Skip to main content

Advertisement

Log in

Improvement of Pregnancy Rate and Live Birth Rate in Poor Ovarian Responders by Intraovarian Administration of Autologous Menstrual Blood Derived- Mesenchymal Stromal Cells: Phase I/II Clinical Trial

  • Published:
Stem Cell Reviews and Reports Aims and scope Submit manuscript

Abstract

The study was aimed to evaluate the safety and efficacy of cell therapy using autologous menstrual blood derived- mesenchymal stromal cells (Men-MSCs) in fertility potential of poor ovarian responders (PORs). POR women were divided into mesenchymal stroma cell (MSC) therapy (n = 15) and routine ICSI (n = 16) groups. The cultured Men-MSCs were autologously injected into left ovary of MSC group after approval by flow cytometry, karyotyping, endotoxin, sterility and mycoplasma tests. Changes in anti-Mullerian hormone (AMH), antral follicles count (AFC), oocytes and embryos number, clinical pregnancy rate and live birth rate were followed in both groups up to one year after treatment. 4 of 15 participants in MSC group got naturally pregnant during 3 months after cell administration, in contrast to no natural conception in control group (P = 0.04). The mean AMH level did not significantly differ with that of previous cycle or control group. Although mean AFC and oocytes number in MSC group did not indicate considerable difference with those of control group, raise of these parameters in comparison with previous cycle was significant (both P = 0.01). Nonetheless, oocyte fertilization rate and embryo number in MSC group were higher than control group (P = 0.04 and P = 0.008, respectively). Altogether, 7 of 15 women in MSC group and 2 of 16 women in routine ICSI group had clinical pregnancy that resulted in 5 live births in main group and one birth in control group. In conclusion, cell therapy using Men-MSCs could be considered as a potential treatment to restore fertility capability of POR women.

The trial registration number (TRN): IRCT20180619040147N2.

Date of registration: 2018-08-21.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

References

  1. Devine, K., Mumford, S. L., Wu, M., DeCherney, A. H., Hill, M. J., & Propst, A. (2015). Diminished ovarian reserve in the United States assisted reproductive technology population: Diagnostic trends among 181,536 cycles from the Society for Assisted Reproductive Technology Clinic Outcomes Reporting System. Fertility and Sterility, 104(3), 612–619.

    PubMed  PubMed Central  Google Scholar 

  2. Papathanasiou, A., Searle, B. J., King, N. M., & Bhattacharya, S. (2016). Trends in ‘poor responder’research: Lessons learned from RCTs in assisted conception. Human Reproduction Update, 22(3), 306–319.

    PubMed  Google Scholar 

  3. Ferraretti, A., La Marca, A., Fauser, B., Tarlatzis, B., Nargund, G., Gianaroli, L., & ESHRE working group on Poor Ovarian Response Definition. (2011). ESHRE consensus on the definition of ‘poor response' to ovarian stimulation for in vitro fertilization: The Bologna criteria. Human Reproduction, 26(7), 1616–1624.

    CAS  PubMed  Google Scholar 

  4. Humaidan, P., Alviggi, C., Fischer, R., & Esteves, S. C. (2016). The novel POSEIDON stratification of ‘low prognosis patients in assisted reproductive Technology’and its proposed marker of successful outcome. F1000Research, 23(5), 2911.

    Google Scholar 

  5. Kyrou, D., Kolibianakis, E. M., Venetis, C. A., Papanikolaou, E. G., Bontis, J., & Tarlatzis, B. C. (2009). How to improve the probability of pregnancy in poor responders undergoing in vitro fertilization: A systematic review and meta-analysis. Fertility and Sterility, 91(3), 749–766.

    CAS  PubMed  Google Scholar 

  6. Alsbjerg, B., Haahr, T., Elbaek, H. O., Laursen, R., Povlsen, B. B., & Humaidan, P. (2019). Dual stimulation using corifollitropin alfa in 54 Bologna criteria poor ovarian responders–a case series. Reproductive Biomedicine Online, 38(5), 677–682.

    CAS  PubMed  Google Scholar 

  7. Lin, L.-T., Vitale, S. G., Chen, S.-N., Wen, Z.-H., Tsai, H.-W., Chern, C.-U., & Tsui, K.-H. (2018). Luteal phase ovarian stimulation may improve oocyte retrieval and oocyte quality in poor ovarian responders undergoing in vitro fertilization: Preliminary results from a single-center prospective pilot study. Advances in Therapy, 35(6), 847–856.

    PubMed  Google Scholar 

  8. Kuroda, K., Kitade, M., Kumakiri, J., Jinushi, M., Shinjo, A., Ozaki, R., Ikemoto, Y., Katoh, N., & Takeda, S. (2016). Minimum ovarian stimulation involving combined clomiphene citrate and estradiol treatment for in vitro fertilization of Bologna-criteria poor ovarian responders. Journal of Obstetrics and Gynaecology Research, 42(2), 178–183.

    CAS  PubMed  Google Scholar 

  9. Chern, C. U., Tsui, K. H., Vitale, S. G., Chen, S. N., Wang, P. H., Cianci, A., Tsai, H. W., Wen, Z. H., & Lin, L. T. (2018). Dehydroepiandrosterone (DHEA) supplementation improves in vitro fertilization outcomes of poor ovarian responders, especially in women with low serum concentration of DHEA-S: A retrospective cohort study. Reproductive Biology and Endocrinology, 16(1), 90.

    CAS  PubMed  Google Scholar 

  10. Zhang, M., Niu, W., Wang, Y., Xu, J., Bao, X., Wang, L., Du, L., & Sun, Y. (2016). Dehydroepiandrosterone treatment in women with poor ovarian response undergoing IVF or ICSI: A systematic review and meta-analysis. Journal of Assisted Reproduction and Genetics, 33(8), 981–991.

    PubMed  PubMed Central  Google Scholar 

  11. Noventa, M., Vitagliano, A., Andrisani, A., Blaganje, M., Viganò, P., Papaelo, E., Scioscia, M., Cavallin, F., Ambrosini, G., & Cozzolino, M. (2019). Testosterone therapy for women with poor ovarian response undergoing IVF: A meta-analysis of randomized controlled trials. Journal of Assisted Reproduction and Genetics, 36(4), 673–683.

    PubMed  PubMed Central  Google Scholar 

  12. Li, X. L., Wang, L., Lv, F., Huang, X. M., Wang, L. P., Pan, Y., & Zhang, X. M. (2017). The influence of different growth hormone addition protocols to poor ovarian responders on clinical outcomes in controlled ovary stimulation cycles: A systematic review and meta-analysis. Medicine (Baltimore), 96(12), e6443.

    CAS  Google Scholar 

  13. Cai, M. H., Liang, X. Y., Wu, Y. Q., Huang, R., & Yang, X. (2019). Six-week pretreatment with growth hormone improves clinical outcomes of poor ovarian responders undergoing in vitro fertilization treatment: A self-controlled clinical study. Journal of Obstetrics and Gynaecology Research, 45(2), 376–381.

    CAS  PubMed  Google Scholar 

  14. Gao, L., Huang, Z., Lin, H., Tian, Y., Li, P., & Lin, S. (2019). Bone marrow mesenchymal stem cells (BMSCs) restore functional endometrium in the rat model for severe Asherman syndrome. Reproductive Sciences, 26(3), 436–444.

    CAS  PubMed  Google Scholar 

  15. Tan, J., Li, P., Wang, Q., Li, Y., Li, X., Zhao, D., Xu, X., & Kong, L. (2016). Autologous menstrual blood-derived stromal cells transplantation for severe Asherman's syndrome. Human Reproduction, 31(12), 2723–2729.

    PubMed  Google Scholar 

  16. He, Y., Chen, D., Yang, L., Hou, Q., Ma, H., & Xu, X. (2018). The therapeutic potential of bone marrow mesenchymal stem cells in premature ovarian failure. Stem Cell Research & Therapy, 9(1), 263.

    CAS  Google Scholar 

  17. Lu, X., Cui, J., Cui, L., Luo, Q., Cao, Q., Yuan, W., & Zhang, H. (2019). The effects of human umbilical cord-derived mesenchymal stem cell transplantation on endometrial receptivity are associated with Th1/Th2 balance change and uNK cell expression of uterine in autoimmune premature ovarian failure mice. Stem Cell Research & Therapy, 10(1), 214.

    Google Scholar 

  18. Liu, R., Zhang, X., Fan, Z., Wang, Y., Yao, G., Wan, X., Liu, Z., Yang, B., & Yu, L. (2019). Human amniotic mesenchymal stem cells improve the follicular microenvironment to recover ovarian function in premature ovarian failure mice. Stem Cell Research & Therapy, 10(1), 299.

    Google Scholar 

  19. Edessy, M., Hosni, H. N., Shady, Y., Waf, Y., Bakr, S., & Kamel, M. (2016). Autologous stem cells therapy, the first baby of idiopathic premature ovarian failure. Acta Medica International, 3(1), 19–23. https://doi.org/10.5530/ami.2016.1.7.

    Article  Google Scholar 

  20. Herraiz, S., Romeu, M., Buigues, A., Martínez, S., Díaz-García, C., Gómez-Seguí, I., Martínez, J., Pellicer, N., & Pellicer, A. (2018). Autologous stem cell ovarian transplantation to increase reproductive potential in patients who are poor responders. Fertility and Sterility, 110(3), 496–505.

    PubMed  Google Scholar 

  21. Chen, L., Qu, J., & Xiang, C. (2019). The multi-functional roles of menstrual blood-derived stem cells in regenerative medicine. Stem Cell Research & Therapy, 10(1), 1.

    Google Scholar 

  22. Fathi-Kazerooni, M., Tavoosidana, G., Taghizadeh-Jahed, M., Khanjani, S., Golshahi, H., Gargett, C. E., Edalatkhah, H., & Kazemnejad, S. (2017). Comparative restoration of acute liver failure by menstrual blood stem cells compared with bone marrow stem cells in mice model. Cytotherapy, 19(12), 1474–1490.

    PubMed  Google Scholar 

  23. Wang, Z., Wang, Y., Yang, T., Li, J., & Yang, X. (2017). Study of the reparative effects of menstrual-derived stem cells on premature ovarian failure in mice. Stem Cell Research & Therapy, 8(1), 11.

    Google Scholar 

  24. Manshadi, M. D., Navid, S., Hoshino, Y., Daneshi, E., Noory, P., & Abbasi, M. (2019). The effects of human menstrual blood stem cells-derived granulosa cells on ovarian follicle formation in a rat model of premature ovarian failure. Microscopy Research and Technique, 82(6), 635–642.

    CAS  PubMed  Google Scholar 

  25. Lai, D., Wang, F., Yao, X., Zhang, Q., Wu, X., & Xiang, C. (2015). Human endometrial mesenchymal stem cells restore ovarian function through improving the renewal of germline stem cells in a mouse model of premature ovarian failure. Journal of Translational Medicine, 13(1), 155.

    PubMed  PubMed Central  Google Scholar 

  26. European Pharmacopoeia. (2018). European directorate for the quality of medicines & healthcare (EDQM). France: Strasbourg.

    Google Scholar 

  27. Musina, R., Belyavski, A., Tarusova, O., Solovyova, E., & Sukhikh, G. (2008). Endometrial mesenchymal stem cells isolated from the menstrual blood. Bulletin of Experimental Biology and Medicine, 145, 539–543.

    CAS  PubMed  Google Scholar 

  28. Cui, C.-H., Uyama, T., Miyado, K., Terai, M., Kyo, S., Kiyono, T., & Umezawa, A. (2007). Menstrual blood-derived cells confer human dystrophin expression in the murine model of Duchenne muscular dystrophy via cell fusion and myogenic transdifferentiation. Molecular Biology of the Cell, 18(5), 1586–1594.

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Khanjani, S., Khanmohammadi, M., Zarnani, A.H., Akhondi, M.M., Ahani, A., Ghaempanah, Z., Naderi, M.M., Eghtesad, S., and Kazemnejad, S. (2014). Comparative evaluation of differentiation potential of menstrual blood- versus bone marrow-derived stem cells into hepatocyte-like cells. PLoS one, 5;9(2):e86075.

  30. Hida, N., Nishiyama, N., Miyoshi, S., Kira, S., Segawa, K., Uyama, T., Mori, T., Miyado, K., Ikegami, Y., & Cui, C. (2008). Novel cardiac precursor-like cells from human menstrual blood-derived mesenchymal cells. Stem Cells, 26, 1695–1704.

    CAS  PubMed  Google Scholar 

  31. Meng, X., Ichim, T. E., Zhong, J., Rogers, A., Yin, Z., Jackson, J., Wang, H., Ge, W., Bogin, V., & Chan, K. W. (2007). Endometrial regenerative cells: a novel stem cell population. Journal of Translational Medicine, 5, 57.

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Borlongan, C. V., Kaneko, Y., Maki, M., Yu, S.-J., Ali, M., Allickson, J. G., Sanberg, C. D., Kuzmin-Nichols, N., & Sanberg, P. R. (2010). Menstrual blood cells display stem cell–like phenotypic markers and exert neuroprotection following transplantation in experimental stroke. Stem Cells and Development, 19, 439–452.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Patel, A. N., Park, E., Kuzman, M., Benetti, F., Silva, F. J., & Allickson, J. G. (2008). Multipotent menstrual blood stromal stem cells: Isolation, characterization, and differentiation. Cell Transplantation, 17, 303–311.

    PubMed  Google Scholar 

  34. Shokri, M. R., Bozorgmehr, M., Ghanavatinejad, A., Falak, R., Aleahmad, M., Kazemnejad, S., Shokri, F., & Zarnani, A. H. (2019). Human menstrual blood-derived stromal/stem cells modulate functional features of natural killer cells. Scientific Reports, 9(1), 10007.

    PubMed  PubMed Central  Google Scholar 

  35. Rajabi, Z., Yazdekhasti, H., Mugahi, S. M. H. N., Abbasi, M., Kazemnejad, S., Shirazi, A., Majidi, M., & Zarnani, A. H. (2018). Mouse preantral follicle growth in 3D co-culture system using human menstrual blood mesenchymal stem cell. Reproductive Biology, 18(1), 122–131.

    PubMed  Google Scholar 

  36. Bhartiya, D., & Patel, H. (2018). Ovarian stem cells-resolving controversies. Journal of Assisted Reproduction and Genetics, 35, 393–398.

    PubMed  Google Scholar 

  37. Bhartiya, D. (2018). Stem cells survive oncotherapy & can regenerate non-functional gonads: A paradigm shift for oncofertility. Indian Journal of Medical Research, 148(Suppl 1), S38–S49.

    PubMed  Google Scholar 

  38. Ye, H., Zheng, T., Li, W., Li, X., Fu, X., Huang, Y., Hu, C., Li, J., Huang, J., Liu, Z., & Zheng, L. (2017). Ovarian stem cell nests in reproduction and ovarian aging. Cellular Physiology and Biochemistry, 43(5), 1917–1925.

    CAS  PubMed  Google Scholar 

  39. Asgari, H. R., Akbari, M., Yazdekhasti, H., Rajabi, Z., Navid, S., Aliakbari, F., Abbasi, N., Aval, F. S., Shams, A., & Abbasi, M. (2017). Comparison of human amniotic, chorionic, and umbilical cord multipotent mesenchymal stem cells regarding their capacity for differentiation toward female germ cells. Cellular Reprogramming (Formerly" Cloning and Stem Cells"), 19(1), 44–53.

    CAS  Google Scholar 

Download references

Acknowledgments

The authors would like to appreciate Dr.Mohammad-Reza Sadeghi (Chairman of Avicenna Research Institute), Dr. Ali Sadeghi-Tabar (Managing director of Avicenna Infertility Clinic), Miss. Haleh Edalatkhah (Quality Control Assistant), Mrs. Somayeh Khorasani (production line assistant), Mrs. Zohreh Fathi (supervisor of embryology lab) and Mrs. Fariba Mohammadi (supervisor of operation room) for their assistance in implementation of this project.

Funding

This study was financially supported by Avicenna Research Institute (grant number: 960110–029).

Author information

Authors and Affiliations

Authors

Contributions

Simin Zafardoust: As a gynaecologist, part of study design, selection and randomization of patients, stem cell injection into ovaries and follow up of participants were performed by her.

Somaieh Kazemnejad: Study design, supervision of entire cell production, quality control and administration along with manuscript writing and editing were performed by her.

Maryam Darzi: She had contribution in stem cell isolation, culture and characterization.

Mina Fathi-Kazerooni: Quality Assurance manager.

Hilda Rastegari: Quality control of culture stem cells was implemented by her.

Afsaneh Mohammadzadeh: She had contribution in participant’s selection and their follow up after stem cell administration.

Corresponding author

Correspondence to Somaieh Kazemnejad.

Ethics declarations

Competing Interests

The authors declare that they have no competing interests.

Ethics Approval

The study was started after providing complete information to patients and obtaining signed written consent. All the procedures in this clinical trial study were conducted according to the Declaration of Helsinki, Good Clinical Practice or good manufacturing practice (GMP) guideline approved by Biomedical Research Ethics Committee of Academic Center for Education, Culture and Research (ACECR, Tehran, Iran). The study was registered at the Iranian Registry of Clinical Trials (IRCT20180619040147N2). The cell manufacturing was performed by STERCO (Tehran, Iran) in GMP clean room authorized by Iran food and drug administration authorities.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Simin Zafardoust and Somaieh Kazemnejad are equally the first author

Electronic supplementary material

Supplementary Fig. 1

Flow chart of isolation, culture, expansion, banking, and infusion of Men-MSCs (JPG 1.42 mb)

Supplementary Fig. 2

Morphology of menstrual blood stem cells in the first passage obtained from each participant in the cell therapy group. Scale bar: 300 μM (JPG 4.74 mb)

Supplementary Fig. 3

Immunophenotypic properties of cultured cells were assessed by flow cytometry. In brief, aliquots of 105 cells/100 μL were incubated separately with PE-conjugated anti-human CD73 (clone AD2; BD Pharmingen) or PE-conjugated anti human CD44 (clone 515;BD Pharmingen), CD90 (clone 5E10; BioLegend), CD45 (clone 2D1; BioLegend) for 30 min at 4 °C. Afterwards, all cell suspensions were washed twice with PBS-fetal bovine serum 2% and analyzed using a flow cytometer (Partec GmbH, Munster, Germany) using appropriate isotype controls. Representative histograms for CD markers of two samples (first participant and 15th participant) are demonstrated (gray). The respective isotype control is shown as black line. (JPG 614 kb)

ESM 4

(DOCX 47 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zafardoust, S., Kazemnejad, S., Darzi, M. et al. Improvement of Pregnancy Rate and Live Birth Rate in Poor Ovarian Responders by Intraovarian Administration of Autologous Menstrual Blood Derived- Mesenchymal Stromal Cells: Phase I/II Clinical Trial. Stem Cell Rev and Rep 16, 755–763 (2020). https://doi.org/10.1007/s12015-020-09969-6

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12015-020-09969-6

Keywords

Navigation