Generic placeholder image

Recent Patents on Anti-Cancer Drug Discovery

Editor-in-Chief

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

Mini-Review Article

An Update of Lysine Specific Demethylase 1 Inhibitor: A Patent Review (2016-2020)

Author(s): Yi-Chao Zheng, Yue-Jiao Liu, Ya Gao, Bo Wang* and Hong-Min Liu*

Volume 17, Issue 1, 2022

Published on: 28 July, 2021

Page: [9 - 25] Pages: 9

DOI: 10.2174/1574892816666210728125224

Price: $65

Abstract

Background: As a FAD (Flavin Adenine Dinucleotide) - dependent histone demethylase discovered in 2004, LSD1 (lysine-specific demethylase 1) was reported to be overexpressed in diverse tumors, regulating target genes transcription associated with cancer development. Hence, LSD1 targeted inhibitors may represent a new insight in anticancer drug discovery. For these reasons, researchers in both the pharmaceutical industry and academia have been actively pursuing LSD1 inhibitors in the quest for new anti-cancer drugs.

Objectives: This review summaries patents about LSD1 inhibitors in recent 5 years in the hope of providing a reference for LSD1 researchers to develop new modulators of LSD1 with higher potency and fewer adverse effects.

Methods: This review collects LSD1 inhibitors disclosed in patents since 2016. The primary ways of patent searching are Espacenet®, Google Patents, and CNKI.

Results: This review covers dozens of patents related to LSD1 inhibitors in recent five years. The compound structures are mainly divided into TCP (Tranylcypromine) derivatives, imidazole derivatives, pyrimidine derivatives, and other natural products and peptides. Meanwhile, the compounds that have entered the clinical phase are also described.

Conclusion: Most of the compounds in these patents have been subjected to activity analysis with LSD1 and multi-cell lines, showing good antitumor activity in vitro and in vivo. These patents exhibited the structural diversity of LSD1 inhibitors and the potential of natural products as novel LSD1 inhibitors.

Keywords: LSD1, cancer, selective inhibitors, patent analysis, tranylcypromine, derivatives.

[1]
Shi Y, Lan F, Matson C, et al. Histone demethylation mediated by the nuclear amine oxidase homolog LSD1. Cell 2004; 119(7): 941-53.
[http://dx.doi.org/10.1016/j.cell.2004.12.012] [PMID: 15620353]
[2]
Sugino N, Kawahara M, Tatsumi G, et al. A novel LSD1 inhibitor NCD38 ameliorates MDS-related leukemia with complex karyotype by attenuating leukemia programs via activating super-enhancers. Leukemia 2017; 31(11): 2303-14.
[http://dx.doi.org/10.1038/leu.2017.59] [PMID: 28210006]
[3]
Stavropoulos P, Blobel G, Hoelz A. Crystal structure and mechanism of human lysine-specific demethylase-1. Nat Struct Mol Biol 2006; 13(7): 626-32.
[http://dx.doi.org/10.1038/nsmb1113] [PMID: 16799558]
[4]
Aravind L, Iyer L M. The SWIRM domain: a conserved module found in chromosomal proteins points to novel chromatin-modifying activities. Genome Biol 2002; 3(8): 1-7.
[5]
Chen Y, Yang Y, Wang F, et al. Crystal structure of human histone lysine-specific demethylase 1 (LSD1). Proc Natl Acad Sci USA 2006; 103(38): 13956-61.
[http://dx.doi.org/10.1073/pnas.0606381103] [PMID: 16956976]
[6]
Peng B, Wang J, Hu Y, et al. Modulation of LSD1 phosphorylation by CK2/WIP1 regulates RNF168-dependent 53BP1 recruitment in response to DNA damage. Nucleic Acids Res 2015; 43(12): 5936-47.
[http://dx.doi.org/10.1093/nar/gkv528] [PMID: 25999347]
[7]
Metzger E, Willmann D, McMillan J, et al. Assembly of methylated KDM1A and CHD1 drives androgen receptor-dependent transcription and translocation. Nat Struct Mol Biol 2016; 23(2): 132-9.
[http://dx.doi.org/10.1038/nsmb.3153] [PMID: 26751641]
[8]
Lee MG, Wynder C, Cooch N, Shiekhattar R. An essential role for CoREST in nucleosomal histone 3 lysine 4 demethylation. Nature 2005; 437(7057): 432-5.
[http://dx.doi.org/10.1038/nature04021] [PMID: 16079794]
[9]
Han X, Gui B, Xiong C, et al. Destabilizing LSD1 by Jade-2 promotes neurogenesis: An antibraking system in neural development. Mol Cell 2014; 55(3): 482-94.
[http://dx.doi.org/10.1016/j.molcel.2014.06.006] [PMID: 25018020]
[10]
Zhang Y, Wu T, Wang Y, et al. The R251Q mutation of LSD1 promotes invasion and migration of luminal breast cancer cells. Int J Biol Macromol 2020; 164: 4000-9.
[http://dx.doi.org/10.1016/j.ijbiomac.2020.08.221] [PMID: 32882276]
[11]
Lim S, Janzer A, Becker A, et al. Lysine-specific demethylase 1 (LSD1) is highly expressed in ER-negative breast cancers and a biomarker predicting aggressive biology. Carcinogenesis 2010; 31(3): 512-20.
[http://dx.doi.org/10.1093/carcin/bgp324] [PMID: 20042638]
[12]
Metzger E, Wissmann M, Yin N, et al. LSD1 demethylates repressive histone marks to promote androgen-receptor-dependent transcription. Nature 2005; 437(7057): 436-9.
[http://dx.doi.org/10.1038/nature04020] [PMID: 16079795]
[13]
Singh MM, Manton CA, Bhat KP, et al. Inhibition of LSD1 sensitizes glioblastoma cells to histone deacetylase inhibitors. Neuro-Oncol 2011; 13(8): 894-903.
[http://dx.doi.org/10.1093/neuonc/nor049] [PMID: 21653597]
[14]
Yi L, Cui Y, Xu Q, Jiang Y. Stabilization of LSD1 by deubiquitinating enzyme USP7 promotes glioblastoma cell tumorigenesis and metastasis through suppression of the p53 signaling pathway. Oncol Rep 2016; 36(5): 2935-45.
[http://dx.doi.org/10.3892/or.2016.5099] [PMID: 27632941]
[15]
Ding J, Zhang ZM, Xia Y, et al. LSD1-mediated epigenetic modification contributes to proliferation and metastasis of colon cancer. Br J Cancer 2013; 109(4): 994-1003.
[http://dx.doi.org/10.1038/bjc.2013.364] [PMID: 23900215]
[16]
Liu YW, Xia R, Lu K, et al. LincRNAFEZF1-AS1 represses p21 expression to promote gastric cancer proliferation through LSD1- Mediated H3K4me2 demethylation. Mol Cancer 2017; 16(1): 39.
[http://dx.doi.org/10.1186/s12943-017-0588-9] [PMID: 28209170]
[17]
Zheng YC, Ma J, Wang Z, et al. A systematic review of histone lysine-specific demethylase 1 and its inhibitors. Med Res Rev 2015; 35(5): 1032-71.
[http://dx.doi.org/10.1002/med.21350] [PMID: 25990136]
[18]
Zheng Y-C, Ma J-L, Liu Y, Liu HM. Writers and erasers of histone lysine methylation with clinically applied modulators: Promising target for cancer therapy. Curr Pharm Des 2016; 22(39): 5943-7.
[http://dx.doi.org/10.2174/1381612822666160715125417] [PMID: 27426130]
[19]
Zheng YC, Yu B, Chen ZS, Liu Y, Liu HM. TCPs: Privileged scaffolds for identifying potent LSD1 inhibitors for cancer therapy. Epigenomics 2016; 8(5): 651-66.
[http://dx.doi.org/10.2217/epi-2015-0002] [PMID: 27102879]
[20]
Zheng YC, Duan YC, Ma JL, et al. Triazole-dithiocarbamate based selective Lysine Specific Demethylase 1 (LSD1) inactivators inhibit gastric cancer cell growth, invasion, and migration. J Med Chem 2013; 56(21): 8543-60.
[http://dx.doi.org/10.1021/jm401002r] [PMID: 24131029]
[21]
Zheng YC, Shen DD, Ren M, et al. Baicalin, a natural LSD1 inhibitor. Bioorg Chem 2016; 69: 129-31.
[http://dx.doi.org/10.1016/j.bioorg.2016.10.004] [PMID: 27814566]
[22]
Duan YC, Guan YY, Zhai XY, et al. Discovery of resveratrol derivatives as novel LSD1 inhibitors: Design, synthesis and their biological evaluation. Eur J Med Chem 2017; 126: 246-58.
[http://dx.doi.org/10.1016/j.ejmech.2016.11.035] [PMID: 27888721]
[23]
Huang J, Sengupta R, Espejo AB, et al. p53 is regulated by the lysine demethylase LSD1. Nature 2007; 449(7158): 105-8.
[http://dx.doi.org/10.1038/nature06092] [PMID: 17805299]
[24]
Scoumanne A, Chen X. The lysine-specific demethylase 1 is required for cell proliferation in both p53-dependent and -independent manners. J Biol Chem 2007; 282(21): 15471-5.
[http://dx.doi.org/10.1074/jbc.M701023200] [PMID: 17409384]
[25]
Kontaki H, Talianidis I. Lysine methylation regulates E2F1-induced cell death. Mol Cell 2010; 39(1): 152-60.
[http://dx.doi.org/10.1016/j.molcel.2010.06.006] [PMID: 20603083]
[26]
Clements EG, Mohammad HP, Leadem BR, et al. DNMT1 modulates gene expression without its catalytic activity partially through its interactions with histone-modifying enzymes. Nucleic Acids Res 2012; 40(10): 4334-46.
[http://dx.doi.org/10.1093/nar/gks031] [PMID: 22278882]
[27]
Jin L, Hanigan CL, Wu Y, et al. Loss of LSD1 (lysine-specific demethylase 1) suppresses growth and alters gene expression of human colon cancer cells in a p53- and DNMT1(DNA methyltransferase 1)-independent manner. Biochem J 2013; 449(2): 459-68.
[http://dx.doi.org/10.1042/BJ20121360] [PMID: 23072722]
[28]
Cho HS, Suzuki T, Dohmae N, et al. Demethylation of RB regulator MYPT1 by histone demethylase LSD1 promotes cell cycle progression in cancer cells. Cancer Res 2011; 71(3): 655-60.
[http://dx.doi.org/10.1158/0008-5472.CAN-10-2446] [PMID: 21115810]
[29]
Niebel D, Kirfel J, Janzen V, Höller T, Majores M, Gütgemann I. Lysine-Specific Demethylase 1 (LSD1) in hematopoietic and lymphoid neoplasms. Blood 2014; 124(1): 151-2.
[http://dx.doi.org/10.1182/blood-2014-04-569525] [PMID: 24993879]
[30]
Harris WJ, Huang X, Lynch JT, et al. The histone demethylase KDM1A sustains the oncogenic potential of MLL-AF9 leukemia stem cells. Cancer Cell 2012; 21(4): 473-87.
[http://dx.doi.org/10.1016/j.ccr.2012.03.014] [PMID: 22464800]
[31]
Schenk T, Chen WC, Göllner S, et al. Inhibition of the LSD1 (KDM1A) demethylase reactivates the all-trans-retinoic acid differentiation pathway in acute myeloid leukemia. Nat Med 2012; 18(4): 605-11.
[http://dx.doi.org/10.1038/nm.2661] [PMID: 22406747]
[32]
Liang Y, Vogel JL, Narayanan A, Peng H, Kristie TM. Inhibition of the histone demethylase LSD1 blocks alpha-herpesvirus lytic replication and reactivation from latency. Nat Med 2009; 15(11): 1312-7.
[http://dx.doi.org/10.1038/nm.2051] [PMID: 19855399]
[33]
Wu L, He C, Qian D-Q, Shen B, Yao W. Cyclopropylamines as LSD1 inhibitors. US2017158633, 2017.
[34]
Wu L, He C, Qian D-Q, Shen B, Yao W. Cyclopropylamines as LSD1 inhibitors. EP3392244, 2016.
[35]
Rocco W L, Liu Y, Li M, Shah T, Wu H. Formulations of an LSD1 inhibitor. US20190307736, 2019.
[36]
Liu H, Li J, Zhu W, et al. Trans-indoline cyclopropylamine chemical compound, and method for preparation, pharmaceutical composition, and use thereof. US20190210998, 2019.
[37]
A BJ, Thor FMC. Lysine demethylase inhibitors for diseases and disorders associated with Flaviviridae. US9790196, 2017.
[38]
Maes T, Arjol CB. Selective LSD1 and dual LSD1/MAO-B inhibitors for modulating diseases associated with alterations in protein conformation. US20160081947, 2016.
[39]
Fyfe MCT, Maes T, Pedemonte MM, De la Cuesta ITF. Lysine demethylase inhibitors for myeloproliferative or lymphoproliferative diseases or disorders. US2017209432, 2017.
[40]
Fyfe MCT, Muñoz AO, Laria JC-P, Pedemonte MM, Estiarte- Martinez MDLA, Vidal NV. Cyclopropylamine derivatives useful as LSD1 inhibitors. US2016052865, 2016.
[41]
Muñoz AO, Fyfe MCT, Pedemonte MM, et al. (Hetero) aryl cyclopropylamine compounds as LSD1 inhibitors. US10329256, 2019.
[42]
Vaisburg A, Marx MA. LSD1 inhibitors. US2017129857, 2017.
[43]
Albrecht BK, Audia JE, Côté A, et al. LSD1 inhibitors and uses thereof. US20180290976, 2018.
[44]
Maes T, Crusat CM, Pozo DR. Method of treating multiple schlerosis employing a LSD1-inhibitor. US20190083469, 2019.
[45]
Zhang H. Novel suicidal LSD1 inhibitors targeting SOX2-expressing. US2016120862, 2016.
[46]
Lifang Y, Yubo Z, Sendong L, et al. 1,1a,6,6a- Tetrahydrocyclopropyl - [a] indene -1- amine derivatives and their preparation and application. CN109535019, 2019.
[47]
González EC, Maes T, Crusat CM, Mun0z AO. Methods to determine KDM1A target engagement and chemoprobes useful therefor. CN106045862, 2019.
[48]
Varasi M, Amici R, Thaler F, Mercurio C, Vianello P, Pasi M. Spirocyclopropylamine derivatives useful as inhibitors of histone demethylases KDM1A. WO2017109061, 2017.
[49]
Biel JH, Warawa EJ. N-cyclopropyl-n'-furfuryl-n'-methyl ethylene diamines. US3471522, 1969.
[50]
Lihua H, Jiawen G, Mingjie H, YunDong F, Yazhen Y. Aromatic ring/aromatic heterocycle-triazole-methylene-TCP derivative and preparation method and application thereof. CN111454252,
[51]
Wu L, Konkol LC, Lajkiewicz N, et al. Imidazopyridines and imidazopyrazines as LSD1 inhibitors. US2019040058, 2019.
[52]
Wu L, Courter JR, He C, et al. Midazopyrazines as LSD1 inhibitors. US20190106426, 2019.
[53]
He C, Li Z, Wu L, Yao W, Zhang F. Eterocyclic compounds as LSD1 inhibitors. US2019055250, 2019.
[54]
Geng PF, Li ZH, Liu HM, et al. Pyrimidine-containing triazole LSD1 inhibitor, preparation method and application thereof. CN106432248, 2017.
[55]
Geng PF, Li ZH, Liu HM, Suo FZ, Zhang T, Zheng YC. Inhibitor with pyrimido-triazole-tetrazole-thione LSD1 (lysine specific demethylase 1), preparation method of inhibitor and application. CN107033148, 2017.
[56]
Geng PF, Li ZH, Liu HM, Suo FZ, Zhang T, Zheng YC. Pyrimidine-containing triazole LSD1 inhibitor, preparation method and application. CN106928235, 2017.
[57]
Liu HM, Wang SA, Yu B, Zhao LJ, Zheng JX. LSD1 inhibitor of pyrimidine-1,2,4-triazole, its preparation method and application. CN106478639, 2017.
[58]
Li ZR, Liu HM, Wang SA, Yu B. yrimidine-1,2,4-triazole compound and preparation method and application thereof. CN109265462, 2019.
[59]
Liu HM, Yu B, Wang SA, Li ZR. Pyrimido-1,2,4-triazolium compound and preparation method and application thereof. CN109293664, 2019.
[60]
Hu B, Liu HM, Ma LY, et al. Pyrimidine derivative containing amino-based urea and terminal alkyne structural unit, preparation method and application thereof. CN104119280, 2014.
[61]
Wu L, Wang X, Yao W, Zhang C. Triazolopyridines and triazolopyrazines as LSD1 inhibitors. US20190152976, 2019.
[62]
Wu L, Sun Y, Wang X, Yao W, Zhang C. Triazolopyridines and triazolopyrazines as LSD1 inhibitors. US20190119272, 2019.
[63]
Liu HM, Liu YJ, Ma LY, et al. Application of 5- cyano-6-phenyl-pyrimidine compound with triazolyl in LSD1 inhibition and LSD1 inhibitor. CN110478352, 2019.
[64]
Duan YC, Guan YY, Liu W, Zhai XY, Zheng YC. A class of resveratrol derivatives, their preparation methods and their application as inhibitors of LSD1. CN106045881, 2016.
[65]
Li F, Liu JF, Ma TF, Wen TY, Zheng YC. Application of α-, β- and γ-mangostin as LSD1 inhibitors in medicine. CN109453161, 2019.
[66]
Kang D, Xu YG, Zha XM, Zhou C. Application of 2-([1,1'-biphenyl]-4-yl)-2-oxoethyl 4-((3-chloro-4-methylphenyl) amino)-4-oxobutanoate in the preparation of LSD1 inhibitors. CN103893163, 2014.
[67]
Dayong S, Lijun W, Bo J, Ning W, Shuaiyu W. A class of LSD1 inhibitors and their applications. CN103961340, 2019.
[68]
Zhongxiang Z, Yuanyuan X, Yingzhong C, et al. Application of piperazine-containing structural compounds in the preparation of LSD1 inhibitors. CN107174584, 2017.
[69]
Zhongxiang Z, Yuanyuan X, Yingzhong C, et al. Histone demethylase LSD1 inhibitor. CN107176927, 2017.
[70]
Yingchao D, Yuanyuan G, Zhaomin L, Xiaoyu Z, Wenping Q. Preparation method and application of a class of 2-phenyl-4-styrylpyridine LSD1 inhibitors. CN107474011, 2017.
[71]
Yingchao D, Xiaoyu Z, Yuanyuan G, Zhiyu X, Wenping Q, Yichao Z. Preparation method and application of a class of trans diarylethene LSD1 inhibitors. CN107501169, 2017.
[72]
Hongmin L, Bin Y, Qisheng M, Yichao Z, Fengzhi S. Astragalus LSD1 inhibitor and preparation method and application thereof. CN107936022, 2018.
[73]
Woster PM. Aminotriazole-and aminotetrazole-based kdm1a inhibitors as epigenetic modulators. US2017001968, 2017.
[74]
Du-Cuny L, He F, Xiao Q, Guoliang X, Zheng Q. Cyano-substituted indole compounds and uses thereof as LSD1 inhibitors. US20190092724, 2019.
[75]
Wang S, Zhang D, Zheng C, et al. Pyrrolo[2,3-c]pyridines and related analogs as LSD-1 inhibitors. WO2018213211, 2018.
[76]
Suzuki T, Itoh Y, Ogasawara D, et al. LSD1-selective inhibitor having lysine structure. US20160039748, 2016.
[77]
Vankayalapati H, Sorna V, Warner S L, Stephens B, Bearss D J, Sharma S. Substituted (E)-N′-(1-phenylethylidene) benzohydrazide analogs as histone demethylase inhibitors. US9266838, 2016.
[78]
Vankayalapati H, Sharma S, Liu X, Coburn C. Substituted benzohydrazide analogs as histone demethylase inhibitors. US20170001970, 2017.
[79]
Xiaoming C, Fei L, Siyuan X, Chen Z, Yungen X. Barbituric acid compound, preparation method and application thereof. CN105541806, 2016.
[80]
Vankayalapati H, Sharma S, Liu X, Kasibhatla S. Substituted 1-h-indol-3-yl-benzamide and 1, 1'-biphenyl analogs as histone demethylase inhibitors. US20170283397, 2017.
[81]
M.J. M, Rienhoff JHY, Michael C, Kasibhatla S. KDM1A inhibitors for the treatment of disease. US2017334873, 2017.
[82]
Rienhoff H Y. Lysine-specific histone demethylase as a novel therapeutic target in myeloproliferative neoplasms. US20190070172, 2019.
[83]
Albrecht BK, Audia JE, Cote A, et al. Therapeutic compounds and uses thereof. US20180009735, 2019.
[84]
Brucelle F, Gehling V S, Khanna A. LSD1 inhibitors and medical uses thereof. US20190274998, 2019.
[85]
Casero RA, Bytheway I, Woster PM. Small molecules as epigenetic modulators of lysine-specific demethylase 1 and methods of treating disorders. US9527805, 2016.
[86]
Quan J, Li Q, Zeng X, et al. Compound simultaneously inhibiting LSD1 and HDAC targets and application thereof. WO2019134087, 2019.
[87]
Casero RA, Woster PM. (Bis) urea and (bis) thiourea compounds as epigenic modulators of lysine-specific demethylase 1 and methods of treating disorders. US9708255, 2017.
[88]
He XR, Hui Z, Xie T, Ye XY. Preparation and application of substituted benzofuran 2-formyl hydrazone LSD1 inhibitor. CN111072610, 2020.
[89]
Li ZH, Ma JL, Qin TT, et al. Medical application of sesquiterpene compound used as LSD1 inhibitor and in preparation of antitumor drug. CN111592513, 2020.
[90]
Chen XH, Lou Y, Ma JL, et al. Compound 3 beta-acetoxyl-atractylenolide I, preparation method and application. CN111892563, 2020.
[91]
Li Z, Ma Y, Yin F. Stable polypeptide inhibitor derived from SNAIL1 based on LSD1 substrate and application thereof. CN111944034, 2020.
[92]
Rao S, Milburn P. Lysine specific histone demethylase-1 inhibitors and uses therefor. AU2017323868, 2019.
[93]
Lin H, Li Z. The role of indole-2,3-dione in anti-neuroblastoma cell lines. CN110151759, 2019.
[94]
Huawei L, Huiqian Y, Yingzi H, Shan S, Ao L, Renjie C. Application of lysine-specific demethylase 1 inhibitor. CN104042616, 2014.
[95]
Furong L, Shuyan Z, Hongjie Y, Hui Q. Method for inducing directed differentiation of human induced pluripotent stem cells into pancreatic cells. CN104726395, 2015.
[96]
Sun Y, Wang J, Chang S, Wang D. Pharmaceutical use and drug product adopting LSD1 inhibitor for preventing and treating triple negative breast cancer. WO2017067454, 2017.
[97]
Peng Z, Fan Z. Application of a small molecule inhibitor of KDM1A in inhibiting tumor cell growth and metastasis. CN106727461, 2017.
[98]
Yu L, Chong C, Baohong W. Application of drug compound. CN109793742, 2019.
[99]
Shailubhai K. Use of a combinational therapy of LSD1 inhibitors with CDK2 inhibitors in the treatment of cancer. US20180311245, 2018.
[100]
Minucci S, Pelicci PG, Hosseini SA. Use of a combinational therapy of LSD1 inhibitors with p21 activators in the treatment of cancer. WO2018197583, 2018.
[101]
Sánchez MIA, Lunardi S, Maes T, Crusat CM. Biomarkers for determining responsiveness to LSD1 inhibitors. US20190256930, 2019.
[102]
Motz G, Mavrakias KJ, Liu J, et al. Immune effector cell therapies with enhanced efficacy. US20190298715, 2019.
[103]
Podardno P, Michaud J, Ferndale M, Descherman-Peltier Y, Seliger L. LSD1 inhibitor as an inducer of skeletal muscle hypertrophy. CN110087644, 2019.
[104]
Puceat M, Jebeniani I, Bonne G, Guenantin A-C, Vignier N. LSD1 inhibitors for the treatment and prevention of cardiomyopathies. WO2019068326, 2019.
[105]
Decaprio JA, Cheng J, Park D. Treating merkel cell carcinoma. WO2019075327, 2019.
[106]
Kristie T, Liang Y, Vogel J. Preventing or treating viral infection using an inhibitor of the LSD1 protein, a MAO inhibitor or an inhibitor of LSD1 and a MAO inhibitor. US9499821, 2016.
[107]
Laria JCCP, Muñoz AO, Guibourt N, Baker JA. Potent selective LSD1 inhibitors and dual LSD1/MAO-B inhibitors for antiviral use. US9616058, 2017.
[108]
Shi YG, Lian CG. Treatment of cancer. US20160271106, 2016.
[109]
Mazzarella L, Minucci S, Pelicci P G, Pallavi R, Durfort T. Combination of Caloric Restriction (CR) or IGF1/INSULIN receptor inhibitor with LSD1 inhibitor. WO2017097865, 2017.
[110]
Fang Y, Liao G, Yu B. LSD1/KDM1A inhibitors in clinical trials: Advances and prospects. J Hematol Oncol 2019; 12(1): 129.
[http://dx.doi.org/10.1186/s13045-019-0811-9] [PMID: 31801559]

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