WO2015056180A1 - Dérivés d'indoline utilisés comme inhibiteurs de perk - Google Patents

Dérivés d'indoline utilisés comme inhibiteurs de perk Download PDF

Info

Publication number
WO2015056180A1
WO2015056180A1 PCT/IB2014/065310 IB2014065310W WO2015056180A1 WO 2015056180 A1 WO2015056180 A1 WO 2015056180A1 IB 2014065310 W IB2014065310 W IB 2014065310W WO 2015056180 A1 WO2015056180 A1 WO 2015056180A1
Authority
WO
WIPO (PCT)
Prior art keywords
cancer
4alkyl
pyrazole
disease
methyl
Prior art date
Application number
PCT/IB2014/065310
Other languages
English (en)
Other versions
WO2015056180A8 (fr
Inventor
Jesus Raul Medina
Adam Kenneth Charnley
Stuart Paul Romeril
Original Assignee
Glaxosmithkline Intellectual Property (No.2) 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 (No.2) Limited filed Critical Glaxosmithkline Intellectual Property (No.2) Limited
Publication of WO2015056180A1 publication Critical patent/WO2015056180A1/fr
Publication of WO2015056180A8 publication Critical patent/WO2015056180A8/fr

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/04Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D401/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom
    • C07D401/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, at least one ring being a six-membered ring with only one nitrogen atom containing three or more hetero rings
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Definitions

  • the present invention relates to substituted indoline 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 and diseases associated with activated unfolded protein response pathways, such as Alzheimer's disease, stroke, Type 1 diabetes, Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, atherosclerosis, and arrhythmias.
  • the unfolded protein response is a signal transduction pathway that allows cells to survive environmental stresses that perturb protein folding and maturation in the endoplasmic reticulum (ER) (Ma and Hendershot, 2004), (Feldman et al., 2005), (Koumenis and Wouters, 2006).
  • Stress stimuli that activate UPR include hypoxia, disruption of protein glycosylation (glucose deprivation), depletion of luminal ER calcium, or changes in ER redox status (Ma and Hendershot, 2004), (Feldman et al., 2005). These perturbations result in the accumulation of unfolded or mis-folded proteins in the ER, which is sensed by resident ER membrane proteins.
  • These proteins activate a coordinated cellular response to alleviate the impact of the stress and enhance cell survival.
  • Responses include an increase in the level of chaperone proteins to enhance protein re-folding, degradation of the mis-folded proteins, and translational arrest to decrease the burden of proteins entering the ER.
  • These pathways also regulate cell survival by modulating apoptosis (Ma and Hendershot, 2004), (Feldman et al., 2005), (Hamanaka et al., 2009) and autophagy (Rouschop et al.), and can trigger cell death under conditions of prolonged ER stress.
  • PPR protein kinase R
  • EIF2AK3 eukaryotic initiation factor 2A kinase 3
  • PEK pancreatic elF2a kinase
  • IRE1 eukaryotic initiation factor 2A kinase 3
  • 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), (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).
  • the major substrate for PERK is the eukaryotic initiation factor 2a (elF2a) at serine-51 (Marciniak et al., 2006). This site is also phosphorylated by other PERK family members [(general control non-derepressed 2 (GCN2), PKR, and heme-regulated kinase (HRI)] in response to different stimuli, and by pharmacological inducers of ER stress such as thapsigargin and tunicamycin. Phosphorylation of elF2a converts it to an inhibitor of elF2B, which hinders the assembly of the 40S ribosome translation initiation complex and consequently reduces the rate of translation initiation.
  • GCN2 general control non-derepressed 2
  • HRI heme-regulated kinase
  • 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 (Delepine et al., 2000).
  • IRE1 is a transmembrane protein with kinase and endonulease (RNAse) functions (Feldman et al., 2005) (Koumenis and Wouters, 2006).
  • XBP1 unspliced X-box binding protein 1
  • ATF6 The third effector of UPR, ATF6, is transported to the golgi upon ER stress, where it is cleaved by proteases to release the cytosolic transcription domain. This domain translocates to the nucleus and activates transcription of UPR genes (Feldman et al., 2005), (Koumenis and Wouters, 2006).
  • 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 and in the treatment of diseases associated with activated unfolded protein response pathways, such as Alzheimer's disease, stroke and Type 1 diabetes.
  • Loss of endoplasmic reticulum homeostasis and accumulation of misfolded proteins can contribute to a number of disease states including cardiovascular and degenerative diseases (Paschen, 2004) such as: Alzheimer's disease (Salminen e.t al., 2009 and O'Connor et. al. 2008), Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis (Kanekura et. al., 2009 and Nassif et. al. 2010), myocardial infarction, cardiovascular disease, atherosclerosis (McAlpine et. al, 2010), and arrhythmias.
  • a PERK inhibitor is expected to have utility in the treatment of such cardiovascular and degenerative diseases in which the underlying pathology and symptoms are associated with dysregulaton of the unfolded protein response.
  • Nrf2 PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress, J Biol Chem 279, 20108-17. Davies, M. P., Barraclough, D. L, Stewart, C, Joyce, K. A., Eccles, R. M., Barraclough, R., Rudland, P. S., and Sibson, D. R. (2008). Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer, Int J Cancer 123, 85-8.
  • PERK and GCN2 contribute to elF2alpha phosphorylation and cell cycle arrest after activation of the unfolded protein response pathway, Mol Biol Cell 16, 5493-501. Hamanaka, R. B., Bobrovnikova-Marjon, E., Ji, X., Liebhaber, S. A., and Diehl, J. A. (2009). PERK-dependent regulation of IAP translation during ER stress, Oncogene 28, 910-20.
  • Cigarette smoke induces endoplasmic reticulum stress and the unfolded protein response in normal and malignant human lung cells, BMC Cancer 8, 229.
  • XBP1 is essential for survival under hypoxic conditions and is required for tumor growth, Cancer Res 64, 5943- 7. Rouschop, K. M., van den Beucken, T., Dubois, L, Niessen, H., Bussink, J., Savelkouls, K., Keulers, T., Mujcic, H., Landuyt, W., Voncken, J. W., et al. The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1 LC3B and ATG5, J Clin Invest 120, 127-41.
  • diseases associated with activated unfolded protein response pathways such as Alzheimer's disease, stroke, Type 1 diabetes, Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, atherosclerosis, and arrhythmias, that comprises administering inhibitors of PERK activity.
  • the invention is directed to substituted indoline derivatives. Specifically, the invention is directed to compounds according to Formula I:
  • Rl R2 and R3 are defined below.
  • 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 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 Type 1 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 state selected from: Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, atherosclerosis, and arrhythmias, which comprises administering to a subject in need thereof an effective amount of a PERK inhibiting compound of Formula (I).
  • a disease state selected from: Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, atherosclerosis, and arrhythmias
  • compositions that comprise a pharmaceutical carrier and compounds useful in the methods of the invention.
  • Also included in the present invention are methods of co-administering the presently invented PERK inhibiting compounds with further active ingredients.
  • This invention relates to novel compounds of Formula (I):
  • aryl substituted with from one to three substituents independently selected from: C-
  • heterocycloalkyl substituted with from one to three substituents independently selected from: C-
  • -4alkyl diCi-4alkylaminoC-
  • heteroaryl substituted with from one to three substituents independently selected from: C-
  • R is selected from:
  • heteroaryl substituted with from one to five substituents independently selected from: fluoro, chloro, bromo, iodo, C-
  • cycloalkyi substituted with from one to five substituents independently selected from: fluoro, chloro, bromo, iodo, C-
  • R is selected from: hydrogen, fluoro, chloro, bromo and iodo; and salts thereof.
  • This invention also relates to pharmaceutically acceptable salts of the compoundsula (I).
  • R1 is heteroaryl substituted with from one substituents independently selected from:
  • aryl substituted with from one to three substituents independently selected from: Ci_4alkyl, diCi-4alkylaminoCi-4alkyl, fluoro, chloro, bromo, iodo and -CF3, heterocycloalkyl,
  • heterocycloalkyl substituted with from one to three substituents independently selected from: C-
  • .Ci-4alkylheterocycloalkyl substituted with from one to three substituents independently selected from: C-
  • heteroaryl substituted with from one to three substituents independently selected from: C-
  • R1 is heteroaryl substituted with from one to three substituents independently selected from:
  • R1 is a substituted heteroaryl where the substituents are as described herein and the heteroaryl is selected from: pyrazole, pyrrole, isoxazole, pyridine, pyrimidine, pyridazine, and imidazole.
  • R1 is a substituted pyrazole where the substituents are as described herein.
  • R1 is , optionally substituted with one or two substituents independently selected from:
  • R2 is selected from:
  • heteroaryl substituted with from one to three substituents independently selected from: fluoro, chloro, bromo, iodo, C-
  • R is selected from: hydrogen, fluoro and chloro.
  • 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 unsatled compound. Accordingly, the invention is further directed to salts, including pharmaceutically-acceptable salts, of the compounds according to Formula I.
  • the 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. The invention includes all such solvates.
  • 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-C4 alkyl refers to an alkyl group having from 1 to 4 member atoms.
  • Alkyl groups may be saturated, unsaturated, straight or branched. Representative branched alkyl groups have one, two, or three branches. Alkyl includes methyl, ethyl, ethylene, propyl (n-propyl and isopropyl), butene, and butyl (n-butyl, isobutyl, and t-butyl).
  • Alkoxy refers to an -O-alkyl group wherein “alkyl” is as defined herein.
  • -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.
  • Aryl groups are monocyclic, bicyclic, and tricyclic ring systems having a total of five to fourteen ring member atoms, wherein at least one ring system is aromatic and wherein each ring in the system contains 3 to 7 member atoms, such as phenyl, naphthalene, tetrahydronaphthalene and biphenyl.
  • aryl is phenyl.
  • Cycloalkyl 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.
  • cycloalkyl examples include: cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutenyl, cyclopentenyl and cyclohexenyl.
  • Halo refers to the halogen radicals fluoro, chloro, bromo, and iodo.
  • Heteroaryl refers to a monocyclic aromatic 4 to 8 member ring containing from 1 to 7 carbon atoms and containing from 1 to 4 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 refers to a saturated or unsaturated non-aromatic ring containing 4 to 12 member atoms, of which 1 to 11 are carbon atoms and from 1 to 6 are heteroatoms. Heterocycloalkyl groups containing more than one heteroatom may contain different heteroatoms. Heterocycloalkyl groups are monocyclic ring systems or a monocyclic ring fused with an aryl ring or to a heteroaryl ring having from 3 to 6 member atoms.
  • Heterocycloalkyl includes: pyrrolidinyl, tetrahydrofuranyl, dihydrofuranyl, pyranyl, tetrahydropyranyl, dihydropyranyl, 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.
  • “Pharmaceutically acceptable” refers to those compounds, materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.
  • ACN acetonitrile
  • AIBN azobis(isobutyronitrile)
  • BINAP (2,2'-bis(diphenylphosphino)-1 , 1'-binaphthyl);
  • BOP Benzotriazole-l-yl-oxy-tris-(dimethylamino)-phosphonium hexafluorophosphate
  • ATP adenosine triphosphate
  • BSA bovine serum albumin
  • C18 refers to 18-carbon alkyl groups on silicon in HPLC stationary phase
  • DIPEA Human's base, N-ethyl-N-(1-methylethyl)-2-propanamine
  • DPPA diphenyl phosphoryl azide
  • EDC N-(3-dimethylaminopropyl)-N'ethylcarbodiimide
  • EDTA ethylenediaminetetraacetic acid
  • HEPES (4-(2-hydroxyethyl)-1-piperazine ethane sulfonic acid);
  • HATU (0-(7-Azabenzotriazol-1-yl)-N,N,N',N'-tetramethyluronium hexafluorophosphate); H OAt ( 1 - hyd roxy-7-azabenzotriazole) ;
  • HMDS hexamethyldisilazide
  • Hunig's Base N,N-Diisopropylethylamine
  • KHMDS potassium hexamethyldisilazide
  • LAH lithium aluminum hydride
  • mCPBA m-chloroperbezoic acid
  • NaHMDS sodium hexamethyldisilazide
  • NBS N-bromosuccinimide
  • PE petroleum ether
  • TFA trifluoroacetic acid
  • 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.
  • a substituent described herein is not compatible with the synthetic methods described herein, the substituent may be protected with a suitable protecting group that is stable to the reaction conditions.
  • the protecting group may be removed at a suitable point in the reaction sequence to provide a desired intermediate or target compound.
  • suitable protecting groups and the methods for protecting and de-protecting different substituents using such suitable protecting groups are well known to those skilled in the art; examples of which may be found in T. Greene and P. Wuts, Protecting Groups in Chemical Synthesis (3rd ed.), John Wley & Sons, NY (1999).
  • a substituent may be specifically selected to be reactive under the reaction conditions used.
  • 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.
  • a coupling reagent e.g., EDC, DCC or HATU
  • Conversion of 2 to the boronate ester and subsequent Suzuki-Miyaura coupling with heteroaryl bromide A affords the product 3.
  • the boronate ester (represented by 4) may be purified and isolated if desired and subjected to the Suzuki- Miyaura coupling in a separate synthetic procedure.
  • the compounds of the invention can be prepared as shown in Scheme 2.
  • the nitrogen of 5-bromoindoline 1 can be protected with the te/f-butylcarbamate (Boc) group to give 8.
  • the analogous 4-F derivative can be prepared from 4-F indole via sodium cyanoborohydride reduction, N-Boc protection, followed by regioselective bromination with NBS. Transformation to the heteroaryl substituted indoline 9 is accomplished with or without isolation of the intermediate boronate ester. Deprotection of the Boc group with HCI affords the indoline 10, which can be converted to 11 using a coupling reagent (e.g., EDC, DCC or HATU) to form the amide bond.
  • a coupling reagent e.g., EDC, DCC or HATU
  • Heteroaryl halide A is prepared as shown in Scheme 3.
  • Dibromopyrazole 14 can be prepared from commercial 3-aminopyrazole-4-carbonitrile 12 using t-Bu nitrite in bromoform followed by bromine; alternatively dibromopyrazole 14 can be prepared by dibromination of commercial 1 H-pyrazole-4-carbonitrile 15 with bromine.
  • Intermediate 14 is alkylated with an alkyl iodide to form intermediate 16 which is treated with 2,4- dimethoxybenzylamine to install the 5-amino group of intermediate 17. Hydrolysis of the nitrile of intermediate 17 with sulfuric acid affords intermediate 18.
  • 3-Aminopyrazole-4- carbonitrile 12 can be brominated with NBS to form intermediate 19 and following hydrolysis with sulfuric acid, gives intermediate 20.
  • 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, cancer and more specifically cancers of the breast, colon, and lung, pancreas and skin. 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,
  • 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 precancerous 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.
  • the precancerous 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
  • the present invention relates to a method for treating or lessening the severity of additional diseases associated with UPR activation including: Type 1 diabetes, Alzheimer's disease, stroke, Parkinson disease, Huntington's disease, amyotrophic lateral sclerosis, myocardial infarction, cardiovascular disease, atherosclerosis, and arrhythmias.
  • 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 can be: edema or neovascularization for any occlusive or inflammatory retinal vascular disease, such as rubeosis irides, neovascular glaucoma, pterygium, vascularized glaucoma filtering blebs, conjunctival papilloma; choroidal neovascularization, such as neovascular age-related macular degeneration (AMD), myopia, prior uveitis, trauma, or idiopathic; macular edema, such as post surgical macular edema, macular edema secondary to uveitis including retinal and/or choroidal inflammation, macular edema secondary to diabetes, and macular edema secondary to retinovascular occlusive disease (i.e.
  • retinal vascular disease such as rubeosis irides, neovascular glaucoma, pterygium,
  • retinal neovascularization due to diabetes such as retinal vein occlusion, uveitis, ocular ischemic syndrome from carotid artery disease, ophthalmic or retinal artery occlusion, sickle cell retinopathy, other ischemic or occlusive neovascular retinopathies, retinopathy of prematurity, or Eale's Disease; and genetic disorders, such as VonHippel- Lindau syndrome.
  • the neovascular age-related macular degeneration is wet age-related macular degeneration. In other embodiments, the neovascular age-related macular degeneration is dry age-related macular degeneration and the patient is characterized as being at increased risk of developing wet age-related macular degeneration.
  • the 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 cancer therapy.
  • the invention is directed to the use of a compound according to Formula I or a pharmaceutically acceptable salt thereof in the preparation of a medicament for the treatment of a disorder characterized by activation of the UPR, such as cancer.
  • treating is meant prophylactic and therapeutic therapy.
  • Prophylactic therapy is appropriate, for example, when a subject has 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 over time as individual patient needs change.
  • a prodrug of a compound of the invention is a functional derivative of the compound which, upon administration to a patient, eventually liberates the compound of the invention in vivo.
  • Administration of a compound of the invention as a prodrug may enable the skilled artisan to do one or more of the following: (a) modify the onset of the compound in vivo; (b) modify the duration of action 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 co-administered with at least one other active agent known to be useful in the treatment of cancer.
  • 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
  • anti-neoplastic agent 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.
  • 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 anticancer 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-11-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. It was first isolated in 1971 by Wani et al. J. Am. Chem, Soc, 93:2325. 1971), who characterized its structure by chemical and X-ray crystallographic methods.
  • 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., 11 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 A 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 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.
  • the dose limiting toxicity of docetaxel is neutropenia.
  • 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, is commercially available as VELBAN® as an injectable solution.
  • 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, 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.
  • the primary dose limiting side effects of cisplatin are nephrotoxicity, which may be controlled by hydration and diuresis, and ototoxicity.
  • 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. Bone marrow suppression is the dose limiting toxicity of carboplatin.
  • 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. Alopecia, nausea, vomiting and leukopenia are the most common dose limiting side effects of cyclophosphamide.
  • 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. Bone marrow suppression is the most common dose limiting side effect of chlorambucil.
  • Busulfan 1 ,4-butanediol dimethanesulfonate, is commercially available as MYLERAN® TABLETS. Busulfan is indicated for the palliative treatment of chronic myelogenous leukemia. Bone marrow suppression is the most common dose limiting side effects of busulfan.
  • 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. Delayed myelosuppression is the most common dose limiting side effects of carmustine.
  • 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. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dacarbazine.
  • 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 know as Actinomycin D, is commercially available in injectable form as COSMEGEN®. Dactinomycin is indicated for the treatment of Wilm's tumor and rhabdomyosarcoma. Nausea, vomiting, and anorexia are the most common dose limiting side effects of dactinomycin.
  • Daunorubicin (8S-cis-)-8-acetyl-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo- hexopyranosyl)oxy]-7,8,9, 10-tetrahydro-6,8, 11-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. Myelosuppression is the most common dose limiting side effect of daunorubicin.
  • Doxorubicin (8S, 10S)-10-[(3-amino-2,3,6-trideoxy-a-L-lyxo-hexopyranosyl)oxy]- 8-glycoloyl, 7,8,9, 10-tetrahydro-6, 8,1 1-trihydroxy-1-methoxy-5, 12 naphthacenedione hydrochloride, is commercially available as an injectable form as RUBEX® or ADRIAMYCIN RDF®.
  • Doxorubicin is primarily indicated for the treatment of acute lymphoblastic leukemia and acute myeloblastic leukemia, but is also a useful component in the treatment of some solid tumors and lymphomas. Myelosuppression is the most common dose limiting side effect of doxorubicin. 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. Pulmonary and cutaneous toxicities are the most common dose limiting side effects of bleomycin.
  • 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]
  • VePESID® an injectable solution or capsules
  • VP-16 an injectable solution or capsules
  • 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. Myelosuppression is the most common side effect of etoposide. The incidence of leucopenia tends to be more severe than thrombocytopenia.
  • 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. Myelosuppression is the most common dose limiting side effect of teniposide.
  • Teniposide can induce both leucopenia and thrombocytopenia.
  • 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.
  • antimetabolite antineoplastic agents include, but are not limited to, fluorouracil, methotrexate, cytarabine, mecaptopurine, thioguanine, and gemcitabine. 5-fluorouracil, 5-fluoro-2,4- (1 H,3H) pyrimidinedione, is commercially available as fluorouracil.
  • 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. Myelosuppression and mucositis are dose limiting side effects of 5- fluorouracil.
  • Other fluoropyrimidine analogs include 5-fluoro deoxyuridine (floxuridine) and 5-fluorodeoxyuridine monophosphate.
  • Cytarabine 4-amino-1-p-D-arabinofuranosyl-2 (I 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). Cytarabine induces leucopenia, thrombocytopenia, and mucositis.
  • Mercaptopurine 1 ,7-dihydro-6H-purine-6-thione monohydrate, is commercially available as PURINETHOL®.
  • Mercaptopurine exhibits cell phase specificity at S-phase by inhibiting DNA synthesis by an as of yet unspecified mechanism.
  • Mercaptopurine is indicated as a single agent or in combination with other chemotherapy agents in the treatment of acute leukemia. Myelosuppression and gastrointestinal mucositis are expected side effects of mercaptopurine at high doses.
  • a useful mercaptopurine analog is azathioprine.
  • Thioguanine 2-amino-1 ,7-dihydro-6H-purine-6-thione
  • 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.
  • Myelosuppression including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of thioguanine administration.
  • 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. Myelosuppression, including leucopenia, thrombocytopenia, and anemia, is the most common dose limiting side effect of gemcitabine administration.
  • 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.
  • Myelosuppression (leucopenia, thrombocytopenia, and anemia) and mucositis are expected side effect of methotrexate administration.
  • 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. It is believed that 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. The dose limiting side effects of irinotecan HCI are myelosuppression, including neutropenia, and Gl effects, including diarrhea.
  • 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.
  • topotecan HCI myelosuppression, primarily neutropenia.
  • camptothecin derivative of Formula A including the racemic mixture (R,S) form as well as the R and S enantiomers:
  • 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, androgens, and anti-androgens such as flutamide, nilutamide, bicalutamide, cyproterone acetate and 5a-reductases
  • 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/SH3 domain blockers, serine/threonine kinases, phosphotidylinositol-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, 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 erbB4
  • VEGFr vascular endothelial growth factor receptor
  • TIE-2 vascular endothelial growth factor receptor
  • TIE-2 t
  • 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.
  • 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 monohydrochlor
  • 5-[[4-[(2,3- dimethyl-2H-indazol-6-yl)methylamino]-2-pyrimidinyl]amino]-2-methylbenzenesulfonamide can be prepared as described in International Application No. PCT/US01/49367.
  • 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.
  • Tyrosine kinases which are not growth factor receptor kinases are termed nonreceptor tyrosine kinases.
  • Non-receptor tyrosine kinases for use in the present invention 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 nonreceptor kinases and agents which inhibit non-receptor tyrosine kinase function are described in Sinh, S. and Corey, S.J., (1999) Journal of Hematotherapy and Stem Cell Research 8 (5): 465 - 80; and Bolen, J.B., Brugge, J.S., (1997) Annual review of Immunology. 15: 371-404.
  • 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
  • PDK1 and TGF beta receptor kinases IkB kinase family
  • 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. 1101-1107; 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.
  • the pharmaceutically active compounds of the invention are used in combination with a MEK inhibitor.
  • N- ⁇ 3-[3-cyclopropyl-5-(2-fluoro-4-iodo-phenylamino)-6,8-dimethyl-2,4,7-trioxo-3,4,6,7- tetrahydro-2H-pyrido[4,3-d]pyrimidin-1-yl]phenyl ⁇ acetamide can be prepared as described in United States Patent Publication No. US 2006/0014768, Published January 19, 2006, the entire disclosure of which is hereby incorporated by reference.
  • the pharmaceutically active compounds of the invention are used in combination with a B-Raf inhibitor.
  • a B-Raf inhibitor e.g., A/- ⁇ 3-[5-(2-Amino-4-pyrimidinyl)-2-(1 , 1- dimethylethyl)-1 ,3-thiazol-4-yl]-2-fluorophenyl ⁇ -2,6-difluorobenzenesulfonamide, or a pharmaceutically acceptable salt thereof, which is disclosed and claimed, in International Application No. PCT/US2009/042682, having an International filing date of May 4, 2009, the entire disclosure of which is hereby incorporated by reference.
  • A/- ⁇ 3-[5-(2-Amino-4- pyrimidinyl)-2-(1 , 1-dimethylethyl)-1 ,3-thiazol-4-yl]-2-fluorophenyl ⁇ -2,6- difluorobenzenesulfonamide can be prepared as described in International Application No. PCT/US2009/042682.
  • the pharmaceutically active compounds of the invention are used in combination with an Akt inhibitor.
  • an Akt inhibitor e.g., N- ⁇ (1 S)-2-amino-1-[(3,4- difluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2- furancarboxamide or a pharmaceutically acceptable salt thereof, which is disclosed and claimed in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008, the entire disclosure of which is hereby incorporated by reference.
  • N- ⁇ (1 S)-2-amino-1-[(3,4-difluorophenyl)methyl]ethyl ⁇ - 5-chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2-furancarboxamide is the compound of example 224 and can be prepared as described in International Application No. PCT/US2008/053269.
  • the pharmaceutically active compounds of the invention are used in combination with an Akt inhibitor.
  • an Akt inhibitor e.g., A/- ⁇ (1 S)-2-amino-1-[(3- fluorophenyl)methyl]ethyl ⁇ -5-chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2- thiophenecarboxamide or a pharmaceutically acceptable salt thereof, which is disclosed and claimed in International Application No. PCT/US2008/053269, having an International filing date of February 7, 2008; International Publication Number WO 2008/098104 and an International Publication date of August 14, 2008, the entire disclosure of which is hereby incorporated by reference.
  • A/- ⁇ (1 S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5- chloro-4-(4-chloro-1-methyl-1 /-/-pyrazol-5-yl)-2-thiophenecarboxamide is the compound of example 96 and can be prepared as described in International Application No. PCT/US2008/053269.
  • A/- ⁇ (1 S)-2-amino-1-[(3-fluorophenyl)methyl]ethyl ⁇ -5- chloro-4-(4-chloro-1-methyl-1 H-pyrazol-5-yl)-2-thiophenecarboxamide is in the form of a hydrochloride salt.
  • the salt form can be prepared by one of skill in the art from the description in International Application No. PCT/US2010/022323, having an International filing date of January 28, 2010.
  • Inhibitors of Phosphotidylinositol-3 Kinase family members including blockers of PI3-kinase, ATM, DNA-PK, and Ku may also be 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 cancer:erbB Family Receptor Tyrosine Kniases, 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).
  • Non-receptor kinase angiogenesis inhibitors may also be useful in the present invention.
  • Inhibitors of angiogenesis related VEGFR and TIE2 are discussed above in regard to signal transduction inhibitors (both receptors are receptor tyrosine kinases).
  • Angiogenesis in general is linked to erbB2/EGFR signaling since inhibitors of erbB2 and EGFR have been shown to inhibit angiogenesis, primarily VEGF expression. Accordingly, non-receptor tyrosine kinase inhibitors may be used in combination with the compounds of the present invention.
  • anti-VEGF antibodies which do not recognize VEGFR (the receptor tyrosine kinase), but bind to the ligand; small molecule inhibitors of integrin (alpha v beta 3 ) that will inhibit angiogenesis; endostatin and angiostatin (non-RTK) may also prove useful in combination with the disclosed compounds.
  • VEGFR the receptor tyrosine kinase
  • small molecule inhibitors of integrin alpha v beta 3
  • endostatin and angiostatin non-RTK
  • Agents used in immunotherapeutic regimens may also be useful in combination with the compounds of Formula (I).
  • immunologic strategies to generate an immune response. These strategies are generally in the realm of tumor vaccinations.
  • the efficacy of immunologic approaches may be greatly enhanced through combined inhibition of signaling pathways using a small molecule inhibitor. Discussion of the immunologic/tumor vaccine approach against erbB2/EGFR are found in Reilly RT et al. (2000), Cancer Res. 60: 3569-3576; and Chen Y, Hu D, Eling DJ, Robbins J, and Kipps TJ. (1998), Cancer Res. 58: 1965-1971.
  • Agents used in proapoptotic regimens may also be used in the combination of the present invention.
  • Members of the Bcl-2 family of proteins block apoptosis. Upregulation of bcl-2 has therefore been linked to chemoresistance.
  • EGF epidermal growth factor
  • 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.
  • p21WAF1/CIP1 has been described as a potent and universal inhibitor of cyclin-dependent kinases (Cdks) (Ball et al., Progress in Cell Cycle Res., 3: 125 (1997)).
  • Cdks cyclin-dependent kinases
  • Compounds that are known to induce expression of p21WAF1/CIP1 have been implicated in the suppression of cell proliferation and as having tumor suppressing activity (Richon et al., Proc. Nat Acad. Sci. U.S.A. 97(18): 10014-10019 (2000)), and are included as cell cycle signaling inhibitors.
  • Histone deacetylase (HDAC) inhibitors are implicated in the transcriptional activation of p21WAF1/CIP1 (Vigushin et al., Anticancer Drugs, 13(1): 1-13 (Jan 2002)), and are suitable cell cycle signaling inhibitors for use herein.
  • HDAC inhibitors examples include:
  • Vorinostat including pharmaceutically acceptable salts thereof. Marks et al., Nature Biotechnology 25, 84 to 90 (2007); Stenger, Community Oncology 4, 384-386 (2007).
  • Vorinostat has the following chemical structure and name:
  • Romidepsin has the following chemical structure and name:
  • Panobinostat including pharmaceutically acceptable salts thereof.
  • Panobinostat has the following chemical structure and name:
  • Valproic acid including pharmaceutically acceptable salts thereof. Gottlich, et al., EM BO J. 20(24): 6969-6978 (2001).
  • Valproic acid has the following chemical structure and name:
  • Mocetinostat (MGCD0103), including pharmaceutically acceptable salts thereof. Balasubramanian et al., Cancer Letters 280: 211-221 (2009).
  • Mocetinostat has the following chemical structure and name:
  • HDAC inhibitors are included in Bertrand European Journal of Medicinal Chemistry 45, (2010) 2095-2116, particularly the compounds of table 3 therein as indicated below.
  • proteasome inhibitors are drugs that block the action of proteasomes, cellular complexes that break down proteins, like the p53 protein.
  • proteasome inhibitors are marketed or are being studied in the treatment of cancer. Suitable proteasome inhibitors for use herein include:
  • Bortezomib has the following chemical structure and name.
  • Disulfiram including pharmaceutically acceptable salts thereof.
  • Disulfiram has the following chemical structure and name.
  • Epigallocatechin gallate has the following chemical structure and name.
  • Salinosporamide A including pharmaceutically acceptable salts thereof. Feling et at., (2003), Angew. Chem. Int. Ed. Engl. 42 (3): 355-7.
  • Salinosporamide A has the following re and name.
  • Carfilzomib including pharmaceutically acceptable salts thereof. Kuhn DJ, et al, Blood, 2007, 1 10:3281-3290.
  • Carfilzomib has the following chemical structure and name.
  • Hsp70s and Hsp90s are a families of ubiquitously expressed heat shock proteins. Hsp70s and Hsp90s are over expressed certain cancer types. Several Hsp70s and Hsp90s inhibitors are being studied in the treatment of cancer. Suitable Hsp70s and Hsp90s inhibitors for use in combination herein include:
  • 17-AAG(Geldanamycin) has the following chemical structure and name.
  • Radicicol has the following chemical structure and name.
  • Inhibitors of cancer metabolism Many tumor cells show a markedly different metabolism from that of normal tissues. For example, the rate of glycolysis, the metabolic process that converts glucose to pyruvate, is increased, and the pyruvate generated is reduced to lactate, rather than being further oxidized in the mitochondria via the tricarboxylic acid (TCA) cycle. This effect is often seen even under aerobic conditions and is known as the Warburg Effect.
  • TCA tricarboxylic acid
  • Lactate dehydrogenase A (LDH-A), an isoform of lactate dehydrogenase expressed in muscle cells, plays a pivotal role in tumor cell metabolism by performing the reduction of pyruvate to lactate, which can then be exported out of the cell.
  • the enzyme has been shown to be upregulated in many tumor types.
  • the alteration of glucose metabolism described in the Warburg effect is critical for growth and proliferation of cancer cells and knocking down LDH-A using RNA-i has been shown to lead to a reduction in cell proliferation and tumor growth in xenograft models.
  • FAS fatty acid synthase
  • the cancer treatment method of the claimed invention includes the co-administration a compound of Formula (I) and/or a pharmaceutically acceptable salt thereof and at least one anti-neoplastic agent, such as one selected from the group consisting of anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor tyrosine kinase angiogenesis inhibitors, immunotherapeutic agents, proapoptotic agents, cell cycle signaling inhibitors; proteasome inhibitors; and inhibitors of cancer metabolism.
  • anti-neoplastic agent such as one selected from the group consisting of anti-microtubule agents, platinum coordination complexes, alkylating agents, antibiotic agents, topoisomerase II inhibitors, antimetabolites, topoisomerase I inhibitors, hormones and hormonal analogues, signal transduction pathway inhibitors, non-receptor t
  • 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, arthritis 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, subcutaneous, intradermal, and parenteral.
  • 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 - 100 mg/kg of active compound, preferably 0.001 - 50 mg/kg.
  • the selected dose is administered preferably from 1-6 times daily, orally or parenterally.
  • Preferred forms of parenteral administration include topically, rectally, 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, which uses lower dosages, is preferred. Parenteral administration, at high dosages, however, also can be used when safe and convenient for the patient.
  • Optimal dosages to be administered may be readily determined by those skilled in the art, and will vary with the particular 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.
  • 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.
  • 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 cancer which comprises a compound of Formula (I) or a pharmaceutically acceptable salt thereof and a pharmaceutically acceptable carrier.
  • the pharmaceutically active compounds of the present invention can be co-administered with further active ingredients, such as other compounds known to treat cancer, or compounds known to have utility when used in combination with a PERK inhibitor.
  • the column was eluted with 400 mL of 1 % DCM in hexane, 400 mL of 1/3 DCM/hexane, 400 mL of 1/1 DCM/hexane, and then 400 mL 1/1 DCM/hexane portions each with 20 mL increment of EtOAc.
  • the desired product eluted from 20 mL to 60 mL EtOAc fractions.
  • the collected fractions (including the one from above Analogix prep) were combined and concentrated in vacuo to about 100 mL volume as a suspension. This suspension was filtered.
  • the mixture was degassed and backflushed with nitrogen four times.
  • the mixture was sealed and heated in an oil bath at 100 °C for two days overnight.
  • the mixture was cooled to room temperature, diluted with EtOAc and the layers separated.
  • the EtOAc layer was concentrated in vacuo to give a pale yellow solid.
  • the solid was purified by flash silica chromatography (SF 15-24 g at CH 2 CI 2 -10% MeOH in CH 2 CI 2 , the compound eluted at 7% MeOH in CH 2 CI 2 ) to afford a solid which was triturated with MeOH, filtered, and dried to afford 5- amino-3-(1- ⁇ [3-(trifluoromethyl)phenyl]acetyl ⁇ -2,3-dihydro-1 H-indol-5-yl)-1 H-pyrazole-4- carboxamide (6 mg, 0.014 mmol) as an off-white solid.
  • LC-MS(ES) m/z 458 [M+H] + .
  • NMP N-methyl-2-pyrrolidone
  • reaction mixture was poured into 1 : 1 saturated aqueous NaCI: H 2 0 (100 ml_) and ethyl acetate (100 ml_), shaken, and filtered through celite. The resulting mixture was separated and the aqueous layer was extracted with two additional portions of ethyl acetate (2 x 50 ml_). The combined organics were dried over sodium sulfate, filtered, and concentrated.
  • reaction mixture was then loaded directly on to an Analogix 24 g column conditioned with hexane and then purified by flash silica chromatography (gradient of 0 to 100% EtOAc in hexane for 15 min then 15 min at 100% EtOAc).
  • the fractions with the desired product were combined then transferred to 40 mL vial with MeCN. Water was added and the mixture was freeze-dried to isolate 5-amino-3-(4-fluoro-1- ⁇ [3- (trifluoromethyl)phenyl]acetyl ⁇ -2,3-dihydro-1 H-indol-5-yl)-1-methyl-1 H-pyrazole-4- carboxamide (60 mg, 0.130 mmol, 40.5 % yield) as a white solid.
  • the reaction was then loaded directly on to an Analogix 24 g column conditioned with hexane and purified by flash silica chromatography (gradient of 0 to 100% EtOAc in hexane for 15 min then 15 min at 100% EtOAc). The fractions with the desired product were combined then transferred to 40 mL vial with MeCN. Water was added and the mixture was freeze-dried.
  • reaction mixture was then loaded directly on to a Analogix 24 g column conditioned with hexane and purified by flash silica chromatography (3 min at 100% hexane then a gradient of 0 to 10% MeOH in DCM over 25 min). The fractions with the desired product were combined then concentrated. The solid was then triturated with EtOAc and hexane then filtered to afford 5-amino-3- ⁇ 4-fluoro-1-[(6-methyl-2-pyridinyl)acetyl]-2,3-dihydro-1 H- indol-5-yl ⁇ -1-methyl-1 H-pyrazole-4-carboxamide (78 mg) as a white solid.
  • reaction mixture was then loaded directly on to a Analogix 24 g column conditioned with hexane and purified by flash silica chromatography (3 min at 100% hexane then a gradient of 0 to 10% MeOH in DCM over 25 min). The fractions with the desired product were combined then concentrated.
  • reaction solution was then loaded directly onto a double stacked (2X) 10 g Biotage SNAP columns first conditioned with hexane and purified by flash silica chromatography (4 min 100% hexane, then 3 min 100% DCM, then 0 to 10% MeOH in DCM for 20 min).
  • product fractions were combined and concentrated then transferred to a 40 mL vial and MeCN and water were added.
  • the sample was freeze-dried to afford 5-amino-1-methyl-3-(1-(2-(3- (trifluoromethyl)phenyl)acetyl)indolin-5-yl)-1 H-pyrazole-4-carboxamide (87 mg) as a white powder.
  • reaction solution was then loaded directly onto a double stacked (2X) 10 g Biotage SNAP column first conditioned with hexane and purified by flash silica chromatography (4 min 100% hexane, then 3 min 100% DCM, then 0 to 10% MeOH in DCM for 20 min).
  • product fractions were combined and concentrated then transferred in to a 40 mL vial and MeCN and water were added.
  • the sample was freeze-dried to afford 5-amino-1-methyl-3-(1-(2-(6-methylpyridin- 2-yl)acetyl)indolin-5-yl)-1 H-pyrazole-4-carboxamide (70 mg) as a white powder.
  • reaction solution was then loaded directly onto a double stacked (2X) 10 g Biotage SNAP column first conditioned with hexane and purified by flash silica chromatography (4 min 100% hexane, then 3 min 100% DCM, then 0 to 10% MeOH in DCM for 20 min).
  • product fractions were combined and concentrated then transferred to a 40 ml_ vial and MeCN and water were added.
  • the sample was freeze- dried to afford 5-amino-1-methyl-3-(1-(2-(6-(trifluoromethyl)pyridin-2-yl)acetyl)indolin-5- yl)-1 H-pyrazole-4-carboxamide (1 12 mg) as a white powder.
  • reaction solution was then loaded directly onto a double stacked (2X) 10 g Biotage SNAP column first conditioned with hexane and purified by flash silica chromatography (4 min 100% hexane, then 3 min 100% DCM, then 0 to 10% MeOH in DCM for 20 min).
  • the product fractions were combined and concentrated and the residue was purified by flash silica chromatography on a 10 g Biotage column conditioned with hexanes (3 min 100% hexane, then 0 to 100% EtOAC for 10 min then 0 to 10% MeOH in EtOAc).
  • the product fractions were combined, concentrated, and then transferred to a 40 ml_ vial and MeCN and water was added.
  • An oral dosage form for administering the present invention is produced by filing a standard two piece hard gelatin capsule with the ingredients in the proportions shown in Table I, below.
  • An injectable form for administering the present invention is produced by stirring 1.7% by weight of 1-Methyl-5-(methylamino)-3-(1- ⁇ [3-(trifluoromethyl)phenyl]acetyl ⁇ -2,3- dihydro-1 H-indol-5-yl)-1 H-pyrazole-4-carboxamide (Compound of Example 2) in 10% by volume propylene glycol in water.
  • sucrose, calcium sulfate dihydrate and a PERK inhibitor as shown in Table II 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.
  • PERK PKR-like Endoplasmic Reticulum Kinase
  • GST-PERK (536-1 1 16) cytoplasmic domain was purchased from Invitrogen (www.invitrogen.com) catalogue#PV5106 (2011).
  • elF2a 6-His-Full-length human elF2a is purified from baculovirus expression in Sf9 insect cells.
  • the elF2 protein is buffer exchanged by dialysis into PBS, chemically modified by NHS-LC-Biotin and then buffer exchanged by dialysis into 50 mm TRIS pH 7.2 /250 mM NaCI/5 mM DTT. Protein is aliquoted and stored at -80oC.
  • the quench solution is freshly prepared and when added to the reactions gives final concentrations of 4 nM elF2a phospho-ser51-Antibody (purchased from Millipore, catalogue #07-760, www.millipore.com), 4 nM Eu-1024 labeled anti-rabbit IgG (purchased from Perkin Elmer, catalogue#AD0083), 40 nM Streptavidin Surelight APC (purchased from Perkin Elmer, catalogue* AD0201) and 15mM EDTA.
  • 4 nM elF2a phospho-ser51-Antibody purchased from Millipore, catalogue #07-760, www.millipore.com
  • 4 nM Eu-1024 labeled anti-rabbit IgG purchased from Perkin Elmer, catalogue#AD0083
  • 40 nM Streptavidin Surelight APC purchased from Perkin Elmer, catalogue* AD0201
  • 15mM EDTA 15mM EDTA.
  • Reactions were performed in black 384-well polystyrene low volume plates (Grenier, #784076) in a final volume of 10 ⁇ .
  • the reaction volume contains, in final concentrations, 10mM HEPES, 5mM MgCI 2 , 5 ⁇ ATP, 1 mM DTT, 2mM CHAPS, 40 nM biotinylated-6-His-EIF2a, and 0.4 nM GST-PERK (536-1116).
  • Assays were performed by adding GST-PERK solution to assay plates containing compounds and pre-incubated for 30 minutes at room temperature. The reaction is initiated by the addition of ATP and EIF2a substrate solution. Quench solution is added following a one hour incubation at room temperature. The plates are covered for 2 hours at room temperature prior to determination of signal. The resulting signal is quantified on a Viewlux Reader (PerkinElmer).
  • the APC Signal is normalized to the Europium signal by transforming the data through an APC/Eu calculation.
  • plC50 -Log10(K).
  • BSA bovine serum albumin
  • Example 1 The compound of Example 1 was tested generally according to the above PERK enzyme assay and in at least one experimental run exhibited a pICso value of 8.4 against PERK.
  • plC50 is defined as -log(IC50) where the IC50 value is expressed in molar units.

Landscapes

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

Abstract

L'invention concerne des dérivés substitués d'indoline. Spécifiquement, l'invention concerne des composés répondant à la Formule I : (I) dans laquelle R1, R2 et R3 sont définis dans la description. Les composés de l'invention sont des inhibiteurs de PERK et peuvent être utiles dans le traitement du cancer et de maladies associées à des voies de réponse de protéine dépliée activée, telles que la maladie d'Alzheimer, un accident vasculaire cérébral, le diabète de Type 1, la maladie de Parkinson, la maladie de Huntington, la sclérose latérale amyotrophique, un infarctus du myocarde, une maladie cardiovasculaire, l'athérosclérose et les arythmies, et plus spécifiquement les cancers du sein, du côlon, du pancréas et du poumon. Ainsi, l'invention concerne en outre des compositions pharmaceutiques comprenant un composé de l'invention. L'invention concerne en outre également des procédés d'inhibition de l'activité de PERK et le traitement de troubles associés à celle-ci au moyen d'un composé de l'invention ou d'une composition pharmaceutique comprenant un composé de l'invention.
PCT/IB2014/065310 2013-10-15 2014-10-14 Dérivés d'indoline utilisés comme inhibiteurs de perk WO2015056180A1 (fr)

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US201361891019P 2013-10-15 2013-10-15
US61/891,019 2013-10-15

Publications (2)

Publication Number Publication Date
WO2015056180A1 true WO2015056180A1 (fr) 2015-04-23
WO2015056180A8 WO2015056180A8 (fr) 2015-12-03

Family

ID=51982658

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/IB2014/065310 WO2015056180A1 (fr) 2013-10-15 2014-10-14 Dérivés d'indoline utilisés comme inhibiteurs de perk

Country Status (1)

Country Link
WO (1) WO2015056180A1 (fr)

Cited By (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN107898793A (zh) * 2017-12-01 2018-04-13 温州医科大学 一种抑制近视的方法及制备药物的应用
WO2020070053A1 (fr) 2018-10-01 2020-04-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de formation de granules de stress pour cibler la régulation de réponses immunitaires
US10633350B2 (en) 2017-09-13 2020-04-28 Novartis Ag Diphenyl derivatives and uses thereof
US10654828B2 (en) 2017-07-28 2020-05-19 Novartis Ag Indole derivatives and uses thereof
US10918650B2 (en) * 2016-06-02 2021-02-16 University Of South Florida Method of treating melanoma using an inhibitor of an atypical protein kinase C
WO2021041976A1 (fr) * 2019-08-29 2021-03-04 Hibercell, Inc. Composés indolinyle inhibiteurs de perk
WO2021083959A1 (fr) 2019-10-29 2021-05-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pour le traitement d'un mélanome uvéal
US11026945B2 (en) 2016-04-29 2021-06-08 The Trustees Of The University Of Pennsylvania Protein kinase RNA-like endoplasmic reticulum kinase (PERK) inhibitors for prevention and/or treatment of lung injury and/or inflammation
CN115645535A (zh) * 2017-03-06 2023-01-31 坪田实验室股份有限公司 小鼠近视诱导模型及用于近视预防/抑制的内质网应激抑制剂
US11655214B2 (en) 2017-04-18 2023-05-23 Eli Lilly And Company Phenyl-2-hydroxy-acetylamino-2-methyl-phenyl compounds for the treatment of pancreatic cancer
CN115645535B (en) * 2017-03-06 2024-07-09 坪田实验室股份有限公司 Mouse myopia induction model and endoplasmic reticulum stress inhibitor for myopia prevention/inhibition

Citations (11)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5342947A (en) 1992-10-09 1994-08-30 Glaxo Inc. Preparation of water soluble camptothecin derivatives
US5491237A (en) 1994-05-03 1996-02-13 Glaxo Wellcome Inc. Intermediates in pharmaceutical camptothecin preparation
US5559235A (en) 1991-10-29 1996-09-24 Glaxo Wellcome Inc. Water soluble camptothecin derivatives
US5681835A (en) 1994-04-25 1997-10-28 Glaxo Wellcome Inc. Non-steroidal ligands for the estrogen receptor
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
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
WO2005121142A1 (fr) 2004-06-11 2005-12-22 Japan Tobacco Inc. Dérivés de 5-amino-2,4,7-trioxo-3,4,7,8-tétrahydro-2h-pyrido’2,3-d! pyrimidine et composés apparentés pour le traitement du cancer
US20060014768A1 (en) 2004-06-11 2006-01-19 Japan Tobacco Inc. Pyrimidine compound and medical use thereof
WO2007026920A2 (fr) * 2005-09-02 2007-03-08 Astellas Pharma Inc. Nouveaux composes
WO2008098104A1 (fr) 2007-02-07 2008-08-14 Smithkline Beecham Corporation Inhibiteurs de l'activité de akt
WO2011119663A1 (fr) * 2010-03-25 2011-09-29 Glaxosmithkline Llc Composés chimiques

Patent Citations (14)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5559235A (en) 1991-10-29 1996-09-24 Glaxo Wellcome Inc. Water soluble camptothecin derivatives
US5342947A (en) 1992-10-09 1994-08-30 Glaxo Inc. Preparation of water soluble camptothecin derivatives
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
US5877219A (en) 1994-04-25 1999-03-02 Glaxo Wellcomeinc. Non-steroidal ligands for the estrogen receptor
US6063923A (en) 1994-05-03 2000-05-16 Glaxo Wellcome Inc. Preparation of a camptothecin derivative by intramolecular cyclisation
US5491237A (en) 1994-05-03 1996-02-13 Glaxo Wellcome Inc. Intermediates in pharmaceutical camptothecin preparation
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
WO2002059110A1 (fr) 2000-12-21 2002-08-01 Glaxo Group Limited Composes chimiques
WO2005121142A1 (fr) 2004-06-11 2005-12-22 Japan Tobacco Inc. Dérivés de 5-amino-2,4,7-trioxo-3,4,7,8-tétrahydro-2h-pyrido’2,3-d! pyrimidine et composés apparentés pour le traitement du cancer
US20060014768A1 (en) 2004-06-11 2006-01-19 Japan Tobacco Inc. Pyrimidine compound and medical use thereof
WO2007026920A2 (fr) * 2005-09-02 2007-03-08 Astellas Pharma Inc. Nouveaux composes
WO2008098104A1 (fr) 2007-02-07 2008-08-14 Smithkline Beecham Corporation Inhibiteurs de l'activité de akt
WO2011119663A1 (fr) * 2010-03-25 2011-09-29 Glaxosmithkline Llc Composés chimiques

Non-Patent Citations (113)

* Cited by examiner, † Cited by third party
Title
"Panobinostat, including pharmaceutically acceptable salts thereof", DRUGS OF THE FUTURE, vol. 32, no. 4, 2007, pages 315 - 322
"Tyrosine Kinase Signalling in Breast cancer:erbB Family Receptor Tyrosine Kniases", BREAST CANCER RES., vol. 2, no. 3, 2000, pages 176 - 183
15 September 2014, AMBINTER STOCK SCREENING COLLECTION *
4 September 2014, CHEMDIV SCREENING COLLECTION *
7 June 2014, AURORA SCREENING LIBRARY *
A. TENNANT, NATURE REVIEWS, 2010, pages 267
ABRAHAM, R.T., CURRENT OPINION IN IMMUNOLOGY, vol. 8, no. 3, 1996, pages 412 - 8
ADAMS J, KAUFFMAN M: "Bortezomib (Vecade®), including pharmaceutically acceptable salts thereof", CANCER INVEST, vol. 22, no. 2, 2004, pages 304 - 11
ALLI ET AL., ONCOGENE, vol. 24, 2005, pages 39 - 46
AMERI, K.; LEWIS, C. E.; RAIDA, M.; SOWTER, H.; HAI, T.; HARRIS, A. L.: "Anoxic induction of ATF-4 through HIF-1-independent pathways of protein stabilization in human cancer cells", BLOOD, vol. 103, 2004, pages 1876 - 82
ASHBY, M.N, CURRENT OPINION IN LIPIDOLOGY, vol. 9, no. 2, 1998, pages 99 - 102
BALASUBRAMANIAN ET AL.: "Mocetinostat (MGCD0103), including pharmaceutically acceptable salts thereof", CANCER LETTERS, vol. 280, 2009, pages 211 - 221
BALL ET AL., PROGRESS IN CELL CYCLE RES., vol. 3, 1997, pages 125
BERTRAND, EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 45, 2010, pages 2095 - 2116
BI, M.; NACZKI, C.; KORITZINSKY, M.; FELS, D.; BLAIS, J.; HU, N.; HARDING, H.; NOVOA, I.; VARIA, M.; RALEIGH, J. ET AL.: "ER stress-regulated translation increases tolerance to extreme hypoxia and promotes tumor growth", EMBO J, vol. 24, 2005, pages 3470 - 81
BIOCHIM. BIOPHYS. ACTA, vol. 1423, no. 3, 1989, pages 19 - 30
BLAIS, J.; BELL, J. C.: "Novel therapeutic target: the PERKs of inhibiting the integrated stress response", CELL CYCLE, vol. 5, 2006, pages 2874 - 7
BOLEN, J.B.; BRUGGE, J.S., ANNUAL REVIEW OF IMMUNOLOGY, vol. 15, 1997, pages 371 - 404
BOUMA ET AL.: "Disulfiram, including pharmaceutically acceptable salts thereof", J. ANTIMICROB. CHEMOTHER., vol. 42, no. 6, 1998, pages 817 - 20
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
BREWER, J. W.; DIEHL, J. A.: "PERK mediates cell-cycle exit during the mammalian unfolded protein response", PROC NATL ACAD SCI U S A, vol. 97, 2000, pages 12625 - 30
BRODT, P; SAMANI, A.; NAVAB, R., BIOCHEMICAL PHARMACOLOGY, vol. 60, 2000, pages 1101 - 1107
BROWN, J. M.; WILSON, W. R.: "Exploiting tumour hypoxia in cancer treatment", NAT REV CANCER, vol. 4, 2004, pages 437 - 47
BRUNS CJ ET AL., CANCER RES., vol. 60, 2000, pages 2926 - 2935
CANMAN, C.E.; LIM, D.S., ONCOGENE, vol. 17, no. 25, 1998, pages 3301 - 3308
CHEN Y; HU D; ELING DJ; ROBBINS J; KIPPS TJ., CANCER RES., vol. 58, 1998, pages 1965 - 1971
CULLINAN, S. B.; DIEHL, J. A.: "PERK-dependent activation of Nrf2 contributes to redox homeostasis and cell survival following endoplasmic reticulum stress", J BIOL CHEM, vol. 279, 2004, pages 20108 - 17
D. G. . KINGSTON ET AL.: "New trends in Natural Products Chemistry 1986", vol. 26, 1986, ELSEVIER, article "Studies in Organic Chemistry", pages: 219 - 235
DATABASE chemcats [online] Chemical Abstacts Service, Columbus Ohio, US; 25 February 2009 (2009-02-25), XP002734831, retrieved from STN Database accession no. F949-0110/ON *
DATABASE Chemcats [online] chemical abstacts service, Columbus, Ohio US; 29 February 2012 (2012-02-29), XP002734824, retrieved from STN Database accession no. Amb16264775/on *
DATABASE chemcats [online] chemical abstacts service, Columbus, Ohio, US; 25 February 2009 (2009-02-25), XP002734832, retrieved from STN Database accession no. F94/02341/ON *
DATABASE chemcats [online] Chemical abstacts service, Columbus, Ohio, US; 29 February 2012 (2012-02-29), XP002734826, retrieved from STN Database accession no. Amb16261182 *
DATABASE chemcats [online] chemical abstacts service, Columbus, Ohio, US; 29 February 2012 (2012-02-29), XP002734828, retrieved from STN Database accession no. K13.172.032/ON *
DATABASE chemcats [online] Chemical abstacts service, Columbus, Ohio; 25 February 2009 (2009-02-25), XP002734827, retrieved from STN Database accession no. Ko6.o24.120/ON *
DATABASE chemcats [online] Chemical Abstracts service, Columbus Ohio, US; 25 February 2009 (2009-02-25), XP002734829, retrieved from STN Database accession no. K06.024.132/ON *
DATABASE Chemcats [online] Chemical abstracts service, Columbus Ohio, US; 29 February 2012 (2012-02-29), XP002734825, retrieved from STN Database accession no. p667-0075/ON *
DATABASE chemcats [online] Chemical Abstracts service, Columbus, Ohio, US; 24 February 2009 (2009-02-24), XP002734830, retrieved from STN Database accession no. F949-0084/ON *
DAVIES, M. P.; BARRACLOUGH, D. L.; STEWART, C.; JOYCE, K. A.; ECCLES, R. M.; BARRACLOUGH, R.; RUDLAND, P. S.; SIBSON, D. R.: "Expression and splicing of the unfolded protein response gene XBP-1 are significantly associated with clinical outcome of endocrine-treated breast cancer", INT J CANCER, vol. 123, 2008, pages 85 - 8
DELEPINE, M.; NICOLINO, M.; BARRETT, T.; GOLAMAULLY, M.; LATHROP, G. M.; JULIER, C.: "EIF2AK3, encoding translation initiation factor 2-alpha kinase 3, is mutated in patients with Wolcott-Rallison syndrome", NAT GENET, vol. 25, 2000, pages 406 - 9
EINZIG, PROC. AM. SOC. CLIN. ONCOL., vol. 20, pages 46
FELDMAN, D. E.; CHAUHAN, V.; KOONG, A. C.: "The unfolded protein response: a novel component of the hypoxic stress response in tumors", MOL CANCER RES, vol. 3, 2005, pages 597 - 605
FELING: "Salinosporamide A, including pharmaceutically acceptable salts thereof", ANGEW. CHEM. INT. ED. ENGL., vol. 42, no. 3, 2003, pages 355 - 7
FORASTIRE, SEM. ONCOL., vol. 20, 1990, pages 56
GOTTLICHER ET AL.: "Valproic acid, including pharmaceutically acceptable salts thereof", EMBO J., vol. 20, no. 24, 2001, pages 6969 - 6978
GREEN, M.C. ET AL., MONOCLONAL ANTIBODY THERAPY FOR SOLID TUMORS, CANCER TREAT. REV., vol. 26, no. 4, 2000, pages 269 - 286
GUPTA, S.; MCGRATH, B.; CAVENER, D. R.: "PERK regulates the proliferation and development of insulin-secreting beta-cell tumors in the endocrine pancreas of mice", PLOS ONE, vol. 4, 2009, pages E8008
HAMANAKA, R. B.; BENNETT, B. S.; CULLINAN, S. B.; DIEHL, J. A.: "PERK and GCN2 contribute to el F2alpha phosphorylation and cell cycle arrest after activation of the unfolded protein response pathway", MOL BIOL CELL, vol. 16, 2005, pages 5493 - 501
HAMANAKA, R. B.; BOBROVNIKOVA-MARJON, E.; JI, X.; LIEBHABER, S. A.; DIEHL, J. A.: "PERK-dependent regulation of IAP translation during ER stress", ONCOGENE, vol. 28, 2009, pages 910 - 20
HARDING, H. P.; ZENG, H.; ZHANG, Y.; JUNGRIES, R.; CHUNG, P.; PLESKEN, H.; SABATINI, D. D.; RON, D.: "Diabetes mellitus and exocrine pancreatic dysfunction in perk-/- mice reveals a role for translational control in secretory cell survival", MOL CELL, vol. 7, 2001, pages 1153 - 63
HOLMES ET AL., J. NAT. CANCER INST., vol. 83, 1991, pages 1797
IGNOFF, R.J.: "Cancer Chemotherapy Pocket Guide", 1998
JACKSON, S.P., INTERNATIONAL JOURNAL OF BIOCHEMISTRY AND CELL BIOLOGY, vol. 29, no. 7, 1997, pages 935 - 8
JIA W ET AL.: "17-AAG(Geldanamydn), induding pharmaceutically acceptable salts thereof", BLOOD, vol. 102, no. 5, 1 September 2003 (2003-09-01), pages 1824 - 32
JORGENSEN, E.; STINSON, A.; SHAN, L.; YANG, J.; GIETL, D.; ALBINO, A. P.: "Cigarette smoke induces endoplasmic reticulum stress and the unfolded protein response in normal and malignant human lung cells", BMC CANCER, vol. 8, 2008, pages 229
KANEKURA, K.; SUZUKI, H.; AISO, S.; MATSUOKA, M., ER STRESS AND UNFOLDED PROTEIN RESPONSE IN AMYOTROPHIC LATERAL SCLEROSIS 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
KITADA S ET AL., ANTISENSE RES. DEV., vol. 4, 1994, pages 71 - 79
KOUMENIS, C.; NACZKI, C.; KORITZINSKY, M.; RASTANI, S.; DIEHL, A.; SONENBERG, N.; KOROMILAS, A.; WOUTERS, B. G.: "Regulation of protein synthesis by hypoxia via activation of the endoplasmic reticulum kinase PERK and phosphorylation of the translation initiation factor elF2alpha", MOL CELL BIOL, vol. 22, 2002, pages 7405 - 16
KOUMENIS, C.; WOUTERS, B. G.: "Translating'' tumor hypoxia: unfolded protein response (UPR)-dependent and UPR-independent pathways", MOL CANCER RES, vol. 4, 2006, pages 423 - 36
KUHN DJ ET AL.: "Carfilzomib, including pharmaceutically acceptable salts thereof", BLOOD, vol. 110, 2007, pages 3281 - 3290
KUMAR, J. BIOL, CHEM, vol. 256, 1981, pages 10435 - 10441
LACKEY, K. ET AL., BIOORGANIC AND MEDICINAL CHEMISTRY LETTERS, no. 10, 2000, pages 223 - 226
LEE ET AL., MOL CELL ENDOCRINOL., vol. 188, 2002, pages 47 - 54
LIDA, K.; LI, Y.; MCGRATH, B. C.; FRANK, A.; CAVENER, D. R.: "PERK eIF2 alpha kinase is required to regulate the viability of the exocrine pancreas in mice", BMC CELL BIOL, vol. 8, 2007, pages 38
LOFTS, F. J. ET AL.: "New Molecular Targets for Cancer Chemotherapy", 1994, CRC PRESS, article "Growth factor receptors as targets"
MA, K.; VATTEM, K. M.; WEK, R. C.: "Dimerization and release of molecular chaperone inhibition facilitate activation of eukaryotic initiation factor-2 kinase in response to endoplasmic reticulum stress", J BIOL CHEM, vol. 277, 2002, pages 18728 - 35
MA, Y.; HENDERSHOT, L. M.: "The role of the unfolded protein response in tumour development: friend or foe?", NAT REV CANCER, vol. 4, 2004, pages 966 - 77
MA, Y.; LU, Y.; ZENG, H.; RON, D.; MO, W.; NEUBERT, T. A.: "Characterization of phosphopeptides from protein digests using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry and nanoelectrospray quadrupole time-of-flight mass spectrometry", RAPID COMMUN MASS SPECTROM, vol. 15, 2001, pages 1693 - 700
MARCINIAK, S. J.; GARCIA-BONILLA, L.; HU, J.; HARDING, H. P.; RON, D.: "Activation-dependent substrate recruitment by the eukaryotic translation initiation factor 2 kinase PERK", J CELL BIOL, vol. 172, 2006, pages 201 - 9
MARKMAN ET AL., YALE JOURNAL OF BIOLOGY AND MEDICINE, vol. 64, 1991, pages 583
MARKS ET AL.: "Vorinostat, including pharmaceutically acceptable salts thereof", NATURE BIOTECHNOLOGY, vol. 25, 2007, pages 84 - 90
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.: "Diabetes, hyperglycemia and accelerated atherosclerosis: evidence supporting a role for endoplasmic reticulum (ER) stress signaling", CARDIOVASCULAR & HEMATOLOGICAL DISORDERS: DRUG TARGETS, vol. 10, no. 2, 2010, pages 151 - 157
MCGUIRE ET AL., ANN. INTEM, MED., vol. 111, 1989, pages 273
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
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: "Phosphorylation of the translation initiation factor eIF2a increases BACE1 levels and promotes amyloidogenesis", NEURON, vol. 60, no. 6, 2008, pages 988 - 1009
P. LEDER, CANCER CELL, vol. 9, 2006, pages 425
PASCHEN, W.: "Endoplasmic reticulum dysfunction in brain pathology: Critical role of protein synthesis", CURRENT NEUROVASCULAR RESEARCH, vol. 1, no. 2, 2004, pages 173 - 181
PEARCE, L.R ET AL., NATURE REVIEWS MOLECULAR CELL BIOLOGY, vol. 11, 2010, pages 9 - 22
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
REILLY RT ET AL., CANCER RES., vol. 60, 2000, pages 3569 - 3576
RICHON ET AL., PROC. NAT ACAD. SCI. U.S.A., vol. 97, no. 18, 2000, pages 10014 - 10019
ROMERO-RAMIREZ, L.; CAO, H.; NELSON, D.; HAMMOND, E.; LEE, A. H.; YOSHIDA, H.; MORI, K.; GLIMCHER, L. H.; DENKO, N. C.; GIACCIA, A: "XBP1 is essential for survival under hypoxic conditions and is required for tumor growth", CANCER RES, vol. 64, 2004, pages 5943 - 7
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.;: "The unfolded protein response protects human tumor cells during hypoxia through regulation of the autophagy genes MAP1LC3B and ATG5", J CLIN INVEST, vol. 120, pages 127 - 41
SALMINEN, A.; KAUPPINEN, A.; SUURONEN, T.; KAARNIRANTA, K.; OJALA, J.: "ER stress in Alzheimer's disease: a novel neuronal trigger for inflammation and Alzheimer's pathology", 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
SCHIFF ET AL., NATURE, vol. 277, 1979, pages 665 - 667
SCHIFF ET AL., PROC. NATL, ACAD, SCI. USA, vol. 77, 1980, pages 1561 - 1565
SCHREIBER AB; WINKLER ME; DERYNCK R, SCIENCE, vol. 232, 1986, pages 1250 - 1253
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.: "Identification and characterization of pancreatic eukaryotic initiation factor 2 alpha-subunit kinase, PEK, involved in translational control", 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: "Pancreatic eukaryotic initiation factor-2alpha kinase (PEK) homologues in humans, Drosophila melanogaster and Caenorhabditis elegans that mediate translational control in response to endoplasmic reticulum stress", BIOCHEM J, vol. 346, 2000, pages 281 - 93
STENGER, COMMUNITY ONCOLOGY, vol. 4, 2007, pages 384 - 386
SU, Q.; WANG, S.; GAO, H. Q.; KAZEMI, S.; HARDING, H. P.; RON, D.; KOROMILAS, A. E.: "Modulation of the eukaryotic initiation factor 2 alpha-subunit kinase PERK by tyrosine phosphorylation", J BIOL CHEM, vol. 283, 2008, pages 469 - 75
T. GREENE; P. WUTS: "Protecting Groups in Chemical Synthesis", 1999, JOHN WILEY & SONS
TABAS, I.; SEIMON, T.; TIMMINS, J.; LI, G.; LIM, W.: "Macrophage apoptosis in advanced atherosclerosis", ANNALS OF THE NEW YORK ACADEMY OF SCIENCES, vol. 1173, no. S1, 2009, pages E40 - E45
V.T. DEVITA AND S. HELLMAN: "Cancer Principles and Practice of Oncology, 6th edition", 15 February 2001, LIPPINCOTT WLLIAMS & WLKINS PUBLISHERS
VIGUSHIN ET AL., ANTICANCER DRUGS, vol. 13, no. 1, January 2002 (2002-01-01), pages 1 - 13
VINODHKUMAR ET AL.: "Romidepsin, including pharmaceutically acceptable salts thereof", BIOMEDICINE & PHARMACOTHERAPY, vol. 62, 2008, pages 85 - 93
WATER JS ET AL., J. CLIN. ONCOL., vol. 18, 2000, pages 1812 - 1823
WLLIAMSON ET AL.: "Epigallocatechin gallate (EGCG), including pharmaceutically acceptable salts thereof", THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY, vol. 118, no. 6, December 2006 (2006-12-01), pages 1369 - 74
WOO, NATURE, vol. 368, 1994, pages 750
YAMAMOTO, T.; TAYA, S.; KAIBUCHI, K., JOURNAL OF BIOCHEMISTRY, vol. 126, no. 5, 1999, pages 799 - 803
YEN L ET AL., ONCOGENE, vol. 19, 2000, pages 3460 - 3469
ZHANG, W.; FENG, D.; LI, Y.; LIDA, K.; MCGRATH, B.; CAVENER, D. R.: "PERK EIF2AK3 control of pancreatic beta cell differentiation and proliferation is required for postnatal glucose homeostasis", CELL METAB, vol. 4, 2006, pages 491 - 7
ZHONG, H. ET AL., CANCER RES, vol. 60, no. 6, 2000, pages 1541 - 1545

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US11026945B2 (en) 2016-04-29 2021-06-08 The Trustees Of The University Of Pennsylvania Protein kinase RNA-like endoplasmic reticulum kinase (PERK) inhibitors for prevention and/or treatment of lung injury and/or inflammation
US10918650B2 (en) * 2016-06-02 2021-02-16 University Of South Florida Method of treating melanoma using an inhibitor of an atypical protein kinase C
US11065266B2 (en) 2016-06-02 2021-07-20 University Of South Florida Method of treating melanoma using an inhibitor of an atypical protein kinase C
CN115645535B (en) * 2017-03-06 2024-07-09 坪田实验室股份有限公司 Mouse myopia induction model and endoplasmic reticulum stress inhibitor for myopia prevention/inhibition
CN115645535A (zh) * 2017-03-06 2023-01-31 坪田实验室股份有限公司 小鼠近视诱导模型及用于近视预防/抑制的内质网应激抑制剂
US11655214B2 (en) 2017-04-18 2023-05-23 Eli Lilly And Company Phenyl-2-hydroxy-acetylamino-2-methyl-phenyl compounds for the treatment of pancreatic cancer
US10654828B2 (en) 2017-07-28 2020-05-19 Novartis Ag Indole derivatives and uses thereof
US10633350B2 (en) 2017-09-13 2020-04-28 Novartis Ag Diphenyl derivatives and uses thereof
CN107898793A (zh) * 2017-12-01 2018-04-13 温州医科大学 一种抑制近视的方法及制备药物的应用
CN107898793B (zh) * 2017-12-01 2019-12-24 温州医科大学 一种抑制近视的方法及制备药物的应用
WO2020070053A1 (fr) 2018-10-01 2020-04-09 INSERM (Institut National de la Santé et de la Recherche Médicale) Utilisation d'inhibiteurs de formation de granules de stress pour cibler la régulation de réponses immunitaires
WO2021041976A1 (fr) * 2019-08-29 2021-03-04 Hibercell, Inc. Composés indolinyle inhibiteurs de perk
WO2021083959A1 (fr) 2019-10-29 2021-05-06 INSERM (Institut National de la Santé et de la Recherche Médicale) Procédés et compositions pour le traitement d'un mélanome uvéal

Also Published As

Publication number Publication date
WO2015056180A8 (fr) 2015-12-03

Similar Documents

Publication Publication Date Title
EP2549868B1 (fr) Composés chimiques
US8598156B2 (en) Chemical compounds
WO2015056180A1 (fr) Dérivés d'indoline utilisés comme inhibiteurs de perk
ES2501518T3 (es) Inhibidores de la actividad de Akt
EP2635279A2 (fr) Composés chimiques
JP5363997B2 (ja) Akt活性の阻害剤
WO2017098421A1 (fr) Composés benzothiadiazine
EP1653961A1 (fr) Inhibiteurs de l'activite de akt
JP2012509335A (ja) 化合物
WO2013096151A1 (fr) Composés chimiques
KR20120133386A (ko) 지방산 신타제 억제제로서의 트리아졸론
WO2019049061A1 (fr) Dérivés de 5-(1 h-benzo[d]imidazo-2-yl)-pyridin-2-amine et de 5-(3h-imidazo[4,5-b]pyridin-6-yl)-pyridin-2-amine utilisés en tant qu'inhibiteurs d'histone acétyltransférase de c-myc et p300/cbp pour le traitement du cancer
WO2013095761A1 (fr) Dérivés d'imidizopyridine comme inhibiteurs de la pi3 kinase
EP2663311A2 (fr) Dérivés de pyrimidinone en tant qu'inhibiteurs de l'acide gras synthase
WO2010120854A1 (fr) Composés chimiques
WO2013028445A1 (fr) Inhibiteurs d'acide gras synthase
JP2013508461A (ja) 脂肪酸シンターゼ阻害剤としてのベンズイミダゾール
WO2019053617A1 (fr) Composés chimiques
JP5554833B2 (ja) Pi3キナーゼ阻害剤としてのチアゾロピリミジノン誘導体
WO2013096153A1 (fr) Composés chimiques

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: 14802945

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: 14802945

Country of ref document: EP

Kind code of ref document: A1