Skip to main content
Top
Published in: Journal of Translational Medicine 1/2018

Open Access 01-12-2018 | Review

Expected advances in human fertility treatments and their likely translational consequences

Author: Norbert Gleicher

Published in: Journal of Translational Medicine | Issue 1/2018

Login to get access

Abstract

Background

Due to rapid research progress in reproductive biology and reproductive clinical endocrinology, many human infertility treatments are close to potential breakthroughs and translational applications. We here review current barriers, where such breakthroughs will likely come from, what they will entail, and their potential clinical applications.

Main text

The radical nature of change will primarily benefit older women, reduce fertility treatment costs and thereby expand access to treatment. A still widely overlooked prerequisite for implantation and normal pregnancy maintenance is timely development of maternal immunological tolerance toward an implanting paternal semi-allograft, if malfunctioning associated with implantation failure and pregnancy loss, while premature termination of tolerance appears associated with premature labor, pre-eclampsia/eclampsia and gestoses of pregnancy. Common denominators between pregnancy and invasive malignancies have again been attracting attention, suggesting that, like in malignant tumors, degrees of embryo aneuploidy may affect invasiveness and ability to “disarm” the immune system’s innate response against implanting embryos. Linking tolerance to implantation, we offer evidence that the so-called “implantation window” is likely immunological rather than hormonally defined.

Conclusions

Because many here outlined treatment changes will disproportionally benefit older women, they will exert a pronounced effect on society, as increasing numbers of women at grandparental ages will become mothers.
Literature
1.
go back to reference Pei D, Beier DW, Levy-Lahad E, Marchant G, Rossant J, Izpisua Belmonte JC, Lovell-Badge R, Jaenisch R, Charo A, Baltimore D. Human embryo editing: opportunities and importance of transnational cooperation. Cell Stem Cell. 2017;21(4):423–6.CrossRefPubMed Pei D, Beier DW, Levy-Lahad E, Marchant G, Rossant J, Izpisua Belmonte JC, Lovell-Badge R, Jaenisch R, Charo A, Baltimore D. Human embryo editing: opportunities and importance of transnational cooperation. Cell Stem Cell. 2017;21(4):423–6.CrossRefPubMed
2.
go back to reference Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, Woodruff TK, Shah RN. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun. 2017;16(8):15261.CrossRef Laronda MM, Rutz AL, Xiao S, Whelan KA, Duncan FE, Roth EW, Woodruff TK, Shah RN. A bioprosthetic ovary created using 3D printed microporous scaffolds restores ovarian function in sterilized mice. Nat Commun. 2017;16(8):15261.CrossRef
3.
go back to reference Hikabe O, Hamazaki N, Nagamatsu G, Obata Y, Hirao Y, Hamada N, Shimamoto S, Imamura T, Nakashima K, Saitou M, Hayashi K. Reconstituting in vitro of the entire cycle of the mouse female germ line. Nature. 2016;539(7628):299–303.CrossRefPubMed Hikabe O, Hamazaki N, Nagamatsu G, Obata Y, Hirao Y, Hamada N, Shimamoto S, Imamura T, Nakashima K, Saitou M, Hayashi K. Reconstituting in vitro of the entire cycle of the mouse female germ line. Nature. 2016;539(7628):299–303.CrossRefPubMed
4.
go back to reference Gleicher N, Kushnir VA, Weghofer A, Barad DH. The “graying” of infertility services: an impending revolution nobody is ready for. Reprod Biol Endocrinol. 2014;9(12):63.CrossRef Gleicher N, Kushnir VA, Weghofer A, Barad DH. The “graying” of infertility services: an impending revolution nobody is ready for. Reprod Biol Endocrinol. 2014;9(12):63.CrossRef
5.
go back to reference Harrison BJ, Hilton TN, Rivière RN, Ferraro ZM, Deonandan R, Walker MC. Advanced maternal age: ethical and medical considerations for assisted reproductive technology. Int J Womens Health. 2017;16(9):561–70.CrossRef Harrison BJ, Hilton TN, Rivière RN, Ferraro ZM, Deonandan R, Walker MC. Advanced maternal age: ethical and medical considerations for assisted reproductive technology. Int J Womens Health. 2017;16(9):561–70.CrossRef
6.
go back to reference Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, Koski A, Ji D, Hayama T, Ahmed R, Darby H, Van Dyken C, Li Y, Kang E, Park AR, Kim D, Kim ST, Gong J, Gu Y, Xu X, Battaglia D, Krieg SA, Lee DM, Wu DH, Wolf DP, Heitner SB, Belmonte JCI, Amato P, Kim JS, Kaul S, Mitalipov S. Correction of pathogenic gene mutation in human embryos. Nature. 2017;548(7668):413–9.CrossRefPubMed Ma H, Marti-Gutierrez N, Park SW, Wu J, Lee Y, Suzuki K, Koski A, Ji D, Hayama T, Ahmed R, Darby H, Van Dyken C, Li Y, Kang E, Park AR, Kim D, Kim ST, Gong J, Gu Y, Xu X, Battaglia D, Krieg SA, Lee DM, Wu DH, Wolf DP, Heitner SB, Belmonte JCI, Amato P, Kim JS, Kaul S, Mitalipov S. Correction of pathogenic gene mutation in human embryos. Nature. 2017;548(7668):413–9.CrossRefPubMed
8.
go back to reference Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978;12(8085):366.CrossRef Steptoe PC, Edwards RG. Birth after the reimplantation of a human embryo. Lancet. 1978;12(8085):366.CrossRef
9.
10.
go back to reference Aach J, Lunshof J, Iyer E, Church GM. Addressing the ethical issues raised by synthetic human entities with embryo-like features. Elife 2017;6. Pii: e20674; Erratum in Elife2017;6. Pii: e27642. Aach J, Lunshof J, Iyer E, Church GM. Addressing the ethical issues raised by synthetic human entities with embryo-like features. Elife 2017;6. Pii: e20674; Erratum in Elife2017;6. Pii: e27642.
11.
go back to reference Wu J, Platero Luengo A, Gil MA, Suzuki K, Cuello C, Morales Valencia M, Parrilla I, Martinez CA, Nohalez A, Roca J, Martinez EA, Izpisua Belmonte JC. Generation of human organs in pigs via interspecies blastocyst complementation. Reprod Domest Anim. 2016;51(Suppl 2):18–24.CrossRefPubMed Wu J, Platero Luengo A, Gil MA, Suzuki K, Cuello C, Morales Valencia M, Parrilla I, Martinez CA, Nohalez A, Roca J, Martinez EA, Izpisua Belmonte JC. Generation of human organs in pigs via interspecies blastocyst complementation. Reprod Domest Anim. 2016;51(Suppl 2):18–24.CrossRefPubMed
12.
go back to reference Wu J, Platero-Luengo A, Sakurai M, Sugawara A, Gil MA, Yamauchi T, Suzuki K, Bogliotti YS, Cuello C, Morales Valencia M, Okumura D, Luo J, Vilariño M, Parrilla I, Soto DA, Martinez CA, Hishida T, Sánchez-Bautista S, Martinez-Martinez ML, Wang H, Nohalez A, Aizawa E, Martinez-Redondo P, Ocampo A, Reddy P, Roca J, Maga EA, Esteban CR, Berggren WT, Nuñez Delicado E, Lajara J, Guillen I, Guillen P, Campistol JM, Martinez EA, Ross PJ, Izpisua Belmonte JC. Interspecies chimerism with mammalian pluripotent stem cells. Cell. 2017;168(3):473–86.CrossRefPubMedPubMedCentral Wu J, Platero-Luengo A, Sakurai M, Sugawara A, Gil MA, Yamauchi T, Suzuki K, Bogliotti YS, Cuello C, Morales Valencia M, Okumura D, Luo J, Vilariño M, Parrilla I, Soto DA, Martinez CA, Hishida T, Sánchez-Bautista S, Martinez-Martinez ML, Wang H, Nohalez A, Aizawa E, Martinez-Redondo P, Ocampo A, Reddy P, Roca J, Maga EA, Esteban CR, Berggren WT, Nuñez Delicado E, Lajara J, Guillen I, Guillen P, Campistol JM, Martinez EA, Ross PJ, Izpisua Belmonte JC. Interspecies chimerism with mammalian pluripotent stem cells. Cell. 2017;168(3):473–86.CrossRefPubMedPubMedCentral
13.
go back to reference Adashi EY, Cohen IG. Going germline: mitochondrial replacement as a guide to genome editing. Cell. 2016;164(5):832–5.CrossRefPubMed Adashi EY, Cohen IG. Going germline: mitochondrial replacement as a guide to genome editing. Cell. 2016;164(5):832–5.CrossRefPubMed
14.
go back to reference Cohen IG, Adashi EY. Mitochondrial replacement therapy: the IOM report and its aftermath. Nat Rev Genet. 2016;17:189–90.CrossRefPubMed Cohen IG, Adashi EY. Mitochondrial replacement therapy: the IOM report and its aftermath. Nat Rev Genet. 2016;17:189–90.CrossRefPubMed
15.
go back to reference Wertz DC. Embryo and stem cell research in the United Sates: history and politics. Gene Ther. 2001;9(11):674–8.CrossRef Wertz DC. Embryo and stem cell research in the United Sates: history and politics. Gene Ther. 2001;9(11):674–8.CrossRef
16.
17.
go back to reference Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y, Hayama T, Van Dyken C, Wang X, Luo S, Ahmed R, Li Y, Ji D, Kayali R, Cinnioglu C, Olson S, Jensen J, Battaglia D, Lee D, Wu D, Huang T, Wolf DP, Temiakov D, Belmonte JC, Amato P, Mitalipov S. Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature. 2016;8(7632):270–5.CrossRef Kang E, Wu J, Gutierrez NM, Koski A, Tippner-Hedges R, Agaronyan K, Platero-Luengo A, Martinez-Redondo P, Ma H, Lee Y, Hayama T, Van Dyken C, Wang X, Luo S, Ahmed R, Li Y, Ji D, Kayali R, Cinnioglu C, Olson S, Jensen J, Battaglia D, Lee D, Wu D, Huang T, Wolf DP, Temiakov D, Belmonte JC, Amato P, Mitalipov S. Mitochondrial replacement in human oocytes carrying pathogenic mitochondrial DNA mutations. Nature. 2016;8(7632):270–5.CrossRef
18.
go back to reference Food and Drug Administration. Advisory on Legal Restrictions on the Use of Mitochondrial Replacement Techniques to Introduce Donor Mitochondria into Reproductive Cells Intended for Transfer into a Human Recipient from the Food and Drug Administration, again reemphasized (https://www.fda.gov/BiologicsBlood Vaccines/Cellular Gene TherapyProducts/ucm570185. htm?); Accessed November 4, 2017. Food and Drug Administration. Advisory on Legal Restrictions on the Use of Mitochondrial Replacement Techniques to Introduce Donor Mitochondria into Reproductive Cells Intended for Transfer into a Human Recipient from the Food and Drug Administration, again reemphasized (https://​www.​fda.​gov/​BiologicsBlood Vaccines/Cellular Gene TherapyProducts/ucm570185. htm?); Accessed November 4, 2017.
19.
go back to reference Zhang J, Liu H, Luo S, Lu Z, J, Liu H, Luo S, Lu Z, Chávez-Badiola A, Liu Z, Yang M, Merhi Z, Silber SJ, Munné S, Konstantinidis M, Wells D, Tang JJ, Huang T. Live birth derived from oocyte spindle transfer to prevent mitochondrial disease. J Reprod Biomed. Online 2017;34(4):361–368; Corrigendum: Reprod Biomed Online. 2017 Jul;35(1): 49; Corrigendum:Reprod Biomed Online. 2017; pii: S1472–6483(17)30317-6. Zhang J, Liu H, Luo S, Lu Z, J, Liu H, Luo S, Lu Z, Chávez-Badiola A, Liu Z, Yang M, Merhi Z, Silber SJ, Munné S, Konstantinidis M, Wells D, Tang JJ, Huang T. Live birth derived from oocyte spindle transfer to prevent mitochondrial disease. J Reprod Biomed. Online 2017;34(4):361–368; Corrigendum: Reprod Biomed Online. 2017 Jul;35(1): 49; Corrigendum:Reprod Biomed Online. 2017; pii: S1472–6483(17)30317-6.
20.
go back to reference Gleicher N, Kushnir VA, Albertini DA, Barad DH. First birth following spindle transfer. Reprod Biomed Online. 2017;35(5):542–3.CrossRefPubMed Gleicher N, Kushnir VA, Albertini DA, Barad DH. First birth following spindle transfer. Reprod Biomed Online. 2017;35(5):542–3.CrossRefPubMed
23.
go back to reference Wolf DP, Mitalipov S. Mitochondrial replacement therapies can circumvent mDNA-based disease transmission. Cell Matab. 2014;20(1):6–8.CrossRef Wolf DP, Mitalipov S. Mitochondrial replacement therapies can circumvent mDNA-based disease transmission. Cell Matab. 2014;20(1):6–8.CrossRef
25.
go back to reference Xie T, Spradling AC. A niche maintaining germ line stem cells in the Drosophilia ovary. Science. 2000;290(5490):328–30.CrossRefPubMed Xie T, Spradling AC. A niche maintaining germ line stem cells in the Drosophilia ovary. Science. 2000;290(5490):328–30.CrossRefPubMed
27.
go back to reference Horan CJ, Williams SA. Oocyte stem cells: fact or fantasy? Reproduction. 2017;15491:R23–35.CrossRef Horan CJ, Williams SA. Oocyte stem cells: fact or fantasy? Reproduction. 2017;15491:R23–35.CrossRef
28.
go back to reference Albertini DF, Gleicher N. A detour in the quest for oogonial stemc cells: methods matter. Nat Med. 2015;21(10):1126–7.CrossRefPubMed Albertini DF, Gleicher N. A detour in the quest for oogonial stemc cells: methods matter. Nat Med. 2015;21(10):1126–7.CrossRefPubMed
29.
go back to reference Hübner K, Fuhrmann G, Christenson LK, Kehler J, Reinbold R, De La Fuente R, Wood J, Strauss JF 3rd, Boiani M, Schöler HR. Derivation of oocytes from mouse embryo stem cells. Science. 2003;23(5623):1251–6.CrossRef Hübner K, Fuhrmann G, Christenson LK, Kehler J, Reinbold R, De La Fuente R, Wood J, Strauss JF 3rd, Boiani M, Schöler HR. Derivation of oocytes from mouse embryo stem cells. Science. 2003;23(5623):1251–6.CrossRef
30.
go back to reference Hayashi K, Saitou M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protocol. 2013;8(8):1513–24.CrossRef Hayashi K, Saitou M. Generation of eggs from mouse embryonic stem cells and induced pluripotent stem cells. Nat Protocol. 2013;8(8):1513–24.CrossRef
31.
go back to reference Hayashi K, Hikabe O, Obata Y, Hirao Y. Reconstitution of mouse oogenesis in a dish from pluripotent stem cells. Nat Protoc. 2017;12(9):1733–44.CrossRefPubMed Hayashi K, Hikabe O, Obata Y, Hirao Y. Reconstitution of mouse oogenesis in a dish from pluripotent stem cells. Nat Protoc. 2017;12(9):1733–44.CrossRefPubMed
32.
go back to reference Ishikura Y, Yabuta Y, Ohta H, Hayashi K, Nakamura T, Okamoto I, Yamamoto T, Kurimoto K, Shirane K, Sasaki H, Saitou M. In vitro derivation and propagation of spermatogonial stem cell activity from mouse pluripotent stem cells. Cell Rep. 2016;17(10):2789–804.CrossRefPubMed Ishikura Y, Yabuta Y, Ohta H, Hayashi K, Nakamura T, Okamoto I, Yamamoto T, Kurimoto K, Shirane K, Sasaki H, Saitou M. In vitro derivation and propagation of spermatogonial stem cell activity from mouse pluripotent stem cells. Cell Rep. 2016;17(10):2789–804.CrossRefPubMed
33.
go back to reference Eskew AM, Omurtag KR. A review on ovarian hyperstimulation syndrome (OHSS) management strategies. Minerva Endocrinol. 2017;43:50.PubMed Eskew AM, Omurtag KR. A review on ovarian hyperstimulation syndrome (OHSS) management strategies. Minerva Endocrinol. 2017;43:50.PubMed
34.
go back to reference Hirota T, Ohta H, Powell BE, Mahadevaiah SK, Ojarikre OA, Saitou M, Turner JMA. Fertile offspring from sterile sex chromosome trisomic mice. Science. 2017;357(6354):932–5.CrossRefPubMedPubMedCentral Hirota T, Ohta H, Powell BE, Mahadevaiah SK, Ojarikre OA, Saitou M, Turner JMA. Fertile offspring from sterile sex chromosome trisomic mice. Science. 2017;357(6354):932–5.CrossRefPubMedPubMedCentral
35.
go back to reference Gleicher N, Orvieto R. Is the hypothesis of preimplantation genetic screening (PGS) still supportable? A review. J Ovarian Res. 2017;27(1):21.CrossRef Gleicher N, Orvieto R. Is the hypothesis of preimplantation genetic screening (PGS) still supportable? A review. J Ovarian Res. 2017;27(1):21.CrossRef
36.
go back to reference Orvieto R, Gleicher N. Should preimplantation genetic screening (PGS) be implemented to routine IVF practice? J Assist Reprod Genet. 2016;33(11):1445–8.CrossRefPubMedPubMedCentral Orvieto R, Gleicher N. Should preimplantation genetic screening (PGS) be implemented to routine IVF practice? J Assist Reprod Genet. 2016;33(11):1445–8.CrossRefPubMedPubMedCentral
38.
go back to reference Bazrgar M, Gourabi H, Valojerdi MR, Yazdi PE, Baharvand H. Self-correction of chromosomal abnormalities in human preimplantation embryos and embryonic stem cells. Stem Cells Dev. 2013;22(17):2449–56.CrossRefPubMed Bazrgar M, Gourabi H, Valojerdi MR, Yazdi PE, Baharvand H. Self-correction of chromosomal abnormalities in human preimplantation embryos and embryonic stem cells. Stem Cells Dev. 2013;22(17):2449–56.CrossRefPubMed
40.
go back to reference Peterson SE, Westra JW, Rehen SK, Young H, Bushman DM, Paczkowski CM, Yung YC, Lynch CL, Tran HT, Nickey KS, Wang YC, Laurent LC, Loring JF, Carpenter MK, Chun J. Normal human pluripotent stem cell lines exhibit pervasive mosaic aneuploidy. PLoS ONE. 2011;6(8):e23018.CrossRefPubMedPubMedCentral Peterson SE, Westra JW, Rehen SK, Young H, Bushman DM, Paczkowski CM, Yung YC, Lynch CL, Tran HT, Nickey KS, Wang YC, Laurent LC, Loring JF, Carpenter MK, Chun J. Normal human pluripotent stem cell lines exhibit pervasive mosaic aneuploidy. PLoS ONE. 2011;6(8):e23018.CrossRefPubMedPubMedCentral
41.
go back to reference Bolton H, Graham SJ, Van der Aa N, Kumar P, Theunis K, Fernandez Gallardo E, Voet T, Zernicka-Goetz M. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat Commun. 2016;29(7):11165.CrossRef Bolton H, Graham SJ, Van der Aa N, Kumar P, Theunis K, Fernandez Gallardo E, Voet T, Zernicka-Goetz M. Mouse model of chromosome mosaicism reveals lineage-specific depletion of aneuploid cells and normal developmental potential. Nat Commun. 2016;29(7):11165.CrossRef
42.
go back to reference Scott LA. Smith S. the successful use of pronuclear embryo transfers the day following oocyte retrieval. Hum Reprod. 1998;13(4):1003–101.CrossRefPubMed Scott LA. Smith S. the successful use of pronuclear embryo transfers the day following oocyte retrieval. Hum Reprod. 1998;13(4):1003–101.CrossRefPubMed
43.
go back to reference Jaroudi K, Al-Hassan S, Sieck U, Al-Sufyan H, Al-Kabra M, Coskun S. Zygote transfer on day 1 versus cleavage stage embryo transfer on day 3: a prospective randomized trial. Hum Reprod. 2004;19(3):645–8.CrossRefPubMed Jaroudi K, Al-Hassan S, Sieck U, Al-Sufyan H, Al-Kabra M, Coskun S. Zygote transfer on day 1 versus cleavage stage embryo transfer on day 3: a prospective randomized trial. Hum Reprod. 2004;19(3):645–8.CrossRefPubMed
44.
go back to reference Gardner DK, Vella P, Lane M, Wagley L, Schlenker T, Schoolcraft WB. Culture and transfer of human blastocysts increases implantation rates and reduces the need for multiple embryo transfers. Fertil Steril. 1998;69(1):84–8.CrossRefPubMed Gardner DK, Vella P, Lane M, Wagley L, Schlenker T, Schoolcraft WB. Culture and transfer of human blastocysts increases implantation rates and reduces the need for multiple embryo transfers. Fertil Steril. 1998;69(1):84–8.CrossRefPubMed
45.
go back to reference Verlinsky Y, Cieslak J, Ivakhnenko V, Evsikov S, Wolf G, White M, Lifchez A, Kaplan B, Moise J, Valle J, Ginsberg N, Strom C, Kuliev A. Preimplantation diagnosis of common aneuploidies by the first- and second-polar body FISH analysis. J Assist Reprod Genet. 1998;15(5):285–9.CrossRefPubMedPubMedCentral Verlinsky Y, Cieslak J, Ivakhnenko V, Evsikov S, Wolf G, White M, Lifchez A, Kaplan B, Moise J, Valle J, Ginsberg N, Strom C, Kuliev A. Preimplantation diagnosis of common aneuploidies by the first- and second-polar body FISH analysis. J Assist Reprod Genet. 1998;15(5):285–9.CrossRefPubMedPubMedCentral
46.
go back to reference Kim K, Ryu SM, Kim ST, Baek G, Kim D, Lim K, Chung E, Kim S, Kim JS. Highly efficient RNA guided base editing in mouse embryos. Nat Biotechnol. 2017;35(5):435–7.CrossRefPubMed Kim K, Ryu SM, Kim ST, Baek G, Kim D, Lim K, Chung E, Kim S, Kim JS. Highly efficient RNA guided base editing in mouse embryos. Nat Biotechnol. 2017;35(5):435–7.CrossRefPubMed
48.
go back to reference Martins WP, Nastri CO, Rienzi L, van der Poel SZ, Gracia C, Racowsky C. Blastocyst vs cleavage-stage embryo transfer: systematic review and meta-analysis of reproductive outcomes. Ultrasound Obstet Gynecol. 2017;49(5):583–91.CrossRefPubMed Martins WP, Nastri CO, Rienzi L, van der Poel SZ, Gracia C, Racowsky C. Blastocyst vs cleavage-stage embryo transfer: systematic review and meta-analysis of reproductive outcomes. Ultrasound Obstet Gynecol. 2017;49(5):583–91.CrossRefPubMed
49.
go back to reference Deglincerti A, Croft GF, Pietila LN, Zernicka-Goetz M, Siggia ED, Brivanlou AH. Self-organization of the in vitro attached human embryo. Nature. 2016;12(7602):251–4.CrossRef Deglincerti A, Croft GF, Pietila LN, Zernicka-Goetz M, Siggia ED, Brivanlou AH. Self-organization of the in vitro attached human embryo. Nature. 2016;12(7602):251–4.CrossRef
50.
go back to reference Shahbazi MN, Jedrusik A, Vuoristo S, Recher G, Hupalowska A, Bolton V, Fogarty NNM, Campbell A, Devito L, Ilic D, Khalaf Y, Niakan KK, Fishel S, Zernicka-Goetz M. Self-organization of the human embryo in the absence of maternal tissues. Nat Cell Biol. 2016;18(6):700–8.CrossRefPubMedPubMedCentral Shahbazi MN, Jedrusik A, Vuoristo S, Recher G, Hupalowska A, Bolton V, Fogarty NNM, Campbell A, Devito L, Ilic D, Khalaf Y, Niakan KK, Fishel S, Zernicka-Goetz M. Self-organization of the human embryo in the absence of maternal tissues. Nat Cell Biol. 2016;18(6):700–8.CrossRefPubMedPubMedCentral
51.
go back to reference Usuda H, Watanabe S, Miura Y, Saito M, Musk GC, Rittenschober-Böhm J, Ikeda H, Sato S, Hanita T, Matsuda T, Jobe AH, Newnham JP, Stock SJ, Kemp MW. Successful maintenance of key physiological parameters in preterm lambs treated with ex vivo uterine environment therapy for a period of 1 week. Am J Obstet Gynecol. 2017;217(4):457.CrossRefPubMed Usuda H, Watanabe S, Miura Y, Saito M, Musk GC, Rittenschober-Böhm J, Ikeda H, Sato S, Hanita T, Matsuda T, Jobe AH, Newnham JP, Stock SJ, Kemp MW. Successful maintenance of key physiological parameters in preterm lambs treated with ex vivo uterine environment therapy for a period of 1 week. Am J Obstet Gynecol. 2017;217(4):457.CrossRefPubMed
52.
go back to reference Brännström M. Womb transplants with live births: an update and the future. Expert Opin Biol Ther. 2017;17(9):1105–12.CrossRefPubMed Brännström M. Womb transplants with live births: an update and the future. Expert Opin Biol Ther. 2017;17(9):1105–12.CrossRefPubMed
53.
go back to reference Ruzo A, Brivanlou AH. At last: gene editing in human embryos to understand human development. Cell Stem Cell. 2017;21(5):564–5.CrossRefPubMed Ruzo A, Brivanlou AH. At last: gene editing in human embryos to understand human development. Cell Stem Cell. 2017;21(5):564–5.CrossRefPubMed
54.
go back to reference Garg V, Morgani S. Hadjantonakis AK capturing identity and fate ex vivo: stem cells from the mouse blastocyst. Curr Top Dev Biol. 2016;120:361–400.CrossRefPubMed Garg V, Morgani S. Hadjantonakis AK capturing identity and fate ex vivo: stem cells from the mouse blastocyst. Curr Top Dev Biol. 2016;120:361–400.CrossRefPubMed
55.
56.
go back to reference Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, Lea R, Elder K, Wamaitha SE, Kim D, Maciulyte V, Kleinjung J, Kim JS, Wells D, Vallier L, Bertero A, Turner JMA, Niakan KK. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017;550(7674):67–73; Erratum: Nature. 2017; https://doi.org/10.1038/nature24292. Fogarty NME, McCarthy A, Snijders KE, Powell BE, Kubikova N, Blakeley P, Lea R, Elder K, Wamaitha SE, Kim D, Maciulyte V, Kleinjung J, Kim JS, Wells D, Vallier L, Bertero A, Turner JMA, Niakan KK. Genome editing reveals a role for OCT4 in human embryogenesis. Nature. 2017;550(7674):67–73; Erratum: Nature. 2017; https://​doi.​org/​10.​1038/​nature24292.
57.
go back to reference Goolam M, Scialdone A, Graham SJL, Macaulay IC, Jedrusik A, Hupalowska A, Voet T, Marioni JC, Zernicka-Goetz M. Heterogeneity in Oct4 and Sox2 targets biases cell fate in 4-Cell mouse embryos. Cell. 2016;24(1):61–74.CrossRef Goolam M, Scialdone A, Graham SJL, Macaulay IC, Jedrusik A, Hupalowska A, Voet T, Marioni JC, Zernicka-Goetz M. Heterogeneity in Oct4 and Sox2 targets biases cell fate in 4-Cell mouse embryos. Cell. 2016;24(1):61–74.CrossRef
58.
go back to reference Tabansky I, Lenarcic A, Draft RW, Loulier K, Keskin DB, Rosains J, Rivera-Feliciano J, Lichtman JW, Livet J, Stern JN, Sanes JR, Eggan K. Developmental bias in cleavage-stage mouse blastomers. Curr Biol. 2013;23(1):21–31.CrossRefPubMed Tabansky I, Lenarcic A, Draft RW, Loulier K, Keskin DB, Rosains J, Rivera-Feliciano J, Lichtman JW, Livet J, Stern JN, Sanes JR, Eggan K. Developmental bias in cleavage-stage mouse blastomers. Curr Biol. 2013;23(1):21–31.CrossRefPubMed
59.
go back to reference Ohnishi Y, Huber W, Tsumura A, Kang M, Xenopoulos P, Kurimoto K, Oleś AK, Araúzo-Bravo MJ, Saitou M, Hadjantonakis AK, Hiiragi T. Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineages. Nat Cell Biol. 2014;16(1):27–37.CrossRefPubMed Ohnishi Y, Huber W, Tsumura A, Kang M, Xenopoulos P, Kurimoto K, Oleś AK, Araúzo-Bravo MJ, Saitou M, Hadjantonakis AK, Hiiragi T. Cell-to-cell expression variability followed by signal reinforcement progressively segregates early mouse lineages. Nat Cell Biol. 2014;16(1):27–37.CrossRefPubMed
60.
go back to reference Saiz N, Williams KM, Seshan VE. Hadjantonakis AK Asynchronous fate decisions by single cells collectively ensure consistent lineage composition in the mouse blastocyst. Nat Commun. 2016;18(7):13463.CrossRef Saiz N, Williams KM, Seshan VE. Hadjantonakis AK Asynchronous fate decisions by single cells collectively ensure consistent lineage composition in the mouse blastocyst. Nat Commun. 2016;18(7):13463.CrossRef
61.
go back to reference Shahbazi MN, Scialdone A, Skorupska N, Weberling A, Recher G, Zhu M, Jedrusik A, Devito LG, Noli L, Macaulay IC, Buecker C, Khalaf Y, Ilic D, Voet T, Marioni JC, Zernicka-Goetz M. Pluripotent state transitions coordinate morphogenesis in mouse and human embryos. Nature. 2017;14(7684):239–43. Shahbazi MN, Scialdone A, Skorupska N, Weberling A, Recher G, Zhu M, Jedrusik A, Devito LG, Noli L, Macaulay IC, Buecker C, Khalaf Y, Ilic D, Voet T, Marioni JC, Zernicka-Goetz M. Pluripotent state transitions coordinate morphogenesis in mouse and human embryos. Nature. 2017;14(7684):239–43.
62.
go back to reference Deglincerti A, Etoc F, Guerra MC, Martyn I, Metzger J, Ruzo A, Simunovic M, Yoney A, Brivanlou AH, Siggia E, Warmflash A. Self-organization of human embryonic stem cells on micropatterns. Nat Protoc. 2016;11(11):2223–32.CrossRefPubMedPubMedCentral Deglincerti A, Etoc F, Guerra MC, Martyn I, Metzger J, Ruzo A, Simunovic M, Yoney A, Brivanlou AH, Siggia E, Warmflash A. Self-organization of human embryonic stem cells on micropatterns. Nat Protoc. 2016;11(11):2223–32.CrossRefPubMedPubMedCentral
63.
go back to reference Gammill HS, Harrington WE. Microchimerism: defining and redefining the pregnancy context—a review. Placenta. 2017;60:130–3.CrossRefPubMed Gammill HS, Harrington WE. Microchimerism: defining and redefining the pregnancy context—a review. Placenta. 2017;60:130–3.CrossRefPubMed
64.
go back to reference Paulson RJ. Preimplantation genetic screening: what is the clinical efficiency? Fertil Steril. 2017;108(2):228–30.CrossRefPubMed Paulson RJ. Preimplantation genetic screening: what is the clinical efficiency? Fertil Steril. 2017;108(2):228–30.CrossRefPubMed
65.
go back to reference Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322):8399.CrossRef Davoli T, Uno H, Wooten EC, Elledge SJ. Tumor aneuploidy correlates with markers of immune evasion and with reduced response to immunotherapy. Science. 2017;355(6322):8399.CrossRef
66.
go back to reference Gleicher N, Kushnir VA, Barad DH. 2017 Redirecting reproductive immunology research toward pregnancy as a period of temporary immune tolerance. J Assist Reprod Genet. 2017;34(4):425–30.CrossRefPubMedPubMedCentral Gleicher N, Kushnir VA, Barad DH. 2017 Redirecting reproductive immunology research toward pregnancy as a period of temporary immune tolerance. J Assist Reprod Genet. 2017;34(4):425–30.CrossRefPubMedPubMedCentral
67.
go back to reference Orvieto R, Shuly Y, Brengauz M, Feldman B. Should preimplantation genetic screening be implemented in routine clinical practice? Gynecol Endocrinol. 2016;12:1–3. Orvieto R, Shuly Y, Brengauz M, Feldman B. Should preimplantation genetic screening be implemented in routine clinical practice? Gynecol Endocrinol. 2016;12:1–3.
68.
go back to reference Gleicher N, Vidali A, Braverman J, Kushnir VA, Albertini DF, Barad DH. Further evidence against use of PGS in poor prognosis patients: report of normal births after transfer of embryos reported as aneuploid. Fertil Steril. 2015;104(Suppl):e9. Gleicher N, Vidali A, Braverman J, Kushnir VA, Albertini DF, Barad DH. Further evidence against use of PGS in poor prognosis patients: report of normal births after transfer of embryos reported as aneuploid. Fertil Steril. 2015;104(Suppl):e9.
69.
go back to reference Gleicher N, Vidali A, Braverman J, Kushnir VA, Barad DH, Hudson C, Wu YG, Wang Q, Zhang L, Albertini DF. Accuracy of preimplantation genetic screening (PGS) is compromised by degree of mosaicism of human embryos. Reprod Biol Endocrinol. 2016;14:54.CrossRefPubMedPubMedCentral Gleicher N, Vidali A, Braverman J, Kushnir VA, Barad DH, Hudson C, Wu YG, Wang Q, Zhang L, Albertini DF. Accuracy of preimplantation genetic screening (PGS) is compromised by degree of mosaicism of human embryos. Reprod Biol Endocrinol. 2016;14:54.CrossRefPubMedPubMedCentral
70.
go back to reference Greco E, Giulia Minasi M, Florentino F. Healthy babies after intrauterine transfer of mosaic aneuploid blastocysts. N Engl J Med. 2015;373:2989–99.CrossRef Greco E, Giulia Minasi M, Florentino F. Healthy babies after intrauterine transfer of mosaic aneuploid blastocysts. N Engl J Med. 2015;373:2989–99.CrossRef
71.
go back to reference Morales R, Lledó B, Ortiz JA, Ten J, Lláce J, Bernabeu R. Embryos showing mosaicism in trophectoderm cells can achieve good pregnancy rates. Hum Reprod. 2016;31(Supl 1):i14. Morales R, Lledó B, Ortiz JA, Ten J, Lláce J, Bernabeu R. Embryos showing mosaicism in trophectoderm cells can achieve good pregnancy rates. Hum Reprod. 2016;31(Supl 1):i14.
72.
go back to reference Munné S, Blazek J, Large M, Martinez-Ortiz PA, Nisson H, Liu E, Tarozzi N, Borini A, Becker A, Zhang J, Maxwell S, Grifo J, Barbariya D, Wells D, Fragouli E. Detailed investigation into the cytogenic constitution and pregnancy outcome of replacing mosaic blastocysts detected with the use of high-resolution next-generation sequencing. Fertil Steril. 2017. https://doi.org/10.1016/j.fernstert.2017.05.002.CrossRef Munné S, Blazek J, Large M, Martinez-Ortiz PA, Nisson H, Liu E, Tarozzi N, Borini A, Becker A, Zhang J, Maxwell S, Grifo J, Barbariya D, Wells D, Fragouli E. Detailed investigation into the cytogenic constitution and pregnancy outcome of replacing mosaic blastocysts detected with the use of high-resolution next-generation sequencing. Fertil Steril. 2017. https://​doi.​org/​10.​1016/​j.​fernstert.​2017.​05.​002.CrossRef
73.
go back to reference Salamonsen LA, Evans J, Nguyen HP, Edgell TA. The microenvironment of human implantation: determinant of reproductive success. Am J Reprod Immunol. 2016;75(3):218–25.CrossRefPubMed Salamonsen LA, Evans J, Nguyen HP, Edgell TA. The microenvironment of human implantation: determinant of reproductive success. Am J Reprod Immunol. 2016;75(3):218–25.CrossRefPubMed
74.
go back to reference Johnson PM, Christmas SE, Vince GS. Immunological aspects of implantation and implantation failure. Hum Reprod. 1999;14(Suppl 2):26–36.CrossRefPubMed Johnson PM, Christmas SE, Vince GS. Immunological aspects of implantation and implantation failure. Hum Reprod. 1999;14(Suppl 2):26–36.CrossRefPubMed
75.
go back to reference Chen C. Early reproductive loss. Aust N Z J Obstet Gynecol. 1986;26(3):215–8.CrossRef Chen C. Early reproductive loss. Aust N Z J Obstet Gynecol. 1986;26(3):215–8.CrossRef
76.
go back to reference Augensen K. Unruptured tubal pregnancy at term with survival of mother and child. Obstet Gynecol. 1963;61(2):259–61. Augensen K. Unruptured tubal pregnancy at term with survival of mother and child. Obstet Gynecol. 1963;61(2):259–61.
77.
78.
go back to reference Dekker GA, Robillard PY, Hulsey TC. Immune maladaptation in the etiology of preeclampsia: a review of corroborative epidemiologic studies. Obstet Gynecol Surv. 1998;53(6):377–82.CrossRefPubMed Dekker GA, Robillard PY, Hulsey TC. Immune maladaptation in the etiology of preeclampsia: a review of corroborative epidemiologic studies. Obstet Gynecol Surv. 1998;53(6):377–82.CrossRefPubMed
79.
go back to reference Saftlas AF, Rubenstein L, Prater K, Harland KK, Field E, Triche EW. Cumulative exposure to paternal seminal fluid prior to conception and subsequent risk of preeclampsia. J Reprod Immunol. 2014;101–102:104–10.CrossRefPubMed Saftlas AF, Rubenstein L, Prater K, Harland KK, Field E, Triche EW. Cumulative exposure to paternal seminal fluid prior to conception and subsequent risk of preeclampsia. J Reprod Immunol. 2014;101–102:104–10.CrossRefPubMed
80.
go back to reference Blackwell AD, Tamayo MA, Beheim B, Trumble BC, Stieglitz J, Hooper PL, Martin M, Kaplan H, Gurven M. Helminth infection, fecundity, and age of first pregnancy in women. Science. 2015;350(6263):970–82.CrossRefPubMedPubMedCentral Blackwell AD, Tamayo MA, Beheim B, Trumble BC, Stieglitz J, Hooper PL, Martin M, Kaplan H, Gurven M. Helminth infection, fecundity, and age of first pregnancy in women. Science. 2015;350(6263):970–82.CrossRefPubMedPubMedCentral
81.
go back to reference Gleicher N. Does the immune system induce labor? Lessons from preterm deliveries in women with autoimmune diseases. Clin rev Allerg Immunol. 2010;39(3):194–206.CrossRef Gleicher N. Does the immune system induce labor? Lessons from preterm deliveries in women with autoimmune diseases. Clin rev Allerg Immunol. 2010;39(3):194–206.CrossRef
82.
go back to reference Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, Mc Henry KT, Pinchback RM, Ligon AH, Cho YJ, Haery L, Greulich H, Reich M, Winckler W, Lawrence MS, Weir BA, Tanaka KE, Chiang DY, Bass AJ, Loo A, Hoffman C, Prensner J, Liefeld T, Gao Q, Yecies D, Signoretti S, Maher E, Kaye FJ, Sasaki H, Tepper JE, Fletcher JA, Tabernero J, Baselga J, Tsao MS, Demichelis F, Rubin MA, Janne PA, Daly MJ, Nucera C, Levine RL, Ebert BL, Gabriel S, Rustgi AK, Antonescu CR, Ladanyi M, Letai A, Garraway LA, Loda M, Beer DG, True LD, Okamoto A, Pomeroy SL, Singer S, Golub TR, Lander ES, Getz G, Sellers WR, Meyerson M. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905.CrossRefPubMedPubMedCentral Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, Mc Henry KT, Pinchback RM, Ligon AH, Cho YJ, Haery L, Greulich H, Reich M, Winckler W, Lawrence MS, Weir BA, Tanaka KE, Chiang DY, Bass AJ, Loo A, Hoffman C, Prensner J, Liefeld T, Gao Q, Yecies D, Signoretti S, Maher E, Kaye FJ, Sasaki H, Tepper JE, Fletcher JA, Tabernero J, Baselga J, Tsao MS, Demichelis F, Rubin MA, Janne PA, Daly MJ, Nucera C, Levine RL, Ebert BL, Gabriel S, Rustgi AK, Antonescu CR, Ladanyi M, Letai A, Garraway LA, Loda M, Beer DG, True LD, Okamoto A, Pomeroy SL, Singer S, Golub TR, Lander ES, Getz G, Sellers WR, Meyerson M. The landscape of somatic copy-number alteration across human cancers. Nature. 2010;463(7283):899–905.CrossRefPubMedPubMedCentral
83.
go back to reference Ghorani E, Kaur B, Fisher RA, Short D, Joneborg U, Carlson JW, Akarca A, Marafioti T, Quezado SA, Sarwar N, Seckl MJ. Pembrolizumab effective for drug-resistant gestational trophoblastic neioplasia. Lancet. 2017;390:2343–5.CrossRefPubMed Ghorani E, Kaur B, Fisher RA, Short D, Joneborg U, Carlson JW, Akarca A, Marafioti T, Quezado SA, Sarwar N, Seckl MJ. Pembrolizumab effective for drug-resistant gestational trophoblastic neioplasia. Lancet. 2017;390:2343–5.CrossRefPubMed
84.
go back to reference Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled-Filhart M. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;9(10027):1540–50.CrossRef Herbst RS, Baas P, Kim DW, Felip E, Pérez-Gracia JL, Han JY, Molina J, Kim JH, Arvis CD, Ahn MJ, Majem M, Fidler MJ, de Castro G, Garrido M, Lubiniecki GM, Shentu Y, Im E, Dolled-Filhart M. Pembrolizumab versus docetaxel for previously treated, PD-L1-positive, advanced non-small-cell lung cancer (KEYNOTE-010): a randomised controlled trial. Lancet. 2016;9(10027):1540–50.CrossRef
85.
go back to reference Guleria I, Khosroshahi A, Ansari MJ, Habicht A, Azuma M, Yagita H, Noelle RJ, Coyle A, Mellor AL, Khoury SJ. A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med. 2005;202(2):231–7.CrossRefPubMedPubMedCentral Guleria I, Khosroshahi A, Ansari MJ, Habicht A, Azuma M, Yagita H, Noelle RJ, Coyle A, Mellor AL, Khoury SJ. A critical role for the programmed death ligand 1 in fetomaternal tolerance. J Exp Med. 2005;202(2):231–7.CrossRefPubMedPubMedCentral
86.
87.
go back to reference Nehar-Belaid D, Courau T, Dérian N, Florez L, Ruocco MG, Klatzmann D. Regulatory T cells orchestrate similar immune evasion of fetuses and tumors in mice. J Immunol. 2016;196(2):678–9.CrossRefPubMed Nehar-Belaid D, Courau T, Dérian N, Florez L, Ruocco MG, Klatzmann D. Regulatory T cells orchestrate similar immune evasion of fetuses and tumors in mice. J Immunol. 2016;196(2):678–9.CrossRefPubMed
88.
go back to reference Donaghay M, Lessey BA. Uterine receptivity: alterations associated with benign gynecological disease. Semin Reprod Med. 2007;5(6):461–75.CrossRef Donaghay M, Lessey BA. Uterine receptivity: alterations associated with benign gynecological disease. Semin Reprod Med. 2007;5(6):461–75.CrossRef
89.
go back to reference Blesa D, Ruiz-Alonso M, Simón C. Clinical management of endometrial receptivity. Semin Reprod Med. 2014;32(5):410–3.CrossRefPubMed Blesa D, Ruiz-Alonso M, Simón C. Clinical management of endometrial receptivity. Semin Reprod Med. 2014;32(5):410–3.CrossRefPubMed
90.
go back to reference Teh WT, McBain J. Rogers P What is the contribution of embryo-endometrial asynchrony to implantation failure? J Assist Reprod Genet. 2016;33(11):1419–30.CrossRefPubMedPubMedCentral Teh WT, McBain J. Rogers P What is the contribution of embryo-endometrial asynchrony to implantation failure? J Assist Reprod Genet. 2016;33(11):1419–30.CrossRefPubMedPubMedCentral
91.
go back to reference Li SJ, Wang TS, Qin FN, Huang Z, Liang XH, Gao F, Song Z, Yang ZM. Differential regulation of receptivity in two uterine horns of a recipient mouse following asynchronous embryo transfer. Sci Rep. 2015;4(5):15897.CrossRef Li SJ, Wang TS, Qin FN, Huang Z, Liang XH, Gao F, Song Z, Yang ZM. Differential regulation of receptivity in two uterine horns of a recipient mouse following asynchronous embryo transfer. Sci Rep. 2015;4(5):15897.CrossRef
92.
go back to reference Macklon NS, Brosens JJ. The human endometrium as a sensor of embryo quality. Biol Reprod. 2014;91(4):98.CrossRefPubMed Macklon NS, Brosens JJ. The human endometrium as a sensor of embryo quality. Biol Reprod. 2014;91(4):98.CrossRefPubMed
93.
go back to reference Sandra O, Constant F, Vitorino Carvalho A, Eozénou C, Valour D, Mauffré V, Hue I, Charpigny G. Maternal organism and embryo biosensoring: insights from ruminants. mJ Reprod Immunol. 2015;108:105–13.CrossRef Sandra O, Constant F, Vitorino Carvalho A, Eozénou C, Valour D, Mauffré V, Hue I, Charpigny G. Maternal organism and embryo biosensoring: insights from ruminants. mJ Reprod Immunol. 2015;108:105–13.CrossRef
94.
go back to reference Leslie M. Two trials test whether influenza vaccinations make the body more accepting of IVF embryos. Science. 2017;355(6331):1247–8.CrossRefPubMed Leslie M. Two trials test whether influenza vaccinations make the body more accepting of IVF embryos. Science. 2017;355(6331):1247–8.CrossRefPubMed
95.
go back to reference Gleicher N, Kushnir VA, Barad DH. Therapeutic interventions into early stages of follicle maturation: a new treatment paradigm after over 50 years of modern infertility therapy. Endocrinology. 2013;154(10):3498–501.CrossRefPubMed Gleicher N, Kushnir VA, Barad DH. Therapeutic interventions into early stages of follicle maturation: a new treatment paradigm after over 50 years of modern infertility therapy. Endocrinology. 2013;154(10):3498–501.CrossRefPubMed
96.
go back to reference Goldman KN, Chenette D, Arju R, Duncan FE, Keefe DL, Grifo JA. Schneider RJmTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proc Natl Acad Sci USA. 2017;114(12):3186–91.CrossRefPubMedPubMedCentral Goldman KN, Chenette D, Arju R, Duncan FE, Keefe DL, Grifo JA. Schneider RJmTORC1/2 inhibition preserves ovarian function and fertility during genotoxic chemotherapy. Proc Natl Acad Sci USA. 2017;114(12):3186–91.CrossRefPubMedPubMedCentral
97.
go back to reference Kushnir VA, Seifer DB, Barad DH, Sen A, Gleicher N. Potential therapeutic applications of human anti-Müllerian hormone (AMH) analouges in reproductive medicine. J Assist Reprod Genet. 2017;34(9):1105–13.CrossRefPubMedPubMedCentral Kushnir VA, Seifer DB, Barad DH, Sen A, Gleicher N. Potential therapeutic applications of human anti-Müllerian hormone (AMH) analouges in reproductive medicine. J Assist Reprod Genet. 2017;34(9):1105–13.CrossRefPubMedPubMedCentral
98.
go back to reference Prizant H, Gleicher N, Sen A. Androgen action in the ovary: balance is key. J Endocrinol. 2014;222(3):R141–51.CrossRefPubMed Prizant H, Gleicher N, Sen A. Androgen action in the ovary: balance is key. J Endocrinol. 2014;222(3):R141–51.CrossRefPubMed
99.
go back to reference Gleicher N, Barad DH. Dehydroepiandrosterone (DHEA) supplementationin diminished ovarian reserve (DOR). Reprod Biol Endocrinol. 2011;17(9):67.CrossRef Gleicher N, Barad DH. Dehydroepiandrosterone (DHEA) supplementationin diminished ovarian reserve (DOR). Reprod Biol Endocrinol. 2011;17(9):67.CrossRef
100.
go back to reference Kuang Y, Chen Q, Hong Q, Lyu Q, Ai A, Fu Y, Shoham Z. Double stimulations during the follicular and luteal phases of poor responders in IVF/ICSI programmes (Shanghai protocol). Reprod Biomed Online. 2014;29(6):684–91.CrossRefPubMed Kuang Y, Chen Q, Hong Q, Lyu Q, Ai A, Fu Y, Shoham Z. Double stimulations during the follicular and luteal phases of poor responders in IVF/ICSI programmes (Shanghai protocol). Reprod Biomed Online. 2014;29(6):684–91.CrossRefPubMed
101.
go back to reference Ubaldi FM, Capalbo A, Vaiarelli A, Cimadomo D, Colamaria S, Alviggi C, Trabucco E, Venturella R, Vajta G, Rienzi L. Follicular versus luteal phase ovarian stimulation during the same menstrual cycle (DuoStim) in a reduced ovarian reserve population results in a similar euploid blastocyst formation rate: new insight in ovarian reserve exploitation. Fertil Steril. 2016;105(6):1488–95.CrossRefPubMed Ubaldi FM, Capalbo A, Vaiarelli A, Cimadomo D, Colamaria S, Alviggi C, Trabucco E, Venturella R, Vajta G, Rienzi L. Follicular versus luteal phase ovarian stimulation during the same menstrual cycle (DuoStim) in a reduced ovarian reserve population results in a similar euploid blastocyst formation rate: new insight in ovarian reserve exploitation. Fertil Steril. 2016;105(6):1488–95.CrossRefPubMed
102.
go back to reference Gemzell CA, Diczfalusy E, Tillinger KG. Clinical effects of human pituitary follicle-stimulating hormone (FSH). J Clin Endocrinol Metab. 1958;18(12):1333–48.CrossRefPubMed Gemzell CA, Diczfalusy E, Tillinger KG. Clinical effects of human pituitary follicle-stimulating hormone (FSH). J Clin Endocrinol Metab. 1958;18(12):1333–48.CrossRefPubMed
103.
go back to reference Albert A, Borth R, Diczfalusy E, Loraine JA, Lunenfeld B, McArthur JW, Rosemberg E. Collaboration assays of two urinary preparations of human pituitary gonadotropins. J Clin Endocrinol Metab. 1958;18(10):1117–23.CrossRefPubMed Albert A, Borth R, Diczfalusy E, Loraine JA, Lunenfeld B, McArthur JW, Rosemberg E. Collaboration assays of two urinary preparations of human pituitary gonadotropins. J Clin Endocrinol Metab. 1958;18(10):1117–23.CrossRefPubMed
104.
go back to reference Palermo G, Joris H, Devroey P, Van Steirteghem AC. Pregnancies after injection of single spermatozoon into an oocyte. Lancet. 1992;340(8810):17–21.CrossRefPubMed Palermo G, Joris H, Devroey P, Van Steirteghem AC. Pregnancies after injection of single spermatozoon into an oocyte. Lancet. 1992;340(8810):17–21.CrossRefPubMed
Metadata
Title
Expected advances in human fertility treatments and their likely translational consequences
Author
Norbert Gleicher
Publication date
01-12-2018
Publisher
BioMed Central
Published in
Journal of Translational Medicine / Issue 1/2018
Electronic ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-018-1525-4

Other articles of this Issue 1/2018

Journal of Translational Medicine 1/2018 Go to the issue