WHY ARE RECOMMENDATIONS ON THE STUDY OF EJACULATE NOT UPDATED?


Cite item

Full Text

Open Access Open Access
Restricted Access Access granted
Restricted Access Subscription or Fee Access

Abstract

In recent years, the quality of germinal cells has been recognized as the main factor determining reproductive potential. Standard sperm analysis is of limited value for predicting both the achievement of a pregnancy and the outcomes of assisted reproductive technologies, which raises the question of the additional significance of functional testing. Most current guidelines recommend to perform additional studies from a scientific point of view, recognizing their importance, but don’t include men in the basic diagnostic schemes. In this review, we try to discuss of the relevance of special sperm testing.

Full Text

Restricted Access

About the authors

E. A Efremov

NMRRC of Minzdrav of Russia

Dr.Med.Sci. Head of the Department of Andrology and Human Reproduction, N.A. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch Moscow, Russia

E. V Kasatonova

NMRRC of Minzdrav of Russia

Email: kasatonova@yandex.ru
researcher of Department of Andrology and Human Reproductive Health Moscow, Russia

Ya. I Melnik

NMRRC of Minzdrav of Russia

Researcher at the Department of Andrology and Human Reproduction, N.A. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch Moscow, Russia

A. A Nikushina

NMRRC of Minzdrav of Russia

Researcher at the Research and Methodology Department of Andrology and HumanReproduction, N.A. Lopatkin Scientific Research Institute of Urology and Interventional Radiology - branch Moscow, Russia

References

  1. Mascarenhas M.N., Flaxman S.R., Boerma T., Vanderpoel S., Stevens G.A. National, regional, and global trends in infertility prevalence since 1990: a systematic analysis of 277 health surveys. PLoS Med. 2012;9(12):e1001356. doi: 10.1371/journal.pmed.1001356.
  2. WHO | Infertility is a global public health issue [online] // Who.int. 2019. URL: http://www.who.int/reproductivehealth/topics/infertility/perspective/ en/. (accessed: 20. 06. 2019).
  3. Andreeva M.V., Khayat S.S., Sorokina T.M., Chernykh V.B., Schileyko L.V., Shtaut M.I, Mashina E.V., Dobrodeeva L.T., Sedova A.O., Kurilo L.F. TYPES OF PATHOZOOSPERMIA IN MEN WITH INFERTILITY IN MARRIAGE АШ/OR DISORDERS OF REPRODUCTIVE SYSTEM. Andrology and Genital Surgery. 2017;18(2):33-38. https://doi.org/10.17650/2070-9781- 2017-18-2-33-38. Russain (Андреева М.В., Хаят С.Ш., Сорокина Т.М., Черных В.Б., Шилейко Л.В., Штаут М.И., Машина Е.В., Добродеева Л.Т., СедоваА.О., Курило Л.Ф. Формы патозооспермии у мужчин с бесплодием в браке и/или с нарушениями репродуктивной системы. Андрология и генитальная хирургия. 2017;2(18): 33-38. doi: 10.17650/2070-9781-2017-18-2-33-38).
  4. Agarwal A., Mulgund A., Hamada A., Chyatte M.R. A unique view on male infertility around the globe. Reprod Biol Endocrinol. 2015;13:37. Doi:10.1186/ s12958-015-0032-1.
  5. Xing J.S., Bai Z.M. Is testicular dysgenesis syndrome a genetic, endocrine, or environmental disease, or an unexplained reproductive disorder? Life Sci. 2018 Feb 1;194:120-129. doi: 10.1016/j.lfs.2017.11.039.
  6. Esteves S.C., Roque M., Bedoschi G., Haahr T., Humaidan P. Intracytoplasmic sperm injection for male infertility and consequences for offspring. Nat Rev Urol. 2018;15(9):535-562. doi: 10.1038/s41585-018-0051-8.
  7. Carrell D.T., De Jonge C.J. The troubling state of the semen analysis. Andrology. 2016;4(5):761-762. doi: 10.1111/andr.12257.
  8. De Geyter C., Calhaz-Jorge C., Kupka M.S., Wyns C., Mocanu E., Motrenko T., Scaravelli G., Smeenk J., Vidakovic S., Goossens V. European IVF-monitoring Consortium (EIM) for the European Society of Human Reproduction and Embryology (ESHRE). ART in Europe, 2014: results generated from European registries by ESHRE: The European IVF-monitoring Consortium (EIM) for the European Society of Human Reproduction and Embryology (ESHRE). Hum Reprod. 2018;33(9):1586-1601. doi: 10.1093/humrep/dey242.
  9. Esteves S.C., Prudencio C., Seol B., Verza S., Knoedler C., Agarwal A. Comparison of sperm retrieval and reproductive outcome in azoospermic men with testicular failure and obstructive azoospermia treated for infertility. Asian J Androl. 2014;16(4):602-606. doi: 10.4103/1008-682X.126015.
  10. Oehninger S., Franken D.R., Ombelet W. Sperm functional tests. FertilSteril. 2014;102(6):1528-1533. doi: 10.1016/j.fertnstert.2014.09.044.
  11. Nagy ZP., Liu J, Joris H., Verheyen G., Tournaye H., Camus M., Derde M.C., Devroey P., Van Steirteghem A.C. The result of intracytoplasmic sperm injection is not related to any of the three basic sperm parameters. Hum Reprod. 1995;10(5):1123-1129. doi: 10.1093/oxfordjournals.humrep.a136104
  12. Guzick D.S., Overstreet J.W., Factor-Litvak P., Brazil C.K., Nakajima S.T., Coutifaris C., Carson S.A., Cisneros P., Steinkampf M.P., Hill J.A., Xu D., Vogel D.L. National Cooperative Reproductive Medicine Network. Sperm morphology, motility, and concentration in fertile and infertile men. N Engl J Med. 20018;345(19):1388-1393. doi: 10.1056/NEJMoa003005
  13. Simon L., Murphy K., Shamsi M.B., Liu L., Emery B., Aston K.I., Hotaling J., Carrell D.T. Paternal influence of sperm DNA integrity on early embryonic development. Hum Reprod. 2014;29(11):2402-2412. doi: 10.1093/humrep/ deu228
  14. Cannarella R., Condorelli R.A., La Vignera S., Calogero A.E. Effects of the insulin-like growth factor system on testicular differentiation and function: a review of the literature. Andrology. 2018 Jan;6(1):3-9. Doi: 10.1111/ andr.12444.
  15. Oehninger S., Ombelet W. Limits of current male fertility testing. FertilSteril. 2019;111(5):835-841. doi: 10.1016/j.fertnstert.2019.03.005.
  16. Liu D.Y., Liu M.L., Garrett C., Baker H.W. Comparison of the frequency of defective sperm-zona pellucida (ZP) binding and the ZP-induced acrosome reaction between subfertile men with normal and abnormal semen. Hum Reprod. 2007;22(7):1878-1884. doi: 10.1093/humrep/dem087.
  17. Jeyendran R.S., Caroppo E., Rouen A., Anderson A., Puscheck E. Selecting the most competent sperm for assisted reproductive technologies. FertilSteril. 2019;111(5):851-863. doi: 10.1016/j.fertnstert.2019.03.024.
  18. Sakkas D. Novel technologies for selecting the best sperm for in vitro fertilization and intracytoplasmic sperm injection. FertilSteril. 2013;99(4):1023-1029. doi: 10.1016/j.fertnstert.2012.12.025.
  19. Lepine S., McDowell S., Searle L.M., Kroon B., Glujovsky D., Yazdani A. Advanced sperm selection techniques for assisted reproduction. Cochrane Database Syst Rev. 2019;7:CD010461. doi: 10.1002/14651858.CD010461. pub3.
  20. Teixeira D.M., Barbosa M.A., Ferriani RA., Navarro P.A., Raine-Fenning N., Nastri C.O., Martins W.P. Regular (ICSI) versus ultra-high magnification (IMSI) sperm selection for assisted reproduction. Cochrane Database Syst Rev. 2013 Jul 25;(7):CD010167. doi: 10.1002/14651858.CD010167.pub2.
  21. World Health Organization. Geneva; World Health Organization; 2010. WHO Laboratory Manual for the Examination and Processing of Human Semen. 5th ed.
  22. World Health Organization. Cambridge; Cambridge University Press; 1999. WHO Laboratory Manual for Examination of Human Semen and Semen- Cervical Mucus Interaction. 4th ed.
  23. Muller C.H. Rationale, interpretation, andrology lab corner validation, and uses of sperm function tests. J Androl. 2000;21:10-30.
  24. Jarow J., Sigman M., Kolettis P.N., et al. The Optimal Evaluation ofthe Infertile Male: Best Practice Statement Reviewed and Validity Confirmed 2011. American Urological Association; 2011. https://www.auanet.org/common/ pdf/education/clinical-guidance/Male-Infertility-d.pdf (обращение от августа 2019).
  25. Practice Committee of the American Society for Reproductive Medicine. Diagnostic evaluation of the infertile male: a committee opinion. FertilSteril. 2015 Mar;103(3):e18-25. doi: 10.1016/j.fertnstert.2014.12.103.
  26. Jungwirth A., Diemer T., Kopa Z., Krausz C., Tournaye H. Male Infertility. EAU Guidelines; 2018 https://uroweb.org/guideline/male-infertility/ (обращение от августа 2019)
  27. Чалый М. Е., Ахвледиани Н. Д., Харчилава Р. Р. Мужское бесплодие. 2017; 7:148.
  28. Simon L., Murphy K., Shamsi M.B., et al. Paternal influence of sperm DNA integrity on early embryonic development. Hum Reprod2014;29:2402-412. doi: 10.1093/humrep/deu228.
  29. Spano M., Bonde J.P., Hjollund H.I., et al. Sperm chromatin damage impairs human fertility. The Danish First Pregnancy Planner Study Team. FertilSteril. 2000;73:43-50. doi: 10.1016/s0015-0282(99)00462-8.
  30. Evenson D.P., Wixon R. Data analysis of two in vivo fertility studies using Sperm Chromatin Structure Assay-derived DNA fragmentation index vs. pregnancy outcome. FertilSteril. 2008;90:1229-1231. Doi: 10.1016/j. fertnstert.2007.10.066.
  31. Bungum M, Humaidan P, Axmon A, Spano M, Bungum L, Erenpreiss J, Giwercman A. Sperm DNA integrity assessment in prediction of assisted reproduction technology outcome. Hum Reprod. 2007 Jan;22(1):174-179. doi: 10.1093/humrep/del326.
  32. Virro M.R., Larson-Cook K.L., Evenson D.P. Sperm chromatin structure assay (SCSA) parameters are related to fertilization, blastocyst development, and ongoing pregnancy in in vitro fertilization and intracytoplasmic sperm injection cycles. FertilSteril. 2004;81:1289-1295. doi: 10.1016/j.fertnstert.2003.09.063.
  33. Collins J.A., BarnhartK.T., SchlegelP.N. Do sperm DNA integrity tests predict pregnancy with in vitro fertilization? FertilSteril. 2008;89:823-831. doi: 10.1016/j.fertnstert.2007.04.055.
  34. Simon L., Proutski I., Stevenson M., Jennings D., McManus J, Lutton D., Lewis S.E. Sperm DNA damage has a negative association with live-birth rates after IVF. Reprod Biomed Online. 2013;26(1):68-78. Doi: 10.1016/j. rbmo.2012.09.019.
  35. Freire Gde C. Surgery or embolization for varicoceles in subfertile men. Sao Paulo Med J. 2013;131(1):67.
  36. Zini A., Boman J.M., Belzile E., Ciampi A. Sperm DNA damage is associated with an increased risk of pregnancy loss after IVF and ICSI: systematic review and meta-analysis. Hum Reprod. 2008 Dec;23(12):2663-2668. Doi: 10.1093/ humrep/den321.
  37. Agarwal A., Majzoub A., Esteves S.C., Ko E., Ramasamy R., Zini A. Clinical utility of sperm DNA fragmentation testing: practice recommendations based on clinical scenarios. TranslAndrol Urol. 2016;5(6):935-950. Doi:10.21037/ tau.2016.10.03
  38. Trost L.W., Nehra A. Guideline-based management of male infertility: Why do we need it?. Indian J Urol. 2011;27(1):49-57. doi: 10.4103/0970-1591.78426
  39. Gandini L., Lombardo F., Paoli D., Caponecchia L., Familiari G., Verlengia C., Dondero F., Lenzi A. Study of apoptotic DNA fragmentation in human spermatozoa. Hum Reprod. 2000 Apr;15(4):830-839. Doi: 10.1093/ humrep/15.4.830
  40. Aitken R.J., De Iuliis G.N., Finnie J.M., Hedges A., McLachlan R.I. Analysis of the relationships between oxidative stress, DNA damage and sperm vitality in a patient population: development of diagnostic criteria. Hum Reprod. 2010;25(10):2415-2426. doi: 10.1093/humrep/deq214.
  41. Hanson H.A., Mayer E.N., Anderson R.E., Aston K.I., Carrell D.T., Berger J., Lowrance W, Smith K.R., Hotaling J.M. Risk of childhood mortality in family members of men with poor semen quality. Hum Reprod. 2017;32(1):239-247. doi: 10.1093/humrep/dew289.
  42. Bisht S., Dada R. Oxidative stress: Major executioner in disease pathology, role in sperm DNA damage and preventive strategies. Front Biosci (Schol Ed) 2017;9:420-447. doi: 10.2741/s495.
  43. Lewis S.E., John Aitken R., Conner S.J., Iuliis G.D., Evenson D.P., Henkel R., Giwercman A., Gharagozloo P. The impact of sperm DNA damage in assisted conception and beyond: recent advances in diagnosis and treatment. Reprod Biomed Online. 2013;27(4):325-337. doi: 10.1016/j.rbmo.2013.06.014.
  44. Smits R.M., Mackenzie-Proctor R., Yazdani A., Stankiewicz M.T., Jordan V., Showell M.G. Antioxidants for male subfertility. Cochrane Database Syst Rev. 2019 Mar 14;3:CD007411. doi: 10.1002/14651858.CD007411.pub4.
  45. Неймарк А.И., Неймарк Б.А., Давыдов А.В., Ноздрачев Н.А., Борисенко Д.В., Архипов Д.О., Борисова О.Г., Семенова Н.А. Методы коррекции фрагментации ДНК сперматозоидов в сочетании с варикоцеле у мужчин с бесплодием в браке. РМЖ. 2017;8:506-510).
  46. Цуканов А.Ю., Делицин А.С. Влияние Андродоза на качество семенной плазмы. Андрология и генитальная хирургия. 2017;18(1):99-101. https://doi. org/10.17650/2070-9781-2017-18-1-99-101).
  47. Попова А.Ю., Овчинников Р.И., Гамидов С.И. Антиоксидантная терапия улучшает показатели НВА-теста у мужчин с бесплодием при подготовке к программам вспомогательных репродуктивных технологий (ЭКО/ИКСИ). Урология. 2019;(1):90-96. https://dx.doi.org/10.18565/urology.20191.90-96.
  48. Неймарк А.И., Клепикова И.И., Неймарк Б.А., Ноздрачев НА. Применение препарата АндроДоз у мужчин с нарушением фертильности. Андрология и генитальная хирургия. 2013;14(4):49-52. https://doi.org/10.17650/2070-9781-2013-4-49-52.
  49. Halpern J.A., Brannigan R.E. Barriers, potential solutions, and alternatives to randomized clinical trials in male reproductive medicine. FertilSteril. 2019;111(5):871-872. doi: 10.1016/j.fertnstert.2019.03.038.
  50. Spencer E.A., Mahtani K.R., Goldacre B., Heneghan C. Claims for fertility interventions: a systematic assessment of statements on UK fertility centre websites. BMJ Open. 2016 Nov 27;6(11):e013940. Doi: 10.1136/ bmjopen-2016-013940.
  51. Battelle Technology Partnership Practice. Biopharmaceutical industry-sponsored clinical trials: impact on state economies. 2015. http://phrma-docs. phrma.org/sites/default/files/pdf/biopharmaceutical-industry-sponsored-clinical-trials-impact-on-state-economies.pdf. (обращение от августа 2019)
  52. Pandiyan N., Pandiyan R., Raja D.R. A perspective on sperm DNA fragmentation. TranslAndrolUrol. 2017;6(4):661-S664. Doi:10.21037/ tau.2017.03.84

Supplementary files

Supplementary Files
Action
1. JATS XML

This website uses cookies

You consent to our cookies if you continue to use our website.

About Cookies