242
Views
1
CrossRef citations to date
0
Altmetric
Patenting perspective

Steroid sulfatase inhibitors: the current landscape

ORCID Icon, , , , , , & ORCID Icon show all
Pages 453-472 | Received 27 Jan 2021, Accepted 25 Mar 2021, Published online: 19 Apr 2021

References

  • Garbacz WG, Jiang M, Xie W. Sex-dependent role of estrogen sulfotransferase and steroid sulfatase in metabolic homeostasis.
  • Gratton AM, Ye L, Brownfoot FC, et al. Steroid sulfatase is increased in the placentas and whole blood of women with early-onset preeclampsia. Placenta. 2016;48:72–79.
  • Shah R, Singh J, Singh D, et al. Sulfatase inhibitors for recidivist breast cancer treatment: a chemical review. Eur J Med Chem. 2016;114:170–190.
  • Sang X, Han H, Poirier D, et al. Steroid sulfatase inhibition success and limitation in breast cancer clinical assays: an underlying mechanism. J Steroid Biochem Mol Biol. 2018;183:80–93.
  • Shin S, Im HJ, Kwon YJ, et al. Human steroid sulfatase induces Wnt/β-catenin signaling and epithelial-mesenchymal transition by upregulating Twist1 and HIF-1α in human prostate and cervical cancer cells. Oncotarget. 2017;8(37):61604–61617.
  • Mungenast F, Aust S, Vergote I, et al. Clinical significance of the estrogen-modifying enzymes steroid sulfatase and estrogen sulfotransferase in epithelial ovarian cancer. Oncol Lett. 2017;13:4047–4054.
  • Zhang M, Huang H, Lin N, et al. X-linked ichthyosis: molecular findings in four pedigrees with inconspicuous clinical manifestations. J Clin Lab Anal. 2020;34:e23201.
  • Malik A, Amer AB, Salama M, et al. X-linked ichthyosis associated with psychosis and behavioral abnormalities: a case report. J Med Case Rep. 2017;1125(1):267.
  • Kent L, Emerton J, Bhadravathi V, et al. X-linked ichthyosis (steroid sulfatase deficiency) is associated with increased risk of attention deficit hyperactivity disorder, autism and social communication deficits. J Med Genet. 2008;45(8):519–524.
  • Chatterjee S, Humby T, Davies W. Behavioural and psychiatric phenotypes in men and boys with x-linked ichthyosis: evidence from a worldwide online survey. PLoS ONE. 2016;11(10):e0164417.
  • Cavenagh A, Chatterjee S, Davies W. Behavioural and psychiatric phenotypes in female carriers of genetic mutations associated with X-linked ichthyosis. PLoS ONE. 2019;14(2):e0212330.
  • Humby T, Davies W. Brain gene expression in a novel mouse model of postpartum mood disorder. Transl Neurosci. 2019;10(1):168–174.
  • Daśko M, Demkowicz S, Biernacki K, et al. Recent progress in the development of steroid sulphatase inhibitors–examples of the novel and most promising compounds from the last decade. J Enz Inhibit Med Chem. 2020;35(1):1163–1184.
  • Morozkina SN, Shavva AG. Estrone sulfatase inhibitors as new anticancer agents. Chem Bio Activity of Steroids. Published online2019. DOI: 10.5772/intechopen.85850
  • Reed MJ, Purohit A, Woo LWL, et al. Steroid sulfatase: molecular biology, regulation, and inhibition. Endocr Rev. 2005;26:171–202.
  • Preusser-Kunze A, Mariappan M, Schmidt B, et al. Molecular characterization of the human Cα-formylglycine-generating enzyme. J Biol Chem. 2005;280(15):14900–14910.
  • Hernandez-Guzman FG, Higashiyama T, Pangborn W, et al. Structure of human estrone sulfatase suggests functional roles of membrane association. J Biol Chem. 2003;278(25):22989–22997.
  • Hernandez-Guzman FG, Higashiyama T, Osawa Y, et al. Purification, characterization and crystallization of human placental estrone/dehydroepiandrosterone sulfatase, a membrane-bound enzyme of the endoplasmic reticulum. J Steroid Biochem Mol Biol. 2001;78(5):441–450.
  • Miki Y, Nakata T, Suzuki T, et al. Systemic distribution of steroid sulfatase and estrogen sulfotransferase in human adult and fetal tissues. J Clin Endocrinol Metab. 2002;87(12):5760–5768.
  • Salido EC, Yen PH, Barajas L, et al. Steroid sulfatase expression in human placenta: immunocytochemistry and in situ hybridization study. J Clin Endocrinol Metab. 1990;70(6):1564–1567.
  • Nardi A, Pomari E, Zambon D, et al. Transcriptional control of human steroid sulfatase. J Steroid Biochem Mol Biol. 2009;115(1–2):68–74.
  • Compagnone NA, Salido E, Shapiro LJ, et al. Expression of steroid sulfatase during embryogenesis. Endocrinology. 1997;138(11):4768–4773.
  • Farnsworth WE. Human prostatic dehydroepiandrosterone sulfate sulfatase. Steroids. 1973;21(5):647–664.
  • Klein H, Molwitz T, Bartsch W. Steroid sulfate sulfatase in human benign prostatic hyperplasia: characterization and quantification of the enzyme in epithelium and stroma. J Steroid Biochem. 1989;33(2):195–200.
  • Baek WS, Aypar U. Neurological manifestations of X-linked Ichthyosis: case report and review of the literature. Case Rep Genet. 2017;2017:9086408.
  • Mueller JW, Gilligan LC, Idkowiak J, et al. The regulation of steroid action by sulfation and desulfation. Endocr Rev. 2015;36(5):526–563.
  • Bi Y, Jiang M, Guo W, et al. Sex-dimorphic and sex hormone–dependent role of steroid sulfatase in adipose inflammation and energy homeostasis. Endocrinology. 2018;159(9):3365–3377.
  • Jiang M, He J, Kucera H, et al. Hepatic overexpression of steroid sulfatase ameliorates mouse models of obesity and type 2 diabetes through sex-specific mechanisms. J Biol Chem. 2014;289(12):8086–8097.
  • Davies W. The steroid sulfate axis and its relationship to maternal behaviour and mental health. J Mol Endocrinol. 2018;61(2):T199–T210.
  • Qaiser MZ, Dolman DEM, Begley DJ, et al. Uptake and metabolism of sulphated steroids by the blood–brain barrier in the adult male rat. J Neurochem. 2017;142(5):672–685.
  • Gibson DA, Foster PA, Simitsidellis I, et al. A role for steroid sulphatase in intracrine regulation of endometrial decidualisation. J Mol Endocrinol. 2018;61(2):M57–M65.
  • Labrie F. All sex steroids are made intracellularly in peripheral tissues by the mechanisms of intracrinology after menopause. J Steroid Biochem Mol Biol. 2015;145:133–138.
  • Paatela H, Wang F, Vihma V, et al. Steroid sulfatase activity in subcutaneous and visceral adipose tissue: a comparison between pre-and postmenopausal women. Eur J Endocrinol. 2016;174(2):167–175.
  • Idkowiak J, Taylor AE, Subtil S, et al. Steroid sulfatase deficiency and androgen activation before and after puberty. J Clin Endocrinol Metab. 2016;101(6):2545–2553.
  • Feng Y, Xie Y, Xu M, et al. Hepatic steroid sulfatase critically determines estrogenic activities of conjugated equine estrogens in human cells in vitro and in mice. J Biol Chem. 2019;294(32):12112–12121.
  • Kurogi K, Yoshihama M, Williams FE, et al. Identification of zebrafish steroid sulfatase and comparative analysis of the enzymatic properties with human steroid sulfatase. J Steroid Biochem Mol Biol. 2019;185:110–117.
  • Liu X, Bai N, Kong X. Genetic analysis of a rare case with Kallman syndrome and steroid sulfatase deficiency. Zhonghua Yi Xue Yi Chuan Xue Za Zhi. 2016;33(3):349–352.
  • Cañueto J, Ciria S, Hernández-Martín A, et al. Analysis of the STS gene in 40 patients with recessive X-linked ichthyosis: a high frequency of partial deletions in a Spanish population. J Eur Acad Dermatol Venereol. 2010;24(10):1226–1229.
  • Alperin ES, Shapiro LJ. Characterization of point mutations in patients with X-linked ichthyosis: effects on the structure and function of the steroid sulfatase protein. J Biol Chem. 1997;272(33):20756–20763.
  • Crane JS, Paller AS Ichthyosis X-linked. statpearls publishing - statpearls treasure Island (FL) 2017. [cited 2021 Mar 23]. Available from:https://www.ncbi.nlm.nih.gov/books/NBK448149/
  • Humby T, Fisher A, Allen C, et al. A genetic variant within STS previously associated with inattention in boys with attention deficit hyperactivity disorder is associated with enhanced cognition in healthy adult males. Brain Behav. 2017;7(3):e00646.
  • Brookes KJ, Hawi Z, Park J, et al. Polymorphisms of the steroid sulfatase (ST) gene are associated with attention deficit hyperactivity disorder and influence brain tissue mRNA expression. Am J Med Genet Part B Neuropsychiatric Genetics. 2010;153B(8):1417–1424.
  • Stergiakouli E, Langley K, Williams H, et al. Steroid sulfatase is a potential modifier of cognition in attention deficit hyperactivity disorder. Genes Brain Behav. 2011;10(3):334–344.
  • Wang LJ, Chan WC, Chou MC, et al. Polymorphisms of STS gene and SULT2A1 gene and neurosteroid levels in Han Chinese boys with attention-deficit/hyperactivity disorder: an exploratory investigation. Sci Rep. 2017;7(1):45595.
  • Yue XH, Tong JQ, Wang ZJ, et al. Steroid sulfatase inhibitor DU-14 protects spatial memory and synaptic plasticity from disruption by amyloid β protein in male rats. Horm Behav. 2016;83:83–92.
  • Rižner TL. The important roles of steroid sulfatase and sulfotransferases in gynecological diseases. Front Pharmacol. 2016;7:7.
  • Secky L, Svoboda M, Klameth L, et al. The sulfatase pathway for estrogen formation: targets for the treatment and diagnosis of hormone-associated tumors. J Drug Deliv. 2013;2013:957605.
  • Valenzuela Scheker E, Kathuria A, Esnakula A, et al. Expression of key androgen-activating enzymes in ovarian steroid cell tumor, not otherwise specified. J Investig Med High Impact Case Rep. 2020;8:232470962093341.
  • Piccinato CA, Malvezzi H, Gibson DA, et al. Contribution of intracrine oestrogens to the aetiology of endometriosis. J Mol Endocrinol. 2018;61(2):T253–T270.
  • Cornel KMC, Delvoux B, Saya T, et al. The sulfatase pathway as estrogen supply in endometrial cancer. Steroids. 2018;139:45–52.
  • Nishimoto M, Toyoshima M, Shiga N, et al. Steroid sulfatase inhibitor reduces proliferation of ishikawa endometrial cancer cells in co-culture systems. Open J Endocr Metab Dis. 2016;06(9):193–204.
  • Piccinato CA, Neme RM, Torres N, et al. Effects of steroid hormone on estrogen sulfotransferase and on steroid sulfatase expression in endometriosis tissue and stromal cells. J Steroid Biochem Mol Biol. 2016;158:117–126.
  • Lee WM, Jang KS, Bae J, et al. The role of steroid sulfatase as a prognostic factor in patients with endometrial cancer. Yonsei Med J. 2016;57(3):754–760.
  • Ye DJ, Kwon YJ, Shin S, et al. Induction of integrin signaling by steroid sulfatase in human cervical cancer cells. Biomol Ther. 2017;25(3):321–328.
  • Wang JG, Miyazu M, Xiang P, et al. Stretch-induced cell proliferation is mediated by FAK-MAPK pathway. Life Sci. 2005;76(24):2817–2825.
  • Cance WG, Harris JE, Iacocca MV, et al. Immunohistochemical analyses of focal adhesion kinase expression in benign and malignant human breast and colon tissues: correlation with preinvasive and invasive phenotypes. Clin Cancer Res. 2000;6(6):2417–2423.
  • Meng XN, Jin Y, Yu Y, et al. Characterisation of fibronectin-mediated FAK signalling pathways in lung cancer cell migration and invasion. Br J Cancer. 2009;101(2):327–334.
  • Sang X, Han H, Li T, et al. Mutual regulations and breast cancer cell control by steroidogenic enzymes: dual sex-hormone receptor modulation upon 17β-HSD7 inhibition. J Steroid Biochem Mol Biol. 2019;193:105411.
  • McNamara KM, Guestini F, Sauer T, et al. In breast cancer subtypes steroid sulfatase (STS) is associated with less aggressive tumour characteristics/692/4028/67/1347/631/67/1459 article. Br J Cancer. 2018;118(9):1208–1216.
  • Wu Y, Tang L, Azabdaftari G, et al. Adrenal androgens rescue prostatic dihydrotestosterone production and growth of prostate cancer cells after castration. Mol Cell Endocrinol. 2019;486:79–88.
  • Nakamura Y, Ise K, McNamara KM, et al. The expression of sex steroid receptors and sex steroid–synthesizing/metabolizing enzymes in metastasized lymph nodes of prostate cancer. Hum Pathol. 2019;84:124–132.
  • Penning TM. Dehydroepiandrosterone (DHEA)-SO4 depot and castration-resistant prostate cancer. In: Vitamins and hormones.
  • Suh B-Y, Jung -J-J, Park N, et al. Induction of steroid sulfatase expression by tumor necrosis factor-α through phosphatidylinositol 3-kinase/Akt signaling pathway in PC-3 human prostate cancer cells. Exp Mol Med. 2011;4328(11):646–652.
  • Shimizu Y, Tamada S, Kato M, et al. Steroid sulfatase promotes invasion through epithelial-mesenchymal transition and predicts the progression of bladder cancer. Exp Ther Med. 2018;16(6):4463–4470.
  • Frycz BA, Murawa D, Borejsza-Wysocki M, et al. mRNA expression of steroidogenic enzymes, steroid hormone receptors and their coregulators in gastric cancer. Oncol Lett. 2017;13(5):3369–3378.
  • Gilligan LC, Rahman HP, Hewitt A-M, et al. Estrogen activation by steroid sulfatase increases colorectal cancer proliferation via GPER. J Clin Endocrinol Metab. 2017;102(12):4435–4447.
  • Gilligan LC, Gondal A, Tang V, et al. Estrone sulfate transport and steroid sulfatase activity in colorectal cancer: implications for hormone replacement therapy. Front Pharmacol. 2017;835:103.
  • Sun N, Kunzke T, Sbiera S, et al. Prognostic relevance of steroid sulfation in adrenocortical carcinoma revealed by molecular phenotyping using high-resolution mass spectrometry imaging. Clin Chem. 2019;65115(10):1276–1286.
  • Whitehead CL, Palmer KR, Nilsson U, et al. Placental expression of a novel primate-specific splice variant of sFlt-1 is upregulated in pregnancies complicated by severe early onset pre-eclampsia. BJOG. 2011;11833(10):1268–1271.
  • Pérez-Jiménez M, Sansigre P, Valladares A, et al. Steroid hormones sulfatase inactivation extends lifespan and ameliorates age-related diseases. Nat Commun. 2021;12(1):49.
  • Reddy DS. Neurosteroids: endogenous role in the human brain and therapeutic potentials. Prog Brain Res. 2010;186:113–137.
  • Honma N, Saji S, Mikami T, et al. Estrogen-related factors in the frontal lobe of alzheimer’s disease patients and importance of body mass index. Sci Rep. 2017;7(1):726.
  • Jiang M, Klein M, Zanger UM, et al. Inflammatory regulation of steroid sulfatase: a novel mechanism to control estrogen homeostasis and inflammation in chronic liver disease. J Hepatol. 2016;64(1):44–52.
  • Barbosa ACS, Feng Y, Yu C, et al. Estrogen sulfotransferase in the metabolism of estrogenic drugs and in the pathogenesis of diseases. Expert Opin Drug Metab Toxicol. 2019;1583(4):329–339.
  • Lerner A, Owens LA, Coates M, et al. Expression of genes controlling steroid metabolism and action in granulosa-lutein cells of women with polycystic ovaries. Mol Cell Endocrinol. 2019;4868:47–54.
  • Jiang M, Xu M, Ren S, et al. Transgenic overexpression of steroid sulfatase alleviates cholestasis. Liver Res. 2017;1(1):63–69.
  • Vihma V, Heinonen S, Naukkarinen J, et al. Increased body fat mass and androgen metabolism – a twin study in healthy young women. Steroids. 2018;140:24–31.
  • Kanda N, Hoashi T, Saeki H. The roles of sex hormones in the course of atopic dermatitis. Int J Mol Sci. 2019;20(19):4660.
  • Tofovic SP, Jackson EK. Estrogens in men: another layer of complexity of estradiol metabolism in pulmonary hypertension. Am J Respir Crit Care Med. 2016;193(10):1087–1089.
  • Konings GFJ, Reynaert NL, Delvoux B, et al. Increased levels of enzymes involved in local estradiol synthesis in chronic obstructive pulmonary disease. Mol Cell Endocrinol. 2017;443:23–31.
  • Mao X, Barger SW. Neuroprotection by dehydroepiandrosteronesulfate: role of an NFkappaB-like factor. NeuroReport. 1998;9(4):759–763.
  • Saldanha CJ, Burstein SR, Duncan KA. Induced synthesis of oestrogens by glia in the songbird brain. J Neuroendocrinol. 2013;25(11):1032–1038.
  • Duncan KA. Estrogen formation and inactivation following tbi: what we know and where we could go. Front Endocrinol. 2020;11:345.
  • Meingassner JG Combination of a steroid sulfatase inhibitor and ascomycin. WO2006097293.
  • Loumaye E, Cayron-Elizondo V, Gotteland J-P Use of steroid sulfatase inhibitors for the treatment of preterm labor. WO2010013187.
  • Potter BVL. SULFATION PATHWAYS: steroid sulfatase sulphatase inhibition via aryl sulfamatessulphamates: clinical progress, mechanism and future prospects. J Mol Endocrinol. 2018;61(2):T233–T252.
  • Parra-Guillen ZP, Cendrós Carreras JP, Peraire C, et al. Population pharmacokinetic modelling of Irosustat in postmenopausal women with oestrogen receptor positive breast cancer incorporating non-linear red blood cell uptake. Pharm Res. 2015;32(4):1493–1504.
  • Zaraei S-O, Abduelkarem AR, Anbar HS, et al. Sulfamates in drug design and discovery: pre-clinical and clinical investigations. Eur J Med Chem. 2019;179:257–271.
  • Palmieri C, Liu X, Stein R. IRIS study: a phase II study of the steroid sulfatase inhibitor Irosustat when added to an aromatase inhibitor in ER-positive breast cancer patients. Breast Cancer Res Treat. 2017;165(2):343–353.
  • Palmieri C, Szydlo R, Miller M. IPET study: an FLT-PET window study to assess the activity of the steroid sulfatase inhibitor irosustat in early breast cancer. Breast Cancer Res Treat. 2017;16664(2):527–539.
  • Munoz MJ, Carrion AM, Perez-Jimenez MM. COMPOSITIONS FOR TREATING AND/OR PREVENTING PROTEIN-AGGREGATION DISEASESS. WO 2019243453 A1.
  • Ruiz M, Jesus M, Perez-Jimenez MM Use of the inhibitor of steroid sulfatases STX64 for treating aging. ES 2514140.
  • Ruiz M, Jesus M, Perez-Jimenez MM. Use of the inhibitor of steroid sulfatases STX64 for treating aging. WO 2014154927 A1.
  • Sun M, Xie M, Qi N, et al. Application of irosustat in treating eimeria tenella. CN 102657642.
  • [cited 2021 Mar 23]. Available from:https://clinicaltrials.gov/ct2/results?cond=&term=irosustat&cntry=&state=&city=&dist=
  • Dasko M, Maslyk M, Kubinski K, et al. Synthesis and steroid sulfatase inhibitory activities of N-phosphorylated 3-(4-aminophenyl)- coumarin-7-O-sulfamates. MedChemComm. 2016;7(6):1146–1150.
  • Demkowicz S, Dasko M, Kozak W, et al. Synthesis and biological evaluation of fluorinated 3-phenylcoumarin-7-o-sulfamate 3-phenylcoumarin-7-O-sulfamate derivatives as steroid sulfatase inhibitors. Chem Biol Drug Des. 2016;87(2):233–238.
  • Dasko M, Przybylowska M, Rachon J, et al. Synthesis and biological evaluation of fluorinated N-benzoyl and N-phenylacetoyl derivatives of 3-(4-aminophenyl)-coumarin-7-O-sulfamate as steroid sulfatase inhibitors. Eur J Med Chem. 2017;128:79–87.
  • Ganeshapillai D, Woo LWL, Thomas MP, et al. C-3- and C-4-substituted bicyclic coumarin sulfamates as potent steroid sulfatase inhibitors. ACS Omega. 2018;3(9):10748–10772.
  • Dasko M, Demkowicz S, Biernacki K, et al. Novel steroid sulfatase inhibitors based on N-thiophosphorylated 3-(4-aminophenyl)-coumarin −7-O-sulfamate3-(4-aminophenyl)-coumarin-7-O-sulfamates. Drug Dev Res. 2019;80(6):857–866.
  • Hng Y, Lin M-H, Lin T-S, et al. Design and synthesis of 3-benzylaminocoumarin-7-O-sulfamate derivatives as steroid sulfatase inhibitors. Bioorg Chem. 2020;96:103618.
  • Poirier D, Maltais R Preparation of 2-benzoyl-1,2,3,4-tetrahydroisoquinolin-7-ol and 2-benzoyl-1,2,3,4-tetrahydroisoquinolin-7-yl sulfamate compounds for the treatment of hormone-dependent diseases. WO 2015024111 A1.
  • Ouellet C, Maltais R, Ouellet E, et al. Discovery of a sulfamate-based steroid sulfatase inhibitor with intrinsic selective estrogen receptor modulator properties. Eur J Med Chem. 2016;119:169–182.
  • Poirier D, Roy J, Maltais R, et al., Antisulfatase, osteogenic, and anticancer activities of steroid sulfatase inhibitor EO-33 in mice. J Med Chem. 62(11): 5512–5521. 2019.
  • El-Gamal MI, Semreen MH, Foster PA, et al. Design, synthesis, and biological evaluation of new arylamide derivatives possessing sulfonate or sulfamate moieties as steroid sulfatase enzyme inhibitors. Bioorg Med Chem. 2016;24(12):2762–2767.
  • Salah M, Abdelsamie AS, Frotscher M. First dual inhibitors of Steroid Sulfatase (STS) and 17β-hydroxysteroid dehydrogenase type 1 (17β-HSD1): designed multiple ligands as novel potential therapeutics for estrogen-dependent diseases. J Med Chem. 2017;60(9):4086–4092.
  • El-Gamal MI, Zaraei S-O, Foster PA, et al. A new series of aryl sulfamate derivatives: design, synthesis, and biological evaluation. Bioorg Med Chem. 2020;28(8):115406.
  • Kaise A, Ohta K, Endo Y. Novel p-carborane-containing multitarget anticancer agents inspired by the metabolism of 17β-estradiol. Bioorg Med Chem. 2017;25(24):6371–6378.
  • Kaise A, Ohta K, Shirata C, et al. Design and synthesis of p-carborane-containing sulfamates as multitarget anti-breast cancer agents. Bioorg Med Chem. 2017;25(24):6417–6426.
  • Dasko M, Rachon J, Maslyk M, et al. Synthesis and biological evaluation ofN-acylated N-acylated tyramine sulfamate sulfamates containing C-F bonds as steroid sulfatase inhibitors. Chem Biol Drug Des. 2017;90(1):156–161.
  • Park J-H ME. Synthesis of androstane-3β-O-sulfonate and -sulfamate derivatives inhibitory effect of steroid sulfatase. Yakhak Hoeji. 2018;62(6):401–409.
  • Bacsa I, Herman BE, Jojart R, et al. Synthesis and structure-activity structure–activity relationships of 2- and/or 4-halogenated 13β 13β- and 13α-estrone derivatives as enzyme inhibitors of estrogen biosynthesis. J Enz Inhibit Med Chem. 2018;33(1):1271–1282.
  • Gao AC, Li P-K Compounds, compositions, and methods for treatment of androgen-mediated disease. WO 2020092972 A1.
  • Maltais R, Djiemeny AN, Roy J, et al., Design and synthesis of dansyl-labeled inhibitors of steroid sulfatase for optical imaging. Bioorg Med Chem. 28(7): 115368. 2020.
  • Grienke U, Foster PA, Zwirchmayr J, et al. 1H NMR-MS-based heterocovariance as a drug discovery tool for fishing bioactive compounds out of a complex mixture of structural analogues. Sci Rep. 2019;9(1):11113.
  • Grienke U, Kaserer T, Kirchweger B, et al. Steroid sulfatase inhibiting lanostane triterpenes – structure activity relationship and in silico insights. Bioorg Chem. 2020;95:103495.
  • Dohle W, Jourdan FL, Menchon G, et al. Quinazolinone-based anticancer agents: synthesis, antiproliferative sar, antitubulin activity, and tubulin co-crystal structure. J Med Chem. 2018;61(3):1031–1044.
  • Dasko M, Demkowicz S, Rachon J, et al. New potent STS inhibitors inHibitors based on fluorinated 4-(1-phenyl-14-(1-pHenyl-1H-(1,2,3)-triazol-4-yl)-phenyl [1,2,3]triazol-4-yl)-pHenyl sulfamates. J Asian Nat Prod Res. 2019;22(11):1037–1044.
  • Demkowicz S, Dasko M, Rachon J Preparation of 4-(1-phenyl-1H-[1,2,3]triazol-4-yl)-phenol and sulfamate derivatives as steroid sulfatase inhibitors and estrogen receptor modulators as antimicrobial medicants and for treatment of cancer. WO 2019245393 A1.
  • Ciupak O, Dasko M, Biernacki K, et al. New potent steroid sulphatase inhibitors sulpHatase inHibitors based on 6-(1-phenyl-16-(1-pHenyl-1H-1,2,3-triazol-4-yl)naphthalen-2-yl sulphamate 1,2,3-triazol-4-yl)napHtHalen-2-yl sulpHamate derivatives. J Enz Inhibit Med Chem. 2021;36(1):238–247.
  • Moi D, Foster PA, Rimmer LG, et al. Synthesis and in vitro evaluation of piperazinyl-ureido sulfamates as steroid sulfatase inhibitors. Eur J Med Chem. 2019;182:111614.
  • Ballabio A, Sorrentino NC, Fraldi A. Therapy of sulfatase deficiencies. WO 2019207167A1 .

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.