WO2019021208A1 - Dérivés d'indazole utiles en tant qu'inhibiteurs de perk - Google Patents

Dérivés d'indazole utiles en tant qu'inhibiteurs de perk Download PDF

Info

Publication number
WO2019021208A1
WO2019021208A1 PCT/IB2018/055548 IB2018055548W WO2019021208A1 WO 2019021208 A1 WO2019021208 A1 WO 2019021208A1 IB 2018055548 W IB2018055548 W IB 2018055548W WO 2019021208 A1 WO2019021208 A1 WO 2019021208A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
disease
compound
fluoro
alkyl
Prior art date
Application number
PCT/IB2018/055548
Other languages
English (en)
Inventor
Jeffrey Michael Axten
Rajendra KRISTAM
Chandregowda VENKATESHAPPA
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 WO2019021208A1 publication Critical patent/WO2019021208A1/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D487/00Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00
    • C07D487/02Heterocyclic compounds containing nitrogen atoms as the only ring hetero atoms in the condensed system, not provided for by groups C07D451/00 - C07D477/00 in which the condensed system contains two hetero rings
    • C07D487/04Ortho-condensed systems
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/519Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim ortho- or peri-condensed with heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia

Definitions

  • the present invention relates to substituted indazole derivatives that are inhibitors of the activity of the protein kinase R (PKR)-like ER kinase, PERK.
  • PPKR protein kinase R
  • the present invention also relates to pharmaceutical compositions comprising such compounds and methods of using such compounds in the treatment of cancer, pre-cancerous syndromes and diseases/injuries associated with activated unfolded protein response pathways, such as Alzheimer's disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, Parkinson's disease, diabetes, metabolic syndrome, metabolic disorders, Huntington's disease, C re utzfeldt- Jakob Disease, and related prion diseases, fatal familial insomnia, Gerstmann-Straussler-Scheinker syndrome, and related prion diseases, amyotrophic lateral sclerosis, progressive supranuclear palsy, myocardial infarction, cardiovascular disease, inflammation, organ fibrosis, chronic and acute diseases of the liver, fatty liver disease
  • the unfolded protein response is a signal transduction pathway that allows cells to survive stress caused by the presence of misfolded or unfolded proteins or protein aggregates (Walter and Ron, 201 1), (Hetz, 2012).
  • UPR activating stress stimuli include hypoxia, disruption of protein glycosylation (glucose deprivation), depletion of luminal ER calcium, or changes in ER redox status, among others (Ma and Hendershot, 2004), (Feldman et al., 2005).
  • PPR protein kinase R
  • PERK protein kinase R
  • EIF2AK3 eukaryotic initiation factor 2A kinase 3
  • PKI pancreatic ER kinase
  • ATF6 activating transcription factor 6
  • PERK is a type I ER membrane protein containing a stress-sensing domain facing the ER lumen, a transmembrane segment, and a cytosolic kinase domain (Shi et al., 1998), (Harding et al., 1999), (Sood et al., 2000). Release of GRP78 from the stress-sensing domain of PERK results in oligomerization and autophosphorylation at multiple serine, threonine and tyrosine residues (Ma et al., 2001), (Su et al., 2008).
  • Phenotypes of PERK knockout mice include diabetes, due to loss of pancreatic islet cells, skeletal abnormalities, and growth retardation (Harding et al., 2001), (Zhang et al., 2006), (lida et al., 2007). These features are similar to those seen in patients with Wolcott-Rallison syndrome, who carry germline mutations in the PERK gene (Julier and Nicolino, 2010).
  • the major substrate for PERK is the eukaryotic initiation factor 2a (elF2a), which PERK phosphorylates at serine-51 (Marciniak et al., 2006) in response to ER stress or treatment with pharmacological inducers of ER stress such as thapsigargin and tunicamycin.
  • This site is also phosphorylated by other EIF2AK family members [(general control non- derepressed 2 (GCN2), PKR, and heme-regulated kinase (HRI)] in response to different stimuli.
  • GCN2 general control non- derepressed 2
  • PKR heme-regulated kinase
  • Phosphorylation of elF2a converts it to an inhibitor of the guanine nucleotide exchange factor (GEF) elF2B which is required for efficient turnover of GDP for GTP in the elF2 protein synthesis complex.
  • GEF guanine nucleotide exchange factor
  • the inhibition of elF2B by P-elF2a causes a decrease in translation initiation and global protein synthesis (Harding et al. 2002).
  • Paradoxically, translation of specific mRNAs is enhanced when the UPR is activated and elF2a is phosphorylated.
  • the transcription factor ATF4 has 5'-upstream open reading frames (uORFs) that normally represses ATF4 synthesis during normal global protein synthesis.
  • PERK is activated under stress and P-elF2a inhibits elF2B
  • low levels of ternary complex allows for selective enhanced translation of ATF4 (Jackson et al. 2010). Therefore, when ER stress ensues, PERK activation causes an increase in ATF4 translation, which transcriptionally upregulates downstream target genes such as CHOP (transcription factor C/EBP homologous protein). This transcriptional reprogramming modulates cell survival pathways and can lead to the induction of pro- apoptotic genes.
  • CHOP transcription factor C/EBP homologous protein
  • PERK and the UPR is associated with human neurodegenerative conditions such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), progressive supranuclear palsy (PSP), dementias, and prion diseases including C re utzfeldt- Jakob Disease (CJD), (Doyle et al. 201 1), (Paschen 2004), (Salminen et al. 2009), (Stutzbach et al. 2013).
  • CJD C re utzfeldt- Jakob Disease
  • malformed/misfolded or aggregated protein deposits e.g tau tangles, Lewy bodies, a-synuclein, ⁇ plaques, mutant prion proteins
  • malformed/misfolded or aggregated protein deposits e.g tau tangles, Lewy bodies, a-synuclein, ⁇ plaques, mutant prion proteins
  • the fate of a cell (e.g a neuron) enduring unfolded or malfolded protein stress is under control of PERK.
  • a cell enduring ER stress may restore proteostasis and return to normal, or if the stress is insurmountable, sustained PERK activation may lead to cell death through ATF4/CHOP signaling coupled with the inability to synthesize vital proteins because of the persistent translational repression.
  • Activated PERK and associated biological markers of PERK activation are detected in post-mortem brain tissue of Alzheimer's disease patients and in human prion disease (Ho et al. 2012), (Hoozemans et al, 2009) (Schberger et al. 2006).
  • P-elF2a the product of PERK activation correlates with levels of BACE1 in post-mortem brain tissue of Alzheimer's disease patients (O'Connor et al. 2008).
  • the small molecule PERK inhibitor GSK2606414 was shown to provide a neuroprotective effect and prevent clinical signs of disease in prion infected mice (Moreno et al. 2013), consistent with previous results derived from genetic manipulation of the UPR/PERK/elF2a pathway (Moreno et al. 2012). Involvement of the pathway in ALS (Kanekura et. al., 2009 and Nassif et. al. 2010), spinal cord injury (Ohri et al. 201 1) and traumatic brain injury (Tajiri et al. 2004) is also reported. Taken together these data suggest that the UPR and PERK represent a promising node of drug intervention as a means to halt or reverse the clinical progression and associated cognitive impairments of a wide range of neurodegenerative diseases.
  • Tumor cells experience episodes of hypoxia and nutrient deprivation during their growth due to inadequate blood supply and aberrant blood vessel function (Brown and Wilson, 2004), (Blais and Bell, 2006). Thus, they are likely to be dependent on active UPR signaling to facilitate their growth.
  • mouse fibroblasts derived from PERK-/-, XBP1 -/-, and ATF4-/- mice, and fibroblasts expressing mutant elF2a show reduced clonogenic growth and increased apoptosis under hypoxic conditions in vitro and grow at substantially reduced rates when implanted as tumors in nude mice (Koumenis et al., 2002), (Romero-Ramirez et al., 2004), (Bi et al., 2005).
  • Human tumor cell lines carrying a dominant negative PERK that lacks kinase activity also showed increased apoptosis in vitro under hypoxia and impaired tumor growth in vivo (Bi et al., 2005).
  • Human tumors including those derived from cervical carcinomas, glioblastomas (Bi et al., 2005), lung cancers (Jorgensen et al., 2008) and breast cancers (Ameri et al., 2004), (Davies et al., 2008) show elevated levels of proteins involved in UPR, compared to normal tissues. Therefore, inhibiting the unfolded protein response with compounds that block the activity of PERK and other components of the UPR is expected to have utility as anticancer agents. Recently, this hypothesis was supported by two small molecule inhibitors of PERK that were shown to inhibit the growth of human tumor xenografts in mice (Axten et al. 2012 and Atkins et al. 2013).
  • Inhibitors of PERK may be therapeutically useful for the treatment of a variety of human diseases such as Alzheimer's disease and frontotemporal dementias, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis (ALS), progressive supranuclear palsy (PSP), and other tauopathies such chronic traumatic encephalopathy (CTE) (Nijholt, D. A., et al. 2012), (Lucke-Wold, B. P., et al.
  • CTE chronic traumatic encephalopathy
  • Inhibitors of PERK may also be useful for effective treatment of cancers, particularly those derived from secretory cell types, such as pancreatic and neuroendocrine cancers, multiple myeloma, or for use in combination as a chemosensitizer to enhance tumor cell killing.
  • a PERK inhibitor may also be useful for myocardial infarction, cardiovascular disease, atherosclerosis (McAlpine et al., 2010, Civelek et al.
  • a PERK inhibitor may also be useful in stem cell or organ transplantation to prevent damage to the organ and in the transportation of organs for transplantation (Inagi et al., 2014), (Cunard, 2015), (Dickhout et al., 201 1), (van Galen, P., et al. (2014).
  • a PERK inhibitor is expected to have diverse utility in the treatment of numerous diseases in which the underlying pathology and symptoms are associated with dysregulaton of the unfolded protein response.
  • compositions that comprise a pharmaceutical carrier and compounds that are inhibitors of PERK.
  • the invention is directed to substituted indazole derivatives and uses thereof. Specifically, the invention is directed to compounds according to Formula (I) and the use of compounds of Formula (I) in treating diseases and conditions:
  • the present invention also relates to the discovery that the compounds of Formula (I) are active as inhibitors of PERK.
  • This invention also relates to a method of treating cancer, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating Alzheimer's disease, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating Parkinson's disease, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating amyotrophic lateral sclerosis, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating Huntington's disease, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating C re utzfeldt- Jakob Disease, and related prion diseases, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating progressive supranuclear palsy
  • PSP which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating dementia, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating spinal cord injury, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating traumatic brain injury, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating ischemic stroke, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating diabetes, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • This invention also relates to a method of treating a disease or condition selected from:, myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, neurological disorders, pain, and arrhythmias, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • a disease or condition selected from:, myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, neurological disorders, pain, and arrhythmias
  • 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.
  • This invention also relates to a method of transporting organs for transplantation with comprises adding a compound of Formula (I) to the solution containg the organ for transplantation.
  • the invention also relates to novel processes and novel intermediates useful in preparing the PERK inhibiting compounds of Formula (I)
  • the invention also relates to methods of co-administering the invented PERK inhibiting compounds of Formula (I) with further active ingredients.
  • 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.
  • the invention also relates to a compound of Formula (I) 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 the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of traumatic brain injury.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of diabetes.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of a disease state selected from:, myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, neurological disorders, pain, and arrhythmias.
  • a disease state selected from:, myocardial infarction, cardiovascular disease, atherosclerosis, ocular diseases, neurological disorders, pain, and arrhythmias.
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of chronic traumatic encephalopathy (CTE).
  • CTE chronic traumatic encephalopathy
  • the invention also relates to the use of a compound of Formula (I) or a pharmaceutically acceptable salt thereof in the preparation of a medicament for use in organ transplantation and in the transportation of organs for transplantation.
  • a pharmaceutical carrier included in the present invention are pharmaceutical compositions that comprise a pharmaceutical carrier 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.
  • This invention relates to compounds of Formula (I) and to the use of compounds of Formula (I) in the methods of the invention:
  • Ci_ 4 alkyl independently selected from: Ci_ 4 alkyl, Ci- 4 alkoxy, -OH, -COOH, -CF 3 , C1-4alkoxyC1-4alkyl, oxo, -N0 2 , -NH 2 and -CN,
  • R 3 , R 4 , and R 5 are each independently selected from hydrogen, fluoro, chloro, bromo, iodo, Ci_ alkyl, Ci- 4 alkoxy, -OH, -COOH, -CF 3 , C1-4alkoxyC1 -4alkyl, -N0 2 , -NH 2 , cycloalkyl, -OC(H)F 2 , -C(H)F 2 , -OCH 2 F, -CH 2 F, -CHF 2 , -OCF 3 , -CN, and cycloalkyl; and R 3 , R 4 , and R 5 are each independently selected from hydrogen, fluoro,
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (I).
  • R is a substituted pyrrolo[2,3-d]pyrimidinyl.
  • R is a substituted pyrazolo[3,4-d]pyrimidinyl.
  • R is a substituted pyrrolo[3,2-c]pyridinyl.
  • R 2 is selected from:
  • R 3 , R 4 , and R 5 are selected from: hydrogen, halo, alkyl, and CF3.
  • R is fluoro
  • R 11 is selected from:
  • substituted bicycloheteroaryl and said substituted heteroaryl are substituted with from one to five substituents independently selected from: fluoro,
  • Ci_ 4 alkyl independently selected from: Ci_ 4 alkyl, Ci- 4 alkoxy, -OH,
  • R 14 , and R 15 are each independently selected from hydrogen, fluoro,
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (II).
  • R is a substituted pyrrolo[2,3-d]pyrimidinyl.
  • R is a substituted pyrazolo[3,4- d]pyrimidinyl.
  • R is a substituted pyrrolo[3,2-c]pyridinyl.
  • R 2 is selected from:
  • R 3 , and R 5 are hydrogen.
  • R 4 is fluoro
  • R 21 is selected from:
  • substituted bicycloheteroaryl is substituted with from one to five substituents independently selected from:
  • Ci-6alkyl substituted with from 1 to 5 substituents independently selected from: fluoro, chloro, bromo, iodo, Ci- 4 alkoxy, -OH, Ci- 4 alkyl, cycloalkyl, -COOH, -CF 3 , -N0 2 , -NH 2 and -CN,
  • Ci_ alkyl independently selected from: Ci_ alkyl, Ci_ alkoxy, -OH, -COOH, -CF 3 , C1-4alkoxyC1-4alkyl, oxo, -N0 2 , -NH 2 and -CN,
  • R 23 , R 24 , and R 25 are each independently selected from hydrogen, fluoro,
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (III).
  • R is a substituted pyrrolo[2,3-c ]pyrimidinyl.
  • R 2 is a substituted pyrazolo[3,4- d]pyrimidinyl.
  • R 2 is a substituted pyrrolo[3,2-c]pyridinyl.
  • R 22 is selected from:
  • R 23 and R 25 are hydrogen.
  • R 24 is fluoro.
  • R 33 , R 34 , and R 35 are each independently selected from hydrogen, fluoro,
  • R is selected from: hydrogen and -CH3;
  • R is selected from:
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (IV).
  • R 32 is selected from:
  • R , and R are hydrogen.
  • R is fluoro
  • R 41 is selected from:
  • R 42 is selected from:
  • R 43 , R 44 , and R 45 are each independently selected from hydrogen, fluoro,
  • R is selected from: hydrogen and Ci -6alkyl
  • R is selected from: hydrogen and -CH3;
  • This invention also relates to pharmaceutically acceptable salts of the compounds of Formula (V).
  • R 42 is selected from:
  • R 43 , and R 45 are hydrogen.
  • R 44 is fluoro.
  • the compound of Formula (I) are selected from the list:
  • 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).
  • the salts, including pharmaceutically acceptable salts, of the compounds of the invention are readily prepared by those of skill in the art.
  • 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.
  • the stereochemistry of a chiral center present in a compound of Formula (I), or in any chemical structure illustrated herein if not specified the structure is intended to encompass all individual stereoisomers and all mixtures thereof.
  • compounds according to Formula (I) containing one or more chiral centers may be used as racemic mixtures, enantiomerically enriched mixtures, or as enantiomerically pure individual stereoisomers.
  • 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.
  • the invention includes all such polymorphs.
  • Alkyl refers to a hydrocarbon chain having the specified number of "member atoms".
  • C-1 -C5 alkyl refers to an alkyl group having from 1 to 6 member atoms.
  • Alkyl groups may be saturated, unsaturated, straight or branched .
  • Representative branched alkyl groups have one, two, or three branches.
  • Alkyl includes, but is not limited to: methyl, ethyl, ethylene, alkynyl (such as ethynyl), propyl (n-propyl and isopropyl), butene, butyl (n-butyl, isobutyl, and t- butyl), pentyl and hexyl.
  • Alkoxy refers to an -O-alkyl group wherein “alkyl” is as defined herein. For example, C-
  • C4alkoxy refers to an alkoxy group having from 1 to 4 member atoms.
  • Representative branched alkoxy groups have one, two, or three branches. Examples of such groups include methoxy, ethoxy, propoxy, and butoxy.
  • Aryl refers to an aromatic hydrocarbon ring systems.
  • Aryl groups are monocyclic, bicyclic, and fused 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 and tetrahydronaphthalene.
  • aryl is phenyl.
  • Bicycloheteroaryl refers to two fused aromatic rings containing from 1 to 6 heteroatoms as ring member atoms. Bicycloheteroaryl groups containing more than one ring heteroatom may contain different heteroatoms. Bicycloheteroaryl rings have from 6 to 1 1 ring member atoms.
  • Bicycloheteroaryl includes: 1 H-pyrazolo[4,3-c]pyridine, furo[2,3-d]pyrimidine, 1 H-pyrazolo[3,4- djpyrimidine, 1 H-pyrrolo[2,3-d]pyrimidine, 7H-pyrrolo[2,3-d]pyrimidine, thieno[3,2-c]pyridine, thieno[2,3-d]pyrimidine, furo[2,3-c]pyridine, indolyl, isoindolyl, indolizinyl, indazolyl, purinyl, quinolinyl, isoquinolinyl, quinoxalinyl, quinazolinyl, pteridinyl, cinnolinyl, azabenzimidazolyl, tetrahydrobenzimidazolyl, benzimidazolyl, benopyranyl, benzoxazolyl, benzofuranyl, iso
  • 1 H-pyrazolo[3,4-d]pyrimidine 1 H-pyrrolo[2,3- d]pyrimidine, thieno[3,2-c]pyridine, thieno[2,3-d]pyrimidine, pyrrolo[2,1 -f][1 ,2,4]triazin-4-amine indazolyl, quinolinyl, quinazolinyl or benzothiazolyl.
  • 1 H-pyrrolo[2,3-d]pyrimidine 1 H-pyrrolo[2,3-d]pyrimidine.
  • Cycloalkyl refers to a saturated or unsaturated non aromatic hydrocarbon ring having from three to seven carbon atoms. Cycloalkyl groups are monocyclic ring systems. For example, C3-C7 cycloalkyl refers to a cycloalkyl group having from 3 to 7 member atoms. Examples of cycloalkyl as used herein include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutenyl, cyclopentenyl, cyclohexenyl and cycloheptyl.
  • Heteroaryl refers to a monocyclic aromatic 5 to 8 member ring containing from 1 to 7 carbon atoms and containing from 1 to 4 ring member heteroatoms, provided that when the number of carbon atoms is 3, the aromatic ring contains at least two heteroatoms. Heteroaryl groups containing more than one heteroatom may contain different heteroatoms.
  • Heteroaryl includes: pyrrolyl, pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, isothiazolyl, furanyl, furazanyl, thienyl, triazolyl, pyridinyl, pyrimidinyl, pyridazinyl, pyrazinyl, triazinyl, tetrazinyl.
  • heteroaryl includes: pyrazole, pyrrole, isoxazole, pyridine, pyrimidine, pyridazine, and imidazole.
  • Heterocycloalkyl or “heterocyclyl” refers to a saturated or unsaturated non-aromatic ring containing 4 to 12 member atoms, of which 1 to 1 1 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, tetrahydrothienyl, pyrazolidinyl, oxazolidinyl, 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 H-benzimidazol,
  • Heteroatom refers to a nitrogen, sulphur or oxygen atom.
  • ACN Aceonitrile
  • AIBN Azobis(isobutyronitrile)
  • CSF Chemical fluoride
  • ATP Addenosine triphosphate
  • Bis-pinacolatodiboron (4,4,4',4',5,5,5',5'-Octamethyl-2,2'-bi-1 ,3,2-dioxaborolane); BSA (Bovine serum albumin);
  • DIPEA Hiinig's base, A/-ethyl-A/-(1-methylethyl)-2-propanamine
  • DPPA Diphenyl phosphoryl azide
  • HMDS Hexamethyldisilazide
  • IPA isopropyl alcohol
  • LAH Lithium aluminum hydride
  • LDA Lithium diisopropylamide
  • LHMDS Lithium hexamethyldisilazide
  • mCPBA m-Chloroperbezoic acid
  • NBS (/V-bromosuccinimide
  • Pd 2 (dba) 3 Tris(dibenzylideneacetone)dipalladium(O); Pd(dppf)CI 2 .DCMComplex ([1 ,1 '-Bis(diphenylphosphino)ferrocene]dichloropalladium(ll) dichloromethane complex);
  • 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 schemes. 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 ortarget 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.
  • final compounds H can be prepared by converting the bicycloheteroaryl bromide G to the corresponding boronate ester G1 , followed by Suzuki-Miyaura coupling with bromo compound E.
  • bicycloheteroaryl bromides G were prepared as per the literature procedures described in J. Med. Chem. , 2012, 55 (16), pp 7193-7207 and J. Med. Chem. , 2015, 58 (3), pp 1426-1441 . WO/2017/046737.
  • R2 Aryl, heteroaryls, etc
  • R4.R5 H, F.
  • X1 Me, iPr, cyclopropyl, trifluoroethyl etc.
  • R2 Aryl, heteroaryls, etc
  • X1 Me, iPr, cyclopropyl, trifluoroethyl etc.
  • R2 Aryl, heteroaryls, etc
  • R4.R5 H, F.
  • X1 Me, iPr, cyclopropyl, trifluoroethyl etc.
  • the compounds according to Formula (I) and pharmaceutically acceptable salts thereof are inhibitors of PERK. These compounds are potentially useful in the treatment of conditions wherein the underlying pathology is attributable to (but not limited to) activation of the UPR 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 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,
  • cell leukemia plasmacytoma, Immunoblastic large cell leukemia, mantle cell leukemia, multiple myeloma, megakaryoblastic leukemia, multiple myeloma, acute megakaryocytic leukemia, promyelocytic leukemia, erythroleukemia, malignant lymphoma, hodgkins lymphoma, non-hodgkins lymphoma, lymphoblastic T cell lymphoma, Burkitt's lymphoma, follicular lymphoma, neuroblastoma, bladder cancer, urothelial cancer, vulval cancer, cervical cancer, endometrial cancer, renal cancer, mesothelioma, esophageal cancer, salivary gland cancer, hepatocellular cancer, gastric cancer, nasopharangeal cancer, buccal cancer, cancer of the mouth, GIST (gastrointestinal stromal tumor), neuroendocrine cancers and testicular cancer.
  • GIST gastrointestinal strom
  • 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), myelodysplasia 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.
  • MGUS monoclonal gammapathy of unknown significance
  • MUS monoclonal gammapathy of unknown significance
  • myelodysplasia syndrome aplastic anemia
  • cervical lesions aplastic anemia
  • cervical lesions skin nevi (pre-melanoma)
  • PIN prostatic intraepith
  • 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, Parkinson disease, metabolic syndrome, metabolic disorders, Huntington's disease, Creutzfeldt-Jakob Disease, and related prion diseases, fatal familial insomnia, Gerstmann-Straussler-Scheinker syndrome, and related prion diseases, progressive supranuclear palsy, amyotrophic lateral sclerosis, and other diseases associated with UPR activation including: diabetes, myocardial infarction, cardiovascular disease, inflammation, fibrosis, chronic and acute diseases of the liver, fatty liver disease, liver steatosis, liver fibrosis chronic and acute diseases of the lung, lung fibrosis, chronic and acute diseases of the kidney, kidney fibrosis, chronic traumatic encephalopathy (CTE), neurodegeneration, dementia, frontotemporal dementias, tauopathies, Pick's disease, Neimann, Ne
  • the present invention relates to a method 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 will comprise the in vivo administration of a compound of Formula (I).
  • the method of preventing organ damage during the transportation of organs for transplantation will comprise adding a compound of Formula (I) to the solution housing the organ during transportation.
  • the compounds of this invention inhibit angiogenesis which is implicated in the treatment of ocular diseases. Nature Reviews Drug Discovery 4, 71 1 -712 (September 2005).
  • the present invention relates to a method for treating or lessening the severity of ocular diseases/angiogenesis.
  • the disorder of ocular diseases, neurological disorders, pain, including vascular leakage 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 glau
  • 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 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, precancerous syndromes, Alzheimer's disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, diabetes, Parkinson disease, metabolic syndrome, metabolic disorders, Huntington's disease, Creutzfeldt-Jakob Disease, and related prion diseases, fatal familial insomnia, Gerstmann-Straussler-Scheinker syndrome, and related prion diseases, amyotrophic lateral sclerosis, progressive supranuclear palsy, myocardial infarction, cardiovascular disease, inflammation, organ fibrosis, chronic and acute diseases of the liver, fatty liver disease, liver steatosis, liver fibrosis, chronic and acute diseases of the lung, lung fibrosis, chronic and acute diseases of the kidney, kidney fibrosis, chronic traumatic encephalopathy (CTE), neurodegeneration, dementias, frontotemporal dementias, tauopathies, Pick's disease
  • treating is meant prophylactic and therapeutic therapy.
  • 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.
  • the term "effective amount” and derivatives thereof means that amount of a drug or pharmaceutical agent that will elicit the biological or medical response of a tissue, system, animal or human that is being sought, for instance, by a researcher or clinician.
  • therapeutically effective amount means any amount which, as compared to a corresponding subject who has not received such amount, results in improved treatment, healing, prevention, or amelioration of a disease, disorder, or side effect, or a decrease in the rate of advancement of a disease or disorder.
  • the term also includes within its scope amounts effective to enhance normal physiological function.
  • patient or “subject” refers to a human or other animal.
  • patient or subject is 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 overtime 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 of the 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 or 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 coadministered 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 a PERK 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. Hellman (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 anti-folate 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 diter
  • Examples of a further active ingredient or ingredients (anti-neoplastic agent) for use in combination or co-administered with the presently invented PERK 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 WO02/0591 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 salt
  • 5-[[4-[(2,3-dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2- methylbenzenesulfonamide is in the form of a monohydrochloride salt.
  • This salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US01/49367, having an International filing date of December 19, 2001 .
  • Pazopanib is implicated in the treatment of cancer and ocular diseases/angiogenesis.
  • the present invention relates to the treatment of cancer and ocular diseases/angiogenesis, suitably age-related macular degeneration, which method comprises the administration of a compound of Formula (I) alone or in combination with pazopanib.
  • the compound of the invention may be employed with other therapeutic methods of cancer treatment.
  • combination therapy with other chemotherapeutic, hormonal, antibody agents as well as surgical and/or radiation treatments other than those mentioned above are envisaged.
  • the further anti-cancer therapy is surgical and/or radiotherapy. In one embodiment, the further anti-cancer therapy is at least one additional antineoplastic agent.
  • a combination comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent.
  • a combination comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in therapy.
  • a combination comprising a compound of Formula (I) or pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, for use in treating cancer and/or pre-cancerous syndromes.
  • a combination comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, in the manufacture of a medicament for the treatment of cancer and/or pre-cancerous syndromes.
  • a method of treating cancer comprising administering to a human in need thereof a therapeutically effective amount of a combination comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent.
  • a pharmaceutical composition comprising a combination comprising a compound of Formula (I) or a pharmaceutically acceptable salt thereof and at least one further therapeutic agent, particularly at least one anti-neoplastic agent and one or more of pharmaceutically acceptable carriers, diluents and excipients.
  • anti-neoplastic agent that has activity versus a susceptible tumor being treated may be utilized in the combination.
  • Typical anti-neoplastic agents useful 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 anti-folate compounds; topoisomerase I inhibitors such as camptothecins; hormones and hormonal analogues; signal transduction pathway inhibitors; non- receptor tyrosine angiogenesis inhibitors; immunotherapeutic agents; proapoptotic agents; cell cycle signaling inhibitors; immuno-oncology agents and immuno
  • Anti-microtubule or anti-mitotic agents are examples of anti-microtubule or anti-mitotic agents.
  • Anti-microtubule or anti-mitotic agents are phase specific agents active against the microtubules of tumor cells during M or the mitosis phase of the cell cycle.
  • anti-microtubule agents include, but are not limited to, diterpenoids and vinca alkaloids.
  • Diterpenoids which are derived from natural sources, are phase specific anti -cancer agents that operate at the G 2 /M phases of the cell cycle. It is believed that the diterpenoids stabilize the ⁇ -tubulin subunit of the microtubules, by binding with this protein. Disassembly of the protein appears then to be inhibited with mitosis being arrested and cell death following. Examples of diterpenoids include, but are not limited to, paclitaxel and its analog docetaxel.
  • Paclitaxel 5p,20-epoxy-1 ,2a,4,7p,10p,13a-hexa-hydroxytax-1 1 -en-9-one 4,10- diacetate 2-benzoate 13-ester with (2R,3S)-N-benzoyl-3-phenylisoserine; is a natural diterpene product isolated from the Pacific yew tree Taxus brevifolia and is commercially available as an injectable solution TAXOL®. It is a member of the taxane family of terpenes.
  • Paclitaxel has been approved for clinical use in the treatment of refractory ovarian cancer in the United States (Markman et al., Yale Journal of Biology and Medicine, 64:583, 1991 ; McGuire et al., Ann. Intern, Med., 1 1 1 :273,1989) and for the treatment of breast cancer (Holmes et al., J. Nat. Cancer Inst., 83:1797,1991 .) It is a potential candidate for treatment of neoplasms in the skin (Einzig et. al., Proc. Am. Soc. Clin. Oncol., 20:46) and head and neck carcinomas (Forastire et. al., Sem. Oncol., 20:56, 1990).
  • the compound also shows potential for the treatment of polycystic kidney disease (Woo et. al., Nature, 368:750. 1994), lung cancer and malaria.
  • Treatment of patients with paclitaxel results in bone marrow suppression (multiple cell lineages, Ignoff, R.J. et. al, Cancer Chemotherapy Pocket Guide ⁇ 1998) related to the duration of dosing above a threshold concentration (50nM) (Kearns, CM. et. al., Seminars in Oncology, 3(6) p.16-23, 1995).
  • Docetaxel (2R.3S)- N-carboxy-3-phenylisoserine,N-fe/?-butyl ester, 13-ester with ⁇ -20-epoxy-l ,2a,4,7p,10p,13a-hexahydroxytax-1 1 -en-9-one 4-acetate 2-benzoate, trihydrate; is commercially available as an injectable solution as TAXOTERE®.
  • Docetaxel is indicated for the treatment of breast cancer.
  • Docetaxel is a semisynthetic derivative of paclitaxel q.v. , prepared using a natural precursor, 10-deacetyl-baccatin III, extracted from the needle of the European Yew tree.
  • Vinca alkaloids are phase specific anti-neoplastic agents derived from the periwinkle plant. Vinca alkaloids act at the M phase (mitosis) of the cell cycle by binding specifically to tubulin. Consequently, the bound tubulin molecule is unable to polymerize into microtubules. Mitosis is believed to be arrested in metaphase with cell death following. Examples of vinca alkaloids include, but are not limited to, vinblastine, vincristine, and vinorelbine.
  • Vinblastine vincaleukoblastine sulfate
  • VELBAN® an injectable solution.
  • Myelosuppression is the dose limiting side effect of vinblastine.
  • Vincristine vincaleukoblastine, 22-oxo-, sulfate
  • ONCOVIN® an injectable solution.
  • Vincristine is indicated for the treatment of acute leukemias and has also found use in treatment regimens for Hodgkin's and non-Hodgkin's malignant lymphomas.
  • Alopecia and neurologic effects are the most common side effect of vincristine and to a lesser extent myelosupression and gastrointestinal mucositis effects occur.
  • Vinorelbine 3',4'-didehydro -4'-deoxy-C'-norvincaleukoblastine [R-(R*,R*)-2,3- dihydroxybutanedioate (1 :2)(salt)], commercially available as an injectable solution of vinorelbine tartrate (NAVELBINE®), is a semisynthetic vinca alkaloid.
  • Vinorelbine is indicated as a single agent or in combination with other chemotherapeutic agents, such as cisplatin, in the treatment of various solid tumors, particularly non-small cell lung, advanced breast, and hormone refractory prostate cancers. Myelosuppression is the most common dose limiting side effect of vinorelbine.
  • Platinum coordination complexes are non-phase specific anti-cancer agents, which are interactive with DNA.
  • the platinum complexes enter tumor cells, undergo, aquation and form intra- and interstrand crosslinks with DNA causing adverse biological effects to the tumor.
  • Examples of platinum coordination complexes include, but are not limited to, oxaliplatin, cisplatin and carboplatin.
  • Cisplatin cis-diamminedichloroplatinum
  • PLATINOL® an injectable solution.
  • Cisplatin is primarily indicated in the treatment of metastatic testicular and ovarian cancer and advanced bladder cancer.
  • Carboplatin platinum, diammine [1 ,1 -cyclobutane-dicarboxylate(2-)-0,0'], is commercially available as PARAPLATIN® as an injectable solution. Carboplatin is primarily indicated in the first and second line treatment of advanced ovarian carcinoma.
  • Alkylating agents are non-phase anti-cancer specific agents and strong electrophiles. Typically, alkylating agents form covalent linkages, by alkylation, to DNA through nucleophilic moieties of the DNA molecule such as phosphate, amino, sulfhydryl, hydroxyl, carboxyl, and imidazole groups. Such alkylation disrupts nucleic acid function leading to cell death.
  • alkylating agents include, but are not limited to, nitrogen mustards such as cyclophosphamide, melphalan, and chlorambucil; alkyl sulfonates such as busulfan; nitrosoureas such as carmustine; and triazenes such as dacarbazine.
  • Cyclophosphamide 2-[bis(2-chloroethyl)amino]tetrahydro-2H-1 ,3,2-oxazaphosphorine 2-oxide monohydrate, is commercially available as an injectable solution or tablets as CYTOXAN®. Cyclophosphamide is indicated as a single agent or in combination with other chemotherapeutic agents, in the treatment of malignant lymphomas, multiple myeloma, and leukemias.
  • Melphalan 4-[bis(2-chloroethyl)amino]-L-phenylalanine, is commercially available as an injectable solution or tablets as ALKERAN®. Melphalan is indicated for the palliative treatment of multiple myeloma and non-resectable epithelial carcinoma of the ovary. Bone marrow suppression is the most common dose limiting side effect of melphalan.
  • Chlorambucil 4-[bis(2-chloroethyl)amino]benzenebutanoic acid, is commercially available as LEUKERAN® tablets. Chlorambucil is indicated for the palliative treatment of chronic lymphatic leukemia, and malignant lymphomas such as lymphosarcoma, giant follicular lymphoma, and Hodgkin's disease.
  • Busulfan 1 ,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia.
  • Carmustine 1 ,3-[bis(2-chloroethyl)-1 -nitrosourea, is commercially available as single vials of lyophilized material as BiCNU®. Carmustine is indicated for the palliative treatment as a single agent or in combination with other agents for brain tumors, multiple myeloma, Hodgkin's disease, and non-Hodgkin's lymphomas.
  • dacarbazine 5-(3,3-dimethyl-1 -triazeno)-imidazole-4-carboxamide, is commercially available as single vials of material as DTIC-Dome®. dacarbazine is indicated for the treatment of metastatic malignant melanoma and in combination with other agents for the second line treatment of Hodgkin's Disease.
  • Antibiotic anti-neoplastics are included in the following:
  • Antibiotic anti-neoplastics are non-phase specific agents, which bind or intercalate with DNA. Typically, such action results in stable DNA complexes or strand breakage, which disrupts ordinary function of the nucleic acids leading to cell death.
  • antibiotic anti-neoplastic agents include, but are not limited to, actinomycins such as dactinomycin, anthrocyclins such as daunorubicin and doxorubicin; and bleomycins.
  • Dactinomycin also known as Actinomycin D
  • Actinomycin D is commercially available in injectable form as COSMEGEN®. Dactinomycin is indicated for the treatment of Wilm's tumor and rh a bd o my osa rco ma .
  • Daunorubicin (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo- hexopyranosyl)oxy]-7,8,9,10-tetrahydro-6,8,1 1 -trihydroxy-1 -methoxy-5,12 naphthacenedione hydrochloride, is commercially available as a liposomal injectable form as DAUNOXOME® or as an injectable as CERUBIDINE®. Daunorubicin is indicated for remission induction in the treatment of acute nonlymphocytic leukemia and advanced HIV associated Kaposi's sarcoma.
  • Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblasts leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas.
  • Bleomycin a mixture of cytotoxic glycopeptide antibiotics isolated from a strain of Streptomyces verticillus, is commercially available as BLENOXANE®. Bleomycin is indicated as a palliative treatment, as a single agent or in combination with other agents, of squamous cell carcinoma, lymphomas, and testicular carcinomas.
  • Topoisomerase II inhibitors include, but are not limited to, epipodophyllotoxins.
  • Epipodophyllotoxins are phase specific anti-neoplastic agents derived from the mandrake plant. Epipodophyllotoxins typically affect cells in the S and G 2 phases of the cell cycle by forming a ternary complex with topoisomerase II and DNA causing DNA strand breaks. The strand breaks accumulate and cell death follows.
  • Examples of epipodophyllotoxins include, but are not limited to, etoposide and teniposide.
  • Etoposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-ethylidene-p-D- glucopyranoside] is commercially available as an injectable solution or capsules as VePESID® and is commonly known as VP-16. Etoposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of testicular and non-small cell lung cancers.
  • Teniposide 4'-demethyl-epipodophyllotoxin 9[4,6-0-(R )-thenylidene-p-D- glucopyranoside], is commercially available as an injectable solution as VUMON® and is commonly known as VM-26. Teniposide is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia in children.
  • Antimetabolite neoplastic agents are:
  • Antimetabolite neoplastic agents are phase specific anti-neoplastic agents that act at S phase (DNA synthesis) of the cell cycle by inhibiting DNA synthesis or by inhibiting purine or pyrimidine base synthesis and thereby limiting DNA synthesis. Consequently, S phase does not proceed and cell death follows.
  • Examples of antimetabolite anti-neoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine.
  • 5-fluorouracil 5-fluoro-2,4- (1 H,3H) pyrimidinedione
  • fluorouracil is commercially available as fluorouracil.
  • Administration of 5-fluorouracil leads to inhibition of thymidylate synthesis and is also incorporated into both RNA and DNA. The result typically is cell death.
  • 5-fluorouracil is indicated as a single agent or in combination with other chemotherapy agents in the treatment of carcinomas of the breast, colon, rectum, stomach and pancreas.
  • Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
  • Cytarabine 4-amino-1-p-D-arabinofuranosyl-2 (1 H)-pyrimidinone, is commercially available as CYTOSAR-U® and is commonly known as Ara-C. It is believed that cytarabine exhibits cell phase specificity at S-phase by inhibiting DNA chain elongation by terminal incorporation of cytarabine into the growing DNA chain. Cytarabine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other cytidine analogs include 5-azacytidine and 2',2'-difluorodeoxycytidine (gemcitabine).
  • Mercaptopunne 1 ,7-dihydro-6H-purine-6-thione monohydrate
  • PURINETHOL® is commercially available as PURINETHOL®.
  • Mercaptopunne exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Mercaptopunne is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia.
  • a useful mercaptopunne analog is azathioprine.
  • Thioguanine, 2-amino-1 ,7-dihydro-6H-purine-6-thione is commercially available as
  • TABLOID® Thioguanine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism. Thioguanine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Other purine analogs include pentostatin, erythrohydroxynonyladenine, fludarabine phosphate, and cladribine.
  • Gemcitabine 2'-deoxy-2', 2'-difluorocytidine monohydrochloride ( ⁇ -isomer), is commercially available as GEMZAR®. Gemcitabine exhibits cell phase specificity at S-phase and by blocking progression of cells through the G1 /S boundary. Gemcitabine is indicated in combination with cisplatin in the treatment of locally advanced non-small cell lung cancer and alone in the treatment of locally advanced pancreatic cancer.
  • Methotrexate N-[4[[(2,4-diamino-6-pteridinyl) methyl]methylamino] benzoyl]-L-glutamic acid, is commercially available as methotrexate sodium. Methotrexate exhibits cell phase effects specifically at S-phase by inhibiting DNA synthesis, repair and/or replication through the inhibition of dyhydrofolic acid reductase which is required for synthesis of purine nucleotides and thymidylate.
  • Methotrexate is indicated as a single agent or in combination with other chemotherapy agents in the treatment of choriocarcinoma, meningeal leukemia, non-Hodgkin's lymphoma, and carcinomas of the breast, head, neck, ovary and bladder.
  • Camptothecins including, camptothecin and camptothecin derivatives are available or under development as Topoisomerase I inhibitors. Camptothecins cytotoxic activity is believed to be related to its Topoisomerase I inhibitory activity. Examples of camptothecins include, but are not limited to irinotecan, topotecan, and the various optical forms of 7-(4-methylpiperazino- methylene)-10,1 1 -ethylenedioxy-20-camptothecin described below.
  • Irinotecan is a derivative of camptothecin which binds, along with its active metabolite SN-38, to the topoisomerase I - DNA complex.
  • cytotoxicity occurs as a result of irreparable double strand breaks caused by interaction of the topoisomerase I : DNA : irintecan or SN-38 ternary complex with replication enzymes.
  • Irinotecan is indicated for treatment of metastatic cancer of the colon or rectum.
  • Topotecan HCI (S)-10-[(dimethylamino)methyl]-4-ethyl-4,9-dihydroxy-1 H- pyrano[3',4',6,7]indolizino[1 ,2-b]quinoline-3,14-(4H,12H)-dione monohydrochloride, is commercially available as the injectable solution HYCAMTIN®.
  • Topotecan is a derivative of camptothecin which binds to the topoisomerase I - DNA complex and prevents religation of singles strand breaks caused by Topoisomerase I in response to torsional strain of the DNA molecule. Topotecan is indicated for second line treatment of metastatic carcinoma of the ovary and small cell lung cancer.
  • Hormones and hormonal analogues are hormones and hormonal analogues:
  • Hormones and hormonal analogues are useful compounds for treating cancers in which there is a relationship between the hormone(s) and growth and/or lack of growth of the cancer.
  • hormones and hormonal analogues useful in cancer treatment include, but are not limited to, adrenocorticosteroids such as prednisone and prednisolone which are useful in the treatment of malignant lymphoma and acute leukemia in children ; aminoglutethimide and other aromatase inhibitors such as anastrozole, letrazole, vorazole, and exemestane useful in the treatment of adrenocortical carcinoma and hormone dependent breast carcinoma containing estrogen receptors; progestrins such as megestrol acetate useful in the treatment of hormone dependent breast cancer and endometrial carcinoma; estrogens, estrogens, and anti-estrogens such as fulvestrant, flutamide, nilutamide, bicalutamide, cyproterone acetate and 5a-re
  • GnRH gonadotropin-releasing hormone
  • LH leutinizing hormone
  • FSH follicle stimulating hormone
  • Signal transduction pathway inhibitors are those inhibitors, which block or inhibit a chemical process which evokes an intracellular change. As used herein this change is cell proliferation or differentiation.
  • Signal tranduction inhibitors useful in the present invention include inhibitors of receptor tyrosine kinases, non-receptor tyrosine kinases, SH2/SH3domain blockers, serine/threonine kinases, phosphotidyl inositol-3 kinases, myo-inositol signaling, and Ras oncogenes.
  • protein tyrosine kinases catalyse the phosphorylation of specific tyrosyl residues in various proteins involved in the regulation of cell growth.
  • protein tyrosine kinases can be broadly classified as receptor or non-receptor kinases.
  • Receptor tyrosine kinases are transmembrane proteins having an extracellular ligand binding domain, a transmembrane domain, and a tyrosine kinase domain. Receptor tyrosine kinases are involved in the regulation of cell growth and are generally termed growth factor receptors. Inappropriate or uncontrolled activation of many of these kinases, i.e. aberrant kinase growth factor receptor activity, for example by over-expression or mutation, has been shown to result in uncontrolled cell growth. Accordingly, the aberrant activity of such kinases has been linked to malignant tissue growth. Consequently, inhibitors of such kinases could provide cancer treatment methods.
  • Growth factor receptors include, for example, epidermal growth factor receptor (EGFr), platelet derived growth factor receptor (PDGFr), erbB2, erbB4, ret, vascular endothelial growth factor receptor (VEGFr), tyrosine kinase with immunoglobulin-like and epidermal growth factor homology domains (TIE-2), insulin growth factor -I (IGFI) receptor, macrophage colony stimulating factor (cfms), BTK, ckit, cmet, fibroblast growth factor (FGF) receptors, Trk receptors (TrkA, TrkB, and TrkC), ephrin (eph) receptors, and the RET protooncogene.
  • EGFr epidermal growth factor receptor
  • PDGFr platelet derived growth factor receptor
  • erbB2 erbB2
  • VEGFr vascular endothelial growth factor receptor
  • TIE-2 immunoglobulin-like and epidermal growth factor homo
  • inhibitors of growth receptors include ligand antagonists, antibodies, tyrosine kinase inhibitors and anti-sense oligonucleotides.
  • Growth factor receptors and agents that inhibit growth factor receptor function are described, for instance, in Kath, John C, Exp. Opin. Ther. Patents (2000) 10(6):803-818; Shawver et al DDT Vol 2, No. 2 February 1997; and Lofts, F. J. et al, "Growth factor receptors as targets", New Molecular Targets for Cancer Chemotherapy, ed. Workman, Paul and Kerr, David, CRC press 1994, London.
  • Non-receptor tyrosine kinases which are targets or potential targets of anti-cancer drugs, include cSrc, Lck, Fyn, Yes, Jak, cAbl, FAK (Focal adhesion kinase), Brutons tyrosine kinase, and Bcr-Abl.
  • Such non-receptor kinases and agents which inhibit non-receptor tyrosine kinase function are described in Sinh, S.
  • SH2/SH3 domain blockers are agents that disrupt SH2 or SH3 domain binding in a variety of enzymes or adaptor proteins including, PI3-K p85 subunit, Src family kinases, adaptor molecules (She, Crk, Nek, Grb2) and Ras-GAP.
  • SH2/SH3 domains as targets for anti-cancer drugs are discussed in Smithgall, T.E. (1995), Journal of Pharmacological and Toxicological Methods. 34(3) 125-32.
  • Inhibitors of Serine/Threonine Kinases including MAP kinase cascade blockers which include blockers of Raf kinases (rafk), Mitogen or Extracellular Regulated Kinase (MEKs), and Extracellular Regulated Kinases (ERKs); and Protein kinase C family member blockers including blockers of PKCs (alpha, beta, gamma, epsilon, mu, lambda, iota, zeta).
  • IkB kinase family IKKa, IKKb
  • PKB family kinases akt kinase family members
  • TGF beta receptor kinases TGF beta receptor kinases.
  • Serine/Threonine kinases and inhibitors thereof are described in Yamamoto, T., Taya, S., Kaibuchi, K., (1999), Journal of Biochemistry. 126 (5) 799-803; Brodt, P, Samani, A., and Navab, R. (2000), Biochemical Pharmacology, 60. 1 101 -1 107; Massague, J., Weis-Garcia, F. (1996) Cancer Surveys. 27:41 -64; Philip, P.A., and Harris, A.L. (1995), Cancer Treatment and Research. 78: 3-27, Lackey, K. et al Bioorganic and Medicinal Chemistry Letters, (10), 2000, 223-226; U.S. Patent No. 6,268,391 ; and Martinez-lacaci, L, et al, Int. J. Cancer (2000), 88(1), 44-52.
  • Inhibitors of Phosphotidyl inositol-3 Kinase family members including blockers of Pekinese, ATM, DNA-PK, and Ku are also useful in the present invention.
  • Such kinases are discussed in Abraham, R.T. (1996), Current Opinion in Immunology. 8 (3) 412-8; Canman, C.E., Lim, D.S. (1998), Oncogene 17 (25) 3301 -3308; Jackson, S.P. (1997), International Journal of Biochemistry and Cell Biology. 29 (7):935-8; and Zhong, H. et al, Cancer res, (2000) 60(6), 1541 - 1545.
  • Myo-inositol signaling inhibitors such as phospholipase C blockers and Myoinositol analogues.
  • signal inhibitors are described in Powis, G., and Kozikowski A., (1994) New Molecular Targets for Cancer Chemotherapy ed., Paul Workman and David Kerr, CRC press 1994, London.
  • Ras Oncogene Another group of signal transduction pathway inhibitors are inhibitors of Ras Oncogene.
  • Such inhibitors include inhibitors of farnesyltransferase, geranyl-geranyl transferase, and CAAX proteases as well as anti-sense oligonucleotides, ribozymes and immunotherapy.
  • Such inhibitors have been shown to block ras activation in cells containing wild type mutant ras , thereby acting as antiproliferation agents.
  • Ras oncogene inhibition is discussed in Scharovsky, O.G., Rozados, V.R., Gervasoni, S.I. Matar, P. (2000), Journal of Biomedical Science. 7(4) 292- 8; Ashby, M.N. (1998), Current Opinion in Lipidology. 9 (2) 99 - 102; and BioChim. Biophys. Acta, (19899) 1423(3):19-30.
  • antibody antagonists to receptor kinase ligand binding may also serve as signal transduction inhibitors.
  • This group of signal transduction pathway inhibitors includes the use of humanized antibodies to the extracellular ligand binding domain of receptor tyrosine kinases.
  • Imclone C225 EGFR specific antibody see Green, M.C. et al, Monoclonal Antibody Therapy for Solid Tumors, Cancer Treat.
  • Herceptin ® erbB2 antibody see Tyrosine Kinase Signalling in Breast cancenerbB Family Receptor Tyrosine Kinases, Breast cancer Res., 2000, 2(3), 176-183
  • 2CB VEGFR2 specific antibody see Brekken, R.A. et al, Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice, Cancer Res. (2000) 60, 51 17- 5124).
  • Anti-angiogenic agents including non-receptor MEK angiogenesis inhibitors may alo be useful.
  • Anti-angiogenic agents such as those which inhibit the effects of vascular edothelial growth factor, (for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM], and compounds that work by other mechanisms (for example linomide, inhibitors of integrin ⁇ 3 function, endostatin and angiostatin);
  • vascular edothelial growth factor for example the anti-vascular endothelial cell growth factor antibody bevacizumab [AvastinTM]
  • linomide inhibitors of integrin ⁇ 3 function, endostatin and angiostatin
  • Immunotherapy approaches including for example ex-vivo and in- vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor, approaches to decrease T-cell anergy, approaches using transfected immune cells such as cytokine-transfected dendritic cells, approaches using cytokine-transfected tumour cell lines and approaches using anti-idiotypic antibodies
  • cytokines such as interleukin 2, interleukin 4 or granulocyte-macrophage colony stimulating factor
  • approaches to decrease T-cell anergy approaches using transfected immune cells such as cytokine-transfected dendritic cells
  • Proapoptotic agents including for example ex-vivo and in- vivo approaches to increase the immunogenecity of patient tumour cells, such as transfection with cyto
  • Agents used in proapoptotic regimens may also be used in the combination of the present invention.
  • Cell cycle signalling inhibitors inhibit molecules involved in the control of the cell cycle.
  • a family of protein kinases called cyclin dependent kinases (CDKs) and their interaction with a family of proteins termed cyclins controls progression through the eukaryotic cell cycle. The coordinate activation and inactivation of different cyclin/CDK complexes is necessary for normal progression through the cell cycle.
  • CDKs cyclin dependent kinases
  • Several inhibitors of cell cycle signalling are under development. For instance, examples of cyclin dependent kinases, including CDK2, CDK4, and CDK6 and inhibitors for the same are described in, for instance, Rosania et al, Exp. Opin. Ther. Patents (2000) 10(2):215-230.
  • the combination of the present invention comprises a compound of Formula I or a salt or solvate thereof and at least one anti-neoplastic agent selected from 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 MEK angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
  • anti-neoplastic agent selected from 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 MEK angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, and cell cycle signaling inhibitors.
  • the combination of the present invention comprises a compound of
  • At least one anti-neoplastic agent agent is a diterpenoid.
  • At least one anti-neoplastic agent is a vinca alkaloid.
  • the combination of the present invention comprises a compound of Formula I or a salt or solvate thereof and at least one anti-neoplastic agent, which is a platinum coordination complex.
  • At least one anti-neoplastic agent is paclitaxel, carboplatin, or vinorelbine.
  • At least one anti-neoplastic agent is carboplatin.
  • At least one anti-neoplastic agent is vinorelbine.
  • At least one anti-neoplastic agent is paclitaxel.
  • the combination of the present invention comprises a compound of Formula I and salts or solvates thereof and at least one anti-neoplastic agent which is a signal transduction pathway inhibitor.
  • the signal transduction pathway inhibitor is an inhibitor of a growth factor receptor kinase VEGFR2, TIE2, PDGFR, BTK, erbB2, EGFr, IGFR-1 , TrkA, TrkB, TrkC, or c-fms.
  • the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase rafk, akt, or PKC-zeta.
  • the signal transduction pathway inhibitor is an inhibitor of a nonreceptor tyrosine kinase selected from the src family of kinases.
  • the signal transduction pathway inhibitor is an inhibitor of c-src.
  • the signal transduction pathway inhibitor is an inhibitor of Ras oncogene selected from inhibitors of farnesyl transferase and geranylgeranyl transferase.
  • the signal transduction pathway inhibitor is an inhibitor of a serine/threonine kinase selected from the group consisting of PI3K.
  • the signal transduction pathway inhibitor is a dual EGFr/erbB2 inhibitor, for example N- ⁇ 3-Chloro-4-[(3-fluorobenzyl) oxy]phenyl ⁇ -6-[5-( ⁇ [2-(methanesulphonyl) ethyl]amino ⁇ methyl)-2-furyl]-4-quinazolinamine (structure below):
  • the combination of the present invention comprises a compound of Formula I or a salt or solvate thereof and at least one anti-neoplastic agent which is a cell cycle signaling inhibitor.
  • cell cycle signaling inhibitor is an inhibitor of CDK2, CDK4 or
  • immunostimulatory agent refers to any agent that can stimulate the immune system.
  • immunostimulatory agents include, but are not limited to, vaccine adjuvants, such as Toll-like receptor agonists, T-cell checkpoint blockers, such as mAbs to PD-1 and CTL4 and T-cell checkpoint agonist, such as agonist mAbs to OX-40 and ICOS.
  • anti-neoplastic agent for use in combination or co-administered with the presently invented compound of Formula (I) 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:
  • WQ2013078174 US Application No. 13/51 1 ,538 (filed August 7, 2012), which is the US National Phase of International Application No. PCT/US10/58007 (filed 2010);
  • 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 Application No. 13/51 1 ,538.
  • the anti-PD-L1 antibody comprises the CDRs of an antibody disclosed in US Application No. 13/51 1 ,538.
  • the antibody to PD-L1 is an antibody disclosed in Application
  • the anti-PD-L1 antibody comprises the CDRs of an antibody disclosed in US Application No. 13/478,51 1 .
  • 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.
  • PD-1 antagonist a further active ingredient or ingredients for use in combination or co-administered with the presently invented compound of Formula (I) are PD-1 antagonist.
  • 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 .
  • Alternative names or synonyms for 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, respectively.
  • 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.
  • PD-1 antagonists useful in the any of the aspects and embodiments of the present invention include an immunoadhesin that specifically binds to PD-1 , and preferably specifically binds to human PD-1 , e.g., a fusion protein containing the extracellular or PD-1 binding portion of PD-L1 or PD-L2 fused to a constant region such as an Fc region of an immunoglobulin molecule.
  • immunoadhesion molecules that specifically bind to PD-1 are described in WO2010/027827 and WO201 1 /066342.
  • Specific 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 8.006,449.
  • immuno-modulators Additional examples of a further active ingredient or ingredients (anti-neoplastic agent) for use in combination or co-administered with the presently invented compound of Formula (I) 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 forthe 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, LAG 3 antibodies, TIM-3 antibodies, 41 BB antibodies and GITR antibodies.
  • Yervoy 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.
  • CD134 also known as OX40
  • OX40 is a member of the T FR-superfamily of receptors which is not constitutively expressed on resting naive T_cejjs, unlike GD28.
  • OX40 is a secondary expressed after 24 to 72 hours following activation; its jjcjand, S fiL is also not expressed on resting antigen presenting cells, but is following their activation. Expression of OX40 is dependent on full activation of the T_cel[; without CD28, expression of OX40 is delayed and of fourfold lower levels.
  • OX-40 antibodies, OX-40 fusion proteins and methods of using them are disclosed in US Patent Nos: US 7,504,101 ; US 7,758,852; US 7,858,765; US 7,550,140; US 7,960,515; WO2012027328; WO2013028231 .
  • Toll-like receptor refers to a member of the Toll-like receptor family of proteins or a fragment thereof that senses a microbial product and/or initiates an adaptive immune response.
  • a TLR activates a dendritic cell (DC).
  • Toll-like receptors are a family of pattern recognition receptors that were initially identified as sensors of the innate immune system that recognize microbial pathogens. TLRs recognize distinct structures in microbes, often referred to as "PAMPs" (pathogen associated molecular patterns). Ligand binding to TLRs invokes a cascade of intra-cellular signaling pathways that induce the production of factors involved in inflammation and immunity.
  • TLRs that are expressed on the surface of cells include TLR-I, -2, -4, -5, and -6, while TLR-3, -7/8, and -9 are expressed with the ER compartment.
  • Human DC subsets can be identified on the basis of distinct TLR expression patterns.
  • the myeloid or "conventional" subset of DC expresses TLRs 1 -8 when stimulated, and a cascade of activation markers (e.g. CD80, CD86, MHC class I and II, CCR7), pro-inflammatory cytokines, and chemokines are produced.
  • a cascade of activation markers e.g. CD80, CD86, MHC class I and II, CCR7
  • DCs acquire an enhanced capacity to take up antigens and present them in an appropriate form to T cells.
  • plasmacytoid subset of DC expresses only TLR7 and TLR9 upon activation, with a resulting activation of NK cells as well as T-cells.
  • activating DC with TLR agonists may be beneficial for priming anti-tumor immunity in an immunotherapy approach to the treatment of cancer. It has also been suggested that successful treatment of breast cancer using radiation and chemotherapy requires TLR4 activation.
  • TLR agonists known in the art and finding use in the present invention include, but are not limited to, the following: Pam3Cys, a TLRI/2 agonist; CFA, a TLR2 agonist; MALP2, a TLR2 agonist; Pam2Cys, a TLR2 agonist; FSL-I, a TLR-2 agonist; Hib-OMPC, a TLR-2 agonist; polyribosinic:polyribocytidic acid (Poly l:C), a TLR3 agonist; polyadenosine-polyuridylic acid (poly AU), a TLR3 agonist; Polyinosinic-Polycytidylic acid stabilized with poly-L-lysine and carboxymethylcellulose (Hiltonol), a TLR3 agonist; bacterial flagellin a TLR5 agonist; imiquimod, a TLR7 agonist; resiquimod, a TLR7/8 agonist; lox
  • TLR agonists known in the art and finding use in the present invention further include, but are not limited to aminoalkyl glucosaminide phosphates (AGPs) which bind to the TLR4 receptor are known to be useful as vaccine adjuvants and immunostimulatory agents for stimulating cytokine production, activating macrophages, promoting innate immune response, and augmenting antibody production in immunized animals.
  • AGPs aminoalkyl glucosaminide phosphates
  • An example of a naturally occurring TLR4 agonist is bacterial LPS.
  • An example of a semisynthetic TLR4 agonist is monophosphoryl lipid A (MPL).
  • AGPs and their immunomodulating effects via TLR4 are disclosed in patent publications such as WO 2006/016997, WO 2001 /090129, and/or U.S. Patent No. 6,1 13,918 and have been reported in the literature. Additional AGP derivatives are disclosed in U.S. Patent No. 7,129,219, U.S. Patent No. 6,525,028 and U.S. Patent No 6,91 1 ,434. Certain AGPs act as agonists of TLR4, while others are recognized as TLR4 antagonist.
  • compositions of the present invention may further comprise one or more additional substances which, because of their adjuvant nature, can act to stimulate the immune system to respond to the cancer antigens present on the inactivated tumor cell(s).
  • adjuvants include, but are not limited to, lipids, liposomes, inactivated bacteria which induce innate immunity (e.g., inactivated or attenuated l/ster/a monocytogenes), compositions which mediate innate immune activation via, (NOD)-like receptors (NLRs), Retinoic acid inducible gene-based (RIG)-l-like receptors (RLRs), and/or C- type lectin receptors (CLRs).
  • NOD non-like receptors
  • RLRs Retinoic acid inducible gene-based
  • CLRs C- type lectin receptors
  • PAMPs examples include lipoproteins, lipopolypeptides, peptidoglycans, zymosan, lipopolysaccharide, neisserial porins, flagellin, profillin, galactoceramide, muramyl dipeptide.
  • Peptidoglycans, lipoproteins, and lipoteichoic acids are cell wall components of Gram-positive. Lipopolysaccharides are expressed by most bacteria, with MPL being one example.
  • Flagellin refers to the structural component of bacterial flagella that is secreted by pathogenic and commensal bacterial.
  • rt.-Galactosylceramide rt.-GalCer
  • Muramyl dipeptide is a bioactive peptidoglycan motif common to all bacteria.
  • TLR agonists are preferably used in combinations with other vaccines, adjuvants and/or immune modulators, and may be combined in various combinations.
  • the herein described compounds of Formula (I) that bind to STING and induce STING-dependent TBKI activation and an inactivated tumor cell which expresses and secretes one or more cytokines which stimulate DC induction, recruitment and/or maturation, as described herein can be administered together with one or more TLR agonists for therapeutic purposes.
  • anti-neoplastic agent for use in combination or co-administered with the presently invented compound of Formula (I) are antibodies to ICOS.
  • CDRs for murine antibodies to human ICOS having agonist activity are shown in PCT/EP2012/055735 (WO 2012/131004).
  • Antibodies to ICOS are also disclosed in WO 2008/137915, WO 2010/056804, EP 1374902, EP1374901 , and EP1 125585.
  • Indoleamine 2,3-dioxygenase 1 is a key immunosuppressive enzyme that modulates the anti-tumor immune response by promoting regulatory T cell generation and blocking effector T cell activation, thereby facilitating tumor growth by allowing cancer cells to avoid immune surveillance.
  • IDO inhibitors Further active ingredients (antineoplastic agents) for use in combination or co-administered with the presently invented compounds of Formula (I) are IDO inhibitors.
  • Epacadostat ((Z)-N-(3-bromo-4-fluorophenyl)-N'- hydroxy-4-[2-(sulfamoylamino)ethylamino]-1 ,2,5-oxadiazole-3-carboxamidine) is a highly potent and selective oral inhibitor of the ID01 enzyme that reverses tumor-associated immune suppression and restores effective anti-tumor immune responses.
  • Epacadostat is disclosed in US patent No. 8,034,953.
  • Additional examples of a further active ingredient or ingredients (anti-neoplastic agent) for use in combination or co-administered with the presently invented compound of Formula (I) are CD73 inhibitors and A2a and A2b adenosine antagonists.
  • 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.
  • a compound of Formula (I) is used as a chemosensitizer to enhance tumor cell killing.
  • a compound of Formula (I) is used in combination as a chemosensitizer to enhance tumor cell killing.
  • a compound of Formula (I) is used in combination with a modulator of ATF-4.
  • a compound of Formula (I) is used in combination with a modulator of ATF-4 to treat diseases/injuries associated with activated unfolded protein response pathways.
  • a compound of Formula (I) is used in combination with a modulator of ATF-4 to treat neurodegenerative diseases.
  • a compound of Formula (I) is used in combination with a modulator of ATF-4 to treat cancer.
  • a compound of Formula (I) is used in combination with a modulator of ATF-4 where the modulator of ATF-4 is ISRIB or another compound that binds to elF2B and enhances global translation.
  • ISRIB is described in International Application PCT/US2014/029568 having an International Filing Date of March 14, 2014, the International Publication Number WO 2014/144952 and an International Publication Date of September 18, 2014.
  • One embodiment of this invention provides a combination comprising:
  • ATF-4 modulation compouns can be identified by the assays described in International Publication Number WO 2014/144952.
  • 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 of Formula (I) and pharmaceutically acceptable salts thereof may be co-administered with at least one other active agent known to be useful for preventing organ damage during and after organ transplantation and in the transportation of organs for transplantation.
  • the pharmaceutically active compounds within the scope of this invention are useful as PERK inhibitors in mammals, particularly humans, in need thereof.
  • the present invention therefore provides a method of treating cancer, neurodegeneration and other conditions requiring PERK 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 PERK 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, transarterial, intradermal, intraocular and parenteral.
  • a PERK 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 PERK inhibitor drug.
  • the pharmaceutically active compounds of the present invention are incorporated into convenient dosage forms such as capsules, tablets, or injectable preparations.
  • 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.
  • 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 - 500 mg/kg of active compound, preferably 0.001 - 100 mg/kg.
  • the selected dose is administered preferably from 1 -6 times daily, orally or parenterally.
  • Preferred forms of parenteral administration include topically, rectally, transdermal ⁇ , 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, with 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 PERK 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.
  • a compound of Formula (I) When administered to prevent organ damage in the transportation of organs for transplantation, a compound of Formula (I) is added to the solution housing the organ during transportation, suitably in a buffered solution.
  • the method of this invention of inducing PERK inhibitory activity in mammals, including humans comprises administering to a subject in need of such activity an effective PERK 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 use as a PERK inhibitor.
  • 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 use in therapy.
  • 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 use in treating cancer, pre-cancerous syndromes, Alzheimer's disease, spinal cord injury, traumatic brain injury, ischemic stroke, stroke, Parkinson disease, diabetes, metabolic syndrome, metabolic disorders, Huntington's disease, Creutzfeldt-Jakob Disease, and related prion diseases, fatal familial insomnia, Gerstmann-Straussler-Scheinker syndrome, and related prion diseases, amyotrophic lateral sclerosis, progressive supranuclear palsy, myocardial infarction, cardiovascular disease, inflammation, organ fibrosis, chronic and acute diseases of the liver, fatty liver disease, liver steatosis, liver fibrosis, chronic and acute diseases of the lung, lung fibrosis, chronic and acute diseases of the kidney, kidney fibrosis, chronic traumatic encephalopathy (CTE), neurodegeneration, dementias, frontotemporal dementias, tauopathies, Pick'
  • 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 use in preventing organ damage during the transportation of organs for transplantation.
  • the invention also provides for a pharmaceutical composition for use as a PERK 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 cancerwhich 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 coadministered with further active ingredients, such as other compounds known to treat cancer, or compounds known to have utility when used in combination with a PERK inhibitor.
  • 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.
  • Step-1 To a stirred solution of 5-bromo-2H-indazole (2.5g ,12.9mmol, 1 equiv) in DMF (40 mL) was added 60% NaH (0.62 g, 15.48mmol, 1 .2 equiv) at 0°C under N 2 atmosphere. The reaction mixture was stirred for 20 min. A solution of 1 -(bromomethyl)-3,5- dimethylbenzene (2.78 g, 40.00mmol, 1 .1 equiv) in DMF ( 10ml_ ) was added and the reaction mixture stirred for 1 h at room temperature. After consumption of the starting material, the reaction mixture was quenched with ice water, the solid was filtered and dried to give crude product.
  • Step-2 To a stirred solution of 5-bromo-2-(3,5-dimethylbenzyl)-2H-indazole (0.5 g, 1 .60mmol, 1 equiv) in 1 ,4-dioxane (15 mL) was added bis(pinacolato)diboron (0.4 g, 1 .60mmol, 1 equiv), and potassium acetate (0.47 g, 4.80mmol, 3 equiv) and the mixture was degassed for 15 minutes with argon.
  • PdCl 2 (dppf)-CH 2 Cl 2 adduct (0.065 g, 0.08mmol, 0.05 equiv) was added and the reaction was heated to 100 °C for 5h and the consumption of the starting material was monitored by LCMS. The reaction mixture was cooled to room temperature. 5-bromo-7-methyl-7H-pyrrolo[2,3-d]pyrimidin-4-amine (0.363 g, 1 .60mmol, 1 equiv) and saturated aqueous NaHC0 3 (5 mL) were added, and the mixture was degassed for 15 minutes. PdCI 2 (dppf)-CH 2 Cl 2 adduct (0.065 g, 0.08mmol, 0.
  • Step 1 To a stirred solution of 3-fluoro-2-methylaniline (2.0 g, 16.00 mmol, 1 equiv) in acetonitrile (50 ml_) was added N-bromosuccinamide (3.13 g, 17.60mmol, 1 .1 equiv) portionwise at 10°C. The resulting reaction mixture was stirred at room temperature for 30 min. Upon completion of the reaction, sat Na 2 S0 3 (50 ml_) was added slowly into the reaction mixture at 10°C. The organic layer was separated and the aqueous layer was extracted with ethyl acetate. The combined organic layers were dried over Na 2 S0 4 and concentrated in vacuo.
  • Step 2 To a stirred solution of 4-bromo-3-fluoro-2-methylaniline (1 .5 g, 7.352mmol, 1 equiv), in acetic acid (45 mL) was added sodium nitrite (0.81 g, 1 1 .764mmol, 1 .2equiv) at 10°C. The resulting mixture was stirred at room temperature for 4h. Then aqueous sodium hydroxide (50%) was added to the reaction mixture to adjust the pH to 7-8. The mixture was extracted with ethyl aecate. The organic layer was dried over Na 2 S0 4 , filtered and concentrated.
  • Step 3 To a stirred solution of 5-bromo-4-fluoro-2H-indazole (0.65g, 3.023mmol, 1 equiv), in DMF (15 mL) was added 1 -(bromomethyl)-3,5-difluorobenzene (0.75 g, 3.627mmol, 1 .2equiv) at room temperature. The reaction mixture was heated to 100°C and stirred for overnight in a sealed tube. The reaction mixture was cooled to room temperature, quenched with ice water basified with sat NaHC0 3 and extracted with EtOAc. The organic layer was dried over Na 2 S0 4 , and concentrated to obtain crude product.
  • Step 4 A mixture of 5-bromo-2-(3,5-difluorobenzyl)-4-fluoro-2H-indazole (0.3 g, 0.879 mmol), bis(pinacolato)diboron (0.33 g, 1 .319mmol, 1 .5equiv) and potassium acetate (0.21 g, 2.199mmol, 2.5 equiv) in 1 ,4-dioxane (30 mL) in a round bottom flask was bubbled with N 2 for 10 min. PdCI 2 (dppf)-CH 2 Cl2 adduct (0.035 g, 0.043mmol, 0.05equiv) was added and the mixture was heated at 1 10°C overnight.
  • Step 5 A mixture of 2-(3,5-difluorobenzyl)-4-fluoro-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-2H-indazole (0.25 g, 0.644mmol, 1 equiv), 5-bromo-7-cyclopropyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine (0.13 g, 0.515mmol, 0.8equiv), potassium phosphate (0.27 g, 1 .288mmol, 2 equiv) and Pd 2 (dba) 3 (0.029 g, 0.032mmol, 0.05 equiv) in 1 ,4-dioxane: water (10 ml_: 2 mL) in a round bottom flask was bubbled with N 2 for 10 min.
  • Tri-tert- butylphosphoniumtetrafluoroborate (0.018 g, 0.064mmol, 0.1 equiv) was added and heated to 1 10°C & stirred for 2h. The reaction mixture was cooled & filtered through celite and the filtrate was concentrated to obtain crude compound. The crude compound was purified by flash column chromatography using silicagel column.
  • Step 1 To a stirred solution of 3-bromo-2,6-difluorobenzaldehyde (5.0 g, 22.624mmol, 1 equiv) in Diethyl Ether (50 mL) was added 1 .4M methyl magnesium bromide in THF oluene (1 :3) (17.77 mL, 24.886mmol, 1 .l equiv) at 5°C over 5 min. The reaction was maintained at 5°C for 30 min. The reaction was quenched by slow addition of water (50 mL). The reaction mixture was extracted with EtOAc (3 x 50 mL).
  • Step 2 To a stirred solution of 1 -(3-bromo-2,6-difluorophenyl)ethan-1 -ol (5.0 g, 22.421 mmol, 1 equiv) in DCM (200 mL) was added water (0.5 mL) followed by Dess-Martin periodinane (14.2 g, 33.632mmol, 1 .5equiv) in portions over 3 min. The resulting thick white suspension was stirred at room temperature for 2h. The reaction mixture was filtered through a celite pad, the filtrate was washed with 1 M aq NaHC0 3 . The organic layer was dried over Na 2 S0 4 and concentrated to give crude product.
  • Step 3 To a stirred solution of 1 -(3-bromo-2,6-difluorophenyl)ethan-1 -one (3.3 g, 14.932mmol, 1 equiv), in ethyleneglycol (30 mL) was added hydrazine hydrate (1 .2 mL, 17.918mmol, 1 .2equiv) and triethylamine (2.5 mL, 17.918mmol, 1 .2equiv) at room temperature. The reaction mixture was heated at 100°C for18h. The reaction mixture was cooled and checked by TLC and LCMS, it showed uncylized product and desired product as a mixture. The reaction mixture was heated at 150°C for an additional 4 h.
  • Step 4 To a stirred solution of 5-bromo-4-fluoro-3-methyl-2H-indazole (0.6g,2.620mmol, 1 equiv), in DMF (10 mL) was added 1 -(bromomethyl)-3,5-difluorobenzene (0.65 g, 3.144mmol, 1 .2equiv) at room temperature. The reaction mixture was heated to 100°C and stirred for overnight in sealed tube. The reaction mixture was cooled to room temperature, quenched with ice water basified with sat NaHC0 3 and extracted with EtOAc (200mL). The organic layer was dried over Na 2 S0 4 , and concentrated to obtain crude product.
  • Step 5 A mixture of 5-bromo-2-(3,5-difluorobenzyl)-4-fluoro-3-methyl-2H-indazole (0.26 g, 0.732mmol), bis(pinacolato)diboron (0.279 g, 1 .098mmol, 1 .5equiv) and potassium acetate (0.21 g, 2.199mmol, 2.5 equiv) in 1 ,4-dioxane (20 mL) in a round bottom flask was bubbled with N 2 for 10 min. PdCI 2 (dppf)-CH 2 Cl2 adduct (0.029 g, 0.036mmol, 0.05equiv) was added and heated at 1 10°C for overnight.
  • Step 6 A mixture of 2-(3,5-difluorobenzyl)-4-fluoro-3-methyl-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-2H-indazole (0.1 1 g, 0.273mmol, 1 equiv), 5-bromo-7-cyclopropyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine (0.069 g, 0.273mmol, 1 equiv), potassium phosphate (0.1 16 g, 0.547mmol, 2 equiv) and Pd 2 (dba) 3 (0.012 g, 0.013mmol, 0.05 equiv) in 1 ,4- dioxane: water (10 ml_: 2 ml_) in a round bottom flask was bubbled with N 2 for 10 min.
  • Tri- tert-butylphosphoniumtetrafluoroborate (0.0079 g, 0.027mmol, 0.1 equiv) was added and heated to 100°C and stirred for 2h. The reaction mixture was cooled, filtered through celite and the filtrate was concentrated to obtain crude compound. The crude compound was purified by flash column chromatography using silicagel column.
  • Step 1 To a stirred solution of 1 -methylcyclopropane-1 -carboxylic acid (2 g, 19.98 mmol, 1 equiv) in t-butanol (10 ml_) was added DPPA (4.8 ml_, 22.17 mmol, 1 .1 equiv) and Triethyl amine(5.6 ml_ 40.18 mmol, 2 equiv) at room temperature, resulting reaction mixture was stirred at 75°C for 16h. Upon completion of the reaction, the reaction mixture was concentrated under reduced pressure.
  • DPPA 4.8 ml_, 22.17 mmol, 1 .1 equiv
  • Triethyl amine 5.6 ml_ 40.18 mmol, 2 equiv
  • Step 2 To a stirred solution of 2-(4,6-dichloropyrimidin-5-yl)acetaldehyde (2 g , 10.47 mmol, 1 equiv), in Ethanol (20 ml_) was added 1 -methylcyclopropan-1 -amine hydrochloride (1 g , 12.66 mmol, 1 .2 equiv) and Triethyl amine( 1 .5 ml_ 10.76 mmol, 1 .1 equiv) at room temperature, resulting reaction mixture was stirred at 75°C for 16h.
  • Step 4 Aqueous a ' mmonia ( 10 mL) was added to a solution of 5-bromo-4-chloro-7-(1 - methylcyclopropyl)-7H-pyrrolo[2,3-d]pyrimidine (1 .1 g, 3.86 mmol, 1 equiv) in 1 , 4- Dioxane ( 5 mL) and heated to 100°C for 12h. The reaction mixture was cooled to room temperature and concentrated under reduced pressure to give crude compound. To the crude compound, water was added and stirred for 15 min. The precipitated solid was filtered.
  • Step 5 To a stirred solution of 5-bromo-2-(3,5-difluorobenzyl)-4-fluoro-2H-indazole (0.5 g,1 .46 mmol) in 1 ,4-dioxane (50 mL) was added bis(pinacolato)diboron (0.45 g, 1 .77 mmol, 1 .2 equiv), and potassium acetate (0.43 g, 4.39 mmol, 3 equiv). The reaction mixture was degassed with N 2 for 10 min, PdCI 2 (dppf)-CH 2 Cl2 adduct (0.12 g, 0.15 mmol, 0.1 equiv) was added.
  • Step 6 To a stirred solution of 2-(3,5-difluorobenzyl)-4-fluoro-5-(4,4,5,5-tetramethyl-1 ,3,2- dioxaborolan-2-yl)-2H-indazole (0.2 g 0.51 mmol) in 1 ,4-dioxane: water (10 mL: 2 mL) was added, 5-bromo-7-(1 -methylcyclopropyl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (0.16 g, 0.60 mmol, 1 .1 equiv), Tri-tert-butylphosphonium tetrafluoroborate ( 0.024 g, 0.05 mmol, 0.1 equiv), Potassium phosphate tribasic (0.22 g, 1 .03 mmol, 3 equiv), and the mixture was degassed with nitrogen for 10 min.
  • Step 1 To a stirred solution of 5-bromo-7-cyclopropyl-7H-pyrrolo [2, 3-d] pyrimidin-4-amine (3.0 g, 1 1 .85 mmol, 1 equiv) in THF (40 mL) was added Boc anhydride (6.8 mL, 29.6 mmol, 2.5 equiv) followed by DMAP (0.3 g, 2.3 mmol, 0.2 equiv). The reaction mixture was stirred at room temperature for 24h. Solvents were completely evaporated and the crude was extracted with ethyl acetate.
  • Step 2 To a stirred solution of N,N-Di(tert-butoxycarbonyl)5-bromo-7-cyclopropyl-7H- pyrrolo[2,3-d]pyrimidin-4-amine (4.0g, 8.83 mmol, 1 equiv), in 1 ,4-Dioxane (40 mL) was added 4,4,5,5-tetramethyl-1 ,3,2-dioxaborolane (5.1 mL, 35.30mmol, 4 equiv) and triethylamine (5 mL, 35.30mmol, 4 equiv). The reaction mixture was degassed for 5 minutes.
  • Step 3 To a stirred solution of 5-bromo-4-fluoro-2H-indazole (0.3g ,1 .395 mmol, 1 equiv), in DMF (10 mL) was added 1 -(bromomethyl)-3,5-dimethylbenzene (0.33 g, 1 .674mmol, 1 .2 equiv) at room temperature and the mixture was heated at 100°C overnight in a sealed tube. The reaction mixture was cooled to room temperature, quenched with ice water and basified with saturated NaHC03 and extracted with EtOAc. The organic layer was separated, dried over Na 2 S0 4 , and concentrated to give crude product.
  • Step 4 A mixture of 5-bromo-2-(3,5-dimethylbenzyl)-4-fluoro-2H-indazole(0.18 g, 0.5405mmol, 1 equiv), N,N-Di(tert-butoxycarbonyl)7-cyclopropyl-5-(4,4,5,5-tetramethyl- 1 ,3,2-dioxaborolan-2-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine (0.243 g, 0.4864mmol, 0.9equiv), potassium phosphate (0.229 g, 1 .081 mmol, 2 equiv) and Pd 2 (dba) 3 (0.024 g, 0.027mmol, 0.05 equiv) in 1 ,4-dioxane: water (10 mL: 2 mL) in a round bottom flask was bubbled with N 2 for 10 min.
  • Tri-tert-butylphosphoniumtetrafluoroborate (0.015 g, 0.054mmol, 0.1 equiv) was added and the mixture heated to 1 10°C for 2h. The reaction mixture was cooled & filtered through celite and the filtrate was concentrated to obtain crude compound. The crude compound was purified by flash column chromatography using silicagel column, and the compound was eluted at 2% MeOH : DCM.
  • Step 5 To a stirred solution of N,N-Di(tert-butoxycarbonyl)7-cyclopropyl-5-(2-(3,5- dimethylbenzyl)-4-fluoro-2H-indazol-5-yl)-7H-pyrrolo[2,3-d]pyrimidin-4-amine in DCM was added TFA at 0°C and the mixture waa stirred at RT for 2 h. The excess of TFA and DCM was concentrated, then diluted with saturated NaHC0 3 and DCM. The organic layer was separated dried and concentrated.
  • An oral dosage form for administering the present invention is produced by filling a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table 2, below.
  • An injectable form for administering the present invention is produced by stirring 1 .7% by weight of 7-cyclopropyl-5-(2-(3,5-difluorobenzyl)-4-fluoro-2H-indazol-5-yl)-7H- pyrrolo[2,3-d]pyrimidin-4-amine (Compound of Example 2) in 10% by volume propylene glycol in water.
  • sucrose, calcium sulfate dihydrate and a PERK inhibitor as shown in Table 3 below are mixed and granulated in the proportions shown 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.
  • Examples 1 to 15 were tested generally according to the above PERK enzyme assay and in at least one experimental run exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value ⁇ 5.8 and ⁇ 8.3 against PERK.
  • Example 1 The compound of Example 1 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 6.3 against PERK.
  • Example 2 The compound of Example 2 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 8.2 against PERK.
  • Example 3 The compound of Example 3 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 8.2 against PERK.
  • Example 4 The compound of Example 4 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 7 against PERK.
  • the compound of Example 8 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 6.7 against PERK.
  • the compound of Example 13 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 8 against PERK.
  • Example 14 The compound of Example 14 was tested generally according to the above PERK enzyme assay and in at least one set of experimental runs exhibited an average PERK Enzyme (500 ⁇ ATP) plC50 value of 7.2 against PERK.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Hospice & Palliative Care (AREA)
  • Psychiatry (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

L'invention concerne des dérivés d'indazole substitués et leurs utilisations. L'invention concerne particulièrement des composés de formule I et l'utilisation de composés de formule (I) dans le traitement d'états pathologiques : (I) où R1, R2, R3, R4 et R5 sont tels que définis ici; ou un sel de ceux-ci comprenant un sel pharmaceutiquement acceptable de ceux-ci. Les composés de l'invention sont des inhibiteurs de PERK et peuvent être utiles dans le traitement du cancer, des syndromes précancéreux et de maladies associées à des voies de réponse des protéines dépliées activées, telles que la maladie d'Alzheimer, une lésion de la moelle épinière, une lésion cérébrale traumatique, un accident ischémique cérébral, un accident vasculaire cérébral, la maladie de Parkinson, le diabète, le syndrome métabolique, les troubles métaboliques, la maladie de Huntington, la maladie de Creutzfeldt-Jakob, et les maladies à prions apparentées, l'insomnie fatale familiale, le syndrome de Gerstmann-Sträussler-Scheinker, et les maladies à prions apparentées, la sclérose latérale amyotrophique, la paralysie supranucléaire progressive, l'infarctus du myocarde, les maladies cardiovasculaires, l'inflammation, la fibrose d'organes, les maladies chroniques et aiguës du foie, la stéatose hépatique, la fibrose du foie, les maladies chroniques et aiguës du poumon, la fibrose pulmonaire, les maladies chroniques et aiguës du rein, la fibrose rénale, l'encéphalopathie traumatique chronique (ETC), la neurodégénérescence, les démences, les démences fronto-temporales, les tauopathies, la maladie de Pick, la maladie de Pick-Neimann, l'amyloïdose, une déficience cognitive, l'athérosclérose, les maladies oculaires, les troubles neurologiques, la douleur, les arythmies, dans une transplantation d'organe et dans le transport d'organes en vue d'une transplantation. Par conséquent, l'invention concerne en outre des compositions pharmaceutiques comprenant un composé de l'invention. L'invention concerne par ailleurs des procédés d'inhibition de l'activité de PERK et le traitement de troubles associés au moyen d'un composé de l'invention ou d'une composition pharmaceutique comprenant un composé de l'invention.
PCT/IB2018/055548 2017-07-27 2018-07-25 Dérivés d'indazole utiles en tant qu'inhibiteurs de perk WO2019021208A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
ININ201711026720 2017-07-27
IN201711026720 2017-07-27

Publications (1)

Publication Number Publication Date
WO2019021208A1 true WO2019021208A1 (fr) 2019-01-31

Family

ID=63371736

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2018/055548 WO2019021208A1 (fr) 2017-07-27 2018-07-25 Dérivés d'indazole utiles en tant qu'inhibiteurs de perk

Country Status (1)

Country Link
WO (1) WO2019021208A1 (fr)

Citations (49)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US6113918A (en) 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US6268391B1 (en) 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
EP1125585A1 (fr) 1999-08-30 2001-08-22 Japan Tobacco Inc. Traitements de maladies immunitaires
WO2001090129A2 (fr) 2000-05-19 2001-11-29 Corixa Corporation Traitement prophylactique et therapeutique de maladies infectieuses et autres avec des composes a base de mono- et disaccharides
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
US6525028B1 (en) 2002-02-04 2003-02-25 Corixa Corporation Immunoeffector compounds
EP1374901A1 (fr) 2001-03-01 2004-01-02 Japan Tobacco, Inc. Dispositifs de rejet du greffon
EP1374902A1 (fr) 2001-03-27 2004-01-02 Japan Tobacco Inc. Remedes contre les affections intestinales inflammatoires
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
US6911434B2 (en) 2002-02-04 2005-06-28 Corixa Corporation Prophylactic and therapeutic treatment of infectious and other diseases with immunoeffector compounds
US6984720B1 (en) 1999-08-24 2006-01-10 Medarex, Inc. Human CTLA-4 antibodies
WO2006016997A2 (fr) 2004-07-08 2006-02-16 Corixa Corporation Composes d'aminoalkylglucosaminide phosphate et leur utilisation
WO2006081230A2 (fr) * 2005-01-26 2006-08-03 Schering Corporation Inhibiteurs de kinase
US7129219B2 (en) 2000-08-04 2006-10-31 Corixa Corporation Immunoeffector compounds
WO2007056170A2 (fr) * 2005-11-02 2007-05-18 Bayer Healthcare Ag Derives pyrrolo-[2,1-f]-[1,2,4]-triazin-4-ylamines inhibiteurs de la igf-1r kinase pour le traitement des cancers et autres maladies hyperproliferatives
WO2007104783A2 (fr) * 2006-03-15 2007-09-20 Ortho-Mcneil-Janssen Pharmaceuticals, Inc. Dérivés de 3-cyanopyridone 1,4-disubstitués et leur emploi en tant que modulateurs allostériques positifs des récepteurs mglur2
WO2008137915A2 (fr) 2007-05-07 2008-11-13 Medimmune, Llc Anticorps anti-icos et leur utilisation en traitement oncologique, de transplantation et maladie auto-immune
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
US7504101B2 (en) 1998-02-24 2009-03-17 Sisters Of Providence In Oregon Methods for enhancing antigen-specific immune response using antibodies that bind OX-40
US7550140B2 (en) 2002-06-13 2009-06-23 Crucell Holland B.V. Antibody to the human OX40 receptor
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
WO2010056804A1 (fr) 2008-11-12 2010-05-20 Medimmune, Llc Formulation d’anticorps
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US7758852B2 (en) 2002-04-03 2010-07-20 Merck Serono Sa OX40R binding agents
WO2010130422A1 (fr) * 2009-05-12 2010-11-18 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Dérivés de la 1,2,4-triazolo [4,3-a] pyridine et leur utilisation en tant que modulateurs allostériques positifs des récepteurs mglur2
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
US7960515B2 (en) 2007-12-14 2011-06-14 Bristol-Myers Squibb Company Binding molecules to the human OX40 receptor
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
WO2011119663A1 (fr) * 2010-03-25 2011-09-29 Glaxosmithkline Llc Composés chimiques
US8034953B2 (en) 2005-05-10 2011-10-11 Incyte Corporation Modulators of indoleamine 2,3-dioxygenase and methods of using the same
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
WO2012027328A2 (fr) 2010-08-23 2012-03-01 Board Of Regents, The University Of Texas System Anticorps anti-ox40 et leurs procédés d'utilisation
US8168757B2 (en) 2008-03-12 2012-05-01 Merck Sharp & Dohme Corp. PD-1 binding proteins
WO2012131004A2 (fr) 2011-03-31 2012-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps dirigés contre icos et utilisation de ceux-ci
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
WO2013028231A1 (fr) 2011-08-23 2013-02-28 Board Of Regents, The University Of Texas System Anticorps anti-ox40 et leurs procédés d'utilisation
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
US20150284383A1 (en) * 2014-04-07 2015-10-08 Purdue Pharma L.P. Indole derivatives and use thereof
WO2017046737A1 (fr) 2015-09-15 2017-03-23 Glaxosmithkline Intellectual Property (No.2) Limited Dérivés de 1-phénylpyrrolidin-2-one comme inhibiteurs de perk

Patent Citations (60)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5877219A (en) 1994-04-25 1999-03-02 Glaxo Wellcomeinc. Non-steroidal ligands for the estrogen receptor
US6207716B1 (en) 1994-04-25 2001-03-27 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
US6113918A (en) 1997-05-08 2000-09-05 Ribi Immunochem Research, Inc. Aminoalkyl glucosamine phosphate compounds and their use as adjuvants and immunoeffectors
US6268391B1 (en) 1997-08-06 2001-07-31 Glaxo Wellcome Inc. Benzylidene-1,3-dihydro-indol-2-one derivatives a receptor tyrosine kinase inhibitors, particularly of Raf kinases
US7504101B2 (en) 1998-02-24 2009-03-17 Sisters Of Providence In Oregon Methods for enhancing antigen-specific immune response using antibodies that bind OX-40
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
EP1125585A1 (fr) 1999-08-30 2001-08-22 Japan Tobacco Inc. Traitements de maladies immunitaires
WO2001090129A2 (fr) 2000-05-19 2001-11-29 Corixa Corporation Traitement prophylactique et therapeutique de maladies infectieuses et autres avec des composes a base de mono- et disaccharides
US7129219B2 (en) 2000-08-04 2006-10-31 Corixa Corporation Immunoeffector compounds
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
EP1374901A1 (fr) 2001-03-01 2004-01-02 Japan Tobacco, Inc. Dispositifs de rejet du greffon
EP1374902A1 (fr) 2001-03-27 2004-01-02 Japan Tobacco Inc. Remedes contre les affections intestinales inflammatoires
US6911434B2 (en) 2002-02-04 2005-06-28 Corixa Corporation Prophylactic and therapeutic treatment of infectious and other diseases with immunoeffector compounds
US6525028B1 (en) 2002-02-04 2003-02-25 Corixa Corporation Immunoeffector compounds
US7858765B2 (en) 2002-04-03 2010-12-28 Merck Serono Sa OX40R binding agents
US7758852B2 (en) 2002-04-03 2010-07-20 Merck Serono Sa OX40R binding agents
US7550140B2 (en) 2002-06-13 2009-06-23 Crucell Holland B.V. Antibody to the human OX40 receptor
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
US7521051B2 (en) 2002-12-23 2009-04-21 Medimmune Limited Methods of upmodulating adaptive immune response using anti-PD-1 antibodies
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
WO2004072286A1 (fr) 2003-01-23 2004-08-26 Ono Pharmaceutical Co., Ltd. Substance specifique a pd-1 humain
WO2006016997A2 (fr) 2004-07-08 2006-02-16 Corixa Corporation Composes d'aminoalkylglucosaminide phosphate et leur utilisation
WO2006081230A2 (fr) * 2005-01-26 2006-08-03 Schering Corporation Inhibiteurs de kinase
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
US8034953B2 (en) 2005-05-10 2011-10-11 Incyte Corporation Modulators of indoleamine 2,3-dioxygenase and methods of using the same
US8383796B2 (en) 2005-07-01 2013-02-26 Medarex, Inc. Nucleic acids encoding monoclonal antibodies to programmed death ligand 1 (PD-L1)
US7943743B2 (en) 2005-07-01 2011-05-17 Medarex, Inc. Human monoclonal antibodies to programmed death ligand 1 (PD-L1)
WO2007056170A2 (fr) * 2005-11-02 2007-05-18 Bayer Healthcare Ag Derives pyrrolo-[2,1-f]-[1,2,4]-triazin-4-ylamines inhibiteurs de la igf-1r kinase pour le traitement des cancers et autres maladies hyperproliferatives
WO2007104783A2 (fr) * 2006-03-15 2007-09-20 Ortho-Mcneil-Janssen Pharmaceuticals, Inc. Dérivés de 3-cyanopyridone 1,4-disubstitués et leur emploi en tant que modulateurs allostériques positifs des récepteurs mglur2
WO2008137915A2 (fr) 2007-05-07 2008-11-13 Medimmune, Llc Anticorps anti-icos et leur utilisation en traitement oncologique, de transplantation et maladie auto-immune
US8354509B2 (en) 2007-06-18 2013-01-15 Msd Oss B.V. Antibodies to human programmed death receptor PD-1
WO2008156712A1 (fr) 2007-06-18 2008-12-24 N. V. Organon Anticorps dirigés contre le récepteur humain de mort programmée pd-1
US7960515B2 (en) 2007-12-14 2011-06-14 Bristol-Myers Squibb Company Binding molecules to the human OX40 receptor
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
US8552154B2 (en) 2008-09-26 2013-10-08 Emory University Anti-PD-L1 antibodies and uses therefor
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
WO2010056804A1 (fr) 2008-11-12 2010-05-20 Medimmune, Llc Formulation d’anticorps
WO2010077634A1 (fr) 2008-12-09 2010-07-08 Genentech, Inc. Anticorps anti-pd-l1 et leur utilisation pour améliorer la fonction des lymphocytes t
US8217149B2 (en) 2008-12-09 2012-07-10 Genentech, Inc. Anti-PD-L1 antibodies, compositions and articles of manufacture
WO2010130422A1 (fr) * 2009-05-12 2010-11-18 Ortho-Mcneil-Janssen Pharmaceuticals, Inc Dérivés de la 1,2,4-triazolo [4,3-a] pyridine et leur utilisation en tant que modulateurs allostériques positifs des récepteurs mglur2
WO2011066342A2 (fr) 2009-11-24 2011-06-03 Amplimmune, Inc. Inhibition simultanée de pd-l1/pd-l2
US20130034559A1 (en) 2009-11-24 2013-02-07 Medlmmune Limited Targeted Binding Agents Against B7-H1
WO2011119663A1 (fr) * 2010-03-25 2011-09-29 Glaxosmithkline Llc Composés chimiques
US20110280877A1 (en) 2010-05-11 2011-11-17 Koji Tamada Inhibition of B7-H1/CD80 interaction and uses thereof
WO2012027328A2 (fr) 2010-08-23 2012-03-01 Board Of Regents, The University Of Texas System Anticorps anti-ox40 et leurs procédés d'utilisation
WO2012131004A2 (fr) 2011-03-31 2012-10-04 INSERM (Institut National de la Santé et de la Recherche Médicale) Anticorps dirigés contre icos et utilisation de ceux-ci
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
WO2013028231A1 (fr) 2011-08-23 2013-02-28 Board Of Regents, The University Of Texas System Anticorps anti-ox40 et leurs procédés d'utilisation
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
US20150284383A1 (en) * 2014-04-07 2015-10-08 Purdue Pharma L.P. Indole derivatives and use thereof
WO2017046737A1 (fr) 2015-09-15 2017-03-23 Glaxosmithkline Intellectual Property (No.2) Limited Dérivés de 1-phénylpyrrolidin-2-one comme inhibiteurs de perk

Non-Patent Citations (105)

* Cited by examiner, † Cited by third party
Title
"Cancer Principles and Practice of Oncology", 15 February 2001, LIPPINCOTT WILLIAMS & WILKINS PUBLISHERS
"NCBI", Database accession no. NP _079515
"NCBI", Database accession no. NP_005009
"NCBI", Database accession no. NP_054862
"Tyrosine Kinase Signalling in Breast cancer:erbB Family Receptor Tyrosine Kinases", BREAST CANCER RES., vol. 2, no. 3, 2000, pages 176 - 183
ABRAHAM, R.T., CURRENT OPINION IN IMMUNOLOGY, vol. 8, no. 3, 1996, pages 412 - 8
AMERI, K.; LEWIS, C. E.; RAIDA, M.; SOWTER, H.; HAI, T.; HARRIS, A. L., BLOOD, vol. 103, 2004, pages 1876 - 82
ASHBY, M.N., CURRENT OPINION IN LIPIDOLOGY, vol. 9, no. 2, 1998, pages 99 - 102
ATKINS, C.; LIU, Q.; MINTHORN, E.; ZHANG, S.; FIGUEROA, D.J.; MOSS, K.; STANLEY, T.B.; SANDERS, B.; GOETZ, A.; GAUL, N., CANCER RES., vol. 74, 2013, pages 1993 - 2002
AXTEN ET AL., J. MED. CHEM., vol. 55, 2012, pages 7193 - 7207
AXTEN, J. M.; MEDINA, J. R.; FENG, Y.; SHU, A.; ROMERIL, S.P.; GRANT, S.W.; LI, W.H.H.; HEERDING, D.A.; MINTHORN, E.; MENCKEN, T., J. MED. CHEM., vol. 55, 2012, pages 7193 - 7207
BI, M.; NACZKI, C.; KORITZINSKY, M.; FELS, D.; BLAIS, J.; HU, N.; HARDING, H.; NOVOA, I.; VARIA, M.; RALEIGH, J. ET AL., EMBO J, vol. 24, 2005, pages 3470 - 81
BIOCHIM. BIOPHYS. ACTA, vol. 1423, no. 3, pages 19 - 30
BLAIS, J.; BELL, J. C., CELL CYCLE, vol. 5, 2006, pages 2874 - 7
BOLEN, J.B.; BRUGGE, J.S., ANNUAL REVIEW OF IMMUNOLOGY, vol. 15, 1997, pages 371 - 404
BREKKEN, R.A. ET AL.: "Selective Inhibition of VEGFR2 Activity by a monoclonal Anti-VEGF antibody blocks tumor growth in mice", CANCER RES., vol. 60, 2000, pages 5117 - 5124, XP002340113
BRODT, P; SAMANI, A.; NAVAB, R., BIOCHEMICAL PHARMACOLOGY, vol. 60, 2000, pages 1101 - 1107
CANMAN, C.E.; LIM, D.S., ONCOGENE, vol. 17, no. 25, 1998, pages 3301 - 3308
CIVELEK, M. ET AL., CIRC RES, vol. 105, no. 5, 2009, pages 453 - 461
CUNARD, R, J. CLIN. MED., vol. 4, 2015, pages 715 - 740
CYBULSKY, A. V. ET AL., J BIOL CHEM, vol. 280, no. 26, 2005, pages 24396 - 24403
DAVIES, M. P.; BARRACLOUGH, D. L.; STEWART, C.; JOYCE, K. A.; ECCLES, R. M.; BARRACLOUGH, R.; RUDLAND, P. S.; SIBSON, D. R., INT J CANCER, vol. 123, 2008, pages 85 - 8
DICKHOUT, J.G.; CARLISLE, R.E.; AUSTIN, R.C., CIRC RES., vol. 108, 2011, pages 629 - 642
DOYLE, K.M.; KENNEDY, D.; GORMAN, A.M.; GUPTA, S.; HEALY, S.J.M.; SAMALI, A., J. CELL. MOL. MED., vol. 15, 2011, pages 2025 - 2039
EINZIG, PROC. AM. SOC. CLIN. ONCOL., vol. 20, pages 46
FELDMAN, D. E.; CHAUHAN, V.; KOONG, A. C., MOL CANCER RES, vol. 3, 2005, pages 597 - 605
FORASTIRE, SEM. ONCOL., vol. 20, 1990, pages 56
GREEN, M.C. ET AL.: "Monoclonal Antibody Therapy for Solid Tumors", CANCER TREAT. REV., vol. 26, no. 4, 2000, pages 269 - 286, XP009019784, DOI: doi:10.1053/ctrv.2000.0176
GUPTA, S.; MCGRATH, B.; CAVENER, D. R., PLOS ONE, vol. 4, 2009, pages e8008
HAMANAKA, R. B.; BENNETT, B. S.; CULLINAN, S. B.; DIEHL, J. A., MOL BIOL CELL, vol. 16, 2005, pages 5493 - 501
HARDING, H. P.; ZENG, H.; ZHANG, Y.; JUNGRIES, R.; CHUNG, P.; PLESKEN, H.; SABATINI, D. D.; RON, D., MOL CELL, vol. 7, 2001, pages 1153 - 63
HARDING, H.P.; CALFON, M.; URANO, F.; NOVOA, I.; RON, D., ANNU. REV. CELL DEV. BIOL., vol. 18, 2002, pages 575 - 599
HARDING, H.P.; ZHANG, Y.; RON, D., NATURE, vol. 397, 1999, pages 271 - 274
HETZ, C., NAT. REV. MOL. CELL BIO., vol. 13, 2012, pages 89 - 102
HO, Y.; YANG, X.; LAU, J.C; HUNG, C.H.; WUWONGSE, S; ZHANG, Q.; WANG, J.; BAUM, L.; SO, K.; CHANG, R.C., J. ALZHEIMER'S DISEASE, vol. 28, 2012, pages 839 - 854
HOLMES ET AL., J. NAT. CANCER INST., vol. 83, 1991, pages 1797
HOOZEMANS. J.J.M.; VAN HAASTERT, E.S.; NIJHOLT, D.A.T.; ROZEMULLER, A.J.M.; EIKELENBOOM, P.; SCHEPER, W., HIPPOCAMPUS AM. J. PATH., vol. 174, 2009, pages 1241 - 1251
IGNOFF, R.J., CANCER CHEMOTHERAPY POCKET GUIDES, 1998
INAGI, R.; ISHIMOTO, Y; NANGAKU, M., NAT. REV. NEPHROL., vol. 10, 2014, pages 369 - 378
J. MED. CHEM., vol. 55, no. 16, 2012, pages 7193 - 7207
J. MED. CHEM., vol. 58, no. 3, 2015, pages 1426 - 1441
JACKSON, R.J.; HELLEN, C.U.T.; PESTOVA, T.V., NAT. REV. MOL. CELL BIOL., vol. 10, 2010, pages 113 - 127
JACKSON, S.P., INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND CELL BIOLOGY, vol. 29, no. 7, 1997, pages 935 - 8
JORGENSEN, E.; STINSON, A.; SHAN, L.; YANG, J.; GIETL, D.; ALBINO, A. P., BMC CANCER, vol. 8, 2008, pages 229
JULIER, C.; NICOLINO, M., J. RARE DISEASES, vol. 5, no. 29, 2010, pages 1 - 13, Retrieved from the Internet <URL:http://www.ojrd.com/content/5/1/29>
KANEKURA, K.; SUZUKI, H.; AISO, S.; MATSUOKA, M., MOLECULAR NEUROBIOLOGY, vol. 39, no. 2, 2009, pages 81 - 89
KATH, JOHN C., EXP. OPIN. THER. PATENTS, vol. 10, no. 6, 2000, pages 803 - 818
KEARNS, C.M., SEMINARS IN ONCOLOGY, vol. 3, no. 6, 1995, pages 16 - 23
KOUMENIS, C.; NACZKI, C.; KORITZINSKY, M.; RASTANI, S.; DIEHL, A.; SONENBERG, N.; KOROMILAS, A.; WOUTERS, B. G., MOL CELL BIOL, vol. 22, 2002, pages 7405 - 16
KOUMENIS, C.; WOUTERS, B. G., MOL CANCER RES, vol. 4, 2006, pages 423 - 36
LACKEY, K. ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, vol. 10, 2000, pages 223 - 226
LEMOS H ET AL., CANCER RES., vol. 76, no. 8, 15 April 2016 (2016-04-15), pages 2076 - 81
LIDA, K.; LI, Y.; MCGRATH, B. C.; FRANK, A.; CAVENER, D. R., BMC CELL BIOL, vol. 8, 2007, pages 38
LIU, M.; S. C. DUDLEY, JR., INT J MOL SCI, vol. 17, no. 1, 2016, pages E52
LOFTS, F. J. ET AL.: "New Molecular Targets for Cancer Chemotherapy", 1994, CRC PRESS, article "Growth factor receptors as targets"
LUCKE-WOLD, B. P. ET AL., J NEUROSURA, vol. 124, no. 3, 2016, pages 687 - 702
MA, K.; VATTEM, K. M.; WEK, R. C., J BIOL CHEM, vol. 277, 2002, pages 18728 - 35
MA, Y.; HENDERSHOT, L. M., NAT REV CANCER, vol. 4, 2004, pages 966 - 77
MA, Y.; LU, Y.; ZENG, H.; RON, D.; MO, W.; NEUBERT, T. A., RAPID COMMUN MASS SPECTROM, vol. 15, 2001, pages 1693 - 700
MARCINIAK, S. J.; GARCIA-BONILLA, L.; HU, J.; HARDING, H. P.; RON, D., J CELL BIOL, vol. 172, 2006, pages 201 - 9
MARKMAN ET AL., YALE JOURNAL OF BIOLOGY AND MEDICINE, vol. 64, 1991, pages 583
MARTINEZ-LACACI, L. ET AL., INT. J. CANCER, vol. 88, no. 1, 2000, pages 44 - 52
MASSAGUE, J.; WEIS-GARCIA, F., CANCER SURVEYS, vol. 27, 1996, pages 41 - 64
MCALPINE, C.S.; BOWES, A.J.; WERSTUCK, G.H., CARDIOVASCULAR & HEMATOLOGICAL DISORDERS: DRUG TARGETS, vol. 10, no. 2, 2010, pages 151 - 157
MCGUIRE ET AL., ANN. INTEM, MED., vol. 111, 1989, pages 273
MORENO, J. A.; HALLIDAY, M.; MOLLOY, C.; RADFORD, H.; VERITY, N.; AXTEN, J. M.; ORTORI, C. A.; WILLIS, A. E.; FISCHER, P. M.; BARR, SCIENCE TRANS. MED, vol. 5, 2013, pages 206ra138
MORENO, J.A; RADFORD, H.; PERETTI, D.; STEINERT, J.R.; VERITY, N.; MARTIN, M.G.; HALLIDAY, M.; MORGAN, J.; DINSDALE, D.; ORTORI, C, NATURE, vol. 485, 2012, pages 507 - 512
MUNN DH, TRENDS IMMUNOL., vol. 37, no. 3, March 2016 (2016-03-01), pages 193 - 207
NASSIF, M.; MATUS, S.; CASTILLO, K.; HETZ, C., AMYOTROPHIC LATERAL SCLEROSIS PATHOGENESIS: A JOURNEY THROUGH THE SECRETORY PATHWAY ANTIOXIDANTS & REDOX SIGNALING, vol. 13, no. 12, 2010, pages 1955 - 1989
NATURE REVIEWS DRUG DISCOVERY, vol. 4, September 2005 (2005-09-01), pages 711 - 712
NIJHOLT, D. A. ET AL., J PATHOL, vol. 226, no. 5, 2012, pages 693 - 702
O'CONNOR, T.; SADLEIR, K.R.; MAUS, E.; VELLIQUETTE, R. A.; ZHAO, J.; COLE, S. L.; EIMER, W. A.; HITT, B.; BEMBINSTER, L. A.; LAMMI, NEURON, vol. 60, no. 6, 2008, pages 988 - 1009
OHRI, S.S.; MADDIE, M.A.; ZHAO, Y.; QIU, M.S.; HETMAN, M.; WHITTEMORE, S.R., GLIA, vol. 59, 2011, pages 1489 - 1502
PASCHEN, W., CURRENT NEUROVASCULAR RESEARCH, vol. 1, no. 2, 2004, pages 173 - 181
PHILIP, P.A.; HARRIS, A.L., CANCER TREATMENT AND RESEARCH, vol. 78, 1995, pages 3 - 27
POWIS, G.; KOZIKOWSKI A.: "New Molecular Targets for Cancer Chemotherapy", 1994, CRC PRESS
PRUSINER, S.B., SCIENCE, vol. 336, 2012, pages 1511 - 1513
ROMERO-RAMIREZ, L.; CAO, H.; NELSON, D.; HAMMOND, E.; LEE, A. H.; YOSHIDA, H.; MORI, K.; GLIMCHER, L. H.; DENKO, N. C.; GIACCIA, A, CANCER RES, vol. 64, 2004, pages 5943 - 7
RON, D., J. CLIN. INVEST., vol. 110, 2002, pages 1383 - 1388
ROSANIA ET AL., EXP. OPIN. THER. PATENTS, vol. 10, no. 2, 2000, pages 215 - 230
ROUSCHOP, K. M.; VAN DEN BEUCKEN, T.; DUBOIS, L.; NIESSEN, H.; BUSSINK, J.; SAVELKOULS, K.; KEULERS, T.; MUJCIC, H.; LANDUYT, W.;, J CLIN INVEST, vol. 120, 2010, pages 127 - 41
SALMINEN, A.; KAUPPINEN, A.; SUURONEN, T.; KAARNIRANTA, K.; OJALA, J., JOURNAL OF NEUROINFLAMMATION, vol. 6, 2009, pages 41
SCHAROVSKY, O.G.; ROZADOS, V.R.; GERVASONI, S.I.; MATAR, P., JOURNAL OF BIOMEDICAL SCIENCE, vol. 7, no. 4, 2000, pages 292 - 8
SHAWVER ET AL., DDT, vol. 2, 2 February 1997 (1997-02-02)
SHI, Y.; VATTEM, K. M.; SOOD, R.; AN, J.; LIANG, J.; STRAMM, L.; WEK, R. C., MOL CELL BIOL, vol. 18, 1998, pages 7499 - 509
SINH, S.; COREY, S.J., JOURNAL OF HEMATOTHERAPY AND STEM CELL RESEARCH, vol. 8, no. 5, 1999, pages 465 - 80
SMITHGALL, T.E., JOURNAL OF PHARMACOLOGICAL AND TOXICOLOGICAL METHODS, vol. 34, no. 3, 1995, pages 125 - 32
SOOD, R.; PORTER, A. C.; MA, K.; QUILLIAM, L. A.; WEK, R. C., BIOCHEM J, vol. 346, 2000, pages 281 - 93
STUTZBACH, L.D.; XIE, S.X.; NAJ, A.C.; ALBIN, R.; GILMAN, S.; LEE, V.M.Y; TROJANOWSKI, J.Q.; DEVLIN, B.; SCHELLENBERG, G.D., ACTA NEUROPATH. COMM., vol. 1, 2013, pages 31, Retrieved from the Internet <URL:http://www.actaneurocomms.org/content/1/1/31>
SU, Q.; WANG, S.; GAO, H. Q.; KAZEMI, S.; HARDING, H. P.; RON, D.; KOROMILAS, A. E., J BIOL CHEM, vol. 283, 2008, pages 469 - 75
T. GREENE; P. WUTS: "Protecting Groups in Organic Synthesis", 2006, JOHN WILEY & SONS
TABAS, I.; SEIMON, T.; TIMMINS, J.; LI, G.; LIM, W.: "Macrophage apoptosis in advanced atherosclerosis Annals of the New York", vol. 1173, 2009, ACADEMY OF SCIENCES, pages: E40 - E45
TAJIRI, S.; OYADOMARI, S.; YANO, S.; MORIOKA, M.; GOTOH, T.; HAMADA, J.I; USHIO, Y.; MORI, M., CELL DEATH AND DIFF., vol. 11, 2004, pages 403 - 415
UNTERBERGER, U.; HOFTBERGER, R.; GELPI, E.; FLICKER, H.; BUDKA, H.; VOIGTLANDER, T., J. NEUROPATHOL. EXP. NEUROL., vol. 65, 2006, pages 348 - 357
VAN GALEN, P. ET AL., NATURE, vol. 510, no. 7504, 2014, pages 268 - 272
WALTER, P.; RON, D., SCIENCE, vol. 334, 2011, pages 1081 - 1086
WEK, R. C.; D. R. CAVENER: "Translational control and the unfolded protein response", ANTIOXID REDOX SIGNAL, vol. 9, no. 12, 2007, pages 2357 - 2371
WHO DRUG INFORMATION, vol. 27, no. 1, 2013, pages 68 - 69
WHO DRUG INFORMATION, vol. 27, no. 2, 2013, pages 161 - 162
WOEHLBIER, U.; HETZ, C., TRENDS BIOCHEM. SCIENCES, vol. 36, pages 329 - 337
WOO, NATURE, vol. 368, 1994, pages 750
YAMAMOTO, T.; TAYA, S.; KAIBUCHI, K., JOURNAL OF BIOCHEMISTRY, vol. 126, no. 5, 1999, pages 799 - 803
ZHANG, K.; R. J. KAUFMAN, NEUROLOGY, vol. 66, no. 2, 2006, pages 102 - 109
ZHANG, W.; FENG, D.; LI, Y.; LIDA, K.; MCGRATH, B.; CAVENER, D. R., CELL METAB, vol. 4, 2006, pages 491 - 7
ZHONG, H. ET AL., CANCER RES, vol. 60, no. 6, 2000, pages 1541 - 1545

Similar Documents

Publication Publication Date Title
JP7119158B2 (ja) タンパク質調節因子として有用な複素環式アミド
JP7291130B2 (ja) インターフェロン遺伝子の刺激物質(sting)の調節物質
EP3487503A1 (fr) Dérivés d&#39;isoquinoléine utilisés comme inhibiteurs de perk
KR101676077B1 (ko) 화합물
JP2020536106A (ja) Hivの処置に有用なインターフェロン遺伝子の刺激物質(sting)の調節物質
US8598156B2 (en) Chemical compounds
EP2477987A1 (fr) Modulateurs des récepteurs de type toll (tlr)
KR20230022402A (ko) PI3Kα 억제제 및 이의 사용 방법
WO2015056180A1 (fr) Dérivés d&#39;indoline utilisés comme inhibiteurs de perk
JP2019508368A (ja) コルチスタチン類縁体及びその使用
WO2017046739A1 (fr) Dérivés d&#39;imidazolidinone comme inhibiteurs de perk
WO2019053617A1 (fr) Composés chimiques
JP2023110094A (ja) Tcr-nck相互作用の阻害剤としてのクロメン誘導体
WO2019021208A1 (fr) Dérivés d&#39;indazole utiles en tant qu&#39;inhibiteurs de perk
EA037671B1 (ru) Гетероциклические амиды, пригодные в качестве модуляторов белков

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: 18759738

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: 18759738

Country of ref document: EP

Kind code of ref document: A1