancer, lobular cancer, lymphatic epithelium cancer, medullary cancer (carcinoma medullare), medullary cancer (medullary carcinoma), melanin cancer, soft cancer, mucous cancer (mucinous carcinoma), mucous cancer (carcinoma muciparum), mucous cell cancer, mucous epidermoid cancer, mucous cancer (carcinoma mucosum), mucous cancer (mucocarpioma), myxoma-like cancer (carcinoma myxomatodes), nasopharyngeal cancer, oat cell cancer, ossified cancer, bone-like cancer, papillary cancer, periportal cancer, peri-portal cancer, malignant tumor, and the like cancer, pre-invasive carcinoma, spiny cell carcinoma, brain-like carcinoma (pultaceous carcinoma), renal cell carcinoma, reserve cell carcinoma, sarcoidosis, schneider's carcinoma, hard carcinoma, scrotum carcinoma, ring cell carcinoma, simple carcinoma, small cell carcinoma, potato-like carcinoma, globular cell carcinoma, spindle cell carcinoma, medullary carcinoma, squamous cell carcinoma, cord bundle carcinoma, vasodilatory carcinoma (carcinoma telangiectaticum), vasodilatory carcinoma (carcinoma telangiectodes), transitional cell carcinoma, nodular skin carcinoma (carcinoma tuberosum), tubular carcinoma, nodular skin carcinoma (tuberous carcinoma), wart-like carcinoma, and villous carcinoma.
Infectious diseases
Examples include, but are not limited to, infections caused by viruses (such as infections caused by HIV-1: human immunodeficiency virus type 1; IAV: influenza A; HCV: hepatitis C; DENV: dengue virus; ASFV: african swine fever virus; EBV: epstein-Barr virus; HSV1: herpes simplex virus 1; chiKV: chikungunya virus; HCMV: human cytomegalovirus; SARS-CoV: severe acute respiratory syndrome coronavirus; SARS-CoV-2: severe acute respiratory syndrome coronavirus 2) and infections caused by bacteria (such as infections caused by Legionella, brucella, ximenechania (Simkania), chlamydia, helicobacter and Campylobacter).
Inflammatory diseases
Examples of inflammatory diseases include, but are not limited to, post-operative cognitive dysfunction (reduced cognitive function after surgery), traumatic brain injury, arthritis, rheumatoid arthritis, psoriatic arthritis, juvenile idiopathic arthritis, multiple sclerosis, systemic Lupus Erythematosus (SLE), myasthenia gravis, juvenile-type diabetes, type 1 diabetes, guillain-barre syndrome, hashimoto's encephalitis, hashimoto's thyroiditis, ankylosing spondylitis, psoriasis, sjogren's syndrome, vasculitis, glomerulonephritis, autoimmune thyroiditis, behcet's disease, crohn's disease, ulcerative colitis, bullous pemphigoid, sarcoidosis, ichthyosis, graves ' eye disease, inflammatory bowel disease, addison's disease, vitiligo, asthma, allergic asthma, acne vulgaris, celiac disease, chronic prostatitis, inflammatory bowel disease, pelvic inflammatory disease, reperfusion injury, sarcoidosis, transplant rejection, interstitial cystitis, atherosclerosis and atopic dermatitis.
Musculoskeletal diseases
Examples of musculoskeletal diseases include, but are not limited to, muscular dystrophy, multiple sclerosis, freidrich's ataxia, muscular dystrophy disorders (e.g., muscular atrophy, sarcopenia, cachexia), inclusion body myopathy, progressive muscular atrophy, motor neuron disease, carpal tunnel syndrome, epicondylitis, tendinitis, back pain, muscle soreness, repetitive strain injury, and paralysis.
Metabolic diseases
Examples of metabolic diseases include, but are not limited to, diabetes (especially type II diabetes), non-alcoholic steatohepatitis (NASH), non-alcoholic fatty liver disease (NAFLD), niemann-Pick disease, liver fibrosis, obesity, heart disease, atherosclerosis, arthritis, cystinosis, phenylketonuria, proliferative retinopathy and Kearns-Sayre disease.
Eye diseases
Examples of ocular diseases include, but are not limited to, oedema or neovascularization of any occlusive or inflammatory retinal vascular disease, such as rubeosis iridis, neovascular glaucoma, pterygium, vascularized glaucoma follicles, conjunctival papilloma; choroidal neovascularization such as neovascular age-related macular degeneration (AMD), myopia, anterior uveitis (priority), trauma or idiopathic; macular oedema, such as post-operative macular oedema, macular oedema secondary to uveitis (including retinal and/or choroidal inflammation), macular oedema secondary to diabetes, and macular oedema secondary to retinal vascular occlusive disease (i.e., retinal branches and central venous occlusions); retinal neovascularization caused by diabetes, such as retinal vein occlusion, uveitis, ocular ischemic syndrome caused by carotid artery disease, ocular or retinal artery occlusion, sickle cell retinopathy, other ischemic or occlusive neovascular retinopathy, retinopathy of prematurity or eirns disease; and genetic disorders such as VonHippel-Lindau syndrome.
Other diseases
Other diseases include, but are not limited to, organ fibrosis (such as liver fibrosis, lung fibrosis or kidney fibrosis), chronic and acute liver disease (such as fatty liver disease or liver sebaceous gland disease), chronic and acute lung disease, chronic and acute kidney disease, myocardial infarction, cardiovascular disease, cardiac arrhythmias, atherosclerosis, spinal cord injury, ischemic stroke, and neuropathic pain.
Yet another aspect of the invention is a pharmaceutical composition comprising at least one compound of the invention or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, and a pharmaceutically acceptable carrier, optionally in combination with one or more other biologically active compounds or pharmaceutical compositions.
Preferably, the one or more bioactive compounds are modulators of the integrated stress response pathway other than the compound of formula (I).
"pharmaceutical composition" refers to one or more active ingredients and one or more inert ingredients comprising a carrier, as well as any product formed directly or indirectly from the combination, complexation or aggregation of any two or more of the ingredients, or any product formed directly or indirectly from the decomposition of one or more of the ingredients, or any product formed directly or indirectly from other types of reactions or interactions of one or more of the ingredients. Accordingly, the pharmaceutical compositions of the present invention include any composition made by mixing a compound of the present invention and a pharmaceutically acceptable carrier.
The pharmaceutical compositions of the invention may comprise one or more additional compounds as active ingredient, such as a mixture of compounds of formula (I) in the composition or other modulators of the integrated stress response pathway.
The active ingredient may be contained in one or more different pharmaceutical compositions (combinations of pharmaceutical compositions).
The term "pharmaceutically acceptable salt" refers to salts prepared from pharmaceutically acceptable non-toxic bases or acids, including inorganic bases or acids and organic bases or acids.
The compositions include those suitable for oral, rectal, topical, parenteral (including subcutaneous, intramuscular and intravenous), ocular (ocular), pulmonary (nasal or buccal inhalation) or nasal administration, although the most suitable route in any given case will depend on the nature and severity of the condition being treated and the nature of the active ingredient. They may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
In practice, the compounds of formula (I) may be combined as an intimate mixture with a pharmaceutical carrier according to conventional pharmaceutical compounding techniques. The carrier may take a variety of forms depending on the form of the formulation intended for administration, for example, orally or parenterally (including intravenously). In preparing the oral dosage form compositions, in the case of oral liquid preparations (e.g., suspensions, elixirs and solutions), any of the usual pharmaceutical media may be employed such as water, glycols, oils, alcohols, flavoring agents, preservatives, coloring agents and the like; or in the case of oral solid preparations such as powders, hard and soft capsules and tablets, carriers such as starches, sugars, microcrystalline cellulose, diluents, granulating agents, lubricants, binders, disintegrating agents and the like may be used, with solid oral preparations being preferred over liquid preparations.
Because of the ease of administration of tablets and capsules, which represent the most advantageous oral dosage unit form, it is apparent that in such cases solid pharmaceutical carriers are used. If desired, the tablets may be coated by standard aqueous or non-aqueous techniques. Such compositions and formulations should contain at least 0.1% active compound. The percentage of active compound in these compositions may of course vary, and may conveniently be from about 2% to about 60% by weight of the unit. The amount of active compound in such therapeutically useful compositions is that amount which achieves an effective dose. The active compounds can also be administered intranasally, for example as liquid drops or sprays.
Tablets, pills, capsules, and the like may also contain: binders such as gum tragacanth, acacia, corn starch or gelatin; excipients such as dicalcium phosphate; disintegrants such as corn starch, potato starch, alginic acid; lubricants such as magnesium stearate; and sweeteners such as sucrose, lactose or saccharin. When the dosage unit form is a capsule, it may contain, in addition to materials of the type described above, a liquid carrier such as a fatty oil.
Various other substances may be present as coatings or used to alter the physical form of the dosage unit. For example, tablets may be coated with shellac, sugar or both. Syrups or elixirs may contain, in addition to the active ingredient: sucrose as a sweetener, methyl and propyl parabens as preservatives, dyes and flavouring agents such as cherry or orange flavouring.
The compounds of formula (I) may also be administered parenterally. Solutions or suspensions of these active compounds may be prepared in water suitably mixed with a surfactant such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof in oils. Under the usual conditions of storage and use, these preparations contain a preservative to prevent the growth of microorganisms.
Pharmaceutical forms suitable for injectable use include sterile aqueous solutions or dispersions, and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. In all cases, this form should be sterile and should be fluid to the extent that easy injection is possible. It should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier may be a solvent or dispersion medium containing, for example, water, ethanol, polyols (e.g., glycerol, propylene glycol, and liquid polyethylene glycol), suitable mixtures thereof, and vegetable oils.
Any suitable route of administration may be employed for providing a mammal, particularly a human, with an effective dose of a compound of the invention. For example, oral, rectal, topical, parenteral, ocular, pulmonary, nasal routes, and the like may be used. Dosage forms include tablets, troches, dispersions, suspensions, solutions, capsules, creams, ointments, aerosols, and the like. The compounds of formula (I) are preferably administered orally.
The effective dosage of the active ingredient employed may vary with the particular compound employed, the mode of administration, the condition being treated, and the severity of the condition being treated. Such dosages can be readily determined by one skilled in the art.
The starting materials for synthesizing the preferred embodiments of the present invention may be purchased from commercially available sources such as Array, sigma Aldrich, acros, fisher, fluka, ABCR, or may be synthesized by one skilled in the art using known methods.
In general, several methods are available for preparing the compounds of the present invention. In some cases, various policies may be combined. Sequential or convergent approaches may be used. Exemplary synthetic pathways are described below.
Examples
I chemical Synthesis
Experimental procedure
The following abbreviations and abbreviations are used:
2-MeTHF 2-methyltetrahydrofuran
aq aqueous
ACN acetonitrile
Silver AgOTf triflate
Saturated solution of brine NaCl in water
BnONH 2 HCl O-benzyl hydroxylamine hydrochloride
Boc t-Butoxycarbonyl group
Boc 2 Di-tert-butyl O dicarbonate
t BuOK potassium tert-butoxide
CDCl 3 Deuterated chloroform
CV column volume
DABCO 1, 4-diazabicyclo [2.2.2] octane
DCM dichloromethane
DCE dichloroethane
DIPEA diisopropylethylamine
DMAP N, N-dimethylpyridine-4-amine
DMSO dimethyl sulfoxide
DMSO-d 6 Deuterated dimethyl sulfoxide
DMF dimethylformamide
ESI + Positive ionization mode
ESI - Negative ionization mode
EtOAc ethyl acetate
EtOH ethanol
Et 2 O-diethyl ether
H 2 SO 4 Sulfuric acid
HATU 1- [ bis (dimethylamino) methylene ] -1H- [1,2,3] triazolo [4,5-b ] pyridin-1-ium 3-oxide hexafluorophosphate
HCl hydrochloric acid
HPLC high performance liquid chromatography
h hours
IPA isopropyl alcohol
K 3 PO 4 Tripotassium phosphate
KHCO 3 Potassium bicarbonate
KOH potassium hydroxide
LiOH·H 2 Lithium hydroxide hydrate of O
m multiple peaks
MeI iodomethane
MeOD deuterated methanol
MeOH methanol
MgSO 4 Magnesium sulfate
min
MsOH methane sulfonic acid
mL of
N 2 Nitrogen atmosphere
Na 2 SO 3 Sodium sulfite
Na 2 SO 4 Sodium sulfate
NaBH 4 Sodium borohydride
NaHCO 3 Sodium bicarbonate
NH 2 -NH 2 ·H 2 O hydrazine hydrate
NH 4 Cl ammonium chloride
NMI 1-methyl-1H-imidazole
NMM 4-methylmorpholine
NMR nuclear magnetic resonance
prep. preparation
r.t. room temperature
RT retention time
satd saturated
TCFH chloro-N, N, N ', N' -tetramethyl formamidine hexafluorophosphate
TsCl 4-methylbenzenesulfonyl chloride
TsOH 4-methylbenzene-1-sulfonic acid
TBME 2-methoxy-2-methylpropane
THF tetrahydrofuran
TFA 2, 2-trifluoro acetic acid
TMSOI trimethylsulfoxonium iodide
ZnBr 2 Zinc dibromide
Analytical LCMS conditions were as follows:
system 1 (S1) acidic IPC method (MS 18 and MS 19)
A Kinetex Core shell C column (2.1 mm. Times.50 mm,5 μm; temperature: 40 ℃) and a gradient of 5-100% B (A=0.1% formic acid/H were used 2 O; b=0.1% formic acid/ACN) for 1.2min, then at 100% B for 0.1min, analytical (MET/CR/1410) HPLC-MS was performed on Shimadzu LCMS system. A second gradient of 100-5% B was then applied over 0.01min, with an injection volume of 3. Mu.L and a flow rate of 1.2mL/min. UV spectra were recorded at 215nm using an SPD-M20A photodiode array detector (spectral range: 200-400 nm). A mass spectrum was obtained using a 2010EV detector. Data were integrated and reported using Shimadzu LCMS-Solutions and PsiPort software.
System 2 (S2) acidic IPC method (MSQ 1, MSQ2 and MSQ 4)
Using WatersBEH TM C18 column (2.1 mm. Times.50 mm,1.7 μm; temperature 40 ℃) and a gradient of 5-100% B (A=0.1% formic acid/H 2 O: b=0.1% formic acid/ACN) for 1.1min, then at 100% B for 0.25min, analytical (MET/uPLC/1704) uHPLC-MS was performed on a Waters Acquity uPLC system. A second gradient of 100-5% B was then applied over 0.05min and held for 0.1min with an injection volume of 1. Mu.L and a flow rate of 0.9mL/min. UV spectra were recorded at 215nm on Waters Acquity PDA having a spectral range of 200-400 nm. Mass spectra were obtained using Waters QDa. Data was integrated and reported using Waters MassLynx and OpenLynx software.
System 3 (S3) alkaline IPC method (MS 16)
Using WatersBEH TM A C18 column (2.1 mM. Times.30 mM,1.7 μm; temperature 40 ℃) and a gradient of 5-100% B (A: 2mM ammonium bicarbonate, buffered to pH 10, B: ACN) were run over 0.75min, then held at 100% B for 0.1min, and analytical (MET/CR/1602) uHPLC-MS was performed on a Waters Acquity uPLC system. A second gradient of 100-5% B was then applied over 0.05min and held for 0.1min with an injection volume of 1. Mu.L and a flow rate of 1mL/min. UV spectra were recorded at 215nm on Waters Acquity PDA having a spectral range of 200-400 nm. A mass spectrum was obtained using Waters Quattro Premier XE. Data was integrated and reported using Waters MassLynx and OpenLynx software.
System 4 (S4) acid final method (MSQ 1 and MSQ 2)
A Phenomenex Kinetex-XB C18 column (2.1 mm. Times.100 mm, 1.7. Mu.M; temperature: 40 ℃) and a gradient of 5-100% B (A=0.1% formic acid/H were used 2 O; b=0.1% formic acid/ACN) for 5.3min, then at 100% B for 0.5min, analytical (MET/uPLC/AB 101) uHPLC-MS was performed on Waters Acquity uPLC system. A second gradient of 100-5% B was then applied over 0.02min and held for 1.18min with an injection volume of 1. Mu.L and a flow rate of 0.6mL/min. UV spectra were recorded at 215nm using a Waters Acquity PDA detector (spectral range: 200-400 nm). Mass spectra were obtained using Waters SQD (MSQ 1) or Waters Acquity QDA (MSQ 2). Data was integrated and reported using Waters MassLynx and OpenLynx software.
System 5 (S5) acid final method (MS 18, MS 19)
A Waters Atlantis dC column (2.1 mm. Times.100 mm,3 μm; temperature: 40 ℃) and a gradient of 5-100% B (A=0.1% formic acid/H were used 2 O; b=0.1% formic acid/ACN) for 5min, then at 100% B for 0.4min, analytical (MET/CR/1416) HPLC-MS was performed on Shimadzu LCMS system. A second gradient of 100-5% B was then applied over 0.02min and held for 1.58min with an injection volume of 3. Mu.L and a flow rate of 0.6mL/min. UV spectra were recorded at 215nm using an SPD-M20A photodiode array detector (spectral range: 200-400 nm). A mass spectrum was obtained using a 2010EV detector. Using Shimadzu LCMS-Solutions and PsiPort softwaresAnd integrating and reporting the data.
System 6 (S6) alkaline Final Process (MS 16)
Using WatersBEH TM The C18 column (2.1 mM. Times.100 mM,1.7 μm column; temperature: 40 ℃) and a gradient of 5-100% (A=2 mM ammonium bicarbonate, buffered to pH 10; B=ACN) were run for 5.3min, then held at 100% B for 0.5min, and analytical (MET/uHPLC/AB 105) uPLC-MS was performed on a Waters Acquity uPLC system. A second gradient of 100-5% B was then applied over 0.02min and held for 1.18min with an injection volume of 1. Mu.L and a flow rate of 0.6mL/min. UV spectra were recorded at 215nm using a Waters acquisition photodiode array detector (spectral range: 200-400 nm). A mass spectrum was obtained using a Waters Quattro Premier XE mass detector. Data was integrated and reported using Waters MassLynx and OpenLynx software.
The purification method is as follows:
method 1 acid early stage method
A Waters Sunfire C18 column (30 mm. Times.100 mm, 10. Mu.M; temperature: room temperature) and a gradient of 10-95% B (A=0.1% formic acid/H were used 2 O; b=0.1% formic acid/ACN) for 14.44min, then held at 95% B for 2.11min, purified (P1) LC on Gilson LC system. A second gradient of 95-10% B was then applied over 0.2min, with an injection volume of 1500. Mu.L and a flow rate of 40mL/min. UV spectra were recorded at 215nm using Gilson detector.
Method 2 acid Standard method
Purification of (P2) LC was performed on a Gilson LC system using a Waters Sunfire C18 column (30 mm x 10mm,10 μm; temperature: room temperature) and a gradient of 30-95% B (a=0.1% formic acid/water; b=0.1% formic acid/ACN) for 11.00min, then 2.10min at 95% B. A second gradient of 95-30% B was then applied over 0.2min, with an injection volume of 1500. Mu.L and a flow rate of 40mL/min. UV spectra were recorded at 215nm using Gilson detector.
Method 3 alkaline early stage method
A Waters X-Bridge C18 column (30 mm. Times.100 mm, 10. Mu.M; temperature: room temperature) and a gradient of 10-95% B (A=0.2% NH) were used 4 OH/H 2 O;B=0.2%NH 4 OH/ACN) for 14.44min, then at 95% B for 2.11min, purified (P3) LC on Gilson LC system. A second gradient of 95-10% B was then applied over 0.2min, with an injection volume of 1500. Mu.L and a flow rate of 40mL/min. UV spectra were recorded at 215nm using Gilson detector.
Method 4 alkaline Standard method
A Waters X-Bridge C18 column (30 mm. Times.10 mm, 10. Mu.M; temperature: room temperature) and a gradient of 30-95% B (A=0.2% NH) were used 4 OH/water; b=0.2% nh 4 OH/ACN) for 11.00min, then at 95% B for 2.10min, purified (P4) LC on Gilson LC system. A second gradient of 95-30% B was then applied over 0.21min, with an injection volume of 1500. Mu.L and a flow rate of 40mL/min. UV spectra were recorded at 215nm using Gilson detector.
Method 5: reversed phase chromatography using acidic pH, standard elution method
A gradient of the appropriate SNAP C18 column and 10% b (a=0.1% formic acid/H was used 2 O; b=0.1% formic acid/ACN) was subjected to 1.7CV, then 10-100% B was subjected to 19.5CV and 100% B was subjected to 2CV, FCC purification on reversed phase silica gel was performed on a Biotage Isolera system (acidic pH, standard elution method).
Method 6: reversed phase chromatography using alkaline pH, standard elution method
A gradient of the appropriate SNAP C18 column and 10% b (a=0.1% nh was used 3 /H 2 O;B=0.1%NH 3 ACN) was subjected to 1.7CV, then 10-100% b was subjected to 19.5CV and 100% b was subjected to 2CV, FCC purification (alkaline pH, standard elution method) on reversed phase silica gel was performed on a Biotage Isolera system.
NMR conditions
Recording at 500MHz, 400MHz or 250MHz on a Bruker Avance III HD 500MHz spectrometer, bruker Avance III HD 400MHz spectrometer or Bruker Avance III HD 250MHz spectrometer, respectively, unless otherwise specified 1 H NMR spectrum. Chemical shift δ is referenced in parts per million (ppm) and refers to the residual solvent peak. The following abbreviations are used to represent multiplicity and general designations: s (singlet), d (doublet), t (triplet), q (tetrad)Doublet), dd (doublet of doublets), ddd (doublet of doublets), dt (doublet of doublets), dq (doublet of quartets), hep (heptadoublet), m (multiplet), pent (quindoublet), td (triplet), qd (tetradoublet), app (distinct), and br. (broad). Coupling constant J is referenced to the nearest 0.1Hz.
General synthesis:
all compounds have been synthesized in a purity equal to or greater than 95% unless otherwise indicated.
Scheme of route 1
Step 1.A: (2R) -5- [ (benzyloxy) imino ] -2- { [ (tert-butoxy) carbonyl ] amino } -6-chlorohexanoic acid ethyl ester
DMSO (75 mL) was added to TMSOI (12.9 g,58.3 mmol) and t BuOK (6.27 g,55.9 mmol) was dissolved in dry THF (62 mL) and the solution was stirred at room temperature for 1h. The reaction mixture was cooled to-12 ℃ and a solution of Boc-D-pyroglutamic acid ethyl ester (12.5 g,48.6 mmol) in anhydrous THF (38 mL) was added and stirred at room temperature for 16h. The reaction mixture was saturated with NH 4 Cl aqueous solution (78 mL), H 2 O (15 mL) and EtOAc (200 mL) were diluted, the organic layer was separated, washed with brine and concentrated in vacuo to about 100mL. Adding BnONH 2 A solution of HCl (8.14 g,51.0 mmol) in EtOAc (62 mL) and the mixture was stirred at reflux for 2h. The reaction mixture was cooled to room temperature and quenched with H 2 O and brine wash. The organic extract was concentrated in vacuo to give the title compound (85% purity, 19.5g,40.1mmol,83% yield) as a colourless oil; 1 H NMR(400MHz,CDCl 3 )δ7.16–7.33(m,5H),5.01–5.06(m,2H),3.95–4.30(m,5H),2.32–2.50(m,2H),1.98–2.13(m,1H),1.75–1.92(m,1H),1.30–1.40(m,9H),1.12–1.24(m,3H)。
step 1.B: (2R) -5- [ (benzyloxy) imino ] piperidine-2-carboxylic acid ethyl ester
To (2R) -5- [ (benzyloxy) imino]-2- { [ (tert-butoxy) carbonyl]A solution of ethyl amino } -6-chlorohexanoate (85% purity, 19.5g,40.1 mmol) in EtOAc (157 mL) was added MsOH (7.8 mL,0.12 mol) and the mixture stirred at 42℃for 2h. The reaction mixture was added to KHCO 3 (20.1 g,0.201 mol) in H 2 The mixture was stirred in a solution of O (100 mL) at 52℃for 2 hours. The solution was cooled to room temperature, the organic layer was separated, washed with brine, and dried over Na 2 SO 4 Drying and concentration in vacuo gave the title compound (85% purity, 13.0g,40.0 mmol) as a dark orange oil in quantitative yield; 1 H NMR(400MHz,CDCl 3 )δ7.20–7.34(m,5H),4.99(d,J=4.8Hz,2H),4.13(q,J=7.1Hz,2H),3.45–3.56(m,1H),3.25(dd,J=14.9,9.8Hz,1H),3.08(dt,J=14.5,4.3Hz,1H),2.01–2.32(m,3H),1.55–1.80(m,1H),1.21(t,J=7.1Hz,3H)。
step 1.C: (2R, 5S) -5- [ (benzyloxy) amino ] piperidine-2-carboxylic acid ethyl ester oxalic acid
Propionic acid (23 mL,0.240 mol) was added to NaBH 4 (3.03 g,80.0 mmol) in EtOAc (95 mL) and the mixture was stirred at room temperature for 1 hr. The reaction mixture was added to (2R) -5- [ (benzyloxy) imino ] at-20 ℃and ]Piperidine-2-carboxylic acid ethyl ester (85% purity, 13.0g,40.0 mmol) in EtOAc (95 mL) and H 2 SO 4 In the solution in (11 mL,0.20 mol), the mixture was stirred at room temperature for 60 hours. The reaction mixture was treated with H 2 O (75 mL) and diluted with NH 4 And (5) neutralizing an OH aqueous solution. The organic layer was separated, washed with brine, and dried over Na 2 SO 4 Dried and concentrated to a volume of 75 mL. The solution was heated to 45℃and MeOH (30 mL) was added followed by oxalic acid (3.60 g,40.0 mmol) in MeOH15 mL) of the solution. The resulting mixture was cooled to 0deg.C and the resulting precipitate was filtered under vacuum, washed with MeOH/EtOH (1:4) and EtOAc to afford the title compound (7.17 g,19.1mmol,48% yield); 1 H NMR(500MHz,DMSO-d 6 )δ=7.25–7.42(m,5H),4.59(s,2H),4.17–4.24(m,2H),3.92(dd,J=12.3,3.2Hz,1H),3.34–3.40(m,1H),3.10(ddd,J=15.1,7.6,3.9Hz,1H),2.64(t,J=11.5Hz,1H),2.13(dt,J=10.2,3.4Hz,1H),1.87(dd,J=9.0,3.8Hz,1H),1.65(qd,J=13.2,3.6Hz,1H),1.40(qd,J=12.8,3.9Hz,1H),1.23(t,J=7.1Hz,3H);M/Z:279,[M+H] + ,ESI + ,RT=0.81(S1)。
intermediate 1 (step 1. D): (2R, 5S) -5- [ (benzyloxy) amino ] piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
At 0℃to (2R, 5S) -5- [ (benzyloxy) amino group]To a solution of piperidine-2-carboxylic acid ethyl ester oxalic acid (4.8 g,12.8 mmol) in anhydrous DCM (64 mL) was added Et 3 N (7.6 mL,54.7 mmol) and DMAP (161 mg,1.32 mmol) followed by Boc 2 O (8.9 mL,38.7 mmol) and the mixture was stirred at room temperature for 17 hours. The reaction mixture was saturated with NH 4 Aqueous Cl solution was diluted, the organic layer was separated, washed with brine, and dried over Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (0-40% etoac/heptane) to give the title compound (3.37 g,8.46mmol,66% yield) as a colorless oil; 1 H NMR(400MHz,CDCl 3 )δ7.39–7.27(m,5H),5.47(s,1H),4.98–4.78(m,1H),4.72(q,J=11.5Hz,3H),4.27–4.07(m,3H),3.16(s,2H),1.95(s,2H),1.74–1.63(m,1H),1.52(s,1H),1.45(s,10H),1.27(t,J=7.1Hz,3H)。
Scheme of route 2
/>
Step 2.A: (2R, 5S) -5- [ (benzyloxy) carbonyl ] amino ] piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
At 0℃to (2R, 5S) -5- [ (benzyloxy) amino group]A solution of piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (3.37 g,8.46mmol, intermediate 1) in DCM (45 mL) was added DMAP (103 mg,0.846 mmol), pyridine (1.44 mL,16.92 mmol) and benzyl chloroformate (3.0 mL,21.1 mmol) and the mixture stirred at room temperature for 24 h. The reaction mixture was treated with H 2 O (50 mL) was diluted and extracted with DCM (2X 50 mL). The combined organic extracts were dried using a phase separator, concentrated in vacuo, and purified by silica gel chromatography (0-30% etoac/heptane) to give the title compound (3.97 g,7.36mmol,87% yield) as a colourless oil; 1 H NMR(400MHz,CDCl 3 )δ7.42–7.27(m,9H),5.33–5.15(m,2H),4.92–4.82(m,2H),4.61–4.48(m,1H),4.33–4.24(m,1H),4.19(q,J=7.1Hz,2H),3.51(dd,J=14.2,5.0Hz,1H),2.29–2.16(m,1H),1.95–1.84(m,2H),1.78–1.67(m,1H),1.57–1.51(m,1H),1.40(s,9H),1.27(t,J=7.1Hz,3H)。
intermediate 2 (step 2. B): (2R, 5S) -5- [ (benzyloxy) carbonyl ] amino ] -1- [ (tert-butoxy) carbonyl ] piperidine-2-carboxylic acid
To (2R, 5S) -5- [ (benzyloxy) carbonyl ]]Amino group]Piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (3.97 g,7.36 mmol) in MeOH (10 mL) and H 2 A solution in O (17 mL) was added to a 2M aqueous LiOH solution (5.8 mL,11.6 mmol) and the mixture was stirred at room temperature for 18 hours. The reaction mixture was cooled to 0 ℃ and acidified to pH2/3 using 1M aqueous HCl. The aqueous solution was extracted with EtOAc (2X 50 mL) and treated with Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 5) to give the title compound (3.24 g,6.35mmol,82% yield) as an off-white solid; 1 H NMR(400MHz,CDCl 3 )δ7.43–7.13(m,10H),5.28–5.17(m,2H),4.92–4.83(m,2H),4.66–4.53(m,1H),4.37–4.21(m,1H),4.18–3.89(m,2H),3.51(dd,J=14.2,4.9Hz,1H),2.31–2.18(m,1H),2.01–1.86(m,2H),1.86–1.72(m,1H),1.40(s,9H);M/Z:483[M-H] - ,ESI - ,RT=1.31(S1)。
scheme of route 3
Intermediate 3 (step 3): [ (E) -4, 4-trifluoro-but-2-enyl ] N-carbamic acid ester
At 0℃under N 2 To a solution of (2E) -4, 4-trifluoro-but-2-en-1-ol (1.00 g,7.93 mmol) in DCM (5 mL) was added a solution of CDI (1.93 g,11.9 mmol) in THF (8.5 mL). After 10 minutes the ice bath was removed and the reaction mixture was stirred at room temperature for 2.5 hours. The mixture was transferred to a dropping funnel and added dropwise to a hydrazine hydrate solution (80%, 2.0ml,31.7 mmol) at 0 ℃ over 20 minutes, followed by stirring overnight at room temperature. H for the reaction mixture 2 O (50 mL) was diluted and the aqueous layer was extracted with EtOAc (3X 20 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous (8X 20 mL) and brine (20 mL) were washed with Na 2 SO 4 Drying and concentration in vacuo gave the title compound (1.02 g,5.26mmol,66% yield) as a colourless oil; 1 H NMR(500MHz,CDCl 3 )δ6.49–6.34(m,1H),6.20(s,1H),5.91–5.78(m,1H),4.81–4.65(m,2H),3.79(s,2H)。
the intermediate compounds in table 1 were synthesized according to general scheme 3 as exemplified for intermediate 3 using the corresponding starting materials.
TABLE 1
Scheme of route 4
Step 4.A: (1 s,3 s) -N' - [ (tert-butoxy) carbonyl ] -3- (trifluoromethoxy) cyclobutane-1-carbohydrazide
/>
To a solution of (1 s,3 s) -3- (trifluoromethoxy) cyclobutane-1-carboxylic acid (87% purity, 2.00g,9.45 mmol) and DIPEA (3.5 mL,19.9 mmol) in anhydrous DMF (18 mL) was added HATU (4.18 g,11.0 mmol), and the mixture was stirred at room temperature for 10 min. Tert-butyl carbazate (1.31 g,9.92 mmol) was added in portions and the mixture was stirred at room temperature for 2 hours. The reaction mixture was diluted with EtOAc (20 mL) and with H 2 O (2X 20 mL) and brine (2X 20 mL). The organic extract was subjected to MgSO 4 Dried, concentrated in vacuo, and purified by silica gel chromatography (0-10% MeOH/DCM). The product containing fractions were combined and concentrated in vacuo and then further purified by silica gel chromatography (KP-NH, 0-100% etoac/heptane) to give the title compound (2.09 g,6.66mmol,70% yield) as a yellow oil; 1 H NMR(500MHz,DMSO-d 6 )δ9.62(s,1H),8.75(s,1H),4.79(p,J=7.6Hz,1H),2.67–2.57(m,1H),2.33–2.19(m,2H),1.41(s,9H);M/Z:199[M-Boc+H] + ,ESI + ,RT=0.77(S2)。
intermediate 5 (step 4. B): (1 s,3 s) -3- (trifluoromethoxy) cyclobutane-1-carbohydrazide
(1 s,3 s) -N' - [ (tert-butoxy) carbonyl]-3- (trifluoromethoxy) cyclobutane-1-carbohydrazide (2.09 g,6.66 mmol) dissolved in 4M HCl/1, 4-dioxane (21 mL) and stirred overnight at room temperature. The reaction mixture was concentrated in vacuo, the residue was suspended in DCM and stirred, then the solid was filtered, taken up in H 2 O was washed and dried using vacuum filtration to give the title compound (91% purity, 1.13g,5.19mmol,78% yield) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ11.10(s,1H),4.83(p,J=7.5Hz,1H),2.89–2.74(m,1H),2.63–2.52(m,2H),2.38–2.23(m,2H);M/Z:199[M+H] + ,ESI + ,RT=0.50(S2)。
Scheme of route 5
Intermediate 7 (step 5.a): (2R, 5S) -5-aminopiperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
At N 2 Downward (2R, 5S) -5- [ (benzyloxy) amino group]A solution of piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (40 g,0.106mol, intermediate 1) in dry EtOH (1L) was added Pd/C (10%, 11.3g,10.6 mmol) and the mixture was taken up in H 2 Stirred for 21 hours. The reaction mixture was filtered through a pad of celite and the filtrate obtained was treated with H 2 O (500 mL) was diluted and acidified to pH4 using 1M aqueous HCl. The aqueous solution was extracted with DCM (3X 500 mL) and the organic extract was set aside. The aqueous layer was then basified to pH8 using 1M aqueous NaOH and extracted with DCM (3×500 mL). The organic extracts were combined, washed with brine (250 mL), and dried over MgSO 4 Dried and concentrated in vacuo to give the title compound (93% purity, 25.6g,87.3mmol,83% yield) as a pale brown oil; 1 H NMR(400MHz,CDCl 3 )δ5.01–4.59(m,1H),4.21(q,J=7.1Hz,2H),3.94–3.72(m,1H),3.33–3.03(m,2H),2.16–1.99(m,2H),1.66–1.54(m,2H),1.54–1.34(m,11H),1.35–1.22(m,3H);M/Z:173[M-Boc+H] + ,ESI + ,RT=0.67(S2)。
step 5.B: (2R, 5S) -5- { [ (benzyloxy) carbonyl ] amino } piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
To a solution of (2 r,5 s) -5-aminopiperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (93% purity, 13.3g,45.2mmol, intermediate 7), DMAP (553 mg,4.52 mmol) and pyridine (7.34 mL,90.7 mmol) in DCM (225 mL) was added benzyl chloroformate (11.3 mL,79.2 mmol) at 0 ℃ and the mixture stirred at room temperature for 72 hours. The reaction mixture was cooled to 0deg.C, additional portions of DMAP (553 mg,4.52 mmol) and benzyl chloroformate (11.3 mL,79.2 mmol) were added and the mixture was stirred at room temperature for 2.5h. The reaction mixture was treated with H 2 O (200 mL) was diluted and extracted with DCM (3X 250 mL). The combined organic extracts were dried using a phase separator, concentrated in vacuo, and purified by silica gel chromatography (0-100% etoac/heptane) to give the title compound (68% purity, 17.1g,28.6mmol,63% yield) as a yellow oil; 1 H NMR(400MHz,CDCl 3 )δ7.35–7.21(m,5H),5.16–4.95(m,3H),4.86–4.52(m,1H),4.13(q,J=6.7Hz,2H),3.99–3.69(m,2H),3.20–2.95(m,1H),2.13–1.95(m,1H),1.91–1.69(m,2H),1.37(s,9H),1.20(t,J=7.1Hz,3H);M/Z:307[M-Boc+H] + ,ESI + ,RT=1.25(S2)。
step 5.C: (2R, 5S) -5- { [ (benzyloxy) carbonyl ] amino } -1- [ (tert-butoxy) carbonyl ] piperidine-2-carboxylic acid
To (2R, 5S) -5- { [ (benzyloxy) carbonyl]Amino } piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (68% purity, 17.1g,28.6 mmol) in EtOH (115 mL): THF (115 mL): H 2 LiOH H was added to the solution in O (115 mL) 2 O (1.36 g,31.7 mmol) and the mixture was stirred at room temperature for 19 hours. The reaction mixture was treated with H 2 O (100 mL) and EtOAc (100 mL) were diluted and the organic layer was discarded. The aqueous layer was acidified by addition of 1M aqueous HCl and extracted with EtOAc (3X 200 mL). The combined organic extracts were washed with brine, over MgSO 4 Dried and concentrated in vacuo to afford the title compound (84%) in quantitative yieldPurity, 14.5g,32.2 mmol) as colorless gum; 1 H NMR(400MHz,CDCl 3 )δ7.45–7.31(m,5H),5.27–5.04(m,3H),5.02–4.71(m,1H),4.09–3.80(m,2H),3.30–3.09(m,1H),2.23–2.05(m,1H),2.05–1.75(m,2H),1.71–1.51(m,1H),1.47(s,9H);M/Z:279[M-Boc+H] + ,ESI + ,RT=0.90(S2)。
intermediate 9 (step 5. D): (2R, 5S) -5- { [ (benzyloxy) carbonyl ] amino } -2- (hydrazinocarbonyl) piperidine-1-carboxylic acid tert-butyl ester
To (2R, 5S) -5- { [ (benzyloxy) carbonyl ]Amino } -1- [ (tert-butoxy) carbonyl group]A solution of piperidine-2-carboxylic acid (84% purity, 6.79g,15.1 mmol) in DMF (60 mL) was added HATU (6.88 g,18.1 mmol) and DIPEA (3.2 mL,18.1 mmol) and the mixture was taken up in N at room temperature 2 Stirred for 30 minutes. The solution was then added dropwise to the NH via cannula 2 -NH 2 ·H 2 A solution of O (1.5 mL,30.1 mmol) in DMF (30 mL) and the mixture was stirred at room temperature for 1.5 h. The reaction mixture was diluted with EtOAc (200 mL) and taken up in H 2 O (4X 50 mL) was washed. The combined organic extracts were purified over Na 2 SO 4 Drying, concentration in vacuo, and purification by silica gel chromatography (0-10% meoh/DCM) gave the title compound (79% purity, 10.2g,20.5 mmol) as a white solid in quantitative yield; 1 H NMR(400MHz,DMSO-d 6 )δ9.04(s,1H),7.46–7.24(m,5H),5.03(s,2H),4.62–4.37(m,1H),4.20(s,2H),4.06–3.82(m,1H),3.76–3.48(m,1H),3.26–3.13(m,1H),2.12–1.93(m,1H),1.80–1.54(m,2H),1.54–1.44(m,1H),1.42–1.20(m,10H);M/Z:293[M-Boc+H] + ,ESI + ,RT=0.79(S2)。
scheme of route 6
Step 6.a: (2R, 5S) -2- (5-amino-1, 3, 4-oxadiazol-2-yl) -5- { [ (benzyloxy) carbonyl ] amino } piperidine-1-carboxylic acid tert-butyl ester
To (2R, 5S) -5- { [ (benzyloxy) carbonyl]A solution of amino } -2- (hydrazinocarbonyl) piperidine-1-carboxylic acid tert-butyl ester (79% purity, 10.2g,20.5mmol, intermediate 9) in 1, 4-dioxane (70 mL) was added NaHCO 3 (2.58 g,30.8 mmol) in H 2 A solution in O (20 mL) was then added BrCN (2.17 g,20.5 mmol) and the mixture was stirred at room temperature for 2.5 hours. H for the reaction mixture 2 Diluting with O, filtering the precipitate under vacuum, and purifying with H 2 O washing afforded the title compound (84% purity, 11.0g,22.2 mmol) as an off-white powder in quantitative yield; 1 H NMR(400MHz,DMSO-d 6 )δ7.51–7.42(m,1H),7.41–7.27(m,5H),7.05–6.95(m,2H),5.30(s,1H),5.09–4.97(m,2H),4.11–3.98(m,1H),2.86–2.76(m,1H),2.29–2.14(m,1H),1.95–1.79(m,2H),1.65–1.53(m,1H),1.44–1.30(m,10H);M/Z:318[M-Boc+H] + ,ESI + ,RT=0.86(S2)。
step 6.B: (2R, 5S) -5- { [ (benzyloxy) carbonyl ] amino } -2- (5-bromo-1, 3, 4-oxadiazol-2-yl) piperidine-1-carboxylic acid tert-butyl ester
To (2 r,5 s) -2- (5-amino-1, 3, 4-oxadiazol-2-yl) -5- { [ (benzyloxy) carbonyl]A solution of tert-butyl amino } piperidine-1-carboxylate (84% purity, 11.0g,22.2 mmol) and CuBr (3 eq,9.54g,66.5 mmol) in anhydrous ACN (400 mL) was added tert-butyl nitrite (90% purity, 17.6mL,133.0 mmol) and the mixture stirred at room temperature for 5 hours. Further portions of CuBr (1.5 eq,4.77g,33.3 mmol) and tert-butyl nitrite (90% purity, 8.79mL,66.5 mmol) were added and the mixture stirred at room temperature for 19h. The reaction mixture was diluted with EtOAc (250 mL) and quenched with Rochelle's salt (2X 200 mL) and H 2 O (3X 200 mL) was washed. The organic extract was purified by Na 2 SO 4 Drying, concentrating in vacuo, and purifying by silica gel chromatographyPurification by means of (0-100% EtOAc/heptane) afforded the title compound (2.02 g,4.03mmol,18% yield) as an off-white solid; 1 H NMR(400MHz,DMSO-d 6 )δ7.52(d,J=6.2Hz,1H),7.41–7.28(m,5H),5.57–5.41(m,1H),5.05(s,2H),4.08–3.91(m,1H),3.65–3.53(m,1H),2.96–2.84(m,1H),2.33–2.23(m,1H),1.99–1.90(m,1H),1.88–1.72(m,1H),1.65–1.57(m,1H),1.38(s,9H);M/Z:383[M-Boc+H] + ,ESI + ,RT=1.09(S2)。
step 6.C: (2R, 5S) -5- { [ (benzyloxy) carbonyl ] amino } -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester
To a solution of 2- (trifluoromethoxy) ethan-1-ol (13%/THF/toluene, 4.50g,4.43 mmol) in anhydrous THF (15 mL) was added NaH (60%, 322mg,8.06 mmol) at 0 ℃ and the resulting mixture was stirred at 0 ℃ for 10 min. (2R, 5S) -5- { [ (benzyloxy) carbonyl group was added in anhydrous THF (10 mL)]Amino } -2- (5-bromo-1, 3, 4-oxadiazol-2-yl) piperidine-1-carboxylic acid tert-butyl ester (2.02 g,4.03 mmol) and the resulting mixture was stirred at room temperature for 2 hours. The reaction mixture was treated with H 2 O (50 mL) was diluted and extracted with EtOAc (3X 100 mL). The combined organic extracts were dried over MgSO 4 Dried, concentrated in vacuo, and purified by silica gel chromatography (0-100% etoac/heptane) to give the title compound (85% purity, 1.60g,2.56mmol,64% yield) as a yellow oil; 1 H NMR(400MHz,DMSO-d 6 )δ7.50(d,J=6.1Hz,1H),7.43–7.26(m,5H),5.46–5.29(m,1H),5.04(s,2H),4.80–4.58(m,2H),4.57–4.41(m,2H),4.43–4.26(m,1H),3.73–3.51(m,1H),2.96–2.80(m,1H),2.32–2.16(m,1H),1.96–1.73(m,2H),1.69–1.49(m,1H),1.37(s,9H);M/Z:531[M-Boc+H] + ,ESI + ,RT=3.83(S4)。
intermediate 10 (step 6.d): (2R, 5S) -5-amino-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester
At N 2 Downward (2R, 5S) -5- { [ (benzyloxy) carbonyl]Amino } -2- {5- [2- (trifluoromethoxy) ethoxy]A solution of tert-butyl-1, 3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (85% purity, 1.60g,2.56 mmol) in EtOH (45 mL) was added Pd/C (10%, 3.27g,3.08 mmol) and the resulting mixture was taken up in H 2 Stirred at room temperature for 18 hours. The reaction mixture was filtered through celite pad and concentrated in vacuo to give the title compound (49% purity, 843mg,1.04mmol,41% yield) as a pale brown oil; 1 H NMR(400MHz,DMSO-d 6 )δ5.39–5.26(m,1H),4.76–4.64(m,2H),4.54–4.42(m,2H),4.43–4.25(m,1H),3.74–3.60(m,1H),3.20–2.91(m,3H),2.30–2.09(m,1H),1.93–1.78(m,1H),1.75–1.59(m,1H),1.53–1.25(m,11H);M/Z:397[M+H] + ,ESI + ,RT=1.76(S4)。
Scheme of route 7
Step 7.a: 7-chloro-6-fluoroquinoline-3-carboxylic acid ethyl ester
To a solution of 2-amino-4-chloro-5-fluorobenzaldehyde (270 mg,1.56 mmol) and ethyl 3, 3-diethoxypropionate (740 mg,3.89 mmol) in toluene (3 mL) was added TsOH (27 mg,0.156 mmol) and the mixture was stirred in a sealed tube at 120℃for 5 hours. The reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc (10 mL) with saturated NaHCO 3 Aqueous solution (10 mL) and H 2 O (10 mL) washing over MgSO 4 Dried and concentrated in vacuo. The residue was suspended in TBME/heptane (1:1), the resulting precipitate was filtered in vacuo and dried in vacuo to give the title compound (230 mg,0.907mmol,58% yield) as an off-white powder; 1 H NMR(400MHz,DMSO-d 6 )δ9.31(d,J=2.0Hz,1H),9.02(d,J=2.0Hz,1H),8.39(d,J=7.2Hz,1H),8.28(d,J=9.7Hz,1H),4.42(q,J=7.1Hz,2H),1.39(t,J=7.1Hz,3H);M/Z:252,254[M+H] + ,ESI + ,RT=1.00(S2)。
intermediate 11 (step 7. B): 7-chloro-6-fluoroquinoline-3-carboxylic acid
To 7-chloro-6-fluoroquinoline-3-carboxylic acid ethyl ester (230 mg,0.907 mmol) in THF (2.5 mL) and H 2 LiOH H was added to a solution in O (2.5 mL) 2 O (46 mg,1.09 mmol) and the mixture was stirred at room temperature overnight. The reaction mixture was concentrated in vacuo to remove THF and the aqueous solution was acidified to-pH 3 using 1M aqueous HCl. The resulting precipitate was collected by vacuum filtration and dried in vacuo to give the title compound (119 mg,0.506mmol,56% yield) as a pale yellow powder; 1 H NMR(400MHz,DMSO-d 6 )δ9.32(d,J=2.0Hz,1H),9.01(d,J=2.0Hz,1H),8.39(d,J=7.2Hz,1H),8.28(d,J=9.7Hz,1H).;M/Z:224,226[M-H] - ,ESI-,RT=0.74(S2)。
Scheme of route 8
Step 8.A: 6-chloroindolizine-2-carboxylic acid methyl ester
At N 2 DABCO (190 mg,1.70 mmol) was added to a solution of 5-chloropyridine-2-carbaldehyde (2.0 g,14.1 mmol) and methyl prop-2-enoate (5.0 mL,55.5 mmol) and the mixture was stirred at room temperature for 12 hours. The reaction mixture was concentrated in vacuo and azeotroped with toluene. The residue was dissolved in acetic anhydride (7.0 mL,74.1 mmol) and stirred at 120℃for 6 hours. The reaction was concentrated in vacuo and purified by silica gel chromatography (10-100%EtOAc/isohexane) to give the title compound (620 mg,2.93mmol,21% yield) as a viscous yellow oil; 1 H NMR(500MHz,DMSO-d 6 )δ8.57(dd,J=1.7,0.9Hz,1H),8.08(d,J=1.1Hz,1H),7.53(d,J=9.6Hz,1H),6.92–6.73(m,2H),3.80(s,3H);M/Z:210,212[M+H] + ,ESI + ,RT=0.92(S2)。
intermediate 12 (step 8. B): 6-chloroindolizine-2-carboxylic acid
6-chloroindolizine-2-carboxylic acid methyl ester (0.62 g,2.96 mmol) and LiOH H 2 O (0.25 g,5.92 mmol) in EtOH (2 mL): THF (2 mL): h 2 The mixture in O (2 mL) was stirred at room temperature for 12 hours. H for the reaction mixture 2 O (10 mL) and EtOAc (10 mL) were diluted and the aqueous layer was acidified with 1M aqueous HCl. The aqueous solution was extracted with EtOAc, washed with brine, and dried over MgSO 4 Drying and concentration in vacuo gave the title compound (380 mg,1.90mmol,64% yield) as a white solid; 1 H NMR(500MHz,DMSO-d 6 )δ12.44(s,1H),8.56(dt,J=1.8,0.9Hz,1H),8.01(d,J=1.0Hz,1H),7.51(d,J=9.6Hz,1H),6.99–6.63(m,2H);M/Z:196,198[M+H] + ,ESI + ,RT=0.74(S2)。
the example intermediates in table 2 were synthesized according to general route 8 as exemplified by intermediate 12 using the corresponding starting materials.
TABLE 2
Scheme of route 9
Step 9.a: 2-amino-4-chloro-3-fluoro-benzaldehyde
Tert-butyl N- (3-chloro-2-fluoro-6-formyl-phenyl) carbamate (1.23 g,4.45 mmol) was added to a solution of 4M HCl/1, 4-dioxane (4.5 mL,18.0 mmol) in anhydrous 1, 4-dioxane (10 mL) and the mixture stirred at room temperature for 3 hours. The reaction mixture was concentrated in vacuo and purified by silica gel chromatography (0-20% etoac/heptane) to give the title compound (326 mg,1.78mmol,40% yield); 1 H NMR(400MHz,DMSO-d 6 )δ9.87(d,J=2.0Hz,1H),7.48(dd,J=8.5,1.6Hz,1H),7.24(s,2H),6.84–6.77(m,1H);M/Z:174,176[M+H] + ,ESI+,RT=0.78(S2)。
step 9.b: 7-chloro-8-fluoro-quinoline-3-carboxylic acid ethyl ester
To a mixture of 2-amino-4-chloro-3-fluoro-benzaldehyde (326 mg,1.78 mmol) and ethyl 3, 3-diethoxypropionate (850 mg,4.47 mmol) in toluene (4 mL) was added TsOH (31 mg,0.180 mmol) and the mixture was stirred in a sealed tube at 120 ℃ for 4h. The reaction mixture was cooled to room temperature and concentrated in vacuo. The residue was dissolved in EtOAc (30 mL) and treated with saturated NaHCO 3 Aqueous solution (10 mL) and H 2 O (10 mL) was washed. The combined organic extracts were purified over Na 2 SO 4 Dried and concentrated in vacuo. The residue was triturated using ACN to give the title compound (249 mg,0.982mmol,55% yield) as an off-white powder; 1 H NMR(500MHz,DMSO-d 6 )δ9.40(d,J=2.0Hz,1H),9.23–9.09(m,1H),8.15(dd,J=8.9,1.4Hz,1H),7.90(dd,J=8.9,6.8Hz,1H),4.45(q,J=7.1Hz,2H),1.40(t,J=7.1Hz,3H);M/Z:254,256[M+H] + ,ESI + ,RT=3.37(S4)。
intermediate 14 (step 9.c): 7-chloro-8-fluoro-quinoline-3-carboxylic acid
To 7-chloro-8-fluoro-quinoline-3-carboxylic acid ethyl ester (249 mg,0.982 mmol) in THF (3 mL) and H 2 LiOH H was added to the solution in O (3 mL) 2 O (49 mg,1.18 mmol) and the mixture was stirred at room temperature for 5 hours. The reaction mixture was concentrated in vacuo and redissolved in H 2 O (10 mL). The aqueous solution was acidified to pH2-3 using 4M aqueous HCl and extracted with EtOAc (2X 50 mL). The combined organic extracts were purified over Na 2 SO 4 Drying and concentration in vacuo gave the title compound (206 mg, 0.284 mmol,92% yield) as an off-white powder; 1 H NMR(500MHz,DMSO-d 6 )δ13.81(s,1H),9.39(d,J=2.0Hz,1H),9.15–9.08(m,1H),8.12(dd,J=9.0,1.4Hz,1H),7.89(dd,J=8.9,6.8Hz,1H);M/Z:226,228[M+H] + ,ESI + ,RT=2.40(S4)。
scheme of route 10
/>
Step 10.A: (2R, 5S) -5- (7-chloroquine-3-amido) piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
To a solution of 7-chloroquinoline-3-carboxylic acid (3.39 g,16.3 mmol) in anhydrous THF (90 mL) at 0deg.C was added NMM (1.9 mL,17.2 mmol), followed by dropwise isobutyl chloroformate (2.1 mL,16.3 mmol) and the mixture was stirred at room temperature for 25 min. A solution of (2R, 5S) -5-aminopiperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (4.45 g,16.3mmol, intermediate 7) in anhydrous THF (90 mL) was added and the mixture stirred at room temperature for 48 hours. T3P (50%/EtOAc, 2.4mL,4.08 mmol) was added and the mixture was stirred at room temperature for 1 hour. The reaction mixture was treated with H 2 O (100 mL) and EtOAc (100 mL) were diluted, and the organic layer was separated, washed with saturated NaHCO 3 Aqueous (100 mL) wash with Na 2 SO 4 Drying and concentration in vacuo gave the title compound (92% purity, 8.09g,16.1mmol,99% yield) as a yellow viscous gel; 1 H NMR(500MHz,DMSO-d 6 )δ9.25(s,1H),8.84(s,1H),8.65(s,1H),8.17(d,J=8.9Hz,2H),7.74(dd,J=8.7,2.1Hz,1H),4.83–4.55(m,1H),4.24–4.08(m,2H),3.15(dd,J=6.4,5.3Hz,1H),2.26(s,1H),1.95(s,1H),1.83–1.73(m,1H),1.60(tt,J=13.3,6.7Hz,1H),1.39–1.24(m,11H),1.18(q,J=7.1Hz,3H);M/Z:462,464
[M+H] + ,ESI + ,RT=0.98(S2)。
intermediate 16 (step 10. B): (2R, 5S) -1- [ (tert-Butoxycarbonyl) -5- (7-chloroquinoline-3-amid-yl) piperidine-2-carboxylic acid
To a solution of (2 r,5 s) -5- (7-chloroquine-3-amido) piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (92% purity, 8.09g,16.1 mmol) in THF (120 mL) was added 2M aqueous LiOH (16 mL,32.2 mmol) and the mixture stirred at room temperature for 2h. An additional 2M aqueous LiOH (16 ml,32.2 mmol) was added and the mixture was stirred at room temperature overnight. An additional 2M aqueous LiOH (16 ml,32.2 mmol) was added and the mixture was stirred at room temperature overnight. Using 2M KHSO 4 Aqueous solution (32 mL,64.4 mmol) and H 2 O (50 mL) quenched the reaction mixture, then stirred at room temperature for 10 minutes, then concentrated in vacuo. EtOAc (100 mL) was added and the organic layer was separated using H 2 Washing with Na 2 SO 4 Dried and concentrated in vacuo. The residue was dissolved in TBME (75 mL), the resulting suspension was filtered, washed with TBME and then dried in vacuo to give the title compound (4.19 g,9.66mmol,60% yield) as a white powder; 1 H NMR(500MHz,DMSO-d 6 )δ12.86(s,1H),9.24(s,1H),8.83(s,1H),8.60(s,1H),8.16(d,J=8.8Hz,2H),7.73(dd,J=8.8,2.0Hz,1H),4.68(s,1H),4.02(q,J=7.1Hz,2H),3.15(d,J=13.6Hz,1H),2.23(s,1H),1.93(d,J=26.7Hz,1H),1.76(d,J=13.1Hz,1H),1.65(s,1H),1.30(d,J=51.9Hz,9H);M/Z:434,436[M+H] + ,ESI + ,RT=0.82(S2)。
the intermediate compounds in table 3 were synthesized according to general route 10 as exemplified by intermediate 16 using the corresponding intermediates.
TABLE 3 Table 3
Scheme of route 11
Step 11.A: (2S, 5R) -5- (3, 4-Dichloroformamido) piperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester
To a solution of 3, 4-dichlorobenzoic acid (0.70 g,3.67 mmol) in DMF (14.284 mL) was added HATU (1.68 g,4.41 mmol) and DIPEA (1.3 mL,7.34 mmol) at 0deg.C and stirred for 10 min. (2S, 5R) -5-aminopiperidine-1, 2-dicarboxylic acid 1-tert-butyl 2-ethyl ester (1.00 g,3.67 mmol) was added, and the mixture was stirred at room temperature for 4 hours. The reaction mixture was diluted with EtOAc (40 mL) and with H 2 O (3X 20 mL) was washed. MgSO for organic layer 4 Dried, concentrated in vacuo and purified by silica gel chromatography (10-100% etoac/heptane) to give the title compound (1.58 g,3.50mmol,95% yield) as a yellow oil; 1 H NMR(400MHz,CDCl 3 )δ7.83(s,1H),7.62–7.47(m,2H),6.44(d,J=94.0Hz,1H),4.86(d,J=79.2Hz,1H),4.32–4.17(m,3H),4.15–4.00(m,1H),3.38–3.20(m,1H),2.27–1.84(m,3H),1.61(s,1H),1.47(s,9H),1.30(t,J=7.1Hz,3H);M/Z:467,469,471[M+Na] + ,ESI + ,RT=1.06(S2)。
intermediate 18 (step 11. B): (2S, 5R) -1-tert-Butoxycarbonyl-5- [ (3, 4-dichlorobenzoyl) amino ] piperidine-2-carboxylic acid
To a solution of 1-tert-butyl 2-ethyl (2S, 5R) -5- (3, 4-dichlorobenzamido) piperidine-1, 2-dicarboxylic acid (1.58 g,3.50 mmol) in EtOH (10 mL) and THF (10 mL) was added 2M LiOH (1.8 mL,3.50 mmol) and the mixture was stirred at room temperature overnight. The reaction mixture was acidified to pH2 using 1M aqueous HCl and extracted with EtOAc (2×50 mL). The combined organic extracts were dried over MgSO 4 Drying and concentration in vacuo gave the title compound (92% purity, 1.55g,3.42mmol,97% yield) as a white solid; 1 H NMR(400MHz,CDCl 3 )δ8.39–7.65(m,2H),7.67–7.40(m,2H),6.76–6.35(m,1H),5.08–4.76(m,1H),4.36–4.22(m,1H),4.12–4.01(m,1H),3.38–3.19(m,1H),2.25–2.09(m,2H),1.94(ddq,J=14.3,10.2,5.1,3.9Hz,1H),1.69(s,1H),1.45(s,9H);M/Z:439,441,443[M+Na] + ,ESI + ,RT=0.90(S2)。
scheme of route 12
Example 1 (step 12. A): (2R, 5S) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethyl) quinoline-2-amid-yl ] piperidine-1-carboxylic acid tert-butyl ester
To a solution of 6- (trifluoromethyl) quinoline-2-carboxylic acid (47 mg,0.194 mmol) in THF (0.5 mL) was added T3P (50%/EtOAc, 0.14mL,0.233 mmol) and DIPEA (41. Mu.L, 0.233 mmol) and the mixture was stirred at room temperature for 10 min. (2R, 5S) -5-amino-2- {5- [2- (trifluoromethoxy) ethoxy ] added in THF (1.5 mL)]-tert-butyl 1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (49% purity, 120mg,0.148mmol, intermediate 10) and the mixture was stirred at room temperature for 4 hours. The reaction mixture was taken up with saturated NaHCO 3 The aqueous solution (2 mL) was diluted and extracted with DCM (2X 5 mL). The combined organic extracts were dried using a phase separator and concentrated in vacuo to afford the title compound, estimated to be quantitative yield (67% pureDegree, 178 mg,0.194 mmol) as orange gum. The crude material was used in the next step without further purification.
Example 2 (step 12. B): n- [ (3S, 6R) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -6- (trifluoromethyl) quinoline-2-carboxamide
To (2R, 5S) -2- {5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethyl) quinolin-2-amido]A solution of tert-butyl piperidine-1-carboxylate (67% purity, 178 mg,0.194mmol, example 1) in DCM (5 mL) was added ZnBr 2 (131 mg,0.580 mmol) and the mixture was stirred at room temperature for 16 h. Adding another part of ZnBr 2 And stirred at room temperature for 3 hours. The reaction mixture was taken up with saturated NaHCO 3 Aqueous solution (2 mL) was diluted and extracted with DCM: IPA (4:1, 2X 2 mL). The combined organic extracts were dried using a phase separator, concentrated in vacuo, and purified by silica gel chromatography (0-100% EtOAc/heptane, then 0-20% MeOH/EtOAc). The residue was purified by preparative HPLC (method 6) to give the title compound (15.3 mg,0.0286mmol,15% yield) as a white powder; 1 H NMR(400MHz,DMSO-d 6 )δ8.80(s,1H),8.78(s,1H),8.65(s,1H),8.37(d,J=8.9Hz,1H),8.29(d,J=8.5Hz,1H),8.13(dd,J=9.0,2.1Hz,1H),4.74–4.66(m,2H),4.53–4.45(m,2H),4.05–3.92(m,1H),3.91–3.82(m,1H),3.15–3.06(m,1H),2.97–2.86(m,1H),2.77–2.65(m,1H),2.09–1.98(m,2H),1.87–1.67(m,2H);M/Z:520[M+H] + ,ESI + ,RT=2.46(S4)。
the example compounds in table 4 were synthesized according to general route 12 as exemplified in example 2 using the corresponding intermediates. The corresponding boc protected intermediates of the numbered examples are also embodiments of the present invention.
TABLE 4 Table 4
/>
/>
/>
Scheme of route 13
Example 7 (step 13. A): (2R, 5S) -5- (6-chloroquinoline-3-carboxamide) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester
To a solution of 6-chloroquinoline-3-carboxylic acid (63 mg,0.303 mmol) in anhydrous DMF (3 mL) was added HATU (138 mg, 0.264 mmol), followed by DIPEA (106. Mu.L, 0.607 mmol) and stirred at room temperature for 10 min. Addition of (2R, 5S) -5-amino-2- {5- [2- (trifluoromethoxy) ethoxy } -]-tert-butyl 1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (120 mg,0.303mmol, intermediate 10) and the mixture was stirred at room temperature for 19 hours. Another portion of HATU (35 mg,0.091 mg) was added to the solution, and the mixture was stirred at room temperature for 1h. The reaction mixture was partitioned between EtOAc (10 mL) and 1M aqueous HCl (10 mL). The organic layer was separated with saturated NaHCO 3 Aqueous (10 mL) and brine (10 mL) were washed with MgSO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (20-100% etoac/heptane) to give the title compound (94% purity, 100mg,0.160mmol,53% yield) as an off-white powder; 1 H NMR(400MHz,DMSO-d 6 )δ9.28–9.20(m,1H),8.82–8.75(m,1H),8.68(d,J=6.2Hz,1H),8.29–8.21(m,1H),8.15–8.08(m,1H),7.87(dd,J=9.0,2.4Hz,1H),5.50–5.38(m,1H),4.75–4.62(m,2H),4.55–4.42(m,2H),4.26–4.13(m,1H),4.12–4.05(m,1H),3.03(dd,J=14.0,2.4Hz,1H),2.07–1.89(m,3H),1.86–1.76(m,1H),1.23(s,9H);M/Z:486,488[M-Boc+H] + ,ESI + ,RT=1.01(S2)。
example 8 (step 13. B): 6-chloro-N- [ (3S, 6R) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide
To (2R, 5S) -5- (6-chloroquinolin-3-ylamino) -2- {5- [2- (trifluoromethoxy) ethoxy]A solution of tert-butyl-1, 3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (94% purity, 98mg,0.157mmol, example 7) in DCM (4 mL) was added ZnBr 2 (106 mg, 0.470 mmol) and the mixture was taken up in N 2 Stirred at room temperature for 20 hours. The reaction mixture was treated with H 2 O (3 mL) was diluted and extracted with DCM/IPA (9:1, 3X 3 mL). The combined organics were dried using a phase separator, concentrated in vacuo and purified by preparative HPLC (method 2). The product-containing fractions were combined using saturated NaHCO 3 The solution was basified to pH9 and extracted with EtOAc (3X 25 mL). The combined organic extracts were purified over Na 2 SO 4 Drying and concentration in vacuo gave the title compound (17 mg,0.0347mmol,22% yield) as a white powder; 1 H NMR(400MHz,DMSO-d 6 )δ9.27(d,J=2.2Hz,1H),8.78(d,J=2.0Hz,1H),8.64(d,J=7.8Hz,1H),8.24(d,J=2.4Hz,1H),8.14–8.07(m,1H),7.87(dd,J=9.0,2.4Hz,1H),4.73–4.64(m,2H),4.52–4.43(m,2H),3.98–3.87(m,1H),3.85–3.77(m,1H),3.18–3.10(m,1H),2.90–2.82(m,1H),2.61–2.53(m,1H),2.10–1.96(m,2H),1.79–1.55(m,2H);M/Z:486,488[M+H] + ,ESI + ,RT=1.98(S4)。
the example compounds in table 5 were synthesized according to the general route 13 as exemplified in example 8 using the corresponding intermediates. The corresponding boc protected intermediates of the numbered examples are also embodiments of the present invention.
TABLE 5
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
/>
Scheme of route 14
Example 28 (step 14. A): (2R, 5S) -5- [4- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester
To a solution of 4- (trifluoromethoxy) benzoic acid (44 mg,0.182 mmol) in anhydrous THF (4 mL) was added DIPEA (95. Mu.L, 0.545 mmol), HATU (83 mg,0.218 mmol) and (2R, 5S) -5-amino-2- {5- [2- (trifluoromethoxy) ethoxy]-tert-butyl 1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (90% purity, 80mg,0.182mmol, intermediate 10) and the mixture was stirred at room temperature for 16 hours. The reaction mixture was taken up with saturated NaHCO 3 Aqueous (5 mL) was diluted and extracted with EtOAc (2X 5 mL). The combined organic extracts were purified over Na 2 SO 4 Drying and concentration in vacuo gave the title compound (65% purity, 160mg,0.178mmol,98% yield) as a yellow oil; M/Z485 [ M-Boc+H ]] + ,ESI + ,RT=1.04(S2)。
Example 29 (step 14. B): 4- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide
To (2R, 5S) -5- [4- (trifluoromethoxy)Benzamido groups]-2- {5- [2- (trifluoromethoxy) ethoxy ]]A solution of tert-butyl-1, 3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (65% purity, 160mg,0.178mmol, example 28) in anhydrous DCM (5 mL) was added ZnBr 2 (109 mg,0.484 mmol) and the mixture was stirred vigorously at room temperature for 16h. The reaction mixture was taken up with DCM/IPA (2:1, 10 mL) and H 2 O (5 mL) dilution, separation of organic layer, na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 3) followed by preparative HPLC (method 1). The relevant fraction was saturated with NaHCO 3 The aqueous solution (5 mL) was diluted and extracted with EtOAc (2X 25 mL). The combined organic extracts were purified over Na 2 SO 4 Dried and concentrated in vacuo. Dissolving the residue in ACN/H 2 O (1:1, 2 mL) and freeze-drying to give the title compound (6.0 mg, 0.01200 mmol,7.4% yield) as a white powder; 1 H NMR(500MHz,MeOD)δ8.01–7.85(m,2H),7.38(d,J=8.1Hz,2H),4.76–4.70(m,2H),4.66–4.54(m,1H),4.48–4.40(m,2H),4.12–4.01(m,1H),3.97–3.87(m,1H),3.34–3.33(m,1H),2.73–2.62(m,1H),2.24–2.14(m,2H),1.97–1.83(m,1H),1.77–1.63(m,1H);M/Z:485[M+H] + ,ESI + ,RT=2.15(S4)。
The example compounds in table 6 were synthesized according to the general route 14 as exemplified in example 29 using the corresponding intermediates. The corresponding boc protected intermediates of the numbered examples are also embodiments of the present invention.
TABLE 6
/>
/>
/>
/>
/>
Scheme of route 15
Step 15.A: 1-methyl-6- (trifluoromethyl) indole-2-carboxylic acid
To a solution of ethyl 6- (trifluoromethyl) -1H-indole-2-carboxylate (100 mg,0.389 mmol) in anhydrous DMF (4 mL) was added KOH powder (128 mg,1.94 mmol) followed by MeI (48. Mu.L, 0.778 mmol) and the mixture was stirred at room temperature for 20 hours. The reaction mixture was taken up in H 2 The layers were partitioned between O (25 mL) and DCM (25 mL) and separated. The organic layer was discarded and the aqueous layer was acidified to pH2/3 by slow dropwise addition of 1M aqueous HCl at 0deg.C. The aqueous layer was then extracted with EtOAc (2X 20 mL) and the combined organics were taken with H 2 O (20 mL) washing with Na 2 SO 4 Dried and concentrated in vacuo to give the title compound (85 mg, 0.345 mmol,88% yield) as an orange solid; 1 H NMR(400MHz,DMSO-d 6 )δ8.02(s,1H),7.89(d,J=8.4Hz,1H),7.39(dd,J=8.4,1.3Hz,1H),7.32(d,J=0.7Hz,1H),4.11(s,3H);M/Z:242[M-H] - ,ESI - ,RT=1.20(S3)。
example 41 (step 15. B): (2R, 5S) -5- [ [ 1-methyl-6- (trifluoromethyl) indole-2-carbonyl ] amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester
Isobutyl chloroformate (42. Mu.L, 0.325 mmol) and NMM (38. Mu.L, 0.343 mmol) were added to a solution of 1-methyl-6- (trifluoromethyl) indole-2-carboxylic acid (85 mg, 0.343mmol) in anhydrous 2-MeTHF (2 mL) at 0deg.C and the mixture was stirred for 15 min. (2R, 5S) -5-amino-2- {5- [2- (trifluoromethoxy) ethoxy ] was added dropwise ]A solution of tert-butyl-1, 3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (141 mg,0.343mmol, intermediate 10) in anhydrous 2-MeTHF (2 mL) and the mixture stirred at room temperature for 3h. The reaction mixture was cooled to 0℃and taken up in H 2 O (5 mL) quench. The layers were separated and the aqueous layer was further extracted with EtOAc (5 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous solution and brine wash with Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (30-100% etoac/heptane) to give the title compound (38 mg,0.0581mmol,17% yield) as a colorless oil; 1 H NMR(400MHz,CDCl 3 )δ7.76–7.65(m,2H),7.38(d,J=8.1Hz,1H),6.86(s,1H),6.76–6.33(m,1H),5.73–5.23(m,1H),4.75–4.66(m,2H),4.38–4.32(m,2H),4.31–4.26(m,1H),4.26–4.19(m,1H),4.10(s,3H),3.38–2.94(m,1H),2.29–2.18(m,1H),2.18–1.99(m,3H),1.54–1.40(m,9H);M/Z:644[M+Na] + ,ESI + ,RT=1.43(S1)。
example 42 (step 15. C): 1-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] -6- (trifluoromethyl) indole-2-carboxamide
To (2R, 5S) -5- [ [ 1-methyl-6- (trifluoromethyl) indole-2-carbonyl]Amino group]-2- [5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl]A solution of tert-butyl piperidine-1-carboxylate (38 mg,0.0581mmol, example 41) in anhydrous DCM (1 mL) was added ZnBr 2 (52 mg,0.232 mmol) and the mixture was stirred at room temperatureMix for 6 hours. The reaction mixture was taken up with saturated NaHCO 3 The aqueous solution was diluted and extracted twice with DCM: IPA (4:1). The combined organic extracts were concentrated in vacuo and purified by preparative HPLC (method 2). The relevant fractions were pooled and saturated NaHCO was used 3 The aqueous solution was basified to pH9 and extracted with EtOAc (2X 10 mL). The combined organics were washed with brine (10 mL), dried over Na 2 SO 4 Drying and concentration in vacuo gave the title compound (6.0 mg,0.0115mmol,20% yield) as a white powder; 1 H NMR(400MHz,DMSO-d 6 )δ8.44(d,J=8.0Hz,1H),7.96(s,1H),7.85(d,J=8.4Hz,1H),7.38(dd,J=8.4,1.3Hz,1H),7.19(s,1H),4.73–4.64(m,2H),4.53–4.41(m,2H),4.05(s,3H),3.93–3.82(m,1H),3.82–3.74(m,1H),3.14–3.06(m,1H),2.89–2.79(m,1H),2.53–2.52(m,1H),2.05–1.96(m,2H),1.76–1.54(m,2H);M/Z:522[M+H] + ,ESI + ,RT=2.52(S4)。
scheme of route 16
Example 43 (step 16. A): (2R, 5S) -5- [ (3-chloro-4-methyl-benzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester
To (2R, 5S) -5-amino-2- [5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl]A solution of piperidine-1-carboxylic acid tert-butyl ester (130 mg, 0.025 mmol, intermediate 10), 3-chloro-4-methyl-benzoic acid (50 mg, 0.025 mmol) and NMI (75 mg, 0.910 mmol) in anhydrous ACN (3 mL) was added TCFH (91 mg,0.325 mmol) and the mixture stirred at room temperature for 2h. H for the reaction mixture 2 O (15 mL) was diluted and the aqueous layer was extracted with EtOAc (2X 15 mL). The combined organic extracts were washed with brine, and with MgSO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (5-100% etoac/heptane) to give the title compound (91% purity, 75mg, 0.12)4mmol,42% yield) as colorless oil; M/Z549,551 [ M+H ]] + ,ESI + ,RT=1.02(S2)。
Example 44 (step 16. B): 3-chloro-4-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide
To (2R, 5S) -5- [ (3-chloro-4-methyl-benzoyl) amino group]-2- [5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl]A stirred solution of tert-butyl piperidine-1-carboxylate (75 mg,0.124mmol, example 43) in anhydrous DCM (4 mL) was added ZnBr 2 (84 mg,0.373 mmol) and the mixture was stirred at room temperature overnight. H for the reaction mixture 2 O (15 mL) was diluted and the aqueous layer was extracted with DCM (3X 15 mL). The combined organic layers were dried over MgSO 4 Dried, concentrated in vacuo, and purified by preparative HPLC (method 3) to give the title compound (9.2 mg,0.0203mmol,16% yield) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ8.27(d,J=7.8Hz,1H),7.90(d,J=1.7Hz,1H),7.73(dd,J=7.9,1.7Hz,1H),7.44(d,J=8.2Hz,1H),4.71–4.66(m,2H),4.50–4.45(m,2H),3.90–3.72(m,2H),3.07(d,J=12.3Hz,1H),2.85–2.77(m,1H),2.37(s,3H),2.04–1.95(m,2H),1.74–1.51(m,2H);M/Z:449,451
[M+H] + ,ESI + ,RT=1.97(S4)。
scheme of route 17
Step 17.A: (2R, 5S) -2- [ [ (4-chlorobenzoyl) amino ] carbamoyl ] -5- [ (7-chloroquinoline-3-carbonyl) amino ] piperidine-1-carboxylic acid tert-butyl ester
(2R, 5S) -1-tert-butylOxycarbonyl-5- [ (7-chloroquinoline-3-carbonyl) amino group]A solution of piperidine-2-carboxylic acid (350 mg, 0.803 mmol, intermediate 16) in anhydrous 2-MeTHF (9 mL) was cooled to 0deg.C and treated with isobutyl chloroformate (99 μL,0.766 mmol) and NMM (89 μL,0.807 mmol). The mixture was stirred for 15 minutes, then 4-chlorobenzoyl hydrazine (138 mg, 0.803 mmol) was added and stirred at room temperature for 1 hour. The reaction mixture was cooled to 0℃and then taken up in H 2 O (1 mL) quench. The solution was dissolved in EtOAc (20 mL) and H 2 O (20 mL) was partitioned between and the layers separated. The aqueous layer was again extracted with EtOAc (10 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous (20 mL) and brine (20 mL) were washed with Na 2 SO 4 Drying and concentration in vacuo gave the title compound (450 mg,0.729mmol,90% yield) as an off-white powder; 1 H NMR(400MHz,CDCl 3 )δ9.24(d,J=2.1Hz,1H),8.88–8.72(m,2H),8.55(d,J=1.8Hz,1H),8.14(d,J=1.7Hz,1H),7.83(d,J=8.7Hz,1H),7.79–7.73(m,2H),7.58(dd,J=8.7,2.0Hz,1H),7.46–7.39(m,2H),4.98(s,1H),4.43–4.25(m,2H),3.52–3.37(m,1H),2.35–2.18(m,1H),2.12–1.99(m,2H),1.99–1.86(m,2H),1.58–1.34(m,9H);M/Z:586,588,590[M+H] + ,ESI + ,RT=1.20(S1)。
example 45 (step 17. B): (2R, 5S) -2- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -5- [ (7-chloroquinoline-3-carbonyl) amino ] piperidine-1-carboxylic acid tert-butyl ester
/>
(2R, 5S) -2- [ [ (4-chlorobenzoyl) amino group]Carbamoyl radicals]-5- [ (7-chloroquinoline-3-carbonyl) amino group]Piperidine-1-carboxylic acid tert-butyl ester (450 mg,0.729 mmol), tsCl (417 mg,2.19 mmol) and K 2 CO 3 A solution of (604 mg,4.37 mmol) in anhydrous ACN (8 mL) was stirred at 80deg.C for 1h. The reaction mixture was cooled to room temperature and taken up with H 2 O (10 mL) quench. The aqueous layer was extracted with EtOAc (2X 20 mL) and the combined organic extracts were extracted with saturated NaHCO 3 Aqueous (3X 20 mL) and brine (20 mL) washed with Na 2 SO 4 Dried and concentrated in vacuo. By chromatography on silica gel (40-100% EtThe residue was purified on OAc/heptane to give the title compound (327 mg,0.546mmol,75% yield) as an off-white solid; 1 H NMR(400MHz,CDCl 3 )δ9.26(d,J=2.2Hz,1H),8.57(d,J=1.7Hz,1H),8.18(d,J=1.8Hz,1H),8.02–7.95(m,2H),7.87(d,J=8.7Hz,1H),7.60(dd,J=8.7,2.0Hz,1H),7.54–7.46(m,2H),6.96–6.42(m,1H),5.96–5.51(m,1H),4.42–4.34(m,1H),4.34–4.24(m,1H),3.44–3.23(m,1H),2.36(d,J=13.2Hz,1H),2.31–2.16(m,2H),2.15–2.05(m,1H),1.51(s,9H);M/Z:568[M+H] + ,ESI + ,RT=1.38(S1)。
example 46 (step 17. C): 7-chloro-N- [ (3 s,6 r) -6- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide
To (2 r,5 s) -2- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl]-5- [ (7-chloroquinoline-3-carbonyl) amino group]A solution of tert-butyl piperidine-1-carboxylate (327 mg,0.546mmol, example 45) in DCM (5 mL) was added TFA (1 mL) and the mixture stirred at room temperature for 1h. The reaction mixture was then added dropwise to saturated NaHCO at 0 ° 3 Quench on aqueous solution (10 mL). The layers were separated and the aqueous layer was extracted with DCM (10 mL). The combined organic layers were saturated with NaHCO 3 Aqueous (20 mL) and brine (20 mL) washed with Na 2 SO 4 Dried and concentrated in vacuo. The crude product was purified by preparative HPLC (method 5) to give the title compound (109 mg,0.233mmol,43% yield) as an off-white powder; 1 H NMR(400MHz,CDCl 3 )δ9.31(d,J=1.9Hz,1H),8.63(s,1H),8.19(s,1H),8.08–7.98(m,2H),7.90(d,J=8.7Hz,1H),7.62(dd,J=8.7,1.8Hz,1H),7.57–7.47(m,2H),6.74(d,J=7.3Hz,1H),4.40–4.26(m,2H),3.52(dd,J=12.0,3.0Hz,1H),2.86(dd,J=12.0,6.9Hz,1H),2.39–2.28(m,1H),2.28–2.11(m,2H),1.95–1.83(m,1H);M/Z:468,470,472[M+H] + ,ESI + ,RT=2.10(S4)。
scheme of route 18
/>
Step 18.A: (2R, 5S) -5- [ (7-chloroquinoline-3-carbonyl) amino ] -2- [ [ [ (E) -4, 4-trifluoro-but-2-yloxy ] carbonylamino ] carbamoyl ] piperidine-1-carboxylic acid tert-butyl ester
(2R, 5S) -1-tert-Butoxycarbonyl-5- [ (7-chloroquinoline-3-carbonyl) amino at 0 ℃C]A solution of piperidine-2-carboxylic acid (410 mg,0.945mmol, intermediate 16) in 2-MeTHF (10 mL) was prepared using [ (E) -4, 4-trifluoro-but-2-enyl ]]N-carbamic acid ester (174 mg,0.945mmol, intermediate 3) and isobutyl chloroformate (0.12 mL,0.898 mmol). The reaction was stirred for 15 minutes at which time a solution of NMM (0.10 mL,0.945 mmol) in 2-MeTHF (10 mL) was added. The reaction was warmed to room temperature and stirred for 1 hour. By H 2 O (10 mL) was slowly quenched and the aqueous layer extracted with EtOAc (2X 10 mL). The combined organic layers were saturated with NaHCO 3 Aqueous (2X 10 mL) and brine (10 mL) were washed with Na 2 SO 4 Drying and vacuum concentration gave the title compound (90% purity, 600mg,0.900mmol,95% yield); 1 H NMR(400MHz,CDCl 3 )δ9.17(d,J=2.1Hz,1H),8.57(s,1H),8.51(s,1H),8.02(s,1H),7.75(d,J=8.8Hz,1H),7.58(s,1H),7.50(dd,J=8.7,1.9Hz,1H),7.17–6.92(m,1H),6.37(d,J=15.3Hz,1H),5.93–5.78(m,1H),4.95–4.81(m,1H),4.77–4.62(m,2H),4.34–4.22(m,2H),3.51–3.38(m,1H),2.22–2.11(m,1H),1.99–1.80(m,3H),1.49–1.27(m,9H);M/Z:600,602[M+H] + ,ESI + ,RT=3.33(S4)。
example 47 (step 18. B): (2R, 5S) -5- [ (7-chloroquinoline-3-carbonyl) amino ] -2- [5- [ (E) -4, 4-trifluoro-but-2-enoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester
(2R, 5S) -5- [ (7-chloroquinoline-3-carbonyl)Amino group]-2- [ [ [ (E) -4, 4-trifluoro-but-2-enoxy group]Carbonylamino group]Carbamoyl radicals]Tert-butyl piperidine-1-carboxylate (90% purity, 505mg,0.758 mmol), tsCl (217 mg,1.14 mmol) and K 3 PO 4 A solution of (480 mg,2.27 mmol) in anhydrous ACN (15 mL) was stirred at 60℃for 4 h. The reaction was cooled to 0deg.C and saturated NH was used 4 Aqueous OH (1 mL) was quenched slowly. The solution was treated with H 2 O (40 mL) was diluted and extracted with EtOAc (2X 40 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous (20 mL) and brine (20 mL) were washed with Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 5) and the relevant fractions were purified with saturated NaHCO 3 The aqueous solution was neutralized and extracted with EtOAc (2X 30 mL). The combined organic extracts were washed with brine (20 mL), and dried over Na 2 SO 4 Dried and concentrated in vacuo to afford the title compound (132 mg,0.218mmol,29% yield); 1 H NMR(400MHz,CDCl 3 )δ9.23(d,J=2.1Hz,1H),8.53(d,J=1.7Hz,1H),8.13(d,J=1.7Hz,1H),7.82(d,J=8.7Hz,1H),7.57(dd,J=8.7,2.0Hz,1H),7.08–6.65(m,1H),6.62–6.50(m,1H),6.16–6.01(m,1H),5.75–5.34(m,1H),5.18–5.03(m,2H),4.42–4.33(m,1H),4.32–4.22(m,1H),3.36–3.13(m,1H),2.30–1.94(m,4H),1.48(s,9H);M/Z:582,584[M+H] + ,ESI + ,RT=3.81(S4)。
example 48 (step 18. C): 7-chloro-N- [ (3 s,6 r) -6- [5- [ (E) -4, 4-trifluoro-but-2-enoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide
To (2R, 5S) -5- [ (7-chloroquinoline-3-carbonyl) amino group]-2- [5- [ (E) -4, 4-trifluoro-but-2-enoxy]-1,3, 4-oxadiazol-2-yl]A solution of tert-butyl piperidine-1-carboxylate (150 mg,0.247mmol, example 47) in anhydrous DCM (3 mL) was added ZnBr 2 (223 mg,0.990 mmol) and the mixture was stirred at room temperature for 3 hours. Additional anhydrous DCM (3 mL) and ZnBr were added 2 (223 mg,0.990 mmol) and the mixture was stirred at room temperature for 6h. The reaction mixture was taken up with saturated NaHCO 3 The aqueous solution (10 mL) was quenched and extracted with DCM: IPA (4:1) (2X 10 mL). The combined organic extracts were washed with brine (10 mL), and with Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 5) and the relevant fractions were purified with saturated NaHCO 3 The aqueous solution was neutralized, combined and extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with brine (10 mL), and dried over Na 2 SO 4 Dried and concentrated in vacuo to afford the title compound (17 mg,0.0346mmol,14% yield) as an off-white powder; 1 H NMR(400MHz,CDCl 3 )δ9.27(d,J=2.1Hz,1H),8.59(d,J=1.9Hz,1H),8.17(s,1H),7.87(d,J=8.7Hz,1H),7.59(dd,J=8.7,2.0Hz,1H),6.70(d,J=6.7Hz,1H),6.62–6.49(m,1H),6.14–5.99(m,1H),5.15–5.06(m,2H),4.37–4.21(m,1H),4.18–4.06(m,1H),3.42(dd,J=12.1,3.1Hz,1H),2.79(dd,J=12.2,6.7Hz,1H),2.26–2.10(m,2H),2.09–2.01(m,1H),1.89–1.75(m,1H);M/Z:482,484[M+H] + ,ESI + ,RT=3.00(S6)。
The example compounds in table 7 were synthesized according to the general route 18 as exemplified in example 48 using the corresponding intermediates. The corresponding boc protected intermediates of the numbered examples are also embodiments of the present invention.
TABLE 7
Scheme of route 19
Step 19.A: (2R, 5S) -5- (7-chloroquine-3-amido) -2- { N' - [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutanecarbonyl ] hydrazinecarbonyl } piperidine-1-carboxylic acid tert-butyl ester
To (1 s,3 s) -3- (trifluoromethoxy) cyclobutane-1-carbohydrazide (237 mg,1.20mmol, intermediate 5), (2R, 5S) -1-tert-butoxycarbonyl-5- [ (7-chloroquinoline-3-carbonyl) amino]A solution of piperidine-2-carboxylic acid (500 mg,1.14mmol, intermediate 16) and DIPEA (0.60 mL,3.42 mmol) in anhydrous DMF (10 mL) was added HATU (521 mg,1.37 mmol) and the mixture stirred at room temperature overnight. The reaction mixture was treated with H 2 O (10 mL) was diluted and extracted with EtOAc (2X 20 mL). The combined organic extracts were washed with brine, dried over Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (0-100% etoac/heptane) to give the title compound (75% purity, 433 mg,0.53 mmol,47% yield) as a red-earth solid; M/Z614, 616[ M+H ]] + ,ESI + ,RT=0.88(S2)。
Example 50 (step 19. B): (2R, 5S) -5- (7-chloroquine-3-amido) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester
(2R, 5S) -5- (7-chloroquine-3-amido) -2- { N' - [ (1 s,3 s) -3- (trifluoro-methoxy) cyclobutanecarbonyl group]T-butyl hydrazinocarbonyl } piperidine-1-carboxylate (75% purity, 433 mg,0.53 mmol), tsCl (305 mg,1.60 mmol) and K 2 CO 3 A solution of (365 mg,2.66 mmol) in ACN (15 mL) was stirred overnight at 65 ℃. The reaction mixture was treated with H 2 O (10 mL) was diluted and extracted with EtOAc (2X 20 mL). The combined organic extracts were purified over Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 5) to give the title compound (124 mg,0.200mmol,38% yield) as a white solid; 1 H NMR(400MHz,DMSO-d 6 )δ9.28(d,J=2.0Hz,1H),8.86(s,1H),8.68(d,J=6.2Hz,1H),8.25–8.08(m,2H),7.75(dd,J=8.8,2.1Hz,1H),5.52(s,1H),4.91(p,J=7.4Hz,1H),4.13(d,J=25.8Hz,2H),3.46(ddd,J=17.6,9.8,7.8Hz,1H),3.06(d,J=11.7Hz,1H),2.91–2.79(m,2H),2.08(d,J=11.7Hz,1H),1.98–1.77(m,2H),1.29(s,9H);M/Z:596,598[M+H] + ,ESI + ,RT=1.02(S2)。
example 51 (step 19. C): 7-chloro-N- [ (3 s,6 r) -6- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide
To (2R, 5S) -5- (7-chloroquine-3-amid-yl) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl]A solution of tert-butyl-1, 3, 4-oxadiazol-2-yl } piperidine-1-carboxylate (124 mg,0.200mmol, example 50) in DCM (10 mL) was added ZnBr 2 (135 mg,0.599 mmol) and the mixture was stirred at room temperature overnight. Adding additional ZnBr 2 (135 mg,0.599 mmol) and the mixture was stirred at room temperature for 3h. The reaction mixture was concentrated in vacuo and the residue was dissolved in EtOAc. Saturated NH for organic layer 4 Washing with aqueous Cl solution, passing through Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 5) to give the title compound (49 mg,0.0931mmol,47% yield) as a white solid; 1 H NMR(500MHz,DMSO-d 6 )δ9.30(d,J=2.2Hz,1H),8.87(d,J=1.8Hz,1H),8.66(d,J=7.7Hz,1H),8.56–8.22(m,1H),8.19–8.13(m,2H),7.74(dd,J=8.7,2.1Hz,1H),4.91(p,J=7.5Hz,1H),3.93(ddd,J=17.7,9.0,3.0Hz,2H),3.17(dd,J=11.8,3.3Hz,1H),2.90–2.81(m,2H),2.62–2.55(m,1H),2.08(d,J=10.4Hz,2H),1.82–1.71(m,1H),1.70–1.58(m,1H);M/Z:496,498[M+H] + ,ESI + ,RT=2.06(S4)。
the example compounds in table 8 were synthesized according to the general route 19 as exemplified in example 51 using the corresponding intermediates. The corresponding boc protected intermediates of the numbered examples are also embodiments of the present invention.
TABLE 8
Scheme of route 20
Step 20.A: (2S, 5R) -5- [ (3, 4-dichlorobenzoyl) amino ] -2- [ [2- (trifluoromethoxy) ethoxycarbonylamino ] carbamoyl ] piperidine-1-carboxylic acid tert-butyl ester
To (2S, 5R) -1-tert-butoxycarbonyl-5- [ (3, 4-dichlorobenzoyl) amino group]Piperidine-2-carboxylic acid (92% purity, 0.50g,1.10mmol, intermediate 18), 2- (trifluoromethoxy) ethyl N-carbamate (207 mg,1.10mmol, intermediate 4) and DIPEA (0.58 mL,3.31 mmol) in anhydrous DMF (10 mL) were added HATU (503 mg,1.32 mmol) and the mixture stirred at room temperature overnight. The reaction mixture was treated with H 2 O (20 mL) was diluted and extracted with EtOAc (2X 30 mL). The combined organic extracts were dried over MgSO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (10-100% etoac/heptane) to give the title compound (80% purity, 4816 mg,0.833mmol,48% yield) as a colorless oil; 1 H NMR(500MHz,CDCl 3 )δ8.05–7.89(m,1H),7.88–7.81(m,1H),7.62–7.56(m,1H),7.55–7.49(m,1H),6.79–6.63(m,1H),6.39–6.15(m,1H),4.90–4.75(m,1H),4.45–4.32(m,2H),4.25–4.15(m,3H),3.38–3.24(m,1H),2.35–2.06(m,2H),2.03–1.63(m,2H),1.55–1.38(m,9H);M/Z:587,589,591[M+H] + ,ESI + ,RT=0.99(S2)。
Example 53 (step 20. B): (2S, 5R) -5- [ (3, 4-dichlorobenzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester
/>
(2S, 5R) -5- [ (3, 4-dichlorobenzoyl) amino)]-2- [ [2- (trifluoromethoxy) ethoxycarbonylamino ]]Carbamoyl radicals]A solution of tert-butyl piperidine-1-carboxylate (80% purity, 250mg, 0.3411 mmol) and TsCl (130 mg,0.681 mmol) in anhydrous ACN (3 mL) was stirred at room temperature for 10 min. Adding K 2 CO 3 (141 mg,1.02 mmol) and the mixture was stirred at 80℃for 2h. The reaction mixture was diluted with EtOAc (15 mL) and saturated NaHCO 3 Aqueous (10 mL) and brine (10 mL). The organic layer was dried over MgSO 4 Dried and concentrated in vacuo. The residue was purified by silica gel chromatography (10-100% etoac/heptane) to give the title compound (71% purity, 80mg,0.0998mmol,29% yield) as a colorless oil; 1 H NMR(500MHz,CDCl 3 )δ7.86–7.81(m,1H),7.61–7.48(m,2H),4.75–4.67(m,2H),4.37–4.32(m,2H),4.28–4.22(m,1H),4.22–4.16(m,1H),3.21(s,1H),2.36–2.05(m,3H),2.03–1.95(m,1H),1.66–1.59(m,2H),1.48(s,9H);M/Z:569,571,573[M+H] + ,ESI + ,RT=1.04(S2)。
example 54 (step 20. C): 3, 4-dichloro-N- [ (3 r,6 s) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide
To (2S, 5R) -5- [ (3, 4-dichlorobenzoyl) amino)]-2- [5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl]A solution of tert-butyl piperidine-1-carboxylate (71% purity, 80mg,0.0998mmol, example 53) in anhydrous DCM (2 mL) was added ZnBr 2 (67 mg,0.299 mmol) and the mixture was stirred at room temperature overnight. The reaction mixture was diluted with EtOAc (10 mL) and with H 2 O (8 mL) was washed. The organic layer was dried over MgSO 4 Dried and concentrated in vacuo. The residue was purified by preparative HPLC (method 3) to give the title compound (14 mg,0.0293mmol,29% yield),as a white solid; 1 H NMR(500MHz,CDCl 3 )δ7.88(d,J=2.0Hz,1H),7.62(dd,J=8.3,2.1Hz,1H),7.53(d,J=8.3Hz,1H),6.50(d,J=5.8Hz,1H),4.74–4.68(m,2H),4.38–4.32(m,2H),4.18(ddt,J=11.0,7.4,3.8Hz,1H),4.09(dd,J=6.5,4.2Hz,1H),3.38–3.32(m,1H),2.75–2.68(m,1H),2.21–1.96(m,4H),1.80–1.72(m,1H);M/Z:469,471,473[M+H] + ,ESI + ,RT=2.04(S4)。
scheme of route 21
Step 21.A:2- [ 2-hydroxy-1- (hydroxymethyl) ethyl ] isoindoline-1, 3-dione
A solution of isobenzofuran-1, 3-dione (254 mg,5.8 mmol) and 2-aminopropane-1, 3-diol (521 mg,5.7 mmol) in toluene (20 mL) was heated at 110℃for 24 hours. The reaction mixture was cooled to 62 ℃ and treated with MTBE (20 mL) resulting in precipitation of a white powder. The suspension of intermediate 4 was stirred for 1 hour and then filtered hot to collect the precipitate. The precipitate was washed with warm MTBE (20 mL) and dried in vacuo to give the title compound (786 mg,3.45mmol,60% yield) as a white powder; 1 H NMR(500MHz,DMSO-d 6 )δ7.87–7.81(m,4H),4.89–4.84(m,2H),4.27–4.20(m,1H),3.83–3.76(m,2H),3.69–3.62(m,2H);M/Z:222[M+H] + ,ESI + ,RT=0.73(S1)。
step 21.B:2- [ trans-2- [ (benzyloxy) methyl ] -1, 3-dioxane-5-yl ] -2, 3-dihydro-1H-isoindole-1, 3-dione
/>
Benzyloxylacetaldehyde (0.25 mL,1.77 mmol), PTSA (28 mg,0.15 mmol) and 2- [ 2-hydroxy-1- (hydroxymethyl) ethyl]IsoindolesA solution of the line-1, 3-dione (336 mg,1.47 mmol) in toluene (30 mL) was heated at reflux under Dean-Stark conditions for 18 hours. The reaction mixture was cooled to room temperature and successively saturated NaHCO 3 Aqueous (2X 10 mL) and brine (10 mL). The organic layer is treated by Na 2 SO 4 Dried, concentrated in vacuo, and purified by silica gel chromatography (0-35% etoac/heptane) to give the title compound (303 mg,0.82mmol,55% yield) as a colorless oil; 1 h NMR (400 MHz, chloroform-d) delta 7.90-7.82 (m, 2H), 7.79-7.71 (m, 2H), 7.41-7.34 (m, 4H), 7.34-7.29 (m, 1H), 4.91 (t, j=4.5 hz, 1H), 4.74-4.66 (m, 1H), 4.64 (s, 2H), 4.52-4.43 (m, 2H), 4.09 (dd, j=10.8, 4.9hz, 2H), 3.60 (d, j=4.5 hz, 2H).
Step 21.C:2- [ trans-2- (hydroxymethyl) -1, 3-dioxane-5-yl ] -2, 3-dihydro-1H-isoindole-1, 3-dione
2- [ trans-2- [ (benzyloxy) methyl ]]-1, 3-dioxan-5-yl]Suspension of (E) -2, 3-dihydro-1H-isoindole-1, 3-dione (393 mg,1.06 mmol) and Pd/C (10%, 112mg,0.11 mmol) in EtOH (10 mL) and EtOAc (6 mL) in H 2 Stirred for 5 hours. The reaction mixture was taken up in N 2 Purged, warmed to near reflux, and then filtered through a pad of celite. The filtrate was concentrated in vacuo to give the title compound (94% purity, 260mg,0.93mmol,88% yield) as a white solid; 1 h NMR (400 MHz, chloroform-d) delta 7.90-7.80 (m, 2H), 7.79-7.66 (m, 2H), 4.78 (t, j=4.3 hz, 1H), 4.69-4.58 (m, 1H), 4.47 (dd, j=10.8 hz, 2H), 4.07 (dd, j=10.7, 4.8hz, 2H), 3.68 (d, j=4.2 hz, 2H), 1.92 (s, 1H).
Step 21.D: trans-5- (1, 3-dioxo-2, 3-dihydro-1H-isoindol-2-yl) -1, 3-dioxane-2-carboxylic acid
TEMPO (0.13 g,0.80 mmol) was added to 2- [ trans-2- (hydroxymethyl) -1, 3-dioxan-5-yl]-2, 3-dihydro-1H-isoindolesThe indole-1, 3-dione (89% purity, 2.37g,8.01 mmol) was purified in ACN (66 mL) and 0.67M NaH 2 PO 4 Solution in aqueous solution (66 mL). The reaction mixture was warmed to 35 ℃. Addition of NaClO 2 (80%, 1.83g,16.0 mmol) in H 2 A solution in O (15 mL) was then added NaOCl (5.0%, 0.5mL,0.41 mmol). The reaction mixture was stirred at 35 ℃ for 20 hours. Additional TEMPO (0.13 g,0.80 mmol), naClO are added 2 (80%, 1.83g,16.0 mmol) and NaOCl (5.0%, 0.5mL,0.41 mmol) and the reaction mixture was stirred at 35℃for 24 h. Additional TEMPO (0.13 g,0.80 mmol), naClO are added 2 (80%, 1.83g,16.0 mmol) and NaOCl (5.0%, 0.5mL,0.41 mmol) and the mixture was stirred at 35℃for 24 h. The reaction mixture was concentrated in vacuo. Using saturated NaHCO 3 The aqueous residue was basified to pH9 and washed with EtOAc (2×20 mL). The aqueous layer was cooled to 0 ℃ and acidified to pH2 by slow addition of 1M aqueous HCl. The aqueous layer was re-extracted with EtOAc (3X 20 mL). The combined organic extracts were treated with H 2 O (50 mL) washing with Na 2 SO 4 Drying and concentration in vacuo gave the title compound (2.10 g,7.50mmol,94% yield) as an off-white powder; 1 h NMR (400 MHz, methanol-d) delta 7.90-7.86 (m, 2H), 7.85-7.81 (m, 2H), 5.11 (s, 1H), 4.63-4.56 (m, 1H), 4.56-4.49 (m, 2H), 4.20-4.14 (m, 2H).
Step 21.E: trans-5- (1, 3-dioxo-2, 3-dihydro-1H-isoindol-2-yl) -N' - { [2- (trifluoromethoxy) ethoxy ] carbonyl } -1, 3-dioxane-2-carbohydrazide
At 0 ℃, at N 2 NMM (0.11 mL,0.959 mmol) and isobutyl chloroformate (0.12 mL,0.912 mmol) were added to a solution of 2- (trifluoromethoxy) ethyl N-carbamate (90% purity, 201mg,0.959mmol, intermediate 4) in anhydrous 2-methyl-THF (11 mL) and stirred for 15 min. Trans-5- (1, 3-dioxo-2, 3-dihydro-1H-isoindol-2-yl) -1, 3-dioxane-2-carboxylic acid (280 mg,0.959 mmol) was added and the mixture was warmed to room temperature. After 1 hour, the reaction mixture was usedH 2 O (1 mL) was quenched and quenched in EtOAc (10 mL) and H 2 O (10 mL) between partitions. The organic layer was separated and washed successively with saturated NaHCO 3 Aqueous (10 mL) and brine (10 mL) washed with Na 2 SO 4 Drying and concentration in vacuo gave the title compound (80% purity, 280mg,0.501mmol,52% yield) as an off-white powder, which was used without further purification; 1 H NMR(400MHz,DMSO-d 6 )δ10.02(s,1H),9.34(s,1H),7.91–7.79(m,4H),5.01(s,1H),4.41–4.09(m,9H)。
Step 21.F:2- [ trans-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -1, 3-dioxan-5-yl ] -2, 3-dihydro-1H-isoindole-1, 3-dione
To trans-5- (1, 3-dioxo-2, 3-dihydro-1H-isoindol-2-yl) -N' - { [2- (trifluoromethoxy) ethoxy ]]A suspension of carbonyl } -1, 3-dioxane-2-carbohydrazide (80% purity, 325mg,0.581 mmol) in anhydrous ACN (14 mL) was added to K 2 CO 3 (480 mg,3.49 mmol) and TsCl (332 mg,1.74 mmol) and stirred at 80℃for 3 hours. The reaction mixture was cooled to room temperature, and taken up in H 2 O (1 mL) was quenched and quenched in EtOAc (10 mL) and H 2 O (10 mL) between partitions. The layers were separated and the aqueous layer was re-extracted with EtOAc (10 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous (10 mL) and brine (10 mL) were washed with Na 2 SO 4 Dried and concentrated in vacuo. The residue was purified by reverse phase column chromatography (method 6) to give the title compound (90% purity, 99mg,0.208mmol,36% yield) as an off-white powder; 1 h NMR (500 MHz, chloroform-d) delta 7.91-7.85 (m, 2H), 7.85-7.75 (m, 2H), 5.85 (s, 1H), 4.88-4.79 (m, 1H), 4.79-4.73 (m, 2H), 4.65 (dd, J=11.3 Hz, 2H), 4.40-4.33 (m, 2H), 4.25 (dd, J=11.0, 4.9Hz, 2H).
Step 21.G: trans-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -1, 3-dioxan-5-amine
To 2- [ trans-2- {5- [2- (trifluoromethoxy) ethoxy ]]-1,3, 4-oxadiazol-2-yl } -1, 3-dioxan-5-yl]A solution of (E) -2, 3-dihydro-1H-isoindole-1, 3-dione (90% purity, 99mg,0.208 mmol) in EtOH (3 mL) was added NH 2 NH 2 ·H 2 O (80% purity, 15. Mu.L, 0.249 mmol) and the mixture was stirred at 40℃for 48 hours. The reaction was cooled to room temperature and the resulting precipitate was removed by filtration and washed with EtOH. The filtrate was concentrated in vacuo to give the title compound (60% purity, 60mg,0.120mmol,58% yield) which was used without further purification; M/Z300 [ M+H ]] + ,ESI + ,RT=0.70(S1)。
Example 55 (step 21. H): 7-chloro-N- [ trans-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -1, 3-dioxane-5-yl ] quinoline-3-carboxamide
A solution of 7-chloroquinoline-3-carboxylic acid (25 mg,0.120 mmol) in anhydrous 2-methyl-THF (2 mL) was treated with NMM (13. Mu.L, 0.120 mmol) and isobutyl chloroformate (15. Mu.L, 0.114 mmol) at 0deg.C. The mixture was stirred for 15 minutes and then trans-2- {5- [2- (trifluoromethoxy) ethoxy ] was slowly added]-1,3, 4-oxadiazol-2-yl } -1, 3-dioxan-5-amine (60% purity, 60mg,0.120 mmol) and then stirred at room temperature for 1 hour. The reaction mixture was cooled to 0℃with H 2 O (5 mL) was quenched and then extracted with EtOAc (5 mL). The combined organic extracts were washed with saturated NaHCO 3 Aqueous (5 mL) and brine (5 mL) washed with Na 2 SO 4 Dried, concentrated in vacuo, and purified by preparative HPLC (method 4). The relevant fractions were concentrated in vacuo at room temperature to give the title compound (4.0 mg,8.10 μmol,6.7% yield) as a white powder; 1 H NMR(500MHz,CDCl 3 )δ9.32(d,J=2.2Hz,1H),8.64(d,J=2.0Hz,1H),8.21(d,J=1.9Hz,1H),7.91(d,J=8.7Hz,1H),7.63(dd,J=8.7,2.0Hz,1H),6.79(d,J=8.3Hz,1H),5.97(s,1H),4.83–4.74(m,2H),4.56(dd,J=11.8,3.1Hz,2H),4.51–4.43(m,1H),4.42–4.36(m,2H),4.00(dd,J=11.9,5.1Hz,2H);M/Z:489,491[M+H] + ,ESI + ,RT=3.24(S4)。
II assay
HEK-ATF4 high content imaging assay
The compounds of the examples were tested in a HEK-ATF4 high content imaging assay to evaluate their pharmacological efficacy against the tunicamycin-induced ISR. Wild HEK293 cells were plated at a density of 12,000 cells per well in growth medium (containing DMEM/F12, 10% FBS, 2mM L-glutamine, 100U/mL penicillin-100. Mu.g/mL streptomycin) in 384-well imaging assay plates and at 37℃5% CO 2 Incubation was performed. After 24 hours, the medium was changed to 50. Mu.L of assay medium (DMEM/F12, 0.3% FBS, 2mM L-glutamine, 100U/mL penicillin-100. Mu.g/mL streptomycin) per well. The compounds of the examples were serially diluted in DMSO, spotted into intermediate plates and pre-diluted with assay medium containing 3.3 μm tunicamycin to give a 11-fold excess of the final assay concentration. In addition to the example compound test areas, the plates contained a plurality of control wells for assay normalization purposes, wells containing tunicamycin but no example compound (high control), and wells containing neither example compound nor tunicamycin (low control). The assay was started by transferring 5 μl from the intermediate plate into the assay plate, followed by 5% co at 37 °c 2 Incubate for 6 hours. Subsequently, cells were fixed (4% PFA/PBS at room temperature for 20 min) and subjected to indirect ATF4 immunofluorescent staining (primary antibody rabbit anti-ATF 4, clone D4B8, cell Signaling Technologies; secondary antibody Alexa Fluor 488 goat anti-rabbit IgG (H+L), thermofisher Scientific). Nuclei were stained using Hoechst dye (Thermofisher Scientific) and plates were imaged on a Opera Phenix High Content imaging platform equipped with 405nm and 488nm excitation. Finally, the image is analyzed using a script-based algorithm. The primary reading HEK-ATF4 monitors the ATF4 signal ratio between the nucleus and cytoplasm. Tunicamycin induces an increase in overall ATF4 ratio signal, which is prevented by the example compounds that modulate ISR. In addition, the number of stained nuclei corresponding to healthy cells is counted, derivedHEK-CellCount readings were generated. This reading served as an internal toxicity control. The example compounds herein did not produce significant reductions in CellCount.
The HEK ATF4 activity of the tested example compounds is provided in table 9 below:
+++=IC 50 1-500nM;++=IC 50 >500-2000nM;+=IC 50 >2000-15000nM。
TABLE 9
/>
Reference to the literature
(1)Pakos-Zebrucka K,Koryga I,Mnich K,Ljujic M,Samali A,Gorman AM.The integrated stress response.EMBO Rep.2016Oct;17(10):1374-1395.Epub 2016Sep 14.
(2)Wek RC,Jiang HY,Anthony TG.Coping with stress:eIF2 kinases and translational control.Biochem Soc Trans.2006Feb;34(Pt 1):7-11.
(3)Donnelly N,Gorman AM,Gupta S,Samali A.The eIF2alpha kinases:their structures and functions.Cell Mol Life Sci.2013Oct;70(19):3493-511
(4)Jackson RJ,Hellen CU,Pestova TV.The mechanism of eukaryotic translation initiation and principles ofits regulation.Nat Rev Mol Cell Biol.2010Feb;11(2):113-27
(5)Lomakin IB,Steitz TA.The initi ation of mammalian protein synthesis and mRNA scanning mechanism.Nature.2013Aug 15;500(7462):307-11
(6)Pain VM.Initiation of protein synthesis in eukaryotic cells.Eur J Biochem.1996Mar 15;236(3):747-71
(7)Payitt GD.Regulation of translation initiation factor eIF2B at the hub of the integrated stress response.Wiley Interdiscip Rev RNA.2018Nov;9(6):e1491.
(8)Krishnamoorthy T,Pavitt GD,Zhang F,Dever TE,Hinnebusch AG.right binding of the phosphorylated alpha subunit of initiation factor 2(eIF2alpha)to the regulatory subunits of guanine nucleotide exchange factor eIF2B is required for inhibition of translation initiation.Mol Cell Biol.2001Aug;21(15):5018-30.
(9)Hinnebusch,A.G.,Ivanov,I.P.,&Sonenberg,N.(2016).Translational control by 5’-untranslated regiohs of eukaryotic mRNAs.Science,352(6292),1413-1416.
(10)Youbg,S.K.,&Wek,R.C.(2016).Upstream open reading frames differentially regulate gene-specific translation in the integrated stress response.The Journal of Biological Chemisty,291(33),1692716935.
(11)Lin JH,Li H,Zhang Y,Ron D,Walter P(2009)Divergent effects of PERK and IRE1 signaling on cell viability.PLoS ONE 4:e4170
(12)Tabas I,Ron D.Nat Cell Biol.2011Mar;13(3):184-90.Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress.
(13)Shore GC,Papa FR,Oakes SA.Curr Opin Cell Biol.2011Apr;23(2):143-9.Signaling cell death from the endoplasmic reticulum stress response.
(14)Bi M,Naczki C,Koritzinsky M,Fels D,Blais J,Hu N,Harding H,Novoa I,Varia M,Raleigh J,Scheuner D,Kaufman RJ,Bell J,Ron D,Wouters BG,Koumenis C.EMBO J.2005Oct 5;24(19):3470-81 ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth.
(15)Bobrovnikova-Marjon E,Grigoriadou C,Pytel D,Zhang F,Ye J,Koumenis C,Cavener D,Diehl JA.Oncogene.2010Jul 8;29(27):3881-95PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage.
(16)Avivar-Valderas A,Salas E,Bobrovnikova-Marjon F,Diehl JA,Nagi c,Debnath J,Aguirre-Ghiso JA.Mol Cell Biol.2011Sep;31(17):3616-29.PERK integrates autophagy and oxidative stress responses to promote survival during extracellular matrix detachment.
(17)Blais,J.D.;Addison,C.L.;Edge,R.;Falls,T.;Zhao,H.;Kishore,W.;Koumenis,C.;Harding,H.P.;Ron,D.;Holcik,M.;Bell,J.C.Mol.Cell.Biol.2006,26,9517-9532.PERK-dependent translational regulation promotes tumor cell adaptation and angiogenesis in response to hypoxic stress.
(18)Taalab YM,Ibrahim N,Maher A,Hassan M,Mohamed W,Moustafa AA,Salama M,Johar D,Bernstein L.Rev Neurosci.2018Jub 27;29(4):387-415.Mechanisms of disordered neurodegenerative function.concepts and tacts about the different roles of the protein kinase RNA-like endoplasmic reticulum kinase(PERK).
(19)Remondelli P,Renna M.Front Mol Neurosci.2017Jun 16;10:187.The Endoplasmic Reticulum Unfolded Protein Response in N eurodegenerative Disorders and Its Potential Therapeutic Significance.
(20)Halliday M,Mallucci GR.Neuropathol Appl Neurobiol.2015Jun;41(4):414-27.Review:Modulating the unfolded protein response to prevent neurodegeneration and enhance memory.
(21)Halliday M,Radford H,Sekine Y,Moreno J,Verity N,le Quesne J,Ortori CA,Barrett DA,Fromont C,Fischer PM,Harding HP,Ron D,Mallucci GR.Cell Death Dis.2015Mar 5;6:e1672.Partial restoration of protein synthesis rates by the small molecule ISRIB prevents neurodegeneration without pancreatic toxicity.
(22)Moreno JA,Radford H,Peretti D,Steinert JR,Verity N,Martin MG,Halliday M,Morgan J,Dinsdale D,Ortori CA,Barrett DA,Tsaytler P,Bertolotti A,Willis AE,Bushell M,Mallucci GR.Nature 2012;485:507-11.Sustained translational repression by eIF2alpha-P mediates prion neurodegeneration.
(23)Skopkova M,Hennig F,Shin BS,Turner CE,Stanikova D,Brennerova K,Stanik J,Fischer U,Henden L,Müller U,Steinberger D,Leshinsky-Silver E,Bottani A,Kurdiova T,Ukropec J,Nyitrayova O,Kolnikova M,Klimes I,Borck G,Bahlo M,Haas SA,Kim JR,Lotspeich-Cole LE,Gasperikova D,Dever TE,Kalscheuer VM.Hum Mutat.2017Apr;38(4):409-425.EIF2S3 Mutations Associated with Severe X-Linked Intellectual Disability Syndrome MEHMO.
(24)Hamilton EMC,van der Lei HDW,Vermeulen G,Gerver JAM,CM,Naidu S,Mierzewska H,Gemke RJBJ,de Vet HCW,Uitdehaag BMJ,Lissenberg-Witte BI;VWM Research Group,van der Knaap MS.Ann Neurol.2018Aug;84(2):274-288.Natural History of Vanishing White Matter.
(25)Bugiani M,Vuong C,Breur M,van der Knaap MS.Brain Pathol.2018May;28(3):408-421.Vanishing white matter:a leukodystrophy due to astrocytic dysfunction.
(26)Wong YL,LeBon L,Edalji R,Lim HB,Sun C,Sidrauski C.Elife.2018Feb 28;7.The small molecule ISRIB rescues the stability and activity of Vanishing White Matter Disease eIF2B mutant complexes.
(27)Wong YL,LeBon L,Basso AM,Kohlhaas KL,Nikkel AL,Robb HM,Donnelly-Roberts DL,Prakash J,Swensen AM,Rubinstein ND,Krishnan S,McAllister FE,Haste NV,O′Brien JJ,Roy M,Ireland A,Frost JM,Shi L,Riedmaier S,Martin K,Dart MJ,Sidrauski C.Elife.2019Jan 9;8.cIF2B activator prevents neurological defects caused by a chronic integrated stress response.
(28)Nguyen HG,Conn CS,Kye Y,Xue L,Forester CM,Cowan JE,Hsieh AC,Cunningham JT,Truillet C,Tameire F,Evans MJ,Evans CP,Yang JC,Hann B,Koumenis C,Walter P,Carroll PR,Ruggero D.Sci Transl Med.2018May 2;10(439).Development of a stress response therapy targeting aggressive prostate cancer.
(29)Waring M,Expert Opinion on Drug Discovery Volume 5,2010-Issue 3,235-248.Lipophilicity in Drug Discovery.
(30)Alelyunas YW,et.al.Bioorg.Med.Chem.Lett.,20(24)2010,7312-7316.Experimental solubility profiling of marketed CNS drugs,exploring solubilitylimit of CNS discovery candidate.
(31)Redfern WS,et.al.,Cardiovascular Research 58(2003),32-45.Relationships between preclinical cardiac electrophysiology,clinical QT interval prolongation and torsade de pointes for a broad range of drugs.

Claims (24)

1. A compound of formula (I) or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof
Wherein the method comprises the steps of
X 1 Is N (R) 4 ) And X is 2 Is CH (R) 4e ) The method comprises the steps of carrying out a first treatment on the surface of the Or X 1 And X 2 Is O;
R 1 is H or C 1-4 Alkyl, preferably H, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
R 2 is phenyl, naphthyl, C 3-7 Cycloalkyl, 3-to 7-membered heterocyclyl or 7-to 12-membered heterobicyclo, wherein R 2 Optionally by one or more of the same or different R 5 With the proviso that if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atom attached to the carbon atom of the amide group is not substituted with H or F;
R 5 independently halogen, CN, C (O) OR 6 、OR 6 、C(O)R 6 、C(O)N(R 6 R 6a )、S(O) 2 N(R 6 R 6a )、S(O)N(R 6 R 6a )、S(O) 2 R 6 、S(O)R 6 、N(R 6 )S(O) 2 N(R 6a R 6b )、SR 6 、N(R 6 R 6a )、NO 2 、OC(O)R 6 、N(R 6 )C(O)R 6a 、N(R 6 )S(O) 2 R 6a 、N(R 6 )S(O)R 6a 、N(R 6 )C(O)OR 6a 、N(R 6 )C(O)N(R 6a R 6b )、OC(O)N(R 6 R 6a ) Oxo (=o), wherein the ring is at least partially saturated, C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 7 Substitution;
R 6 、R 6a 、R 6b independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 7 is halogen, CN, C (O) OR 8 、OR 8 、C(O)R 8 、C(O)N(R 8 R 8a )、S(O) 2 N(R 8 R 8a )、S(O)N(R 8 R 8a )、S(O) 2 R 8 、S(O)R 8 、N(R 8 )S(O) 2 N(R 8a R 8b )、SR 8 、N(R 8 R 8a )、NO 2 、OC(O)R 8 、N(R 8 )C(O)R 8a 、N(R 8 )SO 2 R 8a 、N(R 8 )S(O)R 8a 、N(R 8 )C(O)N(R 8a R 8b )、N(R 8 )C(O)OR 8a Or OC (O) N (R) 8 R 8a );
R 8 、R 8a 、R 8b Independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 3 is OR (OR) 9 、SR 9a 、N(R 9 R 9a )、A 1 、C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 10 Substitution;
R 9 、R 9a independently selected from H, C 1-6 Alkyl, C 2-6 Alkenyl, C 2-6 Alkynyl and A 1 Wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups optionally being substituted by one or more R's, identical or different 11 Substitution;
R 10 is halogen OR 12 CN or A 1
R 11 Is halogen, CN, OR 12 、OA 1 Or A 1
R 12 Is H or C 1-4 Alkyl, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
A 1 is phenyl, C 3-7 Cycloalkyl, C 4-12 Bicycloalkyl or 3-7 membered heterocyclyl, wherein A 1 Optionally by one or more of the same or different R 13 Substitution;
R 13 is R 14 、OH、OR 14 Halogen or CN; and
R 14 is cyclopropyl, C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl, wherein R is 14 Optionally by one or more of the same or different R 15 Substitution; or alternatively
Two R 13 Are linked to form, together with the atoms to which they are attached, a ring A 2
R 15 Is halogen, CN OR OR 16
R 16 Is H or C 1-4 Alkyl, wherein C 1-4 Alkyl is optionally substituted with one or more halogen, the same or different;
A 2 is phenyl, C 3-7 Cycloalkyl or 3-7 membered heterocyclyl, wherein A 2 Optionally by one or more of the same or different R 17 Substitution;
R 17 is C 1-6 Alkyl, C 2-6 Alkenyl or C 2-6 Alkynyl group, wherein C 1-6 Alkyl, C 2-6 Alkenyl and C 2-6 Alkynyl groups are optionally substituted with one or more halo groups, which may be the same or different;
R 4 Is H, C (O) OC 1-4 Alkyl or C 1-4 Alkyl, wherein C (O) OC 1-4 Alkyl and C 1-4 The alkyl group is optionally substituted with one or more substituents selected from the group consisting of: halogen, OH and OC 1-3 Alkyl, wherein the substituents are the same or different;
R 4a 、R 4b 、R 4c 、R 4f independently selected from H, halogen and C 1-4 An alkyl group; and
R 4d 、R 4e independently selected from H, OH, OC 1-4 Alkyl, halogen and C 1-4 An alkyl group;
or R is 4d And R is 4e One and R 4 Forming a methylene or ethylene group;
or R is 4 And R is R 4c Forming an ethylene group;
or R is 4b And R is R 4d Covalent single bonds are formed.
2. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein X in formula (I) 1 Is N (R) 4 ) And X is 2 Is CH (R) 4e ) Obtaining the formula (I-1)
3. The compound of claim 1, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein X in formula (I) 1 And X 2 For O, give the formula (I-2)
4. A compound according to claim 1 or 2, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, wherein R 4 H, CH of a shape of H, CH 3 、CH 2 CH 3 Or CH (CH) 2 CH 2 OCH 3 The method comprises the steps of carrying out a first treatment on the surface of the Preferably H or CH 3 The method comprises the steps of carrying out a first treatment on the surface of the More preferably H.
5. The compound of any one of claims 1-4, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 4a 、R 4b 、R 4c 、R 4f Independently selected from H, halogen and C 1-4 Alkyl and R 4d 、R 4e Independently selected from H, OH, OC 1-4 Alkyl, halogen and C 1-4 An alkyl group; preferably R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e Independently selected from H, F and CH 3 The method comprises the steps of carrying out a first treatment on the surface of the More preferably, R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e Is H.
6. The compound of any one of claims 1-5, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 1 Is H or CH 3 The method comprises the steps of carrying out a first treatment on the surface of the H is preferred.
7. A compound according to any one of claims 1-6, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer thereofOr a stereoisomer, wherein R in formula (I-1) 1 、R 4 、R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 4e For H, to give a compound of formula (Ia-1), or wherein R in formula (I-2) 1 、R 4a 、R 4b 、R 4c 、R 4f 、R 4d H to give the formula (Ia-2)
8. The compound of any one of claims 1-7, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 2 Is phenyl, pyridyl, thienyl, 1H-indolyl, quinolinyl, isoquinolinyl, quinazolinyl, pyrazolo [1,5-a ]]Pyridinyl, pyrrolo [1,2-a ]]Pyrazinyl, indolizinyl, chromeneyl, benzofuranyl or 2H-1, 3-benzodioxolyl; preferably phenyl, pyridin-2-yl, pyridin-3-yl, thiophen-2-yl, 1H-indol-2-yl, quinolin-3-yl, quinolin-6-yl, quinolin-7-yl, isoquinolin-3-yl, quinazolin-2-yl, pyrazolo [1,5-a ]Pyridin-2-yl, pyrrolo [1,2-a ]]Pyrazin-3-yl, indolizin-2-yl, chromen-3-yl, benzofuran-2-yl or 2H-1, 3-benzodioxol-5-yl; and wherein R is 2 Optionally by one or more of the same or different R 5 With the proviso that if R is to be 2 R is bound to a ring atom bound to a carbon atom of an amide group of formula (I) 2 Is oxygen, R is 2 The ring atom attached to the carbon atom of the amide group is not substituted with H or F.
9. The compound of any one of claims 1-8, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 2 By one, two or three identical or different R 5 And (3) substitution.
10. The compound of any one of claims 1-9, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 5 Is F, cl, CH 3 、CF 3 、OCF 3 Or OCH (optical wavelength) 2 CF 3
11. The compound of any one of claims 1-10, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 3 Is OR (OR) 9 And R is 9 Is C 1-6 Alkyl or C 2-6 Alkenyl group, wherein C 1-6 Alkyl and C 2-6 Alkenyl groups being substituted by one or more identical or different R' s 11 And (3) substitution.
12. The compound of any one of claims 1-11, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 3 Is OR (OR) 9 And R is 9 Is C 1-6 Alkyl, preferably ethyl, wherein C 1-6 Alkyl is substituted with one R 11 And (3) substitution.
13. The compound of any one of claims 1-12, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 3 Is OCH 2 CH 2 OCF 3
14. The compound of any one of claims 1-11, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 3 Is OR (OR) 9 And R is 9 Is C 1-6 Alkyl or C 2-6 Alkenyl, preferably but-2-enyl, wherein C 1-6 Alkyl and C 2-6 Alkenyl groups are each substituted with three F; more preferably R 3 Is OCH 2 CH=CHCF 3
15. According to claimA compound of any one of claims 1-10, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 3 Is A 1 Preferably phenyl or cyclobutyl, wherein A is 1 Optionally by one or more of the same or different R 13 And (3) substitution.
16. The compound of any one of claims 1-10 and 15, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein a 1 Is one or two, preferably one R 13 And (3) substitution.
17. The compound of any one of claims 1-10, 15, and 16, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R 13 Is CH 3 、CHF 2 、CF 3 、CH 2 CF 3 、OCHF 2 、OCH 2 CF 3 、OCF 3 、OCH 3 F or Cl, preferably Cl or OCF 3
18. The compound of any one of claims 1-17, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein R in formula (I) is selected 1 、R 2 、R 4a 、R 4b 、R 4c 、R 4f 、R 4d 、R 3 、X 1 、X 2 Obtaining
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethyl) quinoline-2-amid-yl ] piperidine-1-carboxylic acid tert-butyl ester;
n- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -6- (trifluoromethyl) quinoline-2-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2H-chromen-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] isoquinoline-3-carboxamide;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinazoline-2-carboxamide;
(2 r,5 s) -5- (6-chloroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
5-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -1-benzofuran-2-carboxamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-7-carboxamide;
7-chloro-6-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
5-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyrazolo [1,5-a ] pyridine-2-carboxamide;
6- (2, 2-trifluoroethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-3-carboxamide;
n- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2- (trifluoromethyl) quinoline-6-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyrrolo [1,2-a ] pyrazine-3-carboxamide;
6- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-3-carboxamide;
3, 4-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
7-chloro-8-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] indolizine-2-carboxamide;
6-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] indolizine-2-carboxamide;
1-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -5- (trifluoromethyl) -1H-indole-2-carboxamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3, 5-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3, 4-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4, 5-dimethyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] thiophene-2-carboxamide;
4-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
(2 r,5 s) -5- [4- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
4- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
2, 2-difluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -2H-1, 3-benzodioxol-5-carboxamide
4-chloro-3-methyl-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-chloro-3, 5-difluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
3-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -4- (trifluoromethyl) benzamide;
3-chloro-4- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
4-fluoro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -3- (trifluoromethyl) benzamide;
3-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -4- (trifluoromethyl) benzamide;
4-chloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] -3- (trifluoromethyl) benzamide;
4, 5-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-2-carboxamide;
5, 6-dichloro-N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] pyridine-2-carboxamide;
4-chloro-3- (trifluoromethoxy) -N- [ (3 s,6 r) -6- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] benzamide;
(2 r,5 s) -5- [ [ 1-methyl-6- (trifluoromethyl) indole-2-carbonyl ] amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
1-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] -6- (trifluoromethyl) indole-2-carboxamide;
(2 r,5 s) -5- [ (3-chloro-4-methyl-benzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
3-chloro-4-methyl-N- [ (3 s,6 r) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide;
(2 r,5 s) -2- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -5- [ (7-chloroquinoline-3-carbonyl) amino ] piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- [5- (4-chlorophenyl) -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
(2 r,5 s) -5- [ (7-chloroquinoline-3-carbonyl) amino ] -2- [5- [ (E) -4, 4-trifluoro-but-2-yloxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- [5- [ (E) -4, 4-trifluoro-but-2-enoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 r,6 s) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] quinoline-3-carboxamide;
(2 r,5 s) -5- (7-chloroquinoxaline-3-amido) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
7-chloro-N- [ (3 s,6 r) -6- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
7-chloro-N- [ (3 r,6 s) -6- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidin-3-yl ] quinoline-3-carboxamide;
(2 s,5 r) -5- [ (3, 4-dichlorobenzoyl) amino ] -2- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] piperidine-1-carboxylic acid tert-butyl ester;
3, 4-dichloro-N- [ (3 r,6 s) -6- [5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl ] -3-piperidinyl ] benzamide;
(2 s,5 r) -5- (7-chloroquinoxaline-3-amido) -2- {5- [ (1 s,3 s) -3- (trifluoromethoxy) cyclobutyl ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 s,5 r) -5- (7-chloroquinoxaline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-chloro-3- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (5, 6-dichloropyridine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4, 5-dichloropyridine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-chloro-3- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-chloro-4- (trifluoromethoxy) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 4-fluoro-3- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-chloro-4- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 3-fluoro-4- (trifluoromethyl) benzamide ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chloro-3, 5-difluorobenzamide) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chloro-3-methylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (2, 2-difluoro-2H-1, 3-benzodioxol-5-ylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-methylbenzamide) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4, 5-dimethylthiophene-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 4-dimethylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 5-dimethylbenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3-chlorobenzoylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [ 1-methyl-5- (trifluoromethyl) -1H-indole-2-amido ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (6-chloroindolizine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloroindolizine-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-8-fluoroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (4-chlorobenzoylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3, 4-dichlorobenzamido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [6- (trifluoromethoxy) pyridin-3-ylamino ] piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- { 7-chloropyrrolo [1,2-a ] pyrazine-3-amido } -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -5- [2- (trifluoromethyl) quinoline-6-carboxamide ] piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- [6- (2, 2-trifluoroethoxy) pyridin-3-ylamino ] -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- { 5-chloropyrazolo [1,5-a ] pyridine-2-amido } -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-6-fluoroquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (3-chloroquinoxaline-7-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (5-chloro-1-benzofuran-2-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (6-chloroquinazolin-2-ylamino) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloroisoquinoline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2 r,5 s) -5- (7-chloro-2H-chromen-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester;
(2R, 5S) -5- (7-chloroquinoxaline-3-amido) -2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } piperidine-1-carboxylic acid tert-butyl ester or
7-chloro-N- [ trans-2- {5- [2- (trifluoromethoxy) ethoxy ] -1,3, 4-oxadiazol-2-yl } -1, 3-dioxane-5-yl ] quinoline-3-carboxamide.
19. The compound of any one of claims 1-18, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein formula (I) has a stereochemistry shown in formula (Ib)
20. The compound of any one of claims 1-19, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, wherein formula (I) has a stereochemistry shown in formula (Ib-1), (Ib-2), (Ib-3), (Ib-4)
21. A pharmaceutical composition comprising at least one compound of any one of claims 1-20, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer, or stereoisomer thereof, and a pharmaceutically acceptable carrier, optionally in combination with one or more other biologically active compounds or pharmaceutical compositions.
22. A compound according to any one of claims 1-20, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, for use as a medicament.
23. A compound according to any one of claims 1-20, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, or a pharmaceutical composition according to claim 21, for use in a method of treatment or prophylaxis of one or more diseases or disorders associated with an integrated stress response.
24. A compound according to any one of claims 1-20, or a pharmaceutically acceptable salt, solvate, hydrate, tautomer or stereoisomer thereof, or a pharmaceutical composition according to claim 21, for use in a method of treatment or prophylaxis of one or more diseases or disorders selected from the group consisting of: white matter dystrophy, intellectual disability syndrome, neurodegenerative diseases and disorders, neoplastic diseases, infectious diseases, inflammatory diseases, musculoskeletal diseases, metabolic diseases, eye diseases, diseases selected from the group consisting of organ fibrosis, chronic and acute liver diseases, chronic and acute lung diseases, chronic and acute kidney diseases, myocardial infarction, cardiovascular diseases, cardiac arrhythmias, atherosclerosis, spinal cord injury, ischemic stroke and neuropathic pain.
CN202180086795.0A 2020-10-22 2021-10-21 Modulators of integrated stress response pathways Pending CN116761803A (en)

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
EP20203311.4 2020-10-22
EP21192154.9 2021-08-19
EP21192154 2021-08-19
PCT/EP2021/079209 WO2022084447A1 (en) 2020-10-22 2021-10-21 Modulators of the integrated stress response pathway

Publications (1)

Publication Number Publication Date
CN116761803A true CN116761803A (en) 2023-09-15

Family

ID=77411640

Family Applications (1)

Application Number Title Priority Date Filing Date
CN202180086795.0A Pending CN116761803A (en) 2020-10-22 2021-10-21 Modulators of integrated stress response pathways

Country Status (1)

Country Link
CN (1) CN116761803A (en)

Similar Documents

Publication Publication Date Title
CN113993850B (en) Modulators of integrated stress response pathways
KR102399206B1 (en) Isoquinolin-3-yl carboxamide and its preparations and uses
JP5642963B2 (en) Pyridinonyl PDK1 inhibitor
US10604514B2 (en) 6-(5-membered heteroaryl)isoquinolin-3-yl carboxamides and preparation and use thereof
WO2020108590A1 (en) Pyrimidine and five-membered nitrogen heterocycle derivative, preparation method therefor, and medical uses thereof
JP2017531679A (en) Indole carboxamides useful as kinase inhibitors
KR20160134865A (en) Amide derivatives and pharmaceutically acceptable salts thereof, preparation method therefor and medicinal application thereof
JP2022530051A (en) Modulator of integrated stress response pathway
CN114980894A (en) Modulators of integrated stress response pathways
WO2020143763A1 (en) Haloallylamine compounds and application thereof
CN110914253B (en) Isoindolone-imide ring-1,3-diketone-2-alkene compounds, and composition and application thereof
CN115515949A (en) Novel aminopyrimidine EGFR (epidermal growth factor receptor) inhibitor
JP2023517944A (en) Regulators of integrated stress response pathways
CN116761803A (en) Modulators of integrated stress response pathways
CN117098753A (en) Modulators of integrated stress response pathways
CN116964047A (en) Modulators of integrated stress response pathways
JP2023546226A (en) Modulators of integrated stress response pathways
WO2022272106A1 (en) Cdk2 inhibitors and methods of using the same
WO2023246837A1 (en) Class of compounds having pyrimido-six-membered ring structure, pharmaceutical compositions comprising same, and use thereof

Legal Events

Date Code Title Description
PB01 Publication
PB01 Publication
SE01 Entry into force of request for substantive examination
SE01 Entry into force of request for substantive examination
REG Reference to a national code

Ref country code: HK

Ref legal event code: DE

Ref document number: 40100736

Country of ref document: HK