288
Views
7
CrossRef citations to date
0
Altmetric
Reviews

Lessons learned from the preclinical drug discovery of asoprisnil and ulipristal for non-surgical treatment of uterine leiomyomas

(Director) , , , , , , , & show all
Pages 897-911 | Published online: 16 Jul 2011

Bibliography

  • Edozien LC. Hysterectomy for benign conditions. BMJ 2005;330:1457-8
  • Edwards RD, Moss JG, Lumsden MA, Uterine artery embolization versus surgery for symptomatic uterine fibroids. N Engl J Med 2007;356:360-70
  • Farquhar CM, Steiner CA. Hysterectomy rates in the United States 1990-1997. Obstet Gynecol 2002;99:229-34
  • Maruo T, Laoag-Fernandez JB, Matsuo H, Effects of levonorgestrel-releasing intrauterine system on the endometrium and the relevance to the management of menorrhagia caused by uterine myoma and adenomyosis. In: Maruo T, Barlow D, Mardon H, Kennedy S. editors. Cell and molecular biology of endometrium in health and disease. Soeisha; Osaka: 2002. p. 193-207
  • Maruo T, Laoag-Fernandez JB, Pakarinen P, Effects of the levonorgestrel-releasing intrauterine system on proliferation and apoptosis in the endometrium. Hum Reprod 2001;16:2103-8
  • Maruo T. Endometrial effects of levonorgestrel intrauterine delivery. Gynecol Forum 2006;11:27-30
  • Maruo T, Matsuo H, Samoto T, Effects of progesterone on uterine leiomyoma growth and apoptosis. Steroids 2000;65:585-92
  • Maruo T, Ohara N, Matsuo H, Effects of LNG-IUS and progesterone receptor modulator CDB2914 on uterine leiomyomas. Contraception 2007;75:S99-103
  • Jones RL, Critchley HO. Morphological and functional changes in human endometrium following intrauterine levonorgestrel delivery. Hum Reprod 2000;15(Suppl 3):162-72
  • Lukes AS, Reardon B, Arepally G. Use of levonorgestrel-releasing intrauterine system in women with hemostatic disorders. Fertil Steril 2008;90:673-7
  • Chi C, Huq FY, Kadir RA. Levonorgestrel-releasing intrauterine system for the management of heavy menstrual bleeding in women with inherited bleeding disorders: long-term follow up. Contraception 2011;83:242-7
  • Maruo T, Laog-Fernandez JB, Matsuo H, Effects of LNG-IUS on the endometrium and the relevance to the management of menorrhagia. In: Maruo T, Barlow D, Mardon H, Kennedy S. editors. Cell and molecular biology of endometrium. Soeisha; Osaka; 2002. p. 193-207
  • Maruo T. Translational research in women's health: from bedside to bench and from bench to bedside. Int J Gynecol Obstet 2010;109:83-4
  • Rein MS, Barbieri RL, Friedman AJ. Progesterone: a critical role in the pathogenesis of uterine myomas. Am J Obstet Gynecol 1995;172:14-18
  • Matsuo H, Maruo T, Samoto T. Increased expression of Bcl-2 protein in human uterine leiomyoma and its up-regulation by progesterone. J Clin Endocrinol Metab 1997;82:293-9
  • Shimomura Y, Matsuo H, Samoto T, Maruo T. Up-regulation by progesterone of proliferating cell nuclear antigen and epidermal growth factor expression in human uterine leiomyoma. J Clin Endocrinol Metab 1998;83:2192-8
  • Kurachi O, Matsuo H, Samoto T, Maruo T. Tumor necrosis factor- expression in human uterine leiomyoma and its down-regulation by progesterone. J Clin Endocrinol Metab 2001;86:2275-80
  • Maruo T, Ohara N, Wang J, Matsuo H. Sex steroidal regulation of uterine leiomyoma growth and apoptosis. Hum Reprod Update 2004;10:207-20
  • Harrison-Woolrych ML, Charnock-Jones DS, Smith S. Quantification of messenger ribonucleic acid for epidermal growth factor in human myometrium and leiomyomata using reverse transcriptase polymerase chain reaction. J Clin Endocrinol Metab 1994;78:1179-84
  • Maruo T, Samoto T, Matsuo H, Regulation of proliferative potential and Bcl-2 oncoprotein expression in human uterine leiomyoma by sex steroid hormones. In: Kuramoto H, Gurpide E. editors. In vitro biology of sex steroid hormone action. Churchill Livingstone; 1996. p. 251-63
  • Yu L, Saile K, Swartz CD, Differential expression of receptor tyrosine kinases (RTKs) and IGF-I pathway activation in human uterine leiomyomas. Mol Med 2008;14:264-75
  • Gao Z, Matsuo H, Wang Y, Up-regulation by IGF-I of proliferating cell nuclear antigen and Bcl-2 protein expression in human uterine leiomyoma cells. J Clin Endocrinol Metab 2001;86:5593-9
  • Yamada T, Nakago S, Kurachi O, Progesterone down-regulates insulin-like growth factor-I expression in cultured human uterine leiomyoma cells. Hum Reprod 2004;19:1-7
  • Danforth DN Jr, Sgagias MK. Tumour necrosis factor-alpha modulates oestradiol responsiveness of MCF-7 breast cancer cells in vitro. J Endocrinol 1993;138:517-28
  • Terranova PF, Hunter VJ, Roby KF, Hunt JS. Tumor necrosis factor-alpha in the female reproductive tract. Proc Soc Exp Biol Med 1995;209:325-42
  • Haviv R, Stein R. Nerve growth factor inhibits apoptosis induced by tumor necrosis factor in PC12 cells. J Neurosci Res 1999;55:269-77
  • Leonhardt SA, Edwards DP. Mechanism of action of progesterone antagonists. Exp Biol Med 2002;227:969-80
  • Kastner P, Krust A, Turcotte B, Two distinct estrogen-regulated promoters generate transcripts encoding the two functionally different human progesterone receptor forms A and B. EMBO J 1990;9:1603-14
  • Vegeto E, Shahbaz MM, Wen DX, Human progesterone receptor A form is a cell- and promoter-specific repressor of human progesterone receptor B function. Mol Endocrinol 1993;7:1244-55
  • Wen DX, Xu Y-F, Mais DE, The A and B isoforms of the human progesterone receptor operate through distinct signaling pathways within target cells. Mol Cell Biol 1994;14:8356-64
  • Brandon DD, Bethea CL, Strawn EY, Progesterone receptor messenger ribonucleic acid and protein are overexpressed in human uterine leiomyomas. Am J Obstet Gynecol 1993;169:78-85
  • Viville B, Charnock-Jones DS, Sharkey AM, Distribution of the A and B forms of the progesterone receptor messenger ribonucleic acid and protein in uterine leiomyomata and adjacent myometrium. Hum Reprod 1997;12:815-22
  • Nisolle M, Gillerot S, Casanas-Roux F, Immunohistochemical study of the proliferation index, oestrogen receptors and progesterone receptors A and B in leiomyomata and normal myometrium during the menstrual cycle and under gonadotropin-releasing hormone agonist therapy. Hum Reprod 1999;14:2844-50
  • Tiltman AJ. The effect of progestins on the mitotic activity of uterine fibromyomas. Int J Gynecol Pathol 1985;4:89-96
  • Carr BR, Marshburn PB, Weatherall PT, An evaluation of the effect of gonadotropin-releasing hormone analogs and medroxyprogesterone acetate on uterine leiomyomata volume by magnetic resonance imaging: a prospective, randomized, double blind, placebo-controlled, crossover trial. J Clin Endocrinol Metab 1993;76:1217-23
  • Friedman AJ, Daly M, Juneau-Norcross M, A prospective, randomized trial of gonadotropin-releasing hormone agonist plus estrogen-progestin or progestin “add-back” regimens for women with leiomyomata uteri. J Clin Endocrinol Metab 1993;76:1439-45
  • Kettel LM, Murphy AA, Morales AJ, Yen SSC. Clinical efficacy of the antiprogesterone RU486 in the treatment of endometriosis and uterine fibroids. Hum Reprod 1994;9(Suppl 1):116-20
  • Murphy AA, Morales AJ, Kettel LM, Yen SSC. Regression of uterine leiomyomata to the antiprogesterone RU486: dose-response effect. Fertil Steril 1995;64:187-90
  • Eisinger SH, Meldrum S, Fiscella K, Low-dose mifepristone for uterine leiomyomata. Obstet Gynecol 2003;101:243-50
  • Chabbert-Buffet N, Meduri G, Bouchard P, Spitz IM. Selective progesterone receptor modulators and progesterone antagonists: mechanisms of action and clinical applications. Hum Reprod Update 2005;11:293-307
  • Chwalisz K, Perez MC, DeManno D, Selective progesterone receptor modulator development and use in the treatment of leiomyomata and endometriosis. Endocr Rev 2005;26:423-38
  • Chwalisz K, Larsen L, Mattia-Goldberg C, A randomized, controlled trial of asoprisnil, a novel selective progesterone receptor modulator, in women with uterine leiomyomata. Fertil Steril 2007;87:1399-412
  • Attardi BJ, Burgenson J, Hild SA, CDB-4124 and its putative monodemethylated metabolite, CDB-4453, are potent antiprogestins with reduced antiglucocorticoid activity: in vitro comparison to mifepristone and CDB-2914. Mol Cell Endocrinol 2002;188:111-23
  • Blithe DL, Nieman LK, Blye RP, Development of the selective progesterone receptor modulator CDB-2914 for clinical indications. Steroids 2003;68:1013-17
  • Gainer EE, Ulmann A. Pharmacologic properties of CDB(VA)-2914. Steroids 2003;68:1005-11
  • Hild SA, Reel JR, Hoffman LH, Blye RP. CDB-2914: anti-progestational/anti-glucocorticoid profile and post-coital anti-fertility activity in rats and rabbits. Hum Reprod 2000;15:822-9
  • Xu Q, Takekida S, Ohara N, Progesterone receptor modulator CDB-2914 down-regulates proliferative cell nuclear antigen and Bcl-2 protein expression and up-regulates caspase-3 and poly(adenosine 5'-diphosphate-ribose) polymerase expression in cultured human uterine leiomyoma cells. J Clin Endocrinol Metab 2005;90:953-61
  • Chen W, Ohara N, Wang J, A novel selective progesterone receptor modulator asoprisnil (J867) inhibits proliferation and induces apoptosis in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2006;91:1296-304
  • Robbins BA, De la Vega D, Ogata K, Immunohistochemical detection of proliferating cell nuclear antigen in solid human malignancies. Arch Pathol Lab Med 1987;11:841-5
  • Wang J, Ohara N, Wang Z, A novel selective progesterone receptor modulator asoprisnil (J867) down-regulates the expression of EGF, IGF-I, TGFbeta3 and their receptors in cultured uterine leiomyoma cells. Hum Reprod 2006;21:1869-77
  • Singh AB, Harris RC. Autocrine, paracrine and juxtacrine signaling by EGFR ligands. Cell Signal 2005;17:1183-93
  • Lutz M, Knaus P. Integration of the TGF-beta pathway into the cellular signaling network. Cell Signal 2002;14:977-88
  • Wang S, El-Deiry WS. TRAIL and apoptosis induction by TNF-family death receptors. Oncogene 2003;22:8628-33
  • Riedl SJ, Shi Y. Molecular mechanisms of caspase regulation during apoptosis. Nat Rev Mol Cell Biol 2004;5:897-907
  • Shiozaki EN, Shi Y. Caspases, IAPs and Smac/DIABLO: mechanisms from structural biology. Trends Biochem Sci 2004;39:486-94
  • Shi Y. Caspase activation, inhibition, and reactivation: a mechanistic view. Protein Sci 2004;13:1979-87
  • Cohen GM. Caspases: the executioners of apoptosis. Biochem J 1997;326:1-16
  • Bouchard VJ, Rouleau M, Poirier GG. PARP-1, a determinant of cell survival in response to DNA damage. Exp Hematol 2003;31:446-54
  • Pieper AA, Verma A, Zhang J, Snyder SH. Poly(ADP-ribose) polymerase, nitric oxide and cell death. Trends Pharmacol Sci 1999;20:171-81
  • Reed JC, Jurgensmeier JM, Matsuyama S. Bcl-2 family proteins and mitochondria. Biochim Biophys Acta 1998;1366:127-37
  • Yin P, Lin Z, Cheng Y-H, Progesterone receptor regulates bcl-2 gene expression through direct binding to its promoter region in uterine leiomyoma cells. J Clin Endocrinol Metab 2007;92:4459-66
  • Yagita H, Takeda K, Hayakawa Y, TRAIL and its receptors as targets for cancer therapy. Cancer Sci 2004;95:777-83
  • Hoffman PJ, Milliken DB, Gregg LC, Molecular characterization of uterine fibroids and its implication for underlying mechanisms of pathogenesis. Fertil Steril 2004;83:639-49
  • Sasaki H, Ohara N, Xu Q, A novel selective progesterone receptor modulator asoprisnil activates tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated signaling pathway in cultured human uterine leiomyoma cells in the absence of comparable effects on myometrial cells. J Clin Endocrinol Metab 2007;92:616-23
  • Walocha JA, Litwin JA, Miodonski AJ. Vascular system of intramural leiomyomata revealed by corrosion casting and scanning electron microscopy. Hum Reprod 2003;18:1088-93
  • Hague S, Zhang L, Oehler MK, Expression of the hypoxically regulated angiogenic factor adrenomedullin correlates with uterine leiomyoma vascular density. Clin Cancer Res 2000;6:2808-14
  • Gentry CC, Okolo SO, Te Fong LFW, Quantification of vascular endothelial growth factor-A in leiomyomas and adjacent myometrium. Clin Sci 2001;101:691-5
  • Cross MJ, Dixelius J, Matsumoto T, Claesson-Welsh L. VEGF-receptor signal transduction. Trends Biochem Sci 2003;28:488-94
  • Ferrara N, Gerber H-P, LeCouter J. The biology of VEGF and its receptors. Nat Med 2003;9:669-76
  • Brown LF, Detmar M, Tognazzi K, Uterine smooth muscle cells express functional receptors (flt-1 and KDR) for vascular permeability factor/vascular endothelial growth factor. Lab Invest 1997;76:245-55
  • Ribatti D, Guidolin D, Conconi MT, Vinblastine inhibits the angiogenic response induced by adrenomedullin in vitro and in vivo. Oncogene 2003;22:6458-61
  • Kato H, Shichiri M, Marumo F, Hirata Y. Adrenomedullin as an autocrine/paracrine apoptosis survival factor for rat endothelial cells. Endocrinology 1997;138:2615-20
  • Greb RR, Heikinheimo O, Williams RF, Vascular endothelial growth factor in primate endometrium is regulated by oestrogen-receptor and progesterone-receptor ligands in vivo. Hum Reprod 1997;12:1280-92
  • Ancelin M, Buteau-Lozano H, Meduri G, A dynamic shift of VEGF isoforms with a transient and selective progesterone-induced expression of VEGF189 regulates angiogenesis and vascular permeability in human uterus. Proc Natl Acad Sci USA 2002;99:6023-8
  • Thota C, Yallampalli C. Progesterone upregulates calcitonin gene-related peptide and adrenomedullin receptor components and cyclic adenosine 3'5'-monophosphate generation in Eker rat uterine smooth muscle cell line. Biol Reprod 2005;72:416-22
  • Xu Q, Ohara N, Chen W, Progesterone receptor modulator CDB-2914 down-regulates vascular endothelial growth factor, adrenomedullin and their receptors and modulates progesterone receptor content in cultured human uterine leiomyoma cells. Hum Reprod 2006;21:2408-16
  • Gelse K, Poschl E, Aigner TCollagens – structure, function, and biosynthesis. Adv Drug Deliv Rev 2003;55:1531-46
  • Canty EG, Kadler KE. Procollagen trafficking, processing and fibrillogenesis. J Cell Sci 2005;118:1341-53
  • Stewart EA, Friedman AJ, Peck K, Nowak RA. Relative overexpression of collagen type I and collagen type III messenger ribonucleic acids by uterine leiomyomas during the proliferative phase of the menstrual cycle. J Clin Endocrinol Metab 1994;79:900-6
  • Wolanska M, Sobolewski K, Drozdzewicz M, Banknowski E. Extracellular matrix components in uterine leiomyoma and their alteration during the tumour growth. Mol Cell Biochem 1998;189:145-52
  • Berto AGA, Sampaio LO, Franco CRC, A comparative analysis of structure and special distribution of decorin in human leiomyoma and normal myometrium. Biochim Biophys Acta 2003;1619:98-112
  • Wang H, Mahadevappa M, Yamamoto K, Distinctive proliferative phase differences in gene expression in human myometrium and leiomyomata. Fertil Steril 2003;80:266-76
  • Visse R, Nagase H. Matrix metalloproteinases and tissue inhibitors of metalloproteinases. structure, function, and biochemistry. Circ Res 2003;92:827-39
  • Schmidt R, Bultmann A, Ungerer M, Extracellular matrix metalloproteinase inducer regulates matrix metalloproteinase activity in cardiovascular cells: implications in acute myocardial infarction. Circulation 2006;113:834-41
  • Morikawa A, Ohara N, Xu Q, Selective progesterone receptor modulator asoprisnil down-regulates collagen synthesis in cultured human uterine leiomyoma cells through up-regulating extracellular matrix metalloproteinase inhibitor. Hum Reprod 2008;23:944-51
  • Xu Q, Ohara N, Liu J, Progesterone receptor modulator CDB-2914 induces extracellular matrix metalloproteinase inhibitor cultured human uterine leiomyoma cells. Mol Hum Reprod 2008;14:181-91
  • Liang M, Wang H, Zhang Y, Expression and functional analysis of platelet-derived growth factor in uterine leiomyomata. Cancer Biol Ther 2006;5:28-33
  • Catherino WH, Malik M, Driggers P, Novel, Orally active selective progesterone receptor modulator CP8947 inhibits leiomyoma cell proliferation without adversely affecting endometrium or myometrium. J Steroid Biochem Mol Biol 2010;122:279-86
  • Malk M, Norian J, McCarthy-Keith D, Why leiomyomas are called fibroids: the central role of extracellular matrix in symptomatic women. Semin Reprod Med 2010;28:169-79
  • Madauss KP, Grygielko ET, Deng S-J, A structural and in vitro characterization of asoprisnil: a selective progesterone receptor modulator. Mol Endocrinol 2007;21:1066-81
  • Aghajanova L, Velarde MC, Giudice LC. The progesterone receptor coactivator Hic-5 is involved in the pathophysiology of endometriosis. Endocrinology 2009;150:3863-70
  • Wu J, Richer J, Horwitz KB, Hyder SM. Progesterone-dependent induction of vascular endothelial growth factor in human breast cancer cells : preferential regulation by progesterone receptor B. Cancer Res 2004;64:2238-44
  • Chwalisz K, Larsen L, Mattia-Goldberg C, A randomized, controlled trial of asoprisnil, a novel selective progesterone receptor modulator, in women with uterine leiomyomata. Fertil Steril 2007;87:1399-412
  • Wilkens J, Chwalisz K, Han C, Effects of the selective progesterone receptor modulator Asoprisnil on uterine artery blood flow, ovarian activity, and clinical symptoms in patients with uterine leiomyomata scheduled for hysterectomy. J Clin Endocrinol Metab 2008;93:4664-71
  • Levens ED, Potlog-Nahari C, Armstrong AY, CDB-2914 for uterine leiomyomata treatment : a randomized controlled trial. Obstet Gynecol 2008;111:1129-36
  • Nieman LK, Blocker W, Nansel T, Efficacy and tolerability of CDB-2914 treatment for symptomatic uterine fibroids: a randomized, double-blind, placebo-controlled, phase IIb study. Fertil Steril 2011;95:767-72
  • Horne FM, Blithe DL. Progesterone receptor modulators and the endometrium: changes and consequences. Hum Reprod Update 2007;13:567-80
  • Ravet S, Munaut C, Blacher S, Persistence of an intact endometrial matrix and vessels structure in women exposed to VA-2914, a selective progesterone receptor modulator. J Clin Endocrinol Metab 2008;93:4525-31
  • Wilkens J, Williams ARW, Chwalisz K, Effect of asoprisnil on uterine proliferation markers and endometrial expression of the tumour suppressor gene, PTEN. Hum Reprod 2009;1:1-9
  • Mutter GL, Bergeron C, Deligdisch L, The spectrum of endometrial pathology induced by progesterone receptor modulators. Mod Pathol 2008;21:591-8
  • Spitz IM. Clinical utility of progesterone receptor modulators and their effect on the endometrium. Curr Opin Obstet Gynecol 2009;21:318-24

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.