open access

Vol 91, No 9 (2020)
Review paper
Published online: 2020-09-30
Get Citation

Supplementation of dehydroepiandrosterone (DHEA) in pre- and postmenopausal women — position statement of expert panel of Polish Menopause and Andropause Society

Michal Rabijewski1, Lucyna Papierska2, Malgorzata Binkowska3, Radoslaw Maksym1, Katarzyna Jankowska2, Wioletta Skrzypulec-Plinta4, Wojciech Zgliczynski2
·
Pubmed: 33030737
·
Ginekol Pol 2020;91(9):554-562.
Affiliations
  1. Department of Reproductive Health, Center of Postgraduate Medical Education, Warsaw, Poland
  2. Department of Endocrinology of the Centre of Postgraduate Medical Education, Warsaw, Poland, Warsaw, Poland
  3. Department of Gynecological Oncology and Obstetrics, Centre of Postgraduate Medical Education, Warsaw, Poland
  4. Department of Gynecological Disease Prevention and Sexology, Chair of Women’s Health, School of Health Sciences, Medical University of Silesia, Katowice, Poland

open access

Vol 91, No 9 (2020)
REVIEW PAPERS Gynecology
Published online: 2020-09-30

Abstract

Dehydroepiandrosterone (DHEA) concentration decreases with age, therefore, DHEA has been considered a hormone
that reduces the symptoms associated with aging, so the usefulness of DHEA in premenopausal and postmenopausal
women, and the options of hormone therapy have received a large amount of attention. The effectiveness of DHEA in the
premenopausal women remains unclear, while in postmenopausal women with coexisting estrogens deficiency is controversial.
Despite many years of study, the use of DHEA is still controversial, especially regarding its effectiveness. The aim
of present article was to evaluate DHEA specific effects on metabolic parameters, bone mineral density, insulin resistance
as well as the therapeutic potential of DHEA in pre- and postmenopausal women using measures of sexual activity, cognition
and well-being. The summary of this article is the position statement of expert group of the Polish Menopause and
Andropause Society regarding the efficacy and safety of DHEA supplementation in women. We concluded, that currently
available clinical trials and meta-analyses indicate that DHEA supplementation is effective in women with adrenal insufficiency
and chronically treated with exogenous glucocorticoids, postmenopausal women with low bone mineral density
and/or osteoporosis, premenopausal women with sexual disorders and low libido, and in women with vulvovaginal atrophy
due to menopause or genitourinary syndrome of menopause. Currently available clinical trials also suggest that DHEA
supplementation is probably effective in postmenopausal women with hypoactive sexual disorders, infertile women with
diminished ovarian reserve, women suffering from depression and anxiety, and women with obesity and insulin resistance.
No serious adverse effects have been reported.

Abstract

Dehydroepiandrosterone (DHEA) concentration decreases with age, therefore, DHEA has been considered a hormone
that reduces the symptoms associated with aging, so the usefulness of DHEA in premenopausal and postmenopausal
women, and the options of hormone therapy have received a large amount of attention. The effectiveness of DHEA in the
premenopausal women remains unclear, while in postmenopausal women with coexisting estrogens deficiency is controversial.
Despite many years of study, the use of DHEA is still controversial, especially regarding its effectiveness. The aim
of present article was to evaluate DHEA specific effects on metabolic parameters, bone mineral density, insulin resistance
as well as the therapeutic potential of DHEA in pre- and postmenopausal women using measures of sexual activity, cognition
and well-being. The summary of this article is the position statement of expert group of the Polish Menopause and
Andropause Society regarding the efficacy and safety of DHEA supplementation in women. We concluded, that currently
available clinical trials and meta-analyses indicate that DHEA supplementation is effective in women with adrenal insufficiency
and chronically treated with exogenous glucocorticoids, postmenopausal women with low bone mineral density
and/or osteoporosis, premenopausal women with sexual disorders and low libido, and in women with vulvovaginal atrophy
due to menopause or genitourinary syndrome of menopause. Currently available clinical trials also suggest that DHEA
supplementation is probably effective in postmenopausal women with hypoactive sexual disorders, infertile women with
diminished ovarian reserve, women suffering from depression and anxiety, and women with obesity and insulin resistance.
No serious adverse effects have been reported.

Get Citation

Keywords

dehydroepiandrosterone; perimenopause; menopause

About this article
Title

Supplementation of dehydroepiandrosterone (DHEA) in pre- and postmenopausal women — position statement of expert panel of Polish Menopause and Andropause Society

Journal

Ginekologia Polska

Issue

Vol 91, No 9 (2020)

Article type

Review paper

Pages

554-562

Published online

2020-09-30

Page views

6221

Article views/downloads

5176

DOI

10.5603/GP.2020.0091

Pubmed

33030737

Bibliographic record

Ginekol Pol 2020;91(9):554-562.

Keywords

dehydroepiandrosterone
perimenopause
menopause

Authors

Michal Rabijewski
Lucyna Papierska
Malgorzata Binkowska
Radoslaw Maksym
Katarzyna Jankowska
Wioletta Skrzypulec-Plinta
Wojciech Zgliczynski

References (64)
  1. Klinge CM, Clark BJ, Prough RA. Dehydroepiandrosterone Research: Past, Current, and Future. Vitam Horm. 2018; 108: 1–28.
  2. Rutkowski K, Sowa P, Rutkowska-Talipska J, et al. Dehydroepiandrosterone (DHEA): hypes and hopes. Drugs. 2014; 74(11): 1195–1207.
  3. Davis SR, Panjari M, Stanczyk FZ. Replacement for Postmenopausal Women. J Clin Endocrinol Metab. 2011; 96(6): 1642–1653.
  4. Vendola KA, Zhou J, Adesanya OO, et al. Androgens stimulate early stages of follicular growth in the primate ovary. J Clin Invest. 1998; 101(12): 2622–2629.
  5. Sen A, Hammes SR. Granulosa cell-specific androgen receptors are critical regulators of ovarian development and function. Mol Endocrinol. 2010; 24(7): 1393–1403.
  6. Webb SJ, Geoghegan TE, Prough RA, et al. The biological actions of dehydroepiandrosterone involves multiple receptors. Drug Metab Rev. 2006; 38(1-2): 89–116.
  7. Perzyło K, Kulik-Rechberger B, Gałczyński K, et al. [Intracrinology and dehydroepiandrosterone--a new perspective for the use of androgens in hormone replacement therapy in postmenopausal women]. Ginekol Pol. 2011; 82(9): 690–695.
  8. Longcope C. 1 Adrenal and gonadal androgen secretion in normal females. Clin Endocrinol Metab. 1986; 15(2): 213–228.
  9. Chen F, Knecht K, Birzin E, et al. Direct agonist/antagonist functions of dehydroepiandrosterone. Endocrinology. 2005; 146(11): 4568–4576.
  10. Vermeulen A. Androgen replacement therapy in the aging male--a critical evaluation. J Clin Endocrinol Metab. 2001; 86(6): 2380–2390.
  11. Arlt W. Dehydroepiandrosterone replacement therapy. Semin Reprod Med. 2004; 22(4): 379–388.
  12. Jankowski CM, Gozansky WS, Schwartz RS, et al. Effects of dehydroepiandrosterone replacement therapy on bone mineral density in older adults: a randomized, controlled trial. J Clin Endocrinol Metab. 2006; 91(8): 2986–2993.
  13. Lin H, Li L, Wang Qi, et al. A systematic review and meta-analysis of randomized placebo-controlled trials of DHEA supplementation of bone mineral density in healthy adults. Gynecol Endocrinol. 2019; 35(11): 924–931.
  14. Labrie F, Luu-The V, Labrie C, et al. DHEA and its transformation into androgens and estrogens in peripheral target tissues: intracrinology. Front Neuroendocrinol. 2001; 22(3): 185–212.
  15. Pöllänen E, Kangas R, Horttanainen M, et al. Intramuscular sex steroid hormones are associated with skeletal muscle strength and power in women with different hormonal status. Aging Cell. 2015; 14(2): 236–248.
  16. Kong SH, Kim JH, Lee JiH, et al. Dehydroepiandrosterone Sulfate and Free Testosterone but not Estradiol are Related to Muscle Strength and Bone Microarchitecture in Older Adults. Calcif Tissue Int. 2019; 105(3): 285–293.
  17. Villareal DT, Holloszy JO. Effect of DHEA on abdominal fat and insulin action in elderly women and men: a randomized controlled trial. JAMA. 2004; 292(18): 2243–2248.
  18. von Mühlen D, Laughlin GA, Kritz-Silverstein D, et al. Endogenous levels of dehydroepiandrosterone sulfate, but not other sex hormones, are associated with depressed mood in older women: the Rancho Bernardo Study. J Am Geriatr Soc. 1999; 47(6): 685–691.
  19. Moriguchi S, Shinoda Y, Yamamoto Y, et al. Stimulation of the sigma-1 receptor by DHEA enhances synaptic efficacy and neurogenesis in the hippocampal dentate gyrus of olfactory bulbectomized mice. PLoS One. 2013; 8(4): e60863.
  20. Yehuda R, Brand SR, Golier JA, et al. Clinical correlates of DHEA associated with post-traumatic stress disorder. Acta Psychiatr Scand. 2006; 114(3): 187–193.
  21. Pluchino N, Drakopoulos P, Bianchi-Demicheli F, et al. Neurobiology of DHEA and effects on sexuality, mood and cognition. J Steroid Biochem Mol Biol. 2015; 145: 273–280.
  22. Wolkowitz OM, Reus VI, Keebler A, et al. Double-blind treatment of major depression with dehydroepiandrosterone (DHEA). Am J Psychiatry. 1999; 156(4): 646–649.
  23. Schmidt PJ, Daly RC, Bloch M, et al. Dehydroepiandrosterone monotherapy in midlife-onset major and minor depression. Arch Gen Psychiatry. 2005; 62(2): 154–162.
  24. Hough CM, Lindqvist D, Epel ES, et al. Higher serum DHEA concentrations before and after SSRI treatment are associated with remission of major depression. Psychoneuroendocrinology. 2017; 77: 122–130.
  25. Davison SL, Bell R, Donath S, et al. Androgen levels in adult females: changes with age, menopause, and oophorectomy. J Clin Endocrinol Metab. 2005; 90(7): 3847–3853.
  26. Davis SR, Davison SL, Donath S, et al. Circulating androgen levels and self-reported sexual function in women. JAMA. 2005; 294(1): 91–96.
  27. Genazzani AR, Pluchino N. DHEA therapy in postmenopausal women: the need to move forward beyond the lack of evidence. Climacteric. 2010; 13(4): 314–316.
  28. Genazzani AR, Stomati M, Valentino V, et al. Effect of 1-year, low-dose DHEA therapy on climacteric symptoms and female sexuality. Climacteric. 2011; 14(6): 661–668.
  29. Kushnir VA, Darmon SK, Barad DH, et al. Effects of dehydroepiandrosterone (DHEA) supplementation on sexual function in premenopausal infertile women. Endocrine. 2019; 63(3): 632–638.
  30. Davis SR, Baber R, Panay N, et al. Global Consensus Position Statement on the use of Testosterone Therapy for Women. Maturitas. 2019; 128: 89–93.
  31. Labrie F, Diamond P, Cusan L, et al. Effect of 12-month dehydroepiandrosterone replacement therapy on bone, vagina, and endometrium in postmenopausal women. J Clin Endocrinol Metab. 1997; 82(10): 3498–3505.
  32. Baulieu EE, Thomas G, Legrain S, et al. Dehydroepiandrosterone (DHEA), DHEA sulfate, and aging: contribution of the DHEAge Study to a sociobiomedical issue. Proc Natl Acad Sci U S A. 2000; 97(8): 4279–4284.
  33. Løvås K, Loge JH, Husebye ES. Subjective health status in Norwegian patients with Addison's disease. Clin Endocrinol (Oxf). 2002; 56(5): 581–588.
  34. Arlt W. Quality of life in Addison's disease--the case for DHEA replacement. Clin Endocrinol (Oxf). 2002; 56(5): 573–574.
  35. Alkatib AA, Cosma M, Elamin MB, et al. A systematic review and meta-analysis of randomized placebo-controlled trials of DHEA treatment effects on quality of life in women with adrenal insufficiency. J Clin Endocrinol Metab. 2009; 94(10): 3676–3681.
  36. Bornstein SR, Allolio B, Arlt W, et al. Diagnosis and Treatment of Primary Adrenal Insufficiency: An Endocrine Society Clinical Practice Guideline. J Clin Endocrinol Metab. 2016; 101(2): 364–389.
  37. van Vollenhoven RF. Dehydroepiandrosterone for the treatment of systemic lupus erythematosus. Expert Opin Pharmacother. 2002; 3(1): 23–31.
  38. Labrie F, Archer D, Koltun W, et al. Efficacy of intravaginal dehydroepiandrosterone (DHEA) on moderate to severe dyspareunia and vaginal dryness, symptoms of vulvovaginal atrophy, and of the genitourinary syndrome of menopause. Menopause. 2018; 25(11): 1339–1353.
  39. Labrie F, Archer DF, Bouchard C, et al. Members of the VVA Prasterone Group. Prasterone has parallel beneficial effects on the main symptoms of vulvovaginal atrophy: 52-week open-label study. Maturitas. 2015; 81(1): 46–56.
  40. Soman M, Huang LC, Cai WH, et al. Serum androgen profiles in women with premature ovarian insufficiency: a systematic review and meta-analysis. Menopause. 2019; 26(1): 78–93.
  41. Premature Ovarian Failure: An Association with Autoimmune Diseases. JOURNAL OF CLINICAL AND DIAGNOSTIC RESEARCH. 2016.
  42. Daan NMP, Jaspers L, Koster MPH, et al. Androgen levels in women with various forms of ovarian dysfunction: associations with cardiometabolic features. Hum Reprod. 2015; 30(10): 2376–2386.
  43. Ott J, Pecnik P, Promberger R, et al. Dehydroepiandrosterone in women with premature ovarian failure and Hashimoto's thyroiditis. Climacteric. 2014; 17(1): 92–96.
  44. Doldi N, Belvisi L, Bassan M, et al. Premature ovarian failure: steroid synthesis and autoimmunity. Gynecol Endocrinol. 1998; 12(1): 23–28.
  45. Webber L, Davies M, Anderson R, et al. European Society for Human Reproduction and Embryology (ESHRE) Guideline Group on POI. ESHRE Guideline: management of women with premature ovarian insufficiency. Hum Reprod. 2016; 31(5): 926–937.
  46. Qin JC, Fan L, Qin AP. The effect of dehydroepiandrosterone (DHEA) supplementation on women with diminished ovarian reserve (DOR) in IVF cycle: Evidence from a meta-analysis. J Gynecol Obstet Hum Reprod. 2017; 46(1): 1–7.
  47. Lin LT, Tsui KH, Wang PH. Clinical application of dehydroepiandrosterone in reproduction: A review of the evidence. J Chin Med Assoc. 2015; 78(8): 446–453.
  48. Tartagni M, De Pe, Damiani GR, et al. V, Potential benefit of dehydroepiandrosterone supplementation for infertile but not poor responder patients in a IVF program. Minerva Ginecol. 2015; 67(1): 7–12.
  49. Gleicher N, Ryan E, Weghofer A, et al. Miscarriage rates after dehydroepiandrosterone (DHEA) supplementation in women with diminished ovarian reserve: a case control study. Reprod Biol Endocrinol. 2009; 7: 108.
  50. Lin LT, Wang PH, Wen ZH, et al. The Application of Dehydroepiandrosterone on Improving Mitochondrial Function and Reducing Apoptosis of Cumulus Cells in Poor Ovarian Responders. Int J Med Sci. 2017; 14(6): 585–594.
  51. Zhang HH, Xu PY, Wu J, et al. Dehydroepiandrosterone improves follicular fluid bone morphogenetic protein-15 and accumulated embryo score of infertility patients with diminished ovarian reserve undergoing in vitro fertilization: a randomized controlled trial. J Ovarian Res. 2014; 7: 93.
  52. Lin LT, Cheng JT, Wang PH, et al. Dehydroepiandrosterone as a potential agent to slow down ovarian aging. J Obstet Gynaecol Res. 2017; 43(12): 1855–1862.
  53. Hu Q, Hong L, Nie M, et al. The effect of dehydroepiandrosterone supplementation on ovarian response is associated with androgen receptor in diminished ovarian reserve women. J Ovarian Res. 2017; 10(1): 32.
  54. Zhang J, Qiu X, Gui Y, et al. Dehydroepiandrosterone improves the ovarian reserve of women with diminished ovarian reserve and is a potential regulator of the immune response in the ovaries. Biosci Trends. 2015; 9(6): 350–359.
  55. Mamas L, Mamas E. Premature ovarian failure and dehydroepiandrosterone. Fertil Steril. 2009; 91(2): 644–646.
  56. Yilmaz N, Uygur D, Inal H, et al. Dehydroepiandrosterone supplementation improves predictive markers for diminished ovarian reserve: serum AMH, inhibin B and antral follicle count. Eur J Obstet Gynecol Reprod Biol. 2013; 169(2): 257–260.
  57. Singh N, Zangmo R, Kumar S, et al. A prospective study on role of dehydroepiandrosterone (DHEA) on improving the ovarian reserve markers in infertile patients with poor ovarian reserve. Gynecol Endocrinol. 2013; 29(11): 989–992.
  58. Fouany MR, Sharara FI. Is there a role for DHEA supplementation in women with diminished ovarian reserve? J Assist Reprod Genet. 2013; 30(9): 1239–1244.
  59. Agarwal R, Shruthi R, Radhakrishnan G, et al. Evaluation of Dehydroepiandrosterone Supplementation on Diminished Ovarian Reserve: A Randomized, Double-Blinded, Placebo-Controlled Study. J Obstet Gynaecol India. 2017; 67(2): 137–142.
  60. Jankowska K, Maksym R, Zgliczyński W. Dehydroepiandrosterone can restore the function of the ovaries – a series of 5 cases and a review of the literature. J Obstet Gynecol Investig. 2019; 2(1): 11–18.
  61. Hassa H, Aydin Y, Ozatik O, et al. Effects of dehydroepiandrosterone (DHEA) on follicular dynamics in a diminished ovarian reservein vivomodel. Syst Biol Reprod Med. 2015; 61(3): 117–121.
  62. Mahmoud YI, Mahmoud AA, Abo-Zeid FS, et al. Effects of dehydroepiandrosterone on the ovarian reserve and pregnancy outcomes in perimenopausal rats (DHEA and fertility in perimenopausal rats). Life Sci. 2018; 199: 131–138.
  63. Bińkowska M, Paszkowski T, Violetta SP, et al. Position statement by Experts of the Polish Menopause and Andropause Society, and the Polish Society of Aesthetic and Reconstructive Gynaecology on the medicinal product Intrarosa. Prz Menopauzalny. 2019; 18(3): 127–132.
  64. Scheffers C, Armstrong S, Cantineau A, et al. Dehydroepiandrosterone for menopausal women. Cochrane Database of Systematic Reviews. 2014.

Regulations

Important: This website uses cookies. More >>

The cookies allow us to identify your computer and find out details about your last visit. They remembering whether you've visited the site before, so that you remain logged in - or to help us work out how many new website visitors we get each month. Most internet browsers accept cookies automatically, but you can change the settings of your browser to erase cookies or prevent automatic acceptance if you prefer.

By VM Media Group sp. z o.o., ul. Świętokrzyska 73, 80–180 Gdańsk
tel.:+48 58 320 94 94, faks:+48 58 320 94 60, e-mail:  viamedica@viamedica.pl