1,093
Views
38
CrossRef citations to date
0
Altmetric
Review

Tropomyosin receptor kinase inhibitors: an updated patent review for 2010-2016 – Part II

, , &
Pages 831-849 | Received 21 Oct 2016, Accepted 17 Feb 2017, Published online: 08 Mar 2017

References

  • Huang EJ, Reichardt LF. Neurotrophins: roles in neuronal development and function. Annu Rev Neurosci. 2001;24(1):677–736.
  • Klein R, Jing S, Nanduri V, et al. The Trk proto-oncogene encodes a receptor for nerve growth factor. Cell. 1991;65(1):189–197.
  • Kaplan DR, Martin-Zanca D, Parada LF. Tyrosine phosphorylation and tyrosine kinase activity of the Trk proto-oncogene product induced by NGF. Nature. 1991;350(6314):158–160.
  • Klein R, Nanduri V, Jing S, et al. The TrkB tyrosine protein kinase is a receptor for brain-derived neurotrophic factor and neurotrophin-3. Cell. 1991;66(2):395–403.
  • Lamballe F, Klein R, Barbacid M. TrkC, a new member of the Trk family of tyrosine protein kinases, is a receptor for neurotrophin-3. Cell. 1991;66(5):967–979.
  • Bailey JJ, Schirrmacher R, Farrell K, et al. Tropomyosin receptor kinase inhibitors: an updated patent review for 2010-2016 – Part I. Expert Opin Ther Pat. 2017. doi:10.1080/13543776.2017.1297796.
  • Stachel SJ, Sanders JM, Henze DA, et al. Maximizing diversity from a kinase screen: identification of novel and selective pan-Trk inhibitors for chronic pain. J Med Chem. 2014;57(13):5800–5816.
  • Su H-P, Rickert K, Burlein C, et al. Structural characterization of nonactive site, TrkA-selective kinase inhibitors. Proc Natl Acad Sci. 2017;14(3):E297–E306.
  • Shelton DL, Pons J, Rosenthal A. Anti-NGF antibodies and methods using same. WO2004058184. 2004.
  • Lane NE, Schnitzer TJ, Birbara CA, et al. Tanezumab for the treatment of pain from osteoarthritis of the knee. N Engl J Med. 2010;363(16):1521–1531.
  • Pavone F, Marinelli S, Cattaneo A, et al. Novel analgesic treatment with prolonged effect. WO2006131952. 2008.
  • Green A, Li Y, Stachel S. TrkA kinase inhibitors, compositions and methods thereof. WO2012125668. 2012.
  • Green A, Li Y, Stachel S. TrkA kinase inhibitors, compositions and methods thereof. WO2012125667. 2012.
  • Choi H-S, Rucker PV, Wang Z, et al. (R)-2-phenylpyrrolidine substituted imidazopyridazines: a new class of potent and selective pan-TRK inhibitors. ACS Med Chem Lett. 2015;6(5):562–567.
  • Hanney B, Manley P, Rudd MT, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2013009582. 2013.
  • Stachel SJ, Egbertson M, Brnardic E, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2013176970. 2013.
  • Cooke AJ, Stump CA, Zhang XF, et al. TrkA kinase inhibitor, compositions and methods thereof. WO2015042088. 2015.
  • Cooke AJ, Stump CA, Zhang XF, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015042085. 2015.
  • Allen S, Brandhuber BJ, Condroski KR, et al. Bicyclic heteroaryl urea, thiourea, guanidine and cyanoguanidine compounds as TrkA kinase inhibitors. WO2014078408 A1. 2014.
  • Mitchell H, Fraley ME, Cooke AJ, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015143654. 2015.
  • Mitchell H, Fraley ME, Cooke AJ, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015148344. 2015.
  • Mitchell H, Fraley ME, Cooke AJ, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015148350. 2015.
  • Mitchell H, Fraley ME, Cooke AJ, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015148373. 2015.
  • Mitchell H, Wood HB, Li CS, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2015148354. 2015.
  • McElroy WT, Tan Z, Ho G, et al. Potent and selective amidopyrazole inhibitors of IRAK4 that are efficacious in a rodent model of inflammation. ACS Med Chem Lett. 2015;6(6):677–682.
  • Cooke AJ, Pitts D, Johnson A, et al. TrkA kinase inhibitors, compositions and methods thereof. WO2016054807. 2016.
  • Guo L, Parker DL, Zang Y, et al. Discovery and optimization of a novel triazole series of GPR142 agonists for the treatment of type 2 diabetes. ACS Med Chem Lett. 2016;7(12):1107–1111.
  • Brasca MG, Amboldi N, Ballinari D, et al. Identification of N,1,4,4-tetramethyl-8-{[4-(4-methylpiperazin-1-yl)phenyl]amino}-4,5-dihydro-1H-pyrazolo[4,3-h]quinazoline-3-carboxamide (PHA-848125), a potent, orally available cyclin dependent kinase inhibitor. J Med Chem. 2009;52(16):5152–5163.
  • Scaburri A, Pacciarini MA, Ciomei M, et al. Use of a kinase inhibitor for the treatment of thymoma. WO2010106028 A1. 2010.
  • Albanese C, Alzani R, Amboldi N, et al. Anti-tumour efficacy on glioma models of PHA-848125, a multi-kinase inhibitor able to cross the blood–brain barrier. Br J Pharmacol. 2013;169(1):156–166.
  • Weiss GJ, Hidalgo M, Borad MJ, et al. Phase I study of the safety, tolerability and pharmacokinetics of PHA-848125AC, a dual tropomyosin receptor kinase A and cyclin-dependent kinase inhibitor, in patients with advanced solid malignancies. Invest New Drugs. 2012;30(6):2334–2343.
  • Barbugian NA, Forino R, Fumagalli T, et al. Process for the preparation of N-[5-(3,5-difluoro-benzyl)-1H-indazol-3-yl]-4-(4-methyl-piperazin-1-yl)-2-(tetrahydro-pyran-4-ylamino)-benzamide. WO2013174876 A1. 2013.
  • Menichincheri M, Ardini E, Magnaghi P, et al. Discovery of entrectinib: a new 3-aminoindazole as a potent anaplastic lymphoma kinase (ALK), c-ros oncogene 1 kinase (ROS1), and pan-tropomyosin receptor kinases (pan-TRKs) inhibitor. J Med Chem. 2016;59(7):3392–3408.
  • Takeuchi J, Ikura M, Higashino M, et al. Trk-inhibiting compound. WO2013161919 A1. 2013.
  • Takeuchi J, Itadani S, Hashimura K, et al. Trk-inhibiting compound. WO2014129431 A1. 2014.
  • Dumas J, Khire U, Lowinger TB, et al. Inhibition of RAF kinase using substituted heterocyclic ureas. WO1999032106 A1. 1999.
  • Lang H, Gahman TC, Herbert MR, et al. Substituted pyrimidinyloxy ureas useful as inhibitors of protein kinases. WO2007076473 A2. 2007.
  • Albaugh P, Fan Y, Mi Y, et al. Discovery of GNF-5837, a selective TRK inhibitor with efficacy in rodent cancer tumor models. ACS Med Chem Lett. 2012;3(2):140–145.
  • Andrews MD, Bagal SK, Gibson KR, et al. Pyrrolo [2, 3-d] pyrimidine derivatives as inhibitors of tropomyosin-related kinases. WO2012137089 A1. 2012.
  • Marx MA, La GSD, Chen J, et al. Pyrrolopyrimidine derivatives useful in cancer treatment. WO2005116035 A1. 2005.
  • Andrews MD, Bagal SK, Brown DG, et al. Pyrrolo[2,3-d]pyrimidine tropomyosin-related kinase inhibitors. WO2014053967 A1. 2014.
  • Skerratt SE, Andrews M, Bagal SK, et al. The discovery of a potent, selective, and peripherally restricted pan-Trk inhibitor (PF-06273340) for the treatment of pain. J Med Chem. 2016;59(22):10084–10099.
  • Andrews MD, Bagal SK, Brown DG, et al. Pyrrolo[3,2-c]pyridine tropomyosin-related kinase inhibitors. WO2014053968 A1. 2014.
  • Andrews MD, Bagal SK, Skerratt SE. Tropomyosin-related kinase inhibitors. WO2014053965 A1. 2014.
  • Skerratt SE, Bagal SK, Swain NA, et al. N-acylpiperidine ether tropomyosin-related kinase inhibitors. WO2015092610 A1. 2015.
  • Bagal SK, Omoto K, Skerratt SE, et al. N-acylpiperidine ether tropomyosin-related kinase inhibitors. WO2016009296 A1. 2016.
  • Bagal SK, Omoto K, Skerratt SE, et al. N-acylpyrrolidine ether tropomyosin-related kinase inhibitors. WO2016020784 A1. 2016.
  • Bagal SK, Cui JJ, Greasley SE, et al. Tropomyosin-related kinase inhibitors. WO2015170218 A1. 2015.
  • Bagal SK, Cui JJ, Greasley SE, et al. Tropomyosin-related kinase inhibitors containing both a 1H-pyrazole and a pyrimidine moiety. WO2013132376 A1. 2015.
  • Kruczynski A, Creancier L, Kaloun EB, et al. Derivatives of azaindazole or diazaindazole type for treating a cancer overexpressing Trk. WO2014016434 A1. 2014.
  • Lombardi BA, Menichincheri M, Orsini P, et al. Substituted indazole derivatives active as kinase inhibitors. WO2009013126 A1. 2009.
  • Zhang C, Zhang J, Ibrahim PN, et al. Compounds modulating c-fms and/or c-kit activity and uses therefor. WO2008063888 A2. 2008.
  • Visor GC, Ibrahim PN, Spevak W, et al. Solid forms of sulfonamides and amino acids. WO2010129570 A1. 2010.
  • Sasmal PK, Ahmed S, Prabhu G, et al. Substituted heterocyclic compounds as tropomyosin receptor kinase a (TrkA) inhibitors. WO2013088257 A1. 2013.
  • Molteni V, Fan Y, Loren J, et al. Compounds and compositions as Trk inhibitors. WO2012116217 A1. 2012.
  • Sasmal PK, Ahmed S, Tehim A, et al. Substituted pyrazolo[1,5-a] pyridine as tropomyosin receptor kinase (Trk) inhibitors. WO2013088256 A1. 2013.
  • Sasmal PK, Ahmed S, Tehim A, et al. Substituted imidazo[1,2-a]pyridine compounds useful for the treatment of pain. WO2015200341 A1. 2015.
  • Wu JJQ, Wang L Compositions of protein receptor tyrosine kinase inhibitors. WO2010077680 A2. 2010.
  • Wu JJQ, Wang L Crystalline forms of tyrosine kinase inhibitors and their salts. WO2014152663 A1. 2014.
  • Pulciani S, Santos E, Lauver AV, et al. Oncogenes in solid human tumours. Nature. 1982;300(5892):539–542.
  • Martin-Zanca D, Hughes SH, Barbacid M. A human oncogene formed by the fusion of truncated tropomyosin and protein tyrosine kinase sequences. Nature. 1986;319(6056):743–748.
  • Nakagawara A. Trk receptor tyrosine kinases: a bridge between cancer and neural development. Cancer Lett. 2001;169(2):107–114.
  • Brodeur GM. Neuroblastoma: biological insights into a clinical enigma. Nat Rev Cancer. 2003;3(3):203–216.
  • Morrison KB, Tognon CE, Garnett MJ, et al. ETV6-NTRK3 transformation requires insulin-like growth factor 1 receptor signaling and is associated with constitutive IRS-1 tyrosine phosphorylation. Oncogene. 2002;21(37):5684–5695.
  • Douma S, van Laar T, Zevenhoven J, et al. Suppression of anoikis and induction of metastasis by the neurotrophic receptor TrkB. Nature. 2004;430(7003):1034–1039.
  • Laurent D, Eric A, Ikram El Y-B, et al. Nerve growth factor receptors and signaling in breast cancer. Curr Cancer Drug Targets. 2004;4(6):463–470.
  • Weeraratna AT, Dalrymple SL, Lamb JC, et al. Pan-Trk inhibition decreases metastasis and enhances host survival in experimental models as a result of its selective induction of apoptosis of prostate cancer cells. Clin Cancer Res. 2001;7(8):2237–2245.
  • Nakagawara A, Arima-Nakagawara M, Scavarda NJ, et al. Association between high levels of expression of the TRK gene and favorable outcome in human neuroblastoma. N Engl J Med. 1993;328(12):847–854.
  • Schramm A, Schulte JH, Astrahantseff K, et al. Biological effects of TrkA and TrkB receptor signaling in neuroblastoma. Cancer Lett. 2005;228(1–2):143–153.
  • Tacconelli A, Farina AR, Cappabianca L, et al. TrkA alternative splicing: a regulated tumor-promoting switch in human neuroblastoma. Cancer Cell. 2004;6(4):347–360.
  • Reuther GW, Lambert QT, Caligiuri MA, et al. Identification and characterization of an activating TrkA deletion mutation in acute myeloid leukemia. Mol Cell Biol. 2000;20(23):8655–8666.
  • Shabbir M, Stuart R. Lestaurtinib, a multitargeted tyrosinse kinase inhibitor: from bench to bedside. Expert Opin Investig Drugs. 2010;19(3):427–436.
  • Evans AE, Kisselbach KD, Yamashiro DJ, et al. Antitumor activity of CEP-751 (KT-6587) on human neuroblastoma and medulloblastoma xenografts. Clin Cancer Res. 1999;5(11):3594–3602.
  • Evans AE, Kisselbach KD, Liu X, et al. Effect of CEP-751 (KT-6587) on neuroblastoma xenografts expressing TrkB. Med Pediatr Oncol. 2001;36(1):181–184.
  • Iyer R, Evans AE, Qi X, et al. Lestaurtinib enhances the antitumor efficacy of chemotherapy in murine xenograft models of neuroblastoma. Clin Cancer Res. 2010;16(5):1478–1485.
  • Minturn JE, Evans AE, Villablanca JG, et al. Phase I trial of lestaurtinib for children with refractory neuroblastoma: a new approaches to neuroblastoma therapy consortium study. Cancer Chemother Pharmacol. 2011;68(4):1057–1065.
  • Wang T, Yu D, Lamb ML. Trk kinase inhibitors as new treatments for cancer and pain. Expert Opin Ther Pat. 2009;19(3):305–319.
  • Ardini E, Bosotti R, Borgia AL, et al. The TPM3-NTRK1 rearrangement is a recurring event in colorectal carcinoma and is associated with tumor sensitivity to TRKA kinase inhibition. Mol Oncol. 2014;8(8):1495–1507.
  • Rubin BP, Chen C-J, Morgan TW, et al. Congenital mesoblastic nephroma t(12;15) is associated withETV6-NTRK3 gene fusion: cytogenetic and molecular relationship to congenital (infantile) fibrosarcoma. Am J Pathol. 1998;153(5):1451–1458.
  • Euhus DM, Timmons CF, Tomlinson GE. ETV6-NTRK3 – Trk-ing the primary event in human secretory breast cancer. Cancer Cell. 2002;2(5):347–348.
  • Vaishnavi A, Le A, Doebele RC. TRKing down an old oncogene in a new era of targeted therapy. Cancer Discov. 2015;5(1):25–34.
  • Passiglia F, Caparica R, Giovannetti E, et al. The potential of neurotrophic tyrosine kinase (NTRK) inhibitors for treating lung cancer. Expert Opin Investig Drugs. 2016;25(4):385–392.
  • De Braud F, Niger M, Damian S, et al. Alka-372–001: First-in-human,phase I study of entrectinib – an oral pan-Trk, ROS1, and ALK inhibitor – in patients with advanced solid tumors with relevant molecular alterations. J Clin Oncol. 2015;33:suppl; abstr 2517. page 121s.
  • Patel MR, Bauer TM, Liu SV, et al. STARTRK-1: phase 1/2a study of entrectinib, an oral pan-Trk, ROS1, and ALK inhibitor, in patients with advanced solid tumors with relevant molecular alterations. J Clin Oncol. 2015;33:suppl; abstr 2596. page 140s.
  • Burris HA, Brose MS, Shaw AT, et al. A first-in-human study of LOXO-101, a highly selective inhibitor of the tropomyosin receptor kinase (TRK) family. J Clin Oncol. 2015;33:suppl TPS2624. page 147s.
  • Vaishnavi A, Capelletti M, Le AT, et al. Oncogenic and drug-sensitive NTRK1 rearrangements in lung cancer. Nat Med. 2013;19(11):1469–1472.
  • Ardini E, Menichincheri M, Banfi P, et al. Entrectinib, a pan-TRK, ROS1 and ALK inhibitor with activity in multiple molecularly defined cancer indications. Mol Cancer Ther. 2016;15(4):628–629.
  • Iyer R, Wehrmann L, Golden RL, et al. Entrectinib is a potent inhibitor of Trk-driven neuroblastomas in a xenograft mouse model. Cancer Lett. 2016;372(2):179–186.
  • Sartore-Bianchi A, Ardini E, Bosotti R, et al. Sensitivity to entrectinib associated with a novel LMNA-NTRK1 gene fusion in metastatic colorectal cancer. J Natl Cancer Inst. 2016;108:1.
  • Drilon A, Li G, Dogan S, et al. What hides behind the MASC: clinical response and acquired resistance to entrectinib after ETV6-NTRK3 identification in a mammary analogue secretory carcinoma (MASC). Ann Oncol. 2016;27(5):920–926.
  • Farago AF, Le LP, Zheng Z, et al. Durable clinical response to entrectinib in NTRK1-rearranged non-small cell lung cancer. J Thorac Oncol. 2015;10(12):1670–1674.
  • Russo M, Misale S, Wei G, et al. Acquired resistance to the TRK inhibitor entrectinib in colorectal cancer. Cancer Discov. 2016;6(1):36–44.
  • Hong DS, Brose MS, Doebele RC, et al. Abstract PR13: clinical safety and activity from a phase 1 study of LOXO-101, a selective TRKA/B/C inhibitor, in solid-tumor patients with NTRK gene fusions. Mol Cancer Ther. 2015;14(12Supplement 2):PR13–PR13.
  • Doebele RC, Davis LE, Vaishnavi A, et al. An oncogenic NTRK fusion in a patient with soft-tissue sarcoma with response to the tropomyosin-related kinase inhibitor LOXO-101. Cancer Discov. 2015;5(10):1049–1057.
  • Nagasubramanian R, Wei J, Gordon P, et al. Infantile fibrosarcoma with NTRK3–ETV6 fusion successfully treated with the tropomyosin-related kinase inhibitor LOXO-101. Pediatr Blood Cancer. 2016;63(8):1468–1470.
  • Yeh I, Tee MK, Botton T, et al. NTRK3 kinase fusions in Spitz tumours. J Pathol. 2016;240(3):282-290.
  • Lewis RT, Bode CM, Choquette DM, et al. The discovery and optimization of a novel class of potent, selective, and orally bioavailable anaplastic lymphoma kinase (ALK) inhibitors with potential utility for the treatment of cancer. J Med Chem. 2012;55(14):6523–6540.
  • Arkenau H-T, Sachdev JC, Mita MM, et al. Phase (Ph) 1/2a study of TSR-011, a potent inhibitor of ALK and TRK, in advanced solid tumors including crizotinib-resistant ALK positive non-small cell lung cancer. J Clin Oncol. 2015;33:suppl; abstr 8063. page 437s.
  • Watson JJ, Allen SJ, Dawbarn D. Targeting nerve growth factor in pain. Biodrugs. 2008;22(6):349–359.
  • McKelvey L, Shorten GD, O’Keeffe GW. Nerve growth factor-mediated regulation of pain signalling and proposed new intervention strategies in clinical pain management. J Neurochem. 2013;124(3):276–289.
  • Chang DS, Hsu E, Hottinger DG, et al. Anti-nerve growth factor in pain management: current evidence. J Pain Res. 2016;9:373–383.
  • Lieberman H, Yang D, Philbrook CM, et al. Pharmaceutical formulations of tropomyosin related kinase (Trk) inhibitors. WO2016100677 A2. 2016.
  • Redig AJ, Jänne PA. Basket trials and the evolution of clinical trial design in an era of genomic medicine. J Clin Oncol. 2015;33(9):975–977.
  • Andrews SW. Allosteric small molecule inhibitors of the NGF/TrkA pathway, a new approach to treating inflammatory pain. [Internet]. Boulder (CO): Array BioPharma Inc; 2012 [cited 2016 Aug 17]; Available from: http://arraybiopharma.com/files/6313/9810/8021/PubAttachment587.pdf
  • Sanga P, Katz N, Polverejan E, et al. Efficacy, safety, and tolerability of fulranumab, an anti-nerve growth factor antibody, in the treatment of patients with moderate to severe osteoarthritis pain. Pain. 2013;154(10):1910–1919.
  • Tiseo PJ, Kivitz AJ, Ervin JE, et al. Fasinumab (REGN475), an antibody against nerve growth factor for the treatment of pain: results from a double-blind, placebo-controlled exploratory study in osteoarthritis of the knee. Pain. 2014;155:1245–1252.
  • Leite VF, Buehler AM, El Abd O, et al. Anti-nerve growth factor in the treatment of low back pain and radiculopathy: a systematic review and a meta-analysis. Pain Physician. 2014;17(1):E45–60.
  • Hochberg MC, Tive LA, Abramson SB, et al. When is osteonecrosis not osteonecrosis?: adjudication of reported serious adverse joint events in the tanezumab clinical development program. Arthritis Rheumatol. 2016;68(2):382–391.
  • Elkins JM, Fedele V, Szklarz M, et al. Comprehensive characterization of the published kinase inhibitor set. Nat Biotech. 2016;34(1):95–103.
  • Davis MI, Hunt JP, Herrgard S, et al. Comprehensive analysis of kinase inhibitor selectivity. Nat Biotech. 2011;29(11):1046–1051.
  • Anastassiadis T, Deacon SW, Devarajan K, et al. Comprehensive assay of kinase catalytic activity reveals features of kinase inhibitor selectivity. Nat Biotech. 2011;29(11):1039–1045.
  • Regan J, Pargellis CA, Cirillo PF, et al. The kinetics of binding to p38 MAP kinase by analogues of BIRB 796. Bioorg Med Chem Lett. 2003;13(18):3101–3104.
  • Pargellis C, Tong L, Churchill L, et al. Inhibition of p38 MAP kinase by utilizing a novel allosteric binding site. Nat Struct Mol Biol. 2002;9(4):268–272.
  • Perreault M, Feng G, Will S, et al. Activation of TrkB with TAM-163 results in opposite effects on body weight in rodents and non-human primates. PLoS One. 2013;8(5):e62616.
  • McCarthy C, Walker E. Tropomyosin receptor kinase inhibitors: a patent update 2009 – 2013. Expert Opin Ther Pat. 2014;24(7):731–744.
  • Heffron TP. Small molecule kinase inhibitors for the treatment of brain cancer. J Med Chem. 2016;59(22):10030–10066.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.