Current approaches to diagnosing and treating endometrial receptor apparatus disorders in patients with recurrent implantation failures


Cite item

Full Text

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

Abstract

The given review analyzes the data available in the literature on the main etiological factors and pathogenetic mechanisms of endometrial susceptibility disorders in patients with recurrent implantation failures in assisted reproductive technology programs. The paper includes the publications available in the Pubmed database (https://pubmed.ncbi.nlm.nih.gov/) on this topic. It gives information on modern molecular genetic methods for determining the personalized window of implantation and for correcting the timing of embryo transfer, for identifying the pathogenic microorganisms that cause chronic endometritis, and for analyzing the species and quantitative composition of the endometrial microbiota. It summarizes the results of the most effective methods of immunotherapy, which are aimed at improving the receptive properties of the endometrium, by using platelet-rich plasma, mononuclear cells, granulocyte-stimulating factor, growth hormone, and the drugs dexamethasone and letrozole. The paper reflects the expediency of etiotropic antibacterial therapy in patients with chronic endometritis and recurrent implantation failures at the stage of preparation for assisted reproductive technology programs. It shows an association between endometrial scratching and improved reproductive outcomes in patients with recurrent implantation failures. Special attention in overcoming infertility is paid to the possibilities of cell therapy using mesenchymal stem cells in women with Asherman’s syndrome and endometrial hypoplasia. Conclusion. Patients with multiple implantation failures in assisted reproductive technology programs require the in-depth diagnosis of the endometrial receptor apparatus and the application of personalized approach to preparing the endometrium.

Full Text

Restricted Access

About the authors

Maya M. Maslova

I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University)

Email: mayay2012@mail.ru
MD, PhD, Department of Obstetrics, Gynecology, Perinatology and Reproductology

Stanislav V. Pavlovich

I.M. Sechenov First Moscow State Medical University, Ministry of Health of Russia (Sechenov University); Academician V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia

Email: s_pavlovich@oparina4.ru
MD, Academic Secretary

Veronika Yu. Smolnikova

Academician V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology, and Perinatology, Ministry of Health of Russia

Email: veronika.smolnikova@mail.ru
MD, PhD, leading researcher, Department of assistive reproductive technology in the treatment of infertility; Professor of the Department of Obstetrics, Gynecology, Perinatology and Reproductology

References

  1. Thornhill A.R., deDie-Smulders C.E., Geraedts J.P., Harper J.C., Harton G.L., Lavery S.A. et al. ESHRE PGD Consortium “Best practice guidelines for clini cal preimplantation genetic diagnosis (PGD) and preimplantation genetic scre ening (PGS)”. Hum. Reprod. 2005; 20(1): 35-48. https://dx.doi.org/10.1093/ humrep/deh579.
  2. Kolibianakis E.M., Venetis C.A., eds. Recurrent implantation failure. Taylor and Francis Group; 2019. 186p. https://dx.doi.org/10.1201/9781315165707.
  3. Teh W.T., McBain J., Rogers P. What is the contribution of embryo-endometrial asynchrony to implantation failure? J. Assist. Reprod. Genet. 2016; 33(11): 1419-30. https://dx.doi.org/10.1007/s10815-016-0773-6.
  4. Moustafa S., Young S.L. Diagnostic and therapeutic options in recurrent implantation failure. F1000Res. 2020; 9: F1000 Faculty Rev-208. https://dx.doi. org/10.12688/f1000research.22403.1.
  5. Diaz-Gimeno P., Ruiz-Alonso M., Blesa D., Bosch N., Martinez-Conejero J.A., Alama P. et al. The accuracy and reproducibility of the endometrial receptivity array is superior to histology as a diagnostic method for endometrial receptivity. Fertil. Steril. 2013; 99(2): 508-17. https://dx.doi.org/10.1016/j. fertnstert.2012.09.046.
  6. Tan J., Kan A., Hitkari J., Taylor B., Tallon N., Warraich G. et al. The role of the endometrial receptivity array (ERA) in patients who have failed euploid embryo transfers. J. Assist. Reprod. Genet. 2018; 35(4): 683-92. https://dx.doi. org/10.1007/s10815-017-1112-2.
  7. Patel J.A., Patel A.J., Banker J.M., Shah S.I., Banker M.R. Personalized embryo transfer helps in improving In vitro fertilization/ICSI outcomes in patients with recurrent implantation failure. J. Hum. Reprod. Sci. 2019; 12(1): 59-66. https:// dx.doi.org/10.4103/jhrs.JHRS_74_18.
  8. Hashimoto T., Koizumi M., Doshida M., Toy a M., Sagar a E., Oka N. et al. Efficacy of the endometrial receptivity array for repeated implantation failure in Japan: a retrospective, two-centers study. Reprod. Med. Biol. 2017; 16(3): 290-6. https://dx.doi.org/10.1002/rmb2.12041.
  9. Valdes C.T., Schutt A., Simon C. Implantation failure of endometrial origin: it is not pathology, but our failure to synchronize the developing embryo with a receptive endometrium. Fertil. Steril. 2017; 108(1): 15-8. https://dx.doi.org/ 10.1016/j.fertnstert.2017.05.033.
  10. Koler M., Achache H., Tsafrir A., Smith Y., Revel A., Reich R. Disrupted gene pattern in patients with repeated in vitro fertilization (IVF) failure. Hum. Reprod. 2009; 24(10): 2541-8. https://dx.doi.org/10.1093/humrep/dep193.
  11. Tapia A., Gangi L.M., Zegers-Hochschild F., Balmaceda J., Pommer R., Trejo L. et al. Differences in the endometrial transcript profile during the receptive period between women who were refractory to implantation and those who achieved pregnancy. Hum. Reprod. 2008; 23(2): 340-51. https://dx.doi.org/ 10.1093/ humrep/dem319.
  12. Achache H., Tsafrir A., Prus D., Reich R., Revel A. Defective endometrial prostaglandin synthesis identified in patients with repeated implantation failure undergoing in vitro fertilization. Fertil. Steril. 2010; 94(4): 1271-8. https:// dx.doi.org/10.1016/j.fertnstert.2009.07.1668.
  13. Macklon N. Recurrent implantation failure is a pathology with a specific transcriptomic signature. Fertil. Steril. 2017; 108(1): 9-14. https:// dx.doi.org/10.1016/j.fertnstert.2017.05.028.
  14. Puente E., Alonso L., Lagana A.S. Chronic endometritis: old problem, novel insights and future challenges. Int. J. Fertil. Steril. 2020; 13(4): 250-6. https:// dx.doi.org/10.22074/ijfs.2020.5779.
  15. Таболова В.К., Корнеева И.Е., Донников А.Е., Бурменская О.В., Маслова М.А., Смольникова В.Ю. Профиль локальной экспрессии генов ростовых факторов и цитокинов в эндометрии периода «имплантационного окна» при хроническом эндометрите. Акушерство и гинекология. 2014; 12: 74-8. [Tabolova V.K., Korneeva I.E., Donnikov A.E., Burmenskaya O.V., Maslova M.A., Smolnikova V.Yu. The local endometrial expression profile of the growth factor and cytokine genes during the implantation window in chronic endometritis. Akusherstvo i ginekologiya/Obstetrics and Gynecology. 2014; 12: 74-8. (in Russian)].
  16. Moreno I., Cicinelli E., Garcia-Grau I., Gonzalez-Monfort M., Bau D., Vilella F. et al. The diagnosis of chronic endometritis in infertile asymptomatic women: a comparative study of histology, microbial cultures, hysteroscopy, and molecular microbiology. Am. J. Obstet. Gynecol. 2018; 218(6): 602. e1-16. https://dx.doi. org/10.1016/j.ajog.2018.02.012.
  17. Cicinelli E., Matteo M., Tinelli R., Lepera A., Alfonso R., Indraccolo U. et al. Prevalence of chronic endometritis in repeated unexplained implantation failure and the IVF success rate after antibiotic therapy. Hum. Reprod. 2015; 30(2): 323-30. https://dx.doi.org/10.1093/humrep/ deu292.
  18. Kitaya K., Matsubayashi H., Takaya Y., Nishiyama R., Yamaguchi K., Takeuchi T. et al. Live birth rate following oral antibiotic treatment for chronic endometritis in infertile women with repeated implantation failure. Am. J. Reprod. Immunol. 2017; 78(5). https://dx.doi.org/10.1111/aji.12719.
  19. Sfakianoudis K., Simopoulou M., Nikas Y., Rapani A., Nitsos N., Pierouli K. et al. Efficient treatment of chronic endometritis through a novel approach of intrauterine antibiotic infusion: a case series. BMC Womens Health. 2018; 18(1): 197. https://dx.doi.org/10.1186/s12905-018-0688-8.
  20. Zhang Y., Xu H., Liu Y., Zheng S., Zhao W., Wu D. et al. Confirmation of chronic endometritis in repeated implantation failure and success outcome in IVF-ET after intrauterine delivery of the combined administration of antibiotic and dexamethasone. Am. J. Reprod. Immunol. 2019; 82(5): e13177. https://dx.doi. org/10.1111/aji.13177.
  21. Vitagliano A., Saccardi C., Noventa M., Di Spiezio Sardo A., Saccone G., Cicinelli E. et al. Effects of chronic endometritis therapy on in vitro fertilization outcome in women with repeated implantation failure: a systematic review and meta-analysis. Fertil. Steril. 2018; 110(1): 103-12. e1. https://dx.doi. org/10.1016/j.fertnstert.2018.03.017.
  22. Cicinelli E., De Ziegler D., Nicoletti R., Colafiglio G., Saliani N., Resta L. et al. Chronic endometritis: correlation among hysteroscopic, histologic, and bacteriologic findings in a prospective trial with 2190 consecutive office hysteroscopies. Fertil. Steril. 2008; 89(3): 677-84. https://dx.doi.org/ 10.1016/j. fertnstert.2007.03.074.
  23. Kyono K., Hashimoto T., Nagai Y., Sakuraba Y. Analysis of endometrial microbiota by 16S ribosomal RNA gene sequencing among infertile patients: a single-center pilot study. Reprod. Med. Biol. 2018; 17(3): 297-306. https:// dx.doi.org/10.1002/rmb2.12105.
  24. Moreno I., Codoner F.M., Vilella F., Valbuena D., Martinez-Blanch J.F, Jimenez-Almazan J. et al. Evidence that the endometrial microbiota has an effect on implantation success or failure. Am. J. Obstet. Gynecol. 2016; 215(6): 684-703. https://dx.doi.org/10.1016/ j.ajog.2016.09.075.
  25. Kitaya K., Nagai Y., Arai W., Sakuraba Y., Ishikawa T. Characterization of microbiota in endometrial fluid and vaginal secretions in infertile women with repeated implantation failure. Mediators Inflamm. 2019; 2019: 4893437. https:// dx.doi.org/10.1155/2019/4893437.
  26. Igenomix Foundation. EMMA Endometrial Microbiome Metagenomic Analysis. A screening test to evaluate the endometrium at the microbiological level. Available at: https://www.igenomix.com/genetic-solutions/emma-clinics/ Accessed on 23 February 2020.
  27. Hashimoto T., Kyono K. Does dysbiotic endometrium affect blastocyst implantation in IVF patients? J. Assist. Reprod. Genet. 2019; 36(12): 2471-9. https://dx.doi.org/10.1007/s10815-019-01630-7.
  28. Loeb L. The experimental proof changes in the uterine decidua of guinea pig after mating. Zentralbl. Allg. Pathol. 1907; 18: 563-5.
  29. Barash A., Dekel N., Fieldust S., Segal I., Schechtman E., Granot I. Local injury to the endometrium doubles the incidence of successful pregnancies in patients undergoing in vitro fertilization. Fertil. Steril. 2003; 79(6): 1317-22. https:// dx.doi.org/10.1016/s0015-0282(03)00345-5.
  30. Nastri C.O., Lensen S.F., Gibreel A., Raine-Fenning N., Ferriani R.A., Bhattacharya S. et al. Endometrial injury in women undergoing assisted reproductive techniques. Cochrane Database Syst. Rev. 2015; (3): CD009517. https://dx.doi.org/10.1002/14651858.CD009517.pub3.
  31. Tang Z., Hong M., He F., Huang D., Dai Z., Xuan H. et al. Effect of endometrial injury during menstruation on clinical outcomes in frozen-thawed embryo transfer cycles: a randomized control trial. J. Obstet. Gynaecol. Res. 2020; 46(3): 451-8. https://dx.doi.org/10.1111/jog.14193.
  32. Lensen S., Osavlyuk D., Armstrong S., Stadelmann C., Hennes A., Napier E. et al. A randomized trial of endometrial scratching before in vitro fertilization. N. Engl. J. Med. 2019; 380(4): 325-34. https://dx.doi.org/10.1056/NEJMoa1808737.
  33. Vitagliano A., Di Spiezio Sardo A., Saccone G., Valenti G., Sapia F., Kamath M.S. et al. Endometrial scratch injury for women with one or more previous failed embryo transfers: a systematic review and meta-analysis of randomized controlled trials. Fertil. Steril. 2018; 110(4): 687-702. e2. https://dx.doi. org/10.1016/j.fertnstert.2018.04.040.
  34. Tandulwadkar S.R., Naralkar M.V., Surana A.D., Selvakarthick M., Kharat A.H. Autologous intrauterine platelet-rich plasma instillation for suboptimal endometrium in frozen embryo transfer cycles: a pilot study. J. Hum. Reprod. Sci. 2017; 10(3): 208-12. https://dx.doi.org/10.4103/jhrs.JHRS_28_17.
  35. Farimani M., Poorolajal J., Rabiee S., Bahmanzadeh M. Successful pregnancy and live birth after intrauterine administration of autologous platelet-rich plasma in a woman with recurrent implantation failure: a case report. Int. J. Reprod. Biomed. 2017; 15(12): 803-6.
  36. Nazari L., Salehpour S., Hosseini M.S., Hashemi Moghanjoughi P. The effects of autologous platelet-rich plasma in repeated implantation failure: a randomized controlled trial. Hum. Fertil. (Camb). 2020; 23(3): 209-13. https://dx.doi.org/1 0.1080/14647273.2019.1569268
  37. Aghajanzadeh F., Esmaeilzadeh S., Basirat Z., Mahouti T., Heidari F.N., Golsorkhtabaramiri M. Using autologous intrauterine platelet-rich plasma to improve the reproductive outcomes of women with recurrent implantation failure. JBRA Assist. Reprod. 2020; 24(1): 30-3. https://dx.doi.org/10.5935/1518-0557.20190055.
  38. Maleki-Hajiagha A., Razavi M., Rezaeinejad M., Rouholamin S., Almasi-Hashiani A., Pirjani R. et al. Intrauterine administration of autologous peripheral blood mononuclear cells in patients with recurrent implantation failure: a systematic review and meta-analysis. J. Reprod. Immunol. 2019; 131: 50-6. https://dx.doi.org/10.1016/j.jri.2019.01.001.
  39. Амян Т.С., Перминова С.Г., Кречетова Л.В., Вторушина В.В. Эффективность внутриматочного введения аутологичных мононуклеарных клеток периферической крови перед переносом эмбриона у пациенток с повторными неудачами имплантации в программах вспомогательных репродуктивных технологий. Гинекология. 2018; 20(2): 28-33.
  40. Yu N., Zhang B., Xu M., Wang S., Liu R., Wu J. et al. Intrauterine administration of autologous peripheral blood mononuclear cells (PBMCs) activated by HCG improves the implantation and pregnancy rates in patients with repeated implantation failure: a prospective randomized study. Am. J. Reprod. Immunol. 2016; 76(3): 212-6. https://dx.doi.org/10.1111/aji.12542.
  41. Li S., Wang J., Cheng Y., Zhou D., Yin T., Xu W. et al. Intrauterine administration of hCG-activated autologous human peripheral blood mononuclear cells (PBMC) promotes live birth rates in frozen/thawed embryo transfer cycles of patients with repeated implantation failure. J. Reprod. Immunol. 2017; 119: 15-22. https://dx.doi.org/10.1016/j.jri.2016.11.006.
  42. Yakin K., Oktem О., Urman B. Intrauterine administration of peripheral mononuclear cells in recurrent implantation failure: a systematic review and meta-analysis. Sci. Rep. 2019; 9(1): 3897. https://dx.doi.org/10.1038/s41598-019-40521-w.
  43. Gleicher N., Vidali A, Bar ad D.H. Successful treatment of unresponsive thin endometrium. Fertil. Steril. 2011; 95(6): 2123. e13-7. https://dx.doi. org/10.1016/j.fertnstert.2011.01.143.
  44. Xu B., Zhang Q., Hao J., Xu D., Li Y. Two protocols to treat thin endometrium with granulocyte colony-stimulating factor during frozen embryo transfer cycles. Reprod. Biomed. Online. 2015; 30(4): 349-58. https://dx.doi.org/ 10.1016/j. rbmo.2014.12.006.
  45. Li J., Mo S., Chen Y. The effect of G-CSF on infertile women undergoing IVF treatment: a meta-analysis. Syst. Biol. Reprod. Med. 2017; 63(4): 23947. https://dx.doi.org/10.1080/19396368.2017.1287225.
  46. Aleyasin A., Abediasl Z, Nazari A., Sheikh M. Granulocyte colony-stimulating factor in repeated IVF failure, a randomized trial. Reproduction. 2016; 151(6): 637-42. https://dx.doi.org/10.1530/REP-16-0046.
  47. Zeyneloglu H.B., Tohma Y.A., Onalan G., Moran U. Granulocyte colony-stimulating factor for intracytoplasmic sperm injection patients with repeated implantation failure: which route is best? J. Obstet. Gynaecol. 2020; 40(4): 526 30. https://dx.doi.org/10.1080/01443615.2019.1631772
  48. Homburg R., Eshel A., Abdalla H.I., Jacobs H.S. Growth hormone facilitates ovulation induction by gonadotrophins. Clin. Endocrinol. (Oxf). 1988; 29(1): 113-7. https://dx.doi.org/10.1111/j.1365-2265.1988.tb00252.x.
  49. Altmae S., Aghajanova L. Growth hormone and endometrial receptivity. Front. Endocrinol. (Lausanne). 2019; 10: 653. https://dx.doi.org/10.3389/ fendo.2019.00653.
  50. Altmae S., Mendoza-Tesarik R., Mendoza C., Mendoza N., Cucinelli F., Tesarik J. Effect of growth hormone on uterine receptivity in women with repeated implantation failure in an oocyte donation program: a randomized controlled trial. J. Endocr. Soc. 2018; 2(1): 96-105. https://dx.doi.org/10.1210/js.2017-00359.
  51. Chen Y., Liu F., Nong Y., Ruan J., Guo Q., Luo M. et al. Clinical efficacy and mechanism of growth hormone action in patients experiencing repeat implantation failure. Can. J. Physiol. Pharmacol. 2018; 96(9): 929-32. https:// dx.doi.org/10.1139/cjpp-2017-0786.
  52. Zhang T., Huang C., Du Y., Lian R., Mo M., Zeng Y. et al. Successful treatment with intrauterine delivery of dexamethasone for repeated implantation failure. Am. J. Reprod. Immunol. 2017; 78(6). https://dx.doi.org/10.1111/aji.12766.
  53. Miller P.B., Parnell B.A., Bushnell G., Tallman N., Forstein D.A., Higdon H.L. 3rd. et al. Endometrial receptivity defects during IVF cycles with and without letrozole. Hum. Reprod. 2012; 27(3): 881-8. https://dx.doi.org/ 10.1093/ humrep/der452.
  54. Steiner N., Shrem G., Tannus S., Dahan S.Y., Balayla J., Volodarsky-Perel A. et al. Effect of GnRH agonist and letrozole treatment in women with recurrent implantation failure. Fertil. Steril. 2019; 112(1): 98-104. https://dx.doi. org/10.1016/j.fertnstert.2019.03.021.
  55. Chan R.W., Schwab K.E., Gargett C.E. Clonogenicity of human endometrial epithelial and stromal cells. Biol. Reprod. 2004; 70(6): 1738-50. https://dx.doi. org/10.1095/biolreprod.103.024109.
  56. Tersoglio A.E., Tersoglio S., Salatino D.R., Castro M., Gonzalez A., Hinojosa M. et al. Regenerative therapy by endometrial mesenchymal stem cells in thin endometrium with repeated implantation failure. A novel strategy. JBRA Assist. Reprod. 2020; 24(2): 118-27. https://dx.doi.org/10.5935/1518-0557.20190061.
  57. Aizi R., Aghebati-Maleki L., Nouri M., Marofi F., Negargar S., Yousefi M. Stem cell therapy in Asherman syndrome and thin endometrium: stem cell-based therapy. Biomed. Pharmacother. 2018; 102: 333-43. https://dx.doi.org/ 10.1016/j.biopha.2018.03.091.
  58. Кузнецова И.В., Землина Н.С., Мусина Р.А., Мусина К.С., Бекчанова Е.С., Джибладзе Т.А., Коваленко М.А., Тапильская Н.И., Радзинский В.Е. Применение аутологичных мезенхимальных стволовых клеток с целью восстановления эндометрия у пациенток со сниженной фертильностью. Вопросы гинекологии, акушерства и перинатологии. 2019; 18(6): 34-40.

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