1,042
Views
52
CrossRef citations to date
0
Altmetric
Review

A patent review of histone deacetylase 6 inhibitors in neurodegenerative diseases (2014-2019)

ORCID Icon & ORCID Icon
Pages 121-136 | Received 14 Nov 2019, Accepted 20 Dec 2019, Published online: 25 Dec 2019

References

  • Wade PA, Pruss D, Wolffe AP. Histone acetylation: chromatin in action. Trends Biochem Sci. 1997;22:128–132.
  • Egger G, Liang G, Aparicio A, et al. Epigenetics in human disease and prospects for epigenetic therapy. Nature. 2004;429:457–463.
  • Kelly TK, De Carvalho DD, Jones PA. Epigenetic modifications as therapeutic targets. Nat Biotechnol. 2010;28:1069–1078.
  • Rivera CM, Ren B. Mapping human epigenomes. Cell. 2013;155:39–55.
  • d’Ydewalle C, Bogaert E, Van Den Bosch L. HDAC6 at the intersection of neuroprotection and neurodegeneration. Traffic. 2012;13:771–779.
  • Prior R, Van Helleputte L, Klingl YE, et al. HDAC6 as a potential therapeutic target for peripheral nerve disorders. Expert Opin Ther Targets. 2018;22:993–1007.
  • Brindisi M, Saraswati AP, Brogi S, et al. Old but gold: tracking the new guise of histone deacetylase 6 (HDAC6) enzyme as a biomarker and therapeutic target in rare diseases. J Med Chem. 2019: published online. 2019 Aug 15. DOI:10.1021/acs.jmedchem.9b00924.
  • Shen S, Kozikowski AP. Why hydroxamates may not be the best histone deacetylase inhibitors–what some may have forgotten or would rather forget? ChemMedChem. 2016;11:15–21.
  • Wang XX, Wan RZ, Liu ZP. Recent advances in the discovery of potent and selective HDAC6 inhibitors. Eur J Med Chem. 2018;143:1406–1418.
  • Yang FF, Zhao N, Ge D, et al. Next-generation of selective histone deacetylase inhibitors. RSC Adv. 2019;9:19571–19583.
  • Faria Freitas M, Cuendet M, Bertrand P. HDAC inhibitors: a 2013–2017 patent survey. Expert Opin Ther Pat. 2018;28:1–17.
  • Chen S, Owens GC, Makarenkova H, et al. HDAC6 regulates mitochondrial transport in hippocampal neurons. PLoS One. 2010;5:e10848.
  • Liu XA, Rizzo V, Puthanveettil SV. Pathologies of axonal transport in neurodegenerative diseases. Transl Neurosci. 2012;3:355–372.
  • Dou F, Netzer WJ, Tanemura K, et al. Chaperones increase association of tau protein with microtubules. Proc Natl Acad Sci USA. 2003;100:721–726.
  • Cook C, Gendron TF, Scheffel K, et al. Loss of HDAC6, a novel CHIP substrate, alleviates abnormal tau accumulation. Hum Mol Genet. 2012;21:2936–2945.
  • Trachootham D, Lu W, Ogasawara MA, et al. Redox regulation of cell survival. Antioxid Redox Signal. 2008;10:1343–1374.
  • Parmigiani RB, Xu WS, Venta-Perez G, et al. HDAC6 is a specific deacetylase of peroxiredoxins and is involved in redox regulation. Proc Natl Acad Sci USA. 2008;105:9633–9638.
  • Jian W, Wei X, Chen L, et al. Inhibition of HDAC6 increases acetylation of peroxiredoxin1/2 and ameliorates 6-OHDA induced dopaminergic injury. Neurosci Lett. 2017;658:114–120.
  • Seigneurin-Berny D, Verdel A, Curtet S, et al. Identification of components of the murine histone deacetylase 6 complex: link between acetylation and ubiquitination signaling pathways. Mol Cell Biol. 2001;21:8035–8044.
  • Wu Y, Song SW, Sun J, et al. IIp45 inhibits cell migration through inhibition of HDAC6. J Biol Chem. 2010;285:3554–3560.
  • Deribe YL, Wild P, Chandrashaker A, et al. Regulation of epidermal growth factor receptor trafficking by lysine deacetylase HDAC6. Sci Signal. 2009;2:ra84.
  • Ding H, Dolan PJ, Johnson GV. Histone deacetylase 6 interacts with the microtubule-associated protein tau. J Neurochem. 2008;106:2119–2130.
  • Simoes-Pires C, Zwick V, Nurisso A, et al. HDAC6 as a target for neurodegenerative diseases: what makes it different from the other HDACs? Mol Neurodegener. 2013;8:7.
  • Bergman JA, Woan K, Perez-Villarroel P, et al. Selective histone deacetylase 6 inhibitors bearing substituted urea linkers inhibit melanoma cell growth. J Med Chem. 2012;55:9891–9899.
  • Butler KV, Kalin J, Brochier C, et al. Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. J Am Chem Soc. 2010;132:10842–10846.
  • De Vreese R, D’Hooghe M. Synthesis and applications of benzohydroxamic acid-based histone deacetylase inhibitors. Eur J Med Chem. 2017;135:174–195.
  • Miyake Y, Keusch JJ, Wang L, et al. Structural insights into HDAC6 tubulin deacetylation and its selective inhibition. Nat Chem Biol. 2016;12:748–754.
  • Porter NJ, Mahendran A, Breslow R, et al. Unusual zinc-binding mode of HDAC6-selective hydroxamate inhibitors. Proc Natl Acad Sci USA. 2017;114:13459–13464.
  • Hai Y, Christianson DW. Histone deacetylase 6 structure and molecular basis of catalysis and inhibition. Nat Chem Biol. 2016;12:741–747.
  • Porter NJ, Osko JD, Diedrich D, et al. Histone deacetylase 6-selective inhibitors and the influence of capping groups on hydroxamate-zinc denticity. J Med Chem. 2018;61:8054–8060.
  • Shen S, Hadley M, Ustinova K, et al. Discovery of a new isoxazole-3-hydroxamate-based histone deacetylase 6 inhibitor SS-208 with antitumor activity in syngeneic melanoma mouse models. J Med Chem. 2019;62:8557–8577.
  • Vogerl K, Ong N, Senger J, et al. Synthesis and biological investigation of phenothiazine-based benzhydroxamic acids as selective histone deacetylase 6 inhibitors. J Med Chem. 2019;63:1138–1166.
  • Santo L, Hideshima T, Kung AL, et al. Preclinical activity, pharmacodynamic, and pharmacokinetic properties of a selective HDAC6 inhibitor, ACY-1215, in combination with bortezomib in multiple myeloma. Blood. 2012;119:2579–2589.
  • Huang P, Almeciga-Pinto I, Jarpe M, et al. Selective HDAC inhibition by ACY-241 enhances the activity of paclitaxel in solid tumor models. Oncotarget. 2017;8:2694–2707.
  • Krukowski K, Ma J, Golonzhka O, et al. HDAC6 inhibition effectively reverses chemotherapy-induced peripheral neuropathy. Pain. 2017;158:1126–1137.
  • Benoy V, Vanden Berghe P, Jarpe M, et al. Development of improved HDAC6 inhibitors as pharmacological therapy for axonal Charcot-Marie-Tooth disease. Neurotherapeutics. 2017;14:417–428.
  • Ma J, Trinh RT, Mahant ID, et al. Cell-specific role of histone deacetylase 6 in chemotherapy-induced mechanical allodynia and loss of intraepidermal nerve fibers. Pain. 2019: published online. 2019 Jul 24;160:2877–2890. .
  • Choi EW, Song JW, Ha N, et al. CKD-506, a novel HDAC6-selective inhibitor, improves renal outcomes and survival in a mouse model of systemic lupus erythematosus. Sci Rep. 2018;8:17297.
  • Clinical trial status of CDK-506. [cited 2019 Nov]. Available from: http://www.koreabiomed.com/news/articleView.html?idxno=4337
  • Jochems J, Boulden J, Lee BG, et al. Antidepressant-like properties of novel HDAC6-selective inhibitors with improved brain bioavailability. Neuropsychopharmacology. 2014;39:389–400.
  • Shen S, Benoy V, Bergman JA, et al. Bicyclic-capped histone deacetylase 6 inhibitors with improved activity in a model of axonal Charcot-Marie-Tooth disease. ACS Chem Neurosci. 2016;7:240–258.
  • Kozikowski AP, Shen S, Pardo M, et al. Brain penetrable histone deacetylase 6 inhibitor SW-100 ameliorates memory and learning impairments in a mouse model of Fragile X syndrome. ACS Chem Neurosci. 2019;10:1679–1695.
  • Chen B, Petukhov PA, Jung M, et al. Chemistry and biology of mercaptoacetamides as novel histone deacetylase inhibitors. Bioorg Med Chem Lett. 2005;15:1389–1392.
  • Jung M, Kozikowski A, Dritschilo A. Rational design and development of radiation-sensitizing histone deacetylase inhibitors. Chem Biodivers. 2005;2:1452–1461.
  • Sung YM, Lee T, Yoon H, et al. Mercaptoacetamide-based class II HDAC inhibitor lowers Abeta levels and improves learning and memory in a mouse model of Alzheimer’s disease. Exp Neurol. 2013;239:192–201.
  • Song JM, Sung YM, Nam JH, et al. A mercaptoacetamide-based class II HDAC inhibitor lowers Abeta levels and improves learning and memory in a mouse model of Alzheimer’s disease. Exp Neurol. 2013;239:192–201.
  • Lee HY, Fan SJ, Huang FI, et al. 5-Aroylindoles act as selective histone deacetylase 6 inhibitors ameliorating Alzheimer’s disease phenotypes. J Med Chem. 2018;61:7087–7102.
  • Lee HY, Nepali K, Huang FI, et al. (N-Hydroxycarbonylbenylamino)quinolines as selective histone deacetylase 6 inhibitors suppress growth of multiple myeloma in vitro and in vivo. J Med Chem. 2018;61:905–917.
  • Fan SJ, Huang FI, Liou JP, et al. The novel histone de acetylase 6 inhibitor, MPT0G211, ameliorates tau phosphorylation and cognitive deficits in an Alzheimer’s disease model. Cell Death Dis. 2018;9:655.
  • Zhang L, Sheng S, Qin C. The role of HDAC6 in Alzheimer’s disease. J Alzheimers Dis. 2013;33:283–295.
  • Du H, Guo L, Yan S, et al. Early deficits in synaptic mitochondria in an Alzheimer’s disease mouse model. Proc Natl Acad Sci USA. 2010;107:18670–18675.
  • Kim C, Choi H, Jung ES, et al. HDAC6 inhibitor blocks amyloid beta-induced impairment of mitochondrial transport in hippocampal neurons. PLoS One. 2012;7:e42983.
  • Noack M, Leyk J, Richter-Landsberg C. HDAC6 inhibition results in tau acetylation and modulates tau phosphorylation and degradation in oligodendrocytes. Glia. 2014;62:535–547.
  • Ramsden M, Kotilinek L, Forster C, et al. Age-dependent neurofibrillary tangle formation, neuron loss, and memory impairment in a mouse model of human tauopathy (P301L). J Neurosci. 2005;25:10637–10647.
  • Selenica ML, Benner L, Housley SB, et al. Histone deacetylase 6 inhibition improves memory and reduces total tau levels in a mouse model of tau deposition. Alzheimers Res Ther. 2014;6:12.
  • Cook C, Carlomagno Y, Gendron TF, et al. Acetylation of the KXGS motifs in tau is a critical determinant in modulation of tau aggregation and clearance. Hum Mol Genet. 2014;23:104–116.
  • Zhang L, Liu C, Wu J, et al. Tubastatin A/ACY-1215 improves cognition in Alzheimer’s disease transgenic mice. J Alzheimers Dis. 2014;41:1193–1205.
  • Majid T, Griffin D, Criss Z 2nd, et al. Pharmocologic treatment with histone deacetylase 6 inhibitor (ACY-738) recovers Alzheimer’s disease phenotype in amyloid precursor protein/presenilin 1 (APP/PS1) mice. Alzheimers Dement. 2015;1:170–181.
  • Sulistio YA, Heese K. The ubiquitin-proteasome system and molecular chaperone deregulation in Alzheimer’s disease. Mol Neurobiol. 2016;53:905–931.
  • Barisic N, Claeys KG, Sirotkovic-Skerlev M, et al. Charcot-Marie-Tooth disease: a clinico-genetic confrontation. Ann Hum Genet. 2008;72:416–441.
  • Zuchner S, Vance JM. Mechanisms of disease: a molecular genetic update on hereditary axonal neuropathies. Nat Clin Pract Neurol. 2006;2:45–53.
  • d’Ydewalle C, Krishnan J, Chiheb DM, et al. HDAC6 inhibitors reverse axonal loss in a mouse model of mutant HSPB1-induced Charcot-Marie-Tooth disease. Nat Med. 2011;17:968–974.
  • Benoy V, Van Helleputte L, Prior R, et al. HDAC6 is a therapeutic target in mutant GARS-induced Charcot-Marie-Tooth disease. Brain. 2018;141:673–687.
  • Achilli F, Bros-Facer V, Williams HP, et al. An ENU-induced mutation in mouse glycyl-tRNA synthetase (GARS) causes peripheral sensory and motor phenotypes creating a model of Charcot-Marie-Tooth type 2D peripheral neuropathy. Dis Model Mech. 2009;2:359–373.
  • Seburn KL, Nangle LA, Cox GA, et al. An active dominant mutation of glycyl-tRNA synthetase causes neuropathy in a Charcot-Marie-Tooth 2D mouse model. Neuron. 2006;51:715–726.
  • Mo Z, Zhao X, Liu H, et al. Aberrant GlyRS-HDAC6 interaction linked to axonal transport deficits in Charcot-Marie-Tooth neuropathy. Nat Commun. 2018;9:1007.
  • Bilsland LG, Sahai E, Kelly G, et al. Deficits in axonal transport precede ALS symptoms in vivo. Proc Natl Acad Sci USA. 2010;107:20523–20528.
  • Taes I, Timmers M, Hersmus N, et al. Hdac6 deletion delays disease progression in the SOD1G93A mouse model of ALS. Hum Mol Genet. 2013;22:1783–1790.
  • Kim SH, Shanware NP, Bowler MJ, et al. Amyotrophic lateral sclerosis-associated proteins TDP-43 and FUS/TLS function in a common biochemical complex to co-regulate HDAC6 mRNA. J Biol Chem. 2010;285:34097–34105.
  • Petrov D, Mansfield C, Moussy A, et al. ALS clinical trials review: 20 years of failure. Are we any closer to registering a new treatment? Front Aging Neurosci. 2017;9:68.
  • Guo W, Naujock M, Fumagalli L, et al. HDAC6 inhibition reverses axonal transport defects in motor neurons derived from FUS-ALS patients. Nat Commun. 2017;8:861.
  • Rossaert E, Pollari E, Jaspers T, et al. Restoration of histone acetylation ameliorates disease and metabolic abnormalities in a FUS mouse model. Acta Neuropathol Commun. 2019;7:107.
  • Hauser DN, Hastings TG. Mitochondrial dysfunction and oxidative stress in Parkinson’s disease and monogenic parkinsonism. Neurobiol Dis. 2013;51:35–42.
  • Godena VK, Brookes-Hocking N, Moller A, et al. Increasing microtubule acetylation rescues axonal transport and locomotor deficits caused by LRRK2 Roc-COR domain mutations. Nat Commun. 2014;5:5245.
  • Pinho BR, Reis SD, Guedes-Dias P, et al. Pharmacological modulation of HDAC1 and HDAC6 in vivo in a zebrafish model: therapeutic implications for Parkinson’s disease. Pharmacol Res. 2016;103:328–339.
  • Katz DM, Bird A, Coenraads M, et al. Rett Syndrome: crossing the threshold to clinical translation. Trends Neurosci. 2016;39:100–113.
  • Chang Q, Khare G, Dani V, et al. The disease progression of Mecp2 mutant mice is affected by the level of BDNF expression. Neuron. 2006;49:341–348.
  • Zhou Z, Hong EJ, Cohen S, et al. Brain-specific phosphorylation of MeCP2 regulates activity-dependent Bdnf transcription, dendritic growth, and spine maturation. Neuron. 2006;52:255–269.
  • Ogier M, Wang H, Hong E, et al. Brain-derived neurotrophic factor expression and respiratory function improve after ampakine treatment in a mouse model of Rett syndrome. J Neurosci. 2007;27:10912–10917.
  • Reed NA, Cai D, Blasius TL, et al. Microtubule acetylation promotes kinesin-1 binding and transport. Curr Biol. 2006;16:2166–2172.
  • Xu X, Kozikowski AP, Pozzo-Miller L. A selective histone deacetylase-6 inhibitor improves BDNF trafficking in hippocampal neurons from Mecp2 knockout mice: implications for Rett syndrome. Front Cell Neurosci. 2014;8:68.
  • Gold WA, Lacina TA, Cantrill LC, et al. MeCP2 deficiency is associated with reduced levels of tubulin acetylation and can be restored using HDAC6 inhibitors. J Mol Med. 2015;93:63–72.
  • Landucci E, Brindisi M, Bianciardi L, et al. iPSC-derived neurons profiling reveals GABAergic circuit disruption and acetylated alpha-tubulin defect which improves after iHDAC6 treatment in Rett syndrome. Exp Cell Res. 2018;368:225–235.
  • Dompierre JP, Godin JD, Charrin BC, et al. Histone deacetylase 6 inhibition compensates for the transport deficit in Huntington’s disease by increasing tubulin acetylation. J Neurosci. 2007;27:3571–3583.
  • Guedes-Dias P, Oliveira JM. Lysine deacetylases and mitochondrial dynamics in neurodegeneration. Biochim Biophys Acta. 2013;1832:1345–1359.
  • Bobrowska A, Paganetti P, Matthias P, et al. Hdac6 knock-out increases tubulin acetylation but does not modify disease progression in the R6/2 mouse model of Huntington’s disease. PLoS One. 2011;6:e20696.
  • Ragot A, Pietropaolo S, Vincent J, et al. Genetic deletion of the Histone deacetylase 6 exacerbates selected behavioral deficits in the R6/1 mouse model for Huntington’s disease. Brain Behav. 2015;5:e00361.
  • Guedes-Dias P, de Proenca J, Soares TR, et al. HDAC6 inhibition induces mitochondrial fusion, autophagic flux and reduces diffuse mutant huntingtin in striatal neurons. Biochim Biophys Acta. 2015;1852:2484–2493.
  • Ross-Inta C, Omanska-Klusek A, Wong S, et al. Evidence of mitochondrial dysfunction in fragile X-associated tremor/ataxia syndrome. Biochem J. 2010;429:545–552.
  • Kaplan ES, Cao Z, Hulsizer S, et al. Early mitochondrial abnormalities in hippocampal neurons cultured from Fmr1 pre-mutation mouse model. J Neurochem. 2012;123:613–621.
  • Shuttleworth SJ, Tomassi CD, Cecil ARL, et al. Novel histone deacetylase inhibitors and their use in therapy. WO2014072714A1. 2014.
  • Shuttleworth SJ, Cecil ARL, Maccormick S, et al. Diheteroaryl histone deacetylase inhibitors and their use in therapy. WO2016067040A1. 2016.
  • Lee C, Lee J, Song H, et al. Heterocyclicalkyl derivative compounds as selective histone deacetylase inhibitors and pharmaceutical compositions comprising the same. WO2016190630A1. 2016.
  • Grindrod S, Jung M, Dritschilo A Selective histone deacetylase inhibitors for the treatment of human disease. WO2019139921A1. 2019.
  • Mazitschek R, Van Duzer JH Pyrimidine hydroxy amide compounds as HDAC6 selective inhibitors. US20150239869A1. 2015.
  • Yang M, Tamang DL, Jones SS, et al. Pyrimidine hydroxy amide compounds as histone deacetylase inhibitors. WO2015054474A1. 2015.
  • Golonzhka O, Jarpe MB HDAC inhibitors for the treatment of diabetic peripheral neuropathy. WO2017075192A1. 2017.
  • Van Duzer JH, Mazitschek R Phenyl and pyridinyl hydroxamic acids. WO2018089651A1. 2018.
  • Shuttleworth SJ, Tomassi CD, Cecil ARL, et al. Novel histone deacetylase inhibitors. WO2014181137A1. 2014.
  • Shuttleworth SJ, Cecil ARL, Maccormick S, et al. Polyheteroaryl histone deacetylase inhibitors and their use in therapy. WO2016067038A1. 2016.
  • Shuttleworth SJ, Whale AD, Colman LM, et al. Combinations comprising histone deacetylase inhibitors. WO2017208032A1. 2017.
  • Ng Pui Y, Davis H, Bair KW, et al. 3-Spirocyclic-6-hydroxamic acid tetralins as HDAC inhibitors. WO2016168598A1. 2016.
  • Ng Pui Y, Davis H, Bair KW, et al. 3-Spiro-7-hydroxamic acid tetralins as HDAC inhibitors. WO2016168660A1. 2016.
  • Zheng X, Ng Pui Y, Zablocki MM 2-Spiro-5- And 6-hydroxamic acid indanes as HDAC inhibitors. WO2017218950A1. 2017.
  • Tang G, Wong JC, Zhang W, et al. Identification of a novel aminotetralin class of HDAC6 and HDAC8 selective inhibitors. J Med Chem. 2014;57:8026–8034.
  • Zheng X, Ng Pui Y, Han B, et al. 3-Aryl bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors. US20160221973A1. 2016.
  • Zheng X, Ng Pui Y, Han B, et al. 3-Aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors. US20160222022A1. 2016.
  • Zheng X, Ng Pui Y, Rudnitskaya A, et al. Bicyclic [4,6,0] hydroxamic acids as HDAC inhibitors. US20160221997A1. 2016.
  • Lee C, Yang HM, Choi H, et al. Novel compounds for selective histone deacetylase inhibitors, and pharmaceutical composition comprising the same. WO2014178606A1. 2014.
  • Lee C, Yang HM, Lee C, et al. Novel azaindole derivatives as selective histone deacetylase (HDAC) inhibitors and pharmaceutical compositions comprising the same. WO2015087151A1. 2015.
  • Lee C, Yang HM, Kim D, et al. Novel indole derivative compound and pharmaceutical composition comprising the same. WO2015102426A1. 2015.
  • Song H, Lee C, Kwak D, et al. Novel compounds as histone deacetylase 6 inhibitors and pharmaceutical compositions comprising the same. WO2015137750A1. 2015.
  • Vergani B, Caprini G, Fossati G, et al. Selective HDAC6 inhibitors. WO2018189340A1. 2018.
  • Wu H, Wei C, Guo Q, et al. HDAC6 selective inhibitors, preparation method therefor, and application thereof. WO2018130155A1. 2018.
  • Guerriero JL, Sotayo A, Ponichtera HE, et al. Class IIa HDAC inhibition reduces breast tumours and metastases through anti-tumour macrophages. Nature. 2017;543:428–432.
  • Walji A, Berger R, Stump Craig A, et al. 3-Heterocyclyl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (HDAC6) inhibitors. WO 2017/222950 A1. 2017.
  • Walji A, Berger R, Stump CA, et al. 3-Aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (HDAC6) inhibitors. WO2017222951A1. 2017.
  • Walji A, Berger R, Stump Craig A, et al. 3-Heteroaryl substituted 5-trifluoromethyl oxadiazoles as histone deacetylase 6 (HDAC6) inhibitors. WO2017222952A1. 2017.
  • Kaieda A, Ishii N, Nara H, et al. Heterocyclic compound. WO2016031815A1. 2016.
  • Kaieda A, Daini M, Nara H, et al. Heterocyclic compound. WO2017014170A1. 2017.
  • Kaieda A, Toyofuku M, Daini M, et al. Oxadiazole derivatives useful as HDAC inhibitors. WO2017014321A1. 2017.
  • Kaieda A, Ishii N, Nara H, et al. Heterocyclic compound. WO2017033946A1. 2017.
  • Ito M, Sugiyama H, Kubo O, et al. Heterocyclic compound. WO2019027054A1. 2019.
  • Shuttleworth SJ, Gatland AE, Finnemore DJ, et al. Oxadiazolylthiophene derivatives useful as histone deacetylase inhibitors. WO2019166824A1. 2019.
  • Carceller GE, Ortega MA, Salas SJ 1,2,4-Oxadiazole derivatives as histone deacetylase 6 inhibitors. WO2019110663A1. 2019.
  • Lee J, Han Y, Kim Y, et al. 1,3,4-Oxadiazole sulfonamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same. WO2017018803A1. 2017.
  • Lee J, Kim Y, Lee CS, et al. 1,3,4-Oxadiazole derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same. WO2017023133A3. 2017.
  • Kim Y, Lee CS, Oh JT, et al. Oxadiazole amine derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same. WO2017065473A1. 2017.
  • Flipo M, Charton J, Hocine A, et al. Hydroxamates: relationships between structure and plasma stability. J Med Chem. 2009;52:6790–6802.
  • Hermant P, Bosc D, Piveteau C, et al. Controlling plasma stability of hydroxamic acids: a MedChem toolbox. J Med Chem. 2017;60:9067–9089.
  • Wang CY. Mutagenicity of hydroxamic acids for Salmonella typhimurium. Mutat Res. 1977;56:7–12.
  • De Vreese R, Van Steen N, Verhaeghe T, et al. Synthesis of benzothiophene-based hydroxamic acids as potent and selective HDAC6 inhibitors. Chem Commun. 2015;51:9868–9871.
  • Gertz M, Kilford PJ, Houston JB, et al. Drug lipophilicity and microsomal protein concentration as determinants in the prediction of the fraction unbound in microsomal incubations. Drug Metab Dispos. 2008;36:535–542.
  • Porter NJ, Shen S, Barinka C, et al. Molecular basis for the selective inhibition of histone deacetylase 6 by a mercaptoacetamide inhibitor. ACS Med Chem Lett. 2018;9:1301–1305.
  • Segretti MC, Vallerini GP, Brochier C, et al. Thiol-based potent and selective HDAC6 inhibitors promote tubulin acetylation and T-regulatory cell suppressive function. ACS Med Chem Lett. 2015;6:1156–1161.
  • Lv W, Zhang G, Barinka C, et al. Design and synthesis of mercaptoacetamides as potent, selective, and brain permeable histone deacetylase 6 inhibitors. ACS Med Chem Lett. 2017;8:510–515.
  • Bostrom J, Hogner A, Llinas A, et al. Oxadiazoles in medicinal chemistry. J Med Chem. 2012;55:1817–1830.

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.