Main Article Content

Abstract

The discovery, development and utility of selective estrogen receptor modulators (SERMs) are presented in this study. As per literature review SERMs used in the treatment of estrogen hormone responsive diseases like osteoporosis, Alzheimer, angiogenesis, hyperlipidemia, coronary heart disease, atherosclerosis, endometriosis, breast cancer, post-menopausal depression, dysfunctional uterine bleeding, gynacomastia, Albright syndrome, ovarian cancer, dyspareunia, cyclical mastalgia, hypogondism and induced ovulation in sub-fertile woman, etc. Basically world wide a large no. compounds available those function as SERMs successfully or under different phase clinical trials or discontinued because of unwanted side effects during clinical trials. This work describes the specific reference compounds which have created a substitute of estrogen for treating hormone dependent issues.

Keywords

Estrogen receptor Hyperlipidemia Alzheimer Hormone responsive disease Atherosclerosis

Article Details

How to Cite
Kulshrestha, A., & Pandey, J. (2019). Origination Progress and Utility of Selective Estrogen Receptor Modulators in Clinical Practice as an Efficient Substitute of Estrogen for Treating Hormone Dependent Issues. Asian Journal of Organic & Medicinal Chemistry, 4(3), 200–208. https://doi.org/10.14233/ajomc.2019.AJOMC-P188

References

  1. R. Clark, F. Leonessa, J.N. Welch and T.C. Skaar, Cellular and Molecular Pharmacology of Antiestrogen Action and Resistance, Pharmacol. Rev., 53, 25 (2001).
  2. K. Pettersson and J.A. Gustafsson, Role of Estrogen Receptor Beta in Estrogen Action, Annu. Rev. Physiol., 63, 165 (2001); https://doi.org/10.1146/annurev.physiol.63.1.165.
  3. H.U. Bryant, Selective Estrogen Receptor Modulators, Rev. Endocr. Metab. Disord., 3, 231 (2002); https://doi.org/10.1023/A:1020076426727.
  4. M.J. Meegan and D.G. Lloyd, Advances in the Science of Estrogen Receptor Modulation, Curr. Med. Chem., 10, 181 (2003); https://doi.org/10.2174/0929867033368501.
  5. V.C. Jordan, Antiestrogens and Selective Estrogen Receptor Modulators as Multifunctional Medicines. 1. Receptor Interactions, J. Med. Chem., 46, 883 (2003); https://doi.org/10.1021/jm020449y.
  6. B.L. Riggs and L.C. Hartmann, Selective Estrogen-Receptor Modu-lators-Mechanisms of Action and Application to Clinical Practice, N. Engl. J. Med., 348, 618 (2003); https://doi.org/10.1056/NEJMra022219.
  7. J.H. Cha, Y.-S. Cho, H.-Y. Koh, E. Lee, Y.-T. Kim and H. Yang, Prepar-ation of Chromane Derivatives via Indium-Mediated Intramolecular Allylation Reactions, Bull. Korean Chem. Soc., 25, 1123 (2004); https://doi.org/10.5012/bkcs.2004.25.8.1123.
  8. S. Agatonovic-Kustrin and J. Turner, Molecular Structural Character-istics of Estrogen Receptor Modulators as Determinants of Estrogen Receptor Selectivity, Mini Rev. Med. Chem., 8, 943 (2008); https://doi.org/10.2174/138955708785132747.
  9. B.L. Clarke and S. Khosla, New Selective Estrogen and Androgen Receptor Modulators, Curr. Opin. Rheumatol., 21, 374 (2009); https://doi.org/10.1097/BOR.0b013e32832ca447.
  10. F. Ninutolo, M. Macchia, B.S. Katzellenbogen and J.A. Katzellenbogen, Estrogen Receptor b Ligands: Recent Advances and Biomedical Appli-cations, J. A., Med. Res. Rev., 31, 364 (2011); https://doi.org/10.1002/med.20186.
  11. P.Y. Maximov, T.M. Lee and V.C. Jordan, The Discovery and Develop-ment of Selective Estrogen Receptor Modulators (SERMs) for Clinical Practice, Curr. Clin. Pharmacol., 8, 135 (2013); https://doi.org/10.2174/1574884711308020006.
  12. E. Hughes, J. Brown, J.J. Collins and P. Vanderkerchove, Clomiphene Citrate for Unexplained Subfertility in Women, Cochrane Database Syst. Rev., CD000057 (2010); https://doi.org/10.1002/14651858.CD000057.
  13. S. Yilmaz, N. Yilmaz Sezer, I.M. Gönenç, S.E. Ilhan and E. Yilmaz, Safety of Clomiphene Citrate: A Literature Review, Cytotechnology, 70, 489 (2018); https://doi.org/10.1007/s10616-017-0169-1.
  14. J.H. Pickar, T. MacNeil and K. Ohleth, SERMs: Progress and Future Perspectives, Maturitas, 67, 129 (2010); https://doi.org/10.1016/j.maturitas.2010.05.009.
  15. F.H. Duarte, R.S. Jallad and M.D. Bronstein, Estrogens and Selective Estrogen Receptor Modulators in Acromegaly, Endocrine, 54, 306 (2016); https://doi.org/10.1007/s12020-016-1118-z.
  16. V.M. Quirke, Tamoxifen from Failed Contraceptive Pill to Best-Selling Breast Cancer Medicine: A Case-Study in Pharmaceutical Innovation, Front. Pharmacol., 8, 620 (2017); https://doi.org/10.3389/fphar.2017.00620.
  17. A.Z. Steiner, M. Terplan and R.J. Paulson, Comparison of Tamoxifen and Clomiphene Citrate for Ovulation Induction: A Meta-Analysis, Hum. Reprod., 20, 1511 (2005); https://doi.org/10.1093/humrep/deh840.
  18. M.E. Chua, K.G. Escusa, S. Luna, L.C. Tapia, B. Dofitas and M. Morales, Revisiting Oestrogen Antagonists (Clomiphene or Tamoxifen) as Medical Empiric Therapy for Idiopathic Male Infertility: A Meta-Analysis, Andrology, 1, 749 (2013); https://doi.org/10.1111/j.2047-2927.2013.00107.x.
  19. T. Nakamura, Y. Imai, T. Matsumoto, S. Sato, K. Takeuchi, K. Igarashi, Y. Harada, Y. Azuma, A. Krust, Y. Yamamoto, H. Nishina, S. Takeda, H. Takayanagi, D. Metzger, J. Kanno, K. Takaoka, T.J. Martin, P. Chambon and S. Kato, Estrogen Prevents Bone Loss via Estrogen Receptor a and Induction of Fas Ligand in Osteoclasts, Cell, 130, 811 (2007); https://doi.org/10.1016/j.cell.2007.07.025.
  20. B.J.A. Furr and V.C. Jordan, The Pharmacology and Clinical Uses of Tamoxifen, Pharmacol. Ther., 25, 127 (1984); https://doi.org/10.1016/0163-7258(84)90043-3.
  21. V.C. Jordan, Selective Estrogen Receptor Modulation: A Personal Perspective, Cancer Res., 61, 5683 (2001).
  22. L.J. Lerner and V.C. Jordan, Development of Antiestrogens and their use in Breast Cancer: Eighth Cain Memorial Award Lecture, Cancer Res., 50, 4177 (1990).
  23. S. Massarweh, C.K. Osborne, C.J. Creighton, L. Qin, A. Tsimelzon, S. Huang, H. Weiss, M. Rimawi and R. Schiff, Tamoxifen Resistance in Breast Tumors is Driven by Growth Factor Receptor Signaling with Repression of Classic Estrogen Receptor Genomic Function, Cancer Res., 68, 826 (2008); https://doi.org/10.1158/0008-5472.CAN-07-2707.
  24. Y. Jin, Z. Desta, V. Stearns, B. Ward, H. Ho, K.H. Lee, T. Skaar, A.M. Storniolo, L. Li, A. Araba, R. Blanchard, A. Nguyen, L. Ullmer, J. Hayden, S. Lemler, R.M. Weinshilboum, J.M. Rae, D.F. Hayes and D.A. Flockhart, CYP2D6 Genotype, Antidepressant Use and Tamoxifen Metabolism During Adjuvant Breast Cancer Treatment, J. Natl. Cancer Inst., 97, 30 (2005); https://doi.org/10.1093/jnci/dji005.
  25. M.A. Gallo and D. Kaufman, Antagonistic and Agonistic Effects of Tamoxifen: Significance in Human Cancer, Semin. Oncol., 24, S1-71 (1997).
  26. J. Pandey and A. Kulshreshtha, Raloxifene: An Effective Selective Estrogen Receptor Modulator, EC Chemistry, 2, 92 (2015).
  27. D. Hochner-Celnikier, Pharmacokinetics of Raloxifene and its Clinical Application, Eur. J. Obstet. Gynecol. Reprod. Biol., 85, 23 (1999); https://doi.org/10.1016/S0301-2115(98)00278-4.
  28. H.U. Bryant, Mechanism of Action and Preclinical Profile of Raloxifene, A Selective Estrogen Receptor Modulator, Rev. Endocr. Metab. Disord., 2, 129 (2001); https://doi.org/10.1023/A:1010019410881.
  29. D. Thiebaud and R.J. Secrest, Selective Estrogen Receptor Modulators: Mechanism of Action and Clinical Experience. Focus on Raloxifene, Reprod. Fertil. Dev., 13, 331 (2001); https://doi.org/10.1071/RD00109.
  30. W. Lemmo, Anti-Estrogen Withdrawal Effect With Raloxifene? A Case Report, Integr. Cancer Ther., 15, 245 (2016); https://doi.org/10.1177/1534735416658954.
  31. V. Birzniece, S. Sutanto and K.K. Ho, Gender Difference in the Neuro-endocrine Regulation of Growth Hormone Axis by Selective Estrogen Receptor Modulators, J. Clin. Endocrinol. Metab., 97, E521 (2012); https://doi.org/10.1210/jc.2011-3347.
  32. T.H. Ho, R. Nunez-Nateras, Y.-X. Hou, A.H. Bryce, D.W. Northfelt, A.C. Dueck, B. Wong, M.L. Stanton, R.W. Joseph and E.P. Castle, A Study of Combination Bicalutamide and Raloxifene for Patients with Castration-Resistant Prostate Cancer, Clin. Genitourin. Cancer, 15, 196 (2017); https://doi.org/10.1016/j.clgc.2016.08.026.
  33. A. Blum, L. Hathaway, R. Mincemoyer, W.H. Schenke, G. Csako, M.A. Waclawiw, J.A. Panza and R.O. Cannon III, Hormonal, Lipoprotein, and Vascular Effects of the Selective Estrogen Receptor Modulator Raloxifene in Hypercholesterolemic Men, Am. J. Cardiol., 85, 1491 (2000); https://doi.org/10.1016/S0002-9149(00)00802-X.
  34. I.K. Morton and J.M. Hall, Concise Dictionary of Pharmacological Agents, Springer Science & Business Media, pp. 79 (1999).
  35. S. Gizzo, C. Saccardi, T.S. Patrelli, R. Berretta, G. Capobianco, S.D. Gangi, A. Vacilotto, A. Bertocco, M. Noventa, E. Ancona, D. D’Antona and G.B. Nardelli, Update on Raloxifene: Mechanism of Action, Clinical Efficacy, Adverse Effects and Contraindications, Obstet. Gynecol. Surv., 68, 467 (2013); https://doi.org/10.1097/OGX.0b013e31828baef9.
  36. R.C. Gupta, J.K. Paliwal, S. Nityanand, O.P. Asthana and J. Lal, Centchroman: A New Non-Steroidal Oral Contraceptive in Human Milk, Contraception, 52, 301 (1995); https://doi.org/10.1016/0010-7824(95)00214-U.
  37. A. Makker, I. Tandon, M.M. Goel, M. Singh and M.M. Singh, EFfect of Ormeloxifene, A Selective Estrogen Receptor Modulator, On Biomarkers of Endometrial Receptivity and Pinopode Development and its Relation to Fertility and Infertility in Indian Subjects, Fertil. Steril., 91, 2298 (2009); https://doi.org/10.1016/j.fertnstert.2008.04.018.
  38. J. Lal, Clinical Pharmacokinetics and Interaction of Centchroman-A Mini Review, Contraception, 81, 275 (2010); https://doi.org/10.1016/j.contraception.2009.11.007.
  39. J. Lal, S. Nitynand, O.P. Asthana, N.V. Nagaraja and R.C. Gupta, Optimization of Contraceptive Dosage Regimen of Centchroman, Contraception, 63, 47 (2001); https://doi.org/10.1016/S0010-7824(00)00189-X.
  40. M.M. Singh, Centchroman, A Selective Estrogen Receptor Modulator, as a Contraceptive and for the Management of Hormone-Related Clinical Disorders, Med. Res. Rev., 21, 302 (2001); https://doi.org/10.1002/med.1011.
  41. A. Kriplani, V. Kulshrestha and N. Agarwal, Efficacy and Safety of Ormeloxifene in Management of Menorrhagia: A Pilot Study, J. Obstet. Gynaecol. Res., 35, 746 (2009); https://doi.org/10.1111/j.1447-0756.2008.00987.x.
  42. A. Dhar and A. Srivastava, Role of Centchroman in Regression of Mastalgia and Fibroadenoma, World J. Surg., 31, 1180 (2007); https://doi.org/10.1007/s00268-007-9040-4.
  43. W. Shelly, M.W. Draper, V. Krishnan, M. Wong and R.B. Jaffe, Selective Estrogen Receptor Modulators: An Update on Recent Clinical Findings, Obstet. Gynecol. Surv., 63, 163 (2008); https://doi.org/10.1097/OGX.0b013e31816400d7.
  44. M. Nigam, V. Ranjan, S. Srivastava, R. Sharma and A.K. Balapure, Centchroman induces G0/G1 Arrest and Caspase-Dependent Apoptosis involving Mitochondrial Membrane Depolarization in MCF-7 and MDA MB-231 Human Breast Cancer Cells, Life Sci., 82, 577 (2008); https://doi.org/10.1016/j.lfs.2007.11.028.
  45. B. Patil, Communication Strategies In Reproductive Health, Discovery Publishing House Pvt. Ltd.: New Delhi (2009).
  46. V.T. DeVita, T.S. Lawrence and S.A. Rosenberg, DeVita, Hellman, and Rosenberg's Cancer Principles & Practice of Oncology, Wolters Kluwer Health Adis, p. 1126 (2015).
  47. J.M. Dixon, Endocrine Therapy in Breast Cancer, New J. Sci., 2014, Article ID 390618 (2014); https://doi.org/10.1155/2014/390618.
  48. D. Schiff, I. Arrilaga and P.Y. Wen, Cancer Neurology in Clinical Practice, Springer, p. 296 (2017).
  49. T. Qin, Z.Y. Yuan, R.J. Peng, Y.D. Zeng, Y.X. Shi, X.Y. Teng, D.G. Liu, B. Bai and S.S. Wang, Efficacy and Tolerability of Toremifene and Tamoxifen Therapy in Premenopausal Patients with Operable Breast Cancer: A Retrospective Analysis, Curr. Oncol., 20, 196 (2013); https://doi.org/10.3747/co.20.1231.
  50. J. Ellmen, P. Hakulinen, A. Partanen and D.F. Hayes, Estrogenic Effects of Toremifene and Tamoxifen in Postmenopausal Breast Cancer Patients, Breast Cancer Res. Treat., 82, 103 (2003); https://doi.org/10.1023/B:BREA.0000003957.54851.11.
  51. D.M. Biskobing, Update on Bazedoxifene: A Novel Selective Estrogen Receptor Modulator, Clin. Interv. Aging, 2, 299 (2007).
  52. J.D. Fan, B.L. Wagner and D.P. McDonnell, Identification of the Sequences within the Human Complement 3 Promoter Required for Estrogen Responsiveness Provides Insight into the Mechanism of Tamoxifen Mixed Agonist Activity, Mol. Endocrinol., 10, 1605 (1996); https://doi.org/10.1210/mend.10.12.8961270.
  53. P.H. Giangrande, E. A. Kimbrel, D.P. Edwards and D.P. McDonnell, The Opposing Transcriptional Activities of the Two Isoforms of the Human Progesterone Receptor Are Due to Differential Cofactor Binding, Mol. Cell. Biol., 20, 3102 (2000); https://doi.org/10.1128/MCB.20.9.3102-3115.2000.
  54. J.K. Júnior, C.A. Moreira-Kulak and H.S. Taylor, SERMs in the Prevention and Treatment of Postmenopausal Osteoporosis: An Update, Arq. Bras. Endocrinol. Metab., 54, 200 (2010); https://doi.org/10.1590/S0004-27302010000200016.
  55. J.H. Pickar and B.S. Komm, Selective Estrogen Receptor Modulators and the Combination Therapy Conjugated Estrogens/Bazedoxifene: A Review of Effects on the Breast, Post Reprod. Health, 21, 112 (2015); https://doi.org/10.1177/2053369115599090.
  56. M. Abdelrahim, I. Samudio, R. Smith III, R. Burghardt and S. Safe, Small Inhibitory RNA Duplexes for Sp1 mRNA Block Basal and Estrogen-induced Gene Expression and Cell Cycle Progression in MCF-7 Breast Cancer Cells, J. Biol. Chem., 277, 28815 (2002); https://doi.org/10.1074/jbc.M203828200.
  57. L. Gennari, D. Merlotti, G. Martini and R. Nuti, Lasofoxifene: A Third-Generation Selective Estrogen Receptor Modulator for the Prevention and Treatment of Osteoporosis, Expert Opin. Investig. Drugs, 15, 1091 (2006); https://doi.org/10.1517/13543784.15.9.1091.
  58. L. Gennari, D. Merlotti and R. Nuti, Selective Estrogen Receptor Modulator (Serm) for the Treatment of Osteoporosis in Postmenopausal Women: Focus on Lasofoxifene, Clin. Interv. Aging, 5, 19 (2010); https://doi.org/10.2147/CIA.S6083.
  59. S.R. Cummings, K. Ensrud, P.D. Delmas, A.Z. LaCroix, S. Vukicevic, D.M. Reid, S. Goldstein, U. Sriram, A. Lee, J. Thompson, R.A. Armstrong, D.D. Thompson, T. Powles, J. Zanchetta, D. Kendler, P. Neven and R. Eastell, Lasofoxifene in Postmenopausal Women with Osteoporosis, N. Engl. J. Med., 362, 686 (2010); https://doi.org/10.1056/NEJMoa0808692.
  60. L. Gennari, Lasofoxitene: A New Type of Selective Estrogen Receptor Modulator for the Treatment of Osteoporosis, Drugs Today (Barc), 42, 355 (2006); https://doi.org/10.1358/dot.2006.42.6.973583.
  61. L. Gennari, Lasofoxifene, A New Selective Estrogen Receptor Modu-lator for the Treatment of Osteoporosis and Vaginal Atrophy, Expert Opin. Pharmacother., 10, 2209 (2009); https://doi.org/10.1517/14656560903127241.
  62. M. Unkila, S. Kari, E. Yatkin and R. Lammintausta, Vaginal Effects of Ospemifene in the Ovariectomized Rat Preclinical Model of Menopause, J. Steroid Biochem. Mol. Biol., 138, 107 (2013); https://doi.org/10.1016/j.jsbmb.2013.04.004.
  63. G.A. Bachmann and J.O. Komi, Ospemifene Effectively Treats Vulvo-vaginal Atrophy in Postmenopausal Women: Results from a Pivotal Phase 3 Study, Menopause, 17, 480 (2010); https://doi.org/10.1097/gme.0b013e3181c1ac01.
  64. D.J. Portman, G.A. Bachmann and J.A. Simon, Ospemifene, A Novel Selective Estrogen Receptor Modulator for Treating Dyspareunia Asso-ciated with Postmenopausal Vulvar and Vaginal Atrophy, Menopause, 20, 623 (2013); https://doi.org/10.1097/gme.0b013e318279ba64.
  65. J.N. Beverage, T.M. Sissung, A.M. Sion, R. Danesi and W.D. Figg, CYP2D6 Polymorphisms and the Impact on Tamoxifen Therapy, J. Pharm. Sci., 96, 2224 (2007); https://doi.org/10.1002/jps.20892.
  66. Y.C. Lim, Z. Desta, D.A. Flockhart and T.C. Skaar, Endoxifen (4-Hydroxy-N-desmethyl-tamoxifen) has Anti-estrogenic Effects in Breast Cancer Cells with Potency Similar to 4-Hydroxy-tamoxifen, Cancer Chemother. Pharmacol., 55, 471 (2005); https://doi.org/10.1007/s00280-004-0926-7.
  67. X. Wu, J.R. Hawse, M. Subramaniam, M.P. Goetz, J.N. Ingle and T.C. Spelsberg, The Tamoxifen Metabolite, Endoxifen, Is a Potent Anti-estrogen that Targets Estrogen Receptor a for Degradation in Breast Cancer Cells, Cancer Res., 69, 1722 (2009); https://doi.org/10.1158/0008-5472.CAN-08-3933.
  68. S.R.D. Johnston, Endocrinology and Hormone Therapy in Breast Cancer: Selective Oestrogen Receptor Modulators and Downregulators for Breast Cancer-Have They Lost Their Way? Breast Cancer Res., 7, Article number: 119 (2005); https://doi.org/10.1186/bcr1023.
  69. R. McCague, G. Leclercq, N. Legros, J. Goodman, G.M. Blackburn, M. Jarman and A.B. Foster, Derivatives of Tamoxifen. Dependence of Antiestrogenicity on the 4-Substituent, J. Med. Chem., 32, 2527 (1989); https://doi.org/10.1021/jm00132a006.
  70. R. McCague, G.A. Potter and M. Jarman, An Efficient, Large-Scale Synthesis of Idoxifene {(E)-1-[4-[2-(N-pyrrolidino)ethoxy]phenyl]-1-(4-iodophenyl)-2-phenyl-1-butene}, Org. Prep. Proced. Int., 26, 343 (1994); https://doi.org/10.1080/00304949409458432.
  71. H. Liu, W.-C. Park, D.J. Bentrem, K.P. McKian, A.D.L. Reyes, J.A. Loweth, J.M.G. Schafer, J.W. Zapf and V.C. Jordan, Structure-Function Relationships of the Raloxifene-Estrogen Receptor-a Complex for Regulating Transforming Growth Factor-a Expression in Breast Cancer Cells, J. Biol. Chem., 277, 9189 (2002); https://doi.org/10.1074/jbc.M108335200.
  72. L.A. Fitzpatrick, Selective Estrogen Receptor Modulators and Phyto-estrogens: New Therapies for the Postmenopausal Woman, Mayo Clin. Proc., 74, 601 (1999).
  73. A. Buzdar, D. Hayes, A. El-Khoudary, S. Yan, P. Lønning, M. Lichinitser, R. Gopal, G. Falkson, K. Pritchard, A. Lipton, K. Wolter, A. Lee, K. Fly, R. Chew, M. Alderdice, K. Burke and P. Eisenberghase III, Randomized Trial of Droloxifene and Tamoxifen As First-Line Endocrine Treatment of ER/PgR-Positive Advanced Breast Cancer, Breast Cancer Res. Treat., 73, 161 (2002); https://doi.org/10.1023/A:1015229630260.
  74. O. Eckhard and W. Hilmar, Nuclear Receptors as Drug Targets: A Historical Perspective of Modern Drug Discovery, WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim, p. 153 (2008).
  75. E. Chu and V.T. DeVita Jr., Physicians¢ Cancer Chemotherapy Drug Manual, Jones & Bartlett Learning, p. 217 (2003).
  76. V.C. Jordan and B.J.A. Furr, Hormone Therapy in Breast and Prostate Cancer, Springer Science & Business Media, pp. 95-96 (2010).
  77. H.J. Coelingh Bennink, C. Verhoeven, A.E. Dutman and J. Thijssen, The Use of High-dose Estrogens for the Treatment of Breast Cancer, Maturitas, 95, 11 (2017); https://doi.org/10.1016/j.maturitas.2016.10.010.
  78. I. Tadicvana, L. Tasic, N. Vujasinovic-Stupar, K. Ilic and D. Stevanovic, Acta Facult. Med. Naissensis, 31, 29 (2014); https://doi.org/10.2478/afmnai-2014-0003.
  79. M. Prudhomme, Advances in Anticancer Agents in Medicinal Chemistry, Bentham Science Publishers, pp. 368-369 (2013).
  80. P.J. Shughrue, M.V. Lane and I. Merchenthaler, Comparative Distri-bution of Estrogen Eeceptor-a and -b mRNA in the Rat Central Nervous System, J. Comp. Neurol., 388, 507 (1997); https://doi.org/10.1002/(SICI)1096-9861(19971201)388:4<507::AID-CNE1>3.0.CO;2-6.
  81. G.G.J.M. Kuiper, P.J. Shughrue, I. Merchenthaler and J.-A. Gustafsson, The Estrogen Receptor Subtype: A Novel Mediator of Estrogen Action in Neuroendocrine Systems, Front. Neuroendocrinol., 19, 253 (1998); https://doi.org/10.1006/frne.1998.0170.
  82. Z. Desta, B.A. Ward, N.V. Soukhova and D.A. Flockhart, Comprehen-sive Evaluation of Tamoxifen Sequential Biotransformation by the Human Cytochrome P450 System in vitro: Prominent Roles for CYP3A and CYP2D6, J. Pharmacol. Exp. Ther., 310, 1062 (2004); https://doi.org/10.1124/jpet.104.065607.
  83. I. Jatoi and A. Rody, Management of Breast Diseases, Springer, p. 77 (2016).
  84. R. Mansel, A. Goyal, E.L. Nestour, V. Masini-Etévé and K. O’Connell, A Phase II Trial of Afimoxifene (4-Hydroxytamoxifen Gel) for Cyclical Mastalgia in Premenopausal Women, Breast Cancer Res. Treat., 106, 389 (2007); https://doi.org/10.1007/s10549-007-9507-x.
  85. J. MacCallum, J. Cummings, J.M. Dixon and W.R. Miller, Concentra-tions of Tamoxifen and Its Major Metabolites in Hormone Responsive and Resistant Breast Tumours, Br. J. Cancer, 82, 1629 (2000); https://doi.org/10.1054/bjoc.2000.1120.
  86. C.R. Overk, K.W. Peng, R.T. Asghodom, I. Kastrati, D.D. Lantvit, Z. Qin, J. Frasor, J.L. Bolton and G.R.J. Thatcher, Structure-Activity Relation-ships for a Family of Benzothiophene Selective Estrogen Receptor Modulators Including Raloxifene and Arzoxifene, ChemMedChem, 2, 1520 (2007); https://doi.org/10.1002/cmdc.200700104.
  87. S. Martinkovich, D. Shah, S.L. Planey and J.A. Arnott, Selective Estrogen Receptor Modulators: Tissue Specificity and Clinical Utility, Clin. Interv. Aging, 9, 1437 (2014); https://doi.org/10.2147/CIA.S66690.
  88. M. Bolognese, J.H. Krege, W.H. Utian, R. Feldman, S. Broy, D.L. Meats, J. Alam, M. Lakshmanan and M. Omizo, Effects of Arzoxifene on Bone Mineral Density and Endometrium in Postmenopausal Women with Normal or Low Bone Mass, J. Clin. Endocrinol. Metab., 94, 2284 (2009); https://doi.org/10.1210/jc.2008-2143.
  89. S. Mocellin, P. Pilati, M. Briarava and D. Nitti, Breast Cancer Chemo-prevention: A Network Meta-Analysis of Randomized Controlled Trials, J. Natl. Cancer Inst., 108, djv318 (2016); https://doi.org/10.1093/jnci/djv318.
  90. A. Ahmad, S.M. Ali, M.U. Ahmad, S. Sheikh and I. Ahmad, Orally Administered Endoxifen is a New Therapeutic Agent for Breast Cancer, Breast Cancer Res. Treat., 122, 579 (2010); https://doi.org/10.1007/s10549-009-0704-7.
  91. A. Ahmad, S. Sheikh, T. Shah, M.S. Reddy, B.S.V. Prasad, K.K. Verma, B.B. Chandrakant, M. Paithankar, P. Kale, R.V. Solanki, R. Patel, H. Barkate and I. Ahmad, Endoxifen, a New Treatment Option for Mania: A Double-Blind, Active-Controlled Trial Demonstrates the Antimanic Efficacy of Endoxifen, Clin. Transl. Sci., 9, 252 (2016); https://doi.org/10.1111/cts.12407.
  92. P. Ravn, T.F. Nielsen and C. Christiansen, What can be Learned from the Levormeloxifene Experience? Acta Obstet. Gynecol. Scand., 85, 135 (2006); https://doi.org/10.1080/00016340500345691.
  93. N.N. Yang, H.U. Bryant, S. Hardikar, M. Sato, R.J.S. Galvin, A.L. Glasebrook and J.D. Termine, Estrogen and Raloxifene Stimulate Trans-forming Growth Factor-beta 3 Gene Expression in Rat Bone: A Potential Mechanism for Estrogen- or Raloxifene-Mediated Bone Maintenance, Endocrinology, 137, 2075 (1996); https://doi.org/10.1210/endo.137.5.8612550.
  94. N.N. Yang, M. Venugopalan, S. Hardikar and A. Glasebrook, Erratum: Correction: Raloxifene Response Needs More than an Element, Science, 276, 1245 (1997); https://doi.org/10.1126/science.275.5304.1245f.
  95. C. Gruber and D. Gruber, Bazedoxifene (Wyeth), Curr. Opin. Investig. Drugs, 5, 1086 (2004).
  96. T.A. Blizzard, Estrogen Receptor Modulator Medicinal Chemistry at Merck-A Review, Curr. Top. Med. Chem., 8, 792 (2008); https://doi.org/10.2174/156802608784535066.
  97. A. Yenesew, S. Derese, J.O. Midiwo, M. Heydenreich and M.G. Peter, Effect of Rotenoids from the Seeds of Millettia dura on Larvae of Aedes aegypti, Pest Manag. Sci., 59, 1159 (2003); https://doi.org/10.1002/ps.740.
  98. M. Cabizza, A. Angioni, M. Melis, M. Cabras, C.V. Tuberoso and P. Cabras, Rotenone and Rotenoids in Cubè Resins, Formulations, and Residues on Olives, J. Agric. Food Chem., 52, 288 (2004); https://doi.org/10.1021/jf034987a.
  99. G.G. Kuiper, J.G. Lemmen, B. Carlsson, J.C. Corton, S.H. Safe, P.T. van der Saag, B. van der Burg and J.-Å. Gustafsson, Interaction of Estrogenic Chemicals and Phytoestrogens with Estrogen Receptor b, Endocrinology, 139, 4252 (1998); https://doi.org/10.1210/endo.139.10.6216.
  100. M.A. Torres and D.E. Furst, Treatment of Generalized Systemic Sclerosis, Rheum. Dis. Clin. North Am., 16, 217 (1990).
  101. V. Ruiz-velasco, J. Rosas-Arceo and M.M. Matute, Chemical Inducers of Ovulation: Comparative Results, Int. J. Fertil., 24, 61 (1979).
  102. V.J. Stella, R.T. Borchardt, M.J. Hageman, R. Oliyai, H. Maag and J.W. Tilley, Prodrugs: Challenges and Rewards. Parts 1 and 2, Springer Science & Business Media, pp. 168-169 (2007).
  103. M. Oettel and E. Schillinger, Estrogens and Antiestrogens I: Physiology and Mechanisms of Action of Estrogens and Antiestrogens, Springer Science & Business Media, pp. 58-60 (2012).
  104. Y. Yang, Y. Chen, H. Aloysius, D. Inoyama and L. Hu, eds.: H.C. Yang, W.K. Yeh, and J.R. McCarthy, Enzymes and Targeted Activation of Prodrugs, In: Enzyme Technologies: Pluripotent Players in Discovering Therapeutic Agents, Wiley & Sons: New York, pp 165-235 (2014).
  105. G.J. Kelloff, E.T. Hawk and C.C. Sigman, Cancer Chemoprevention: Strategies for Cancer Chemoprevention, Springer, vol. 2, pp. 251 (2008).
  106. J. Elks, The Dictionary of Drugs: Chemical Data, Springer, pp. 298 (2014).
  107. K.M. Rodriguez, A.W. Pastuszak and L.I. Lipshultz, Enclomiphene Citrate for the Treatment of Secondary Male Hypogonadism, Expert Opin. Pharmacother., 17, 1561 (2016); https://doi.org/10.1080/14656566.2016.1204294.
  108. S. Hill, V. Arutchelvam and R. Quinton, Enclomiphene, An Estrogen Receptor Antagonist For The Treatment Of Testosterone Deficiency In Men., IDrugs, 12, 109 (2009).
  109. A. Ahmad, S. Shahabuddin, S. Sheikh, P. Kale, M. Krishnappa, R.C. Rane and I. Ahmad, Endoxifen, a New Cornerstone of Breast Cancer Therapy: Demonstration of Safety, Tolerability, and Systemic Bioavailability in Healthy Human Subjects, Clin. Pharmacol. Ther., 88, 814 (2010); https://doi.org/10.1038/clpt.2010.196.
  110. S.E. Wardell, E.R. Nelson, C.A. Chao, H.M. Alley and D.P. McDonnell, Evaluation of the Pharmacological Activities of RAD1901, A Selective Estrogen Receptor Degrader, Endocr. Rel. Cancer, 22, 713 (2015); https://doi.org/10.1530/ERC-15-0287.
  111. F. Garner, M. Shomali, D. Paquin, C.R. Lyttle and G. Hattersley, RAD1901: A Novel, Orally Bioavailable Selective Estrogen Receptor Degrader that Demonstrates Antitumor Activity in Breast Cancer Xenograft Models, Anticancer Drugs, 26, 948 (2015); https://doi.org/10.1097/CAD.0000000000000271.
  112. G.J. Kelloff, J.A. Crowell, V.E. Steele, R.A. Lubet, C.W. Boone, W.A. Malone, E.T. Hawk, R. Lieberman, J.A. Lawrence, L. Kopelovich, I. Ali, J.L. Viner, and C.C. Sigman, Progress in Cancer Chemoprevention, Ann. N.Y. Acad. Sci., 889, 1 (2008); https://doi.org/10.1111/j.1749-6632.1999.tb08718.x.
  113. C.E. Connor, J.D. Norris, G. Broadwater, T.M. Willson, M.M. Gottardis, M.W. Dewhirst and D.P. McDonnell, Circumventing Tamoxifen Resistance in Breast Cancers using Antiestrogens that Induce Unique Conformational Changes in the Estrogen Receptor, Cancer Res., 61, 2917 (2001).
  114. R.C. Dardes, R.M. Regan, C. Gajdos, S.P. Robinson, D. Bentrem, A. De Los Reyes and V.C. Jordan, Effects of a New Clinically Relevant Antiestrogen (GW5638) Related to Tamoxifen on Breast and Endometrial Cancer Growth in vivo, Clin. Cancer Res., 8, 1995 (2002).
  115. A. Lai, M. Kahraman, S. Govek, J. Nagasawa, C. Bonnefous, J. Julien, K. Douglas, J. Sensintaffar, N. Lu, K. Lee, A. Aparicio, J. Kaufman, J. Qian, G. Shao, R. Prudente, M.J. Moon, J.D. Joseph, B. Darimont, D. Brigham, K. Grillot, R. Heyman, P.J. Rix, J.H. Hager and N.D. Smith, Identification of GDC-0810 (ARN-810), An Orally Bioavailable Selec-tive Estrogen Receptor Degrader (SERD) that Demonstrates Robust Activity in Tamoxifen-Resistant Breast Cancer Xenografts, J. Med. Chem., 58, 4888 (2015); https://doi.org/10.1021/acs.jmedchem.5b00054.
  116. S.P. Govek, J.Y. Nagasawa, K.L. Douglas, A.G. Lai, M. Kahraman, C. Bonnefous, A.M. Aparicio, B.D. Darimont, K.L. Grillot, J.D. Joseph, J.A. Kaufman, K.-J. Lee, N. Lu, M.J. Moon, R.Y. Prudente, P.J. Rix, J. Sensintaffar, J.H. Hager and N.D. Smith, Optimization of an Indazole Series of Selective Estrogen Receptor Degraders: Tumor Regression in a Tamoxifen-Resistant Breast Cancer Xenograft, Bioorg. Med. Chem. Lett., 25, 5163 (2015); https://doi.org/10.1016/j.bmcl.2015.09.074.
  117. M. Oettel and E. Schillinger, Estrogens and Antiestrogens I: Physiology and Mechanisms of Action of Estrogens and Antiestrogens, Springer Science & Business Media, p. 604 (2012).
  118. J. Borvendég, I. Hermann and O. Csuka, Antiestrogens, Antiandrogens, Acta Physiol. Hung., 84, 405 (1996).
  119. K.-C. An, Selective Estrogen Receptor Modulators, Asian Spine J., 10, 787 (2016); https://doi.org/10.4184/asj.2016.10.4.787.
  120. A.J. Ellis, V.M. Hendrick, R. Williams and B.S. Komm, Selective Estrogen Receptor Modulators in Clinical Practice: A Safety Overview, Expert Opin. Drug Saf., 14, 921 (2015); https://doi.org/10.1517/14740338.2015.1014799.
  121. J.V. Pinkerton and S.R. Goldstein, Endometrial Safety: A Key Hurdle for Selective Estrogen Receptor Modulators in Development, Menopause, 17, 642 (2010); https://doi.org/10.1097/gme.0b013e3181c4f1d6.