Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

ISSN (Print): 1574-8928
ISSN (Online): 2212-3970

Review Article

Novel Selective Histone Deacetylase 6 (HDAC6) Inhibitors: A Patent Review (2016-2019)

Author(s): Xingrui He, Zhen Li, Xiao-Tao Zhuo, Zi Hui, Tian Xie* and Xiang-Yang Ye*

Volume 15, Issue 1, 2020

Page: [32 - 48] Pages: 17

DOI: 10.2174/1574892815666200217125419

Price: $65

Abstract

Background: Many human diseases are associated with dysregulation of HDACs. HDAC6 exhibits deacetylase activity not only to histone protein but also to non-histone proteins such as α- tubulin, HSP90, cortactin, and peroxiredoxin. These unique functions of HDAC6 have gained significant attention in the medicinal chemistry community in recent years. Thus a great deal of effort has devoted to developing selective HDAC6 inhibitors for therapy with the hope to minimize the side effects caused by pan-HDAC inhibition.

Objective: The review intends to analyze the structural feature of the scaffolds, to provide useful information for those who are interested in this field, as well as to spark the future design of the new inhibitors.

Methods: The primary tool used for patent searching is SciFinder. All patents are retrieved from the following websites: the World Intellectual Property Organization (WIPO®), the United States Patent Trademark Office (USPTO®), Espacenet®, and Google Patents. The years of patents covered in this review are between 2016 and 2019.

Results: Thirty-six patents from seventeen companies/academic institutes were classified into three categories based on the structure of ZBG: hydroxamic acid, 1,3,4-oxadiazole, and 1,2,4-oxadiazole. ZBG connects to the cap group through a linker. The cap group can tolerate different functional groups, including amide, urea, sulfonamide, sulfamide, etc. The cap group appears to modulate the selectivity of HDAC6 over other HDAC subtypes.

Conclusion: Selectively targeting HDAC6 over other subtypes represents two fold advantages: it maximizes the pharmacological effects and minimizes the side effects seen in pan-HDAC inhibitors. Many small molecule selective HDAC6 inhibitors have advanced to clinical studies in recent years. We anticipate the approval of selective HDAC6 inhibitors as therapeutic agents in the near future.

Keywords: Cancer, epigenetic mechanism, Histone Deacetylase 6 (HDAC6), hydroxamic acid, non-epigenetic mechanism, 1, 3, 4-oxadiazole, patent analysis.

[1]
Berger SL. Histone modifications in transcriptional regulation. Curr Opin Genet Dev 2002; 12(2): 142-8.
[http://dx.doi.org/10.1016/s0959-437x(02)00279-4] [PMID: 11893486]
[2]
Simões-Pires CA, Bertrand P, Cuendet M. Novel histone deacetylase 6 (HDAC6) selective inhibitors: a patent evaluation (WO2014181137). Expert Opin Ther Pat 2017; 27(3): 229-36.
[http://dx.doi.org/10.1080/13543776.2017.1282945] [PMID: 28092474]
[3]
Franci G, Miceli M, Altucci L. Targeting epigenetic networks with polypharmacology: a new avenue to tackle cancer. Epigenomics 2010; 2(6): 731-42.
[http://dx.doi.org/10.2217/epi.10.62] [PMID: 22122079]
[4]
Dallavalle S, Pisano C, Zunino F. Development and therapeutic impact of HDAC6-selective inhibitors. Biochem Pharmacol 2012; 84(6): 756-65.
[http://dx.doi.org/10.1016/j.bcp.2012.06.014] [PMID: 22728920]
[5]
Chang C, Zhao S, Guo J, Zhao Y, Fei C, Gu S, et al. Chidamide, a novel histone deacetylase inhibitor, displays potent antitumor activity against MDS cells mainly through JAK2/STAT3 signaling inhibition. Blood 2015; 126(23): 5233.
[http://dx.doi.org/10.1182/blood.V126.23.5233.5233]
[6]
Song Y, Lim J, Seo YH. A novel class of anthraquinone-based HDAC6 inhibitors. Eur J Med Chem 2019; 164: 263-72.
[http://dx.doi.org/10.1016/j.ejmech.2018.12.056] [PMID: 30597327]
[7]
Zou H, Wu Y, Navre M, Sang B-C. Characterization of the two catalytic domains in histone deacetylase 6. Biochem Biophys Res Commun 2006; 341(1): 45-50.
[http://dx.doi.org/10.1016/j.bbrc.2005.12.144] [PMID: 16412385]
[8]
Liu Y, Li L, Min J. Structural biology: HDAC6 finally crystal clear. Nat Chem Biol 2016; 12(9): 660-1.
[http://dx.doi.org/10.1038/nchembio.2158] [PMID: 27538024]
[9]
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(8) e42983
[http://dx.doi.org/10.1371/journal.pone.0042983] [PMID: 22937007]
[10]
Kawaguchi Y, Kovacs JJ, McLaurin A, Vance JM, Ito A, Yao T-P. The deacetylase HDAC6 regulates aggresome formation and cell viability in response to misfolded protein stress. Cell 2003; 115(6): 727-38.
[http://dx.doi.org/10.1016/s0092-8674(03)00939-5] [PMID: 14675537]
[11]
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(28): 9633-8.
[http://dx.doi.org/10.1073/pnas.0803749105] [PMID: 18606987]
[12]
Chen S, Owens GC, Makarenkova H, Edelman DB. HDAC6 regulates mitochondrial transport in hippocampal neurons. PLoS One 2010; 5(5) e10848
[http://dx.doi.org/10.1371/journal.pone.0010848] [PMID: 20520769]
[13]
Yano M, Katoh T, Miyazawa M, et al. Clinicopathological correlation of ARID1A status with HDAC6 and its related factors in ovarian clear cell carcinoma. Sci Rep 2019; 9(1): 2397.
[http://dx.doi.org/10.1038/s41598-019-38653-0] [PMID: 30787326]
[14]
Carew JS, Espitia CM, Zhao W, et al. Rational cotargeting of HDAC6 and BET proteins yields synergistic antimyeloma activity. Blood Adv 2019; 3(8): 1318-29.
[http://dx.doi.org/10.1182/bloodadvances.2018026484] [PMID: 31015208]
[15]
Zhang Y, Ying JB, Hong JJ, et al. How does chirality determine the selective inhibition of histone deacetylase 6? A lesson from trichostatin A enantiomers based on molecular dynamics. ACS Chem Neurosci 2019; 10(5): 2467-80.
[http://dx.doi.org/10.1021/acschemneuro.8b00729] [PMID: 30784262]
[16]
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(18): 8557-77.
[http://dx.doi.org/10.1021/acs.jmedchem.9b00946] [PMID: 31414801]
[17]
Nam G, Jung JM, Park H-J, et al. Structure-activity relationship study of thiazolyl-hydroxamate derivatives as selective histone deacetylase 6 inhibitors. Bioorg Med Chem 2019; 27(15): 3408-20.
[http://dx.doi.org/10.1016/j.bmc.2019.06.036] [PMID: 31235266]
[18]
Pérez-Salvia M, Aldaba E, Vara Y, et al. In vitro and in vivo activity of a new small-molecule inhibitor of HDAC6 in mantle cell lymphoma. Haematologica 2018; 103(11): e537-40.
[http://dx.doi.org/10.3324/haematol.2018.189241] [PMID: 29880608]
[19]
Tang C, Du Y, Liang Q, Cheng Z, Tian J. A selenium-containing selective histone deacetylase 6 inhibitor for targeted in vivo breast tumor imaging and therapy. J Mater Chem B 2019; 7: 3528-36.
[http://dx.doi.org/10.1039/C9TB00383E]
[20]
Lee DH, Kim GW, Kwon SH. The HDAC6-selective inhibitor is effective against non-Hodgkin lymphoma and synergizes with ibrutinib in follicular lymphoma. Mol Carcinog 2019; 58(6): 944-56.
[http://dx.doi.org/10.1002/mc.22983] [PMID: 30693983]
[21]
Yadav R, Mishra P, Yadav D. Histone deacetylase inhibitors: A prospect in drug discovery. Turk J Pharma Sci 2019; 16(1): 101-14.
[http://dx.doi.org/10.4274/tjps.75047]
[22]
Liang T, Hou X, Zhou Y, Yang X, Fang H. Design, synthesis, and biological evaluation of 2,4-imidazolinedione derivatives as HDAC6 isoform-selective inhibitors. ACS Med Chem Lett 2019; 10(8): 1122-7.
[http://dx.doi.org/10.1021/acsmedchemlett.9b00084] [PMID: 31413795]
[23]
Laino AS, Betts BC, Veerapathran A, et al. HDAC6 selective inhibition of melanoma patient T-cells augments anti-tumor characteristics. J Immunother Cancer 2019; 7: 33.
[http://dx.doi.org/10.1186/s40425-019-0517-0] [PMID: 30728070]
[24]
Li T, Zhang C, Hassan S, et al. Histone deacetylase 6 in cancer. J Hematol Oncol 2018; 11: 111.
[http://dx.doi.org/10.1186/s13045-018-0654-9] [PMID: 30176876]
[25]
Yuan H, Li H, Yu P, et al. Involvement of HDAC6 in ischaemia and reperfusion-induced rat retinal injury. BMC Ophthalmol 2018; 18(1): 300.
[http://dx.doi.org/10.1186/s12886-018-0951-7] [PMID: 30453928]
[26]
Kaliszczak M, van Hechanova E, Li Y, et al. The HDAC6 inhibitor C1A modulates autophagy substrates in diverse cancer cells and induces cell death. Br J Cancer 2018; 119(10): 1278-87.
[http://dx.doi.org/10.1038/s41416-018-0232-5] [PMID: 30318510]
[27]
Brindisi M, Saraswati AP, Brogi S, Gemma S, Butini S, Campiani G. 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 2020; 63(1): 23-39.
[http://dx.doi.org/10.1021/acs.jmedchem.9b00924] [PMID: 31415174]
[28]
Wang F, Zheng L, Yi Y, et al. SKLB-23bb, a HDAC6-selective inhibitor, exhibits superior and broad-spectrum antitumor activity via additionally targeting microtubules. Mol Cancer Ther 2018; 17(4): 763-75.
[http://dx.doi.org/10.1158/1535-7163.MCT-17-0332] [PMID: 29610282]
[29]
Fedele P, Orlando L, Cinieri S. Targeting triple negative breast cancer with histone deacetylase inhibitors. Expert Opin Investig Drugs 2017; 26(11): 1199-206.
[http://dx.doi.org/10.1080/13543784.2017.1386172] [PMID: 28952409]
[30]
Vogl DT, Raje N, Jagannath S, et al. Ricolinostat, the first selective histone deacetylase 6 inhibitor, in combination with bortezomib and dexamethasone for relapsed or refractory multiple myeloma. Clin Cancer Res 2017; 23(13): 3307-15.
[http://dx.doi.org/10.1158/1078-0432.CCR-16-2526] [PMID: 28053023]
[31]
Andreopoulou E, Kelly CM, McDaid HM. Therapeutic advances and new directions for triple-negative breast cancer. Breast Care 2017; 12(1): 21-8.
[http://dx.doi.org/10.1159/000455821] [PMID: 28611537]
[32]
Batchu SN, Brijmohan AS, Advani A. The therapeutic hope for HDAC6 inhibitors in malignancy and chronic disease. Clin Sci 2016; 130(12): 987-1003.
[http://dx.doi.org/10.1042/CS20160084] [PMID: 27154743]
[33]
Mu S, Kuroda Y, Shibayama H, et al. Panobinostat PK/PD profile in combination with bortezomib and dexamethasone in patients with relapsed and relapsed/refractory multiple myeloma. Eur J Clin Pharmacol 2016; 72(2): 153-61.
[http://dx.doi.org/10.1007/s00228-015-1967-z] [PMID: 26494130]
[34]
Depetter Y, Geurs S, De Vreese R, et al. Selective pharmacological inhibitors of HDAC6 reveal biochemical activity but functional tolerance in cancer models. Int J Cancer 2019; 145: 735-47.
[http://dx.doi.org/10.1002/ijc.32169] [PMID: 30694564]
[35]
Sun X, Xie Y, Sun X, et al. The selective HDAC6 inhibitor Nexturastat A induces apoptosis, overcomes drug resistance and inhibits tumor growth in multiple myeloma. Biosci Rep 2019; 39(3) BSR20181916
[http://dx.doi.org/10.1042/BSR20181916] [PMID: 30782785]
[36]
Newbold A, Falkenberg KJ, Prince HM, Johnstone RW. How do tumor cells respond to HDAC inhibition? FEBS J 2016; 283(22): 4032-46.
[http://dx.doi.org/10.1111/febs.13746] [PMID: 27112360]
[37]
Yu F, Ran J, Zhou J. Ciliopathies: Does HDAC6 represent a new therapeutic target? Trends Pharmacol Sci 2016; 37(2): 114-9.
[http://dx.doi.org/10.1016/j.tips.2015.11.002] [PMID: 26651415]
[38]
Hideshima T, Qi J, Paranal R, et al. Discovery of selective small-molecule HDAC6 inhibitor for overcoming proteasome inhibitor resistance in multiple myeloma. Proc Natl Acad Sci USA 2016; 113(46): 13162-7.
[http://dx.doi.org/10.1073/pnas.1608067113] [PMID: 27799547]
[39]
Marek L, Hamacher A, Hansen FK, et al. Histone deacetylase (HDAC) inhibitors with a novel connecting unit linker region reveal a selectivity profile for HDAC4 and HDAC5 with improved activity against chemoresistant cancer cells. J Med Chem 2013; 56(2): 427-36.
[http://dx.doi.org/10.1021/jm301254q] [PMID: 23252603]
[40]
Tate CR, Rhodes LV, Segar HC, et al. Targeting triple-negative breast cancer cells with the histone deacetylase inhibitor panobinostat. Breast Cancer Res 2012; 14(3): R79.
[http://dx.doi.org/10.1186/bcr3192] [PMID: 22613095]
[41]
Inks ES, Josey BJ, Jesinkey SR, Chou CJ. A novel class of small molecule inhibitors of HDAC6. ACS Chem Biol 2012; 7(2): 331-9.
[http://dx.doi.org/10.1021/cb200134p] [PMID: 22047054]
[42]
Johnstone RW. Histone-deacetylase inhibitors: Novel drugs for the treatment of cancer. Nat Rev Drug Discov 2002; 1(4): 287-99.
[PMID: 12120280]
[43]
Zeb A, Park C, Rampogu S, Son M, Lee GH, Lee KW. Structure-based drug designing recommends HDAC6 inhibitors to attenuate microtubule-associated Tau-pathogenesis. ACS Chem Neurosci 2019; 10(3): 1326-35.
[http://dx.doi.org/10.1038/nrd772] [PMID: 12120280]
[44]
Zhou YF, Zhou YX, Lu F, Peng XS, Liang JQ. Application of Histone Deacetylase 6 (HDAC6) inhibitor in transplantation of bone marrow mesenchymal stem cell (BMSC) CN107189981. (2017).
[45]
Silva J, Yu J. Identifying and treating cancer patients who are suitable for a targeted therapy using an Histone Deacetylase 6 (HDAC6) inhibitor WO2017070441. (2017).
[46]
Iaconelli J, Xuan L, Karmacharya R. HDAC6 modulates signaling pathways relevant to synaptic biology and neuronal differentiation in human stem cell-derived neurons. Int J Mol Sci 2019; 20 E1605
[http://dx.doi.org/10.3390/ijms20071605] [PMID: 30935091]
[47]
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(3): 1679-95.
[http://dx.doi.org/10.1021/acschemneuro.8b00600] [PMID: 30511829]
[48]
Iaconelli J, Lalonde J, Watmuff B, et al. Lysine deacetylation by HDAC6 regulates the kinase activity of AKT in human neural progenitor cells. ACS Chem Biol 2017; 12(8): 2139-48.
[http://dx.doi.org/10.1021/acschembio.6b01014] [PMID: 28628306]
[49]
Jochems J, Boulden J, Lee BG, et al. Antidepressant-like properties of novel HDAC6-selective inhibitors with improved brain bioavailability. Neuropsychopharmacology 2014; 39(2): 389-400.
[http://dx.doi.org/10.1038/npp.2013.207] [PMID: 23954848]
[50]
Iaconelli J, Huang JH, Berkovitch SS, et al. HDAC6 inhibitors modulate Lys49 acetylation and membrane localization of β-catenin in human iPSC-derived neuronal cells. ACS Chem Biol 2015; 10(3): 883-90.
[http://dx.doi.org/10.1021/cb500838r] [PMID: 25546293]
[51]
Liu N, Zhuang SG, Shi YF, Xu LQ, Tao M, Tang JH. Application of Histone Deacetylase HDAC6 inhibitor in preparing medicine for preventing and treating acute renal injury CN107158390. (2017).
[52]
Dere R, Walker CL, Jonasch E. Use of HDAC6 inhibitors for preventing and treating renal cystogenesis, renal cell carcinoma, and renal ciliopathies WO2018039581. (2018).
[53]
Jang JH, Yoon JA. Pharmaceutical composition containing HDAC6 inhibitor as active ingredient for prevention or treatment of itching WO2019139244. (2019).
[54]
Matthias PD, Saito M. HDAC6 inhibitors for treating a degenerative disease associated with intracellular granules formed from protein aggregation WO2019030692. (2019).
[55]
Liu N, Zhuang SG, Xu LQ, et al. Application of histone deacetylase HDAC6 inhibitor in manufacture of medicine for preventing and treating peritoneal fibrosis after peritoneal dialysis CN107260730. (2017).
[56]
Hwang JS, Yoon EY, Nam SH, et al. Composition for HDAC6 enzyme inhibitor for enhancing intestinal epithelial cell barrier function KR1734072. (2017).
[57]
Butler KV, Kozikowski AP. Chemical origins of isoform selectivity in histone deacetylase inhibitors. Curr Pharm Des 2008; 14(6): 505-28.
[http://dx.doi.org/10.2174/138161208783885353] [PMID: 18336297]
[58]
Mai A, Massa S, Rotili D, et al. Exploring the connection unit in the HDAC inhibitor pharmacophore model: novel uracil-based hydroxamates. Bioorg Med Chem Lett 2005; 15(21): 4656-61.
[http://dx.doi.org/10.1016/j.bmcl.2005.07.081] [PMID: 16165353]
[59]
Mehndiratta S, Lin M-H, Wu Y-W, et al. N-alkyl-hydroxybenzoyl anilide hydroxamates as dual inhibitors of HDAC and HSP90, downregulating IFN-γ induced PD-L1 expression. Eur J Med Chem 2020; 185 111725
[http://dx.doi.org/10.1016/j.ejmech.2019.111725] [PMID: 31655430]
[60]
El-Din MMG, El-Gamal MI, Abdel-Maksoud MS, Yoo KH, Oh C-H. Synthesis and in vitro antiproliferative activity of new 1,3,4-oxadiazole derivatives possessing sulfonamide moiety. Eur J Med Chem 2015; 90: 45-52.
[http://dx.doi.org/10.1016/j.ejmech.2014.11.011] [PMID: 25461310]
[61]
Leonhardt M, Sellmer A, Krämer OH, et al. Design and biological evaluation of tetrahydro-β-carboline derivatives as highly potent histone deacetylase 6 (HDAC6) inhibitors. Eur J Med Chem 2018; 152: 329-57.
[http://dx.doi.org/10.1016/j.ejmech.2018.04.046] [PMID: 29738953]
[62]
Senger J, Melesina J, Marek M, et al. Synthesis and biological investigation of oxazole hydroxamates as highly selective Histone Deacetylase 6 (HDAC6) inhibitors. J Med Chem 2016; 59(4): 1545-55.
[http://dx.doi.org/10.1021/acs.jmedchem.5b01493] [PMID: 26653328]
[63]
Vögerl 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; 62(3): 1138-66.
[http://dx.doi.org/10.1021/acs.jmedchem.8b01090] [PMID: 30645113]
[64]
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(11): 2579-89.
[http://dx.doi.org/10.1182/blood-2011-10-387365] [PMID: 22262760]
[65]
Niesvizky R, Richardson PG, Gabrail NY, Madan S, Yee AJ, Quayle SN, et al. ACY-241, a novel, HDAC6 selective inhibitor: Synergy with immunomodulatory (IMiD®) drugs in Multiple Myeloma (MM) cells and early clinical results (ACE-MM-200 Study). Blood 2015; 126(23): 3040.
[http://dx.doi.org/10.1182/blood.V126.23.3040.3040]
[66]
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(2): 2694.
[http://dx.doi.org/10.18632/oncotarget.13738] [PMID: 27926524]
[67]
Kaliszczak M, Trousil S, Åberg O, Perumal M, Nguyen Q-D, Aboagye EO. A novel small molecule hydroxamate preferentially inhibits HDAC6 activity and tumour growth. Br J Cancer 2013; 108(2): 342-50.
[http://dx.doi.org/10.1038/bjc.2012.576] [PMID: 23322205]
[68]
Hideshima T, Mazitschek R, Qi J, et al. HDAC6 inhibitor WT161 downregulates growth factor receptors in breast cancer. Oncotarget 2017; 8(46): 80109-23.
[http://dx.doi.org/10.18632/oncotarget.19019] [PMID: 29113288]
[69]
Suzuki T, Kouketsu A, Itoh Y, et al. Highly potent and selective histone deacetylase 6 inhibitors designed based on a small-molecular substrate. J Med Chem 2006; 49(16): 4809-12.
[http://dx.doi.org/10.1021/jm060554y] [PMID: 16884291]
[70]
Goracci L, Deschamps N, Randazzo GM, et al. A rational approach for the identification of non-hydroxamate HDAC6-selective inhibitors. Sci Rep 2016; 6: 29086.
[http://dx.doi.org/10.1038/srep29086] [PMID: 27404291]
[71]
Wang X-X, Wan R-Z, Liu Z-P. Recent advances in the discovery of potent and selective HDAC6 inhibitors. Eur J Med Chem 2018; 143: 1406-18.
[http://dx.doi.org/10.1016/j.ejmech.2017.10.040] [PMID: 29133060]
[72]
Qin H-T, Li H-Q, Liu F. Selective histone deacetylase small molecule inhibitors: recent progress and perspectives. Expert Opin Ther Pat 2017; 27(5): 621-36.
[http://dx.doi.org/10.1080/13543776.2017.1276565] [PMID: 28033734]
[73]
Freitas M, Cuendet M, Bertrand P. HDAC inhibitors: a 2013-2017 patent survey. Expert Opin Ther Pat 2018; 28(5): 365-81.
[http://dx.doi.org/10.1080/13543776.2018.1459568] [PMID: 29671355]
[74]
Lee C, Lee J, Song H, Bae D, Ha N. Heterocyclicalkyl derivative compounds as selective histone deacetylase inhibitors and pharmaceutical compositions comprising the same WO2016190630. (2016).
[75]
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 WO2017018803. (2017).
[76]
Lee J, Han Y, Kim Y, et al. 1,3,4-Oxadiazole sulfamide derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same WO2017018805. (2017).
[77]
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 WO2017023133. (2017).
[78]
Kim Y, Lee CS, Oh JT, Song H, Choi J, Lee J. Oxadiazole amine derivative compounds as histone deacetylase 6 inhibitor, and the pharmaceutical composition comprising the same WO2017065473. (2017).
[79]
Kim SJ. Pharmaceutical combinations of histone deacetylase inhibitor and proteasome inhibitor or immunomodulatory drug for the treatment of hematological cancer WO2018066946. (2018).
[80]
Lee J, Ko MS, Han Y, Kim Y. 1,3,4-oxadiazole amide derivative compound as histone deacetylase 6 inhibitor, and pharmaceutical composition containing same WO2017018804. (2017).
[81]
Van DJ, Mazitschek R, Ogier W, et al. Reverse amide compounds as protein deacetylase inhibitors and methods of use thereof WO2011091213. (2011).
[82]
Lee SS, Lee KJ, Lee CS, et al. Novel hydroxamate derivative, a production method for the same, and a pharmaceutical composition comprising the same WO2010110545. (2010).
[83]
Shim Y, Kim S, Velinova M, Arold G, Kim S, Choi YI, et al. Safety, tolerability, pharmacokinetics and pharmacodynamics of ckd-506, a novel, Histone Deacetylase 6 (HDAC6) inhibitor, in healthy volunteers. ACR/ARHP Annual Meeting 2018. Abstract number: 2535.
[84]
Shin J, Ha N, Bae D, Suh D-h, Baek J-y, Jun JH, et al. OP23 CKD- 506, a novel Histone Deacetylase (HDAC) 6 inhibitor, ameliorates colitis in various animal models. J Crohn's Colitis 2019; 13(Suppl_1): S015-6.
[85]
Choi EW, Song JW, Ha N, Choi YI, Kim S. CKD-506, a novel HDAC6-selective inhibitor, improves renal outcomes and survival in a mouse model of systemic lupus erythematosus. Sci Rep 2018; 8(1): 17297.
[http://dx.doi.org/10.1038/s41598-018-35602-1] [PMID: 30470828]
[86]
Korea Drug Development Fund, R&D pipeline, page 37, CKD-506. [cited 2020 February 28]. Available from:. https://kasbp.org/resources/Documents/KDDF%202018.pdf
[87]
Shuttleworth SJ, Cecil AR, MacCormick S, Nodes WJ, Tomassi CD, Silva FA. Polyheteroaryl histone deacetylase inhibitors and their use in therapy. WO2016067038. ( 2016).
[88]
Shuttleworth SJ, Cecil ARL, MacCormick S, Nodes WJ, Tomassi CD, Silva FA. Diheteroaryl histone deacetylase inhibitors and their use in therapy WO2016067040. (2016).
[89]
Shuttleworth SJ, Whale AD. Compositions comprising a PI3K inhibitor and an HDAC inhibitor WO2017029514. (2017).
[90]
Shuttleworth SJ, Whale AD, Colman LM, Rogers HL. Combinations comprising histone deacetylase inhibitors WO2017208032. (2017).
[91]
Safety,tolerability and MTD KA2507 (HDAC6 Inhibitor) in patients with solid tumours. [cited 2019 Sep 18]. [cited 2019 September 28] Available from:. https://www.cancer.gov/about-cancer/treatment/clinical-trials/search/v?id=NCI-2017-00819&r=1
[92]
Shuttleworth SJ. KA2237 and KA2507: Novel, oral cancer immunotherapeutics targeting PI3K-p110β/p110δ and HDAC6 with single-agent and combination activity. AACR 107th Annual Meeting 2016. Abstract 3996..
[93]
Karus Therapeutics press release. [cited 2020 February 28]. Available from:. https://karustherapeutics.com/portfolio/hdac6-selective-inhibitors/
[94]
Zheng XZ, Ng PY, Han BS, et al. 3-Aryl-4-amido-bicyclic [4,5,0] hydroxamic acids as HDAC inhibitors. US2016222022. (2016).
[95]
Zheng XZ, Ng PY, Zablocki M-M. 2-Spiro-5- and 6- hydroxamic acid indanes as HDAC inhibitors. US2019201384. (2019).
[96]
Mazitschek R, van Duzer DJ. Pyrimidine hydroxy amide compounds as HDAC6 selective inhibitors. US9464073. (2016).
[97]
van Duzer DJ, Mazitschek R. Preparation of phenyl and pyridinyl hydroxamic acids as HDAC6 inhibitors. patent US20180127356. (2018).
[98]
Breslow R, Marks P, Mahendran A, Yao YS. Selective HDAC6 inhibitors. US2018118709. (2018).
[99]
Walji A, Berger R, Stump CA, et al. 3-Heterocyclyl substituted 5-trifluoromethyl oxadiazoles as Histone Deacetylase 6 (HDAC6) inhibitors WO2017222950. (2017).
[100]
Walji A, Berger R, Stump CA, et al. 3-Aryl and heteroaryl substituted 5-trifluoromethyl oxadiazoles as Histone Deacetylase 6 (HDAC6) inhibitors WO2017222951. (2017).
[101]
Walji A, Berger R, Stump CA, et al. 3-Heteroaryl substituted 5-trifluoromethyl oxadiazoles as Histone Deacetylase 6 (HDAC6) inhibitors WO2017222952. (2017).
[102]
Wu H, Wei C, Guo Q, et al. HDAC6 selective inhibitor, preparation method, and application thereof WO2018130155. (2018).
[103]
Guo Q, Wei C, Xiao Y, Fan L, Qian W. Preparation of pyridine derivative in polymorph as HDAC6-selective inhibitor WO2020007329. (2020).
[104]
Villagra A, Kozikowski AP, Shen S. Preparation of isoxazole hydroxamic acids as histone deacetylase 6 inhibitors useful in mono- and combination therapy of diseases WO2018183701. (2018).
[105]
Vergani B, Caprini G, Fossati G, et al. Preparation of novel benzohydroxamic compounds as selective HDAC6 inhibitors WO2018189340. (2018).
[106]
Teng C-M, Liou J-P, Pan S-L, Yang C-R. Preparation of benzohydroxamic acid and (E)-3-phenylpropenohydroxamic acid derivatives as selective histone deacetylase 6 inhibitors and use thereof WO2017200966. (2017).
[107]
Mahboobi S, Pongratz H, Gruenstein E. Preparation of tetracyclic fused pyridoindoles as HDAC6 inhibitors with improved solubility, useful in mono- and combination therapy of cancer and other diseases WO2019007836. (2019).
[108]
Ma H, Feng Y, Wang Y. Preparation of spiro compounds-based Histone Deacetylase (HDAC) inhibitors and their uses WO2019169257. (2019).
[109]
Liu Z, Wang X, Liu P, Shi X, He X, Li X. Histone Deacetylase HDAC6 inhibitor and preparation method and application thereof CN109020921. (2018).
[110]
Liu D, Zhao L, Xu Q, Cheng G, Wang T. Hydroxamic acid compound having HDAC6 inhibition activity and application thereof CN109574936. (2019).
[111]
Liu D, Zhao L, Wen J, Niu Q, Xu Q, Jing Y. Thiols compound with HDAC6 inhibitory activity and its application in preparing drug for treating and/or preventing prostatic cancer, breast carcinoma, cervical cancer or leukemia CN109384793. (2019).
[112]
Ito M, Sugiyama H, Kubo O, Kikuchi F, Yasui T, Kakegawa K. Preparation of heterocyclic oxadiazoles as HDAC inhibitors WO2019027054. (2019).
[113]
Carceller González E, Ortega Muñoz A, Salas Solana J. Preparation of 3-pyridinyl-1,2,4-oxadiazole derivatives as histone deacetylase 6 inhibitors useful in treatment of diseases WO2019110663. (2019).
[114]
Grindrod S, Jung M, Dritschilo A. Selective Histone Deacetylase inhibitors (HDAC) for the treatment of human disease WO2019139921. (2019).
[115]
Li D, Yan G, Li D, Tao L. HDAC6 selective inhibitor for antitumor drug synthesis CN110294759. (2019).
[116]
Drug profile of CKD-506. [cited 2020 February 28]. Available from:. https://adisinsight.springer.com/drugs/800050914
[117]
CStone receives IND approval in China for HDAC6 inhibitor CS3003. [cited 2020 February 28]. Available from:. https://www.biospectrumasia.com/news/48/13012/cstone-receives-ind-approval-in-china-for-hdac6-inhibitor-cs3003.html

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy