WO2020031107A1 - Composés chimiques - Google Patents

Composés chimiques Download PDF

Info

Publication number
WO2020031107A1
WO2020031107A1 PCT/IB2019/056721 IB2019056721W WO2020031107A1 WO 2020031107 A1 WO2020031107 A1 WO 2020031107A1 IB 2019056721 W IB2019056721 W IB 2019056721W WO 2020031107 A1 WO2020031107 A1 WO 2020031107A1
Authority
WO
WIPO (PCT)
Prior art keywords
4alkyl
cycloalkyl
substituted
disease
heterocycloalkyl
Prior art date
Application number
PCT/IB2019/056721
Other languages
English (en)
Inventor
Karen Anderson Evans
Drew Roger HEITZ
Joseph E. Pero
Original Assignee
Glaxosmithkline Intellectual Property Development Limited
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 Glaxosmithkline Intellectual Property Development Limited filed Critical Glaxosmithkline Intellectual Property Development Limited
Publication of WO2020031107A1 publication Critical patent/WO2020031107A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D307/00Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom
    • C07D307/02Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings
    • C07D307/04Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members
    • C07D307/06Heterocyclic compounds containing five-membered rings having one oxygen atom as the only ring hetero atom not condensed with other rings having no double bonds between ring members or between ring members and non-ring members with only hydrogen atoms or radicals containing only hydrogen and carbon atoms, directly attached to ring carbon atoms
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D407/00Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00
    • C07D407/02Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings
    • C07D407/12Heterocyclic compounds containing two or more hetero rings, at least one ring having oxygen atoms as the only ring hetero atoms, not provided for by group C07D405/00 containing two hetero rings linked by a chain containing hetero atoms as chain links

Definitions

  • the present invention relates to substituted tetrahydrofuran derivatives that are inhibitors of the ATF4 pathway.
  • the present invention also relates to pharmaceutical compositions comprising such compounds and methods of using such compounds in the treatment of diseases/injuries associated with activated unfolded protein response pathways, such as cancer, pre-cancerous syndromes, Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, Creutzfeldt-Jakob Disease, and related prion diseases, progressive supranuclear palsy, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, cognitive impairment, atherosclerosis, ocular diseases, neurological disorders, pain, arrhythmias, in organ transplantation and in the transportation of organs for transplantation.
  • elF2a phosphorylation results in an attenuation of translation with consequences that allow cells to cope with the varied stresses (1).
  • elF2 (which is comprised of three subunits, a, b , a n d y) binds GTP and the initiator Met-tRNA to form the ternary complex (elF2-GTP-Met-tRNAi), which, in turn, associates with the 40S ribosomal subunit scanning the 5'UTR ofmRNAs to select the initiating AUG codon.
  • elF2 Upon phosphorylation of its a-subunit, elF2 becomes a competitive inhibitor of its GTP-exchange factor (GEF), elF2B (2).
  • GEF GTP-exchange factor
  • elF2B The tight and nonproductive binding of phosphorylated elF2 to elF2B prevents loading of the elF2 complex with GTP thus blocking ternary complex formation and reducing translation initiation (3).
  • GTP-exchange factor GTP-exchange factor
  • mRNAs that contain upstream open reading frames (uORFs) in their 5'UTR are translationally up-regulated (4,5).
  • uORFs upstream open reading frames
  • mRNAs that contain upstream open reading frames (uORFs) in their 5'UTR are translationally up-regulated (4,5).
  • ATF4 a cAMP element binding (CREB) transcription factor
  • CHOP a pro-apoptotic transcription factor
  • ATF4 regulates the expression of many genes involved in metabolism and nutrient uptake and additional transcription factors, such as CHOP, which is under both translational and transcriptional control (9).
  • Phosphorylation of elF2a thus leads to preferential translation of key regulatory molecules and directs diverse changes in the transcriptome of cells upon cellular stress.
  • UPR unfolded protein response
  • the UPR is activated by unfolded or misfolded proteins that accumulate in the ER lumen because of an imbalance between protein folding load and protein folding capacity, a condition known as "ER stress".
  • the UPR is comprised of three signaling branches mediated by ER- localized transmembrane sensors, PERK, IRE1 , and ATF6.
  • PERK and IRE1 are homologous and likely activated in analogous ways by direct binding to unfolded peptides (12). This binding event leads to oligomerization and trans- autophosphorylation of their cytosolic kinase domains, and, for PERK, phosphorylation of its only known substrate, elF2a. In this way, PERK activation results in a quick reduction in the load of newly synthesized proteins that are translocated into the ER- lumen (13).
  • both the transcription factor XBP1 s produced as the consequence of a non-conventional mRNA splicing reaction initiated by IRE1
  • the transcription factor ATF6 produced by proteolysis and release from the ER membrane
  • ATF4 Upon ER stress, both the transcription factor XBP1 s, produced as the consequence of a non-conventional mRNA splicing reaction initiated by IRE1 , and the transcription factor ATF6, produced by proteolysis and release from the ER membrane, collaborate with ATF4 to induce the vast UPR transcriptional response.
  • Transcriptional targets of the UPR include the ER protein folding machinery, the ER-associated degradation machinery, and many other components functioning in the secretory pathway (14).
  • Small-molecule therapeutics that inhibit the UPR and/or the Integrated Stress Response could be used in cancer as a single agent or in combination with other chemotherapeutics ( 1 7 , 1 8 , 1 9 ) , for enhancement of long-term memory (24,25) , in neurodegenerative and prion associated diseases (20) , in white matter disease (VWM) (23) and in biotechnology applications that would benefit from increased protein translation.
  • chemotherapeutics 1 7 , 1 8 , 1 9
  • compositions that comprise a pharmaceutically acceptable excipient and compounds of Formula (I).
  • Alzheimer’s disease spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease
  • the invention is directed to substituted tetrahydrofuran derivatives. Specifically, the invention is directed to compounds according to Formula (I):
  • C, D, L 1 , L 2 , R 1 , R 2 , R 3 , R 4 , R 5 , R 6 , Y 1 , Y 2 , z 1 , z 2 , z 3 , z 4 , X 1 , X 2 , X 3 , X 4 , X 5 , and X® are as defined below; or a salt thereof including a pharmaceutically acceptable salt thereof.
  • the present invention also relates to the discovery that the compounds of Formula (I) are active as inhibitors of the ATF4 pathway.
  • the present invention also relates to the discovery that the compounds of Formula (I) prevent the translation of ATF4.
  • This invention also relates to a method of treating Alzheimer’s disease, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating Parkinson’s disease, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating amyotrophic lateral sclerosis, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating Huntington’s disease, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating C re utzfeldt- Jakob Disease, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating progressive supranuclear palsy (PSP), which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • PSP progressive supranuclear palsy
  • This invention also relates to a method of treating dementia, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating spinal cord injury, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating traumatic brain injury, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating ischemic stroke, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating diabetes, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating a disease state selected from: myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, and arrhythmias, which comprises administering to a human in need thereof an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • This invention also relates to a method of treating an integrated stress response-associated disease in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, to the patient.
  • This invention also relates to a method of treating a disease associated with phosphorylation of elF2a in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to the patient.
  • This invention also relates to a method of treating a disease in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, to the patient, wherein the disease is selected from the group consisting of cancer, a neurodegenerative disease, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and an intellectual disability syndrome.
  • This invention also relates to a method of improving long-term memory in a patient, which comprises administering a therapeutically effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, to the patient.
  • This invention also relates to a method of increasing protein expression of a cell or in vitro expression system, which comprises administering an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof, to the cell or expression system.
  • This invention also relates to a method of treating an inflammatory disease in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to the patient.
  • This invention also relates to a method of using the compounds of Formula (I) in organ transplantation and in the transportation of organs for transplantation.
  • Also included in the present invention are methods of co-administering the presently invented compounds with further active ingredients.
  • Included in the present invention is a method for treating neurodegenerative diseases, cancer, and other diseases/injuries associated with activated unfolded protein response pathways, such as: Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeldt- Jakob Disease, and related prion diseases, amyotrophic lateral sclerosis, progressive supranuclear palsy, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementias, atherosclerosis, ocular diseases, arrhythmias, in organ transplantation and in the transportation of organs for transplantation that comprises administering the compounds of Formula (I).
  • Alzheimer’s disease spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeld
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in therapy.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of Alzheimer’s disease.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of Parkinson’s disease syndromes.
  • the invention also relates to a compound of Formula 0 or a pharmaceutically acceptable salt thereof for use in the treatment of amyotrophic lateral sclerosis.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of Huntington’s disease.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of Creutzfeldt-Jakob Disease.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of progressive supranuclear palsy (PSP).
  • PSP progressive supranuclear palsy
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of dementia.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of spinal cord injury.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of traumatic brain injury.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of ischemic stroke.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of diabetes.
  • the invention also relates to a compound of Formula (I) or a pharmaceutically acceptable salt thereof for use in the treatment of a disease state selected from: myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, and arrhythmias.
  • a disease state selected from: myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, and arrhythmias.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of an integrated stress response-associated disease.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a disease associated with phosphorylation of elF2a.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a disease selected from the group consisting of: cancer, a
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for improving long-term memory.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for increasing protein expression of a cell or in vitro expression system.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of inflammatory disease.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament in organ transplantation and in the transportation of organs for transplantation.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for the treatment of a disease state selected from: neurodegenerative diseases, cancer, and other diseases/injuries associated with activated unfolded protein response pathways such as: Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeldt- Jakob Disease, and related prion diseases, amyotrophic lateral sclerosis, progressive supranuclear palsy, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementias, atherosclerosis, ocular diseases, neurological disorders, pain, arrhythmias, in organ transplantation and in the transportation of organs for transplantation.
  • a disease state selected from: neurodegenerative diseases,
  • compositions that comprise a pharmaceutical excipient and a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the invention also relates to a pharmaceutical composition as defined above for use in therapy.
  • the invention also relates to a combination for use in therapy which comprises a therapeutically effective amount of (i) a compound of Formula (I) or a pharmaceutically acceptable salt thereof; and (ii) further active ingredients.
  • c1 and X ⁇ are independently selected from: hydrogen, Ci-4alkyl, Ci-4alkyl substituted from 1 to 3 times by fluoro, hydroxy, and -NH2;
  • X® and X® are independently selected from: hydrogen, Ci-4alkyl, Ci-4alkyl substituted from 1 to 3 times by fluoro, hydroxy, and -NH2, or X® and X® are taken together to form oxo;
  • X ⁇ and X® are independently selected from: hydrogen, Ci-4alkyl, Ci-4alkyl substituted from 1 to 3 times by fluoro, hydroxy, and -NH2, or X ⁇ and X® are taken together to form oxo; and y2 are independently selected from: -C(O)-, Ci-3alkyene, -S(0)2-, -S(O)-,
  • i is selected from: a bond, -NH-, -N(Cl -4alkyl)-, -N(substituted Cl-4alkyl)-, -0-, -S-, -S(O)-, -S(0)2-, cycloalkyl, -O-cycloalkyl, cycloalkyl-O-, -NH-cycloalkyl, cycloalkyl-NH-, -CH2-cycloalkyl, cycloalkyl-CH2-, azetidinyl, -O-azetidinyl, azetidinyl-O-, -N-azetidinyl, azetidinyl-N-, substituted or unsubstituted Ci-6alkylene and substituted or unsubstituted Ci-6he
  • heterocycloalkyl heterocycloalkyl-O-, heterocycloalkyl-NH-,
  • U is taken together with an R® substituent adjacent to the point of attachment of i to C to form a cycloalkyl ring, a heterocycloalkyl ring, or heteroaryl ring fused to C;
  • l_2 is selected from: a bond, -NH-, -N(Cl -4alkyl)-, -N(substituted Cl-4alkyl)-, -0-, -S-, -S(O)-, -S(0)2-, cycloalkyl, -O-cycloalkyl, cycloalkyl-O-, -NH- cycloalkyl, cycloalkyl-NH-, -CH2-cycloalkyl, cycloalkyl-CH2-, azetidinyl, -O-azetidinyl, azetidinyl-O-, -N-azetidinyl, azetidinyl-N-, substituted or unsubstituted Ci-6alkylene and substituted or unsubstituted Ci -6heteroalkylene, or,
  • heterocycloalkyl heterocycloalkyl-O-, heterocycloalkyl-NH-,
  • l_2 is taken together with an R® substituent adjacent to the point of attachment of l_2 to D to form a cycloalkyl ring, a heterocycloalkyl ring, or heteroaryl ring fused to D; are independently NR ⁇ , O, or S;
  • R is selected from: hydrogen, Ci -6alkyl and Ci -6alkyl substituted 1 to 6 times by fluoro;
  • is selected from: fluoro, chloro, bromo, iodo, -C(0)0Cl-4alkyl, -OH, -NH2, -C(0)NHCl-4alkyl, -OCl-4alkyl, -OCH2Ph, -C(0)Ph, -CF3, -CN, -S(0)CH3,
  • R® substituents can combine to form a cycloalkyl ring, a heterocycloalkyl ring, or a heteroaryl ring fused to C,
  • an R® substituent adjacent to the point of attachment of i to C combines with U to form a cycloalkyl ring, a heterocycloalkyl ring, or a heteroaryl ring fused to C;
  • is selected from: fluoro, chloro, bromo, iodo, -C(0)0Ci-4alkyl, -OH, -IMH2,
  • R x is selected from hydrogen, Ci-4alkyl, and cycloalkyl, and RY is selected from Ci-4alkyl, and cycloalkyl
  • R x is selected from hydrogen, Ci-4alkyl, and cycloalkyl
  • RY is selected from Ci-4alkyl, and cycloalkyl
  • substituted or unsubstituted Ci-6alkyl substituted or unsubstituted Ci-6heteroalkyl
  • substituted or unsubstituted cycloalkyl substituted or unsubstituted heterocycloalkyl, substituted or
  • an R® substituent adjacent to the point of attachment of l_2 to D combines with l_2 to form a cycloalkyl ring, a heterocycloalkyl ring, or a heteroaryl ring fused to D;
  • C and D are independently phenyl or pyridyl
  • R 1 is selected from: hydrogen, Ci-6alkyl, substituted Ci -6alkyl, cycloalkyl, and heterocycloalkyl, or R ⁇ is taken together with i to form: heterocycloalkyl, heterocycloalkyl-O-, heterocycloalkyl-NH-, heterocycloalkyl-CH2-, oxoheterocycloalkyl, oxoheterocycloalkyl-O-, oxoheterocycloalkyl-N-, or oxoheterocycloalkyl-CH2-;
  • R2 is selected from: hydrogen, Ci-6alkyl, substituted Ci -6alkyl, cycloalkyl, and heterocycloalkyl, or R ⁇ is taken together with l_2 to form: heterocycloalkyl, heterocycloalkyl-O-, heterocycloalkyl-NH-, heterocycloalkyl-CH2-, oxoheterocycloalkyl, oxoheterocycloalkyl-O-, oxoheterocycloalkyl-N-, or oxoheterocycloalkyl-CH2-; are independently 0 or 1 ; and z : 3 and z ⁇ are independently an integer from 0 to 5; or a salt thereof including a pharmaceutically acceptable salt thereof.
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (I).
  • c ⁇ ⁇ and c12 are independently selected from: hydrogen, Ci -4alkyl, and Ci -4alkyl substituted from 1 to 3 times by fluoro;
  • X 1 3 and X 1 5 are independently selected from: hydrogen, Ci -4alkyl, and Ci -4alkyl substituted from 1 to 3 times by fluoro, or X 3 and X 3 are taken together to form oxo ;
  • c14 and c ⁇ ® are independently selected from: hydrogen, Ci -4alkyl, and Ci -4alkyl substituted from 1 to 3 times by fluoro, or X ⁇ and X® are taken together to form oxo ;
  • g1 ⁇ and g12 are independently selected from: -C(O)-, Ci -3alkyene, -S(0)2-, and -S(O)-;
  • L1 1 is selected from: a bond, -NH-, -N(Cl -4alkyl)-, -N (substituted Cl -4alkyl)-, -0-, -S-, -S(O)-, -S(0)2-, cycloalkyl, -O-cycloalkyl, cycloalkyl-O-, -NH-cycloalkyl, cycloalkyl-NH-, -CH2-cycloalkyl, cycloalkyl-CH2-, azetidinyl, -O-azetidinyl, azetidinyl-O-, -N-azetidinyl, azetidinyl-N-, substituted or unsubstituted Ci -6alkylene and substituted or unsubstituted Ci -6heteroalkylene, or,
  • heterocycloalkyl heterocycloalkyl, heterocycloalkyl-O-, heterocycloalkyl-NH-, heterocycloalkyl-CH2-, oxoheterocycloalkyl, oxoheterocycloalkyl-O-, oxoheterocycloalkyl-N-, or oxoheterocycloalkyl-CH2-,
  • i 1 is taken together with an 5 substituent adjacent to the point of attachment of i 1 to form a cycloalkyl ring, a heterocycloalkyl ring, or heteroaryl ring fused t
  • heterocycloalkyl heterocycloalkyl-O-, heterocycloalkyl-NH-,
  • R 1 7 is selected from: hydrogen, Ci-6alkyl and Ci -6alkyl substituted 1 to 6 times by fluoro;
  • R 1 5 is selected from: fluoro, chloro, bromo, iodo, -C(0)0Cl -4alkyl, -OH, -NH2,
  • R x is selected from hydrogen, Ci-4alkyl, and cycloalkyl, and RY is selected from Ci-4alkyl, and cycloalkyl
  • R x is selected from hydrogen, Ci-4alkyl, and cycloalkyl
  • RY is selected from Ci-4alkyl, and cycloalkyl
  • substituted or unsubstituted Ci-6alkyl substituted or unsubstituted Ci-6heteroalkyl
  • substituted or unsubstituted cycloalkyl substituted or unsubstituted heterocycloalkyl, substituted or
  • two adjacent R ⁇ substituents can combine to form a cycloalkyl ring, a heterocycloalkyl ring, or a heteroaryl ring fused to C ⁇ ,
  • js selected from: fluoro, chloro, bromo, iodo, -C(0)0Cl -4alkyl, -OH, -NH2,
  • R ⁇ ® substituents combine to form a cycloalkyl ring, a heterocycloalkyl ring, or heteroaryl ring fused to ,
  • 2 to combines with
  • C ' ' and D ' ' are independently phenyl or pyridyl;
  • R 1 1 is selected from: hydrogen, Ci -6alkyl, substituted Ci -6alkyl, cycloalkyl, and heterocycloalkyl, or R ⁇ ⁇ is taken together with i ⁇ to form: heterocycloalkyl, heterocycloalkyl-O-, heterocycloalkyl-NH-, heterocycloalkyl-CH2-, oxoheterocycloalkyl, oxoheterocycloalkyl-O-, oxoheterocycloalkyl-N-, or oxoheterocycloalkyl-CH2-;
  • R12 is selected from: hydrogen, Ci -6alkyl, substituted Ci -6alkyl, cycloalkyl, and heterocycloalkyl, or R ⁇ js taken together with to form: heterocycloalkyl, heterocycloalkyl-O-, heterocycloalkyl-NH-, heterocycloalkyl-CH2-, oxoheterocycloalkyl, oxoheterocycloalkyl-O-, oxoheterocycloalkyl-N-, or oxoheterocycloalkyl-CH2-; are independently 0 or 1 ; and
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (II).
  • L21 is selected from: a bond, -CH2-, -NH-, -N(Cl -4alkyl)-, -N(Cl-4alkyl substituted from 1 to 3 times by fluoro)-, -0-, -CH2-O-, -O-CH2-, -O-CH2-CH2-, -CH2-CH2-O-, cyclopropyl, -O-cyclopropyl, cyclopropyl-O-,
  • l_22 is selected from: a bond, -CH2-, -NH-, -N(Cl -4alkyl)-, -N(Cl-4alkyl substituted from 1 to 3 times by fluoro)-, -0-, -CH2-O-, -O-CH2-, -O-CH2-CH2-, -CH2-CH2-O-, cyclopropyl, -O-cyclopropyl, cyclopropyl-O-, cyclopropyl-O-, cyclopropyl-O-,
  • imidazolidinyl imidazolidinyl, imidazolidinyl-CH2-, pyrrolidinyl, pyrrolidinyl-O-, pyrrolidinyl-NH-, pyrrolidinyl-CH2-, oxopyrrolidinyl, oxopyrrolidinyl-O-, oxopyrrolidinyl-NH-, or oxopyrrolidinyl-CH2-;
  • R23 and R24 are independently O, or S;
  • R25 is selected from: fluoro, chloro, bromo, iodo, Cl -4alkyl, -C(0)0Cl -4alkyl, -OH, -NH2, -C(0)NHCl-4alkyl, -OCl-4alkyl, -OCH2Ph, -C(0)Ph, -CF3, -CN,
  • R26 is selected from: fluoro, chloro, bromo, iodo, Cl -4alkyl, -C(0)0Cl -4alkyl, -OH, -NH2, -C(0)NHCl-4alkyl, -OCl-4alkyl, -OCH2Ph, -C(0)Ph, -CF3, -CN,
  • c2 and D2 are independently phenyl or pyridyl
  • R21 is selected from: hydrogen, Ci -6alkyl, and cyclopropyl
  • R21 is taken together with l_21 to form:
  • imidazolidinyl imidazolidinyl, imidazolidinyl-CH2-, pyrrolidinyl, pyrrolidinyl-O-, pyrrolidinyl-NH-, pyrrolidinyl-CH2-, oxopyrrolidinyl, oxopyrrolidinyl-O-, oxopyrrolidinyl-NH-, or oxopyrrolidinyl-CH2-;
  • R22 is selected from: hydrogen, Ci -6alkyl, and cyclopropyl
  • R22 is taken together with l_22 to form:
  • z23 and z ⁇ 4 are independently an integer from 0 to 3; or a salt thereof including a pharmaceutically acceptable salt thereof.
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (III). Included in the compounds of the invention are:
  • the compounds of the invention include:
  • the adjacent“R*” and“L*” moieties form a ring, such as a heterocycloalkyl, for example a pyrrolidinyl, the“R*” and“L*” moieties do not have to be adjacent in the ring.
  • R 5 , R 6 , R 15 , R 16 , R 25 and R 26 are indicated by“is selected from...”.
  • each R 5 can be a different substituent.
  • one R 5 can be F and the other R 5 can be Cl.
  • R 5 is selected from: fluoro, chloro, bromo, iodo, -OCH3, -OCH2Ph, -C(0)Ph, -CF3, -CN, -S(0)CH3, -OH, -NH2, -COOH, -CONH2,
  • R 5 is independently fluoro, chloro, bromo, iodo,
  • R 5 is -F. In embodiments,
  • R 5 is -Cl. In embodiments, R 5 is -Br. In embodiments, R 5 is -I. In embodiments, R 5 is substituted or unsubstituted Ci_ 6 alkyl, substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 5 is unsubstituted Ci_ 6 alkyl, unsubstituted heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl. In embodiments, R 5 is -
  • R 5 is -OCH2Ph. In embodiments, R 5 is -CH3. In embodiments,
  • R 5 is -OH. In embodiments, R 5 is -CF3. In embodiments, R 5 is -CN. In embodiments, R 5 is -S(0)CH3. In embodiments, R 5 is -NO2. In embodiments, R 5 is -C(0)CH3. In embodiments, R 5 is -C(0)Ph. In embodiments, R 5 is -CH(CH3)2. In embodiments, R 5 is -
  • R 5 is -CH2 SCH. In embodiments, R 5 is -SO3H. In embodiments, R 5 is -SO2NH2. In embodiments, R 5 is -NHC(0)NH2. In embodiments, R 5 is -NHC(0)H. In embodiments, R 5 is -NHOH. In embodiments, R is -OCF3. In embodiments, R 5 is -OCHF2.
  • R 6 is selected from: fluoro, chloro, bromo, iodo, -OCH3,
  • Ci_ 6 alkyl substituted or unsubstituted heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 6 is independently fluoro, chloro, bromo, iodo, -OCH3, - OCH2Ph, -CH3, -OH, -CF3, -CN, -S(0)CH3, -NO2, -C(0)CH3, -C(0)Ph, -CH(CH3)2, or -C ⁇ CH.
  • R 6 is -F.
  • R 6 is -Cl.
  • R 6 is -Br.
  • R 6 is -I.
  • R 6 is substituted or unsubstituted Ci_ 6 alkyl, substituted or unsubstituted C1 -6heteroalkyl, substituted or unsubstituted cycloalkyl, substituted or unsubstituted heterocycloalkyl, substituted or unsubstituted aryl, or substituted or unsubstituted heteroaryl.
  • R 6 is unsubstituted Ci_ 6 alkyl, unsubstituted C1 -6heteroalkyl, unsubstituted cycloalkyl, unsubstituted heterocycloalkyl, unsubstituted aryl, or unsubstituted heteroaryl.
  • R 6 is -OCH3. In embodiments, R 6 is -OCH2Ph. In embodiments, R 6 is - CH3. In embodiments, R 6 is -OH. In embodiments, R 6 is -CF3. In embodiments, R 6 is - CN. In embodiments, R 6 is -S(0)CH3. In embodiments, R 6 is -NO2. In embodiments, R 6 is -C(0)CH3. In embodiments, R 6 is -C(0)Ph. In embodiments, R 6 is -CH(CH3)2. In embodiments, R 6 is -C ⁇ CH. In embodiments, R 6 is -CH2C ⁇ CH. In embodiments, R 6 is - SO3H. In embodiments, R 6 is -SO2NH2.
  • R 6 is -NHC(0)NH2. In embodiments, R 6 is -NHC(0)H. In embodiments, R 6 is -NHOH. In embodiments, R 6 is - OCF3. In embodiments, R 6 is -OCHF2.
  • R 3 is NR a .
  • R 3 is NH. In embodiments, R 3 is O. In embodiments, R 3 is S. In embodiments, R 3 is CH2.
  • R 4 is NR a .
  • R 4 is NH. In embodiments, R 4 is O. In embodiments, R 4 is S. In embodiments, R 4 is CH2.
  • R 3 and R 4 are NH. In embodiments, R 3 and R 4 are O. In embodiments, R 3 and R 4 are S. In embodiments, R 3 and R 4 are NR a .
  • L 2 is a bond. In embodiments, L 2 is a substituted or unsubstituted Ci_ 6 alkylene. In embodiments, L 2 is a substituted or unsubstituted Ci- 6 heteroalkylene. In embodiments, L 2 is a bond, -O-, -S-, -NH-, -S(O)-, or -S(0)2-. In embodiments, L 2 is a bond or substituted or unsubstituted Ci- 6 alkylene. In embodiments, L 2 is a bond, -O-, or - NH-. In embodiments, L 2 is a bond. In embodiments, L 2 is -O-. In embodiments, L 2 is - S-.
  • L 2 is -NH-. In embodiments, L 2 is -S(O)-. In embodiments, L 2 is - S(0)2-. In embodiments, L 2 is a substituted or unsubstituted C-i-Csheteroalkylene. In embodiments, L 2 is an unsubstituted C-i-Csheteroalkylene. In embodiments, L 2 is a substituted or unsubstituted Ci-C 4 heteroalkylene. In embodiments, L 2 is an
  • L 2 is a substituted or unsubstituted Ci-C3heteroalkylene. In embodiments, L 2 is an unsubstituted C1-C3 heteroalkylene. In embodiments, L 2 is a substituted C-i-Csheteroalkylene. In embodiments, L 2 is a substituted Ci-C6heteroalkylene. In embodiments, L 2 is a substituted C1- C 4 heteroalkylene. In embodiments, L 2 is a Ci-C6heteroalkylene substituted with -CF3.
  • L 2 is cyclopropyl. In embodiments, L 2 is -CH2-cycloalkyl. In embodiments, L 2 is cycloalkyl-CH2-.
  • L 1 is a bond. In embodiments, L 1 is a substituted or unsubstituted Ci_ 6 alkylene. In embodiments, L 1 is a substituted or unsubstituted Ci- 6 heteroalkylene. In embodiments, L 1 is a bond, -0-, -S-, -NH-, -S(O)-, or -S(0)2-. In embodiments, L 1 is a bond or substituted or unsubstituted Ci- 6 alkylene. In embodiments, L 1 is a bond, -0-, or - NH-. In embodiments, L 1 is a bond. In embodiments, L 1 is -0-. In embodiments, L 1 is - S-.
  • L 1 is -NH-. In embodiments, L 1 is -S(O)-. In embodiments, L 1 is - S(0)2-. In embodiments, L 1 is a substituted or unsubstituted C-i-Csheteroalkylene. In embodiments, L 1 is an unsubstituted C-i-Csheteroalkylene. In embodiments, L 1 is a substituted or unsubstituted Ci-C 4 heteroalkylene. In embodiments, L 1 is an
  • L 1 is a substituted or unsubstituted Ci-C3heteroalkylene. In embodiments, L 1 is an unsubstituted C1-C3 heteroalkylene. In embodiments, L 1 is a substituted C-i-Csheteroalkylene. In embodiments, L 1 is a substituted Ci-C6heteroalkylene. In embodiments, L 1 is a substituted C1- C 4 heteroalkylene. In embodiments, L 1 is a Ci-C6heteroalkylene substituted with -CF3. In embodiments, L 1 is cyclopropyl. In embodiments, L 1 is -CH2-cycloalkyl.
  • L 1 is cycloalkyl-CH2-. In embodiments, L 1 is taken together with R 1 to form heterocycloalkyl.
  • the heterocycloalkyl is imidazolidinyl or pyrrolidinyl.
  • the heterocycloalkyl is imidazolidinyl.
  • the heterocycloalkyl is pyrrolidinyl.
  • L 2 is taken together with R 2 to form heterocycloalkyl.
  • the heterocycloalkyl is imidazolidinyl or pyrrolidinyl.
  • the heterocycloalkyl is imidazolidinyl.
  • the heterocycloalkyl is pyrrolidinyl.
  • L 12 is taken together with R 12 to form heterocycloalkyl.
  • the heterocycloalkyl is imidazolidinyl or pyrrolidinyl.
  • the heterocycloalkyl is imidazolidinyl.
  • the heterocycloalkyl is pyrrolidinyl.
  • L 1 3 is taken together with R 1 3 to form heterocycloalkyl.
  • the heterocycloalkyl is imidazolidinyl or pyrrolidinyl.
  • the heterocycloalkyl is imidazolidinyl.
  • the heterocycloalkyl is pyrrolidinyl.
  • the symbol z ⁇ is 0. In embodiments, the symbol z ⁇ is 1 . In embodiments, the symbol z ⁇ is 0. In embodiments, the symbol z ⁇ is 1 . In embodiments, the symbols z2 and z ⁇ are 0. In embodiments, the symbols z2 and z ⁇ are 1 . In embodiments, the symbol z is 0. In embodiments, the symbol z ⁇ is 1 . In embodiments, the symbol z ⁇ is 2. In embodiments, the symbol z ⁇ is 3. In embodiments, the symbol z ⁇ is 4. In embodiments, the symbol z ⁇ is 0. In embodiments, the symbol z ⁇ is 1 . In embodiments, the symbol z ⁇ is 2. In embodiments, the symbol z ⁇ is 3. In embodiments, the symbol z ⁇ is 4. In embodiments, the symbol z ⁇ is 0. In embodiments, the symbol z ⁇ is 1 . In embodiments, the symbol z ⁇ is 2. In embodiments, the symbol z ⁇ is 3. In embodiments, the symbol z ⁇ is 4. In embodiments, the symbol z
  • the compounds of the invention are symmetrical.
  • R21 are equal to R26, Z ⁇ 4, D ⁇ ,
  • salts, including pharmaceutically acceptable salts, of the compounds according to Formula (I) may be prepared. Indeed, in certain embodiments of the invention, salts including pharmaceutically-acceptable salts of the compounds according to Formula (I) may be preferred over the respective free or unsalted compound. Accordingly, the invention is further directed to salts, including pharmaceutically-acceptable salts, of the compounds according to Formula (I).
  • salts including pharmaceutically acceptable salts, of the compounds of the invention are readily prepared by those of skill in the art.
  • the salts of the present invention are pharmaceutically acceptable salts.
  • Salts encompassed within the term “pharmaceutically acceptable salts” refer to non-toxic salts of the compounds of this invention.
  • Representative pharmaceutically acceptable acid addition salts include, but are not limited to, 4-acetamidobenzoate, acetate, adipate, alginate, ascorbate, aspartate, benzenesulfonate (besylate), benzoate, bisulfate, bitartrate, butyrate, calcium edetate, camphorate, camphorsulfonate (camsylate), caprate (decanoate), caproate (hexanoate), caprylate (octanoate), cinnamate, citrate, cyclamate, digluconate, 2,5-dihydroxybenzoate, disuccinate, dodecylsulfate (estolate), edetate (ethylenediaminetetraacetate), estolate (lauryl sulfate), ethane-1 ,2-disulfonate (edisylate), ethanesulfonate (esylate), formate, fumarate, galactarate
  • Representative pharmaceutically acceptable base addition salts include, but are not limited to, aluminium, 2-amino-2-(hydroxymethyl)-1 ,3-propanediol (TRIS,
  • tromethamine arginine, benethamine (/V-benzylphenethylamine), benzathine (A/, A/ - dibenzylethylenediamine), b/s-(2-hydroxyethyl)amine, bismuth, calcium, chloroprocaine, choline, clemizole (1-p chlorobenzyl-2-pyrrolildine-1’-ylmethylbenzimidazole),
  • cyclohexylamine dibenzylethylenediamine, diethylamine, diethyltriamine, dimethylamine, dimethylethanolamine, dopamine, ethanolamine, ethylenediamine, L-histidine, iron, isoquinoline, lepidine, lithium, lysine, magnesium, meglumine (/V-methylglucamine), piperazine, piperidine, potassium, procaine, quinine, quinoline, sodium, strontium, t- butylamine, and zinc.
  • the compounds according to Formula (I) may contain one or more asymmetric centers (also referred to as a chiral center) and may, therefore, exist as individual enantiomers, diastereomers, or other stereoisomeric forms, or as mixtures thereof.
  • Chiral centers such as chiral carbon atoms, may be present in a substituent such as an alkyl group.
  • compounds according to Formula (I) containing one or more chiral centers may be used as racemic mixtures, enantiomerically or diastereomerically enriched mixtures, or as enantiomerically or diastereomerically pure individual stereoisomers.
  • the compounds according to Formula (I) and pharmaceutically acceptable salts thereof may contain isotopically-labelled compounds, which are identical to those recited in Formula (I) and following, but for the fact that one or more atoms are replaced by an atom having an atomic mass or mass number different from the atomic mass or mass number usually found in nature.
  • isotopes include isotopes of hydrogen, carbon, nitrogen, oxygen, phosphorous, sulphur, fluorine, iodine, and chlorine, such as 2H, 3H, 11 C, 13C, 14C, 15N, 170, 180, 31 P, 32P, 35S, 18F, 36CI, 1231 and 1251.
  • Isotopically-labelled compounds for example those into which radioactive isotopes such as 3H or 14C are incorporated, are useful in drug and/or substrate tissue distribution assays. Tritiated, i.e., 3H, and carbon-14, i.e., 14C, isotopes are particularly preferred for their ease of preparation and detectability. 11 C and 18F isotopes are particularly useful in PET (positron emission tomography), and 1251 isotopes are particularly useful in SPECT (single photon emission computerized tomography), both are useful in brain imaging.
  • Isotopically labelled compounds can generally be prepared by substituting a readily available isotopically labelled reagent for a non-isotopically labelled reagent.
  • the compounds according to Formula (I) may also contain double bonds or other centers of geometric asymmetry. Where the stereochemistry of a center of geometric asymmetry present in Formula (I), or in any chemical structure illustrated herein, is not specified, the structure is intended to encompass the trans (E) geometric isomer, the cis (Z) geometric isomer, and all mixtures thereof. Likewise, all tautomeric forms are also included in Formula (I) whether such tautomers exist in equilibrium or predominately in one form.
  • the compounds of Formula (I) or salts, including pharmaceutically acceptable salts, thereof may exist in solid or liquid form.
  • the compounds of the invention may exist in crystalline or noncrystalline form, or as a mixture thereof.
  • pharmaceutically acceptable solvates may be formed wherein solvent molecules are incorporated into the crystalline lattice during crystallization.
  • Solvates wherein water is the solvent that is incorporated into the crystalline lattice are typically referred to as "hydrates.” Hydrates include stoichiometric hydrates as well as compositions containing vaiable amounts of water.
  • polymorphs may have the same chemical composition but differ in packing, geometrical arrangement, and other descriptive properties of the crystalline solid state. Polymorphs, therefore, may have different physical properties such as shape, density, hardness, deformability, stability, and dissolution properties. Polymorphs typically exhibit different melting points, IR spectra, and X-ray powder diffraction patterns, which may be used for identification.
  • polymorphs may be produced, for example, by changing or adjusting the reaction conditions or reagents, used in making the compound. For example, changes in temperature, pressure, or solvent may result in polymorphs. In addition, one polymorph may spontaneously convert to another polymorph under certain conditions.
  • Alkyl and“alkylene”, and derivatives thereof, refer to a hydrocarbon chain having the specified number of“member atoms”. Alkyl being monovalent and alkylene being bivalent. For example, C- j -Cg alkyl refers to an alkyl group having from 1 to 6 member atoms. Alkyl and alkylene groups may be saturated, unsaturated, straight or branched. Representative branched alkyl groups have one, two, or three branches.
  • Alkyl and alkylene include: methyl, ethyl, ethylene, propyl (n-propyl and isopropyl), butene, butyl (n-butyl, isobutyl, and t-butyl), pentyl and hexyl.
  • Aryl refers to an aromatic hydrocarbon ring.
  • Aryl groups are monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring member atoms, wherein at least one ring system is aromatic and wherein each ring in the system contains 3 to 7 member atoms, such as phenyl, naphthalene, tetrahydronaphthalene and biphenyl.
  • aryl is phenyl.
  • Cycloalkyl unless otherwise defined, refers to a saturated or unsaturated non aromatic hydrocarbon ring having from three to seven carbon atoms. Cycloalkyl groups are monocyclic ring systems.
  • C3-C7 cycloalkyl refers to a cycloalkyl group having from 3 to 7 member atoms.
  • cycloalkyl as used herein include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptyl.
  • cycolalkyl is selected from: cyclopropyl, cyclobutyl and cyclohexyl.
  • cycolalkyl is cyclopropyl.
  • Halo refers to fluoro, chloro, bromo, and iodo.
  • Heteroaryl refers to a monocyclic aromatic 4 to 8 member ring containing 1 to 7 carbon atoms and containing 1 to 4 heteroatoms, provided that when the number of carbon atoms is 3, the aromatic ring contains at least two heteroatoms, or to such aromatic ring is fused one or more rings, such as heteroaryl rings, aryl rings, heterocyclic rings, or cycloalkyl rings. Heteroaryl groups containing more than one heteroatom may contain different heteroatoms.
  • Heteroaryl includes but is not limited to: benzoimidazolyl, benzothiazolyl, benzothiophenyl, benzopyrazinyl, benzotriazolyl, benzotriazinyl, benzo[1 ,4]dioxanyl, benzofuranyl, 9/-/-a-carbolinyl, cinnolinyl, furanyl, pyrazolyl, imidazoiy!, indolizinyl, naphthyridinyl, oxazolyl, oxothiadiazolyl, oxadiazolyl, phthalazinyl, pyridyl, pyrrolyl, purinyl, pteridinyl, phenazinyl, pyrazolopyrimidinyl, pyrazolopyridinyl, pyrrolizinyl, pyrimidyl, isothiazolyl, furazanyl, pyrimidinyl,
  • heteroaryl is selected from: pyrazolyl, imidazolyi, oxazolyl and thienyl.
  • heteroaryl is a pyridyl group or an imidazolyi group.
  • heteroaryl is a pyridyl.
  • Heterocycloalkyl refers to a saturated or unsaturated non-aromatic ring containing 4 to 12 member atoms, of which 1 to 11 are carbon atoms and from 1 to 6 are heteroatoms. Heterocycloalkyl groups containing more than one heteroatom may contain different heteroatoms. Heterocycloalkyl groups are monocyclic ring systems or a monocyclic ring fused with an aryl ring or to a heteroaryl ring having from 3 to 6 member atoms.
  • Heterocycloalkyl includes: pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, pyranyl, tetrahydropyranyl, dihydropyranyl, tetra hydrothienyl, pyrazolidinyl, oxazolidinyl, imidazolidinyl, oxetanyl, thiazolidinyl, piperidinyl, homopiperidinyl, piperazinyl, morpholinyl, thiamorpholinyl, 1 ,3-dioxolanyl, 1 ,3-dioxanyl, 1 ,4-dioxanyl, 1 ,3-oxathiolanyl, 1 ,3-oxathianyl, 1 ,3-dithianyl, 1 ,3oxazolidin-2-one, hexahydro-1 H-azepin, 4,5,6,7,tetrahydro-1
  • heterocycloalkyl includes: piperidinyl, tetrahydrofuranyl, tetrahydropyranyl, imidazolidinyl, oxetanyl, and pyrrolidinyl.
  • heterocycloalkyl is selected from: imidazolidinyl, tetrahydropyranyl and pyrrolidinyl.
  • Heteroatom refers to a nitrogen, sulfur or oxygen atom.
  • Heteroalkyl and“heteroalkylene” by itself or in combination with another term, means, unless otherwise stated, a non-cyclic stable straight or branched chain, or combinations thereof, including at least one carbon atom (and up to the number specified) and at least one heteroatom selected from the group consisting of O, N, P, Si, and S, and wherein the nitrogen and sulfur atoms may optionally be oxidized, and the nitrogen heteroatom may optionally be quaternized.
  • Ci-6heteroalkyl(ene) contains at least one and up to 6 carbon atoms, in addition to at least one heteroatom.
  • Heteroalkyl being monovalent and heteroalkylene being bivalent.
  • heteroalkyl and heteroalkylene groups may be taken together with another substituent to form a heterocycloalkyl group.
  • the heteroatom(s) O, N, P, S, and Si may be placed at any interior position of the heteroalkyl or heteroalkylene group or at the position at which the alkyl group is attached to the remainder of the molecule.
  • Heteroalkyl examples include, but are not limited to:
  • Heteroalkylene examples include, but are not limited to:
  • -O-CH2-CH2- Up to two or three heteroatoms may be consecutive, such as, for example, -CH2-NH-OCH3 and -CH2-0-Si(CH3)3.
  • “Substituted” as used herein, unless otherwise defined, is meant that the subject chemical moiety has from one to nine substituents, suitably from one to five substituents, selected from the group consisting of: fluoro,
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents independently selected from: fluoro, oxo, -OH,
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents independently selected from: fluoro, oxo, -OH, -COOH, -NH2, and -CN,
  • R x is selected from Ci -6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x is selected from Ci -6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x1 and R 5 2 are each independently selected from Ci -6alkyl, and Ci -6alkyl substituted with from 1 to 6
  • Ci -6alkyl selected from Ci -6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x1 and R 5 2 are each independently selected from Ci-6alkyl, and Ci-6alkyl substituted with from 1 to 6
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x1 and R 5 2 are each independently selected from Ci-6alkyl, and Ci-6alkyl substituted with from 1 to 6
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x is selected from Ci-6alkyl, and Ci -6alkyl substituted with from 1 to 6 substituents
  • R x1 and R 5 2 are each independently selected from Ci-6alkyl, and Ci-6alkyl substituted with from 1 to 6
  • Substituents independently selected from: fluoro, oxo, -OH, -COOH, -NH2, and -CN,
  • Suitably“substituted” means the subject chemical moiety has from one to four substituents selected from the group consisting of: fluoro,
  • -OCi -4alkyl substituted with from 1 to 4 substituents independently selected from: fluoro, oxo, -OH, -COOH, -NH2, and -CN,
  • R x is selected from Ci-4alkyl, and Ci -6alkyl substituted one to 4 times by fluoro,
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro,
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro,
  • Suitably“substituted” means the subject chemical moiety has from one to four substituents selected from the group consisting of: fluoro,
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to 4 times by fluoro
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro,
  • R x is selected from Ci-4alkyl, and Ci -6alkyl substituted one to 4 times by fluoro
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x is selected from Ci-4alkyl, and Ci -4alkyl substituted one to four times by fluoro,
  • R x1 and R 5 2 are each independently selected from Ci-4alkyl, and Ci-4alkyl substituted one to four times by fluoro,
  • ACN acetonitrile
  • Boc tert-Butoxycarbonyl
  • CAN cerric ammonium nitrate
  • C18 refers to 18-carbon alkyl groups on silicon in HPLC stationary phase); CH 3 CN (acetonitrile);
  • DIAD diisopropyl azodicarboxylate
  • HEPES (4-(2-hydroxyethyl)-1 -piperazine ethane sulfonic acid);
  • IPA isopropyl alcohol
  • Li0H.H 2 0 lithium hydroxide monohydrate
  • NaCNBH 3 sodium cyanoborohydride
  • NaOH sodium hydroxide
  • TFA trifluoroacetic acid
  • THF tetrahydrofuran
  • T3P® ® (2,4,6-tripropyM ,3,5,2,4,6-trioxatriphosphorinane-2,4,6-trioxide).
  • the compounds according to Formula (I) are prepared using conventional organic synthetic methods.
  • a suitable synthetic route is depicted below in the following general reaction scheme. All of the starting materials are commercially available or are readily prepared from commercially available starting materials by those of skill in the art.
  • a substituent described herein is not compatible with the synthetic methods described herein, the substituent may be protected with a suitable protecting group that is stable to the reaction conditions.
  • the protecting group may be removed at a suitable point in the reaction sequence to provide a desired intermediate or target compound.
  • suitable protecting groups and the methods for protecting and de-protecting different substituents using such suitable protecting groups are well known to those skilled in the art; examples of which may be found in T. Greene and P. Wuts, Protecting Groups in Organic Synthesis (4th ed.), John Wiley & Sons, NY (2006).
  • a substituent may be specifically selected to be reactive under the reaction conditions used. Under these circumstances, the reaction conditions convert the selected substituent into another substituent that is either useful as an intermediate compound or is a desired substituent in a target compound.
  • r represents all corresponding positional combinations on all of the Formulas disclosed herein.
  • Substituted tetrahydrofurans with general structure (I) can be prepared through the synthetic sequence outlined below. Reduction of commercially-available dimethyl furan-3,4-dicarboxylate (A) provides a separable mixture of syn- (B) and anti- (C) isomers. Syn-isomer B can be subsequently reduced with LAH to provide diol D, which can be converted to bis-mesylate E through standard conditions. Wth E in hand, treatment with NaN 3 followed by palladium-mediated hydrogenation procures diamine G. Diamidation with carboxylic acid H mediated by a coupling reagent such as T3P can provide targeted compounds of generic structure I. The corresponding anf/ ' -isomer of I can be analogously synthesized from C.
  • the compounds according to Formula (I) and pharmaceutically acceptable salts thereof are inhibitors of the ATF4 pathway.
  • Compounds which are inhibitors of the ATF4 pathway are readily identified by exhibiting activity in the ATF4 Cell Based Assay below. These compounds are potentially useful in the treatment of conditions wherein the underlying pathology is attributable to (but not limited to) modulation of the elF2alpha pathway, for example, neurodegenerative disorders, cancer, cardiovascular and metabolic diseases. Accordingly, in another aspect the invention is directed to methods of treating such conditions.
  • the pharmaceutically active compounds within the scope of this invention are useful as ATF4 pathway inhibitors in mammals, particularly humans, in need thereof.
  • the Integrated Stress Response is a collection of cellular stress response pathways that converge in phosphorylation of the translation initiation factor elF2a resulting in a reduction in overall translation in cells.
  • Mammalian cells have four elF2a kinases that phosphorylate this initiation factor in the same residue (serine 51); PERK is activated by the accumulation of unfolded proteins in the endoplasmic reticulum (ER), GCN2 is activated by amino acid starvation, PKR by viral infection and HRI by heme deficiency. Activation of these kinases decreases bulk protein synthesis but it also culminates in increased expression of specific mRNAs that contain uORFs.
  • An integrated stress response-associated disease is a disease characterized by increased activity in the integrated stress response (e.g. increased phosphorylation of elF2a by an elF2a kinase compared to a control such as a subject without the disease).
  • a disease associated with phosphorylation of elF2a is disease characterized by an increase in phosphorylation of elF2a relative to a control, such as a subject without the disease.
  • PERK Activation of PERK occurs upon ER stress and hypoxic conditions and its activation and effect on translation has been shown to be cytoprotective for tumor cells [17] Adaptation to hypoxia in the tumor microenvironment is critical for survival and metastatic potential. PERK has also been shown to promote cancer proliferation by limiting oxidative DNA damage and death [18, 19] Moreover, a newly identified PERK inhibitor has been shown to have antitumor activity in a human pancreatic tumor xenograft model [20] Compounds disclosed herein decrease the viability of cells that are subjected to ER-stress. Thus, pharmacological and acute inhibition of the PERK branch with the compounds disclosed herein results in reduced cellular fitness. During tumor growth, compounds disclosed herein, that block the cytoprotective effects of elF2a phosphorylation upon stress may prove to be potent anti-proliferative agents.
  • Prolonged ER stress leads to the accumulation of CHOP, a pro-apoptotic molecule.
  • CHOP a pro-apoptotic molecule.
  • overexpression of the phosphatase of elF2a increased survival of prion- infected mice whereas sustained elF2a phosphorylation decreased survival [22]
  • the restoration of protein translation rates during prion disease was shown to rescue synaptic deficits and neuronal loss.
  • the compounds disclosed herein that make cells insensitive to elF2a phosphorylation sustain protein translation. Compounds disclosed herein could prove potent inhibitors of neuronal cell death in prion disease by blocking the deleterious effects of prolonged elF2a phosphorylation.
  • tissue-specific pathology that is linked to heightened elF2a phosphorylation is the fatal brain disorder, vanishing white matter disease (VWM) or childhood ataxia with CNS hypomyelination (CACH).
  • VWM vanishing white matter disease
  • CACH CNS hypomyelination
  • This disease has been linked to mutation in elF2B, the GTP exchange factor that is necessary for elF2 function in translation [23] elF2a phosphorylation inhibits the activity of elF2B and mutations in this exchange factor that reduce its exchange activity exacerbate the effects of elF2a phosphorylation.
  • the severe consequences of the CACH mutations point to the dangers of UPR hyper-activation, especially as it pertains to the myelin-producing oligodendrocyte.
  • Small molecules, such as compounds disclosed herein, that block signaling through elF2a phosphorylation may reduce the deleterious effects of its hyperactivation in VWM.
  • a method of improving long-term memory in a patient which comprises administering a therapeutically effective amount of a compound of Formu la (I) to the patient.
  • the patient is human.
  • the patient is a mammal.
  • the compounds of this invention inhibit the integrated stress response which is implicated in the pathogenesis of neurological disorders.
  • the present invention relates to a method for treating or lessening the severity of neurological disorders.
  • the disorders treatable with the compounds of the invention include: Alcoholism, Anxiety, Depression, Schizophrenia, Bipolar Disorder, Obsessive Compulsive Disorder, Panic Disorder, Chronic Pain, Obesity, Senile Dementia, Migraine, Bulimia, Anorexia, Social Phobia, Pre-Menstrual Syndrome (PMS), Adolescent Depression, Trichotillomania, Dysthymia and Substance Abuse.
  • the neurological disorder is treated in a human patient.
  • Visceral pain is pain associated with the viscera, which encompass the internal organs of the body. These organs include, e.g., the heart, lungs, reproductive organs, bladder, ureters, the digestive organs, liver, pancreas, spleen, and kidneys. There are a variety of conditions in which visceral pain may exist, such as, for exampie, pancreatitis, iabor, abdominal surgery associated with ileus, cystitis, menstrual period, or dysmenorrhea.
  • kidney pain, epigastric pain, pleural pain, and painful biliary colic, appendicitis pain may all be considered to be visceral pain.
  • Substerna! pain or pressure from early myocardial infarction is also visceral.
  • Diseases of the stomach, dudenum or colon can cause visceral pain.
  • Commonly encountered gastrointestinal (Gl) disorders that cause visceral pain include functional bowel disorder (FBD) and inflammatory bowel disease (IBD).
  • Gl disorders include a wide range of disease states that are currently only moderately controlled, including, with respect to FBD, gastroesophageal reflux, dyspepsia, irritable bowel syndrome (IBS) and functional abdominal pain syndrome (PARS), and, with respect to IBD, Crohn’s disease, ileitis and ulcerative colitis, ail of which regularly produce visceral pain.
  • the present invention relates to a method for treating or lessening the severity of pain.
  • the invention can alleviate pain from many causes, including but not limited to shock; limb amputation; severe chemical or thermal bum Injury; sprains, ligament tears, fractures, wounds and other tissue injuries; dental surgery, procedures and maladies; labor and delivery; migraine; during physical therapy; post operative pain; radiation poisoning; cancer: acquired immunodeficiency syndrome (AIDS); epidural (or peridural) fibrosis; failed back surgery and failed laminectomy; sciatica; painful sickle ceil crisis; arthritis; autoimmune disease; intractable bladder pain; and the like.
  • the present invention is directed to the treatment of intractible pain, whatever its cause.
  • pain is treated in a human patient.
  • the compounds of this invention inhibit the unfolded protein response which is implicated in the pathogenesis of inter vertebral disc degeneration.
  • the present invention relates to a method for treating or lessening the severity of vertebral disc degeneration.
  • the compounds set forth herein are provided as pharmaceutical compositions comprising the compound and a pharmaceutically acceptable excipient.
  • the compound, or a pharmaceutically acceptable salt thereof is co-adminstered with a second agent (e.g. therapeutic agent).
  • the compound, or a pharmaceutically acceptable salt thereof is co-adminstered with a second agent (e.g. therapeutic agent), which is administered in a therapeutically effective amount.
  • the second agent is an agent for improving memory.
  • elF2a kinase/s responsible for phosphorylation of elF2a in the brain compounds disclosed herein th at block translation and ATF4 production make them ideal molecules to block the effects of this phosphorylation event on memory.
  • Pharmacological treatment with compounds disclosed herein increase spatial memory and enhance auditory and contextual fear conditioning.
  • Regulators of translation such as the compounds of Formula (I), could serve as therapeutic agents that improve memory in human disorders associated with memory loss such as Alzheimer's disease and in other neurological disorders that activate the UPR in neurons and thus could have negative effects on memory consolidation such as Parkinson's disease, Amyotrophic lateral sclerosis and prion diseases.
  • the compounds of Formula (I) are also useful in applications where increasing protein production output is desirable, such as in vitro cell free systems for protein production.
  • In vitro systems have basal levels of elF2a phosphorylation that reduce translational output [28, 29]
  • production of antibodies by hybridomas may also be improved by addition of compounds disclosed herein.
  • a method of increasing protein expression of a cell or in vitro expression system which comprises administering an effective amount of a compound of Formula (I) to the cell or expression system.
  • the method is a method of increasing protein expression by a cell and includes administering an effective amount of a compound of Formula (I) to the cell.
  • the method is a method of increasing protein expression by an in vitro protein expression system and includes administering an effective amount of a compound of Formu la (I) to the in vitro (e.g. cell free) protein expression system.
  • the compounds set forth herein are provided as pharmaceutical compositions comprising the compound and a pharmaceutically acceptable excipient.
  • the compound, or a pharmaceutically acceptable salt thereof is co-adminstered with a second agent.
  • the compound, or a pharmaceutically acceptable salt thereof is co-adminstered with a second agent, which is administered in a therapeutically effective amount.
  • the second agent is an agent for improving protein expression.
  • the present invention relates to a method for treating or lessening the severity of breast cancer, including inflammatory breast cancer, ductal carcinoma, and lobular carcinoma.
  • the present invention relates to a method for treating or lessening the severity of colon cancer.
  • the present invention relates to a method for treating or lessening the severity of pancreatic cancer, including insulinomas, adenocarcinoma, ductal adenocarcinoma, adenosquamous carcinoma, acinar cell carcinoma, and glucagonoma.
  • the present invention relates to a method for treating or lessening the severity of skin cancer, including melanoma, including metastatic melanoma.
  • the present invention relates to a method for treating or lessening the severity of lung cancer including small cell lung cancer, non-small cell lung cancer, squamous cell carcinoma, adenocarcinoma, and large cell carcinoma.
  • the present invention relates to a method for treating or lessening the severity of cancers selected from the group consisting of brain (gliomas), glioblastomas, astrocytomas, glioblastoma multiforme, Bannayan-Zonana syndrome, Cowden disease, Lhermitte-Duclos disease, Wilm's tumor, Ewing's sarcoma, Rhabdomyosarcoma, ependymoma, medulloblastoma, head and neck, kidney, liver, melanoma, ovarian, pancreatic, adenocarcinoma, ductal adenocarcinoma, adenosquamous carcinoma, acinar cell carcinoma, glucagonoma, insulinoma, prostate, sarcoma, osteosarcoma, giant cell tumor of bone, thyroid, lymphoblastic T cell leukemia, chronic myelogenous leukemia, chronic lymphocytic leukemia,
  • the present invention relates to a method for treating or lessening the severity of pre-cancerous syndromes in a mammal, including a human, wherein the pre- cancerous syndrome is selected from: cervical intraepithelial neoplasia, monoclonal gammapathy of unknown significance (MGUS), myelodysplastic syndrome, aplastic anemia, cervical lesions, skin nevi (pre-melanoma), prostatic intraepithleial (intraductal) neoplasia (PIN), Ductal Carcinoma in situ (DCIS), colon polyps and severe hepatitis or cirrhosis.
  • the pre- cancerous syndrome is selected from: cervical intraepithelial neoplasia, monoclonal gammapathy of unknown significance (MGUS), myelodysplastic syndrome, aplastic anemia, cervical lesions, skin nevi (pre-melanoma), prostatic intraepithleial (intraductal) ne
  • the present invention relates to a method for treating or lessening the severity of neurodegenerative diseases/injury, such as Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeldt- Jakob Disease, and related prion diseases, progressive supranuclear palsy, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, cognitive impairment, atherosclerosis, ocular diseases, arrhythmias, in organ transplantation and in the transportation of organs for transplantation.
  • neurodegenerative diseases/injury such as Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeldt- Jakob Disease,
  • the present invention relates to a method for preventing organ damage during and after organ transplantation and in the transportation of organs for transplantation.
  • the method of preventing organ damage during and after organ transplantation comprises the in vivo administration of a compound of Formula (I).
  • the method of preventing organ damage during the transportation of organs for transplantation comprises adding a compound of Formula (I) to the solution housing the organ during transportation.
  • the present invention relates to a method for treating or lessening the severity of neurodegernative ocular diseases, wherein the disease is retinitis pigmentosa.
  • the present invention relates to a method for treating or lessening the severity of ocular diseases, wherein the disease is selected from retinal dystrophies and corneal dystrophies, such as Fuch’s corneal dystrophy.
  • the present invention relates to a method for treating or lessening the severity of ocular diseases/angiogenesis.
  • the method of treating or lessening the severity of ocular diseases/angiogenesis comprises the in vivo administration of a compound of Formula (I).
  • the disorder of ocular diseases can be: edema or neovascularization for any occlusive or inflammatory retinal vascular disease, such as rubeosis irides, neovascular glaucoma, pterygium, vascularized glaucoma filtering blebs, conjunctival papilloma; choroidal neovascularization, such as neovascular age-related macular degeneration (AMD), myopia, prior uveitis, trauma, or idiopathic; macular edema, such as post surgical macular edema, macular edema secondary to uveitis including retinal and/or choroidal inflammation, macular edema secondary to diabetes, and macular edema secondary to retinovascular occlusive disease (i.e.
  • retinal vascular disease such as rubeosis irides, neovascular glaucoma, pterygium,
  • retinal neovascularization due to diabetes such as retinal vein occlusion, uveitis, ocular ischemic syndrome from carotid artery disease, ophthalmic or retinal artery occlusion, sickle cell retinopathy, other ischemic or occlusive neovascular retinopathies, retinopathy of prematurity, or Eale's Disease; and genetic disorders, such as VonHippel-Lindau syndrome.
  • the neovascular age-related macular degeneration is wet age-related macular degeneration. In other embodiments, the neovascular age-related macular degeneration is dry age-related macular degeneration and the patient is characterized as being at increased risk of developing wet age-related macular degeneration.
  • the ocular disease is treated in a human patient.
  • the methods of treatment of the invention comprise administering an effective amount of a compound according to Formula (I) or a pharmaceutically acceptable salt, thereof to a patient in need thereof.
  • the invention also provides a compound according to Formula (I) or a pharmaceutically-acceptable salt thereof for use in medical therapy, and particularly in therapy for: cancer, pre-cancerous syndromes, Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, C re utzfeldt- Jakob Disease, and related prion diseases, progressive supranuclear palsy, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, cognitive impairment, atherosclerosis, ocular diseases, in organ transplantation and ar
  • the invention also provides a compound according to Formula (I) or a pharmaceutically-acceptable salt thereof for use in preventing organ damage during the transportation of organs for transplantation.
  • the invention is directed to the use of a compound according to Formula (I) or a pharmaceutically acceptable salt thereof in the manfacture of a medicament for the treatment of a disorder characterized by activation of the UPR, such as cancer, pre- cancerous syndromes, Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease, Creutzfeldt- Jakob Disease, and related prion diseases, progressive supranuclear palsy, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, cognitive impairment, atherosclerosis, ocular diseases, in organ transplantation
  • the methods of treatment of the invention comprise administering a safe and effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof to a mammal, suitably a human, in need thereof.
  • treating in reference to a condition means: (1) to ameliorate the condition or one or more of the biological manifestations of the condition, (2) to interfere with (a) one or more points in the biological cascade that leads to or is responsible for the condition or (b) one or more of the biological manifestations of the condition, (3) to alleviate one or more of the symptoms or effects associated with the condition, or (4) to slow the progression of the condition or one or more of the biological manifestations of the condition.
  • treating and derivatives thereof refers to therapeutic therapy.
  • Therapeutic therapy is appropriate to alleviate symptions or to treat at early signs of disease or its progression.
  • Prophylactic therapy or prevention therapy is appropriate when a subject has, for example, a strong family history of neurodegenerative diseases.
  • Prophylactic therapy is appropriate when a subject has, for example, a strong family history of cancer or is otherwise considered at high risk for developing cancer, or when a subject has been exposed to a carcinogen.
  • prevention is not an absolute term. In medicine, “prevention” is understood to refer to the prophylactic administration of a drug to substantially diminish the likelihood or severity of a condition or biological manifestation thereof, or to delay the onset of such condition or biological manifestation thereof.
  • safe and effective amount in reference to a compound of Formula (I), or a pharmaceutically acceptable salt thereof, means an amount of the compound sufficient to treat the patient's condition but low enough to avoid serious side effects (at a reasonable benefit/risk ratio) within the scope of sound medical judgment.
  • a safe and effective amount of the compound will vary with the particular route of administration chosen; the condition being treated; the severity of the condition being treated; the age, size, weight, and physical condition of the patient being treated; the medical history of the patient to be treated; the duration of the treatment; the nature of concurrent therapy; the desired therapeutic effect; and like factors, but can nevertheless be routinely determined by the skilled artisan.
  • patient refers to a human or other mammal, suitably a human.
  • the compounds of Formula (I) or pharmaceutically acceptable salts thereof may be administered by any suitable route of administration, including systemic administration.
  • Systemic administration includes oral administration, and parenteral administration.
  • Parenteral administration refers to routes of administration other than enteral, transdermal, or by inhalation, and is typically by injection or infusion.
  • Parenteral administration includes intravenous, intramuscular, and subcutaneous injection or infusion.
  • the compounds of Formula (I) or pharmaceutically acceptable salts thereof may be administered once or according to a dosing regimen wherein a number of doses are administered at varying intervals of time for a given period of time. For example, doses may be administered one, two, three, or four times per day. Doses may be administered until the desired therapeutic effect is achieved or indefinitely to maintain the desired therapeutic effect. Suitable dosing regimens for a compound of the invention depend on the pharmacokinetic properties of that compound, such as absorption, distribution, and half- life, which can be determined by the skilled artisan.
  • suitable dosing regimens including the duration such regimens are administered, for a compound of the invention depend on the condition being treated, the severity of the condition being treated, the age and physical condition of the patient being treated, the medical history of the patient to be treated, the nature of concurrent therapy, the desired therapeutic effect, and like factors within the knowledge and expertise of the skilled artisan. It will be further understood by such skilled artisans that suitable dosing regimens may require adjustment given an individual patient's response to the dosing regimen or over time as individual patient needs change.
  • a prodrug of a compound of the invention is a functional derivative of the compound which, upon administration to a patient, eventually liberates the compound of the invention in vivo.
  • Administration of a compound of the invention as a prodrug may enable the skilled artisan to do one or more of the following: (a) modify the onset of the compound in vivo; (b) modify the duration of action ofthe compound in vivo; (c) modify the transportation or distribution of the compound in vivo; (d) modify the solubility of the compound in vivo; and (e) overcome a side effect or other difficulty encountered with the compound.
  • esters can be employed, for example methyl, ethyl, and the like for -COOH, and acetate, maleate, and the like for -OH, and those esters known in the art for modifying solubility or hydrolysis characteristics.
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful in the treatment of cancer or pre-cancerous syndromes.
  • co-administration is meant either simultaneous administration or any manner of separate sequential administration of an ATF4 pathway inhibiting compound, as described herein, and a further active agent or agents, known to be useful in the treatment of cancer, including chemotherapy and radiation treatment.
  • further active agent or agents includes any compound or therapeutic agent known to or that demonstrates advantageous properties when administered to a patient in need of treatment for cancer.
  • the compounds are administered in a close time proximity to each other.
  • the compounds are administered in the same dosage form, e.g. one compound may be administered by injection and another compound may be administered orally.
  • any anti-neoplastic agent that has activity versus a susceptible tumor being treated may be co-administered in the treatment of cancer in the present invention.
  • examples of such agents can be found in Cancer Principles and Practice of Oncology by V.T. Devita and S. Heilman (editors), 6 th edition (February 15, 2001), Lippincott Williams & Wilkins Publishers.
  • a person of ordinary skill in the art would be able to discern which combinations of agents would be useful based on the particular characteristics of the drugs and the cancer involved.
  • Typical anti-neoplastic agents useful in the present invention include, but are not limited to, anti-microtubule agents such as diterpenoids and vinca alkaloids; platinum coordination complexes; alkylating agents such as nitrogen mustards, oxazaphosphorines, alkylsulfonates, nitrosoureas, and triazenes; antibiotic agents such as anthracyclins, actinomycins and bleomycins; topoisomerase II inhibitors such as epipodophyllotoxins; antimetabolites such as purine and pyrimidine analogues and antifolate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non-receptor tyrosine kinase angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; cell cycle signaling inhibitors; proteasome inhibitors; and inhibitors of cancer metabolism.
  • anti-microtubule agents such as di
  • Examples of a further active ingredient or ingredients (anti-neoplastic agent) for use in combination or co-administered with the presently invented ATF4 pathway inhibiting compounds are chemotherapeutic agents.
  • the pharmaceutically active compounds of the invention are used in combination with a VEGFR inhibitor, suitably 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride salt thereof, which is disclosed and claimed in in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001 , International Publication Number W002/0591 10 and an International Publication date of August 1 , 2002, the entire disclosure of which is hereby incorporated by reference, and which is the compound of Example 69.
  • a VEGFR inhibitor suitably 5-[[4-[(2,3-dimethyl-2H-indazol-6- yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide, or a pharmaceutically acceptable salt, suitably the monohydrochloride
  • the cancer treatment method of the claimed invention includes the co-administration a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, such as one selected from the group consisting of anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine kinase angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, cell cycle signaling inhibitors; proteasome inhibitors; and inhibitors of cancer metabolism.
  • anti-neoplastic agent such as one selected from the group consisting of anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor t
  • “Chemotherapeutic” or “chemotherapeutic agent” is used in accordance with its plain ordinary meaning and refers to a chemical composition or compound having antineoplastic properties or the ability to inhibit the growth or proliferation of cells.
  • the compounds described herein can be co-administered with conventional immunotherapeutic agents including, but not limited to, immunostimulants (e.g., Bacillus Calmette-Guerin (BCG), levamisole, interleukin-2, alpha-interferon, etc.), monoclonal antibodies (e.g., anti-CD20, anti-HER2, anti-CD52, anti-HLA-DR, and anti- VEGF monoclonal antibodies), immunotoxins (e.g., anti-CD33 monoclonal antibody- calicheamicin conjugate, anti-CD22 monoclonal antibody-pseudomonas exotoxin conjugate, etc.), and radioimmunotherapy (e.g., anti-CD20 monoclonal antibody conjugated to 1 11 1n, 90 Y, or 131 1 , etc.).
  • immunostimulants e.g., Bacillus Calmette-Guerin (BCG), levamisole, interleukin-2, alpha-interfer
  • the compounds described herein can be co-administered with conventional radiotherapeutic agents including, but not limited to, radionuclides such as 47 Sc, 64 C 67 C, 89 Sr, 86 Y, 87 Y, and 212 Bi, optionally conjugated to antibodies directed against tumor antigens.
  • conventional radiotherapeutic agents including, but not limited to, radionuclides such as 47 Sc, 64 C 67 C, 89 Sr, 86 Y, 87 Y, and 212 Bi, optionally conjugated to antibodies directed against tumor antigens.
  • anti-neoplastic agent for use in combination or co-administered with the presently invented ATF4 pathway inhibiting compounds are anti-PD-L1 agents.
  • Anti-PD-L1 antibodies and methods of making the same are known in the art.
  • Such antibodies to PD-L1 may be polyclonal or monoclonal, and/or recombinant, and/or humanized.
  • Exemplary PD-L1 antibodies are disclosed in:
  • antibodies are in development as immuno-modulatory agents for the treatment of cancer.
  • the antibody to PD-L1 is an antibody disclosed in US Patent No. 8,217,149.
  • the anti-PD-L1 antibody comprises the CDRs of an antibody disclosed in US Patent No. 8,217,149.
  • the antibody to PD-L1 is an antibody disclosed in US
  • the anti-PD-L1 antibody comprises the CDRs of an antibody disclosed in US Application No. 13/511 ,538.
  • the antibody to PD-L1 is an antibody disclosed in Application No. 13/478,511.
  • the anti-PD-L1 antibody comprises the CDRs of an antibody disclosed in US Application No. 13/478,511 .
  • the anti-PD-L1 antibody is BMS-936559 (MDX-1 105). In another embodiment, the anti-PD-L1 antibody is MPDL3280A (RG7446). In another embodiment, the anti-PD-L1 antibody is MEDI4736. In another embodiment, the anti-PD- L1 antibody is atezolizumab. In another embodiment, the anti-PD-L1 antibody is avelumab. In another embodiment, the anti-PD-L1 antibody is durvalumab.
  • PD-1 antagonist means any chemical compound or biological molecule that blocks binding of PD-L1 expressed on a cancer cell to PD-1 expressed on an immune cell (T cell, B cell or NKT cell) and preferably also blocks binding of PD-L2 expressed on a cancer cell to the immune-cell expressed PD-1.
  • PD-1 and its ligands include: PDCD1 , PD1 , CD279 and SLEB2 for PD-1 ; PDCD1 L1 , PDL1 , B7H1 , B7-4, CD274 and B7-H for PD-L1 ; and PDCD1 L2, PDL2, B7- DC, Btdc and CD273 for PD-L2.
  • the PD-1 antagonist blocks binding of human PD-L1 to human PD-1 , and preferably blocks binding of both human PD-L1 and PD-L2 to human PD-1.
  • Human PD-1 amino acid sequences can be found in NCBI Locus No.: NP_005009.
  • Human PD-L1 and PD-L2 amino acid sequences can be found in NCBI Locus No.: NP_054862 and NP_079515,
  • PD-1 antagonists useful in the any of the aspects of the present invention include a monoclonal antibody (mAb), or antigen binding fragment thereof, which specifically binds to PD-1 or PD-L1 , and preferably specifically binds to human PD-1 or human PD-L1 .
  • the mAb may be a human antibody, a humanized antibody or a chimeric antibody, and may include a human constant region.
  • the human constant region is selected from the group consisting of lgG1 , lgG2, lgG3 and lgG4 constant regions, and in preferred embodiments, the human constant region is an lgG1 or lgG4 constant region.
  • the antigen binding fragment is selected from the group consisting of Fab, Fab'-SH, F(ab')2, scFv and Fv fragments.
  • Specific anti-human PD-1 mAbs useful as the PD-1 antagonist in any of the aspects and embodiments of the present invention include: MK-3475, a humanized lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No. 2, pages 161 -162 (2013) and which comprises the heavy and light chain amino acid sequences shown in Figure 6; nivolumab, a human lgG4 mAb with the structure described in WHO Drug Information, Vol. 27, No.
  • immunoadhesion molecules that specifically bind to PD-1 are described in WO2010/027827 and
  • fusion proteins useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include AMP-224 (also known as B7-DCIg), which is a PD-L2-FC fusion protein and binds to human PD- 1 .
  • mAbs that bind to human PD-L1 are described in WO2013/019906, W02010/077634 A1 and US8383796.
  • Specific anti-human PD-L1 mAbs useful as the PD-1 antagonist in the treatment method, medicaments and uses of the present invention include MPDL3280A, BMS-936559, MEDI4736, MSB0010718C.
  • KEYTRUDA ® /pembrolizumab is an anti-PD-1 antibody marketed for the treatment of lung cancer by Merck.
  • the amino acid sequence of pembrolizumab and methods of using are disclosed in US Patent No. 8,168,757.
  • Opdivo ® /nivolumab is a fully human monoclonal antibody marketed by Bristol Myers Squibb directed against the negative immunoregulatory human cell surface receptor PD-1 (programmed death-1 or programmed cell death-1 /PCD-1) with immunopotentiation activity.
  • Nivolumab binds to and blocks the activation of PD-1 , an Ig superfamily transmembrane protein, by its ligands PD-L1 and PD-L2, resulting in the activation of T-cells and cell-mediated immune responses against tumor cells or pathogens.
  • Activated PD-1 negatively regulates T-cell activation and effector function through the suppression of P13k/Akt pathway activation.
  • Other names for nivolumab include: BMS-936558, MDX-1 106, and ONO-4538. The amino acid sequence for nivolumab and methods of using and making are disclosed in US Patent No. US
  • Additional examples of a further active ingredient or ingredients (anti-neoplastic agent) for use in combination or co-administered with the presently invented ATF4 pathway inhibiting compounds are immuno-modulators.
  • immuno-modulators refer to any substance including monoclonal antibodies that affects the immune system.
  • the ICOS binding proteins of the present invention can be considered immune-modulators.
  • Immuno-modulators can be used as anti-neoplastic agents for the treatment of cancer.
  • immune-modulators include, but are not limited to, anti-CTLA-4 antibodies such as ipilimumab (YERVOY ® ) and anti-PD-1 antibodies (Opdivo ® /nivolumab and Keytruda ® /pembrolizumab).
  • Other immuno-modulators include, but are not limited to, OX-40 antibodies, PD-L1 antibodies, LAG3 antibodies, TIM-3 antibodies, 41 BB antibodies and GITR antibodies.
  • Yervoy ® (ipilimumab) is a fully human CTLA-4 antibody marketed by Bristol Myers Squibb.
  • the protein structure of ipilimumab and methods are using are described in US Patent Nos. 6,984,720 and 7,605,238.
  • the compounds of the invention are combined with an inhibitor of the activity of the protein kinase R (PKR)-like ER kinase, PERK.
  • PSR protein kinase R
  • the compounds of the invention are combined with an inhibitor of the activity of the elF2a kinases protein kinase R, (PKR), Heme-regulated elF2a kinase (HRI), or general control non-derepressible 2 (GCN2).
  • PPKR protein kinase R
  • HRI Heme-regulated elF2a kinase
  • GCN2 general control non-derepressible 2
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful in the treatment of neurodegenerative diseases/injury.
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful in the treatment of diabetes.
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful in the treatment of cardiovascular disease.
  • the compounds of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful in the treatment of ocular diseases.
  • the compounds described herein can be used in combination with one another, with other active agents known to be useful in treating cancer (e.g. pancreatic cancer, breast cancer, multiple myeloma, or cancers of secretory cells), neurodegenerative diseases, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and/or intellectual disability syndromes (e.g. associated with impaired function of elF2 or components in a signal transduction pathway including elF2), or with adjunctive agents that may not be effective alone, but may contribute to the efficacy of the active agent.
  • the compounds set forth herein are provided as pharmaceutical compositions comprising the compound and a pharmaceutically acceptable excipient.
  • the compound, or a pharmaceutically acceptable salt thereof is co- adminstered with a second agent (e.g. therapeutic agent).
  • a second agent e.g. therapeutic agent
  • the compound, or a pharmaceutically acceptable salt thereof is co-adminstered with a second agent (e.g. therapeutic agent), which is administered in a therapeutically effective amount.
  • the second agent is an agent for treating cancer (e.g. pancreatic cancer, breast cancer, multiple myeloma, or cancers of secretory cells), neurodegenerative diseases, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and/or intellectual disability syndromes (e.g.
  • the second agent is an anti-cancer agent.
  • the second agent is a chemotherapeutic.
  • the second agent is an agent for improving memory.
  • the second agent is an agent for treating a neurodegenerative disease.
  • the second agent is an agent for treating vanishing white matter disease.
  • the second agent is an agent for treating childhood ataxia with CNS hypo-myelination.
  • the second agent is an agent for treating an intellectual disability syndrome.
  • the second agent is an agent for treating pancreatic cancer.
  • the second agent is an agent for treating breast cancer. In embodiments, the second agent is an agent for treating multiple myeloma. In embodiments, the second agent is an agent for treating myeloma. In embodiments, the second agent is an agent for treating a cancer of a secretory cell. In embodiments, the second agent is an agent for reducing elF2a phosphorylation. In embodiments, the second agent is an agent for inhibiting a pathway activated by elF2a phosphorylation. In embodiments, the second agent is an agent for inhibiting the integrated stress response. In embodiments, the second agent is an anti-inflammatory agent.
  • elF2alpha refers to the protein "Eukaryotic translation initiation factor 2A".
  • elF2alpha refers to the human protein. Included in the term “elF2alpha” or “elF2a” are the wildtype and mutant forms of the protein.
  • elF2alpha refers to the protein associated with Entrez Gene 83939, OMIM 609234, UniProt Q9BY44, and/or RefSeq (protein) NP 114414.
  • the present invention relates to a method for treating an integrated stress response associated disease in a patient in need of such treatment, the method including administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to the patient.
  • the integrated stress response-associated disease is cancer.
  • the integrated stress response-associated disease is a neurodegenerative disease.
  • the integrated stress response-associated disease is vanishing white matter disease.
  • the integrated stress response-associated disease is childhood ataxia with CNS hypomyelination.
  • the integrated stress response-associated disease is an intellectual disability syndrome.
  • the present invention relates to a method for treating a disease associated with phosphorylation of elF2a in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to the patient.
  • the disease associated with phosphorylation of elF2 a is cancer.
  • the disease associated with phosphorylation of elF2 a is a neurodegenerative disease.
  • the disease associated with phosphorylation of elF2 a is vanishing white matter disease.
  • the disease associated with phosphorylation of elF2 a is childhood ataxia with CNS hypomyelination.
  • the disease associated with phosphorylation of elF2 a is an intellectual disability syndrome.
  • the present invention relates to a method for treating a disease selected from the group consisting of cancer, a neurodegenerative disease, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and an intellectual disability syndrome.
  • the present invention relates to a method for treating an inflammatory disease in a patient in need of such treatment, which comprises administering a therapeutically effective amount of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, to the patient.
  • the inflammatory disease is associated with neurological inflammation.
  • the inflammatory disease is postoperative cognitive dysfunction.
  • the inflammatory disease is traumatic brain injury or chronic traumatic encephalopathy (CTE).
  • the disease is selected from the group consisting of cancer, a neurodegenerative disease, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and an intellectual disability syndrome.
  • the disease is cancer.
  • the disease is a neurodegenerative disease.
  • the disease is vanishing white matter disease.
  • the disease is childhood ataxia with CNS hypomyelination.
  • the method is an intellectual disability syndrome.
  • the disease is associated with phosphorylation of elF2a. In embodiments of the method of treating a disease, the disease is associated with an elF2a signaling pathway. In embodiments of the method of treating a disease, the disease is a cancer of a secretory cell type. In embodiments of the method of treating a disease, the disease is pancreatic cancer. In embodiments of the method of treating a disease, the disease is breast cancer. In embodiments of the method oftreating a disease, the disease is multiple myeloma. In embodiments of the method of treating a disease, the disease is lymphoma. In embodiments of the method of treating a disease, the disease is leukemia. In embodiments of the method of treating a disease, the disease is a hematopoietic cell cancer.
  • the disease is Alzheimer's disease. In embodiments of the method of treating a disease, the disease is Amyotrophic lateral sclerosis. In embodiments of the method of treating a disease, the disease is C re utzfeldt- Jakob disease. In embodiments of the method of treating a disease, the disease is frontotemporal dementia. In embodiments of the method of treating a disease, the disease is Gerstmann-Straussler-Scheinker syndrome. In embodiments of the method of treating a disease, the disease is Huntington's disease. In embodiments of the method of treating a disease, the disease is HIV-associated dementia. In embodiments of the method of treating a disease, the disease is kuru.
  • the disease is Lewy body dementia. In embodiments of the method of treating a disease, the disease is Multiple sclerosis. In embodiments of the method of treating a disease, the disease is Parkinson's disease. In embodiments of the method of treating a disease, the disease is a Prion disease.
  • the disease is an inflammatory disease.
  • the inflammatory disease is postoperative cognitive dysfunction.
  • the inflammatory disease is traumatic brain injury.
  • the inflammatory disease is arthritis.
  • the inflammatory disease is rheumatoid arthritis.
  • the inflammatory disease is psoriatic arthritis.
  • the inflammatory disease is juvenile idiopathic arthritis.
  • the inflammatory disease is multiple sclerosis.
  • the inflammatory disease is systemic lupus erythematosus (SLE).
  • the inflammatory disease is myasthenia gravis.
  • the inflammatory disease is juvenile onset diabetes.
  • the inflammatory disease is diabetes mellitus type 1 .
  • the inflammatory disease is Guillain-Barre syndrome.
  • the inflammatory disease is Hashimoto's encephalitis.
  • the inflammatory disease is Hashimoto's thyroiditis.
  • the inflammatory disease is ankylosing spondylitis.
  • the inflammatory disease is psoriasis.
  • the inflammatory disease is Sjogren's syndrome.
  • the inflammatory disease is vasculitis.
  • the inflammatory disease is glomerulonephritis.
  • the inflammatory disease is auto-immune thyroiditis.
  • the inflammatory disease is Behcet's disease.
  • the inflammatory disease is Crohn's disease. In embodiments, the inflammatory disease is ulcerative colitis. In embodiments, the inflammatory disease is bullous pemphigoid. In embodiments, the inflammatory disease is sarcoidosis. In embodiments, the inflammatory disease is ichthyosis. In embodiments, the inflammatory disease is Graves ophthalmopathy. In embodiments, the inflammatory disease is inflammatory bowel disease. In embodiments, the inflammatory disease is Addison's disease. In embodiments, the inflammatory disease is Vitiligo. In embodiments, the inflammatory disease is asthma. In embodiments, the inflammatory disease is allergic asthma. In embodiments, the inflammatory disease is acne vulgaris.
  • the inflammatory disease is celiac disease. In embodiments, the inflammatory disease is chronic prostatitis. In embodiments, the inflammatory disease is inflammatory bowel disease. In embodiments, the inflammatory disease is pelvic inflammatory disease. In embodiments, the inflammatory disease is reperfusion injury. In embodiments, the inflammatory disease is sarcoidosis. In embodiments, the inflammatory disease is transplant rejection. In embodiments, the inflammatory disease is interstitial cystitis. In embodiments, the inflammatory disease is atherosclerosis. In embodiments, the inflammatory disease is atopic dermatitis.
  • the method of treatment is a method of prevention.
  • a method of treating postsurgical cognitive dysfunction may include preventing postsurgical cognitive dysfunction or a symptom of postsurgical cognitive dysfunction or reducing the severity of a symptom of postsurgical cognitive dysfunction by administering a compound described herein prior to surgery.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease selected from the group consisting of cancer, a neurodegenerative disease, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and an intellectual disability syndrome.
  • a disease selected from the group consisting of cancer, a neurodegenerative disease, vanishing white matter disease, childhood ataxia with CNS hypomyelination, and an intellectual disability syndrome.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of an integrated stress response associated disease.
  • this invention provides a compound of Formula (I), or a pharmaceutically acceptable salt thereof, for use in the treatment of a disease associated with phosphorylation of elF2a.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease selected from the group consisting of cancer, a
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment an integrated stress response associated disease.
  • this invention provides for the use of a compound of Formula (I), or a pharmaceutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a disease associated with phosphorylation of elF2a.
  • the present invention therefore provides a method of treating cancer, neurodegeneration and other conditions requiring ATF4 pathway inhibition, which comprises administering an effective amount of a compound of Formula (I) or a pharmaceutically acceptable salt thereof.
  • the compounds of Formula (I) also provide for a method of treating the above indicated disease states because of their demonstrated ability to act as ATF4 pathway inhibitors.
  • the drug may be administered to a patient in need thereof by any conventional route of administration, including, but not limited to, intravenous, intramuscular, oral, topical, subcutaneous, intradermal, intraocular and parenteral.
  • a ATF4 pathway inhibitor may be delivered directly to the brain by intrathecal or intraventricular route, or implanted at an appropriate anatomical location within a device or pump that continuously releases the ATF4 pathway inhibiting drug.
  • Solid or liquid pharmaceutical carriers are employed.
  • Solid carriers include, starch, lactose, calcium sulfate dihydrate, terra alba, sucrose, talc, gelatin, agar, pectin, acacia, magnesium stearate, and stearic acid.
  • Liquid carriers include syrup, peanut oil, olive oil, saline, and water.
  • the carrier or diluent may include any prolonged release material, such as glyceryl monostearate or glyceryl distearate, alone or with a wax.
  • the amount of solid carrier varies widely but, preferably, will be from about 25 mg to about 1 g per dosage unit.
  • the preparation will be in the form of a syrup, elixir, emulsion, soft gelatin capsule, sterile injectable liquid such as an ampoule, or an aqueous or nonaqueous liquid suspension.
  • the pharmaceutical compositions are made following conventional techniques of a pharmaceutical chemist involving mixing, granulating, and compressing, when necessary, for tablet forms, or mixing, filling and dissolving the ingredients, as appropriate, to give the desired oral or parenteral products.
  • Doses of the presently invented pharmaceutically active compounds in a pharmaceutical dosage unit as described above will be an efficacious, nontoxic quantity preferably selected from the range of 0.001 - 100 mg/kg of active compound, preferably 0.001 - 50 mg/kg.
  • the selected dose is administered preferably from 1 -6 times daily, orally or parenterally.
  • Preferred forms of parenteral administration include topically, rectally, transdermally, by injection and continuously by infusion.
  • Oral dosage units for human administration preferably contain from 0.05 to 3500 mg of active compound.
  • Oral administration, which uses lower dosages, is preferred. Parenteral administration, at high dosages, however, also can be used when safe and convenient for the patient.
  • Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular ATF4 pathway inhibitor in use, the strength of the preparation, the mode of administration, and the advancement of the disease condition. Additional factors depending on the particular patient being treated will result in a need to adjust dosages, including patient age, weight, diet, and time of administration.
  • the method of this invention of inducing ATF4 pathway inhibitory activity in mammals, including humans, comprises administering to a subject in need of such activity an effective ATF4 pathway inhibiting amount of a pharmaceutically active compound of the present invention.
  • the invention also provides for the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for inhibiting the ATF4 pathway.
  • the invention also provides for the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for treating cancer, pre-cancerous syndromes, Alzheimer’s disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, Huntington's disease,
  • C re utzfeldt- Jakob Disease, and related prion diseases progressive supranuclear palsy, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, chronic and acute diseases of the lung, chronic and acute diseases of the kidney, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, cognitive impairment, atherosclerosis, ocular diseases, arrhythmias, in organ transplantation and in the transportation of organs for
  • the invention also provides for the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the manufacture of a medicament for preventing organ damage during the transportation of organs for transplantation.
  • the invention also provides for a pharmaceutical composition for use as a ATF4 pathway inhibitor which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the invention also provides for a pharmaceutical composition for use in the treatment of cancer which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutically active compounds of the present invention can be co-administered with further active ingredients, such as other compounds known to treat cancer, or compounds known to have utility when used in combination with a ATF4 pathway inhibitor.
  • further active ingredients such as other compounds known to treat cancer, or compounds known to have utility when used in combination with a ATF4 pathway inhibitor.
  • the invention also provides novel processes and novel intermedites useful in preparing the presently invented compounds.
  • the invention also provides a pharmaceutical composition comprising from 0.5 to 1 ,000 mg of a compound of Formula (I) or pharmaceutically acceptable salt thereof and from 0.5 to 1 ,000 mg of a pharmaceutically acceptable excipient.
  • Examples 2-5 of Table 1 were prepared from intermediate i or intermediate ii generally according to the above schemes and the procedure described for Example 1 :
  • ATF4 Cell Based Assay measures the effect of thapsigargin-induced cellular stress on ATF4 expression.
  • a stable cell line was created by transfecting SH-SY5Y cells with a plasmid containing the NanoLuc® luciferase gene fused to the 5’-UTR of ATF4, under the control of the CMV promoter.
  • the ATF4 5’-UTR contains two open reading frames which mediate the cellular stress-dependent translation of the reporter gene. Clones stably expressing the reporter construct were isolated and selected based on the luminescence response to thapsigargin and inhibition of this signal by test compounds.
  • SH-SY5Y-ATF4-NanoLuc cells were challenged with thapsigargin to determine the stress effect with or without test compounds.
  • Cells were propagated in DMEM/F12 growth media containing 10% FBS (Invitrogen 10999-141) and 0.5 mg/ml_ geneticin (Corning 30-234-CR). Aliquots of cells were cryopreserved in dialyzed FBS containing 10% DMSO.
  • Test compounds were prepared in neat DMSO at a concentration of 10 mM. Assay plates were prepared by adding 250nl_ of compound stock solution to test wells in a 384- well white tissue culture-treated plate (Greiner 781073). For inhibition curves, compounds were diluted using a three-fold serial dilution and tested at 11 concentrations (10 pM - 0.17 nM). Aliquots of frozen cells were thawed with a 37C water bath. The cells were washed using DMEM/F12 (1 :1) (1X) (Gibco 1 1039-021). The cells were re-suspended in the DMEM/F12 and the suspension was counted. A final suspension of 7.5e5 cells/ml was prepared.
  • a volume of 20uL of cell suspension was added to compound plates (15K cells/well). Cells were incubated for 1 hour at 37°C. A volume of 5pL of 1 uM
  • the measurement of luciferase produced by the ATF4 constructs was measured using Nano-Glo Luciferase Assay reagent, Promega N1150.
  • the components of the Promega kit are: Nano-Glo® Luciferase Assay Substrate, N113C, Nano-Glo® Luciferase Assay Buffer, N1128.
  • the buffer is brought to room temperature, and a solution of 50:1 buffensubstrate were prepared.
  • the cell plates were equilibrated to room temperature.
  • a volume of 20 microliters/well of the mixed Nano-Glo reagent were dispensed into assay and control wells. The plates were read on a Viewlux plate reader.
  • Examples 1 to 5 were tested generally according to the above ATF4 cell based assay and in a set of two or more experimental runs exhibited the following average ATF4 pathway inhibitory activities (plC50).
  • Example 1 The compound of Example 1 was tested generally according to the above ATF4 cell based assay and in at least one set of two or more experimental runs exhibited an average ATF4 pathway inhibitory activity IC50 of 7.9 nM.
  • Example 2 The compound of Example 2 was tested generally according to the above ATF4 cell based assay and in at least one set of two or more experimental runs exhibited an average ATF4 pathway inhibitory activity IC50 of 12.6 nM.
  • the compound of Example 3 was tested generally according to the above ATF4 cell based assay and in at least one set of two or more experimental runs exhibited an average ATF4 pathway inhibitory activity IC50 of 20 nM.
  • the compound of Example 4 was tested generally according to the above ATF4 cell based assay and in at least one set of two or more experimental runs exhibited an average ATF4 pathway inhibitory activity IC50 of 1995 nM.
  • Example 5 The compound of Example 5 was tested generally according to the above ATF4 cell based assay and in at least one set of two or more experimental runs exhibited an average ATF4 pathway inhibitory activity IC50 of 631 nM.
  • An oral dosage form for administering the present invention is produced by filing a standard two-piece hard gelatin capsule with the ingredients shown in Formulation Table 3, below.
  • sucrose, calcium sulfate dihydrate and an ATF4 pathway inhibitor as shown in FormulationTable 4 below are mixed and granulated with a 10% gelatin solution.
  • the wet granules are screened, dried, mixed with the starch, talc and stearic acid, screened and compressed into a tablet.
  • Ron D Walter P. Signal integration in the endoplasmic reticulum unfolded protein response. Nat Rev Mol Cell Biol. 2007 Jul;8(7):519-29.
  • PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des dérivés de tétrahydrofurane substitués. En particulier, l'invention concerne des composés selon la formule I : (I) dans laquelle C, D, L1, L2, R1, R2, R3, R4, R5, R6, Y1, Y2, z1, z2, z3, z4, X1, X2, X3, X4, X5, et X6 sont tels que définis dans la description ; ou des sels de ceux-ci.
PCT/IB2019/056721 2018-08-08 2019-08-07 Composés chimiques WO2020031107A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201862715886P 2018-08-08 2018-08-08
US62/715,886 2018-08-08

Publications (1)

Publication Number Publication Date
WO2020031107A1 true WO2020031107A1 (fr) 2020-02-13

Family

ID=68072871

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2019/056721 WO2020031107A1 (fr) 2018-08-08 2019-08-07 Composés chimiques

Country Status (1)

Country Link
WO (1) WO2020031107A1 (fr)

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021180774A1 (fr) 2020-03-11 2021-09-16 Evotec International Gmbh Modulateurs de la voie de réponse au stress intégrée
US11166942B2 (en) 2018-06-05 2021-11-09 Praxis Biotech LLC Inhibitors of integrated stress response pathway
US11230542B2 (en) 2017-12-13 2022-01-25 Praxis Biotech LLC Inhibitors of integrated stress response pathway
US11236100B2 (en) 2017-08-09 2022-02-01 Denali Therapeutics Inc. Modulators of eukaryotic initiation factor 2B, compositions and methods
US11306077B2 (en) 2019-02-13 2022-04-19 Denali Therapeutics Inc. Compounds, compositions and methods
WO2022084448A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse intégrée au stress
WO2022084446A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse intégrée au stress
WO2022084447A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse de stress intégrée
US11318133B2 (en) 2019-06-12 2022-05-03 Praxis Biotech LLC Modulators of integrated stress response pathway
US11993613B2 (en) 2022-03-31 2024-05-28 Abbvie Inc. Thiazolo[5,4-b]pyridine MALT-1 inhibitors

Citations (22)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
WO2004072286A1 (fr) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specifique a pd-1 humain
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US20110280877A1 (en) 2010-05-11 2011-11-17 Koji Tamada Inhibition of B7-H1/CD80 interaction and uses thereof
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US20130309250A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
WO2014144952A2 (fr) 2013-03-15 2014-09-18 Peter Walter Modulateurs de la voie eif2 alpha
WO2017212423A1 (fr) * 2016-06-08 2017-12-14 Glaxosmithkline Intellectual Property Development Limited Composés chimiques

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
US7605238B2 (en) 1999-08-24 2009-10-20 Medarex, Inc. Human CTLA-4 antibodies and their uses
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
WO2004004771A1 (fr) 2002-07-03 2004-01-15 Ono Pharmaceutical Co., Ltd. Compositions immunostimulantes
US8168179B2 (en) 2002-07-03 2012-05-01 Ono Pharmaceutical Co., Ltd. Treatment method using anti-PD-L1 antibody
US7595048B2 (en) 2002-07-03 2009-09-29 Ono Pharmaceutical Co., Ltd. Method for treatment of cancer by inhibiting the immunosuppressive signal induced by PD-1
WO2004056875A1 (fr) 2002-12-23 2004-07-08 Wyeth Anticorps anti pd-1 et utilisations
US7488802B2 (en) 2002-12-23 2009-02-10 Wyeth Antibodies against PD-1
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
WO2004072286A1 (fr) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specifique a pd-1 humain
US8008449B2 (en) 2005-05-09 2011-08-30 Medarex, Inc. Human monoclonal antibodies to programmed death 1 (PD-1) and methods for treating cancer using anti-PD-1 antibodies alone or in combination with other immunotherapeutics
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2010027827A2 (fr) 2008-08-25 2010-03-11 Amplimmune, Inc. Polypeptides co-stimulateurs ciblés et leurs procédés d'utilisation dans le traitement du cancer
US20110271358A1 (en) 2008-09-26 2011-11-03 Dana-Farber Cancer Institute, Inc. Human anti-pd-1, pd-l1, and pd-l2 antibodies and uses therefor
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US20110280877A1 (en) 2010-05-11 2011-11-17 Koji Tamada Inhibition of B7-H1/CD80 interaction and uses thereof
WO2013019906A1 (fr) 2011-08-01 2013-02-07 Genentech, Inc. Procédés de traitement du cancer à l'aide d'antagonistes se liant à l'axe pd-1 et inhibiteurs de mek
WO2013079174A1 (fr) 2011-11-28 2013-06-06 Merck Patent Gmbh Anticorps anti-pd-l1 et utilisations associées
US20130309250A1 (en) 2012-05-15 2013-11-21 Bristol-Myers Squibb Company Cancer immunotherapy by disrupting pd-1/pd-l1 signaling
WO2014055897A2 (fr) 2012-10-04 2014-04-10 Dana-Farber Cancer Institute, Inc. Anticorps monoclonaux humains anti pd-l1 et procédés d'utilisation
WO2014144952A2 (fr) 2013-03-15 2014-09-18 Peter Walter Modulateurs de la voie eif2 alpha
WO2017212423A1 (fr) * 2016-06-08 2017-12-14 Glaxosmithkline Intellectual Property Development Limited Composés chimiques

Non-Patent Citations (35)

* Cited by examiner, † Cited by third party
Title
"An efficient mammalian cell-free translation system supplemented with translation factors", PROTEIN EXPR. PURIF., vol. 46, pages 348 - 357
"Cancer Principles and Practice of Oncology", 15 February 2001
"NCBI", Database accession no. NP_079515
AVIVAR-VALDERAS ABOBROVNIKOVA-MARJON EDIEHL ANAGI CDEBNATH JAGUIRRE-GUISO JA: "PERK integrates autophagy and oxidative stress responses to promote survival during extracellular matrix detachment", MOL CEL BIOL, vol. 31, 2011, pages 3616 - 3629
AXTEN JM.MEDINA J. R.FENG Y.SHU A.ROMERIL S. P. ET AL.: "Discovery of 7-methy-5(1-([3-10 (trifluoromethyl)phenyl]acetyl)-2, 3-dihydro-1 H-indo 1-5yl)-7H-pyrrolo [2,3-d]pyrimidin-4 amine (GSK2606414), a potent and selective first-in class inhibitor of protein kinase R (PKR)-like endplasmic reticulum kinase (PERK)", J. MED. CHEM., vol. 55, no. 16, 2012, pages 7193 - 7207
BLAIS JHU NHARKING HNOVOA IVARIA MRALEIGH JSCHEUNER DKAUFMAN RJBELL JRON D: "ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth", EMBO J., vol. 24, 2005, pages 3470 - 3481
BOBROVNIKOVA-MARJON EPYTEL DVAITES LPSINGH NKORETZKY GADIEHL JA: "PERK promotes cancer cell proliferation and tumor growth by limiting oxidative DNA damage", ONCOGENE, vol. 29, 2010, pages 3881 - 3895
BORCK G.SHIN B.S.STILLER B. ET AL.: "elF2y mutation that disrupts elF2 complex integrity links intellectual disability to impaired translation 30 initiation", MOL CELL, vol. 48, 2012, pages 1 - 6
COSTA-MATTIOLI M.GOBERT D.HARDING H.HERDY B.AZZI M.BRUNO M. ET AL.: "Translational control of hippocampal synaptic plasticity and memory by the elF2n kinase GCN2", NATURE, vol. 436, 2005, pages 1166 - 1173
COSTA-MATTIOLI M.GOBERT D.STERN E.GARNACHE K.COLINA RICUELLO C.SOSSIN W.KAUFMAN R.PELLETIER J., ROSENBLUM ET AL.: "elF2n phosphorylation bidirectionally regulates the switch from short to long term synaptic plasticity and memory", CELL, vol. 129, 2007, pages 195 - 206
GARDNER BMWALTER P: "Unfolded proteins are Irel-activating ligands that directly induce the unfolded protein response", SCIENCE, vol. 333, no. 6051, 30 September 2011 (2011-09-30), pages 1891 - 4
HARDING HPNOVOA IZHANG YZENG HWEK RSCHAPIRA M ET AL.: "Regulated translation initiation controls stress-induced gene expression in mammalian cells", MOL. CELL., vol. 6, no. 5, November 2000 (2000-11-01), pages 1099 - 108, XP002226689, DOI: 10.1016/S1097-2765(00)00108-8
HARDING HPZHANG YBERTOLOTTI AZENG HRON D: "Perk is essential for translational regulation and cell survival during the unfolded protein response", MOL CELL, vol. 5, no. 5, May 2000 (2000-05-01), pages 897 - 904, XP002226688, DOI: 10.1016/S1097-2765(00)80330-5
HINNEBUSCH AG: "Translational regulation of GCN4 and the general amino acid control of yeast", ANNU. REV. MICROBIOL., vol. 59, 2005, pages 407 - 50, XP055132693, DOI: 10.1146/annurev.micro.59.031805.133833
HINNEBUSCH AGLORSCH JR: "The mechanism of eukaryotic translation initiation: new insights and challenges", COLD SPRING HARB PERSPECT BIOL., vol. 4, no. 10, 2012, pages a011544
JACKSON RJHELLEN CUTPESTOVA TV: "he mechanism of eukaryotic translation initiation and principles of its regulation", NAT REV MOL CELL BIOL., vol. 11, no. 2, 1 February 2010 (2010-02-01), pages 113 - 27, XP009135819, DOI: 10.1038/nrm2838
JOURNAL OF THE AMERICAN CHEMICAL SOCIETY
KRISHNAMOORTHY TPAVITT GDZHANG FDEVER TEHINNEBUSCH AG: "Tight binding of the phosphorylated alpha subunit of initiation factor 2 (eIF2alpha) to the regulatory subunits of guanine nucleotide exchange factor elF2B is required for inhibition of translation initiation", MOL CELL BIOL., vol. 21, no. 15, August 2001 (2001-08-01), pages 5018 - 30
MA YBREWER JWDIEHL JAHENDERSHOT LM: "Two distinct stress signaling pathways converge upon the CHOP promoter during the mammalian unfolded protein response", J. MOL. BIOL., vol. 318, no. 5, 17 May 2002 (2002-05-17), pages 1351 - 65
MORENO JARADFORD HPERETTI DSTEINERT JRVERITY NMARTIN MGHALLIDAY MMORGAN JDINSDALE DORTORI CA: "Sustained translational repression by elF2n-P mediates prion neurodegeneration", NATURE, vol. 485, 2012, pages 507 - 511
PALAM LRBAIRD TDWEK RC: "Phosphorylation ofelF2 facilitates ribosomal bypass of an inhibitory upstream ORF to enhance CHOP translation", JOURNAL OF BIOLOGICAL CHEMISTRY, vol. 286, no. 13, 1 April 2011 (2011-04-01), pages 10939 - 49
PAVITT GDPROUD CG: "Protein synthesis and its control in neuronal cells with a focus on vanishing white matter disease", BIOCHEM SOC TRANS, vol. 37, 2009
PAVITT GDRON D: "New insights into translational regulation in the endoplasmic reticulum unfolded protein response", COLD SPRING HARB PERSPECT BIOL., vol. 4, no. 6, June 2012 (2012-06-01), pages a012278
RON DWALTER P: "Signal integration in the endoplasmic reticulum unfolded protein response", NAT REV MOL CELL BIOL., vol. 8, no. 7, July 2007 (2007-07-01), pages 519 - 29, XP009151293
SHORE GCGPAPA FRFOAKES SAS: "Signaling cell death from the endoplasmic reticulum stress response", CURRENT OPINION IN CELL BIOLOGY, vol. 23, no. 2, 1 April 2011 (2011-04-01), pages 143 - 9, XP028193835, DOI: 10.1016/j.ceb.2010.11.003
T. GREENEP. WUTS: "Protecting Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
TABAS IRON D: "Integrating the mechanisms of apoptosis induced by endoplasmic reticulum stress", NAT CELL BIOL., vol. 13, no. 3, 1 March 2011 (2011-03-01), pages 184 - 90
VATTEM KMWEK RC: "Reinitiation involving upstream ORFs regulates ATF4 mRNA translation in mammalian cells", PROC NATL ACAD SCI USA, vol. 101, no. 31, 3 August 2004 (2004-08-03), pages 11269 - 74, XP055369009, DOI: 10.1073/pnas.0400541101
VINCENZO TUMIATTI ET AL: "Structure-Activity Relationships of Acetylcholinesterase Noncovalent Inhibitors Based on a Polyamine Backbone. 4. Further Investigation on the Inner Spacer", JOURNAL OF MEDICINAL CHEMISTRY, vol. 51, no. 22, 27 November 2008 (2008-11-27), US, pages 7308 - 7312, XP055304506, ISSN: 0022-2623, DOI: 10.1021/jm8009684 *
WALTER PRON D: "The unfolded protein response: from stress pathway to homeostatic regulation", SCIENCE, vol. 334, no. 6059, 25 November 2011 (2011-11-25), pages 1081 - 6
WEK RCJIANG H-YANTHONY TG: "Coping with stress: elF2 kinases and translational control", BIOCHEM. SOC. TRANS., vol. 34, February 2006 (2006-02-01), pages 7 - 11
WHO DRUG INFORMATION, vol. 27, no. 1, 2013, pages 161 - 162
YE J.KUMANOVA M.HART L. S.SLOANE K.ZHANG H. ET AL.: "The GCN2-ATF4 pathway is critical for tumour cell survival and proliferation in response to nutrient deprivation", EMBO J., vol. 29, 2010, pages 2082 - 2096, XP055429392, DOI: 10.1038/emboj.2010.81
ZEENKO V. V.WANG CMAJUMDER MKOMAR A. A.SNIDER M. D.MERRICK W. C.KAUFMAN R. J.HATZOGLOU M.: "An efficient in vitro translation system from mammalian cell lacking translational inhibition caused by elF2 phosphorylation", RNA, vol. 14, 2008, pages 593 - 602
ZHU P. JHUAN W.KALIKULOV D.YOO J. W.PLACZEK A. N.STOICA LZHOU H.BELL J. C.FRIELANDER M. J.KRNJEVIC K.: "Suppression of PKR promotes network excitability and enhanced cognition by interferon-7-mediated disinhibition", CE, vol. 147, 2011, pages 1384 - 1396, XP028392401, DOI: 10.1016/j.cell.2011.11.029

Cited By (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11236100B2 (en) 2017-08-09 2022-02-01 Denali Therapeutics Inc. Modulators of eukaryotic initiation factor 2B, compositions and methods
US11851440B2 (en) 2017-08-09 2023-12-26 Denali Therapeutics Inc. Modulators of eukaryotic initiation factor 2B, compositions and methods
US11230542B2 (en) 2017-12-13 2022-01-25 Praxis Biotech LLC Inhibitors of integrated stress response pathway
US11166942B2 (en) 2018-06-05 2021-11-09 Praxis Biotech LLC Inhibitors of integrated stress response pathway
US11306077B2 (en) 2019-02-13 2022-04-19 Denali Therapeutics Inc. Compounds, compositions and methods
US11958840B2 (en) 2019-02-13 2024-04-16 Denali Therapeutics Inc. Compounds, compositions and methods
US11318133B2 (en) 2019-06-12 2022-05-03 Praxis Biotech LLC Modulators of integrated stress response pathway
WO2021180774A1 (fr) 2020-03-11 2021-09-16 Evotec International Gmbh Modulateurs de la voie de réponse au stress intégrée
WO2022084448A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse intégrée au stress
WO2022084446A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse intégrée au stress
WO2022084447A1 (fr) 2020-10-22 2022-04-28 Evotec International Gmbh Modulateurs de la voie de réponse de stress intégrée
US11993613B2 (en) 2022-03-31 2024-05-28 Abbvie Inc. Thiazolo[5,4-b]pyridine MALT-1 inhibitors

Similar Documents

Publication Publication Date Title
US11547704B2 (en) Chemical compounds
US10851053B2 (en) Chemical compounds
WO2020031107A1 (fr) Composés chimiques
WO2018225093A1 (fr) Composés chimiques utilisés comme inhibiteurs de la voie atf4
WO2020012339A1 (fr) Composés chimiques
EP3649106A1 (fr) Dérivés de n-(3-(2-(4-chlorophénoxy)acétamido)bicyclo[1.1.1]pentan-1-yl)-2-cyclobutane-1-carboxamide et composés apparentés en tant qu'inhibiteurs atf4 pour le traitement du cancer et d'autres maladies
WO2019193541A1 (fr) Dérivés de cycle aromatiques bicycliques de formule (i) utilisés en tant qu'inhibiteurs d'atf4
WO2019193540A1 (fr) Dérivés hétéroaryles de formule (i) utilisés en tant qu'inhibiteurs d'atf4
CA3068395A1 (fr) Derives de 2-(4-chlorophenoxy)-n-((1-(2-(4-chlorophenoxy)ethynazetidine-3-yl)methyl)acetamide et composes apparentes servant d'inhibiteurs d'atf4 pour le traitement du cancer et d'autres maladies
EP3634952A1 (fr) Composés chimiques utilisés comme inhibiteurs de la voie atf4
US20210093619A1 (en) Chemical Compounds as ATF-4 Pathway Inhibitors

Legal Events

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

Ref document number: 19779120

Country of ref document: EP

Kind code of ref document: A1

NENP Non-entry into the national phase

Ref country code: DE

122 Ep: pct application non-entry in european phase

Ref document number: 19779120

Country of ref document: EP

Kind code of ref document: A1