Skip to main content Accessibility help
×
Hostname: page-component-848d4c4894-v5vhk Total loading time: 0 Render date: 2024-06-20T14:37:15.417Z Has data issue: false hasContentIssue false

Chapter 3 - Ulipristal and Other Medical Interventions for Treatment of Uterine Fibroids

Published online by Cambridge University Press:  10 October 2020

Mostafa Metwally
Affiliation:
University of Sheffield
Tin-Chiu Li
Affiliation:
The Chinese University of Hong Kong
Get access

Summary

Uterine fibroids (UFs) are the most common non-malignant neoplasms affecting women of reproductive age. Some estimates suspect nearly 70–80% of all women will develop at least one fibroid during their lifetime [1]. Women suffering from UFs can present with heavy or prolonged vaginal bleeding, pain or pressure in pelvic region, dysmenorrhoea, dyspareunia, bladder problems, constipation, subfertility, and even loss of pregnancy [2]. Many women with fibroids experience heavy menstrual bleeding (HMB), thereby making them more prone to developing iron deficiency anaemia [3]. Many women suffering from severe symptomatic fibroids choose to have a hysterectomy, making it the second most commonly performed procedure in the United States [4]. Unfortunately the risks involved with surgery, in conjunction with the possibility of eradicating any hope of future pregnancies, make it a less favourable option for women. Thus understanding the pathogenesis behind fibroid formation is paramount for the development of novel therapeutic strategies.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2020

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Baird, DD, Dunson, DB, Hill, MC, Cousins, D, Schectman, JM. High cumulative incidence of uterine leiomyoma in black and white women: ultrasound evidence. Am J Obstet Gynecol 2003;188(1):100–7.Google Scholar
Stewart, EA. Uterine fibroids. Nat Rev Dis Primers 2016;2:16043.Google Scholar
Pritts, EA, Parker, WH, Olive, DL. Fibroids and infertility: an updated systematic review of the evidence. Fertil Steril 2009;91(4):1215–23.Google Scholar
Cardozo, ER, Clark, AD, Banks, NK, et al. The estimated annual cost of uterine leiomyomata in the United States. Am J Obstet Gynecol 2012;206(3):211.e1–9.Google Scholar
Bulun, SE. Uterine fibroids. N Engl J Med 2013;369(14):1344–55.Google Scholar
Borahay, MA, Asoglu, MR, Mas, A, et al. Estrogen receptors and signaling in fibroids: role in pathobiology and therapeutic implications. Reprod Sci 2017;24(9):1235–44.Google Scholar
Ciavattini, A, Di Giuseppe, J, Stortoni, P, et al. Uterine fibroids: pathogenesis and interactions with endometrium and endomyometrial junction. Obstet Gynecol Int 2013;2013:173184.Google Scholar
Lusher, SJ, Raaijmakers, HC, Vu-Pham, D, et al. Structural basis for agonism and antagonism for a set of chemically related progesterone receptor modulators. J Biol Chem 2011;286(40):35079–86.CrossRefGoogle ScholarPubMed
Bouchard, P. Selective progesterone receptor modulators: a class with multiple actions and applications in reproductive endocrinology, and gynecology. Gynecol Endocrinol 2014;30(10):683–4.CrossRefGoogle Scholar
Wagenfeld, A, Saunders, PT, Whitaker, L, Critchley, HO. Selective progesterone receptor modulators (SPRMs): progesterone receptor action, mode of action on the endometrium and treatment options in gynecological therapies. Expert Opin Ther Targets 2016;20(9):1045–54.Google Scholar
Donnez, J, Donnez, O, Dolmans, MM. The current place of medical therapy in uterine fibroid management. Best Pract Res Clin Obstet Gynaecol 2017.Google Scholar
Reis, FM, Bloise, E, Ortiga-Carvalho, TM. Hormones and pathogenesis of uterine fibroids. Best Pract Res Clin Obstet Gynaecol 2016;34:1324.Google Scholar
Ali, M, Al-Hendy, A. Selective progesterone receptor modulators for fertility preservation in women with symptomatic uterine fibroids. Biol Reprod 2017;97(3):337–52.Google Scholar
Donnez, J, Tomaszewski, J, Vázquez, F, et al. Ulipristal acetate versus leuprolide acetate for uterine fibroids. N Engl J Med 2012;366(5):421–32.Google Scholar
Donnez, J, Donnez, O, Courtoy, GE, Dolmans, MM. The place of selective progesterone receptor modulators in myoma therapy. Minerva Ginecol 2016;68(3):313–20.Google ScholarPubMed
Yoshida, S, Ohara, N, Xu, Q, et al. Cell-type specific actions of progesterone receptor modulators in the regulation of uterine leiomyoma growth. Semin Reprod Med 2010;28(3):260–73.Google Scholar
Xu, Q, Ohara, N, Liu, J, et al. Progesterone receptor modulator CDB-2914 induces extracellular matrix metalloproteinase inducer in cultured human uterine leiomyoma cells. Mol Hum Reprod 2008;14(3):181–91.CrossRefGoogle ScholarPubMed
Courtoy, GE, Donnez, J, Marbaix, E, Dolmans, MM. In vivo mechanisms of uterine myoma volume reduction with ulipristal acetate treatment. Fertil Steril 2015;104(2):426–34.e1.Google Scholar
Murji, A, Whitaker, L, Chow, TL, Sobel, ML. Selective progesterone receptor modulators (SPRMs) for uterine fibroids. Cochrane Database Syst Rev 2017;4:CD010770.Google Scholar
Donnez, J, Tatarchuk, TF, Bouchard, P, et al. Ulipristal acetate versus placebo for fibroid treatment before surgery. N Engl J Med 2012;366(5):409–20.Google Scholar
Donnez, J, Donnez, O, Matule, D, et al. Long-term medical management of uterine fibroids with ulipristal acetate. Fertil Steril 2016;105(1):165–73.e4.CrossRefGoogle ScholarPubMed
Fauser, BC, Donnez, J, Bouchard, P, et al. Safety after extended repeated use of ulipristal acetate for uterine fibroids. PLoS One 2017;12(3):e0173523.Google Scholar
Shen, Q, Hua, Y, Jiang, W, et al. Effects of mifepristone on uterine leiomyoma in premenopausal women: a meta-analysis. Fertil Steril 2013;100(6):1722–6.e1–10.CrossRefGoogle ScholarPubMed
Luo, X, Yin, P, Coon, V JS et al. The selective progesterone receptor modulator CDB4124 inhibits proliferation and induces apoptosis in uterine leiomyoma cells. Fertil Steril 2010;93(8):2668–73.Google Scholar
Wilkens, J, Chwalisz, K, Han, C, et al. 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(12):4664–71.CrossRefGoogle ScholarPubMed
Schutt, B, Kaiser, A, Schultze-Mosgau, MH, et al. Pharmacodynamics and safety of the novel selective progesterone receptor modulator vilaprisan: a double-blind, randomized, placebo-controlled phase 1 trial in healthy women. Hum Reprod 2016;31(8):1703–12.Google Scholar
Lethaby, A, Vollenhoven, B, Sowter, M. Efficacy of preoperative gonadotrophin hormone releasing analogues for women with uterine fibroids undergoing hysterectomy or myomectomy: a systematic review. BJOG 2002;109(10):1097–108.Google Scholar
Lee, MJ, Yun, BS, Seong, SJ, et al. Uterine fibroid shrinkage after short-term use of selective progesterone receptor modulator or gonadotropin-releasing hormone agonist. Obstet Gynecol Sci 2017;60(1):6973.Google Scholar
Moroni, R, Vieira, CS, Ferriani, RA, Candido-dos-Reis, FJ, Brito, LGO. Pharmacological treatment of uterine fibroids. Ann Med Health Sci Res 2014;4(Suppl 3):S185–92.CrossRefGoogle ScholarPubMed
Struthers, RS, Nicholls, AJ, Grundy, J, et al. Suppression of gonadotropins and estradiol in premenopausal women by oral administration of the nonpeptide gonadotropin-releasing hormone antagonist elagolix. J Clin Endocrinol Metab 2009;94(2):545–51.CrossRefGoogle ScholarPubMed
Archer, DF, Stewart, EA, Jain, RI, et al. Elagolix for the management of heavy menstrual bleeding associated with uterine fibroids: results from a phase 2a proof-of-concept study. Fertil Steril 2017;108(1):152–60.e4.Google Scholar
MacLean, DB, Shi, H, Faessel, HM, Saad, F. Medical castration using the investigational oral GnRH antagonist TAK-385 (relugolix): phase 1 study in healthy males. J Clin Endocrinol Metab 2015;100(12):4579–87.Google Scholar
Sumitani, H, Shozu, M, Segawa, T, et al. In situ oestrogen synthesized by aromatase P450 in uterine leiomyoma cells promotes cell growth probably via an autocrine/intracrine mechanism. Endocrinology 2000;141(10):3852–61.Google Scholar
Hadji, P, Body, JJ, Aapro, MS, et al. Practical guidance for the management of aromatase inhibitor-associated bone loss. Ann Oncol 2008;19(8):1407–16.Google Scholar
Deng, L, Wu, T, Chen, XY, Xie, L, Yang, J. Selective oestrogen receptor modulators (SERMs) for uterine leiomyomas. Cochrane Database Syst Rev 2012;10:CD005287.Google Scholar

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

Available formats
×